通用中文 | 氘代丁苯那嗪片 | 通用外文 | Deutetrabenazine |
品牌中文 | 品牌外文 | Austedo | |
其他名称 | |||
公司 | 梯瓦(TEVA) | 产地 | 美国(USA) |
含量 | 9mg | 包装 | 60片/盒 |
剂型给药 | 储存 | 室温 | |
适用范围 | 亨丁顿舞蹈病 |
通用中文 | 氘代丁苯那嗪片 |
通用外文 | Deutetrabenazine |
品牌中文 | |
品牌外文 | Austedo |
其他名称 | |
公司 | 梯瓦(TEVA) |
产地 | 美国(USA) |
含量 | 9mg |
包装 | 60片/盒 |
剂型给药 | |
储存 | 室温 |
适用范围 | 亨丁顿舞蹈病 |
Austedo(deutetrabenazine)片说明书
2017年4月第一版
批准日前:2017年4月3日;
公司:Teva Pharmaceutical Industries Ltd.
为治疗:亨丁顿舞蹈病
这些重点不包括安全和有效使用AUSTEDO需所有资料。请参阅AUSTEDO完整处方资料。
AUSTEDO™(deutetrabenazine)片,为口服使用
美国初次批准:2017
适应证和用途
AUSTEDO是一种血管单按转运蛋白2(VMAT2)抑制剂适用为伴随亨丁顿病[Huntington’s disease]舞蹈症的治疗(1)
剂量和给药方法
● 开始剂量是6 mg每天1次。在每周间隔向上滴定调整by 6 mg每天至一个减低舞蹈症的耐受剂量,至一个最大每天推荐剂量48 mg(24 mg每天2次)(2.1)
● 在两个分次剂量中给予每天总剂量12 mg或以上 (2.1)
● 与食物给予(2.1)
● 吞咽整片;不要咀嚼,粉碎或破碎片(2.1)
● 如患者从四苯嗪[tetrabenazine]转换,终止四苯嗪和在后面天开始AUSTEDO。对推荐的转换片见完整处方资料(2.2)
● AUSTEDO在CYP2D6差代谢者中最大推荐剂量为36 mg每天(即,18 mg每天2次)(2.4,8.7)
剂型和规格
片:6 mg,9 mg,和12 mg(3)
禁忌证
● 有自杀倾向的,或未治疗的/不佳地治疗抑郁(4,5.2)
● 肝受损(4,8.6,12.3)
● 用MAOIs,利血平[reserpine],或四苯嗪(XENAZINE®)(4,7.2,7.3,7.7)
警告和注意事项
● 抗精神病药物恶性症候群[Neuroleptic Malignant Syndrome](NMS):如这发生终止(5.3,7.4)
● 静坐不能,激动,不安,和帕金森症:如这发生减低剂量或终止。(5.4,5.5)
● 镇静/嗜睡:可能损害患者驾驶或操作复杂机械的能力(5.6)
不良反应
最常见不良反应(>8%的AUSTEDO-治疗患者和大于安慰剂)为:嗜睡,腹泻,口干,和疲乏(6.1)
药物相互作用
●强CYP2D6抑制剂的同时使用:AUSTEDO的最大推荐剂量是36 mg每天(18 mg每天2次)(2.3,7.1)
● 酒精或其他镇静药物:可能有相加镇静和嗜睡(7.5)
在特殊人群中使用
妊娠:根据动物数据,可能致胎儿危害(8.1)
完整处方资料
1 适应证和用途
AUSTEDO™是适用为伴随亨丁顿病舞蹈症的治疗。
2 剂量和给药方法
2.1 给药资料
AUSTEDO的剂量被个体地根据舞蹈症的减低和耐受性确定。当首次处方给予不是正在从四苯嗪(一个相关的VMAT2抑制剂)转换患者,AUSTEDO的推荐开始剂量是6 mg口服给予每天1次。
● AUSTEDO的剂量可能以每周间隔在每天增量6 mg至一个最大每天推荐剂量48 mg。
● 在两个分次剂量中给予每天总剂量12 mg或以上.
● 与食物给予AUSTEDO [见临床药理学(12.3)].
● 整吞AUSTEDO片。不要咀嚼,粉碎或破碎片。
2.2 患者从四苯嗪(XENAZINE®)转换至AUSTEDO
在以下天终止四苯嗪(XENAZINE®)和开始AUSTEDO。在表1显示患者从四苯嗪(XENAZINE®)转换转换至AUSTEDO推荐的初始给药方案。
患者被转换至AUSTEDO后,剂量可能在每周间隔被调整[见剂量和给药方法(2.1)]。
2.3 与强CYP2D6抑制剂剂量调整
在接受强CYP2D6抑制剂患者中(如,奎尼丁[quinidine],抗抑郁剂例如帕罗西汀[paroxetine],氟西汀[fluoxetine],和安非他酮[bupropion]),AUSTEDO的总每天剂量不应超过36 mg(最大单次剂量18 mg)[见药物相互作用(7.1)和临床药理学(12.3)]。
2.4 在CYP2D6差代谢者中剂量调整
在患者是CYP2D6差代谢者,AUSTEDO的每天总剂量不应超过36 mg(最大单次剂量18 mg)[见在特殊人群中使用(8.7)]。
2.5 治疗的终止和中断
用AUSTEDO治疗可能无滴定调整被终止。治疗的中断后大于一周,当恢复时AUSTEDO治疗应被再次滴定调整。对治疗中断少于一周,治疗可能没有滴定调整在以前维持剂量被恢复。
3 剂型和规格
AUSTEDO片可得到以下规格:
● 6 mg片是圆形,紫色-涂层片,在一侧黑墨水打印有“SD”上面“6”.
● 9 mg片是圆形,蓝色-涂层片,在一侧黑墨水打印有 “SD”上面“9”.
● 12 mg片是圆形,米色-涂层片,在一侧黑墨水打印有 “SD”上面“12”.
4 禁忌证
在以下患者禁忌AUSTEDO:
● 患者是有自杀倾向的,或n有未治疗或不适当地治疗的抑郁患者[见警告和注意事项(5.2)].
● 有肝受损[见在特殊人群中使用(8.6),临床药理学(12.3)].
● 用单胺氧化酶抑制剂(MAOIs)。AUSTEDO不应与一个MAOI联用,或用一个MAOI治疗终止的14天内[见药物相互作用(7.3)]。
● 开始AUSTEDO前用,利血平应停止后过后至少20天[见药物相互作用(7.2)]。
● 用四苯嗪(XENAZINE®)[见药物相互作用(7.7)].
5 警告和注意事项
5.1 临床恶化和不良事件
亨丁顿病是一种进行性疾患特征是情绪,认知,舞蹈症,强直,和功能性能力随时间。VMAT2 抑制剂,包括AUSTEDO,可能致一个情绪,认知,强直,和功能性能力恶化。
处方者应定期地再评价对AUSTEDO需要在他们的患者通过评价对舞蹈症效应和可能不良效应,包括镇静/嗜睡,抑郁和自杀倾向,帕金森症,静坐不能,不安,和认知下降。可能难以区分不良反应和潜在疾病的进展间;减低剂量或停止药物可能有助于临床医生区分两种可能性。在有些患者,潜在舞蹈症本身可能随时间改善,减低对AUSTEDO需求。
5.2 抑郁和自杀倾向
有亨丁顿病患者是处于对抑郁,和有自杀倾向的观念或行为(自杀倾向)的风险增加。在有亨丁顿病患者中AUSTEDO可能增加对自杀倾向风险。.
在一项12-周,双盲,安慰剂-对照试验,报道2%的用AUSTEDO治疗患者有自杀倾向的观念,相比较无用安慰剂患者;被报道无自杀企图和无已完成的自杀。用AUSTEDO治疗患者报告抑郁为4%。
当考虑AUSTEDO的使用,应平衡自杀倾向的风险与对舞蹈症的治疗的需求。所有用AUSTEDO治疗患者应被观察对新或恶化的抑郁或自杀倾向。如抑郁或自杀倾向没有解决,考虑终止用AUSTEDO治疗。
患者,他们的护理人员,和家庭应被告知伴随AUSTEDO抑郁,恶化 抑郁,和自杀倾向的风险,和应被指导报告关注的行为及时地至治疗医生。有亨丁顿病患者表现有自杀倾向的观念 应立即被评价。
5.3 抗精神病药物恶性症候群(NMS)
曽被报道一种潜在地致命症状复合有时被称为抗精神病药物恶性症候群(NMS)在伴随药物减低多巴胺传导[见药物相互作用(7.4)]。而在接受AUSTEDO患者中未曽观察到NMS,在接受四苯嗪(一种密切相关的VMAT2抑制剂)患者曽被观察到。
临床医生应警惕伴随NMS体征和症状变化。NMS的临床表现是高热,肌肉强直,改变的精神状态,和自主神经不稳定的证据(不规则脉搏或血压,心动过速,出汗,和心律失常),附加征象可能包括升高的肌酐磷酸化酶,肌红蛋白尿,横纹肌溶解症,和急性肾衰。NMS的诊断可能被复杂化;其他严重的医学疾患(如,肺炎,全身感染)和可能存在未治疗或治疗不足的锥体外系疾病与相似体征和症状。在鉴别诊断中其他重要考虑包括中枢抗胆碱能毒性,热中暑,药物热,和主要中枢神经系统病理学。
NMS的处理应包括(1) 立即终止AUSTEDO;(2) 强化对症治疗和医学监视;和(3) 可得到的任何同时严重医学问题的治疗特殊治疗。对NMS有关特殊药理学治疗没有一般一致的方案。
曽报告NMS的复发与药物治疗的恢复。如从NMS恢复后需要用AUSTEDO治疗,患者应被监视对复发征象。
5.4 静坐不能,激动,和不安
在有亨丁顿病患者中AUSTEDO可能增加静坐不能,激动,和不安的风险。在一项12-周,双盲,安慰剂-对照试验, 4%用AUSTEDO治疗患者报道静坐不能,激动,或不安与之比较用安慰剂患者为2%。
接受AUSTEDO患者应被监视不安和激动的体征和症状,因为这些可能是发生静坐不能的指示指标。如一例患者用AUSTEDO治疗期间发生静坐不能,AUSTEDO剂量应被减低;有些患者可能需要终止治疗。
5.5 帕金森症
在有亨丁顿病患者中AUSTEDO可能致帕金森症。
因为在亨丁顿病强直可能发展作为潜在疾病过程的一部分,可能这个潜在的药物-诱发不良反应和潜在疾病过程的进展间难以区分。对有些有亨丁顿病患者药物-诱发帕金森症有潜能致比未治疗的舞蹈症的更功能性残疾。如一例患者用AUSTEDO治疗期间发生帕金森症, AUSTEDO剂量应被减低;有些患者可能需要终止治疗。
5.6 镇静和嗜睡
镇静是AUSTEDO常见剂量-限制不良反应。在一项12-周,双盲,安慰剂-对照试验,11%的AUSTEDO-治疗患者报告嗜睡与之比较用安慰剂患者为4%和9%的AUSTEDO-治疗患者报告疲乏相比较有4%的安慰剂-治疗患者。
患者不应进行需要精神警惕性活动以保持他们自身或其他人安全性,例如操作汽车或操作危害性机械,直至他们是用维持剂量的AUSTEDO和知道药物如何影响他们。
5.7 QTc延长
四苯嗪,一种密切相关的VMAT2 抑制剂,在校正的QT(QTc)间隔中致一个增加(约8 msec)[见临床药理学(12.2)]。
在有些用AUSTEDO治疗患者是CYP2D6差代谢者或被一个强CYP2D6抑制剂共同给药,可能发生一个临床上相关的QT延长[见临床药理学(12.2,12.3)]。
AUSTEDO的使用应避免与其他已知延长QTc药物联用[见药物相互作用(7.6)]。
在有先天性长QT综合证患者和在有一个心脏心律失常病史患者也应避免使用AUSTEDO。某些情况可能增加尖端扭转型室性心动过速的发生的风险和/或突然死亡在伴随延长QTc间期药物的使用,包括(1) 心动过缓;(2) 低钾血症或低镁血症;(3) 延长QTc间期其他药物的同时使用;和(4) 先天性QT间期的延长的存在。
5.8 高催乳素血症
在AUSTEDO开发计划没有评价血清催乳素水平。
四苯嗪,一种密切相关的VMAT2 抑制剂,在人中升高血清催乳素浓度。给予25 mg 的四苯嗪至健康志愿者后,血浆催乳素峰水平增加4-至5-倍。.
组织培养实验表明约三分之一人乳癌是体外催乳素-依赖,一种潜在重要性因子如AUSTEDO 正在被考虑为一个以前检测到乳癌患者。虽然无月经,乳溢症,男性乳房增大症,和阳痿可能被升高的血清催乳素浓度引起,对大多数患者不知道升高的血清催乳素浓度的临床意义。
血清催乳素水平中慢性增加(虽然在AUSTEDO或四苯嗪开放计划中没有评价)曽被伴随低水平雌激素和增加骨质疏松症风险。应做是否有一个症状性高催乳素血症的临床怀疑,适当实验室测试和应被考虑给予终止AUSTEDO。
5.9 结合至含黑色素组织
因为deutetrabenazine或其代谢物结合至含黑色素组织,随时间它可能蓄积在这些组织。广泛使用后这个升高有可能性AUSTEDO可能在这些组织中致毒性。在有色素动物种属例如犬的慢性毒性研究没有眼科学也没有进行眼显微镜检查。
在人中眼科学监视不足以排除长期接触后发生的损伤的可能性.
不知道deutetrabenazine的结合至含黑色素组织的临床相关性。
尽管对定期地眼科学监视没有专门建议,处方者应认识到长期眼科学效应的可能性[见临床药理学(12.2)]。
6 不良反应
在说明书的其他节更详细讨论以下严重的不良反应:
● 抑郁和自杀倾向[见警告和注意事项(5.2)]
● 抗精神病药物恶性症候群(NMS)[见警告和注意事项(5.3)]
● 静坐不能,激动,和不安[见警告和注意事项(5.4)]
● 帕金森症[见警告和注意事项(5.5)]
● 镇静和嗜睡[见警告和注意事项(5.6)]
● QTc延长[见警告和注意事项(5.7)]
● 高催乳素血症[见警告和注意事项(5.8)]
● 结合至含黑色素组织[见警告和注意事项(5.9)]
6.1 临床试验经验
因为临床试验是在广泛不同情况下进行的,临床试验观察到不良反应率不能与另一种药临床试验发生率直接比较而且可能不反映实践中观察到的发生率。
研究1是在有舞蹈症伴随亨丁顿病患者一项随机化,12-周,安慰剂-对照研究。总共45例患者接受AUSTEDO,和45例患者接受安慰剂。患者年龄范围在年龄23和74岁间(均数54岁);56% 为男性,和92%为高加索人。最常见不良反应发生在大于8%的AUSTEDO-治疗患者为嗜睡,腹泻,口干,和疲乏。表2中总结发生在4%或更多用AUSTEDO治疗患者,和比用安慰剂患者更大发生率的不良反应。
在研究1在7%的患者中一种或更多不良反应导致研究药物的剂量减低。在接受AUSTEDO患者中最常不良反应导致剂量减低为眩晕(4%).
在研究1中激动导致在2%的用AUSTEDO治疗患者终止。
7 药物相互作用
7.1 强CYP2D6抑制剂
在维持用一个稳定剂量的AUSTEDO患者当添加一个强CYP2D6抑制剂在AUSTEDO剂量可能被需要一个减低。使用强CYP2D6抑制剂的同时(如,帕罗西汀,氟西汀,奎尼丁,安非他酮)曽被显示增加对deutetrabenazine活性双氢-代谢物的全身暴露约3-倍。AUSTEDO的每天剂量不应超过36 mg每天,而在用强CYP2D6抑制剂患者AUSTEDO的最大单次剂量不应超过18 mg [见剂量和给药方法(2.3)和临床药理学(12.3)]。
7.2 利血平
利血平与VMAT2不可逆地结合和它的效应时间为几天。给予AUSTEDO前处方者应等待对舞蹈症重现有助于减低药物过量的风险和在中枢神经系统中5羟色胺和去甲肾上腺素重大耗竭。
停止利血平后开始AUSTEDO前至少要经过20天。AUSTEDO和利血平不应同时使用[见禁忌证(4)]。
7.3 单胺氧化酶抑制剂(MAOIs)
在用MAOIs患者中禁忌AUSTEDO。AUSTEDO不应与一个MAOI联用,或治疗用一个MAOI终止14天内使用[见禁忌证(4)]。
7.4 神经抑制药药物[Neuroleptic Drugs]
对帕金森症,NMS,和静坐不能的风险可能被增加AUSTEDO和多巴胺[dopamine]拮抗剂或抗精神活动的药物的同时使用增加。
7.5 酒精或其他镇静药物
酒精或其他镇静药物的同时使用可能有相加效应和恶化镇静和嗜睡[见警告和注意事项(5.6)]。
7.6 致QTc延长药物
四苯嗪,一种密切相关的VMAT2抑制剂,致一个小增加在校正的QT(QTc)间期。用AUSTEDO也可能发生临床上相关的QT延长[见警告和注意事项(5.7),临床药理学(12.2)]。
AUSTEDO的使用应被避免与已知延长QTc其他药物联用,包括抗精神病药物(如,氯丙嗪[chlorpromazine],氟哌啶醇[haloperidol],甲硫达嗪[thioridazine],齐拉西酮[ziprasidone]),抗生素(如,莫西沙星[moxifloxacin]),类型1A(如,奎尼丁,普鲁卡因酰胺[procainamide]),和类型III(如,胺碘酮[amiodarone],苏特罗[sotalol])抗心律失常药物或任何其他延长QTc间期药物。
7.7 四苯嗪
在当前用四苯嗪患者禁忌AUSTEDO。四苯嗪终止后天可开始AUSTEDO[见剂量和给药方法(2.2)]。
8 在特殊人群中使用
8.1 妊娠
风险总结
在妊娠妇女AUSTEDO的使用伴随对发育风险没有适当的数据。器官形成期时给予大鼠deutetrabenazine对胚胎胎儿发育没有产生明确的不良效应。但是,对大鼠妊娠和哺乳自始至终给予四苯嗪导致死胎增加和产后子代死亡率的增加[见数据]。
在美国一般人群,重大出生缺陷和在临床上认可妊娠流产的估算背景风险分别为2-4%和15-20%。不知道在适应证人群重大出生缺陷和流产的背景风险。
数据
动物数据
器官形成期时Deutetrabenazine的口服给药(5,10,或30 mg/kg/day)或四苯嗪(30 mg/kg/day)至妊娠大鼠对胚胎胎儿发育无明确的影响。在体表面积(mg/m2)基础最高被测试的剂量为最大推荐人剂量48 mg/day的6倍。
未曽评估器官形成期时对兔或妊娠和哺乳时对大鼠给予deutetrabenazine的影响。
当妊娠兔器官形成期阶段时在口服剂量至60 mg/kg/day给予四苯嗪对胚胎胎儿发育无影响。当四苯嗪被给予至雌性大鼠(剂量5,15,和30 mg/kg/day)从器官形成期开始至哺乳阶段,在15和30 mg/kg/day观察到死胎和产后出生的子代死亡率增加,和在所有剂量观察到幼畜成熟延迟。
8.2 哺乳
风险总结
没有在人乳汁中deutetrabenazine或其代谢物的存在,对哺乳喂养婴儿的影响,或药物对乳汁产生影响的数据。哺乳喂养的发育和健康获益应与对AUSTEDO母亲的临床需求和对哺乳喂养婴儿来自AUSTEDO或来自母体潜在情况任何潜在不良效应一并考虑。
8.4 儿童使用
尚未在儿童患者中确定安全性和有效性。
8.5 老年人使用
AUSTEDO的临床研究没有包括充分数量受试者年龄65和以上以确定是否他们反应不同于较年轻受试者。其他被报道的临床经验没有确定老年和较年轻患者间反应中差别。一般说来,对一位老年患者剂量选择应谨慎,通常开始在给药范围的低端,反映肝,肾和心脏功能失调,和同时疾病或其他药物治疗的更大频数。
8.6 肝受损
尚未研究肝受损对deutetrabenazine及其主要代谢物的药代动力学的影响;但是,在用四苯嗪进行的一项临床研究,一种密切相关的VMAT2抑制剂,在有肝受损患者对四苯嗪及其活性代谢物暴露有巨大增加。尚未评价这个增加暴露的临床意义,但是因为关注对严重的不良反应更大风险,在有肝受损患者禁忌使用AUSTEDO[见禁忌证(4),临床药理学(12.3)]。
8.7 CYP2D6差代谢者
尽管尚未曽在没有表达药物代谢酶患者中系统地评价deutetrabenazine及其代谢物的药代动力学,它是可能对α-HTBZ和β-HTBZ暴露将是增加相似于用一个强CYP2D6抑制剂(约3-倍)。在患者是CYP2D6 poor差代谢者,AUSTEDO的每天剂量不应超过36 mg(最大单次剂量为18 mg)[见剂量和给药方法(2.4)和临床药理学(12.3)]。
10 药物过量
在文献中用四苯嗪,一种密切相关的VMAT2抑制剂,曽被报道药物过量范围从100 mg至1 g。用过量给药发生以下不良反应:急性肌张力障碍,眼动危象,恶心和呕吐,出汗,镇静,低血压,混乱,腹泻,幻觉,发红,和震颤。治疗应那些与任何中枢神经系统-活性药物过量处理中应用一般措施组成。建议一般支持和对症措施。应监视心脏节律和生命征象。在处理药物过量中,应经常被考虑涉及多种药物的可能性。医生应考虑联系一个对任何过量的治疗毒物控制中心。在美国经认证的毒物控制中心美国协会网址www.aapcc.org.列出电话号码。
11 一般描述
AUSTEDO(deutetrabenazine)是一种为口服给药的血管单按转运蛋白2(VMAT2)抑制剂。 Deutetrabenazine的分子量为323.46;pKa为6.31。Deutetrabenazine为一种hexahydro-dimethoxybenzoquinolizine衍生物和有以下化学名:(RR,SS)-1,3,4,6,7,11b-hexahydro-9,10-di(methoxy-d3)-3-(2-methylpropyl)-2H-benzo[a]quinolizin-2-one.
Deutetrabenazine的分子式为C19H21D6NO3。Deutetrabenazine是一种消旋混合物含以下结构。
Deutetrabenazine是白色至浅黄色结晶粉为微溶于水和溶于乙醇。AUSTEDO片含6 mg,9 mg,或12 mg deutetrabenazine,和以下无活性成分:氢氧化铵,黑色氧化铁,n-丁醇,丁基羟基苯甲醚,丁基羟基甲苯,硬脂酸镁,甘露醇,微晶纤维素,聚乙二醇,聚氧乙烯,聚山梨醇80,聚乙烯醇,聚维酮,丙二醇,虫胶,滑石,二氧化钛,和FD&C蓝#2 lake。6 mg片还含FD&C红#40 lake。12 mg片还含FD&C黄#6 lake。
12 临床药理学
12.1 作用机制
不知道AUSTEDO(deutetrabenazine)发挥其抗-舞蹈症作用的精准机制,但被相信是与它的作用为一种来自神经神经终端器[nerve terminals]单胺(例如多巴胺,5羟色胺,去甲肾上腺素,和组胺)可逆性耗竭剂[depletory]相关。Deutetrabenazine的主要的循环代谢物(α-双氢tetrabenazine[HTBZ]和β-HTBZ),是VMAT2的可逆性抑制剂,导致单胺的摄取入突触囊泡减低和单胺贮存的耗竭。.
12.2 药效动力学
心脏电生理学
在一项随机化,双盲,安慰剂-对照交叉研究在健康男性和女性受试者用莫西沙星作为阳性对照研究一个单次12-mg或24-mg剂量的AUSTEDO对QT间期的影响。在24 mg时,AUSTEDO 引起一个约4.5 msec均数增加在QTc(90% CI:2.4,6.5 msec)。未曽评价在较高暴露对AUSTEDO或其代谢物暴露的影响。.
在一项随机化,双盲,安慰剂-对照交叉研究在健康男性和女性受试者用莫西沙星作为阳性对照研究一个单次25-mg或50-mg剂量的四苯嗪,一种密切相关的VMAT2抑制剂,对QT间期的影响。在50 mg时,四苯嗪引起一个约8 msec均数增加在QTc(90% CI:5.0,10.4 msec)。未曽评价在较高暴露对或四苯嗪或其代谢物影响[见警告和注意事项(5.7)和药物相互作用(7.6)]。
黑色素结合
在有色素大鼠中Deutetrabenazine或其代谢物结合至含黑色素组织(即,眼,皮肤,皮毛)。在单次口服剂量放射性标记的deutetrabenazine,在给药后35天时仍在眼和皮毛中检测到放射性[见警告和注意事项(5.9)]。
12.3 药代动力学
口服给药至25 mg后,deutetrabenazine的血浆浓度是一般地低于检测低限是因为广泛的肝代谢deutetrabenazine至活性氘化的二氢[dihydro]代谢物(HTBZ),α-HTBZ和β-HTBZ。观察到对活性代谢物单次或多剂量后(6 mg至24 mg和7.5 mg每天2次至22.5 mg每天2次) Cmax和AUC的线性剂量依赖性。
吸收
Deutetrabenazine的口服给药后,吸收的程度是至少80%。口服给药后Deutetrabenazine的血浆浓度是一般地低于检测低限。在给药后3至4小时内达到氘化的α-HTBZ和β-HTBZ的血浆峰浓度(Cmax)。
食物的影响
在受试者给予一个单次剂量有和无食物研究食物对AUSTEDO生物利用度的影响。食物对α-HTBZ或β-HTBZ的血浆浓度时间曲线下面积(AUC)没有影响,尽管存在食物中Cmax是增加约50%[见剂量和给药方法(2.1)]。
分布
AUSTEDO的α-HTBZ,和β-HTBZ代谢物的中位分布容积(Vc/F)是分别约500 L和730 L。
在人中PET-扫描研究结果显示11C-标记的四苯嗪或α-HTBZ静脉注射后,放射性被迅速地分布至大脑,有最高结合在纹状体中和最低结合在皮质中。.
在人血浆中对浓度范围从50至200 ng/mL检查四苯嗪,α-HTBZ,和β-HTBZ的体外的蛋白结合。四苯嗪结合范围从82%至85%,α-HTBZ结合范围从60%至68%,和β-HTBZ结合范围从59%至63%。
消除
AUSTEDO是主要地在代谢物型式肾脏地消除。
从deutetrabenazine总(α+β)-HTBZ的半衰期是约9至10小时。在亨丁顿病人群患者中AUSTEDO的α-HTBZ,和β-HTBZ代谢物中位清除率值(CL/F)为分别约47 L/hour和70 L/hour。
代谢
在人肝微粒体体外实验显示deutetrabenazine被广泛地生物转化,主要地通过羰基还原酶,至其主要活性代谢物,α-HTBZ和β-HTBZ,它们随后主要地被CYP2D6代谢,与CYP1A2和CYP3A4/5有次要贡献,形成几种次要代谢物。
排泄
在6例健康受试者中一项物料平衡研究,75%至86%的deutetrabenazine剂量在尿中被排泄,而粪回收占剂量的8%至11%。来自deutetrabenazine的α-HTBZ和β-HTBZ代谢物尿排泄各占低于给药剂量的10%。Deutetrabenazine的α-HTBZ和β-HTBZ代谢物的硫酸和葡萄糖醛酸结合物,以及氧化代谢的产物,占尿中代谢物的多数。
特殊人群
男性和女性患者
性别对deutetrabenazine的α-HTBZ和β-HTBZ的药代动力学无明显影响。.
有肾受损患者
未曽进行临床研究评估肾受损对AUSTEDO的PK的影响。.
有肝受损患者
未曽研究肝受损对deutetrabenazine及其主要代谢物的药代动力学的影响。但是,进行在临床研究评估肝受损对四苯嗪药代动力学影响,一种密切相关VMAT2抑制剂,在有肝受损患者对α-HTBZ和β-HTBZ的暴露是较大至40%,而在有肝受损患者均数四苯嗪的Cmax为至190-倍较高于健康受试者[见禁忌证(4),在特殊人群中使用(8.6)]。
CYP2D6差代谢者
尽管未曽在不表达药物代谢酶CYP2D6患者系统地评价deutetrabenazine及其代谢物的药代动力学,他可能对α-HTBZ和β-HTBZ的暴露将与用强CYP2D 抑制剂相似地被增加(约3-倍)[见剂量和给药方法(2.4),药物相互作用(7.1)].
药物相互作用研究
未曽在体外研究评价Deutetrabenazine,α-HTBZ,和β-HTBZ对CYP 酶诱导作用或抑制作用或与P-糖蛋白相互作用。四苯嗪的体外研究结果不提示四苯嗪或其α-HTBZ或β-HTBZ代谢物是可能地导致CYP2D6,CYP1A2,CYP2B6,CYP2C8,CYP2C9,CYP2C19,CYP2E1,或CYP3A临床上意义的的抑制作用。体外研究提示四苯嗪不像是其α-HTBZ或β-HTBZ代谢物也不像是导致CYP1A2,CYP3A4,CYP2B6,CYP2C8,CYP2C9,或CYP2C19的临床意义的诱导作用。四苯嗪不像其α-HTBZ或β-HTBZ代谢物也不像是在体内在临床上相关浓度是P-糖蛋白的底物或抑制剂。
在一组的体外药物-药物相互作用研究曽评价Deutetrabenazine的代谢物,β-HTBZ的2-甲基丙酸(M1)和单羟基四苯嗪(M4);结果表明M1/M4预期不会致临床相关药物相互作用。
CYP2D6抑制剂
体外研究表明deutetrabenazine的α-HTBZ和β-HTBZ代谢物是对CYP2D6底物。在24例健康受试者每天的给予强CYP2D6抑制剂帕罗西汀20 mg后8天,给予单次22.5 mg剂量的deutetrabenazine后研究对deutetrabenazine及其代谢物的药代动力学研究CYP2D6抑制作用的影响。在存在帕罗西汀,α-HTBZ的全身暴露(AUCinf)为1.9-倍较高和β-HTBZ为6.5-倍较高,导致对总(α+β)-HTBZ的AUCinf约3-倍增加。帕罗西汀减低AUSTEDO的α-HTBZ和β-HTBZ代谢物的清除率有相应的均数半衰期分别约1.5-倍和2.7-倍。在存在帕罗西汀中,α-HTBZ和β-HTBZ的Cmax为分别1.2-倍和2.2-倍较高。
未曽评价中度或弱CYP2D6抑制剂例如度洛西汀[duloxetine],特比萘芬[terbinafine],胺碘酮[amiodarone],或舍曲林[sertraline]对deutetrabenazine及其代谢物暴露的影响。
地高辛[Digoxin]
未曽评价AUSTEDO对与地高辛相互作用。地高辛是对P糖蛋白的底物。在健康受试者一项研究显示四苯嗪(25 mg每天2次共3天)不影响地高辛的生物利用度,提示在这个剂量,四苯嗪不影响在肠道中P-糖蛋白。体外研究也不提示四苯嗪或其代谢物是P-糖蛋白抑制剂。
13 非临床毒理学
13.1 癌发生,突变发生,生育力受损
癌发生
未进行用deutetrabenazine致癌性研究。.
用p53+/–转基因小鼠用口服四苯嗪在剂量0,5,15,和30 mg/kg/day治疗共26周未观察到肿瘤增加。
突变发生
Deutetrabenazine及其氘化的α-HTBZ和β-HTBZ代谢物在体外(细菌回复突变和在人外周血淋巴细胞染色体畸变)试验在存在或缺乏代谢活化和在体内小鼠中微核试验为阴性。
生育力受损
尚未评价deutetrabenazine对生育力的影响。口服给予deutetrabenazine(剂量5,10,或30 mg/kg/day)至雌性大鼠共3个月在所有剂量导致动情周期破坏;在体表面积(mg/m2)的基础上最低测试剂量是相似于人最大推荐剂量(48 mg/day)。
雌性大鼠交配前和自始至终,四苯嗪的口服给药(剂量5,15,或30 mg/kg/day),和继续至妊娠天7,导致破坏的动情周期性在剂量大于5 mg/kg/day。对雄性交配和生育力指数或精子参数(运动力,计数,密度),当未治疗雌性交配前和自始至终用四苯嗪口服治疗在剂量5,15或30 mg/kg/day未观察到影响。
14 临床研究
双盲,安慰剂-对照研究
在研究1, 90例能走动的患者有表现舞蹈症伴随亨丁顿病进行一项随机化,双盲,安慰剂-对照,多中心试验主要地确定AUSTEDO作为对舞蹈症伴随亨丁顿病治疗的疗效。亨丁顿病的诊断是根据家族史,神经学检查,和遗传测试。治疗时间为周,包括一个 8-周给药滴定调整阶段和一个4-周维持阶段,接着被一个1-周冲洗。患者不是盲态至终止。AUSTEDO在6 mg被开始每天和向上滴定调整,在每周间隔,在6 mg增量直至实现舞蹈症满意的的治疗,发生不可耐受的副作用,或直至达到一个最大剂量的48 mg每天。主要疗效终点为总最大舞蹈症评分,统一的亨丁顿病计分量表(UHDRS)的一个项目。在这个量表,舞蹈症对机体的7个不同部分被计分从0至4(用0代表无舞蹈症)。总评分范围从0至28。在90例被纳入患者中,87患者完成该研究。均数年龄为54(范围23至74)。患者为56%男性和92%高加索人。在滴定调整后均数剂量为40 mg每天。表3和图1总结了根据总最大舞蹈症评分AUSTEDO对舞蹈症的影响。对接受AUSTEDO患者总最大舞蹈症评分从基线改善约4.4单位至平均维持阶段(周9和周12的平均),与之比较在安慰剂组约1.9单位。-2.5单位的效应为统计显著(p<0.0001)。维持终点为对周9和周12访问时总最大舞蹈症评分的均数。在周13随访时(1研究药物的终止后1周),曽接受AUSTEDO患者的总最大舞蹈症评分返回至基线(图1).
图1. 在研究1中总最大舞蹈症评分随时间变化
图2. 在研究1中总最大舞蹈症评分变化的分布。
图2显示研究1中总最大舞蹈症评分值的分布。阴性值表明舞蹈症中一个减低和阳性数量表示舞蹈症增加。
一个患者如何计分他们的总体亨丁顿病症状-评估的变化的计数全面印象。51%的用AUSTEDO治疗患者在治疗结束时计分他们的症状为“大为改进[Much Improved]”或“非常大改进[Very Much Improved]”,与之比较,安慰剂-治疗患者为20%。在医生计分的临床变化全面印象中,在治疗结束时用AUSTEDO治疗患者42%他们的症状被计为“大为改进”或“非常大改进”,与之比较安慰剂-治疗患者为13%。
16 如何供应/贮存和处置
16.1 如何供应
AUSTEDO片可得到以下的规格和包装:
6 mg:圆形,紫色-涂层片,在一侧黑墨水打印有“SD”上面“6”。
60片瓶:NDC 68546-170-60.
9 mg:圆形,蓝色-涂层片,在一侧黑墨水打印有“SD”上面“9”.
60片瓶:NDC 68546-171-60.
12 mg:圆形,米色-涂层片,在一侧黑墨水打印有“SD”上面“12”.
60片瓶:NDC 68546-172-60.
16.2 贮存
贮存在25º C(77ºF);外出允许至15°C至30ºC(59°F至86ºF)[见USP控制室温]。避光和潮湿保护。
17 患者咨询资料
建议患者或护理人员阅读FDA-批准的患者说明书(用药指南)。
给药指导
建议患者与食物服用AUSTEDO。AUSTEDO片应被整吞和不要咀嚼,压碎或破碎[见剂量和给药方法(2.1)]。
抑郁和自杀的风险
忠告患者,他们的护理人员,和家庭AUSTEDO可能增加抑郁,恶化 抑郁,和自杀倾向的风险,和任何症状立即报告给卫生保健提供者[见禁忌证(4),警告和注意事项(5.2)]。
镇静和嗜睡的风险
忠告患者AUSTEDO可能致镇静和嗜睡和可能损害进行需要复杂运动和精神技能任务的能力。直至他们学习任何他们对稳定剂量AUSTEDO反应,患者应小心做需要他们警觉的活动,例如驾驶车辆或操作机械[见警告和注意事项(5.6)]。
与酒精或其他镇静药物相互作用
忠告患者酒精或其他致想睡将恶化嗜睡药物[见药物相互作用(7.5)]。
同时药物
建议患者告知他们的医生自己服用的所有药物和因为一个潜在的相互作用在开始任何新药物前咨询他们的卫生保健提供者[见禁忌证(4)和药物相互作用(7.1,7.4,7.5,7.6)]
Austedo(deutetrabenazine)片说明书
2017年4月第一版
批准日前:2017年4月3日;
公司:Teva Pharmaceutical Industries Ltd.
为治疗:亨丁顿舞蹈病
这些重点不包括安全和有效使用AUSTEDO需所有资料。请参阅AUSTEDO完整处方资料。
AUSTEDO™(deutetrabenazine)片,为口服使用
美国初次批准:2017
适应证和用途
AUSTEDO是一种血管单按转运蛋白2(VMAT2)抑制剂适用为伴随亨丁顿病[Huntington’s disease]舞蹈症的治疗(1)
剂量和给药方法
● 开始剂量是6 mg每天1次。在每周间隔向上滴定调整by 6 mg每天至一个减低舞蹈症的耐受剂量,至一个最大每天推荐剂量48 mg(24 mg每天2次)(2.1)
● 在两个分次剂量中给予每天总剂量12 mg或以上 (2.1)
● 与食物给予(2.1)
● 吞咽整片;不要咀嚼,粉碎或破碎片(2.1)
● 如患者从四苯嗪[tetrabenazine]转换,终止四苯嗪和在后面天开始AUSTEDO。对推荐的转换片见完整处方资料(2.2)
● AUSTEDO在CYP2D6差代谢者中最大推荐剂量为36 mg每天(即,18 mg每天2次)(2.4,8.7)
剂型和规格
片:6 mg,9 mg,和12 mg(3)
禁忌证
● 有自杀倾向的,或未治疗的/不佳地治疗抑郁(4,5.2)
● 肝受损(4,8.6,12.3)
● 用MAOIs,利血平[reserpine],或四苯嗪(XENAZINE®)(4,7.2,7.3,7.7)
警告和注意事项
● 抗精神病药物恶性症候群[Neuroleptic Malignant Syndrome](NMS):如这发生终止(5.3,7.4)
● 静坐不能,激动,不安,和帕金森症:如这发生减低剂量或终止。(5.4,5.5)
● 镇静/嗜睡:可能损害患者驾驶或操作复杂机械的能力(5.6)
不良反应
最常见不良反应(>8%的AUSTEDO-治疗患者和大于安慰剂)为:嗜睡,腹泻,口干,和疲乏(6.1)
药物相互作用
●强CYP2D6抑制剂的同时使用:AUSTEDO的最大推荐剂量是36 mg每天(18 mg每天2次)(2.3,7.1)
● 酒精或其他镇静药物:可能有相加镇静和嗜睡(7.5)
在特殊人群中使用
妊娠:根据动物数据,可能致胎儿危害(8.1)
完整处方资料
1 适应证和用途
AUSTEDO™是适用为伴随亨丁顿病舞蹈症的治疗。
2 剂量和给药方法
2.1 给药资料
AUSTEDO的剂量被个体地根据舞蹈症的减低和耐受性确定。当首次处方给予不是正在从四苯嗪(一个相关的VMAT2抑制剂)转换患者,AUSTEDO的推荐开始剂量是6 mg口服给予每天1次。
● AUSTEDO的剂量可能以每周间隔在每天增量6 mg至一个最大每天推荐剂量48 mg。
● 在两个分次剂量中给予每天总剂量12 mg或以上.
● 与食物给予AUSTEDO [见临床药理学(12.3)].
● 整吞AUSTEDO片。不要咀嚼,粉碎或破碎片。
2.2 患者从四苯嗪(XENAZINE®)转换至AUSTEDO
在以下天终止四苯嗪(XENAZINE®)和开始AUSTEDO。在表1显示患者从四苯嗪(XENAZINE®)转换转换至AUSTEDO推荐的初始给药方案。
患者被转换至AUSTEDO后,剂量可能在每周间隔被调整[见剂量和给药方法(2.1)]。
2.3 与强CYP2D6抑制剂剂量调整
在接受强CYP2D6抑制剂患者中(如,奎尼丁[quinidine],抗抑郁剂例如帕罗西汀[paroxetine],氟西汀[fluoxetine],和安非他酮[bupropion]),AUSTEDO的总每天剂量不应超过36 mg(最大单次剂量18 mg)[见药物相互作用(7.1)和临床药理学(12.3)]。
2.4 在CYP2D6差代谢者中剂量调整
在患者是CYP2D6差代谢者,AUSTEDO的每天总剂量不应超过36 mg(最大单次剂量18 mg)[见在特殊人群中使用(8.7)]。
2.5 治疗的终止和中断
用AUSTEDO治疗可能无滴定调整被终止。治疗的中断后大于一周,当恢复时AUSTEDO治疗应被再次滴定调整。对治疗中断少于一周,治疗可能没有滴定调整在以前维持剂量被恢复。
3 剂型和规格
AUSTEDO片可得到以下规格:
● 6 mg片是圆形,紫色-涂层片,在一侧黑墨水打印有“SD”上面“6”.
● 9 mg片是圆形,蓝色-涂层片,在一侧黑墨水打印有 “SD”上面“9”.
● 12 mg片是圆形,米色-涂层片,在一侧黑墨水打印有 “SD”上面“12”.
4 禁忌证
在以下患者禁忌AUSTEDO:
● 患者是有自杀倾向的,或n有未治疗或不适当地治疗的抑郁患者[见警告和注意事项(5.2)].
● 有肝受损[见在特殊人群中使用(8.6),临床药理学(12.3)].
● 用单胺氧化酶抑制剂(MAOIs)。AUSTEDO不应与一个MAOI联用,或用一个MAOI治疗终止的14天内[见药物相互作用(7.3)]。
● 开始AUSTEDO前用,利血平应停止后过后至少20天[见药物相互作用(7.2)]。
● 用四苯嗪(XENAZINE®)[见药物相互作用(7.7)].
5 警告和注意事项
5.1 临床恶化和不良事件
亨丁顿病是一种进行性疾患特征是情绪,认知,舞蹈症,强直,和功能性能力随时间。VMAT2 抑制剂,包括AUSTEDO,可能致一个情绪,认知,强直,和功能性能力恶化。
处方者应定期地再评价对AUSTEDO需要在他们的患者通过评价对舞蹈症效应和可能不良效应,包括镇静/嗜睡,抑郁和自杀倾向,帕金森症,静坐不能,不安,和认知下降。可能难以区分不良反应和潜在疾病的进展间;减低剂量或停止药物可能有助于临床医生区分两种可能性。在有些患者,潜在舞蹈症本身可能随时间改善,减低对AUSTEDO需求。
5.2 抑郁和自杀倾向
有亨丁顿病患者是处于对抑郁,和有自杀倾向的观念或行为(自杀倾向)的风险增加。在有亨丁顿病患者中AUSTEDO可能增加对自杀倾向风险。.
在一项12-周,双盲,安慰剂-对照试验,报道2%的用AUSTEDO治疗患者有自杀倾向的观念,相比较无用安慰剂患者;被报道无自杀企图和无已完成的自杀。用AUSTEDO治疗患者报告抑郁为4%。
当考虑AUSTEDO的使用,应平衡自杀倾向的风险与对舞蹈症的治疗的需求。所有用AUSTEDO治疗患者应被观察对新或恶化的抑郁或自杀倾向。如抑郁或自杀倾向没有解决,考虑终止用AUSTEDO治疗。
患者,他们的护理人员,和家庭应被告知伴随AUSTEDO抑郁,恶化 抑郁,和自杀倾向的风险,和应被指导报告关注的行为及时地至治疗医生。有亨丁顿病患者表现有自杀倾向的观念 应立即被评价。
5.3 抗精神病药物恶性症候群(NMS)
曽被报道一种潜在地致命症状复合有时被称为抗精神病药物恶性症候群(NMS)在伴随药物减低多巴胺传导[见药物相互作用(7.4)]。而在接受AUSTEDO患者中未曽观察到NMS,在接受四苯嗪(一种密切相关的VMAT2抑制剂)患者曽被观察到。
临床医生应警惕伴随NMS体征和症状变化。NMS的临床表现是高热,肌肉强直,改变的精神状态,和自主神经不稳定的证据(不规则脉搏或血压,心动过速,出汗,和心律失常),附加征象可能包括升高的肌酐磷酸化酶,肌红蛋白尿,横纹肌溶解症,和急性肾衰。NMS的诊断可能被复杂化;其他严重的医学疾患(如,肺炎,全身感染)和可能存在未治疗或治疗不足的锥体外系疾病与相似体征和症状。在鉴别诊断中其他重要考虑包括中枢抗胆碱能毒性,热中暑,药物热,和主要中枢神经系统病理学。
NMS的处理应包括(1) 立即终止AUSTEDO;(2) 强化对症治疗和医学监视;和(3) 可得到的任何同时严重医学问题的治疗特殊治疗。对NMS有关特殊药理学治疗没有一般一致的方案。
曽报告NMS的复发与药物治疗的恢复。如从NMS恢复后需要用AUSTEDO治疗,患者应被监视对复发征象。
5.4 静坐不能,激动,和不安
在有亨丁顿病患者中AUSTEDO可能增加静坐不能,激动,和不安的风险。在一项12-周,双盲,安慰剂-对照试验, 4%用AUSTEDO治疗患者报道静坐不能,激动,或不安与之比较用安慰剂患者为2%。
接受AUSTEDO患者应被监视不安和激动的体征和症状,因为这些可能是发生静坐不能的指示指标。如一例患者用AUSTEDO治疗期间发生静坐不能,AUSTEDO剂量应被减低;有些患者可能需要终止治疗。
5.5 帕金森症
在有亨丁顿病患者中AUSTEDO可能致帕金森症。
因为在亨丁顿病强直可能发展作为潜在疾病过程的一部分,可能这个潜在的药物-诱发不良反应和潜在疾病过程的进展间难以区分。对有些有亨丁顿病患者药物-诱发帕金森症有潜能致比未治疗的舞蹈症的更功能性残疾。如一例患者用AUSTEDO治疗期间发生帕金森症, AUSTEDO剂量应被减低;有些患者可能需要终止治疗。
5.6 镇静和嗜睡
镇静是AUSTEDO常见剂量-限制不良反应。在一项12-周,双盲,安慰剂-对照试验,11%的AUSTEDO-治疗患者报告嗜睡与之比较用安慰剂患者为4%和9%的AUSTEDO-治疗患者报告疲乏相比较有4%的安慰剂-治疗患者。
患者不应进行需要精神警惕性活动以保持他们自身或其他人安全性,例如操作汽车或操作危害性机械,直至他们是用维持剂量的AUSTEDO和知道药物如何影响他们。
5.7 QTc延长
四苯嗪,一种密切相关的VMAT2 抑制剂,在校正的QT(QTc)间隔中致一个增加(约8 msec)[见临床药理学(12.2)]。
在有些用AUSTEDO治疗患者是CYP2D6差代谢者或被一个强CYP2D6抑制剂共同给药,可能发生一个临床上相关的QT延长[见临床药理学(12.2,12.3)]。
AUSTEDO的使用应避免与其他已知延长QTc药物联用[见药物相互作用(7.6)]。
在有先天性长QT综合证患者和在有一个心脏心律失常病史患者也应避免使用AUSTEDO。某些情况可能增加尖端扭转型室性心动过速的发生的风险和/或突然死亡在伴随延长QTc间期药物的使用,包括(1) 心动过缓;(2) 低钾血症或低镁血症;(3) 延长QTc间期其他药物的同时使用;和(4) 先天性QT间期的延长的存在。
5.8 高催乳素血症
在AUSTEDO开发计划没有评价血清催乳素水平。
四苯嗪,一种密切相关的VMAT2 抑制剂,在人中升高血清催乳素浓度。给予25 mg 的四苯嗪至健康志愿者后,血浆催乳素峰水平增加4-至5-倍。.
组织培养实验表明约三分之一人乳癌是体外催乳素-依赖,一种潜在重要性因子如AUSTEDO 正在被考虑为一个以前检测到乳癌患者。虽然无月经,乳溢症,男性乳房增大症,和阳痿可能被升高的血清催乳素浓度引起,对大多数患者不知道升高的血清催乳素浓度的临床意义。
血清催乳素水平中慢性增加(虽然在AUSTEDO或四苯嗪开放计划中没有评价)曽被伴随低水平雌激素和增加骨质疏松症风险。应做是否有一个症状性高催乳素血症的临床怀疑,适当实验室测试和应被考虑给予终止AUSTEDO。
5.9 结合至含黑色素组织
因为deutetrabenazine或其代谢物结合至含黑色素组织,随时间它可能蓄积在这些组织。广泛使用后这个升高有可能性AUSTEDO可能在这些组织中致毒性。在有色素动物种属例如犬的慢性毒性研究没有眼科学也没有进行眼显微镜检查。
在人中眼科学监视不足以排除长期接触后发生的损伤的可能性.
不知道deutetrabenazine的结合至含黑色素组织的临床相关性。
尽管对定期地眼科学监视没有专门建议,处方者应认识到长期眼科学效应的可能性[见临床药理学(12.2)]。
6 不良反应
在说明书的其他节更详细讨论以下严重的不良反应:
● 抑郁和自杀倾向[见警告和注意事项(5.2)]
● 抗精神病药物恶性症候群(NMS)[见警告和注意事项(5.3)]
● 静坐不能,激动,和不安[见警告和注意事项(5.4)]
● 帕金森症[见警告和注意事项(5.5)]
● 镇静和嗜睡[见警告和注意事项(5.6)]
● QTc延长[见警告和注意事项(5.7)]
● 高催乳素血症[见警告和注意事项(5.8)]
● 结合至含黑色素组织[见警告和注意事项(5.9)]
6.1 临床试验经验
因为临床试验是在广泛不同情况下进行的,临床试验观察到不良反应率不能与另一种药临床试验发生率直接比较而且可能不反映实践中观察到的发生率。
研究1是在有舞蹈症伴随亨丁顿病患者一项随机化,12-周,安慰剂-对照研究。总共45例患者接受AUSTEDO,和45例患者接受安慰剂。患者年龄范围在年龄23和74岁间(均数54岁);56% 为男性,和92%为高加索人。最常见不良反应发生在大于8%的AUSTEDO-治疗患者为嗜睡,腹泻,口干,和疲乏。表2中总结发生在4%或更多用AUSTEDO治疗患者,和比用安慰剂患者更大发生率的不良反应。
在研究1在7%的患者中一种或更多不良反应导致研究药物的剂量减低。在接受AUSTEDO患者中最常不良反应导致剂量减低为眩晕(4%).
在研究1中激动导致在2%的用AUSTEDO治疗患者终止。
7 药物相互作用
7.1 强CYP2D6抑制剂
在维持用一个稳定剂量的AUSTEDO患者当添加一个强CYP2D6抑制剂在AUSTEDO剂量可能被需要一个减低。使用强CYP2D6抑制剂的同时(如,帕罗西汀,氟西汀,奎尼丁,安非他酮)曽被显示增加对deutetrabenazine活性双氢-代谢物的全身暴露约3-倍。AUSTEDO的每天剂量不应超过36 mg每天,而在用强CYP2D6抑制剂患者AUSTEDO的最大单次剂量不应超过18 mg [见剂量和给药方法(2.3)和临床药理学(12.3)]。
7.2 利血平
利血平与VMAT2不可逆地结合和它的效应时间为几天。给予AUSTEDO前处方者应等待对舞蹈症重现有助于减低药物过量的风险和在中枢神经系统中5羟色胺和去甲肾上腺素重大耗竭。
停止利血平后开始AUSTEDO前至少要经过20天。AUSTEDO和利血平不应同时使用[见禁忌证(4)]。
7.3 单胺氧化酶抑制剂(MAOIs)
在用MAOIs患者中禁忌AUSTEDO。AUSTEDO不应与一个MAOI联用,或治疗用一个MAOI终止14天内使用[见禁忌证(4)]。
7.4 神经抑制药药物[Neuroleptic Drugs]
对帕金森症,NMS,和静坐不能的风险可能被增加AUSTEDO和多巴胺[dopamine]拮抗剂或抗精神活动的药物的同时使用增加。
7.5 酒精或其他镇静药物
酒精或其他镇静药物的同时使用可能有相加效应和恶化镇静和嗜睡[见警告和注意事项(5.6)]。
7.6 致QTc延长药物
四苯嗪,一种密切相关的VMAT2抑制剂,致一个小增加在校正的QT(QTc)间期。用AUSTEDO也可能发生临床上相关的QT延长[见警告和注意事项(5.7),临床药理学(12.2)]。
AUSTEDO的使用应被避免与已知延长QTc其他药物联用,包括抗精神病药物(如,氯丙嗪[chlorpromazine],氟哌啶醇[haloperidol],甲硫达嗪[thioridazine],齐拉西酮[ziprasidone]),抗生素(如,莫西沙星[moxifloxacin]),类型1A(如,奎尼丁,普鲁卡因酰胺[procainamide]),和类型III(如,胺碘酮[amiodarone],苏特罗[sotalol])抗心律失常药物或任何其他延长QTc间期药物。
7.7 四苯嗪
在当前用四苯嗪患者禁忌AUSTEDO。四苯嗪终止后天可开始AUSTEDO[见剂量和给药方法(2.2)]。
8 在特殊人群中使用
8.1 妊娠
风险总结
在妊娠妇女AUSTEDO的使用伴随对发育风险没有适当的数据。器官形成期时给予大鼠deutetrabenazine对胚胎胎儿发育没有产生明确的不良效应。但是,对大鼠妊娠和哺乳自始至终给予四苯嗪导致死胎增加和产后子代死亡率的增加[见数据]。
在美国一般人群,重大出生缺陷和在临床上认可妊娠流产的估算背景风险分别为2-4%和15-20%。不知道在适应证人群重大出生缺陷和流产的背景风险。
数据
动物数据
器官形成期时Deutetrabenazine的口服给药(5,10,或30 mg/kg/day)或四苯嗪(30 mg/kg/day)至妊娠大鼠对胚胎胎儿发育无明确的影响。在体表面积(mg/m2)基础最高被测试的剂量为最大推荐人剂量48 mg/day的6倍。
未曽评估器官形成期时对兔或妊娠和哺乳时对大鼠给予deutetrabenazine的影响。
当妊娠兔器官形成期阶段时在口服剂量至60 mg/kg/day给予四苯嗪对胚胎胎儿发育无影响。当四苯嗪被给予至雌性大鼠(剂量5,15,和30 mg/kg/day)从器官形成期开始至哺乳阶段,在15和30 mg/kg/day观察到死胎和产后出生的子代死亡率增加,和在所有剂量观察到幼畜成熟延迟。
8.2 哺乳
风险总结
没有在人乳汁中deutetrabenazine或其代谢物的存在,对哺乳喂养婴儿的影响,或药物对乳汁产生影响的数据。哺乳喂养的发育和健康获益应与对AUSTEDO母亲的临床需求和对哺乳喂养婴儿来自AUSTEDO或来自母体潜在情况任何潜在不良效应一并考虑。
8.4 儿童使用
尚未在儿童患者中确定安全性和有效性。
8.5 老年人使用
AUSTEDO的临床研究没有包括充分数量受试者年龄65和以上以确定是否他们反应不同于较年轻受试者。其他被报道的临床经验没有确定老年和较年轻患者间反应中差别。一般说来,对一位老年患者剂量选择应谨慎,通常开始在给药范围的低端,反映肝,肾和心脏功能失调,和同时疾病或其他药物治疗的更大频数。
8.6 肝受损
尚未研究肝受损对deutetrabenazine及其主要代谢物的药代动力学的影响;但是,在用四苯嗪进行的一项临床研究,一种密切相关的VMAT2抑制剂,在有肝受损患者对四苯嗪及其活性代谢物暴露有巨大增加。尚未评价这个增加暴露的临床意义,但是因为关注对严重的不良反应更大风险,在有肝受损患者禁忌使用AUSTEDO[见禁忌证(4),临床药理学(12.3)]。
8.7 CYP2D6差代谢者
尽管尚未曽在没有表达药物代谢酶患者中系统地评价deutetrabenazine及其代谢物的药代动力学,它是可能对α-HTBZ和β-HTBZ暴露将是增加相似于用一个强CYP2D6抑制剂(约3-倍)。在患者是CYP2D6 poor差代谢者,AUSTEDO的每天剂量不应超过36 mg(最大单次剂量为18 mg)[见剂量和给药方法(2.4)和临床药理学(12.3)]。
10 药物过量
在文献中用四苯嗪,一种密切相关的VMAT2抑制剂,曽被报道药物过量范围从100 mg至1 g。用过量给药发生以下不良反应:急性肌张力障碍,眼动危象,恶心和呕吐,出汗,镇静,低血压,混乱,腹泻,幻觉,发红,和震颤。治疗应那些与任何中枢神经系统-活性药物过量处理中应用一般措施组成。建议一般支持和对症措施。应监视心脏节律和生命征象。在处理药物过量中,应经常被考虑涉及多种药物的可能性。医生应考虑联系一个对任何过量的治疗毒物控制中心。在美国经认证的毒物控制中心美国协会网址www.aapcc.org.列出电话号码。
11 一般描述
AUSTEDO(deutetrabenazine)是一种为口服给药的血管单按转运蛋白2(VMAT2)抑制剂。 Deutetrabenazine的分子量为323.46;pKa为6.31。Deutetrabenazine为一种hexahydro-dimethoxybenzoquinolizine衍生物和有以下化学名:(RR,SS)-1,3,4,6,7,11b-hexahydro-9,10-di(methoxy-d3)-3-(2-methylpropyl)-2H-benzo[a]quinolizin-2-one.
Deutetrabenazine的分子式为C19H21D6NO3。Deutetrabenazine是一种消旋混合物含以下结构。
Deutetrabenazine是白色至浅黄色结晶粉为微溶于水和溶于乙醇。AUSTEDO片含6 mg,9 mg,或12 mg deutetrabenazine,和以下无活性成分:氢氧化铵,黑色氧化铁,n-丁醇,丁基羟基苯甲醚,丁基羟基甲苯,硬脂酸镁,甘露醇,微晶纤维素,聚乙二醇,聚氧乙烯,聚山梨醇80,聚乙烯醇,聚维酮,丙二醇,虫胶,滑石,二氧化钛,和FD&C蓝#2 lake。6 mg片还含FD&C红#40 lake。12 mg片还含FD&C黄#6 lake。
12 临床药理学
12.1 作用机制
不知道AUSTEDO(deutetrabenazine)发挥其抗-舞蹈症作用的精准机制,但被相信是与它的作用为一种来自神经神经终端器[nerve terminals]单胺(例如多巴胺,5羟色胺,去甲肾上腺素,和组胺)可逆性耗竭剂[depletory]相关。Deutetrabenazine的主要的循环代谢物(α-双氢tetrabenazine[HTBZ]和β-HTBZ),是VMAT2的可逆性抑制剂,导致单胺的摄取入突触囊泡减低和单胺贮存的耗竭。.
12.2 药效动力学
心脏电生理学
在一项随机化,双盲,安慰剂-对照交叉研究在健康男性和女性受试者用莫西沙星作为阳性对照研究一个单次12-mg或24-mg剂量的AUSTEDO对QT间期的影响。在24 mg时,AUSTEDO 引起一个约4.5 msec均数增加在QTc(90% CI:2.4,6.5 msec)。未曽评价在较高暴露对AUSTEDO或其代谢物暴露的影响。.
在一项随机化,双盲,安慰剂-对照交叉研究在健康男性和女性受试者用莫西沙星作为阳性对照研究一个单次25-mg或50-mg剂量的四苯嗪,一种密切相关的VMAT2抑制剂,对QT间期的影响。在50 mg时,四苯嗪引起一个约8 msec均数增加在QTc(90% CI:5.0,10.4 msec)。未曽评价在较高暴露对或四苯嗪或其代谢物影响[见警告和注意事项(5.7)和药物相互作用(7.6)]。
黑色素结合
在有色素大鼠中Deutetrabenazine或其代谢物结合至含黑色素组织(即,眼,皮肤,皮毛)。在单次口服剂量放射性标记的deutetrabenazine,在给药后35天时仍在眼和皮毛中检测到放射性[见警告和注意事项(5.9)]。
12.3 药代动力学
口服给药至25 mg后,deutetrabenazine的血浆浓度是一般地低于检测低限是因为广泛的肝代谢deutetrabenazine至活性氘化的二氢[dihydro]代谢物(HTBZ),α-HTBZ和β-HTBZ。观察到对活性代谢物单次或多剂量后(6 mg至24 mg和7.5 mg每天2次至22.5 mg每天2次) Cmax和AUC的线性剂量依赖性。
吸收
Deutetrabenazine的口服给药后,吸收的程度是至少80%。口服给药后Deutetrabenazine的血浆浓度是一般地低于检测低限。在给药后3至4小时内达到氘化的α-HTBZ和β-HTBZ的血浆峰浓度(Cmax)。
食物的影响
在受试者给予一个单次剂量有和无食物研究食物对AUSTEDO生物利用度的影响。食物对α-HTBZ或β-HTBZ的血浆浓度时间曲线下面积(AUC)没有影响,尽管存在食物中Cmax是增加约50%[见剂量和给药方法(2.1)]。
分布
AUSTEDO的α-HTBZ,和β-HTBZ代谢物的中位分布容积(Vc/F)是分别约500 L和730 L。
在人中PET-扫描研究结果显示11C-标记的四苯嗪或α-HTBZ静脉注射后,放射性被迅速地分布至大脑,有最高结合在纹状体中和最低结合在皮质中。.
在人血浆中对浓度范围从50至200 ng/mL检查四苯嗪,α-HTBZ,和β-HTBZ的体外的蛋白结合。四苯嗪结合范围从82%至85%,α-HTBZ结合范围从60%至68%,和β-HTBZ结合范围从59%至63%。
消除
AUSTEDO是主要地在代谢物型式肾脏地消除。
从deutetrabenazine总(α+β)-HTBZ的半衰期是约9至10小时。在亨丁顿病人群患者中AUSTEDO的α-HTBZ,和β-HTBZ代谢物中位清除率值(CL/F)为分别约47 L/hour和70 L/hour。
代谢
在人肝微粒体体外实验显示deutetrabenazine被广泛地生物转化,主要地通过羰基还原酶,至其主要活性代谢物,α-HTBZ和β-HTBZ,它们随后主要地被CYP2D6代谢,与CYP1A2和CYP3A4/5有次要贡献,形成几种次要代谢物。
排泄
在6例健康受试者中一项物料平衡研究,75%至86%的deutetrabenazine剂量在尿中被排泄,而粪回收占剂量的8%至11%。来自deutetrabenazine的α-HTBZ和β-HTBZ代谢物尿排泄各占低于给药剂量的10%。Deutetrabenazine的α-HTBZ和β-HTBZ代谢物的硫酸和葡萄糖醛酸结合物,以及氧化代谢的产物,占尿中代谢物的多数。
特殊人群
男性和女性患者
性别对deutetrabenazine的α-HTBZ和β-HTBZ的药代动力学无明显影响。.
有肾受损患者
未曽进行临床研究评估肾受损对AUSTEDO的PK的影响。.
有肝受损患者
未曽研究肝受损对deutetrabenazine及其主要代谢物的药代动力学的影响。但是,进行在临床研究评估肝受损对四苯嗪药代动力学影响,一种密切相关VMAT2抑制剂,在有肝受损患者对α-HTBZ和β-HTBZ的暴露是较大至40%,而在有肝受损患者均数四苯嗪的Cmax为至190-倍较高于健康受试者[见禁忌证(4),在特殊人群中使用(8.6)]。
CYP2D6差代谢者
尽管未曽在不表达药物代谢酶CYP2D6患者系统地评价deutetrabenazine及其代谢物的药代动力学,他可能对α-HTBZ和β-HTBZ的暴露将与用强CYP2D 抑制剂相似地被增加(约3-倍)[见剂量和给药方法(2.4),药物相互作用(7.1)].
药物相互作用研究
未曽在体外研究评价Deutetrabenazine,α-HTBZ,和β-HTBZ对CYP 酶诱导作用或抑制作用或与P-糖蛋白相互作用。四苯嗪的体外研究结果不提示四苯嗪或其α-HTBZ或β-HTBZ代谢物是可能地导致CYP2D6,CYP1A2,CYP2B6,CYP2C8,CYP2C9,CYP2C19,CYP2E1,或CYP3A临床上意义的的抑制作用。体外研究提示四苯嗪不像是其α-HTBZ或β-HTBZ代谢物也不像是导致CYP1A2,CYP3A4,CYP2B6,CYP2C8,CYP2C9,或CYP2C19的临床意义的诱导作用。四苯嗪不像其α-HTBZ或β-HTBZ代谢物也不像是在体内在临床上相关浓度是P-糖蛋白的底物或抑制剂。
在一组的体外药物-药物相互作用研究曽评价Deutetrabenazine的代谢物,β-HTBZ的2-甲基丙酸(M1)和单羟基四苯嗪(M4);结果表明M1/M4预期不会致临床相关药物相互作用。
CYP2D6抑制剂
体外研究表明deutetrabenazine的α-HTBZ和β-HTBZ代谢物是对CYP2D6底物。在24例健康受试者每天的给予强CYP2D6抑制剂帕罗西汀20 mg后8天,给予单次22.5 mg剂量的deutetrabenazine后研究对deutetrabenazine及其代谢物的药代动力学研究CYP2D6抑制作用的影响。在存在帕罗西汀,α-HTBZ的全身暴露(AUCinf)为1.9-倍较高和β-HTBZ为6.5-倍较高,导致对总(α+β)-HTBZ的AUCinf约3-倍增加。帕罗西汀减低AUSTEDO的α-HTBZ和β-HTBZ代谢物的清除率有相应的均数半衰期分别约1.5-倍和2.7-倍。在存在帕罗西汀中,α-HTBZ和β-HTBZ的Cmax为分别1.2-倍和2.2-倍较高。
未曽评价中度或弱CYP2D6抑制剂例如度洛西汀[duloxetine],特比萘芬[terbinafine],胺碘酮[amiodarone],或舍曲林[sertraline]对deutetrabenazine及其代谢物暴露的影响。
地高辛[Digoxin]
未曽评价AUSTEDO对与地高辛相互作用。地高辛是对P糖蛋白的底物。在健康受试者一项研究显示四苯嗪(25 mg每天2次共3天)不影响地高辛的生物利用度,提示在这个剂量,四苯嗪不影响在肠道中P-糖蛋白。体外研究也不提示四苯嗪或其代谢物是P-糖蛋白抑制剂。
13 非临床毒理学
13.1 癌发生,突变发生,生育力受损
癌发生
未进行用deutetrabenazine致癌性研究。.
用p53+/–转基因小鼠用口服四苯嗪在剂量0,5,15,和30 mg/kg/day治疗共26周未观察到肿瘤增加。
突变发生
Deutetrabenazine及其氘化的α-HTBZ和β-HTBZ代谢物在体外(细菌回复突变和在人外周血淋巴细胞染色体畸变)试验在存在或缺乏代谢活化和在体内小鼠中微核试验为阴性。
生育力受损
尚未评价deutetrabenazine对生育力的影响。口服给予deutetrabenazine(剂量5,10,或30 mg/kg/day)至雌性大鼠共3个月在所有剂量导致动情周期破坏;在体表面积(mg/m2)的基础上最低测试剂量是相似于人最大推荐剂量(48 mg/day)。
雌性大鼠交配前和自始至终,四苯嗪的口服给药(剂量5,15,或30 mg/kg/day),和继续至妊娠天7,导致破坏的动情周期性在剂量大于5 mg/kg/day。对雄性交配和生育力指数或精子参数(运动力,计数,密度),当未治疗雌性交配前和自始至终用四苯嗪口服治疗在剂量5,15或30 mg/kg/day未观察到影响。
14 临床研究
双盲,安慰剂-对照研究
在研究1, 90例能走动的患者有表现舞蹈症伴随亨丁顿病进行一项随机化,双盲,安慰剂-对照,多中心试验主要地确定AUSTEDO作为对舞蹈症伴随亨丁顿病治疗的疗效。亨丁顿病的诊断是根据家族史,神经学检查,和遗传测试。治疗时间为周,包括一个 8-周给药滴定调整阶段和一个4-周维持阶段,接着被一个1-周冲洗。患者不是盲态至终止。AUSTEDO在6 mg被开始每天和向上滴定调整,在每周间隔,在6 mg增量直至实现舞蹈症满意的的治疗,发生不可耐受的副作用,或直至达到一个最大剂量的48 mg每天。主要疗效终点为总最大舞蹈症评分,统一的亨丁顿病计分量表(UHDRS)的一个项目。在这个量表,舞蹈症对机体的7个不同部分被计分从0至4(用0代表无舞蹈症)。总评分范围从0至28。在90例被纳入患者中,87患者完成该研究。均数年龄为54(范围23至74)。患者为56%男性和92%高加索人。在滴定调整后均数剂量为40 mg每天。表3和图1总结了根据总最大舞蹈症评分AUSTEDO对舞蹈症的影响。对接受AUSTEDO患者总最大舞蹈症评分从基线改善约4.4单位至平均维持阶段(周9和周12的平均),与之比较在安慰剂组约1.9单位。-2.5单位的效应为统计显著(p<0.0001)。维持终点为对周9和周12访问时总最大舞蹈症评分的均数。在周13随访时(1研究药物的终止后1周),曽接受AUSTEDO患者的总最大舞蹈症评分返回至基线(图1).
图1. 在研究1中总最大舞蹈症评分随时间变化
图2. 在研究1中总最大舞蹈症评分变化的分布。
图2显示研究1中总最大舞蹈症评分值的分布。阴性值表明舞蹈症中一个减低和阳性数量表示舞蹈症增加。
一个患者如何计分他们的总体亨丁顿病症状-评估的变化的计数全面印象。51%的用AUSTEDO治疗患者在治疗结束时计分他们的症状为“大为改进[Much Improved]”或“非常大改进[Very Much Improved]”,与之比较,安慰剂-治疗患者为20%。在医生计分的临床变化全面印象中,在治疗结束时用AUSTEDO治疗患者42%他们的症状被计为“大为改进”或“非常大改进”,与之比较安慰剂-治疗患者为13%。
16 如何供应/贮存和处置
16.1 如何供应
AUSTEDO片可得到以下的规格和包装:
6 mg:圆形,紫色-涂层片,在一侧黑墨水打印有“SD”上面“6”。
60片瓶:NDC 68546-170-60.
9 mg:圆形,蓝色-涂层片,在一侧黑墨水打印有“SD”上面“9”.
60片瓶:NDC 68546-171-60.
12 mg:圆形,米色-涂层片,在一侧黑墨水打印有“SD”上面“12”.
60片瓶:NDC 68546-172-60.
16.2 贮存
贮存在25º C(77ºF);外出允许至15°C至30ºC(59°F至86ºF)[见USP控制室温]。避光和潮湿保护。
17 患者咨询资料
建议患者或护理人员阅读FDA-批准的患者说明书(用药指南)。
给药指导
建议患者与食物服用AUSTEDO。AUSTEDO片应被整吞和不要咀嚼,压碎或破碎[见剂量和给药方法(2.1)]。
抑郁和自杀的风险
忠告患者,他们的护理人员,和家庭AUSTEDO可能增加抑郁,恶化 抑郁,和自杀倾向的风险,和任何症状立即报告给卫生保健提供者[见禁忌证(4),警告和注意事项(5.2)]。
镇静和嗜睡的风险
忠告患者AUSTEDO可能致镇静和嗜睡和可能损害进行需要复杂运动和精神技能任务的能力。直至他们学习任何他们对稳定剂量AUSTEDO反应,患者应小心做需要他们警觉的活动,例如驾驶车辆或操作机械[见警告和注意事项(5.6)]。
与酒精或其他镇静药物相互作用
忠告患者酒精或其他致想睡将恶化嗜睡药物[见药物相互作用(7.5)]。
同时药物
建议患者告知他们的医生自己服用的所有药物和因为一个潜在的相互作用在开始任何新药物前咨询他们的卫生保健提供者[见禁忌证(4)和药物相互作用(7.1,7.4,7.5,7.6)]
Austedo
Generic Name: deutetrabenazine
Dosage Form: tablet, coated
Medically reviewed by Drugs.com. Last updated on Jul 1, 2019.
WARNING: DEPRESSION AND SUICIDALITY IN PATIENTS WITH HUNTINGTON’S DISEASE
Austedo can increase the risk of depression and suicidal thoughts and behavior (suicidality) in patients with Huntington’s disease. Anyone considering the use of Austedo must balance the risks of depression and suicidality with the clinical need for treatment of chorea. Closely monitor patients for the emergence or worsening of depression, suicidality, or unusual changes in behavior. Patients, their caregivers, and families should be informed of the risk of depression and suicidality and should be instructed to report behaviors of concern promptly to the treating physician.
Particular caution should be exercised in treating patients with a history of depression or prior suicide attempts or ideation, which are increased in frequency in Huntington’s disease. Austedo is contraindicated in patients who are suicidal, and in patients with untreated or inadequately treated depression [see Contraindications (4) and Warnings and Precautions (5.1)].
Austedo® is indicated for the treatment of:
chorea associated with Huntington’s disease [see Clinical Studies (14.1)]tardive dyskinesia in adults [see Clinical Studies (14.2)]Austedo Dosage and AdministrationDosing InformationThe dose of Austedo is determined individually for each patient based on reduction of chorea or tardive dyskinesia and tolerability. When first prescribed to patients who are not being switched from tetrabenazine (a related VMAT2 inhibitor), the recommended starting dose of Austedo is 6 mg administered orally once daily for patients with Huntington’s disease and 12 mg per day (6 mg twice daily) for patients with tardive dyskinesia.
The dose of Austedo may be increased at weekly intervals in increments of 6 mg per day to a maximum recommended daily dosage of 48 mg.Administer total daily dosages of 12 mg or above in two divided doses.Administer Austedo with food [see Clinical Pharmacology (12.3)].Swallow Austedo whole. Do not chew, crush, or break tablets.For patients at risk for QT prolongation, assess the QT interval before and after increasing total Austedo dosage above 24 mg per day [see Warnings and Precautions (5.3) and Drug Interactions (7.2)].Switching Patients from Tetrabenazine (XENAZINE®) to AustedoDiscontinue tetrabenazine (XENAZINE®) and initiate Austedo the following day. The recommended initial dosing regimen of Austedo in patients switching from tetrabenazine (XENAZINE®) to Austedo is shown in Table 1.
Table 1: Recommended Initial Dosing Regimen when Switching from Tetrabenazine (XENAZINE®) to Austedo |
|
Current tetrabenazine |
Initial regimen of |
12.5 mg |
6 mg once daily |
25 mg |
6 mg twice daily |
37.5 mg |
9 mg twice daily |
50 mg |
12 mg twice daily |
62.5 mg |
15 mg twice daily |
75 mg |
18 mg twice daily |
87.5 mg |
21 mg twice daily |
100 mg |
24 mg twice daily |
After patients are switched to Austedo, the dose may be adjusted at weekly intervals [see Dosage and Administration (2.1)].
Dosage Adjustment with Strong CYP2D6 InhibitorsIn patients receiving strong CYP2D6 inhibitors (e.g., quinidine, antidepressants such as paroxetine, fluoxetine, and bupropion), the total daily dosage of Austedo should not exceed 36 mg (maximum single dose of 18 mg) [see Drug Interactions (7.1) and Clinical Pharmacology (12.3)].
Dosage Adjustment in Poor CYP2D6 MetabolizersIn patients who are poor CYP2D6 metabolizers, the total daily dosage of Austedo should not exceed 36 mg (maximum single dose of 18 mg) [see Use in Specific Populations (8.7)].
Discontinuation and Interruption of TreatmentTreatment with Austedo can be discontinued without tapering. Following treatment interruption of greater than one week, Austedo therapy should be re-titrated when resumed. For treatment interruption of less than one week, treatment can be resumed at the previous maintenance dose without titration.
Dosage Forms and StrengthsAustedo tablets are available in the following strengths:
The 6 mg tablets are round, purple-coated tablets, with “SD” over “6” printed in black ink on one side.The 9 mg tablets are round, blue-coated tablets, with “SD” over “9” printed in black ink on one side.The 12 mg tablets are round, beige-coated tablets, with “SD” over “12” printed in black ink on one side.ContraindicationsAustedo is contraindicated in patients:
With Huntington’s disease who are suicidal, or have untreated or inadequately treated depression [see Warnings and Precautions (5.1)].With hepatic impairment [see Use in Specific Populations (8.6), Clinical Pharmacology (12.3)].Taking reserpine. At least 20 days should elapse after stopping reserpine before starting Austedo [see Drug Interactions (7.3)].Taking monoamine oxidase inhibitors (MAOIs). Austedo should not be used in combination with an MAOI, or within 14 days of discontinuing therapy with an MAOI [see Drug Interactions (7.4)].Taking tetrabenazine (XENAZINE®) or valbenazine [see Drug Interactions (7.7)].Warnings and PrecautionsDepression and Suicidality in Patients with Huntington’s DiseasePatients with Huntington’s disease are at increased risk for depression, and suicidal ideation or behaviors (suicidality). Austedo may increase the risk for suicidality in patients with Huntington’s disease.
In a 12-week, double-blind, placebo-controlled trial, suicidal ideation was reported by 2% of patients treated with Austedo, compared to no patients on placebo; no suicide attempts and no completed suicides were reported. Depression was reported by 4% of patients treated with Austedo.
When considering the use of Austedo, the risk of suicidality should be balanced against the need for treatment of chorea. All patients treated with Austedo should be observed for new or worsening depression or suicidality. If depression or suicidality does not resolve, consider discontinuing treatment with Austedo.
Patients, their caregivers, and families should be informed of the risks of depression, worsening depression, and suicidality associated with Austedo, and should be instructed to report behaviors of concern promptly to the treating physician. Patients with Huntington’s disease who express suicidal ideation should be evaluated immediately.
Clinical Worsening and Adverse Events in Patients with Huntington’s DiseaseHuntington’s disease is a progressive disorder characterized by changes in mood, cognition, chorea, rigidity, and functional capacity over time. VMAT2 inhibitors, including Austedo, may cause a worsening in mood, cognition, rigidity, and functional capacity.
Prescribers should periodically re-evaluate the need for Austedo in their patients by assessing the effect on chorea and possible adverse effects, including sedation/somnolence, depression and suicidality, parkinsonism, akathisia, restlessness, and cognitive decline. It may be difficult to distinguish between adverse reactions and progression of the underlying disease; decreasing the dose or stopping the drug may help the clinician to distinguish between the two possibilities. In some patients, the underlying chorea itself may improve over time, decreasing the need for Austedo.
QTc ProlongationTetrabenazine, a closely related VMAT2 inhibitor, causes an increase (about 8 msec) in the corrected QT (QTc) interval.
A clinically relevant QT prolongation may occur in some patients treated with Austedo who are CYP2D6 poor metabolizers or are co-administered a strong CYP2D6 inhibitor [see Clinical Pharmacology (12.2, 12.3)].
For patients who are CYP2D6 poor metabolizers or are taking a strong CYP2D6 inhibitor, dose reduction may be necessary [see Dosage and Administration (2.3, 2.4)]. The use of Austedo in combination with other drugs that are known to prolong QTc may result in clinically significant QT prolongations [see Drug Interactions (7.2)].
For patients requiring Austedo doses greater than 24 mg per day who are using Austedo with other drugs known to prolong QTc, assess the QTc interval before and after increasing the dose of Austedo or other medications that are known to prolong QTc.
Austedo should also be avoided in patients with congenital long QT syndrome and in patients with a history of cardiac arrhythmias. Certain circumstances may increase the risk of the occurrence of torsade de pointes and/or sudden death in association with the use of drugs that prolong the QTc interval, including (1) bradycardia; (2) hypokalemia or hypomagnesemia; (3) concomitant use of other drugs that prolong the QTc interval; and (4) presence of congenital prolongation of the QT interval.
Neuroleptic Malignant Syndrome (NMS)A potentially fatal symptom complex sometimes referred to as Neuroleptic Malignant Syndrome (NMS) has been reported in association with drugs that reduce dopaminergic transmission. While NMS has not been observed in patients receiving Austedo, it has been observed in patients receiving tetrabenazine (a closely related VMAT2 inhibitor). Clinicians should be alerted to the signs and symptoms associated with NMS. Clinical manifestations of NMS are hyperpyrexia, muscle rigidity, altered mental status, and evidence of autonomic instability (irregular pulse or blood pressure, tachycardia, diaphoresis, and cardiac dysrhythmia). Additional signs may include elevated creatinine phosphokinase, myoglobinuria, rhabdomyolysis, and acute renal failure. The diagnosis of NMS can be complicated; other serious medical illness (e.g., pneumonia, systemic infection) and untreated or inadequately treated extrapyramidal disorders can present with similar signs and symptoms. Other important considerations in the differential diagnosis include central anticholinergic toxicity, heat stroke, drug fever, and primary central nervous system pathology.
The management of NMS should include (1) immediate discontinuation of Austedo; (2) intensive symptomatic treatment and medical monitoring; and (3) treatment of any concomitant serious medical problems for which specific treatments are available. There is no general agreement about specific pharmacological treatment regimens for NMS.
Recurrence of NMS has been reported with resumption of drug therapy. If treatment with Austedo is needed after recovery from NMS, patients should be monitored for signs of recurrence.
Akathisia, Agitation, and RestlessnessAustedo may increase the risk of akathisia, agitation, and restlessness in patients with Huntington’s disease and tardive dyskinesia.
In a 12-week, double-blind, placebo-controlled trial in Huntington’s disease patients, akathisia, agitation, or restlessness was reported by 4% of patients treated with Austedo, compared to 2% of patients on placebo; in patients with tardive dyskinesia, 2% of patients treated with Austedo and 1% of patients on placebo experienced these events.
Patients receiving Austedo should be monitored for signs and symptoms of restlessness and agitation, as these may be indicators of developing akathisia. If a patient develops akathisia during treatment with Austedo, the Austedo dose should be reduced; some patients may require discontinuation of therapy.
ParkinsonismAustedo may cause parkinsonism in patients with Huntington’s disease or tardive dyskinesia. Parkinsonism has also been observed with other VMAT2 inhibitors.
Because rigidity can develop as part of the underlying disease process in Huntington’s disease, it may be difficult to distinguish between potential drug-induced parkinsonism and progression of underlying Huntington’s disease. Drug-induced parkinsonism has the potential to cause more functional disability than untreated chorea for some patients with Huntington’s disease.
Postmarketing cases of parkinsonism in patients treated with Austedo for tardive dyskinesia have been reported. Signs and symptoms in reported cases have included bradykinesia, gait disturbances, which led to falls in some cases, and the emergence or worsening of tremor. In most cases, the development of parkinsonism occurred within the first two weeks after starting or increasing the dose of Austedo. In cases in which follow-up clinical information was available, parkinsonism was reported to resolve following discontinuation of Austedo therapy.
If a patient develops parkinsonism during treatment with Austedo, the Austedo dose should be reduced; some patients may require discontinuation of therapy.
Sedation and SomnolenceSedation is a common dose-limiting adverse reaction of Austedo. In a 12-week, double-blind, placebo-controlled trial examining patients with Huntington’s disease, 11% of Austedo-treated patients reported somnolence compared with 4% of patients on placebo and 9% of Austedo-treated patients reported fatigue compared with 4% of placebo-treated patients.
Patients should not perform activities requiring mental alertness to maintain the safety of themselves or others, such as operating a motor vehicle or operating hazardous machinery, until they are on a maintenance dose of Austedo and know how the drug affects them.
HyperprolactinemiaSerum prolactin levels were not evaluated in the Austedo development program. Tetrabenazine, a closely related VMAT2 inhibitor, elevates serum prolactin concentrations in humans. Following administration of 25 mg of tetrabenazine to healthy volunteers, peak plasma prolactin levels increased 4- to 5-fold.
Tissue culture experiments indicate that approximately one-third of human breast cancers are prolactin-dependent in vitro, a factor of potential importance if Austedo is being considered for a patient with previously detected breast cancer. Although amenorrhea, galactorrhea, gynecomastia, and impotence can be caused by elevated serum prolactin concentrations, the clinical significance of elevated serum prolactin concentrations for most patients is unknown.
Chronic increase in serum prolactin levels (although not evaluated in the Austedo or tetrabenazine development programs) has been associated with low levels of estrogen and increased risk of osteoporosis. If there is a clinical suspicion of symptomatic hyperprolactinemia, appropriate laboratory testing should be done and consideration should be given to discontinuation of Austedo.
Binding to Melanin-Containing TissuesSince deutetrabenazine or its metabolites bind to melanin-containing tissues, it could accumulate in these tissues over time. This raises the possibility that Austedo may cause toxicity in these tissues after extended use. Neither ophthalmologic nor microscopic examination of the eye has been conducted in the chronic toxicity studies in a pigmented species such as dogs. Ophthalmologic monitoring in humans was inadequate to exclude the possibility of injury occurring after long-term exposure.
The clinical relevance of deutetrabenazine’s binding to melanin-containing tissues is unknown. Although there are no specific recommendations for periodic ophthalmologic monitoring, prescribers should be aware of the possibility of long-term ophthalmologic effects [see Clinical Pharmacology (12.2)].
Adverse ReactionsThe following serious adverse reactions are discussed in greater detail in other sections of the labeling:
Depression and Suicidality in Patients with Huntington’s disease [see Warnings and Precautions (5.1)]QTc Prolongation [see Warnings and Precautions (5.3)]Neuroleptic Malignant Syndrome (NMS) [see Warnings and Precautions (5.4)]Akathisia, Agitation, and Restlessness [see Warnings and Precautions (5.5)]Parkinsonism [see Warnings and Precautions (5.6)]Sedation and Somnolence [see Warnings and Precautions (5.7)]Hyperprolactinemia [see Warnings and Precautions (5.8)]Binding to Melanin-Containing Tissues [see Warnings and Precautions (5.9)]Clinical Trials ExperienceBecause 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 practice.
Patients with Huntington’s Disease
Study 1 was a randomized, 12-week, placebo-controlled study in patients with chorea associated with Huntington’s disease. A total of 45 patients received Austedo, and 45 patients received placebo. Patients ranged in age between 23 and 74 years (mean 54 years); 56% were male, and 92% were Caucasian. The most common adverse reactions occurring in greater than 8% of Austedo-treated patients were somnolence, diarrhea, dry mouth, and fatigue. Adverse reactions occurring in 4% or more of patients treated with Austedo, and with a greater incidence than in patients on placebo, are summarized in Table 2.
Table 2: Adverse Reactions in Patients with Huntington’s Disease (Study 1) Experienced by at Least 4% of Patients on Austedo and with a Greater Incidence than on Placebo |
||
Adverse Reaction |
Austedo |
Placebo |
Somnolence |
11 |
4 |
Diarrhea |
9 |
0 |
Dry mouth |
9 |
7 |
Fatigue |
9 |
4 |
Urinary tract infection |
7 |
2 |
Insomnia |
7 |
4 |
Anxiety |
4 |
2 |
Constipation |
4 |
2 |
Contusion |
4 |
2 |
One or more adverse reactions resulted in a reduction of the dose of study medication in 7% of patients in Study 1. The most common adverse reaction resulting in dose reduction in patients receiving Austedo was dizziness (4%).
Agitation led to discontinuation in 2% of patients treated with Austedo in Study 1.
Patients with Tardive Dyskinesia
The data described below reflect 410 tardive dyskinesia patients participating in clinical trials. Austedo was studied primarily in two 12-week, placebo-controlled trials (fixed dose, dose escalation). The population was 18 to 80 years of age, and had tardive dyskinesia and had concurrent diagnoses of mood disorder (33%) or schizophrenia/schizoaffective disorder (63%). In these studies, Austedo was administered in doses ranging from 12-48 mg per day. All patients continued on previous stable regimens of antipsychotics; 71% and 14% respective atypical and typical antipsychotic medications at study entry.
The most common adverse reactions occurring in greater than 3% of Austedo-treated patients and greater than placebo were nasopharyngitis and insomnia. The adverse reactions occurring in >2% or more patients treated with Austedo (12-48 mg per day) and greater than in placebo patients in two double-blind, placebo-controlled studies in patients with tardive dyskinesia (Study 1 and Study 2) are summarized in Table 3.
Table 3: Adverse Reactions in 2 Placebo-Controlled Tardive Dyskinesia Studies (Study 1 and Study 2) of 12-week Treatment on Austedo Reported in at Least 2% of Patients and Greater than Placebo |
||
Preferred Term |
Austedo |
Placebo |
Nasopharyngitis |
4 |
2 |
Insomnia |
4 |
1 |
Depression/ Dysthymic disorder |
2 |
1 |
Akathisia/Agitation/Restlessness |
2 |
1 |
One or more adverse reactions resulted in a reduction of the dose of study medication in 4% of Austedo-treated patients and in 2% of placebo-treated patients.
Drug InteractionsStrong CYP2D6 InhibitorsA reduction in Austedo dose may be necessary when adding a strong CYP2D6 inhibitor in patients maintained on a stable dose of Austedo. Concomitant use of strong CYP2D6 inhibitors (e.g., paroxetine, fluoxetine, quinidine, bupropion) has been shown to increase the systemic exposure to the active dihydro-metabolites of deutetrabenazine by approximately 3-fold. The daily dose of Austedo should not exceed 36 mg per day, and the maximum single dose of Austedo should not exceed 18 mg in patients taking strong CYP2D6 inhibitors [see Dosage and Administration (2.3) and Clinical Pharmacology (12.3)].
Drugs that Cause QTc ProlongationTetrabenazine, a closely related VMAT2 inhibitor, may cause an increase in the corrected QT (QTc) interval. Clinically relevant QT prolongation may also occur with Austedo [see Warnings and Precautions (5.3), Clinical Pharmacology (12.2)].
For patients requiring Austedo doses above 24 mg per day, who are using Austedo in combination with other drugs known to prolong QTc, assess the QTc interval before and after increasing the dose of Austedo or other medications that are known to prolong QTc. Drugs known to prolong QTc include antipsychotic medications (e.g., chlorpromazine, haloperidol, thioridazine, ziprasidone), antibiotics (e.g., moxifloxacin), Class 1A (e.g., quinidine, procainamide), and Class III (e.g., amiodarone, sotalol) antiarrhythmic medications.
ReserpineReserpine binds irreversibly to VMAT2 and the duration of its effect is several days. Prescribers should wait for chorea or dyskinesia to reemerge before administering Austedo to help reduce the risk of overdosage and major depletion of serotonin and norepinephrine in the central nervous system. At least 20 days should elapse after stopping reserpine before starting Austedo. Austedo and reserpine should not be used concomitantly [see Contraindications (4)].
Monoamine Oxidase Inhibitors (MAOIs)Austedo is contraindicated in patients taking MAOIs. Austedo should not be used in combination with an MAOI, or within 14 days of discontinuing therapy with an MAOI [see Contraindications (4)].
Neuroleptic DrugsThe risk of parkinsonism, NMS, and akathisia may be increased by concomitant use of Austedo and dopamine antagonists or antipsychotics.
Alcohol or Other Sedating DrugsConcomitant use of alcohol or other sedating drugs may have additive effects and worsen sedation and somnolence [see Warnings and Precautions (5.7)].
Concomitant Tetrabenazine or ValbenazineAustedo is contraindicated in patients currently taking tetrabenazine or valbenazine. Austedo may be initiated the day following discontinuation of tetrabenazine [see Dosage and Administration (2.2)].
USE IN SPECIFIC POPULATIONSPregnancyRisk Summary
There are no adequate data on the developmental risk associated with the use of Austedo in pregnant women. Administration of deutetrabenazine to rats during organogenesis produced no clear adverse effect on embryofetal development. However, administration of tetrabenazine to rats throughout pregnancy and lactation resulted in an increase in stillbirths and postnatal offspring mortality [see Data].
In the U.S. general population, the estimated background risk of major birth defects and miscarriage in clinically recognized pregnancies is 2-4% and 15-20%, respectively. The background risk of major birth defects and miscarriage for the indicated population is unknown.
Data
Animal Data
Oral administration of deutetrabenazine (5, 10, or 30 mg/kg/day) or tetrabenazine (30 mg/kg/day) to pregnant rats during organogenesis had no clear effect on embryofetal development. The highest dose tested was 6 times the maximum recommended human dose of 48 mg/day, on a body surface area (mg/m2) basis.
The effects of deutetrabenazine when administered during organogenesis to rabbits or during pregnancy and lactation to rats have not been assessed.
Tetrabenazine had no effects on embryofetal development when administered to pregnant rabbits during the period of organogenesis at oral doses up to 60 mg/kg/day. When tetrabenazine was administered to female rats (doses of 5, 15, and 30 mg/kg/day) from the beginning of organogenesis through the lactation period, an increase in stillbirths and offspring postnatal mortality was observed at 15 and 30 mg/kg/day, and delayed pup maturation was observed at all doses.
LactationRisk Summary
There are no data on the presence of deutetrabenazine or its metabolites in human milk, the effects on the breastfed infant, or the effects of the drug on milk production.
The developmental and health benefits of breastfeeding should be considered along with the mother’s clinical need for Austedo and any potential adverse effects on the breastfed infant from Austedo or from the underlying maternal condition.
Pediatric UseSafety and effectiveness in pediatric patients have not been established.
Geriatric UseClinical studies of Austedo did not include sufficient numbers of subjects aged 65 and over to determine whether they respond differently from younger subjects. Other reported clinical experience has not identified differences in responses between the elderly and younger patients. In general, dose selection for an elderly patient should be cautious, usually starting at the low end of the dosing range, reflecting the greater frequency of hepatic, renal, and cardiac dysfunction, and of concomitant disease or other drug therapy.
Hepatic ImpairmentThe effect of hepatic impairment on the pharmacokinetics of deutetrabenazine and its primary metabolites has not been studied; however, in a clinical study conducted with tetrabenazine, a closely related VMAT2 inhibitor, there was a large increase in exposure to tetrabenazine and its active metabolites in patients with hepatic impairment. The clinical significance of this increased exposure has not been assessed, but because of concerns for a greater risk for serious adverse reactions, the use of Austedo in patients with hepatic impairment is contraindicated [see Contraindications (4), Clinical Pharmacology (12.3)].
Poor CYP2D6 MetabolizersAlthough the pharmacokinetics of deutetrabenazine and its metabolites have not been systematically evaluated in patients who do not express the drug metabolizing enzyme, it is likely that the exposure to α-HTBZ and β-HTBZ would be increased similarly to taking a strong CYP2D6 inhibitor (approximately 3-fold). In patients who are CYP2D6 poor metabolizers, the daily dose of Austedo should not exceed 36 mg (maximum single dose of 18 mg) [see Dosage and Administration (2.4) and Clinical Pharmacology (12.3)].
OverdosageOverdoses ranging from 100 mg to 1 g have been reported in the literature with tetrabenazine, a closely related VMAT2 inhibitor. The following adverse reactions occurred with overdosing: acute dystonia, oculogyric crisis, nausea and vomiting, sweating, sedation, hypotension, confusion, diarrhea, hallucinations, rubor, and tremor.
Treatment should consist of those general measures employed in the management of overdosage with any central nervous system-active drug. General supportive and symptomatic measures are recommended. Cardiac rhythm and vital signs should be monitored. In managing overdosage, the possibility of multiple drug involvement should always be considered. The physician should consider contacting a poison control center on the treatment of any overdose. Telephone numbers for certified poison control centers are listed on the American Association of Poison Control Centers website www.aapcc.org.
Austedo DescriptionAustedo (deutetrabenazine) is a vesicular monoamine transporter 2 (VMAT2) inhibitor for oral administration. The molecular weight of deutetrabenazine is 323.46; the pKa is 6.31. Deutetrabenazine is a hexahydro-dimethoxybenzoquinolizine derivative and has the following chemical name: (RR, SS)-1, 3, 4, 6, 7, 11b-hexahydro-9, 10-di(methoxy-d3)-3-(2-methylpropyl)-2H-benzo[a]quinolizin-2-one.
The molecular formula for deutetrabenazine is C19H21D6NO3. Deutetrabenazine is a racemic mixture containing the following structures:
Deutetrabenazine is a white to slightly yellow crystalline powder that is sparingly soluble in water and soluble in ethanol.
Austedo tablets contain 6 mg, 9 mg, or 12 mg deutetrabenazine, and the following inactive ingredients: ammonium hydroxide, black iron oxide, n-butyl alcohol, butylated hydroxyanisole, butylated hydroxytoluene, magnesium stearate, mannitol, microcrystalline cellulose, polyethylene glycol, polyethylene oxide, polysorbate 80, polyvinyl alcohol, povidone, propylene glycol, shellac, talc, titanium dioxide, and FD&C blue #2 lake. The 6 mg tablets also contain FD&C red #40 lake. The 12 mg tablets also contain FD&C yellow #6 lake.
Austedo - Clinical PharmacologyMechanism of ActionThe precise mechanism by which deutetrabenazine exerts its effects in the treatment of tardive dyskinesia and chorea in patients with Huntington’s disease is unknown but is believed to be related to its effect as a reversible depletor of monoamines (such as dopamine, serotonin, norepinephrine, and histamine) from nerve terminals. The major circulating metabolites (α-dihydrotetrabenazine [HTBZ] and β-HTBZ) of deutetrabenazine, are reversible inhibitors of VMAT2, resulting in decreased uptake of monoamines into synaptic vesicles and depletion of monoamine stores.
PharmacodynamicsCardiac Electrophysiology
The effect of a single 12-mg or 24-mg dose of Austedo on the QT interval was studied in a randomized, double-blind, placebo-controlled crossover study in healthy male and female subjects with moxifloxacin as a positive control. At 24 mg, Austedo caused an approximately 4.5 msec mean increase in QTc (90% CI: 2.4, 6.5 msec). Effects at higher exposures to Austedo or its metabolites have not been evaluated.
Melanin Binding
Deutetrabenazine or its metabolites bind to melanin-containing tissues (i.e., eye, skin, fur) in pigmented rats. After a single oral dose of radiolabeled deutetrabenazine, radioactivity was still detected in eye and fur at 35 days following dosing [see Warnings and Precautions (5.9)].
PharmacokineticsAfter oral dosing up to 25 mg, plasma concentrations of deutetrabenazine are generally below the limit of detection because of the extensive hepatic metabolism of deutetrabenazine to the active deuterated dihydro metabolites (HTBZ), α-HTBZ and β-HTBZ. Linear dose dependence of Cmax and AUC was observed for the active metabolites following single or multiple doses of deutetrabenazine (6 mg to 24 mg and 7.5 mg twice daily to 22.5 mg twice daily).
Absorption
Following oral administration of deutetrabenazine, the extent of absorption is at least 80%.
Plasma concentrations of deutetrabenazine are generally below the limit of detection after oral dosing. Peak plasma concentrations (Cmax) of deuterated α-HTBZ and β-HTBZ are reached within 3 to 4 hours after dosing.
Effect of Food
The effects of food on the bioavailability of Austedo were studied in subjects administered a single dose with and without food. Food had no effect on the area under the plasma concentration-time curve (AUC) of α-HTBZ or β-HTBZ, although Cmax was increased by approximately 50% in the presence of food [see Dosage and Administration (2.1)].
Distribution
The median volume of distribution (Vc/F) of the α-HTBZ, and the β-HTBZ metabolites of Austedo are approximately 500 L and 730 L, respectively.
Results of PET-scan studies in humans show that following intravenous injection of 11C-labeled tetrabenazine or α-HTBZ, radioactivity is rapidly distributed to the brain, with the highest binding in the striatum and lowest binding in the cortex.
The in vitro protein binding of tetrabenazine, α-HTBZ, and β-HTBZ was examined in human plasma for concentrations ranging from 50 to 200 ng/mL. Tetrabenazine binding ranged from 82% to 85%, α-HTBZ binding ranged from 60% to 68%, and β-HTBZ binding ranged from 59% to 63%.
Elimination
Austedo is primarily renally eliminated in the form of metabolites.
The half-life of total (α+β)-HTBZ from deutetrabenazine is approximately 9 to 10 hours.
The median clearance values (CL/F) of the α-HTBZ, and the β-HTBZ metabolites of Austedo are approximately 47 L/hour and 70 L/hour, respectively, in the Huntington’s disease patient population.
Metabolism
In vitro experiments in human liver microsomes demonstrate that deutetrabenazine is extensively biotransformed, mainly by carbonyl reductase, to its major active metabolites, α-HTBZ and β-HTBZ, which are subsequently metabolized primarily by CYP2D6, with minor contributions of CYP1A2 and CYP3A4/5, to form several minor metabolites.
Excretion
In a mass balance study in 6 healthy subjects, 75% to 86% of the deutetrabenazine dose was excreted in the urine, and fecal recovery accounted for 8% to 11% of the dose. Urinary excretion of the α-HTBZ and β-HTBZ metabolites from deutetrabenazine each accounted for less than 10% of the administered dose. Sulfate and glucuronide conjugates of the α-HTBZ and β-HTBZ metabolites of deutetrabenazine, as well as products of oxidative metabolism, accounted for the majority of metabolites in the urine.
Specific Populations
Male and Female Patients
There is no apparent effect of gender on the pharmacokinetics of α-HTBZ and β‑HTBZ of deutetrabenazine.
Patients with Renal Impairment
No clinical studies have been conducted to assess the effect of renal impairment on the PK of Austedo.
Patients with Hepatic Impairment
The effect of hepatic impairment on the pharmacokinetics of deutetrabenazine and its primary metabolites has not been studied. However, in a clinical study conducted to assess the effect of hepatic impairment on the pharmacokinetics of tetrabenazine, a closely related VMAT2 inhibitor, the exposure to α-HTBZ and β-HTBZ was up to 40% greater in patients with hepatic impairment, and the mean tetrabenazine Cmax in patients with hepatic impairment was up to 190-fold higher than in healthy subjects [see Contraindications (4), Use in Specific Populations (8.6)].
Poor CYP2D6 Metabolizers
Although the pharmacokinetics of deutetrabenazine and its metabolites have not been systematically evaluated in patients who do not express the drug metabolizing enzyme CYP2D6, it is likely that the exposure to α-HTBZ and β-HTBZ would be increased similarly to taking strong CYP2D6 inhibitors (approximately 3-fold) [see Dosage and Administration (2.4), Drug Interactions (7.1)].
Drug Interaction Studies
Deutetrabenazine, α-HTBZ, and β-HTBZ have not been evaluated in in vitro studies for induction or inhibition of CYP enzymes or interaction with P-glycoprotein. The results of in vitro studies of tetrabenazine do not suggest that tetrabenazine or its α-HTBZ or β-HTBZ metabolites are likely to result in clinically significant inhibition of CYP2D6, CYP1A2, CYP2B6, CYP2C8, CYP2C9, CYP2C19, CYP2E1, or CYP3A. In vitro studies suggest that neither tetrabenazine nor its α-HTBZ or β-HTBZ metabolites are likely to result in clinically significant induction of CYP1A2, CYP3A4, CYP2B6, CYP2C8, CYP2C9, or CYP2C19. Neither tetrabenazine nor its α-HTBZ or β-HTBZ metabolites are likely to be a substrate or inhibitor of P-glycoprotein at clinically relevant concentrations in vivo.
The deutetrabenazine metabolites, 2-methylpropanoic acid of β-HTBZ (M1) and monohydroxy tetrabenazine (M4), have been evaluated in a panel of in vitro drug-drug interaction studies; the results indicate that M1/M4 are not expected to cause clinically relevant drug interactions.
CYP2D6 Inhibitors
In vitro studies indicate that the α-HTBZ and β-HTBZ metabolites of deutetrabenazine are substrates for CYP2D6. The effect of CYP2D6 inhibition on the pharmacokinetics of deutetrabenazine and its metabolites was studied in 24 healthy subjects following a single 22.5 mg dose of deutetrabenazine given after 8 days of administration of the strong CYP2D6 inhibitor paroxetine 20 mg daily. In the presence of paroxetine, systemic exposure (AUCinf) of α-HTBZ was 1.9-fold higher and β-HTBZ was 6.5-fold higher, resulting in approximately 3-fold increase in AUCinf for total (α+β)-HTBZ. Paroxetine decreased the clearance of α-HTBZ and β-HTBZ metabolites of Austedo with corresponding increases in mean half-life of approximately 1.5-fold and 2.7-fold, respectively. In the presence of paroxetine, Cmax of α-HTBZ and β-HTBZ were 1.2-fold and 2.2-fold higher, respectively.
The effect of moderate or weak CYP2D6 inhibitors such as duloxetine, terbinafine, amiodarone, or sertraline on the exposure of deutetrabenazine and its metabolites has not been evaluated.
Digoxin
Austedo was not evaluated for interaction with digoxin. Digoxin is a substrate for P-glycoprotein. A study in healthy subjects showed that tetrabenazine (25 mg twice daily for 3 days) did not affect the bioavailability of digoxin, suggesting that at this dose, tetrabenazine does not affect P‑glycoprotein in the intestinal tract. In vitro studies also do not suggest that tetrabenazine or its metabolites are P-glycoprotein inhibitors.
Nonclinical ToxicologyCarcinogenesis, Mutagenesis, Impairment of FertilityCarcinogenesis
No carcinogenicity studies were performed with deutetrabenazine.
No increase in tumors was observed in p53+/– transgenic mice treated orally with tetrabenazine at doses of 0, 5, 15, and 30 mg/kg/day for 26 weeks.
Mutagenesis
Deutetrabenazine and its deuterated α-HTBZ and β-HTBZ metabolites were negative in in vitro (bacterial reverse mutation and chromosome aberration in human peripheral blood lymphocytes) assays in the presence or absence of metabolic activation and in the in vivo micronucleus assay in mice.
Impairment of Fertility
The effects of deutetrabenazine on fertility have not been evaluated. Oral administration of deutetrabenazine (doses of 5, 10, or 30 mg/kg/day) to female rats for 3 months resulted in estrous cycle disruption at all doses; the lowest dose tested was similar to the maximum recommended human dose (48 mg/day) on a body surface area (mg/m2) basis.
Oral administration of tetrabenazine (doses of 5, 15, or 30 mg/kg/day) to female rats prior to and throughout mating, and continuing through day 7 of gestation, resulted in disrupted estrous cyclicity at doses greater than 5 mg/kg/day. No effects on mating and fertility indices or sperm parameters (motility, count, density) were observed when males were treated orally with tetrabenazine at doses of 5, 15 or 30 mg/kg/day prior to and throughout mating with untreated females.
Clinical StudiesChorea Associated with Huntington’s DiseaseDouble-Blind, Placebo-Controlled Study
The efficacy of Austedo as a treatment for chorea associated with Huntington's disease was established primarily in Study 1, a randomized, double-blind, placebo-controlled, multi-center trial conducted in 90 ambulatory patients with manifest chorea associated with Huntington’s disease. The diagnosis of Huntington’s disease was based on family history, neurological exam, and genetic testing. Treatment duration was 12 weeks, including an 8-week dose titration period and a 4-week maintenance period, followed by a 1-week washout. Patients were not blinded to discontinuation. Austedo was started at 6 mg per day and titrated upward, at weekly intervals, in 6 mg increments until satisfactory treatment of chorea was achieved, intolerable side effects occurred, or until a maximal dose of 48 mg per day was reached. The primary efficacy endpoint was the Total Maximal Chorea Score, an item of the Unified Huntington's Disease Rating Scale (UHDRS). On this scale, chorea is rated from 0 to 4 (with 0 representing no chorea) for 7 different parts of the body. The total score ranges from 0 to 28.
Of the 90 patients enrolled, 87 patients completed the study. The mean age was 54 (range 23 to 74). Patients were 56% male and 92% Caucasian. The mean dose after titration was 40 mg per day. Table 4 and Figure 1 summarize the effects of Austedo on chorea based on the Total Maximal Chorea Score. Total Maximal Chorea Scores for patients receiving Austedo improved by approximately 4.4 units from baseline to the maintenance period (average of Week 9 and Week 12), compared to approximately 1.9 units in the placebo group. The treatment effect of -2.5 units was statistically significant (p<0.0001). The Maintenance Endpoint is the mean of the Total Maximal Chorea Scores for the Week 9 and Week 12 visits. At the Week 13 follow-up visit (1 week after discontinuation of the study medication), the Total Maximal Chorea Scores of patients who had received Austedo returned to baseline (Figure 1).
Table 4: Change from Baseline to Maintenance Therapy in Total Maximal Chorea (TMC)a Score in Patients with Huntington’s Disease Treated with Austedo in Study 1 |
|||
Motor Endpoint |
Austedo |
Placebo |
p value |
Change in Total Chorea Scorea from Baseline to Maintenance Therapyb |
-4.4 |
-1.9 |
<0.0001 |
a TMC is a subscale of the Unified Huntington's Disease Rating Scale (UHDRS)
b Primary efficacy endpoint
Figure 1: Total Maximal Chorea Score Over Time in Study 1
Figure 2: Distribution of the Change in Total Maximal Chorea Scores in Study 1
Figure 2 shows the distribution of values for the change in Total Maximal Chorea Score in Study 1. Negative values indicate a reduction in chorea and positive numbers indicate an increase in chorea.
A patient-rated global impression of change assessed how patients rated their overall Huntington’s disease symptoms. Fifty-one percent of patients treated with Austedo rated their symptoms as “Much Improved” or “Very Much Improved” at the end of treatment, compared to 20% of placebo-treated patients.
In a physician-rated clinical global impression of change, 42% percent of patients treated with Austedo rated their symptoms as “Much Improved” or “Very Much Improved” at the end of treatment compared to 13% of placebo-treated patients.
Tardive DyskinesiaThe efficacy of Austedo in the treatment for tardive dyskinesia was established in two 12‑week, randomized, double-blind, placebo-controlled, multi-center trials conducted in 335 adult ambulatory patients with tardive dyskinesia caused by use of dopamine receptor antagonists. Patients had a history of using a dopamine receptor antagonist (antipsychotics, metoclopramide) for at least 3 months (or 1 month in patients 60 years of age and older). Concurrent diagnoses included schizophrenia/schizoaffective disorder (62%) and mood disorder (33%). With respect to concurrent antipsychotic use, 64% of patients were receiving atypical antipsychotics, 12% were receiving typical or combination antipsychotics, and 24% were not receiving antipsychotics.
The Abnormal Involuntary Movement Scale (AIMS) was the primary efficacy measure for the assessment of tardive dyskinesia severity. The AIMS is a 12-item scale; items 1 to 7 assess the severity of involuntary movements across body regions and these items were used in this study. Each of the 7 items was scored on a 0 to 4 scale, rated as: 0=not present; 1=minimal, may be extreme normal (abnormal movements occur infrequently and/or are difficult to detect); 2=mild (abnormal movements occur infrequently and are easy to detect); 3=moderate (abnormal movements occur frequently and are easy to detect) or 4 =severe (abnormal movements occur almost continuously and/or of extreme intensity). The AIMS total score (sum of items 1 to 7) could thus range from 0 to 28, with a decrease in score indicating improvement.
In Study 1, a 12-week, placebo-controlled, fixed-dose trial, adults with tardive dyskinesia were randomized 1:1:1:1 to 12 mg Austedo, 24 mg Austedo, 36 mg Austedo, or placebo. Treatment duration included a 4-week dose escalation period and an 8-week maintenance period followed by a 1-week washout. The dose of Austedo was started at 12 mg per day and increased at weekly intervals in 6 mg/day increments to a dose target of 12 mg, 24 mg or 36 mg per day. The population (n= 222) was 21 to 81 years old (mean 57 years), 48% male, and 79% Caucasian. In Study 1, the AIMS total score for patients receiving Austedo demonstrated statistically significant improvement, from baseline to Week 12, of 3.3 and 3.2 units for the 36 mg and 24 mg arms, respectively, compared with 1.4 units in placebo (Study 1 in Table 5). The improvements on the AIMS total score over the course of the study are displayed in Figure 3. Data did not suggest substantial differences in efficacy across various demographic groups. The treatment response rate distribution, based on magnitude of AIMS total score from baseline to week 12 is displayed in Figure 4.
The mean changes in the AIMS total score by visit are shown in Figure 3.
In Study 2, a 12-week, placebo-controlled, flexible-dose trial, adults with tardive dyskinesia (n=113) received daily doses of placebo or Austedo, starting at 12 mg per day with increases allowed in 6-mg increments at 1-week intervals until satisfactory control of dyskinesia was achieved, until intolerable side effects occurred, or until a maximal dose of 48 mg per day was reached. Treatment duration included a 6-week dose titration period and a 6-week maintenance period followed by a 1-week washout. The population was 25 to 75 years old (mean 55 years), 48% male, and 70% Caucasian. Patients were titrated to an optimal dose over 6 weeks. The average dose of Austedo after treatment was 38.3 mg per day. There was no evidence suggesting substantial differences in efficacy across various demographic groups. In Study 2, AIMS total score for patients receiving Austedo demonstrated statistically significant improvement by 3.0 units from baseline to endpoint (Week 12), compared with 1.6 units in the placebo group with a treatment effect of -1.4 units. Table 5 summarizes the effects of Austedo on tardive dyskinesia based on the AIMS.
Table 5: Improvement in AIMS Total Score in Patients Treated with Austedo in Study 1 and Study 2 |
|||||
Study |
Treatment Group |
Primary Efficacy Measure: AIMS Total Score |
|
||
Mean Baseline Score (SD) |
LS Mean Change from Baseline (SE) |
Treatment Effect (95% CI) |
|
||
Study 1 |
Austedo 36 mg* (n= 55) |
10.1 (3.21) |
-3.3 (0.42) |
-1.9 (-3.09, -0.79) |
|
Austedo 24 mg (n= 49) |
9.4 (2.93) |
-3.2 (0.45) |
-1.8 (-3.00, -0.63) |
|
|
Austedo 12 mg (n= 60) |
9.6 (2.40) |
-2.1 (0.42) |
-0.7 (-1.84, 0.42) |
|
|
Placebo (n= 58) |
9.5 (2.71) |
-1.4 (0.41) |
|
|
|
Study 2 |
Austedo (12-48 mg/day)* (n= 56) |
9.7 (4.14) |
-3.0 (0.45) |
-1.4 (-2.6, -0.2) |
|
Placebo (n= 57) |
9.6 (3.78) |
-1.6 (0.46) |
|
|
*Dose that was statistically significantly different from placebo after adjusting for multiplicity.
LS Mean = Least-squares mean; SD = Standard deviation; SE = Standard error; CI = 2-sided 95% confidence interval
Figure 3: Least Square Means of Change in AIMS Total Score from Baseline for Austedo Compared to Placebo (Study 1)
SE = Standard error
Figure 4: Percent of Patients with Specified Magnitude of AIMS Total Score Improvement at the End of Week 12 (Study 1)
How Supplied/Storage and HandlingHow Supplied
Austedo tablets are available in the following strengths and packages:
6 mg: round, purple-coated tablets, with “SD” over “6” printed in black ink on one side.
Bottles of 60 tablets: NDC 68546-170-60.
9 mg: round, blue-coated tablets, with “SD” over “9” printed in black ink on one side.
Bottles of 60 tablets: NDC 68546-171-60.
12 mg: round, beige-coated tablets, with “SD” over “12” printed in black ink on one side.
Bottles of 60 tablets: NDC 68546-172-60.
StorageStore at 25ºC (77ºF); excursions permitted to 15ºC to 30ºC (59ºF to 86ºF) [see USP Controlled Room Temperature]. Protect from light and moisture.
Patient Counseling InformationAdvise the patient or caregiver to read the FDA-approved patient labeling (Medication Guide).
Administration Instructions
Advise patients to take Austedo with food. Austedo tablets should be swallowed whole and not chewed, crushed, or broken [see Dosage and Administration (2.1)].
Risk of Depression and Suicide in Patients with Huntington’s Disease
Advise patients, their caregivers, and families that Austedo may increase the risk of depression, worsening depression, and suicidality, and to immediately report any symptoms to a healthcare provider [see Contraindications (4), Warnings and Precautions (5.2)].
Prolongation of the QTc Interval
Inform patients to consult their physician immediately if they feel faint, lose consciousness, or have heart palpitations [see Warnings and Precautions (5.3)]. Advise patients to inform physicians that they are taking Austedo before any new drug is taken.
Parkinsonism
Inform patients that Austedo may cause Parkinson-like symptoms, which could be severe. Advise patients to consult their healthcare provider if they experience slight shaking, body stiffness, trouble moving, trouble keeping their balance, or falls [see Warnings and Precautions (5.6)].
Risk of Sedation and Somnolence
Advise patients that Austedo may cause sedation and somnolence and may impair the ability to perform tasks that require complex motor and mental skills. Until they learn how they respond to a stable dose of Austedo, patients should be careful doing activities that require them to be alert, such as driving a car or operating machinery [see Warnings and Precautions (5.7)].
Interaction with Alcohol or Other Sedating Drugs
Advise patients that alcohol or other drugs that cause sleepiness will worsen somnolence [see Drug Interactions (7.6)].
Concomitant Medications
Advise patients to notify their physician of all medications they are taking and to consult with their healthcare provider before starting any new medications because of a potential for interactions [see Contraindications (4) and Drug Interactions (7.1, 7.5)].
Distributed by:
Teva Pharmaceuticals USA, Inc.
North Wales, PA 19454
AUS-003
U.S. Patent Nos: 8,524,733; 9,233,959; 9,296,739; 9,550,780
XENAZINE® is a trademark of Valeant Pharmaceuticals Luxembourg S.A.R.L.
MEDICATION GUIDE
MEDICATION GUIDE |
What is the most important information I should know about Austedo? Austedo can cause serious side effects in people with Huntington’s disease, including:depressionsuicidal thoughtssuicidal actionsDo not start taking Austedo if you have Huntington’s disease and are depressed (have untreated depression or depression that is not well controlled by medicine) or have suicidal thoughts.Pay close attention to any changes, especially sudden changes, in mood, behaviors, thoughts, or feelings. This is especially important when Austedo is started and when the dose is changed.Call your healthcare provider right away if you become depressed or have any of the following symptoms, especially if they are new, worse, or worry you: feel sad or have crying spellslose interest in seeing your friends or doing things you used to enjoysleep a lot more or a lot less than usualfeel unimportantfeel guiltyfeel hopeless or helplessfeel more irritable, angry, or aggressive than usualfeel more or less hungry than usual or notice a big change in your body weighthave trouble paying attentionfeel tired or sleepy all the timehave thoughts about hurting yourself or ending your life |
What is Austedo? Austedo is a prescription medicine that is used to treat: the involuntary movements (chorea) of Huntington’s disease. Austedo does not cure the cause of the involuntary movements, and it does not treat other symptoms of Huntington’s disease, such as problems with thinking or emotions.movements in the face, tongue, or other body parts that cannot be controlled (tardive dyskinesia).It is not known if Austedo is safe and effective in children. |
Who should not take Austedo? Do not take Austedo if you: have Huntington’s disease and are depressed or have thoughts of suicide. See “What is the most important information I should know about Austedo?”have liver problems.are taking a monoamine oxidase inhibitor (MAOI) medicine. Do not take an MAOI within 14 days after you stop taking Austedo. Do not start Austedo if you stopped taking an MAOI in the last 14 days. Ask your healthcare provider or pharmacist if you are not sure.are taking reserpine. Do not take medicines that contain reserpine (such as Serpalan and Renese-R) with Austedo. If your healthcare provider plans to switch you from taking reserpine to Austedo, you must wait at least 20 days after your last dose of reserpine before you start taking Austedo.are taking tetrabenazine (Xenazine). If your healthcare provider plans to switch you from tetrabenazine (Xenazine) to Austedo, take your first dose of Austedo on the day after your last dose of tetrabenazine (Xenazine).are taking valbenazine (Ingrezza). |
Before taking Austedo, tell your healthcare provider about all of your medical conditions, including if you: have emotional or mental problems (for example, depression, nervousness, anxiety, anger, agitation, psychosis, previous suicidal thoughts or suicide attempts).have liver disease.have an irregular heart rhythm or heartbeat (QT prolongation, cardiac arrhythmia) or a heart problem called congenital long QT syndrome.have low levels of potassium or magnesium in your blood (hypokalemia or hypomagnesemia).have breast cancer or a history of breast cancer.are pregnant or plan to become pregnant. It is not known if Austedo can harm your unborn baby.are breastfeeding or plan to breastfeed. It is not known if Austedo passes into breast milk.Tell your healthcare provider about all of the medicines you take, including prescription and over-the-counter medicines, vitamins, and herbal supplements. Taking Austedo with certain other medicines may cause side effects. Do not start any new medicines while taking Austedo without talking to your healthcare provider first. |
How should I take Austedo? Take Austedo exactly as your healthcare provider tells you to take it.Take Austedo by mouth and with food.Swallow Austedo tablets whole with water. Do not chew, crush, or break Austedo tablets before swallowing. If you cannot swallow Austedo tablets whole, tell your healthcare provider. You may need a different medicine.If your dose of Austedo is 12 mg or more each day, take Austedo tablets 2 times a day in equal doses with food.Your healthcare provider will increase your dose of Austedo each week for several weeks, until you and your healthcare provider find the right dose for you.Tell your healthcare provider if you stop taking Austedo for more than 1 week. Do not take another dose until you talk to your healthcare provider. |
What should I avoid while taking Austedo? Sleepiness (sedation) is a common side effect of Austedo. While taking Austedo, do not drive a car or operate dangerous machinery until you know how Austedo affects you. Drinking alcohol and taking other drugs that may also cause sleepiness while you are taking Austedo may increase any sleepiness caused by Austedo. |
What are the possible side effects of Austedo? Austedo can cause serious side effects, including: Depression and suicidal thoughts or actions in people with Huntington’s disease. See “What is the most important information I should know about Austedo?”Irregular heartbeat (QT prolongation). Austedo increases your chance of having certain changes in the electrical activity in your heart. These changes can lead to a dangerous abnormal heartbeat. Taking Austedo with certain medicines may increase this chance.If you are at risk of QT prolongation, your healthcare provider should check your heart before and after increasing your Austedo dose above 24 mg a day.Neuroleptic Malignant Syndrome (NMS). Call your healthcare provider right away and go to the nearest emergency room if you develop these signs and symptoms that do not have another obvious cause:high fever problems thinkingincreased sweatingstiff musclesvery fast or uneven heartbeatRestlessness. You may get a condition where you feel a strong urge to move. This is called akathisia.Parkinsonism. Symptoms of parkinsonism include: slight shaking, body stiffness, trouble moving, trouble keeping your balance, or falls.The most common side effects of Austedo in people with Huntington’s disease include: sleepiness (sedation)diarrheatirednessdry mouthThe most common side effects of Austedo in people with tardive dyskinesia include: inflammation of the nose and throat (nasopharyngitis)problems sleeping (insomnia)These are not all the possible side effects of Austedo. Call your doctor for medical advice about side effects. You may report side effects to FDA at 1-800-FDA-1088. |
How should I store Austedo? Store Austedo tablets at room temperature, between 68°F to 77°F (20°C to 25°C).Keep the bottle tightly closed to protect Austedo from light and moisture.Keep Austedo tablets and all medications out of reach of children. |
General information about the safe and effective use of Austedo. Medicines are sometimes prescribed for purposes other than those listed in a Medication Guide. Do not use Austedo for a condition for which it was not prescribed. Do not give Austedo to other people, even if they have the same symptoms you have. It may harm them. You can ask your pharmacist or healthcare provider for information about Austedo that is written for health professionals. |
What are the ingredients in Austedo? Active ingredient: deutetrabenazine Inactive ingredients: ammonium hydroxide, black iron oxide, n‑butyl alcohol, butylated hydroxyanisole, butylated hydroxytoluene, magnesium stearate, mannitol, microcrystalline cellulose, polyethylene glycol, polyethylene oxide, polysorbate 80, polyvinyl alcohol, povidone, propylene glycol, shellac, talc, titanium dioxide, and FD&C blue #2 lake. The 6 mg tablets also contain FD&C red #40 lake. The 12 mg tablets also contain FD&C yellow #6 lake.
Distributed by: AUSMG-003 For more information, go to www.Austedo.com or call 1-888-483-8279. |
This Medication Guide has been approved by the U.S. Food and Drug Administration. Revised: July 2019 |
NDC 68546-170-60
Austedo™ (deutetrabenazine) tablets
6 mg
60 tablets
Medication Guide Required: Each time Austedo is dispensed, give the patient a Medication Guide.
Package/Label Display Panel
NDC 68546-171-60
Austedo™ (deutetrabenazine) tablets
9 mg
60 tablets
Medication Guide Required: Each time Austedo is dispensed, give the patient a Medication Guide.
Package/Label Display Panel
NDC 68546-172-60
Austedo™ (deutetrabenazine) tablets
12 mg
60 tablets
Medication Guide Required: Each time Austedo is dispensed, give the patient a Medication Guide.
Austedo |
||||||||||||||||||||||||||||||||||||||||||
|
||||||||||||||||||||||||||||||||||||||||||
|
||||||||||||||||||||||||||||||||||||||||||
|
||||||||||||||||||||||||||||||||||||||||||
|
||||||||||||||||||||||||||||||||||||||||||
|
||||||||||||||||||||||||||||||||||||||||||
|
||||||||||||||||||||||||||||||||||||||||||
|
||||||||||||||||||||||||||||||||||||||||||
Austedo |
||||||||||||||||||||||||||||||||||||||||||
|
||||||||||||||||||||||||||||||||||||||||||
|
||||||||||||||||||||||||||||||||||||||||||
|
||||||||||||||||||||||||||||||||||||||||||
|
||||||||||||||||||||||||||||||||||||||||||
|
||||||||||||||||||||||||||||||||||||||||||
|
||||||||||||||||||||||||||||||||||||||||||
|
Austedo |
||||||||||||||||||||||||||||||||||||||||||
|
||||||||||||||||||||||||||||||||||||||||||
|
||||||||||||||||||||||||||||||||||||||||||
|
||||||||||||||||||||||||||||||||||||||||||
|
||||||||||||||||||||||||||||||||||||||||||
|
||||||||||||||||||||||||||||||||||||||||||
|
||||||||||||||||||||||||||||||||||||||||||
|
||||||||||||||||||||||||||||||||||||||||||
Labeler - Teva Neuroscience, Inc. (009906397) |
Teva Neuroscience, Inc.