通用中文 | 克唑替尼胶囊 | 通用外文 | Crizotinib Capsules |
品牌中文 | 品牌外文 | Xalkori | |
其他名称 | CRIZALK赛可瑞靶点ALK MET ROSI | ||
公司 | 辉瑞(Pfizer) | 产地 | 德国(Germany) |
含量 | 250mg | 包装 | 60粒/盒 |
剂型给药 | 口服 | 储存 | 室温 |
适用范围 | 晚期,转移性,非小细胞肺癌(NSCLC),变性淋巴瘤激酶(ALK),ROS1, |
通用中文 | 克唑替尼胶囊 |
通用外文 | Crizotinib Capsules |
品牌中文 | |
品牌外文 | Xalkori |
其他名称 | CRIZALK赛可瑞靶点ALK MET ROSI |
公司 | 辉瑞(Pfizer) |
产地 | 德国(Germany) |
含量 | 250mg |
包装 | 60粒/盒 |
剂型给药 | 口服 |
储存 | 室温 |
适用范围 | 晚期,转移性,非小细胞肺癌(NSCLC),变性淋巴瘤激酶(ALK),ROS1, |
以下资料仅供参考:
文案整理:Dr. Jasmine Ding
克唑替尼使用说明书:
美国FDA初次批准:2011
请仔细阅读说明书并在医师指导下使用:
【商品名称】
通用名称:克唑替尼
品牌名称:XALKORI
通用英文名称:Crizotinib
其他名称:Crizonix,赛可瑞 CRIZALK
【成分】
本品主要成分为克唑替尼
化学名:(R)-3- [1-(2,6-二氯-3-氟苯基)乙氧基] -5- [1-(哌啶-4-基)-1H-吡唑-4-基]吡啶-2-胺。
分子式:C21H22Cl2FN5O
分子量:450.34克/摩尔。
【适应症/功能主治】
克唑替尼是一种口服受体酪氨酸激酶抑制剂。适用于可用于经 FDA 批准的检测方法确定的变性淋巴瘤激酶(ALK)-阳性和ROS1阳性的局部晚期或转移非小细胞肺癌(NSCLC)患者的治疗。
【规格与型号】250 mg*60粒/瓶
【用法用量]
患者选择
本品必须在有使用经验的医疗机构中并在特定的专业技术人员指导下使用。服用本品前, 必须获得经充分验证的检测方法证实的 ALK 阳性评估结果。
推荐剂量
克唑替尼胶囊的推荐剂量为 250mg 口服,每日两次,直至疾病进展或患者无法耐受。对于无需透析的 严重肾损害(肌酐清除率<30ml/分钟)患者,克唑替尼胶囊的推荐剂量为 250 mg 口服,每日一次。胶 囊应整粒吞服。克唑替尼胶囊与食物同服或不同服均可。若漏服一剂克唑替尼胶囊,则补服漏服剂量的 药物,除非距下次服药时间短于 6 小时。如果在服药后呕吐,则在正常时间服用下一剂药物。
剂量调整
如果患者出现美国国立癌症研究所(NCI)不良事件通用术语标准(CTCAE,第 4.0 版)规定的严重 程度为 3 级或 4 级的不良事件,需一次或多次减少剂量,按以下方法减少剂量: 第一次减少剂量:口服,200 mg,每日两次 第二次减少剂量:口服,250 mg,每日一次 如果每日一次口服 250 mg 克唑替尼胶囊仍无法耐受,则永久停服 剂量减少指南参见表 1 和表 2。
表 1. 克唑替尼胶囊剂量调整——血液学毒性
CTCAE 级别 3 级 暂停给药直至恢复至≤2 级或以下,继续服用同一剂量。
4 级 暂停给药直至恢复至≤2 级或以下,继续按下一个较低剂量给药
( 淋巴细胞减少除外(除非伴随临床事件,例如,机会性感染)。
表 2. 克唑替尼胶囊剂量调整——非血液学毒性
标准 克唑替尼胶囊剂量
丙氨酸氨基转移酶(ALT)或天门冬氨 酸氨基转移酶(AST)升高大于正常值 上限(ULN)5 倍并伴有总胆红素小于 或等于正常值上限 1.5 倍 者 , 应暂停给药直至恢复至基线水平或者小于或等于正常值上限 3 倍,继续减少剂量给药
ALT 或 AST 升高大于正常值上限 3 倍, 同时总胆红素升高大于正常值上限 1.5 倍(未出现胆汁淤积或溶血)者应 永久停用本药
任何级别的药物相关间质性肺病/非感 染性肺炎 应永久停用本药
至少 2 个单独的 ECG 上 QTc 大于 500 ms 暂停给药直至基线水平或 QTc 小于 481 ms,继续减少剂量给 药
QTc 大于 500 ms 或与基线相比的变化 大于或等于 60 ms,并伴有尖端扭转型 室速、多形性室性心动过速或严重心律 失常的症状/体征者 应永久停用本药
心动过缓 a:(有症状、可能严重、具有 医学意义、需要医疗干预)者 应暂停用药,直到恢复为无症状性心动过缓或心率为 60 bpm 或 以上。并 评估已知会引起心动过缓的合并用药以及降压药物 如果确定并停用了导致心动过缓的合并用药或调整了其剂量, 继续服用先前恢复为无症状性心动过缓或心率为 60bpm 或以 上时的剂量 。如果确定没有引起心动过缓的合并用药,或未停用或调整引起 心动过缓的合并用药,继续减少剂量给药直至恢复为无症状性 心动过缓或心率为 60 bpm 或以上
心动过缓 a,b(危及生命、需要紧急干预): 如果确定没有引起心动过缓的合并用药,则永久停用本药 如果确定并停用了引起心动过缓的合并用药或调整了其剂量, 在恢复为无症状性心动过缓或心率为 60 bpm 或以上时,可在 频繁监测下继续用药 250 mg,每日一次
备注:a) 心率低于 60 次/分(bpm)。 b) 如果再次出现,永久停用。
应在每月和出现临床症状时监测包括白细胞分类计数的全血细胞计数,如果出现 3 或 4 级异常或发热 或感染时,应增加监测频度。
肝损害患者
目前尚未对肝损害的患者使用克唑替尼的情况进行研究。由于克唑替尼主要在肝脏代谢,肝损害很可 能升高克唑替尼的血浆浓度。因此,肝损害的患者使用克唑替尼胶囊进行治疗时应谨慎。(见【药代 动力学】)
肾损害患者
根据群体药代动力学分析,对轻度(肌酐清除率[CLcr]为 60 至 89 ml /分钟)和中度([CLcr]为 30 至 59ml/分钟)肾损害的患者不需要进行起始剂量调整。在无需透析的严重肾损伤([CLcr]小于 30ml/分钟) 患者中,克唑替尼的暴露量增加,推荐克唑替尼起始剂量为 250 mg,口服,每日一次。(见【药代动 力学】项下“特殊人群”)
【不良反应】 以下不良反应在说明书的其他部分有更加详细的论述:
肝毒性[见注意事项]。
间质性肺病/非感染性肺炎[见注意事项]
QT 间期延长[见注意事项]
心动过缓[见注意事项] 安全性数据主要来自超过 1200 例接受克唑替尼胶囊单药治疗的 ALK 阳性的转移性非小细胞肺癌患者, 起始剂量 250mg,口服,每日两次,连续用药。在研究 A8081005、A8081007 和 A8081014 中包括了 252 例中国患者。
由于各个临床研究的情况不尽相同,直接比较两种药物在不同临床研究中的不良反应发生率是不恰当 的,临床研究中的不良反应发生率也可能与临床实践中的情况有所不同。
克唑替尼胶囊最常见的不良反应(≥25%)为视觉异常、恶心、腹泻、呕吐、便秘、水肿、转氨酶升高 及疲乏。
ALK 阳性的转移性非小细胞肺癌-研究 A8081007 表 3 中的数据来自一项随机、多中心、活性药物对照、开放的试验(研究 A8081007)中入组的 343 例 ALK 阳性的转移性非小细胞肺癌患者。克唑替尼胶囊组(n=172)的患者口服克唑替尼胶囊 250 mg, 每日两次,直到记录到疾病进展、无法耐受治疗或研究人员确定患者不再获得临床效益为止。化疗组 总共有 171 例患者接受了培美曲塞 500 mg/m2(n=99)或多西他赛 75mg/m2 (n=72),通过静脉输注 给药,每隔三周一次,直到记录到疾病进展、无法耐受治疗或研究人员确定患者不再获得临床效益为 止。化疗组患者均接受培美曲塞,除非在一线或维持性治疗中已经接受了培美曲塞。接受克唑替尼胶 囊和接受化疗治疗的患者的中位研究治疗持续时间分别为 7.1 个月和 2.8 个月。 64 例(37.2%)接受克唑替尼胶囊治疗的患者和 40 例(23.4%)化疗组患者报告了严重不良反应。接 受克唑替尼胶囊治疗的患者报告的最常见严重不良反应为肺炎(4.1%)、肺栓塞(3.5%)、呼吸困难 (2.3%)和间质性肺病(ILD;2.9%)。有 9 例(5%)接受克唑替尼胶囊治疗的患者出现了致死性不 良反应,包括:急性呼吸窘迫综合症、心律不齐、呼吸困难、肺炎、非感染性肺炎、肺栓塞、ILD、呼 吸衰竭和脓毒血症。 接受克唑替尼胶囊治疗的患者中,16%的患者因不良反应而需要减少剂量。导致克唑替尼胶囊减量的最 常见不良反应有 ALT 升高(7.6%),还包括一些同时伴随 AST 升高、QTc 间期延长(2.9%)和中性 粒细胞减少(2.3%)的患者。 接受克唑替尼胶囊治疗的患者中,因不良反应而停止治疗的比例为 17.0%。因接受克唑替尼胶囊治疗而 停止治疗的患者最常见不良反应为 ILD(1.7%)、ALT 和 AST 升高(1.2%)、呼吸困难(1.2%)和肺栓 塞(1.2%)。表 3 和表 4 汇总了接受克唑替尼胶囊治疗的患者常见不良反应和实验室检查异常。
表 3.研究 A8081007中报告的克唑替尼胶囊组高于化疗组的不良反应发生率(对于所有级别,高≥5%,或对于 3/4级,高≥2%)
不良反应 |
克唑替尼组(N=172) |
|
化疗组 (培美曲塞或多西他赛) (N=171) |
||
|
所有级别 (%) |
|
3/4级 (%) |
所有级别 (%) |
3/4级 (%) |
神经系统异常头晕 a 味觉障碍 晕厥 |
22 26 3 |
|
1 0 3 |
8 9 0 |
0 0 0 |
眼部异常视觉异常b |
60 |
|
0 |
9 |
0 |
心脏异常 心电图QT延长心动过缓c |
5 5 |
|
3 0 |
0 0 |
0 0 |
实验室检查 体重下降 |
10 |
|
1 |
4 |
0 |
胃肠道异常 呕吐 恶心 腹泻 便秘 消化不良 |
47 55 60 42 8 |
|
1 1 0 2 0 |
18 37 19 23 3 |
0 1 1 0 0 |
感染和侵染 上呼吸道感染 d |
26 |
|
0 |
13 |
1 |
呼吸道、胸部及纵隔异常 肺栓塞 e |
6 |
|
5 |
2 |
2 |
全身性疾病及给药部位各种反应 水肿 f |
31 |
|
0 |
16 |
0 |
a 头晕(平衡障碍、头晕、体位性头晕)
b 视觉异常(复视、畏光、闪光幻觉、视物模糊、视敏度降低、视觉损害、玻璃体飞蚊症) c 心动过缓(心动过缓、窦性心动过缓)
d 上呼吸道感染(喉炎、鼻咽炎、咽炎、鼻炎、上呼吸道感染) e 肺栓塞(肺动脉血栓形成、肺栓塞)
f 水肿(面部水肿、全身水肿、局部肿胀、局部水肿、水肿、外周水肿、眼周水肿)
接受克唑替尼胶囊治疗的患者中,总体发生率在 1%和 30%之间的其他不良反应包括食欲减退(27%)、 疲乏(27%)、神经病变(19%;感觉迟钝、步态障碍、感觉减退、肌无力、神经痛、周围神经病变、 感觉错乱、外周感觉神经病变、多发性神经病、皮肤烧灼感)、皮疹(9%)、ILD(4%;急性呼吸窘 迫综合症、ILD、非感染性肺炎)、肾囊肿(4%)和肝功能衰竭(1%)。
A8081007 研究中中国受试者的安全性信息(数据截至 2012 年 3 月 30 日) 研究 A8081007 中,29 例 ALK 阳性的转移性非小细胞肺癌(经 Vysis ALK Break-Apart FISH 检测鉴定) 中国(包括中国大陆、台湾和香港)患者被随机分配接受克唑替尼治疗(12 例,口服 250 mg,每日两 次)或化疗(17 例)。截至 2012 年 3 月 30 日,10 例患者(9 例接受克唑替尼治疗,1 例化疗)仍在 研究中。在此小群体中,未有非预期安全性相关发现。1 例接受克唑替尼患者和 3 例化疗患者因治疗相关的不良事件而永久停药。最常见严重不良反应(≥30%)包括腹泻(67%)、呕吐(67%)、视觉异 常*(50%)、中性粒细胞减少*(50%)、转氨酶升高*(42%)、恶心(33%)、咳嗽* (33%)、晕眩 *(33%)、贫血*(33%)、口腔炎*(33%)、低蛋白血症(33%)和白细胞降低 (33%)。
表 4.接受克唑替尼胶囊治疗的患者治疗后出现的 3 级或 4 级实验室检查结果异常汇总(发生率≥4%)
实验室检查异常 克唑替尼组 化疗组
任何级别 3/4 级 任何级别 3/4 级
血液学
中性粒细胞减少 49% 12% 28% 12%
淋巴细胞减少 51% 9% 60% 25%
血生化
ALT 升高 76% 17% 38% 4%
AST 升高 61% 9% 33% 0%
低钾血症 18% 4% 10% 1%
低磷血症 28% 5% 25% 6%
ALK 阳性的转移性非小细胞肺癌-研究
A8081005 研究 A8081005 中的安全性分析人群包括在临床试验中接受克唑替尼胶囊治疗的 934 例 ALK 阳性的转 移性非小细胞肺癌患者。他们的中位治疗持续时间为 23 周。分别有 23%和 12%的患者因治疗相关不良 事件而中断用药和减量。因治疗相关不良事件而导致永久停药的比例为 5%。最常见的不良反应(≥25%) 包括视觉异常(55%)、恶心(51%)、呕吐(46%)、腹泻(46%)、水肿(39%)、便秘(38%) 和疲乏(26%)。
A8081005 研究中中国受试者的安全性信息(数据截至 2013 年 11 月 30 日) 研究 A8081005 中的安全性分析人群包括 234 例来自中国(包括中国大陆、台湾和香港)的 ALK 阳性 的转移性非小细胞肺癌(经任一种 ALK 诊断检测鉴定)患者,在临床试验中接受克唑替尼治疗(口服 250 mg,每日两次。至截止日期,64 例患者(27.4%)仍在研究中。他们的中位治疗持续时间为 42.1 周。分别有 19.2%和 10.3%的患者因治疗相关不良事件而中断用药和减量。因治疗相关不良事件而导致永久停药的比例为 5.1%。最常见严重不良反应(≥30%)包括转氨酶升高*(55%)、呕吐(54%)、 视觉异常*(50%)、便秘(45%)、腹泻(44%)、恶心(37%)、白细胞减少症* (34%)、食欲减退 (34%)、水肿*(32%)。 *群集项(因某些特定医学概念或情况的频率可能因依赖单一 MedDRA 首选术语而低估,一些不良事件采用美国群集项 分析。)
特定药物不良反应描述
视觉异常
在临床试验中(n=1225),691 例(56%)患者出现了视觉异常,最常见的有视觉损害、闪光幻觉、视 物模糊或玻璃体飞蚊症。大多数(99%)患者都出现了 1 级或 2 级视觉不良反应。在临床研究中,1 例 患者出现了 3 级治疗相关视觉异常。 根据视觉症状评估问卷(Visual Symptom Assessment Questionnaire,VSAQ-ALK),研究 A8081007 中 接受克唑替尼胶囊治疗的患者报告的视觉障碍发生率高于化疗组患者。视觉异常一般在用药的第一周 内开始出现。研究 A8081007 中的大多数克唑替尼胶囊组患者(>50%)报告了视觉障碍;根据在患者 问卷中获得的结果,这些视觉障碍的发生率为每周 4-7 天,最长持续 1 分钟,对日常生活有轻微影响或 没有影响(评分为 0-3 分,最高分为 10 分)。
神经病变
在 1225 例患者中,有 235 例(19%)出现了神经病变,最常见的实际上为感觉神经病变。大多数事件 (95%)的严重程度为 1 级或 2 级。
肾囊肿
在研究 A8081007 中,克唑替尼胶囊治疗组和化疗组患者中分别有 7 例(4%)和 1 例(1%)出现肾囊 肿。接受克唑替尼胶囊治疗的患者出现的大部分肾囊肿为复杂性肾囊肿。观察到局部囊性侵及肾脏, 在某些病例中影像特征表明脓肿形成。但是,在临床试验中微生物检测均未确认肾脓肿。
【禁忌】 禁用于对克唑替尼或本品中任一成分过敏的患者。禁用于严重肝损害患者。
【注意事项】
肝毒性
在三项主要临床试验中,接受克唑替尼胶囊治疗的 1225 例患者中有 2 例(0.2%)发生了药物引起的致 命性肝毒性反应。7 例患者(0.6%)出现了 ALT 升高大于正常值上限 3 倍同时总胆红素升高大于正常 值上限 2 倍而碱性磷酸酶正常。此外,109 例患者(9.2%)的 ALT 升高大于正常值上限五倍。8 例患 者(0.7%)因转氨酶升高而需要永久停药。这些实验室结果通常没有症状,且一般在中断给药后可以 恢复。转氨酶升高通常发生在治疗的前 2 个月内。
肝功能检查包括ALT和总胆红素,在治疗开始的最初两个月应每两周检测一次,之后每月检测一次, 并且根据临床状况对转氨酶水平升高的患者更频繁地进行重复检测转氨酶、碱性磷酸酶或总胆红素升 高水平。根据表2的说明进行临时暂停给药、减量或永久停药。(见【用法用量】和【不良反应】)
间质性肺病(非感染性肺炎)
接受克唑替尼胶囊治疗的患者可能出现严重的、危及生命或致命性间质性肺病(ILD)/非感染性肺炎。 在临床试验中(n=1225),31 例(2.5%)接受克唑替尼胶囊治疗的患者出现了不同级别的 ILD,11 例 (0.9%)患者出现了 3 级或 4 级 ILD、6 例(0.5%)患者出现了致命事件。这些事件通常发生在开始治 疗后的最初 2 个月内。 应密切监测患者 ILD/非感染性肺炎的肺部症状指标。并排除其他潜在原因引起的 ILD/非感染性肺炎。 一旦患者出现治疗相关的 ILD/非感染性肺炎,应永久停止克唑替尼的治疗(参见【用法用量】及【不 良反应】)。
QT间期延长
接受克唑替尼胶囊治疗的患者可能出现 QTc 间期延长。在临床试验中(n=1225),34 例(2.7%)患者 出现 QTc 间期延长(所有级别),17 例(1.4%)患者在至少 2 个单独的 ECG 出现 QTc 大于 500 ms。。 先天性长 QT 综合征患者应避免服用克唑替尼胶囊。对于充血性心力衰竭、缓慢性心律失常和电解质异 常患者,以及正在服用已知可致 QT 间期延长药物的患者,使用本品治疗时应定期监测其心电图与电解 质。 QTc 大于 500 ms 或与基线相比的变化大于或等于 60 ms 并伴有尖端扭转型室速、多形性 室性心动过速或严重心律失常症状/体征的患者应永久停用克唑替尼胶囊。在至少 2 个单独 的 ECG 上 QTc 大于 500 ms 的患者应暂停使用克唑替尼胶囊,直到恢复至 QTc 小于或等 于 480 ms,然后继续按表 2 中描述的减少剂量(参见【用法用量】和【药代动力学】)。
心动过缓
接受克唑替尼胶囊治疗的患者可能会出现有症状的心动过缓。在不同的临床试验里接受克唑替尼治疗 的 1174 例患者中,出现心动过缓且心率<50 次/分的比例为 11%。在研究 A8081007 中,2.9%接受 XALKORI 胶囊治疗的患者出现了 3 级晕厥,接受化疗的患者未出现此类事件。 尽可能避免克唑替尼与其他已知可引起心动过缓的药物(如 β-受体阻滞剂、非二氢吡啶类钙通道阻滞 剂、可乐定和地高辛)同时使用。应定期监测心率和血压。如果出现不会危及生命的症状性心动过缓, 暂停使用克唑替尼胶囊直到恢复为无症状性心动过缓或心率为 60 bpm 或以上,重新评估合并用药,并 调整克唑替尼胶囊的剂量。如果克唑替尼胶囊引起危及生命的心动过缓,应永久停用本药;如果与已 知可引起心动过缓或低血压的合并用药有关,应暂停使用克唑替尼胶囊直到恢复为无症状性心动过缓 或心率为 60 bpm 或以上,如果可以停用或调整合并用药的剂量,则可在频繁监测下继续使用克唑替尼 胶囊 250 mg,每日一次(见【用法用量】和【不良反应】)。
胚胎毒性
根据克唑替尼胶囊的作用机制,妊娠妇女服用后可能会给胎儿带来伤害。
在大鼠非临床研究中,在暴露量约等于人体临床推荐剂量(250 mg,每日两次)时克唑替尼胶囊具有胚胎毒性和胎儿毒性。目前 尚未针对妊娠妇女服用克唑替尼胶囊进行充分且良好对照的研究。若在妊娠期间服用克唑替尼胶囊, 或患者在服药期间怀孕,则应告知其该药对胎儿的潜在危害(见【孕妇及哺乳期妇女用药】)。
ALK 检测 2013 年 6 月发布的《中国间变性淋巴瘤激酶(ALK)阳性非小细胞肺癌诊断专家共识》(2013 版)、 《中国表皮生长因子受体基因突变和间变性淋巴瘤激酶融合基因阳性非小细胞肺癌诊断治疗指南》 (2013 版)推荐,目前针对 ALK 融合基因检测常用的方法主要有 3 种:荧光原位杂交(FISH)、基 于聚合酶链反应(PCR)扩增基础上的技术和针对融合蛋白表达的免疫组织化学法(IHC)。应根据目 前 ALK 融合基因检测各种方法的优缺点、临床样本的特点和实验室的条件,按合理的检测流程,选择 合适的检测方法。 在选择使用克唑替尼治疗的患者时,必须由受过培训的专业技术人员采用经充分验证过的检测方法诊 断为 ALK 阳性非小细胞肺癌(NSCLC)。在临床研究 A8081005、A8081007 及 A8081014 中使用的是 雅培公司的 Vysis ALK Break Apart FISH(荧光原位杂交)探针试剂盒。该试剂盒正在中国注册申报中。 在中国,罗氏诊断产品(上海)有限公司的 Ventana anti-ALK 抗体诊断试剂盒(免疫组织化学法)及 厦门艾德生物医药科技有限公司的 EML4-ALK 融合基因检测试剂盒(荧光 PCR 法)均已经获得 CFDA 批准用于 ALK 融合基因的检测。
【孕妇及哺乳期妇女用药】
孕妇
基于克唑替尼胶囊的作用机制,妊娠妇女服用可能会给胎儿带来伤害。目前尚无对妊娠妇女服用克唑 替尼胶囊进行足够的且良好对照的研究。
在大鼠的非临床研究中,在暴露于近似于人体临床推荐剂量 (250 mg,每日两次)的情况下具有胚胎毒性和胎儿毒性。对器官形成期间的妊娠大鼠和家兔给予克 唑替尼,以研究药物对胚胎/胎儿发育的影响。当大鼠用药为≥50 mg/kg/天时(根据曲线下面积,大约 为推荐人体临床用药剂量的 0.6 倍),出现着床后流产增加。当大鼠用药剂量达 200 mg/kg/天(根据曲 线下面积,大约为推荐人体临床剂量时的 2.7 倍)或家兔用药剂量达 60 mg/kg/天(曲线下面积约为推 荐人体临床剂量时的 1.6 倍)时,胚胎的体重有所降低,但未出现致畸作用。
育龄妇女在服用克唑替尼胶囊进行治疗时应尽量避免怀孕。
服用本药的育龄妇女或服用本药的育龄妇女的伴侣,在治疗过程中以及完成治疗至少 90 天内应采取适 当的方法进行避孕。
若在妊娠期间服用本药,或患者或其伴侣在用药期间怀孕,则应告知其本品对胎 儿具有潜在危害。
哺乳期妇女
目前尚不明确克唑替尼及其代谢物是否会从乳汁中分泌。由于多数药物通常都会从乳汁中分泌,以及 婴儿若暴露于克唑替尼会发生潜在的严重不良反应,因此,决定哺乳期妇女是否终止哺乳或停止用药 非常重要。
【儿童用药】
目前尚无儿科患者使用克唑替尼胶囊的有效性和安全性数据。研究发现,给予幼鼠克唑替尼 150mg/kg/ 天,每日一次,连续 28 天后(根据曲线下面积,大约为 5.4 倍的推荐人体临床剂量),其长骨生长过程 中骨形成减少。关于儿科患者的其他潜在毒性,尚未在幼年动物中进行评价。
【老年用药】
研究A8081007中有 27例(16%)接受克唑替尼胶囊治疗的患者年龄为65 岁或65岁以上;研究 A8081005中有 152 例(16%)为 65 岁或 65 岁以上;研究 A8081001 中有 16 例(13%)为 65 岁或 65 岁以上。 未见这些患者与年轻患者在安全性或有效性方面存在总体差异。
【药物相互作用】
可能会增加克唑替尼血药浓度的药物
克唑替尼与 CYP3A 强抑制剂合用可能会导致克唑替尼血药浓度升高(参见【药代动力学】)。应避免 合并使用下列 CYP3A 强抑制剂(包括但不仅限于):阿扎那韦、克拉霉素、印地那韦、伊曲康唑、酮 康唑、奈法唑酮、奈非那韦、利托那韦、沙奎那韦、克拉霉素、泰利霉素、醋竹桃霉素和伏立康唑。 而西柚或西柚汁也可能会增加克唑替尼的血药浓度,应避免同时食用。与中度 CYP3A 抑制剂合并用药 时应谨慎。
可能会降低克唑替尼血药浓度的药物
克唑替尼与 CYP3A 强诱导剂合用可能会导致克唑替尼血药浓度降低(参见【药代动力学】)。应避免 合并使用下列 CYP3A 强诱导剂(包括但不仅限于):卡马西平、苯巴比妥、苯妥英钠、利福平、利福 布丁和圣约翰草。
克唑替尼可能改变其血药浓度的药物
克唑替尼在体内或体外均可抑制 CYP3A(参见【药代动力学】)。
服用克唑替尼的患者应避免与治疗 指数较窄的 CYP3A 底物(包括但不限于阿芬太尼、环孢霉素、双氢麦角胺、麦角胺、芬太尼、匹莫齐 特、奎尼丁、西罗莫司和他克莫司)合并使用。如果服用克唑替尼胶囊的患者需要合并使用这些治疗 指数较窄的 CYP3A 底物,可能需要减少 CYP3A 底物的剂量,因为药物合用可产生不良反应。
【药物过量】
目前尚无已知的克唑替尼胶囊药物过量的病例。目前尚无克唑替尼胶囊解毒剂。
【临床试验】
随机对照研究-研究 A8081007 一项随机、多中心、开放、活性药物对照研究(A8081007)证明了克唑替尼胶囊单药治疗 347 例 ALK 阳性的转移性非小细胞肺癌患者的疗效和安全性。在该研究中,患者之前接受过一种含铂化疗方案的 治疗。主要疗效结果为经独立影像学评价(IRR)确定的无进展生存期(PFS)。其他疗效结果包括经独 立影像学评价的客观缓解率(ORR)和总生存期(OS)。 患者被随机分配接受克唑替尼治疗(口服 250 mg,每日两次)(n=173)或化疗(n=174)。化疗包括培 美曲塞 500 mg/m2(如果此前未接受过培美曲塞治疗;n=99)或多西他赛 75 mg/m2(n=72),静脉给药 (IV),21 天为一个治疗周期。
两个治疗组的患者都继续治疗直到记录到疾病进展、无法耐受治疗或研 究人员确定患者不再有临床获益为止。随机分组按 ECOG 体力状态评分(0-1,2)、脑部转移(存在, 不存在)和先前 EGFR 酪氨酸激酶抑制剂治疗(是,否)进行分层。在随机分组之前,患者需诊断确 认为 ALK 阳性的非小细胞肺癌,使用经 FDA 批准的检测探针试剂盒 Vysis ALK Break-Apart FISH(荧 光原位杂交)鉴定。 总体研究人群的人口统计学特征为:56%为女性,中位年龄为 50 岁,基线 ECOG 体力状态评分为 0 分 (39%)或 1 分(52%),白人占 52%,亚洲人占 45%,4%患者吸烟,33%患者有吸烟史,63%患者从 不吸烟。至少 95%的患者疾病特征为转移性,至少 93%患者的肿瘤组织类型为腺癌。
研究 A8081007 结果证明,接受克唑替尼胶囊治疗的患者,其无进展生存期显著改善,且具有统计学意 义。总生存数据两组未显示统计学差异,因最初被随机分配到化疗组的 112 例(64%)患者在疾病进展后交叉接受了克唑替尼胶囊治疗,不能排除交叉治疗对于结果的影响。且目前总生存数据尚不成熟。 疗效结果总结于表 5 和图 1 中。
表 5. ALK 阳性的转移性非小细胞肺癌-疗效结果
克唑替尼组 (N=173 化疗组(N=174)
无进展生存期(基于独立影像学评价)
事件数(%) 100(58%) 127(73%)
疾病进展 84(49%) 119(68%)
死亡 16(9%) 8(5%)
中值, 月数(95% CI) 7.7(6.0,8.8) 3.0a(2.6,4.3)
HR(95% CI)b 0.49(0.37,0.64)
P 值 c <0.001
总生存期 d
事件数(%) 49(28%) 47(27%)
中值,月数(95% CI) 20.3(18.1,NR) 22.8(18.6,NR)
HR(95% CI)b 1.02(0.68,1.54)
P 值 c 0.54
肿瘤缓解率(基于独立影像学评价)
客观缓解率%(95% CI) 65%(58,72) 20%(14,26)
CR,n(%) 1(0.6%) 0
PR,n(%) 112(65%) 34(20%)
P 值 e <0.001
缓解持续时间
中值,月数(95% CI) 7.4(6.1,9.7) 5.6(3.4,8.3)
HR=风险比;CI=置信区间;NR=未达到;CR=完全缓解;PR=部分缓解
a 培美曲塞的中位无进展生存期为 4.2 个月。多西他赛的中位无进展生存期为 2.6 个月。
b 基于 Cox 比例风险分层分析
c 基于分层对数秩检验
d 对最终分析需要的总事件的 40%进行的中期总生存期分析
e 基于分层 Cochran-Mantel-Haenszel 检验
图 1. 研究 A8081007 中经独立影像学评价评估的无进展生存期的 Kaplan-Meier 曲线
A8081007 研究中中国受试者的疗效信息(数据截至 2012 年 3 月 30 日) 研究 A8081007 中,29 例 ALK 阳性的转移性非小细胞肺癌(经 Vysis ALK Break-Apart FISH 检测鉴定) 中国(包括中国大陆、台湾和香港)患者被随机分配接受克唑替尼治疗(12 例,口服 250 mg,每日两 次)或化疗(17 例)。其疗效结果与总体人群观察到的结果一致。中国受试者中,随机接受克唑替尼 胶囊治疗和化疗患者的中位无进展生存期分别为 5.6 个月(95% CI: 2.9, 8.5)和 1.5 个月(95% CI: 1.3, 4.0)。风险比率为 0.07(95 % CI: 0.01, 0.52),P 值为 0.0006(单侧,基于分层对数秩检验)。分析 时,克唑替尼胶囊治疗组和化疗组分别有 8 例(66.7%)(6 例疾病进展,2 例死亡)和 15 例(88.2 %) (14 例疾病进展,1 例死亡)PFS 事件数;分别有 4 例(33.3 %)和 0 例(0%)仍在随访。根据独立 影像学评价,与化疗组相比,克唑替尼胶囊也显著提高客观缓解率,P 值为 0.0019(双侧分层检验)。 随机分配接受克唑替尼治疗和化疗患者的客观缓解率分别为 75 %(95 % CI: 43 %, 95 %)和 6 %(95 % CI: 0.1 %, 29%),中位缓解持续时间分别为 24.1 周和 5.6 周。
单臂研究-研究 A8081005 和研究 A8081001 两项多国家、单臂研究(研究 A8081005 和研究 A8081001)证明了克唑替尼胶囊单药治疗 ALK 阳性的 转移性非小细胞肺癌的安全性和抗癌活性。两项研究的主要结果均为研究人员根据实体瘤疗效评价标 准(RECIST)评估的客观缓解率。两项研究中患者均口服 250 mg 克唑替尼胶囊,每日两次。 在研究 A8081005(n=934)中,57%为女性,中位年龄为 52 岁,基线 ECOG 体力状态评分为 0/1 分(82%) 或 2/3 分(18%),白人占 52%,亚洲人占 44%,4%患者吸烟,30%患者有吸烟史,66%患者从不吸烟。 92%患者的疾病特征为转移性;94%的患者被组织结构分类为腺癌。 研究 A8081005 中接受克唑替尼胶囊治疗的 934 例 ALK 阳性的转移性非小细胞肺癌患者中,765 例经 Vysis ALK Break-Apart FISH(荧光原位杂交)探针试剂盒确定为 ALK 阳性,且肿瘤缓解可评估;人口统计学特征与该研究的总体人群相似。他们的中位治疗持续时间为 5.5 个月。
据研究人员评估,有 8 例患者完全缓解,357 例患者部分缓解,客观缓解率为 48%(95% CI:44%,51%),中位缓解持续时 间为 11.0 个月。 A8081005 研究中中国受试者的疗效信息(数据截至 2013 年 11 月 30 日) 研究 A8081005 中,209 例 ALK 阳性的转移性非小细胞肺癌(经 Vysis ALK Break-Apart FISH 检测鉴定) 中国(包括中国大陆、台湾和香港)患者接受克唑替尼治疗(起始剂量 250 mg,每日两次),且肿瘤 缓解可评估。这些患者中,有 2 例患者完全缓解,96 例患者部分缓解,客观缓解率为 47% (95% CI: 40%, 54%),中位缓解持续时间为 36.0 周。在最初 8 周治疗期间,肿瘤客观缓解率为 79%。中位缓解持续 时间为 41.9 周。观察到 157 例(75.1%)PFS 事件数(131 例疾病进展,26 例死亡),中位无进展生存 期为 6.9 个月(95% CI: 5.5, 8.3),仍有 31 例(14.8%)患者在随访。
在研究 A8081001(n=119)中,50%为女性,中位年龄为 51 岁,基线 ECOG 体力状态评分为 0 分(35%) 或 1 分(53%),白人占 62%,亚洲人占 29%,不到 1%的患者吸烟,27%患者有吸烟史,72%患者从不吸 烟。96%的患者疾病特征为转移性;98%的患者癌症组织结构分类为腺癌,且 13%患者既往未因疾病转移 接受过全身治疗。
在研究 A8081001 中,119 例 ALK 阳性的转移性非小细胞肺癌患者接受克唑替尼胶囊治疗,中位治疗持 续时间为 32 周。根据研究人员的评估,客观缓解率为 61%(95% CI:52%,70%),中位缓解持续时间 为 11.1 个月。 尚有 2 项包括中国患者的随机对照国际多中心临床研究正在进行中: 研究 A8081014 是一项随机、对照、开放的 III 期研究,比较了以 250 mg BID 起始剂量口服克唑替尼与 培美曲塞/顺铂或培美曲塞/卡铂化疗一线治疗晚期 ALK 阳性非鳞状 NSCLC 的安全有效性。计划入组 334 例患者以 1:1 比例随机分配至 A 组(克唑替尼)或 B 组(化疗:培美曲塞/顺铂或培美曲塞/卡铂) 中。预计 2015 年第 4 季度完成。 研究 A8081029 是一项正在进行的多中心、随机、对照、开放的 III 期研究,在既往未经治疗的东亚 ALK 阳性晚期非鳞状 NSCLC 患者中比较了克唑替尼与化疗(即培美曲塞/顺铂或培美曲塞/卡铂)的安 全有效性。入组患者来自中国、台湾、香港、泰国和马来西亚。计划入组 200 例患者按 1:1 比例随机 分配至 A 组(克唑替尼)或 B 组(化疗:培美曲塞/顺铂或培美曲塞/卡铂)中。其中,150 例患者来自 中国,而其余 50 例患者来自其他亚洲国家。
预计 2015 年第 4 季度完成。 本品是按照有条件批准程序获准注册上市,这意味着申办方后续需提供进一步的临床研究数据包括在 ALK 阳性 NSCLC 患者中进行的与标准化疗比较的随机对照研究结果,CFDA 将每半年到一年对更新的 研究信息进行评价,并将必要的更新信息纳入说明书中。
【药理毒理】
药理作用
克唑替尼是一种酪氨酸激酶受体抑制剂,包括 ALK、肝细胞生长因子受体(HGFR,c-Met)、ROS1(c-cos) 和 RON。易位可促使 ALK 基因引起致癌融合蛋白的表达。ALK 融合蛋白形成可引起基因表达和信号 的激活和失调,进而促使表达这些蛋白的肿瘤细胞增殖和存活。克唑替尼在肿瘤细胞株中对 ALK、ROS1 和 c-Met 在细胞水平检测的磷酸化具有浓度依赖性抑制作用,对表达 EML4-ALK 或 NPM-ALK 融合蛋 白或 c-Met 的异种移植荷瘤小鼠具有抗肿瘤活性。 辉瑞机密 第 14 页 共 17 页 Version No: 20140918
毒理研究
遗传毒性:克唑替尼体外中国仓鼠卵巢细胞微核试验、人淋巴细胞染色体畸变试验和大鼠体内骨髓微 核试验结果均为阳性,Ames 试验结果阴性。
生殖毒性:克唑替尼未开展特定动物试验评价其对生育力的影响。大鼠重复给药毒性试验结果提示克 唑替尼对人类生育力和生殖功能具有潜在损害作用。研究显示,大鼠持续给药 28 天,给药剂量 为>50mg/kg/天时(AUC 约为推荐人临床用药剂量的 1.7 倍),雄性动物出现睾丸粗线期精母细胞退化。 大鼠重复给药 3 天,500mg/kg/天时(AUC 约为推荐人临床用药剂量 10 倍),雌性动物出现卵泡单细 胞坏死。 克唑替尼未进行致癌性试验研究。
【药代动力学】
吸收
口服单剂量克唑替尼,平均 4~6 小时克唑替尼的吸收达到峰值。每日服用 250mg 克唑替尼两次,15 天 内可达到并保持稳态血药浓度,平均累积率为 4.8。当剂量超出每日两次、每次 200~300 mg 的剂量范 围,稳态系统药物暴露(Cmin和 AUC)的增加略高于剂量的增加比例。 单剂量口服给药 250mg 后,克唑替尼的平均绝对生物利用度为 43%(范围:32%~66%)。 高脂膳食可使克唑替尼的 AUCinf和 Cmax降低约 14%。克唑替尼与食物同服或不同服均可。(参见【用 法用量】)。
分布
静脉注射 50mg 克唑替尼,药物几何平均分布容积(Vss)为 1772 升,说明药物自血浆广泛分布至组 织内。 在体外克唑替尼与人体血浆蛋白结合率为 91%,与药物浓度无关。体外研究表明克唑替尼为 P-糖蛋白 (P-gp)的底物。血液-血浆浓度比率约为 1。
代谢
参与克唑替尼代谢消除的主要酶是 CYP3A4/5。克唑替尼在人体的主要代谢途径是哌啶环氧化得到克唑 替尼酰胺和 O-脱羟产物,并在随后的第二步中 O-脱羟产物形成共轭。
清除
克唑替尼单剂量给药后,表观终末半衰期为 42 小时。 健康志愿者在服用单剂量 250mg 放射物标记的克唑替尼后,在其粪便和尿液中分别发现给药剂量 63% 和 22%的放射物标记的克唑替尼。粪便与尿液中克唑替尼原型药物分别约占给药剂量 53%和 2.3%。 克唑替尼 250mg 每日两次给药后在稳态时的平均表观清除率(CL/F)(60 升/小时)低于单剂量 250mg 口服给药后的(100 升/小时),可能归因于克唑替尼多次给药后 CYP3A 的自动抑制。
药物相互作用
CYP3A 抑制剂
克唑替尼单剂量口服 150mg,合并 CYP3A 强抑制剂酮康唑(200mg,每日两次),克唑替尼的 AUCinf 和 Cmax值与单独服用克唑替尼相比,分别增加约 3.2 倍和 1.4 倍。但是目前尚未确立 CYP3A 抑制剂对 稳态克唑替尼暴露量影响的大小 (参见【药物相互作用】)。
CYP3A 诱导剂
克唑替尼单剂量口服 250mg,合并服用 CYP3A 强诱导剂利福平(600mg,每日一次),克唑替尼的 AUCinf和 Cmax与单独服用克唑替尼相比分别降低 82%和 69%。但是尚未确立 CYP3A 诱导剂对于稳态 克唑替尼暴露量的影响的大小 (参见【药物相互作用】)。
提高胃内 PH 值的药物:在正常受试者中,连续 5 日每日服用埃索美拉唑 40 mg 后,同时单剂量口服 250 mg 克唑替尼,未发现克唑替尼暴露量出现临床相关变化(AUCinf降低 10%,Cmax无变化)。
CYP3A 底物:合用咪达唑仑与克唑替尼(每日两次,每次 250 mg,连续服用 28 天)的患者的 AUCinf 与单独口服咪达唑仑时相比增加了 3.7 倍,这表明克唑替尼是 CYP3A 的一种中度抑制剂(参加【药物 相互作用】)。
其他 CYP 底物
体外研究表明,尽管克唑替尼是 CYP1A2,CYP2C8,CYP2C9,CYP2C19 或 CYP2D6 等底物代谢的介 导抑制剂,但在临床上不会发生药物相互作用。 克唑替尼是 CYP2B6 的体外抑制剂。因此,克唑替尼与主要经 CYP2B6 代谢的药物合并使用时可能会 增加其血药浓度。 体外研究表明,尽管克唑替尼是 CYP1A2,CYP2B6,CYP2C8,CYP2C9,CYP2C19 或 CYP3A 底物代 谢的介导诱导剂,但在临床上不会发生药物相互作用。
UGT 底物
体外研究表明,尽管克唑替尼介导抑制属于UGT1A1、UGT1A4、UGT1A6、UGT1A9或UGT2B7底物 的药物代谢,但在临床上不会发生药物相互作用。
转运底物
在体外,临床相关浓度的克唑替尼会抑制 P-糖蛋白(P-gp)。因此,当克唑替尼与属于 P-糖蛋白底物 的药物合并使用时可能增加其血药浓度。
体外研究表明,临床相关浓度的克唑替尼会抑制肝脏摄取转运蛋白、有机阳离子转运蛋白 1(OCT1)、 肾脏摄取转运蛋白和有机阳离子转运蛋白 2(OCT2)。因此,当克唑替尼与属于 OCT1 或 OCT2 底物 的药物合并使用时可能增加其血药浓度。体外研究发现,临床相关浓度的克唑替尼不会抑制人类肝脏摄取转运蛋白 OATP1B1 或 OATP1B3 或者 肾脏摄取转运蛋白 OAT1 或 OAT3。
对其他转运蛋白的作用
体外研究表明,临床相关浓度的克唑替尼不会抑制肝脏外排胆盐输出泵转运蛋白(BSEP)。
特殊人群
肝损害:
目前尚未对肝损害的患者使用克唑替尼的情况进行研究。由于克唑替尼主要在肝脏代谢,肝 损害很可能升高克唑替尼的血浆浓度。临床研究排除纳入 AST 或 ALT>2.5 倍的正常值上限或由于肿瘤 肝转移而>5.0 倍正常值上限或总胆红素>1.5 倍的正常值上限的患者。因此,肝损害的患者使用克唑替 尼胶囊进行治疗时应谨慎。对来自研究 A8081007、A8081005 和 A8081001 的数据进行群体药代动力学 分析表明,基线总胆红素(0.1 至 2.1 mg/dL)或 AST 水平(7 至 124 U/L)不会对克唑替尼暴露量产生 临床相关影响。
肾损害:
研究 A8081007、A8081005、A8081001 中轻度([CLcr]为 60 至 89 ml/分钟,N=433)和中度 ([CLcr]为 30 至 59ml/分钟,N=137)肾损害患者的稳态谷浓度进行群体药代动力学分析,评估了克 唑替尼的药代动力学。轻度或中度肾损害不会对克唑替尼暴露量产生临床相关影响。。对 7 名无需透 析的严重肾损害(CLcr<30 ml /分钟)患者及 8 名肾功能正常([CLcr]≥90ml/分钟)的患者进行了一项研究。所有患者都接受 250 mg 单次口服剂量的克唑替尼胶囊。相对于肾功能正常的患者,严重肾损害 患者的克唑替尼平均 AUCinf和平均 Cmax分别升高 79%和 34%。且观察到克唑替尼活性代谢产物的 AUCinf和 Cmax也有类似变化(见【用法用量】)。
种族:亚洲患者(N=523)和非亚洲患者(N=691)的克唑替尼暴露量不存在临床相关差异。
年龄:根据研究 A8081007、A8081005 和 A8081001 的群体药代动力学分析,年龄不会对克唑替尼的暴 露量产生任何影响。
体重和性别:根据研究 A8081007、A8081005 和 A8081001 的群体药代动力学分析,体重或性别不会对 克唑替尼暴露量产生任何临床相关影响。
[心脏电生理 ]
研究对服用克唑替尼 250mg,每日两次的所有患者的 QT 间期延长的可能性进行了评估。单剂给药后, 达稳态时连续采集患者的三份心电图以评价克唑替尼对 QT 间期的影响。通过对心电图自动记录的评估 发现,1167 例患者中有 16 例(1.4%)QTcF≥500msec(采用 Fridericia 法校正 QT),1136 例患者中有 51 例(4.4%)其 QTcF 相比基线的增加≥60msec。一项药动学/药效学分析表明 QTcF 增加具有浓度依 赖性(见【注意事项】)。
【贮藏】
应在25°C以下;外出允许至15°-30°C。
文案整理:Dr. Jasmine Ding
HIGHLIGHTS OF PRESCRIBING INFORMATION
These highlights do not include all the information needed to use XALKORI® safely and effectively. See full prescribing information for XALKORI.
XALKORI® (crizotinib) capsules, for oral use
Initial U.S. Approval: 2011
RECENT MAJOR CHANGES
Indications and Usage (1) 7/2017
Dosage and Administration, Patient Selection (2.1) 7/2017
INDICATIONS AND USAGE
XALKORI is a kinase inhibitor indicated for the treatment of patients with metastatic non-small cell lung cancer (NSCLC) whose tumors are anaplastic lymphoma kinase (ALK) or ROS1-positive as detected by an FDA-approved test
DOSAGE AND ADMINISTRATION
· Recommended Dose: 250 mg orally, twice daily. (2.2)
· Renal Impairment: 250 mg orally, once daily in patients with severe renal impairment (creatinine clearance
5 WARNINGS AND PRECAUTIONS
. QT Interval Prolongation: Occurred in 2.1% of patients. Monitor electrocardiograms and electrolytes in patients who have a history of or predisposition for QTc prolongation, or who are taking medications that prolong QT. Temporarily suspend, dose reduce, or permanently discontinue XALKORI. (5.3)
· Bradycardia: XALKORI can cause bradycardia. Monitor heart rate and blood pressure regularly. Temporarily suspend, dose reduce, or permanently discontinue XALKORI. (5.4)
· Severe Visual Loss: Reported in 0.2% of patients. Discontinue XALKORI in patients with severe visual loss. Perform an ophthalmological evaluation. (5.5)
· Embryo-Fetal Toxicity: Can cause fetal harm. Advise females of reproductive potential of the potential risk to a fetus and use of effective contraception. (5.6, 8.1, 8.3)
ADVERSE REACTIONS
The most common adverse reactions (≥25%) are vision disorders, nausea, diarrhea, vomiting, edema, constipation, elevated transaminases, fatigue, decreased appetite, upper respiratory infection, dizziness, and neuropathy.
(6) To report SUSPECTED ADVERSE REACTIONS, contact Pfizer Inc. at 1-800-438-1985 or FDA at 1-800-FDA-1088 or www.fda.gov/medwatch
DRUG INTERACTIONS
· CYP3A Inhibitors: Avoid concurrent use of XALKORI with strong CYP3A inhibitors. (7.1)
· CYP3A Inducers: Avoid concurrent use of XALKORI with strong CYP3A inducers. (7.2)
· CYP3A Substrates: Avoid concurrent use of XALKORI with CYP3A substrates with narrow therapeutic indices. (7.3)
USE IN SPECIFIC POPULATIONS
Lactation: Do not breastfeed while taking XALKORI. (8.2) See 17 for PATIENT COUNSELING INFORMATION and FDA‐approved patient labeling. Revised: 7/2017
FULL PRESCRIBING INFORMATION
1 INDICATIONS AND USAGE
XALKORI is indicated for the treatment of patients with metastatic non-small cell lung cancer (NSCLC) whose tumors are anaplastic lymphoma kinase (ALK) or ROS1-positive as detected by an FDA-approved test [see Clinical Studies (14.1 and 14.2)].
2 DOSAGE AND ADMINISTRATION
2.1 Patient Selection
Select patients for the treatment of metastatic NSCLC with XALKORI based on the presence of ALK or ROS1 positivity in tumor specimens [see Indications and Usage (1) and Clinical Studies (14.1, 14.2)].
Information on FDA-approved tests for the detection of ALK and ROS1 rearrangements in NSCLC is available at http://www.fda.gov/MedicalDevices/ProductsandMedicalProcedures/InVitroDiagnostics/ucm301431.htm. 2.2 Recommended Dosing The recommended dose of XALKORI is 250 mg orally, twice daily until disease progression or no longer tolerated by the patient. The recommended dose of XALKORI in patients with severe renal impairment [creatinine clearance (CLcr)
Table 1. XALKORI Dose Modification – Hematologic Toxicitiesa
Grade 3 Withhold until recovery to Grade 2 or less, then resume at the same dose schedule
Grade 4 Withhold until recovery to Grade 2 or less, then resume at next lower dose a Except lymphopenia (unless associated with clinical events, e.g., opportunistic infections).
Table 2. XALKORI Dose Modification – Non-Hematologic Toxicities Criteria XALKORI Dosing Alanine aminotransferase (ALT) or aspartate aminotransferase (AST) elevation greater than 5 times upper limit of normal (ULN) with total bilirubin less than or equal to 1.5 times ULN Withhold until recovery to baseline or less than or equal to 3 times ULN, then resume at reduced dose. ALT or AST elevation greater than 3 times ULN with concurrent total bilirubin elevation greater than 1.5 times ULN (in the absence of cholestasis or hemolysis) Permanently discontinue. Any grade drug-related interstitial lung disease/pneumonitis Permanently discontinue. QT corrected for heart rate (QTc) greater than 500 ms on at least 2 separate electrocardiograms (ECGs) Withhold until recovery to baseline or to a QTc less than 481 ms, then resume at reduced dose. QTc greater than 500 ms or greater than or equal to 60 ms change from baseline with Torsade de pointes or polymorphic ventricular tachycardia or signs/symptoms of serious arrhythmia Permanently discontinue. Bradycardiaa (symptomatic, may be severe and medically significant, medical intervention indicated) Withhold until recovery to asymptomatic bradycardia or to a heart rate of 60 bpm or above. Evaluate concomitant medications known to cause bradycardia, as well as antihypertensive medications. If contributing concomitant medication is identified and discontinued, or its dose is adjusted, resume at previous dose upon recovery to asymptomatic bradycardia or to a heart rate of 60 bpm or above. If no contributing concomitant medication is identified, or if contributing concomitant medications are not discontinued or dose modified, resume at reduced dose upon recovery to asymptomatic bradycardia or to a heart rate of 60 bpm or above. Bradycardiaa,b (life-threatening consequences, urgent intervention indicated) Permanently discontinue if no contributing concomitant medication is identified. If contributing concomitant medication is identified and discontinued, or its dose is adjusted, resume at 250 mg once daily upon recovery to asymptomatic bradycardia or to a heart rate of 60 bpm or above, with frequent monitoring. Visual Loss (Grade 4 Ocular Disorder) Discontinue during evaluation of severe vision loss. a Heart rate less than 60 beats per minute (bpm). b Permanently discontinue for recurrence. Monitor complete blood counts including differential white blood cell counts monthly and as clinically indicated, with more frequent repeat testing if Grade 3 or 4 abnormalities are observed, or if fever or infection occurs.
3 DOSAGE FORMS AND STRENGTHS
· 250 mg capsules: hard gelatin capsule, size 0, pink opaque cap and body, with “Pfizer” on the cap and “CRZ 250” on the body.
· 200 mg capsules: hard gelatin capsule, size 1, white opaque body and pink opaque cap, with “Pfizer” on the cap and “CRZ 200” on the body.
4 CONTRAINDICATIONS
None.
5 WARNINGS AND PRECAUTIONS
5.1 Hepatotoxicity
Drug-induced hepatotoxicity with fatal outcome occurred in 2 (0.1%) of the 1719 patients treated with XALKORI across clinical trials. Concurrent elevations in alanine aminotransferase (ALT) or aspartate aminotransferase (AST) greater than or equal to 3 times the upper limit of normal (ULN) and total bilirubin greater than or equal to 2 times the ULN, with normal alkaline phosphatase, occurred in 10 patients ((Elevations in ALT or AST greater than 5 times the ULN occurred in 187 (11.2%) and 95 (5.7%) patients, respectively. Seventeen patients (1.0%) required permanent discontinuation due to elevated transaminases. Transaminase elevations generally occurred within the first 2 months of treatment.
Monitor liver function tests, including ALT, AST, and total bilirubin, every 2 weeks during the first 2 months of treatment, then once a month, and as clinically indicated, with more frequent repeat testing for increased liver transaminases, alkaline phosphatase, or total bilirubin in patients who develop transaminase elevations. Temporarily suspend, dose reduce, or permanently discontinue XALKORI as described in Table 2 [see Dosage and Administration (2.3) and Adverse Reactions (6)].
5.2 Interstitial Lung Disease (Pneumonitis)
Severe, life-threatening, or fatal interstitial lung disease (ILD)/pneumonitis can occur in patients treated with XALKORI. Across clinical trials (n=1719), 50 XALKORI-treated patients (2.9%) had ILD of any grade, 18 patients (1.0%) had Grade 3 or 4 ILD, and 8 patients (0.5%) had fatal ILD. Interstitial lung disease generally occurred within 3 months after the initiation of XALKORI. Monitor patients for pulmonary symptoms indicative of ILD/pneumonitis. Exclude other potential causes of ILD/pneumonitis, and permanently discontinue XALKORI in patients diagnosed with drug-related ILD/pneumonitis [see Dosage and Administration (2.3) and Adverse Reactions (6)].
5.3 QT Interval Prolongation
QTc prolongation can occur in patients treated with XALKORI. Across clinical trials, 34 of 1616 patients (2.1%) had QTcF (corrected QT for heart rate by the Fridericia method) greater than or equal to 500 ms and 79 of 1582 patients (5.0%) had an increase from baseline QTcF greater than or equal to 60 ms by automated machine-read evaluation of ECGs. Avoid use of XALKORI in patients with congenital long QT syndrome. Monitor ECGs and electrolytes in patients with congestive heart failure, bradyarrhythmias, electrolyte abnormalities, or who are takingmedications that are known to prolong the QT interval. Permanently discontinue XALKORI in patients who develop QTc greater than 500 ms or greater than or equal to 60 ms change from baseline with Torsade de pointes or polymorphic ventricular tachycardia or signs/symptoms of serious arrhythmia. Withhold XALKORI in patients who develop QTc greater than 500 ms on at least 2 separate ECGs until recovery to a QTc less than or equal to 480 ms, then resume XALKORI at a reduced dose as described in Table 2 [see Dosage and Administration (2.3) and Clinical Pharmacology (12.2)].
5.4 Bradycardia
Symptomatic bradycardia can occur in patients receiving XALKORI. Across clinical trials, bradycardia occurred in 219 (12.7%) of 1719 patients treated with XALKORI. Grade 3 syncope occurred in 2.4% of XALKORI-treated patients and in 0.6% of the chemotherapy-treated patients. Avoid using XALKORI in combination with other agents known to cause bradycardia (e.g., beta-blockers, non-dihydropyridine calcium channel blockers, clonidine, and digoxin) to the extent possible. Monitor heart rate and blood pressure regularly. In cases of symptomatic bradycardia that is not life-threatening, hold XALKORI until recovery to asymptomatic bradycardia or to a heart rate of 60 bpm or above, re-evaluate the use of concomitant medications, and adjust the dose of XALKORI. Permanently discontinue for life-threatening bradycardia due to XALKORI; however, if associated with concomitant medications known to cause bradycardia or hypotension, hold XALKORI until recovery to asymptomatic bradycardia or to a heart rate of 60 bpm or above, and if concomitant medications can be adjusted or discontinued, restart XALKORI at 250 mg once daily with frequent monitoring [see Dosage and Administration (2.3) and Adverse Reactions (6)].
5.5 Severe Visual Loss
Across all clinical trials, the incidence of Grade 4 visual field defect with vision loss was 0.2% (4/1719). Optic atrophy and optic nerve disorder have been reported as potential causes of vision loss. Discontinue XALKORI in patients with new onset of severe visual loss (best corrected vision less than 20/200 in one or both eyes). Perform an ophthalmological evaluation consisting of best corrected visual acuity, retinal photographs, visual fields, optical coherence tomography (OCT) and other evaluations as appropriate for new onset of severe visual loss. There is insufficient information to characterize the risks of resumption of XALKORI in patients with a severe visual loss; a decision to resume XALKORI should consider the potential benefits to the patient.
5.6 Embryo-Fetal Toxicity
Based on its mechanism of action, XALKORI can cause fetal harm when administered to a pregnant woman. In animal reproduction studies, oral administration of crizotinib in pregnant rats during organogenesis at exposures similar to those observed with the maximum recommended human dose resulted in embryotoxicity and fetotoxicity. Advise pregnant women of the potential risk to a fetus. Advise females of reproductive potential to use effective contraception during treatment with XALKORI and for at least 45 days following the final dose [see Use in Specific Populations (8.1, 8.3)]. Advise male patients with female partners of reproductive potential to use condoms during treatment with XALKORI and for at least 90 days after the final dose [see Use in Specific Populations (8.3) and Nonclinical Toxicology (13.1)].
6 ADVERSE REACTIONS
The following adverse reactions are discussed in greater detail in other sections of the labeling: · Hepatotoxicity [see Warnings and Precautions (5.1)]
· Interstitial Lung Disease (Pneumonitis)
[see Warnings and Precautions (5.2)] · QT Interval Prolongation [see Warnings and Precautions (5.3)] · Bradycardia [see Warnings and Precautions (5.4)] · Severe Visual Loss [see Warnings and Precautions (5.5)] 6.1 Clinical Trials Experience Because clinical trials are conducted under widely varying conditions, adverse reaction rates observed in the clinical trials of a drug cannot be directly compared to rates in the clinical trials of another drug and may not reflect the rates observed in clinical practice. The data in the Warnings and Precautions section reflect exposure to XALKORI in 1719 patients who received XALKORI 250 mg twice daily enrolled on Studies 1 (including an additional 109 patients who crossed over from the control arm), 2, 3, a single arm trial (n=1063) of ALK-positive NSCLC, and an additional ALK-positive NSCLC expansion cohort of a dose finding study (n=154) [see Warnings and Precautions (5)]. The data described below is based primarily on 343 patients with ALK-positive metastatic NSCLC who received XALKORI 250 mg twice daily from 2 open-label, randomized, active-controlled trials (Studies 1 and 2). The safety of XALKORI was also evaluated in 50 patients with ROS1-positive metastatic NSCLC from a single-arm study (Study 3). The most common adverse reactions (≥25%) of XALKORI are vision disorders, nausea, diarrhea, vomiting, edema, constipation, elevated transaminases, fatigue, decreased appetite, upper respiratory infection, dizziness, and neuropathy.
Previously Untreated ALK-Positive Metastatic NSCLC - Study 1
The data in Table 3 are derived from 340 patients with ALK-positive metastatic NSCLC who had not received previous systemic treatment for advanced disease who received treatment in a randomized, multicenter, open-label, active-controlled trial (Study 1). Patients in the XALKORI arm (n=171) received XALKORI 250 mg orally twice daily until documented disease progression, intolerance to therapy, or the investigator determined that the patient was no longer experiencing clinical benefit. A total of 169 patients in the chemotherapy arm received pemetrexed 500 mg/m2 in combination with cisplatin 75 mg/m2 (n=91) or carboplatin at a dose calculated to produce an area under the concentration-time curve (AUC) of 5 or 6 mg min/mL (n=78). Chemotherapy was given by intravenous infusion every 3 weeks for up to 6 cycles, in the absence of dose-limiting chemotherapy-related toxicities. After 6 cycles, patients remained on study with no additional anticancer treatment, and tumor assessments continued until documented disease progression. The median duration of study treatment was 10.9 months for patients in the XALKORI arm and 4.1 months for patients in the chemotherapy arm. Median duration of treatment was 5.2 months for patients who received XALKORI after cross over from chemotherapy. Across the 340 patients who were treated in Study 1, the median age was 53 years; 16% of patients were older than 65 years. A total of 62% of patients were female and 46% were Asian. Serious adverse events were reported in 58 patients (34%) treated with XALKORI. The most frequent serious adverse events reported in patients treated with XALKORI were dyspnea (4.1%) and pulmonary embolism (2.9%). Fatal adverse events in XALKORI-treated patients occurred in 2.3% patients, consisting of septic shock, acute respiratory failure, and diabetic ketoacidosis. 6 Reference ID: 4127887 Dose reductions due to adverse reactions were required in 6.4% of XALKORI-treated patients. The most frequent adverse reactions that led to dose reduction in these patients were nausea (1.8%) and elevated transaminases (1.8%). Permanent discontinuation of XALKORI treatment for adverse reactions was 8.2%. The most frequent adverse reactions that led to permanent discontinuation in XALKORI-treated patients were elevated transaminases (1.2%), hepatotoxicity (1.2%), and ILD (1.2%).
Tables 3 and 4 summarize common adverse reactions and laboratory abnormalities in XALKORI-treated patients.
Table 3. Adverse Reactions Reported at a Higher Incidence (≥5% Higher for All Grades or ≥2% Higher for Grades 3-4) with XALKORI than Chemotherapy in Study 1† Adverse Reaction XALKORI (N=171) Chemotherapy (Pemetrexed/Cisplatin or Pemetrexed/Carboplatin) (N=169) All Grades (%) Grade 3-4 (%) All Grades (%) Grade 3-4 (%) Cardiac Disorders Electrocardiogram QT prolonged Bradycardiaa 6 14 2 1 2 1 0 0 Eye Disorders Vision disorderb 71 1 10 0 Gastrointestinal Disorders Vomiting Diarrhea Constipation Dyspepsia Dysphagia Abdominal painc Esophagitisd 46 61 43 14 10 26 6 2 2 2 0 1 0 2 36 13 30 2 2 12 1 3 1 0 0 1 0 0 General Disorders and Administration Site Conditions Edemae Pyrexia 49 19 1 0 12 11 1 1 Infections and Infestations Upper respiratory infectionf 32 0 12 1 Investigations Increased weight 8 1 2 0 Musculoskeletal and Connective Tissue Disorders Pain in extremity Muscle spasm 16 8 0 0 7 2 0 1 Nervous System Disorders Dizzinessg Dysgeusia Headache 18 26 22 0 0 1 10 5 15 1 0 0 † Adverse reactions were graded using NCI CTCAE version 4.0. Includes cases reported within the clustered terms: a Bradycardia (Bradycardia, Sinus bradycardia). b Vision Disorder (Diplopia, Photophobia, Photopsia, Reduced visual acuity, Blurred vision, Vitreous floaters, Visual impairment). c Abdominal pain (Abdominal discomfort, Abdominal pain, Lower abdominal pain, Upper abdominal pain, Abdominal tenderness). d Esophagitis (Esophagitis, Esophageal ulcer). e Edema (Edema, Peripheral edema, Face edema, Generalized edema, Local swelling, Periorbital edema). f Upper respiratory infection (Nasopharyngitis, Pharyngitis, Rhinitis, Upper respiratory tract infection). g Dizziness (Balance disorder, Dizziness, Postural dizziness, Presyncope). Additional adverse reactions occurring at an overall incidence between 1% and 60% in patients treated with XALKORI included nausea (56%), decreased appetite (30%), fatigue (29%), neuropathy (21%; gait disturbance, hypoesthesia, muscular weakness, neuralgia, neuropathy peripheral, paresthesia, peripheral sensory neuropathy, polyneuropathy, sensory disturbance), rash (11%), renal cyst (5%), ILD (1%; ILD, pneumonitis), syncope (1%), and decreased blood testosterone (1%; hypogonadism).
Table 4. Laboratory Abnormalities with Grade 3 or 4 Incidence of ≥4% in XALKORI-Treated Patients in Study 1 Laboratory Abnormality XALKORI Any Grade (%) Grade 3-4 (%) Chemotherapy Any Grade (%) Grade 3-4 (%) Hematology Neutropenia Lymphopenia 52 48 11 7 59 53 16 13 Chemistry ALT elevation AST elevation Hypophosphatemia 79 66 32 15 8 10 33 28 21 2 1 6 Additional laboratory test abnormality in patients treated with XALKORI was an increase in creatinine (Any Grade: 99%; Grade 3: 2%; Grade 4: 0%) compared to the chemotherapy arm (Any Grade: 92%; Grade 3: 0%; Grade 4: 1%). Previously Treated ALK-Positive Metastatic NSCLC - Study 2 The data in Table 5 are derived from 343 patients with ALK-positive metastatic NSCLC enrolled in a randomized, multicenter, active-controlled, open-label trial (Study 2). Patients in the XALKORI arm (n=172) received XALKORI 250 mg orally twice daily until documented disease progression, intolerance to therapy, or the investigator determined that the patient was no longer experiencing clinical benefit. A total of 171 patients in the chemotherapy arm received pemetrexed 500 mg/m2 (n=99) or docetaxel 75 mg/m2 (n=72) by intravenous infusion every 3 weeks until documented disease progression, intolerance to therapy, or the investigator determined that the patient was no longer experiencing clinical benefit. Patients in the chemotherapy arm received pemetrexed unless they had received pemetrexed as part of first-line or maintenance treatment. The median duration of study treatment was 7.1 months for patients who received XALKORI and 2.8 months for patients who received chemotherapy. Across the 347 patients who were randomized to study treatment (343 received at least 1 dose of study treatment), the median age was 50 years; 14% of patients were older than 65 years. A total of 56% of patients were female and 45% of patients were Asian. Serious adverse reactions were reported in 64 patients (37.2%) treated with XALKORI and 40 patients (23.4%) in the chemotherapy arm. The most frequent serious adverse reactions reported in patients treated with XALKORI were pneumonia (4.1%), pulmonary embolism (3.5%), dyspnea (2.3%), and ILD (2.9%). Fatal adverse reactions in XALKORI-treated patients in Study 2 occurred in 9 (5%) patients, consisting of: acute respiratory distress syndrome, arrhythmia, dyspnea, pneumonia, pneumonitis, pulmonary embolism, ILD, respiratory failure, and sepsis. Dose reductions due to adverse reactions were required in 16% of XALKORI-treated patients. The most frequent adverse reactions that led to dose reduction in the patients treated with XALKORI were ALT elevation (7.6%) including some patients with concurrent AST elevation, QTc prolongation (2.9%), and neutropenia (2.3%). XALKORI was discontinued for adverse reactions in 15% of patients. The most frequent adverse reactions that led to discontinuation of XALKORI were ILD (1.7%), ALT and AST elevation (1.2%), dyspnea (1.2%), and pulmonary embolism (1.2%). Tables 5 and 6 summarize common adverse reactions and laboratory abnormalities in XALKORI-treated patients. 9 Reference ID: 4127887 Table 5. Adverse Reactions Reported at a Higher Incidence (≥5% Higher for All Grades or ≥2% Higher for Grades 3/4) with XALKORI than Chemotherapy in Study 2† Adverse Reaction XALKORI (N=172) Chemotherapy (Pemetrexed or Docetaxel) (N=171) All Grades (%) Grade 3-4 (%) All Grades (%) Grade 3-4 (%) Nervous System Disorders Dizzinessa Dysgeusia Syncope 22 26 3 1 0 3 8 9 0 0 0 0 Eye Disorders Vision disorderb 60 0 9 0 Cardiac Disorders Electrocardiogram QT prolonged Bradycardiac 5 5 3 0 0 0 0 0 Investigations
Decreased weight 10 1 4 0 Gastrointestinal Disorders Vomiting Nausea Diarrhea Constipation Dyspepsia 47 55 60 42 8 1 1 0 2 0 18 37 19 23 3 0 1 1 0 0 Infections and Infestations Upper respiratory infectiond 26 0 13 1 Respiratory, Thoracic and Mediastinal Disorders Pulmonary embolisme 6 5 2 2 General Disorders and Administration Site Conditions Edemaf 31 0 16 0 † Adverse reactions were graded using NCI CTCAE version 4.0. Includes cases reported within the clustered terms: a Dizziness (Balance disorder, Dizziness, Postural dizziness). b Vision Disorder (Diplopia, Photophobia, Photopsia, Blurred vision, Reduced visual acuity, Visual impairment, Vitreous floaters). c Bradycardia (Bradycardia, Sinus bradycardia). d Upper respiratory infection (Laryngitis, Nasopharyngitis, Pharyngitis, Rhinitis, Upper respiratory tract infection). e Pulmonary embolism (Pulmonary artery thrombosis, Pulmonary embolism). f Edema (Face edema, Generalized edema, Local swelling, Localized edema, Edema, Peripheral edema, Periorbital edema). Additional adverse reactions occurring at an overall incidence between 1% and 30% in patients treated with XALKORI included decreased appetite (27%), fatigue (27%), neuropathy (19%; dysesthesia, gait disturbance, hypoesthesia, muscular weakness, neuralgia, peripheral neuropathy, paresthesia, peripheral sensory neuropathy, polyneuropathy, burning sensation in skin), rash (9%), ILD (4%; acute respiratory distress syndrome, ILD, pneumonitis), renal cyst (4%), esophagitis (2%), hepatic failure (1%), and decreased blood testosterone (1%; hypogonadism). 10 Reference ID: 4127887 Table 6. Laboratory Abnormalities with Grade 3 or 4 Incidence of ≥4% in XALKORI-Treated Patients in Study 2 Laboratory Abnormality XALKORI Any Grade (%) Grade 3-4 (%) Chemotherapy Any Grade (%) Grade 3-4 (%) Hematology Neutropenia Lymphopenia 49 51 12 9 28 60 12 25 Chemistry ALT elevation AST elevation Hypokalemia Hypophosphatemia 76 61 18 28 17 9 4 5 38 33 10 25 4 0 1 6 Additional laboratory test abnormality in patients treated with XALKORI was an increase in creatinine (Any Grade: 96%; Grade 3: 1%; Grade 4: 0%) compared to the chemotherapy arm (Any Grade: 72%; Grade 3: 0%; Grade 4: 0%).
ROS1-Positive Metastatic NSCLC - Study 3
The safety profile of XALKORI from Study 3, which was evaluated in 50 patients with ROS1-positive metastatic NSCLC, was generally consistent with the safety profile of XALKORI evaluated in patients with ALK-positive metastatic NSCLC (n=1669). Vision disorders occurred in 92% of patients in Study 3; 90% were Grade 1 and 2% were Grade 2. The median duration of exposure to XALKORI was 34.4 months.
Description of Selected Adverse Drug Reactions
Vision disorders
Vision disorders, most commonly visual impairment, photopsia, blurred vision, or vitreous floaters, occurred in 1084 (63.1%) of 1719 patients. The majority (95%) of these patients had Grade 1 visual adverse reactions. There were 13 (0.8%) patients with Grade 3 and 4 (0.2%) patients with Grade 4 visual impairment. Based on the Visual Symptom Assessment Questionnaire (VSAQ-ALK), patients treated with XALKORI in Studies 1 and 2 reported a higher incidence of visual disturbances compared to patients treated with chemotherapy. The onset of vision disorder generally was within the first week of drug administration. The majority of patients on the XALKORI arms in Studies 1 and 2 (>50%) reported visual disturbances which occurred at a frequency of 4-7 days each week, lasted up to 1 minute, and had mild or no impact (scores 0 to 3 out of a maximum score of 10) on daily activities as captured in the VSAQ-ALK questionnaire.
Neuropathy
Neuropathy, most commonly sensory in nature, occurred in 435 (25%) of 1719 patients. Most events (95%) were Grade 1 or Grade 2 in severity.
Renal cysts
Renal cysts were experienced by 52 (3%) of 1719 patients. The majority of renal cysts in XALKORI-treated patients were complex. Local cystic invasion beyond the kidney occurred, in some cases with imaging characteristics suggestive of abscess formation. However, across clinical trials no renal abscesses were confirmed by microbiology tests.
Renal impairment
The estimated glomerular filtration rate (eGFR) decreased from a baseline median of 96.42 mL/min/1.73 m2 (n=1681) to a median of 80.23 mL/min/1.73 m2 at 2 weeks (n=1499) in patients with ALK-positive advanced 11 Reference ID: 4127887 NSCLC who received XALKORI in clinical trials. No clinically relevant changes occurred in median eGFR from 12 to 104 weeks of treatment. Median eGFR slightly increased (83.02 mL/min/1.73 m2 ) 4 weeks after the last dose of XALKORI
7 DRUG INTERACTIONS
7.1 Drugs That May Increase Crizotinib Plasma Concentrations Coadministration of crizotinib with strong cytochrome P450 (CYP) 3A inhibitors increases crizotinib plasma concentrations [see Clinical Pharmacology (12.3)]. Avoid concomitant use of strong CYP3A inhibitors, including but not limited to atazanavir, clarithromycin, indinavir, itraconazole, ketoconazole, nefazodone, nelfinavir, ritonavir, saquinavir, telithromycin, troleandomycin, and voriconazole. Avoid grapefruit or grapefruit juice which may also increase plasma concentrations of crizotinib. Exercise caution with concomitant use of moderate CYP3A inhibitors.
7.2 Drugs That May Decrease Crizotinib Plasma Concentrations Coadministration of crizotinib with strong CYP3A inducers decreases crizotinib plasma concentrations [see Clinical Pharmacology (12.3)]. Avoid concomitant use of strong CYP3A inducers, including but not limited to carbamazepine, phenobarbital, phenytoin, rifabutin, rifampin, and St. John’s Wort.
7.3 Drugs Whose Plasma Concentrations May Be Altered By Crizotinib Crizotinib inhibits CYP3A both in vitro and in vivo [see Clinical Pharmacology (12.3)]. Avoid concomitant use of CYP3A substrates with narrow therapeutic range, including but not limited to alfentanil, cyclosporine, dihydroergotamine, ergotamine, fentanyl, pimozide, quinidine, sirolimus, and tacrolimus in patients taking XALKORI. If concomitant use of these CYP3A substrates with narrow therapeutic range is required in patients taking XALKORI, dose reductions of the CYP3A substrates may be required due to adverse reactions.
8 USE IN SPECIFIC POPULATIONS
8.1 Pregnancy Risk Summary
Based on its mechanism of action, XALKORI can cause fetal harm when administered to a pregnant woman [see Clinical Pharmacology (12.1)]. There are no available data on the use of XALKORI during pregnancy. In animal reproduction studies, oral administration of crizotinib in pregnant rats during organogenesis at exposures similar to those expected with the maximum recommended human dose resulted in embryotoxicity and fetotoxicity [see Data]. Advise pregnant women of the potential risk to a fetus. The estimated background risk of major birth defects and miscarriage for the indicated population is unknown. In the U.S. general population, the estimated background risk of major birth defects and miscarriage in clinically recognized pregnancies is 2 to 4% and 15 to 20%, respectively.
Animal Data Crizotinib was administered to pregnant rats and rabbits during organogenesis to study the effects on embryo-fetal development. Postimplantation loss was increased at doses ≥50 mg/kg/day (approximately 0.6 times the recommended human dose based on AUC) in rats. No teratogenic effects were observed in rats at doses up to the maternally toxic dose of 200 mg/kg/day (approximately 2.7 times the recommended human dose based on AUC) or in rabbits at doses of up to 60 mg/kg/day (approximately 1.6 times the recommended human dose based on AUC), though fetal body weights were reduced at these doses.
8.2 Lactation Risk Summary
There is no information regarding the presence of crizotinib in human milk, the effects on the breastfed infant, or the effects on milk production. Because of the potential for adverse reactions in breastfed infants, do not breastfeed during treatment with XALKORI and for 45 days after the final dose.
8.3 Females and Males of Reproductive Potential Contraception Females XALKORI can cause fetal harm when administered to a pregnant woman [see Use in Specific Populations (8.1)]. Advise females of reproductive potential to use effective contraception during treatment with XALKORI and for at least 45 days after the final dose. Males Because of the potential for genotoxicity, advise males with female partners of reproductive potential to use condoms during treatment with XALKORI and for at least 90 days after the final dose [see Nonclinical Toxicology (13.1)]. Infertility Based on reproductive organ findings in animals, XALKORI may cause reduced fertility in females and males of reproductive potential. It is not known whether these effects on fertility are reversible [see Nonclinical Toxicology (13.1)]. 8.4 Pediatric Use The safety and efficacy of XALKORI in pediatric patients has not been established. Animal Data Decreased bone formation in growing long bones was observed in immature rats at 150 mg/kg/day following once daily dosing for 28 days (approximately 5.4 times the recommended human dose based on AUC). Other toxicities of potential concern to pediatric patients have not been evaluated in juvenile animals.
8.5 Geriatric Use
Of the total number of patients with ALK-positive metastatic NSCLC in clinical studies of XALKORI (n=1669), 16% were 65 years or older and 3.8% were 75 years or older. No overall differences in safety or effectiveness were observed between these patients and younger patients. Clinical studies of XALKORI in patients with ROS1-positive metastatic NSCLC did not include sufficient numbers of patients age 65 years and older to determine whether they respond differently from younger patients.
8.6 Hepatic Impairment
XALKORI has not been studied in patients with hepatic impairment. As crizotinib is extensively metabolized in the liver, hepatic impairment is likely to increase plasma crizotinib concentrations. Clinical studies excluded patients with AST or ALT greater than 2.5 times ULN, or greater than 5 times ULN, if due to liver metastases. Patients with total bilirubin greater than 1.5 times ULN were also excluded. Therefore, use caution in patients with hepatic impairment [see Clinical Pharmacology (12.3)].
8.7 Renal Impairment
No starting dose adjustment is needed for patients with mild (CLcr 60-89 mL/min) or moderate (CLcr 30-59 mL/min) renal impairment based on a population pharmacokinetic analysis. Increased exposure to crizotinib occurred in patients with severe renal impairment
10 OVERDOSAGE
There have been no known cases of XALKORI overdose. There is no antidote for XALKORI.
11 DESCRIPTION
XALKORI (crizotinib) is an oral receptor tyrosine kinase inhibitor. The molecular formula for crizotinib is C21H22Cl2FN5O. The molecular weight is 450.34 daltons. Crizotinib is described chemically as (R)-3-[1-(2,6-Dichloro-3-fluorophenyl)ethoxy]-5-[1-(piperidin-4-yl)-1H-pyrazol-4-yl]pyridin-2-amine.
Crizotinib is a white to pale-yellow powder with a pKa of 9.4 (piperidinium cation) and 5.6 (pyridinium cation). The solubility of crizotinib in aqueous media decreases over the range pH 1.6 to pH 8.2 from greater than 10 mg/mL to less than 0.1 mg/mL. The log of the distribution coefficient (octanol/water) at pH 7.4 is 1.65. XALKORI capsules are supplied as printed hard-shell capsules containing 250 mg or 200 mg of crizotinib together with colloidal silicon dioxide, microcrystalline cellulose, anhydrous dibasic calcium phosphate, sodium starch glycolate, magnesium stearate, and hard gelatin capsule shells as inactive ingredients. The pink opaque capsule shell components contain gelatin, titanium dioxide, and red iron oxide. The white opaque capsule shell components contain gelatin and titanium dioxide. The printing ink contains shellac, propylene glycol, strong ammonia solution, potassium hydroxide, and black iron oxide.
12 CLINICAL PHARMACOLOGY
12.1 Mechanism of Action
Crizotinib is an inhibitor of receptor tyrosine kinases including ALK, Hepatocyte Growth Factor Receptor (HGFR, c-Met), ROS1 (c-ros), and Recepteur d’Origine Nantais (RON). Translocations can affect the ALK gene resulting in the expression of oncogenic fusion proteins. The formation of ALK fusion proteins results in activation and dysregulation of the gene’s expression and signaling which can contribute to increased cell proliferation and survival in tumors expressing these proteins. Crizotinib demonstrated concentration-dependent inhibition of ALK, ROS1, and c-Met phosphorylation in cell-based assays using tumor cell lines and demonstrated antitumor activity in mice bearing tumor xenografts that expressed echinoderm microtubule-associated protein-like 4 (EML4)- or nucleophosmin (NPM)-ALK fusion proteins or c-Met.
12.2 Pharmacodynamics
Cardiac electrophysiology In an ECG substudy conducted in 52 patients with ALK-positive NSCLC who received crizotinib 250 mg twice daily, the maximum mean QTcF (corrected QT by the Fridericia method) change from baseline was 12.3 ms (2-sided 90% upper CI: 19.5 ms). An exposure-QT analysis suggested a crizotinib plasma concentrationdependent increase in QTcF [see Warnings and Precautions (5.3)]
12.3 Pharmacokinetics Absorption
Following a single oral dose, crizotinib was absorbed with median time to achieve peak concentration of 4 to 6 hours. Following crizotinib 250 mg twice daily, steady state was reached within 15 days and remained stable, with a median accumulation ratio of 4.8. Steady-state systemic exposure [observed minimum concentration (Cmin) and AUC] appeared to increase in a greater than dose-proportional manner over the dose range of 200-300 mg twice daily. The mean absolute bioavailability of crizotinib was 43% (range: 32% to 66%) following a single 250 mg oral dose. A high-fat meal reduced crizotinib AUC from time zero to infinity (AUCinf) and maximum observed plasma concentration (Cmax) by approximately 14%. XALKORI can be administered with or without food [see Dosage and Administration (2.2)
Distribution
The geometric mean volume of distribution (Vss) of crizotinib was 1772 L following intravenous administration of a 50 mg dose, indicating extensive distribution into tissues from the plasma. Binding of crizotinib to human plasma proteins in vitro is 91% and is independent of drug concentration. In vitro studies suggested that crizotinib is a substrate for P-glycoprotein (P-gp). The blood-to-plasma concentration ratio is approximately 1.
Elimination
Following single doses of crizotinib, the mean apparent plasma terminal half-life of crizotinib was 42 hours in patients. Following the administration of a single 250 mg radiolabeled crizotinib dose to healthy subjects, 63% and 22% of the administered dose was recovered in feces and urine, respectively. Unchanged crizotinib represented approximately 53% and 2.3% of the administered dose in feces and urine, respectively. The mean apparent clearance (CL/F) of crizotinib was lower at steady state (60 L/h) after 250 mg twice daily than after a single 250 mg oral dose (100 L/h), which was likely due to autoinhibition of CYP3A by crizotinib after multiple dosing.
Metabolism
Crizotinib is predominantly metabolized by CYP3A4/5. The primary metabolic pathways in humans were oxidation of the piperidine ring to crizotinib lactam and O-dealkylation, with subsequent Phase 2 conjugation of O-dealkylated metabolites. Specific populations Hepatic impairment: As crizotinib is extensively metabolized in the liver, hepatic impairment is likely to increase plasma crizotinib concentrations. However, XALKORI has not been studied in patients with hepatic impairment. Clinical studies excluded patients with ALT or AST greater than 2.5 times ULN or greater than 16 Reference ID: 4127887 5 times ULN if due to liver metastases. Patients with total bilirubin greater than 1.5 times ULN were also excluded [see Use in Specific Populations (8.6)]. The population pharmacokinetic analysis using the data from approximately 1200 patients with cancer who received XALKORI suggested that baseline total bilirubin (0.1 to 2.1 mg/dL) or AST levels (7 to 124 U/L) did not have a clinically relevant effect on the exposure of crizotinib.
Renal impairment:
The pharmacokinetics of crizotinib were evaluated using the population pharmacokinetic analysis in patients with mild (CLcr 60-89 mL/min, n=433) and moderate (CLcr 30-59 mL/min, n=137) renal impairment. Mild or moderate renal impairment has no clinically relevant effect on the exposure of crizotinib. A study was conducted in 7 patients with severe renal impairment (CLcr
Ethnicity:
No clinically relevant difference in the exposure of crizotinib between Asian patients (n=523) and non-Asian patients (n=691).
Age: Age has no effect on the exposure of crizotinib based on the population pharmacokinetic analysis. Body weight and gender: No clinically relevant effect of body weight or gender on the exposure of crizotinib based on the population pharmacokinetic analysis.
Drug interactions
Effect of Other Drugs on Crizotinib
Strong CYP3A inhibitors: Coadministration of a single 150 mg oral dose of crizotinib with ketoconazole (200 mg twice daily), a strong CYP3A inhibitor, increased crizotinib AUCinf and Cmax values by approximately 3.2-fold and 1.4-fold, respectively, compared to crizotinib alone. However, the magnitude of effect of CYP3A inhibitors on steady-state crizotinib exposure has not been evaluated [see Drug Interactions (7.1)].
Strong CYP3A inducers: Coadministration of crizotinib (250 mg twice daily) with rifampin (600 mg once daily), a strong CYP3A inducer, decreased crizotinib steady-state AUCtau and Cmax by 84% and 79%, respectively, compared to crizotinib alone [see Drug Interactions (7.2)].
Gastric pH elevating medications: In healthy subjects, coadministration of a single 250 mg oral dose of crizotinib following administration of esomeprazole 40 mg daily for 5 days did not result in a clinically relevant change in crizotinib exposure (AUCinf decreased by 10% and no change in Cmax).
Effect of Crizotinib on Other Drugs CYP3A substrates: Coadministration of crizotinib (250 mg twice daily for 28 days) in patients increased the AUCinf of oral midazolam 3.7-fold compared to midazolam alone, suggesting that crizotinib is a moderate inhibitor of CYP3A [see Drug Interactions (7.3)].
Other CYP substrates: In vitro studies suggest that clinical drug-drug interactions as a result of crizotinib-mediated inhibition of the metabolism of substrates for CYP1A2, CYP2C8, CYP2C9, CYP2C19, or CYP2D6 are unlikely to occur.
Crizotinib is an inhibitor of CYP2B6 in vitro. Therefore, crizotinib may increase plasma concentrations of coadministered drugs that are predominantly metabolized by CYP2B6. An in vitro study suggests that clinical drug-drug interactions as a result of crizotinib-mediated induction of the metabolism of substrates for CYP1A2, CYP2B6, CYP2C8, CYP2C9, CYP2C19, or CYP3A are unlikely to occur.
UGT substrates: In vitro studies suggest that clinical drug-drug interactions as a result of crizotinib-mediated inhibition of the metabolism of drugs that are substrates for uridine diphosphate glucuronosyltransferase (UGT)1A1, UGT1A4, UGT1A6, UGT1A9 or UGT2B7 are unlikely to occur.
Substrates of transporters: Crizotinib inhibited P-gp in vitro at clinically relevant concentrations. Therefore, crizotinib has the potential to increase plasma concentrations of coadministered drugs that are substrates of P-gp.
Crizotinib inhibited the hepatic uptake transporter, organic cation transporter (OCT) 1, and renal uptake transporter, OCT2, in vitro at clinically relevant concentrations. Therefore, crizotinib has the potential to increase plasma concentrations of coadministered drugs that are substrates of OCT1 or OCT2. Crizotinib did not inhibit the human hepatic uptake transport proteins, organic anion transporting polypeptides (OATP) B1 or OATP1B3, or the renal uptake transport proteins organic anion transporter (OAT) 1 or OAT3 in vitro at clinically relevant concentrations.
Other transporters: Crizotinib did not inhibit the hepatic efflux bile salt export pump transporter (BSEP) in vitro at clinically relevant concentrations.
13 NONCLINICAL TOXICOLOGY
13.1 Carcinogenesis, Mutagenesis, Impairment of Fertility
Carcinogenicity studies with crizotinib have not been conducted.
Crizotinib was genotoxic in an in vitro micronucleus assay in Chinese Hamster Ovary cultures, in an in vitro human lymphocyte chromosome aberration assay, and in in vivo rat bone marrow micronucleus assays. Crizotinib was not mutagenic in vitro in the bacterial reverse mutation (Ames) assay.
No specific studies with crizotinib have been conducted in animals to evaluate the effect on fertility; however, crizotinib is considered to have the potential to impair reproductive function and fertility in humans based on findings in repeat-dose toxicity studies in the rat. Findings observed in the male reproductive tract included testicular pachytene spermatocyte degeneration in rats given greater than or equal to 50 mg/kg/day for 28 days (greater than 1.7 times the recommended human dose based on AUC). Findings observed in the female reproductive tract included single-cell necrosis of ovarian follicles of a rat given 500 mg/kg/day (approximately 10 times the recommended human dose based on body surface area) for 3 days.
14 CLINICAL STUDIES
14.1 ALK-Positive Metastatic NSCLC Previously Untreated ALK-Positive Metastatic NSCLC - Study 1
The efficacy and safety of XALKORI for the treatment of patients with ALK-positive metastatic NSCLC, who had not received previous systemic treatment for advanced disease, was demonstrated in a randomized, multicenter, open-label, active-controlled study (Study 1). Patients were required to have ALK-positive NSCLC as identified by the FDA-approved assay, Vysis ALK Break-Apart fluorescence in situ hybridization (FISH) Probe Kit, prior to randomization. The major efficacy outcome measure was progression-free survival (PFS) according to Response Evaluation Criteria in Solid Tumors (RECIST) version 1.1 as assessed by independent radiology review (IRR) committee. Additional efficacy outcome measures included objective response rate (ORR) as assessed by IRR, duration of response (DOR), and overall survival (OS). Patient-reported lung cancer symptoms were assessed at baseline and periodically during treatment.
Patients were randomized to receive XALKORI (n=172) or chemotherapy (n=171). Randomization was stratified by Eastern Cooperative Oncology Group (ECOG) performance status (0-1, 2), race (Asian, non-Asian), and brain metastases (present, absent). Patients in the XALKORI arm received XALKORI 250 mg orally twice daily until documented disease progression, intolerance to therapy, or the investigator determined that the patient was no longer experiencing clinical benefit. Chemotherapy consisted of pemetrexed 500 mg/m2 with cisplatin 75 mg/m2 or carboplatin AUC of 5 or 6 mg·min/mL by intravenous infusion every 3 weeks for up to 6 cycles. Patients in the chemotherapy arm were not permitted to receive maintenance chemotherapy. At the time of documented disease progression, as per independent radiology review, patients randomized to chemotherapy were offered XALKORI.
The demographic characteristics of the overall study population were 62% female, median age of 53 years, baseline ECOG performance status 0 or 1 (95%), 51% White and 46% Asian, 4% current smokers, 32% past smokers, and 64% never smokers. The disease characteristics of the overall study population were metastatic disease in 98% of patients, 92% of patients’ tumors were classified as adenocarcinoma histology, 27% of patients had brain metastases, and 7% received systemic chemotherapy as adjuvant or neoadjuvant therapy. Of those randomized to chemotherapy, 70% received XALKORI after IRR documented progression.
Study 1 demonstrated a statistically significant improvement in PFS in the patients treated with XALKORI. The OS analysis conducted at the time of the PFS analysis did not suggest a difference in survival between arms. Table 7 and Figure 1 summarize the efficacy results. Exploratory patient-reported symptom measures of baseline and post-treatment dyspnea, cough, and chest pain suggested a delay in time to development of or worsening of dyspnea, but not cough or chest pain, in patients treated with XALKORI as compared to chemotherapy. The patient-reported delay in onset or worsening of dyspnea may be an overestimation, because patients were not blinded to treatment assignment.
Table 7. Previously Untreated ALK-Positive Metastatic NSCLC - Efficacy Results XALKORI (N=172) Chemotherapy (N=171) Progression-Free Survival (Based on IRR) Number of Events (%) 100 (58%) 137 (80%) Progressive Disease 89 (52%) 132 (77%) Death 11 (6%) 5 (3%) Median, Months (95% CI) 10.9 (8.3, 13.9) 7.0 (6.8, 8.2) HR (95% CI)a 0.45 (0.35, 0.60) p-valueb
Figure 1. Kaplan-Meier Curves of Progression-Free Survival as Assessed by IRR in Study 1
Previously Treated ALK-Positive Metastatic NSCLC - Study 2
The efficacy and safety of XALKORI as monotherapy for the treatment of 347 patients with ALK-positive metastatic NSCLC, previously treated with 1 platinum-based chemotherapy regimen, were demonstrated in a randomized, multicenter, open-label, active-controlled study (Study 2). The major efficacy outcome was PFS according to RECIST version 1.1 as assessed by IRR. Additional efficacy outcomes included ORR as assessed by IRR, DOR, and OS.
Patients were randomized to receive XALKORI 250 mg orally twice daily (n=173) or chemotherapy (n=174). Chemotherapy consisted of pemetrexed 500 mg/m2 (if pemetrexed-naïve; n=99) or docetaxel 75 mg/m2 (n=72) intravenously (IV) every 21 days. Patients in both treatment arms continued treatment until documented disease progression, intolerance to therapy, or the investigator determined that the patient was no longer experiencing clinical benefit. Randomization was stratified by ECOG performance status (0-1, 2), brain metastases (present, absent), and prior EGFR tyrosine kinase inhibitor treatment (yes, no). Patients were required to have ALK-positive NSCLC as identified by the FDA-approved assay, Vysis ALK Break-Apart FISH Probe Kit, prior to randomization. At the time of the final analysis of overall survival, 154 (89%) patients randomized to the chemotherapy arm subsequently received XALKORI.
The demographic characteristics of the overall study population were 56% female, median age of 50 years, baseline ECOG performance status 0 or 1 (90%), 52% White and 45% Asian, 4% current smokers, 33% past smokers, and 63% never smokers. The disease characteristics of the overall study population were metastatic disease in at least 95% of patients and at least 93% of patients’ tumors were classified as adenocarcinoma histology.
Study 2 demonstrated a statistically significant improvement in PFS in the patients treated with XALKORI. Table 8 and Figure 2 summarize the efficacy results.
Table 8. Previously Treated ALK-Positive Metastatic NSCLC - Efficacy Results XALKORI (N=173) Chemotherapy (N=174) Progression-Free Survival (Based on IRR) Number of Events (%) 100 (58%) 127 (73%) Progressive Disease 84 (49%) 119 (68%) Death 16 (9%) 8 (5%) Median, Months (95% CI) 7.7 (6.0, 8.8) 3.0a (2.6, 4.3) HR (95% CI)b 0.49 (0.37, 0.64) p-valuec
Figure 2. Kaplan-Meier Curves of Progression-Free Survival as Assessed by IRR in Study 2
14.2 ROS1-Positive Metastatic NSCLC The efficacy and safety of XALKORI was investigated in a multicenter, single-arm study (Study 3), in which patients with ROS1-positive metastatic NSCLC received XALKORI 250 mg orally twice daily. Patients were required to have histologically-confirmed advanced NSCLC with a ROS1 rearrangement, age 18 years or older, ECOG performance status of 0, 1, or 2, adequate organ function, and measurable disease. The efficacy outcome measures were ORR and DOR according to RECIST version 1.0 as assessed by IRR and investigator, with imaging performed every 8 weeks for the first 60 weeks. Baseline demographic and disease characteristics were female (56%), median age of 53 years, baseline ECOG performance status of 0 or 1 (98%), White (54%), Asian (42%), past smokers (22%), never smokers (78%), metastatic disease (92%), adenocarcinoma (96%), no prior systemic therapy for metastatic disease (14%), and prior platinum-based chemotherapy for metastatic disease (80%). The ROS1 status of NSCLC tissue samples was determined by laboratory-developed break-apart FISH (96%) or RT-PCR (4%) clinical trial assays. For assessment by FISH, ROS1 positivity required that ≥15% of a minimum of 50 evaluated nuclei contained a ROS1 gene rearrangement. Efficacy results are summarized in Table 9. 23 Reference ID: 4127887 Table 9. ROS1-Positive Metastatic NSCLC - Efficacy Results* Efficacy Parameters IRR (N=50) Investigator-Assessed (N=50) Objective Response Rate (95% CI) 66% (51, 79) 72% (58, 84) Complete Response, n 1 5 Partial Response, n 32 31 Duration of Response Median, Months (95% CI) 18.3 (12.7, NR) NR (14.5, NR) IRR=independent radiology review; CI=confidence interval; NR=not reached. * As assessed by RECIST version 1.0.
16 HOW SUPPLIED/STORAGE AND HANDLING
· 250 mg capsules Hard gelatin capsule with pink opaque cap and body, printed with black ink “Pfizer” on the cap, “CRZ 250” on the body; available in: Bottles of 60 capsules: NDC 0069-8140-20
· 200 mg capsules Hard gelatin capsule with pink opaque cap and white opaque body, printed with black ink “Pfizer” on the cap, “CRZ 200” on the body; available in: Bottles of 60 capsules: NDC 0069-8141-20
Store at room temperature 20° to 25°C (68° to 77°F); excursions permitted between 15° to 30°C (59° to 86°F) [see USP Controlled Room Temperature].
17 PATIENT COUNSELING INFORMATION
Advise the patient to read the FDA-approved patient labeling (Patient Information).
Hepatotoxicity Inform patients to immediately report symptoms of hepatotoxicity [see Warnings and Precautions (5.1)].
Interstitial Lung Disease (Pneumonitis)
Advise patients to immediately report any new or worsening pulmonary symptoms [see Warnings and Precautions (5.2)].
Bradycardia Advise patients to report any symptoms of bradycardia and to inform their healthcare provider about the use of any heart or blood pressure medications [see Warnings and Precautions (5.4)].
Severe Visual Loss Inform patients of the potential risk of severe visual loss and to immediately contact their healthcare provider if they develop severe visual loss. Inform patients that visual changes such as perceived flashes of light, blurry vision, light sensitivity, and floaters are commonly reported adverse events and may occur while driving or operating machinery. The onset of visual disorders most commonly occurs during the first week of treatment [see Warnings and Precautions (5.5) and Adverse Reactions (6)]. 24
Drug Interactions Inform patients to avoid grapefruit or grapefruit juice while taking XALKORI.
Advise patients to inform their healthcare providers of all concomitant medications, including prescription medicines, over-the-counter drugs, vitamins, and herbal products [see Drug Interactions (7)].
Dosing and Administration Advise patients to take XALKORI with or without food and swallow XALKORI capsules whole. If a patient misses a dose, advise the patient to take it as soon as remembered unless it is less than 6 hours until the next dose, in which case, advise the patient not to take the missed dose.
If a patient vomits after taking a dose of XALKORI, advise the patient not to take an extra dose, but to take the next dose at the regular time.
Embryo-Fetal Toxicity Advise females of reproductive potential of the potential risk to a fetus and to inform their healthcare provider of a known or suspected pregnancy [see Warnings and Precautions (5.6) and Use in Specific Populations (8.1)]. Advise females of reproductive potential to use effective contraception during treatment with XALKORI and for at least 45 days after the final dose [see Use in Specific Populations (8.3)].
Females and Males of Reproductive Potential Advise females and males of reproductive potential of the potential for reduced fertility from XALKORI [see Use in Specific Populations (8.3) and Nonclinical Toxicology (13.1)].
Advise male patients with female partners of reproductive potential to use condoms during treatment with XALKORI and for at least 90 days after the final dose [see Use in Specific Populations (8.3) and Nonclinical Toxicology (13.1)].
Lactation Advise females not to breastfeed during treatment with XALKORI and for 45 days after the final dose [see Use in Specific Populations (8.2)].
Infertility Advise females and males of reproductive potential of the potential for reduced fertility from XALKORI [see Use in Specific Populations (8.3)].
This product’s labeling may have been updated. For full prescribing information, please visit www.XALKORI.com.