Treatment outcomes with telaprevir-based therapy for HIV/hepatitis C coinfected patients are comparable with hepatitis C monoinfected patients

C. O’Neil, J. Pang, Samuel S. Lee, M. Swain, K. Burak, P. Klein, R. Myers, Jeff Kapler, M. Gill, Martin Labrie, C. Coffin
{"title":"Treatment outcomes with telaprevir-based therapy for HIV/hepatitis C coinfected patients are comparable with hepatitis C monoinfected patients","authors":"C. O’Neil, J. Pang, Samuel S. Lee, M. Swain, K. Burak, P. Klein, R. Myers, Jeff Kapler, M. Gill, Martin Labrie, C. Coffin","doi":"10.1155/2015/974871","DOIUrl":null,"url":null,"abstract":"To the Editor: Hepatitis C virus (HCV) infection is an important cause of morbidity and mortality among individuals living with HIV (1). Before the introduction of direct-acting antivirals (DAAs), pegylated interferon (peg-IFN) and ribavirin (RBV) were standard of care for coinfected patients with dismal sustained virological response (SVR) rates of <30% (2,3). Telaprevir (TVR), an NS3/4A protease inhibitor, was a first-generation DAA approved for HCV treatment in Canada, in November 2012. In randomized trials, the rate of SVR to TVR/peg-IFN/RBV was 65% to 75% in monoinfected patients and similar in coinfected patients (4-8). Few studies have reported treatment outcomes of TVR-based therapy outside of clinical trials. Our objective was to compare clinical outcomes of HCV-monoinfected and HIV-HCV coinfected patients treated with TVR-based triple therapy at a regional referral centre in Alberta. Patients who initiated TVR/pegIFN/RBV combination therapy from June 2011 to December 2013, were included in the study. Patients were treated according to Canadian guidelines for HCV treatment (9,10). All patients with HCV genotype 1 were eligible for therapy and were treated at the discretion of their HCV care provider. Demographic, clinical and laboratory data were collected at baseline and during therapy. Parameters of interest included HIV-coinfection, body mass index (BMI), Child-Pugh classification, previous injection drug use, haemophilia, liver transplantation, hepatitis B coinfection and previous HCV treatment. Fibrosis was determined using transient elastography by FibroScan (Echosens, France) with the following parameters: F0 to F1 ≤7.0, F2 7.1 to 9.4, F3 9.5 to 12.4, F4 (cirrhosis) ≥12.5 (11). Where applicable, HIV viral load and CD4+ T cell count were collected. Severe treatment-related anemia and thrombocytopenia were defined as nadir of hemoglobin ≤80 g/L and platelet count ≤50×109, respectively. Treatment response was determined using established definitions according to Canadian guidelines (9). Patients lost to follow-up were considered to have virological failure. In total, 103 patients received TVR at our clinics (Table 1). This included 13 (12.6%) HIV-HCV coinfected patients and seven (6.7%) patients who experienced recurrent HCV after liver transplantation. The median age at treatment onset was 56 years (interquartile range [IQR]: 51 to 59 years); 72% of patients were male and 86% were Caucasian. One-third (37%) of patients reported a history of injection drug use, nine (10%) had hemophilia and three (3%) were HCVhepatitis B virus coinfected. The median BMI was 26.8 kg/m2 (IQR 24.0 kg/m2 to 30.5 kg/m2). Forty-seven percent (n=45) of patients had been previously treated with pegIFN-RBV and 13% (n=12) were previous null responders. Most patients were HCV genotype 1a and IL28B non-CC genotype (71% and 70%, respectively). The majority (60%) of patients had advanced fibrosis or cirrhosis (F3 or F4). One patient had decompensated Child-Pugh B cirrhosis. HCV-HIV coinfected patients did not differ significantly with respect to previous anti-HCV therapy, HCV genotype subtype, interleukin (IL)28B genotype or degree of fibrosis. Coinfected patients were more likely to report injection drug use (P=0.05) and to have hemophilia (P=0.03). Most (92%) HIV coinfected patients had undetectable HIV RNA while receiving antiretroviral therapy, with a median baseline CD4 count of 490 cells/mm3 (IQR 250 cells/mm3 to 639 cells/mm3). Most (85%) required adjustment of their antiretroviral regimen before TVR initiation. Integrase-based antiretroviral therapy was the most commonly (77%) used regimen. The overall rate of SVR in our cohort was 66% (Table 2). The rate of SVR among HIV-HCV coinfected patients was 62% (eight of 13). Patients with cirrhosis and previous null responders had a lower SVR rate (54% and 42%, respectively). Fifty-seven percent (four of seven) of post-liver transplant recipients achieved SVR. Outcomes for postliver transplant patients have been previously reported (12). Among treatment failures, discontinuation due to adverse events was the most common (20%), followed by virological relapse (15%). Five (5%) patients discontinued therapy due to hepatic decompensation. Two (2%) patients were lost to follow-up. Two (2%) patients died; one patient died due to drug and alcohol intoxication while on therapy. The other patient had Child-Pugh B cirrhosis at baseline and died from complications of decompensated cirrhosis. The most commonly reported side effects were fatigue (65%), rash (68%), mood symptoms (42%), anorectal symptoms (43%) and infections (17%). Severe anemia occurred in 15% of participants and warranted red blood cell transfusion or erythropoietin in 11% and 2%, respectively. Severe thrombocytopenia occurred in 24% of participants. Most (57%) patients required RBV dose reduction. Comparing HCV monoinfected with HIV coinfected patients, there was no significant difference with regard to SVR (67% versus 62%, P=0.76). There was no difference between monoinfected and coinfected patients in treatment discontinuation due to adverse events (20% versus 15%; P=1.00) or virological relapse (13% versus 23%; P=0.40). One patient with HIV coinfection discontinued therapy due to hepatic decompensation, but nevertheless achieved SVR. There were no deaths among HIV-coinfected patients. Patients with HIV coinfection were more likely to have infections (12% versus 48%; P≤0.01), severe anemia (11% versus 38%; P=0.02) and to require peg-IFN dose adjustment (6% versus 46%; P≤0.01). In HIVcoinfected patients, infectious complications consisted of cellulitis (n=2), sepsis (n=1), gastroenteritis (n=1) and urinary tract infections (n=1). All HIV-coinfected patients maintained undetectable HIV RNA while receiving therapy. In a bivariate analysis, variables associated with increased rate of SVR included lower BMI (26.0 kg/m2 [IQR 24.0 kg/m2 to 29.1 kg/m2] versus 29.1 kg/m2 [IQR 26.6 kg/m2 to 32.0 kg/m2]; P=0.05), IL28B genotype CC (37% versus 12%; P=0.02) and cirrhosis (37% versus 60%; P=0.03). In a multivariate analysis, only fibrosis class (F0 to F2 versus F3 to F4; adjusted OR 0.34 [95% CI 0.12 to 0.99]; P=0.05) remained significantly associated with SVR. HIV status, a history of injection drug use and previous response to peg-IFN therapy did not predict SVR. There have been few reports of the effectiveness of TVR-based therapy in HIV-coinfected patients outside of clinical trials. In our study, the overall SVR was 67% in HCV-monoinfected patients and 62% in coinfected patients. This is comparable with clinical trials, despite a higher percentage of patients with cirrhosis (45%) in our cohort (4-6). Lower BMI, IL28B CC genotype and degree of fibrosis were associated with increased probability of SVR, although in multivariate analysis, only degree of fibrosis remained a significant predictor of SVR. Additional negative predictors of SVR with TVR-based therapy included African American race and previous treatment response (13). We also demonstrated that HIV coinfection is not a negative predictor of SVR. This was consistent with trials involving second-generation DAAs such as ledipasvir and sofosbuvir. Data from patients with HCV monoinfection treated","PeriodicalId":22481,"journal":{"name":"The Canadian Journal of Infectious Diseases & Medical Microbiology = Journal Canadien des Maladies Infectieuses et de la Microbiologie Médicale","volume":null,"pages":null},"PeriodicalIF":0.0000,"publicationDate":"2015-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"3","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"The Canadian Journal of Infectious Diseases & Medical Microbiology = Journal Canadien des Maladies Infectieuses et de la Microbiologie Médicale","FirstCategoryId":"1085","ListUrlMain":"https://doi.org/10.1155/2015/974871","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"","JCRName":"","Score":null,"Total":0}
引用次数: 3

Abstract

To the Editor: Hepatitis C virus (HCV) infection is an important cause of morbidity and mortality among individuals living with HIV (1). Before the introduction of direct-acting antivirals (DAAs), pegylated interferon (peg-IFN) and ribavirin (RBV) were standard of care for coinfected patients with dismal sustained virological response (SVR) rates of <30% (2,3). Telaprevir (TVR), an NS3/4A protease inhibitor, was a first-generation DAA approved for HCV treatment in Canada, in November 2012. In randomized trials, the rate of SVR to TVR/peg-IFN/RBV was 65% to 75% in monoinfected patients and similar in coinfected patients (4-8). Few studies have reported treatment outcomes of TVR-based therapy outside of clinical trials. Our objective was to compare clinical outcomes of HCV-monoinfected and HIV-HCV coinfected patients treated with TVR-based triple therapy at a regional referral centre in Alberta. Patients who initiated TVR/pegIFN/RBV combination therapy from June 2011 to December 2013, were included in the study. Patients were treated according to Canadian guidelines for HCV treatment (9,10). All patients with HCV genotype 1 were eligible for therapy and were treated at the discretion of their HCV care provider. Demographic, clinical and laboratory data were collected at baseline and during therapy. Parameters of interest included HIV-coinfection, body mass index (BMI), Child-Pugh classification, previous injection drug use, haemophilia, liver transplantation, hepatitis B coinfection and previous HCV treatment. Fibrosis was determined using transient elastography by FibroScan (Echosens, France) with the following parameters: F0 to F1 ≤7.0, F2 7.1 to 9.4, F3 9.5 to 12.4, F4 (cirrhosis) ≥12.5 (11). Where applicable, HIV viral load and CD4+ T cell count were collected. Severe treatment-related anemia and thrombocytopenia were defined as nadir of hemoglobin ≤80 g/L and platelet count ≤50×109, respectively. Treatment response was determined using established definitions according to Canadian guidelines (9). Patients lost to follow-up were considered to have virological failure. In total, 103 patients received TVR at our clinics (Table 1). This included 13 (12.6%) HIV-HCV coinfected patients and seven (6.7%) patients who experienced recurrent HCV after liver transplantation. The median age at treatment onset was 56 years (interquartile range [IQR]: 51 to 59 years); 72% of patients were male and 86% were Caucasian. One-third (37%) of patients reported a history of injection drug use, nine (10%) had hemophilia and three (3%) were HCVhepatitis B virus coinfected. The median BMI was 26.8 kg/m2 (IQR 24.0 kg/m2 to 30.5 kg/m2). Forty-seven percent (n=45) of patients had been previously treated with pegIFN-RBV and 13% (n=12) were previous null responders. Most patients were HCV genotype 1a and IL28B non-CC genotype (71% and 70%, respectively). The majority (60%) of patients had advanced fibrosis or cirrhosis (F3 or F4). One patient had decompensated Child-Pugh B cirrhosis. HCV-HIV coinfected patients did not differ significantly with respect to previous anti-HCV therapy, HCV genotype subtype, interleukin (IL)28B genotype or degree of fibrosis. Coinfected patients were more likely to report injection drug use (P=0.05) and to have hemophilia (P=0.03). Most (92%) HIV coinfected patients had undetectable HIV RNA while receiving antiretroviral therapy, with a median baseline CD4 count of 490 cells/mm3 (IQR 250 cells/mm3 to 639 cells/mm3). Most (85%) required adjustment of their antiretroviral regimen before TVR initiation. Integrase-based antiretroviral therapy was the most commonly (77%) used regimen. The overall rate of SVR in our cohort was 66% (Table 2). The rate of SVR among HIV-HCV coinfected patients was 62% (eight of 13). Patients with cirrhosis and previous null responders had a lower SVR rate (54% and 42%, respectively). Fifty-seven percent (four of seven) of post-liver transplant recipients achieved SVR. Outcomes for postliver transplant patients have been previously reported (12). Among treatment failures, discontinuation due to adverse events was the most common (20%), followed by virological relapse (15%). Five (5%) patients discontinued therapy due to hepatic decompensation. Two (2%) patients were lost to follow-up. Two (2%) patients died; one patient died due to drug and alcohol intoxication while on therapy. The other patient had Child-Pugh B cirrhosis at baseline and died from complications of decompensated cirrhosis. The most commonly reported side effects were fatigue (65%), rash (68%), mood symptoms (42%), anorectal symptoms (43%) and infections (17%). Severe anemia occurred in 15% of participants and warranted red blood cell transfusion or erythropoietin in 11% and 2%, respectively. Severe thrombocytopenia occurred in 24% of participants. Most (57%) patients required RBV dose reduction. Comparing HCV monoinfected with HIV coinfected patients, there was no significant difference with regard to SVR (67% versus 62%, P=0.76). There was no difference between monoinfected and coinfected patients in treatment discontinuation due to adverse events (20% versus 15%; P=1.00) or virological relapse (13% versus 23%; P=0.40). One patient with HIV coinfection discontinued therapy due to hepatic decompensation, but nevertheless achieved SVR. There were no deaths among HIV-coinfected patients. Patients with HIV coinfection were more likely to have infections (12% versus 48%; P≤0.01), severe anemia (11% versus 38%; P=0.02) and to require peg-IFN dose adjustment (6% versus 46%; P≤0.01). In HIVcoinfected patients, infectious complications consisted of cellulitis (n=2), sepsis (n=1), gastroenteritis (n=1) and urinary tract infections (n=1). All HIV-coinfected patients maintained undetectable HIV RNA while receiving therapy. In a bivariate analysis, variables associated with increased rate of SVR included lower BMI (26.0 kg/m2 [IQR 24.0 kg/m2 to 29.1 kg/m2] versus 29.1 kg/m2 [IQR 26.6 kg/m2 to 32.0 kg/m2]; P=0.05), IL28B genotype CC (37% versus 12%; P=0.02) and cirrhosis (37% versus 60%; P=0.03). In a multivariate analysis, only fibrosis class (F0 to F2 versus F3 to F4; adjusted OR 0.34 [95% CI 0.12 to 0.99]; P=0.05) remained significantly associated with SVR. HIV status, a history of injection drug use and previous response to peg-IFN therapy did not predict SVR. There have been few reports of the effectiveness of TVR-based therapy in HIV-coinfected patients outside of clinical trials. In our study, the overall SVR was 67% in HCV-monoinfected patients and 62% in coinfected patients. This is comparable with clinical trials, despite a higher percentage of patients with cirrhosis (45%) in our cohort (4-6). Lower BMI, IL28B CC genotype and degree of fibrosis were associated with increased probability of SVR, although in multivariate analysis, only degree of fibrosis remained a significant predictor of SVR. Additional negative predictors of SVR with TVR-based therapy included African American race and previous treatment response (13). We also demonstrated that HIV coinfection is not a negative predictor of SVR. This was consistent with trials involving second-generation DAAs such as ledipasvir and sofosbuvir. Data from patients with HCV monoinfection treated
查看原文
分享 分享
微信好友 朋友圈 QQ好友 复制链接
本刊更多论文
以替雷韦为基础的治疗HIV/丙型肝炎合并感染患者的治疗结果与单一丙型肝炎感染患者相当
致编辑:丙型肝炎病毒(HCV)感染是HIV感染者发病和死亡的重要原因(1)。在引入直接作用抗病毒药物(DAAs)之前,聚乙二醇化干扰素(peg-IFN)和利巴韦林(RBV)是治疗持续病毒学反应(SVR)率<30%的合并感染患者的标准治疗方法(2,3)。Telaprevir (TVR)是一种NS3/4A蛋白酶抑制剂,是加拿大于2012年11月批准用于HCV治疗的第一代DAA。在随机试验中,单感染患者对TVR/peg-IFN/RBV的SVR率为65%至75%,合并感染患者的SVR率相似(4-8)。在临床试验之外,很少有研究报道基于tvr的治疗结果。我们的目的是比较在阿尔伯塔省的一个区域转诊中心接受基于tvr的三联疗法治疗的单hcv感染和HIV-HCV合并感染患者的临床结果。研究纳入2011年6月至2013年12月期间接受TVR/pegIFN/RBV联合治疗的患者。患者按照加拿大HCV治疗指南进行治疗(9,10)。所有HCV基因型为1的患者均符合治疗条件,并由其HCV医护人员自行决定治疗。在基线和治疗期间收集人口统计学、临床和实验室数据。感兴趣的参数包括hiv合并感染、体重指数(BMI)、Child-Pugh分类、既往注射药物使用、血友病、肝移植、乙型肝炎合并感染和既往HCV治疗。纤维化采用FibroScan (Echosens, France)瞬时弹性成像测定,参数为F0 ~ F1≤7.0,F2 7.1 ~ 9.4, F3 9.5 ~ 12.4, F4(肝硬化)≥12.5(11)。在适用的情况下,收集HIV病毒载量和CD4+ T细胞计数。重度治疗相关性贫血和血小板减少的定义分别为血红蛋白≤80 g/L和血小板计数≤50×109的最低点。根据加拿大指南的既定定义确定治疗效果(9)。未能随访的患者被认为是病毒学失败。共有103例患者在我们的诊所接受了TVR(表1)。其中包括13例(12.6%)HIV-HCV合并感染患者和7例(6.7%)肝移植后复发的HCV患者。治疗开始时的中位年龄为56岁(四分位数间距[IQR]: 51至59岁);72%的患者为男性,86%为白种人。三分之一(37%)的患者报告有注射吸毒史,9例(10%)有血友病,3例(3%)合并感染hcvhbv病毒。中位BMI为26.8 kg/m2 (IQR为24.0 kg/m2至30.5 kg/m2)。47% (n=45)的患者以前曾接受过pegIFN-RBV治疗,13% (n=12)的患者以前是无效应答者。大多数患者为HCV基因型1a和IL28B非cc基因型(分别为71%和70%)。大多数(60%)患者有晚期纤维化或肝硬化(F3或F4)。1例失代偿Child-Pugh B肝硬化。HCV- hiv共感染患者在既往抗HCV治疗、HCV基因型亚型、白细胞介素(IL)28B基因型或纤维化程度方面无显著差异。合并感染患者报告注射用药(P=0.05)和血友病(P=0.03)的可能性更大。大多数(92%)HIV合并感染患者在接受抗逆转录病毒治疗时无法检测到HIV RNA,基线CD4细胞计数中位数为490细胞/mm3 (IQR为250细胞/mm3至639细胞/mm3)。大多数(85%)患者在TVR开始前需要调整抗逆转录病毒治疗方案。以整合酶为基础的抗逆转录病毒治疗是最常用的方案(77%)。在我们的队列中,SVR的总比率为66%(表2)。在HIV-HCV合并感染的患者中,SVR的比率为62%(13人中有8人)。肝硬化和既往无应答者的SVR率较低(分别为54%和42%)。57%(七分之四)的肝移植后受者达到了SVR。肝移植后患者的预后既往有报道(12)。在治疗失败中,因不良事件而停药最为常见(20%),其次是病毒学复发(15%)。5例(5%)患者因肝功能失代偿而停止治疗。2例(2%)患者失访。2例(2%)患者死亡;一名患者在治疗期间因药物和酒精中毒死亡。另一位患者基线时为Child-Pugh B肝硬化,死于失代偿肝硬化并发症。最常见的副作用是疲劳(65%)、皮疹(68%)、情绪症状(42%)、肛肠症状(43%)和感染(17%)。15%的参与者出现严重贫血,11%和2%的参与者需要输血或使用促红细胞生成素。24%的参与者出现了严重的血小板减少症。大多数(57%)患者需要减少RBV剂量。 比较HCV单感染者和HIV合并感染者,SVR无显著差异(67% vs 62%, P=0.76)。单感染和合并感染患者因不良事件而停止治疗的比例无差异(20% vs 15%;P=1.00)或病毒学复发(13% vs 23%;P = 0.40)。一名合并HIV感染的患者因肝功能失代偿而停止治疗,但仍然达到了SVR。合并感染艾滋病毒的患者中没有死亡病例。合并HIV感染的患者更容易发生感染(12%对48%;P≤0.01),重度贫血(11%对38%;P=0.02),需要调整peg-IFN剂量(6% vs 46%;P≤0.01)。在hiv感染患者中,感染性并发症包括蜂窝织炎(n=2)、败血症(n=1)、胃肠炎(n=1)和尿路感染(n=1)。所有HIV合并感染的患者在接受治疗时都维持着检测不到的HIV RNA。在双变量分析中,与SVR增加率相关的变量包括较低的BMI (26.0 kg/m2 [IQR 24.0 kg/m2至29.1 kg/m2] vs . 29.1 kg/m2 [IQR 26.6 kg/m2至32.0 kg/m2];P=0.05), IL28B基因型CC (37% vs 12%;P=0.02)和肝硬化(37% vs 60%;P = 0.03)。在多变量分析中,只有纤维化级别(F0至F2 vs F3至F4;调整OR 0.34 [95% CI 0.12 ~ 0.99];P=0.05)仍与SVR显著相关。HIV状态、注射药物使用史和既往对peg-IFN治疗的反应不能预测SVR。在临床试验之外,关于基于tvr的治疗在hiv合并感染患者中的有效性的报道很少。在我们的研究中,hcv单感染患者的总SVR为67%,合并感染患者的SVR为62%。这与临床试验相当,尽管在我们的队列中肝硬化患者的比例更高(45%)(4-6)。较低的BMI、IL28B CC基因型和纤维化程度与SVR的概率增加相关,尽管在多变量分析中,只有纤维化程度仍然是SVR的重要预测因子。基于tvr治疗的SVR的其他负面预测因素包括非裔美国人种族和既往治疗反应(13)。我们还证明HIV合并感染不是SVR的负向预测因子。这与涉及第二代daa(如ledipasvir和sofosbuvir)的试验一致。数据来自接受治疗的HCV单感染患者
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 去求助
来源期刊
自引率
0.00%
发文量
0
期刊最新文献
Circulating Dynamics of SARS-CoV-2 Variants between April 2021 and February 2022 in Turkey Global Occupational Exposure to Blood and Body Fluids among Healthcare Workers: Systematic Review and Meta-Analysis Baptism of Fire: Modeling the Effects of Prescribed Fire on Lyme Disease Spatiotemporal Study of COVID-19 in Fars Province, Iran, October-November 2020: Establishment of Early Warning System Emergence of SARS-CoV-2 New Variants and Their Clinical Significance
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
已复制链接
已复制链接
快去分享给好友吧!
我知道了
×
扫码分享
扫码分享
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1