Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2016 Mar 9.
Published in final edited form as: J Hepatol. 2013 Nov 23;60(4):872–884. doi: 10.1016/j.jhep.2013.11.013

Drug-drug interactions and Idiosyncratic Hepatotoxicity in the Liver Transplant setting

Sarah Tischer 1, Robert J Fontana 2
PMCID: PMC4784678  NIHMSID: NIHMS764594  PMID: 24280292

Abstract

Preliminary studies of boceprevir and telaprevir based antiviral therapy in liver transplant (LT) recipients with hepatitis C have demonstrated dramatic increases in tacrolimus, cyclosporine, and the mTOR inhibitor exposure. In addition to empiric dose reductions, daily monitoring of immunosuppressant blood levels is required when initiating as well as discontinuing the protease inhibitors to maximize patient safety. Although improved suppression of HCV replication is anticipated, 20 to 40% of treated subjects have required early treatment discontinuation due to various adverse events including anemia (100%), infection (30%), nephrotoxicity (20%) and rejection (5 to 10%). Simeprevir and faldepravir are 2nd generation protease inhibitors which may have improved efficacy and tolerability profiles but potential drug interactions with other OATP1B1 substrates and unconjugated hyperbilirubinemia are expected. In contrast, sofosbuvir and daclatasvir based therapies are not expected to lead to clinically significant drug-drug interactions in LT recipients but confirmatory studies are needed. Liver transplant recipients may also be at increased risk of developing drug induced liver injury (DILI). Establishing a diagnosis of DILI in the transplant setting is very difficult with the variable latency, laboratory features and histopathological manifestations of hepatotoxicity associated with a given drug, the need to exclude competing causes of allograft injury, and the lack of an objective and verifiable confirmatory test. Nonetheless, a heightened awareness of the possibility of DILI is warranted in light of the large number of medications used in LT recipients and the potential adverse impact that DILI may have on patient outcomes.


The calcineurin inhibitors (CNI), tacrolimus and cyclosporine, as well as the mammalian target of rapamycin inhibitors (mTORi), sirolimus and everolimus, are the backbone of modern immunosuppression in solid organ transplantation. Both of these drug classes are substrates of cytochrome–P450 (CYP) isoenzymes 3A4/5 and the drug-transporter, P-glycoprotein (P-gp). These metabolic pathways are also primarily involved in the elimination of 40 to 60% of all marketed drugs and in vivo expression of both CYP3A4/5 and P-gp vary substantially between individuals (16). As a result, administration of a drug that is a CYP3A or P-gp substrate/inhibitor to a liver transplant (LT) recipient can lead to dangerously high immunosuppressant blood levels, while intake of CYP3A inducers can predispose to subtherapeutic dosing and rejection (4,5). Therefore, transplant practitioners must be knowledgeable of the pharmacokinetic and potential drug-drug interaction (DDI) profiles of many drugs.

The azole antifungals and non-dihydropyridine calcium channel blockers are commonly prescribed drugs that can increase the blood levels of CNI’s and mTORi’s. For example, a 200 mg dose of fluconazole will increase the area under the curve (AUC) of cyclosporine by 1.8-fold and increase the tacrolimus trough concentration by 5-fold in transplant recipients (7). Similarly, intake of CYP3A inducers such as carbamazepine, St. John’s wort, and rifampin can lead to increased metabolism and reduced bioavailability of both CNI’s and mTORi’s (8). Boceprevir (BOC) and telaprevir (TPV) are NS3 protease inhibitors approved for use in combination with peginterferon (PEG-IFN) and ribavirin (RBV) for patients with chronic hepatitis C virus (HCV) genotype 1 infection. Both BOC and TPV are potent substrates and inhibitors of CYP3A and have demonstrated significant interactions with the CNI’s and mTORi’s in healthy volunteers as well as LT recipients. In this article, potential drug-interactions of BOC and TPV with immunosuppressants and other commonly used medications will be reviewed. In addition, preliminary safety and efficacy data of these drugs as well as other newer direct acting antiviral agents (DAA’s) in LT recipients will be provided. Lastly, a review of the incidence, presentation, and outcomes of drug induced liver injury (DILI) in LT recipients will be provided.

The first generation HCV protease inhibitors: Boceprevir and Telaprevir

Hepatitis C remains the leading indication for LT in most western countries and is associated with nearly universal recurrence of HCV replication and damage in the allograft (9, 10). The rate of liver disease and fibrosis progression in LT recipients is greatly accelerated compared to non-transplant patients with ~ 20% developing cirrhosis within 5 years of transplant and ~ 1 to 5% developing rapidly progressive and frequently fatal fibrosing cholestatic hepatitis (FCH) (11). As a result, PEG-IFN and RBV combination therapy is frequently used in selected LT recipients (12, 13). However, many LT recipients have contraindications to PEG-IFN therapy and rates of sustained virologic response (SVR) are substantially lower in LT recipients compared to non-transplant patients (e.g., 20% to 30% vs. 45% in HCV genotype 1) (12,13). The lower observed SVR rates are attributed to the use of immunosuppressant agents that enhance viral replication and the need for frequent antiviral dose reductions (50 to 70%) and early antiviral treatment discontinuation (20 to 40%) (12,14). Furthermore, there are increasing reports of immune-mediated allograft dysfunction due to PEG-IFN that may not only require early discontinuation of treatment, but also lead to premature graft failure and/or death (1517). However, since LT recipients who achieve SVR have a significantly improved survival compared to non-responders, there is an urgent unmet medical need to develop safer and more effective therapies for LT recipients (18, 19).

BOC and TPV in combination with PEG-IFN and RBV significantly improve SVR rates in both treatment naïve and previously treated patients with HCV genotype 1 infection compared to PEG-IFN and RBV alone (20, 21). In addition, only 6 months of response guided therapy is required in 50 to 60% of non-cirrhotic patients (20,2225). However, use of these agents is also associated with various adverse events including rash (50%), anorectal symptoms (30%), and anemia (50%) with TPV and dysgeusia (30%) and anemia (50%) with BOC treatment (26,27). Although both of these agents are considered investigational in LT recipients due to potential DDI’s with CNI’s and mTORi’s, the anticipated improvement in antiviral efficacy has generated a great deal of interest in using them in the transplant setting(28).

Drug-drug interactions with Boceprevir and Telaprevir

Boceprevir and TPV are extensively metabolized in the liver and both drugs are substrates and inhibitors of CYP3A. Telaprevir is also a potent substrate and inhibitor of Pg-p. Since elimination of BOC is dependent on multiple routes of metabolism, BOC is anticipated to be associated with less severe DDI’s with CYP3A substrates compared to TPV (28, 29).

Co-administration of BOC and TPV with drugs metabolized by CYP3A can lead to increased pharmacodynamic effects of those concomitant drugs due to reduced metabolism and increased bioavailability in the non-transplant setting (3034). For example, the area-under the curve (AUC) and maximum concentration (Cmax) of a 20 mg dose of atorvastatin increased 7.9 and 10.6-fold, respectively, with TPV co-administration while BOC increased the AUC and Cmax of a single 40 mg dose of atorvastatin by 2.3- and 2.7-fold, respectively (33,35). Therefore, atorvastatin should not be co-administered with TPV and the lowest possible dose of atorvastatin should be used in patients receiving BOC. Alternatively, pravastatin which is a weak inhibitor of CYP3A may be a suitable alternative (33). Similarly, the dose of intravenous midazolam should be reduced by at least 50% in patients receiving BOC or TPV (30,36). Digoxin levels are increased 18% when co-administered with BOC and increased 85% when co-administered with TPV (30,36). These latter data suggest that TPV is a moderate inhibitor of Pg-p while BOC appears to be a mild P-gp inhibitor (31).

Use of BOC and TPV may also alter the bioavailability and pharmacodynamic effect of some concomitantly administered medications. For example, both BOC and TPV lower the AUC of ethinyl estradiol by approximately 25%, which may result in the loss of contraceptive efficacy (30, 37). In addition, BOC and TPV have differing effects on the bioavailability of the progestin component of oral contraceptives (30). Since ribavirin is highly teratogenic, two alternative forms of contraception, such as an intrauterine device and barrier methods, are recommended during and after treatment with BOC or TPV based therapy (26,27,30).

Concomitant administration of CYP3A inhibitors and inducers may also alter the pharmacokinetics and pharmacodynamics of BOC and TPV during antiviral therapy (Supplemental Table 1). For example, administration of carbamazepine, a CYP3A inducer, may lower serum BOC and TPV levels and increase the risk of drug resistant variants developing in HCV patients. In contrast, drugs that are CYP3A inhibitors, such as the macrolide antibiotics, may lead to increased BOC or TPV exposure and increase the severity and frequency of adverse events (26,27,34). Therefore, reviewing all concomitant medications prior to BOC or TPV based therapy is required. If a concomitant medication(s) metabolized by CYP3A or P-gp is required, the lowest effective dose should be used or an agent that is not heavily dependent on CYP3A could be considered (Table 1).

Table 1.

Selected drugs that should be used with caution in subjects receiving boceprevir or telaprevir based antiviral treatment

Drug Class Effect on concomitant drug bioavailability (Clinical impact) Alternative agent(s) and management

Macrolide antibiotics

Clarithromycin
Erythromycin
Telithromycin
Increased (QT prolongation; Torsade de Pointes) Amoxicillin
Cefazolin
Clindamycin
Trimethoprim/sulfamethoxazole
Ciprofloxacin
Levofloxacin
Metronidazole

Antidepressants

Escitalopram* Decreased (Decreased efficacy) Citalopram
Sertraline
Venalfaxine
Duloxetine

Trazodone
Despiramine**
Increased (Dizziness, hypotension, nausea) As above
Use lower dose of trazodone

Anti-fungals

Itraconazole
Ketoconazole
Posaconazole
Voriconazole ***
Increased (QT prolongation, diarrhea, vomiting) Ketoconazole dose not to exceed 200 mg/day
Fluconazole
Micafungin
Caspofungin

Calcium channel blockers

Amlodipine
Diltiazem
Nicardipine
Nifedipine
Verapamil
Increased (Hypotension, bradycardia) Consider amlodipine dose reduction
Metoprolol, atenolol
Hydrochlorothiazide
Lisinopril, benazepril
Losartan, valsartan
Clonidine

Immunosuppressants

Cyclosporine
Everolimus
Sirolimus
Tacrolimus
Increased (Nephrotoxicity, hypertension, neurotoxicity) Significant dose reductions and close monitoring of drug levels

Prednisone
Methylprednisolone
Increased (hyperglycemia, osteoporosis, insomnia) Risk verses benefit
Use lowest effective dose.

Anti-arrhtymic

Amiodarone
Propafenone
Lidocaine
Quinidine
Increased (Proarrhtymic)

Digoxin Increased (Digoxin toxicity) Use lowest dose and monitor digoxin levels.
*

Only reported with TPV

**

Only reported with BOC

***

Not recommended to be used with TPV. TPV co-administration may increase or decrease voriconazole.

Adapted from boceprevir and telaprevir package insert (26, 27)

Please consult package inserts for complete list of known drug interactions and recommended management.

Effects of Telaprevir and Boceprevir on immunosuppressant drug levels

One of the greatest challenges of using BOC and TPV in the LT population is the dramatic effect that BOC and TPV have on CNI and mTORi blood levels (28,30,38). In one study of healthy volunteers, the AUC of cyclosporine increased 4.6 and 2.7-fold when co-administered with TPV and BOC, respectively (Supplemental Table 2). In addition, the AUC of tacrolimus increased 70.3- and 17.1-fold when co-administered with TPV and BOC in healthy individuals, respectively (39,40). Lastly, a study of BOC with single dose sirolimus in healthy volunteers showed a significant increase in the AUC and Cmax of sirolimus by 8.1 and 4.8-fold, respectively (41). Currently, use of BOC and TPV in subjects receiving CNI’s and mTORi is considered a relative to absolute contraindication until additional safety data are obtained (26,27).

Despite the aforementioned concerns, several studies have begun to explore the use of BOC and TPV in combination with PEG-IFN and RBV in carefully monitored LT recipients (Table 2). A substantial reduction in the clearance of tacrolimus (~80%), cyclosporine (~50%), and everolimus (53%) was reported in LT recipients receiving BOC with PEG-IFN and RBV (42). In addition, a significant reduction in the clearance of both cyclosporine and tacrolimus in LT recipients receiving TPV and PEG-IFN and RBV therapy was reported (43). The median weekly dose of tacrolimus and cyclosporine during TPV treatment was 4% and 14% of the pretreatment dose, respectively (43). Similarly, the AUC of sirolimus increased 26-fold and the mean terminal half-life increased 1.5-fold in 5 patients receiving TPV and PEG-IFN and RBV (44). During the 12 weeks of TPV therapy, patients required only 3 to 33% (mean 11%) of the pretreatment sirolimus dose with doses ranging from 0.5 to 1 mg every 5 to 22 days (44).

Table 2.

Ongoing studies of Boceprevir and Telaprevir with PEG-IFN and RBV in LT recipients with HCV genotype 1

Study (ref) Antiviral regimen Immunosuppressant % HCV-RNA undetectable Immunosuppressant dose adjustments % Adverse events
Coily (Ref #47)
N=37
BOC
4 week PEG-IFN + RBV then BOC + PEG-IFN/RBV
N=18
Mean Rx = 41 weeks


TPV
4 week PEG-IFN + RBV; TPV + PEG-IFN + RBV × 12 week then PEG-IFN + RBV
N=19
Mean Rx = 41 weeks
BOC
CSA =12
TAC =6


TPV
CSA =10
TAC =9
BOC
56% Week 8
89% Week 16
72% Week 48
11% early viral breakthrough


TPV
47% Week 8
58% Week 16
21% Week 48
21% early viral breakthrough
BOC
CSA 36% of original dose
TAC 22% of original dose


TPV
CSA 54% of original dose
TAC 5% of original dose

All patients hospitalized for CNI dose adjustments
BOC
100 % Anemia
61% Neutropenia
50% Thrombocytopenia
5% Rash
5% Nephrotoxicity
26% Infections
33% Hospitalization
11% DeathTPV
84% Anemia
21% Neutropenia
15% Thrombocytopenia
5% Rash
21% Nephrotoxicity
27% Infections
31% Hospitalization
5% Death
Pungpapong, (Ref #46)
(n=60)
BOC
4 week PEG-IFN + RBV then BOC + PEG-IFN/RBV (n=25)

Mean Rx=39 wk


TPV
TPV + PEG-IFN/RBV × 12 wk then PEGIFN/RIB (n=35)

Mean Rx = 32 wk
BOC
CSA = 23
TAC= 2


TPV
CSA = 33
SRL =1
TAC =1
BOC
24% Week 8
40% Week 12
12% early viral breakthrough


TPV
17% Week 4
80% Week 12
8% early viral breakthrough
BOC
CSA 33–100% (mean 56%) of original dosed every 12 hours
TAC 86% reduction of original dose dosed twice weekly to every 48 hours


TPV
CSA 50–100% (mean 70%) of original dose every 12 hours
SRL = 0.5 mg every 4 days
TAC = 0.5 mg every 7 days
BOC
100% Anemia
76% Leukopenia
36% increase SCr > 0.5 mg/dL
32% Rash
4 % Infections
4% Acute rejection
4% Death
TPV
88% Anemia
77% Leukopenia
11% SCr increase > 0.5 mg/dL
31% Rash
6% Infections
6% Acute rejection
3% Death
Werner (Ref #49)
(n=9)
TPV
TPV + PEG-IFN/RBV × 12 weeks then PEG-IFN/RBV

Mean Rx= 12 wks
TPV
CSA = 4
TAC = 4
SRL = 1
TPV
44% Week 4
88% Week 12
TPV- Fold dose reduction
TAC = 22 (96%)
SRL = 7 (86%)
CSA = 2.5 (60%)

Immunosuppressant interval
TAC – single dose per week
SIRL – single dose per week
CSA – single dose daily
TPV
66% Anemia
66% Leukopenia
44% Thrombocytopenia
33% Rash
44% Hospitalized infection
11% Renal failure
Brown (Ref #48)
(n=26)
TPV
TPV+PEG-IFN/RBV × 12 wks then PEG-IFN/RBV

Mean Rx = 12 wks
TPV
TAC = 23
CSA =3
TPV
47% Week 4
82% Week 12
TPV
CSA reduced 4-fold
TAC reduced 10-fold

Med time to 1st TAC dose =71 hrs
Median TAC dose =0.5 mg every 7 days
Med time to 1st CSA dose 22 hrs
Med CSA dose =25 every 24 hrs
TPV
39% Anemia
35% Rash (mild)
17% Anorectal symptoms
13% Pruritus
13% Renal insufficiency
4% Supratherapeutic CNI level (discontinued TPV)
O’Leary (Ref #44)
(n= 120**)
TPV/BOC
TPV (n=107) or BOC (n=13) + PEG/IFN ± PEG-IFN/RBV lead-in (n=116)

Median Rx = 148 days
TPV/BOC
TAC =35
CSA = 73
TPV/BOC
63% Week 4
78% Week 12
72% Week 24

7% Viral breakthrough
TPV/BOC
Median daily dose prior and after initiation of TPV or BOC
CSA = 200 mg/50 mg
TAC = 1.0 mg/0.19 mg
TPV/BOC
79% Anemia
45% Blood transfusion
43% GCSF
20% Hospitalization
33% Scr increased > 0.5 mg/dL
6% Acute rejection
2% Death

Abbreviations: BOC, boceprevir; CSA, cyclosporine; EVRL, everolimus; IS, immunosuppression; MMF, mycophenolate mofetil; PEG-IFN, peginterferon; RBV, ribavirin; Rx, treatment duration; Scr, serum creatinine; SIRL, sirolimus; TAC, tacrolimus; TPV, telaprevir

*

One patient received prolonged-release (once daily) tacrolimus

**

6 liver-kidney transplant recipients

Prednisone and methylprednisolone are also substrates of CYP3A and one study demonstrated a 37% increase in prednisolone AUC when co-administered with BOC (26, 27, 31, 45). However, the increase in prednisolone concentration is unlikely to be clinically significant, so no dose adjustments are recommended (26,27, 45).

Boceprevir and Telaprevir based antiviral therapy in LT recipients

There are several ongoing studies of BOC and TPV in combination with PEG-IFN and RBV in LT recipients with recurrent HCV genotype 1 (Table 2) (4652). In one study, 35 patients treated with TPV, PEG-IFN and RBV were followed for a mean of 32 weeks and 25 BOC treated patients were followed for a mean of 39 weeks (46). Prior to initiation of treatment, 92% of the patients were converted to cyclosporine. Thus far, 14 (67%) TPV and 10 (45%) BOC treated patients had undetectable HCV RNA at week 24 and 3 (5%) had developed viral breakthrough. Despite restricting the initial ribavirin dose to 800 mg/day, anemia was encountered in 100% of the patients and 50% required a blood transfusion. In addition, biopsy-proven rejection due to subtherapeutic cyclosporine levels occurred in two patients during TPV therapy and another patient following discontinuation of BOC. Of the two patients that died, one with FCH developed sepsis after treatment of rejection and the other patient had decompensation prior to starting antiviral therapy.

The preliminary results of a multicenter French study of 37 LT recipients treated with either BOC or TPV were recently published (47). Sixteen percent of these patients had FCH and 51% had received prior antiviral therapy post LT. A 4-week lead-in of PEG-IFN and RBV was given to 84% of patients and all of the patients were hospitalized when BOC or TPV was started to monitor CNI levels. Quite remarkably, 89% of the BOC and 58% of the TPV treated patients had an undetectable HCV RNA at week 16. However, early discontinuation of therapy was required in 58% of the TPV treated patients due to severe infections or a lack of response, while only 28% of the BOC treated patients required early discontinuation of therapy. Although follow-up is ongoing, 71% of the BOC treated patients and 20% of the TPV treated patients with a week-48 response have remained HCV RNA negative at post-treatment week 12. Anemia was encountered in 100% of the patients and 35% required a blood transfusion. Only one episode of mild rejection was reported, but 8% died of liver-related complications.

The week 12 results of the ongoing REFRESH study demonstrate more promising outcomes with TPV use in LT recipients with 47% and 82% of patients achieving undetectable HCV RNA at weeks 4 and 12, respectively (48). The most frequent adverse events include anemia (39%) and rash (35%) but follow-up is ongoing.

Overall, these preliminary data suggest that the addition of TPV or BOC to PEG-IFN and RBV can lead to increased rates of HCV RNA suppression in LT recipients compared to historical controls. However, the dose of CNI needs to be markedly reduced during BOC and TPV therapy with highly variable dosing intervals necessitating the need for frequent therapeutic drug monitoring. In addition, a rapid increase in the CNI dosing and frequency is required within 1 to 2 days of discontinuing BOC or TPV to minimize the risk of under immunosuppression and rejection (53).

Adverse effects of Boceprevir and Telaprevir in LT recipients

Anemia has been a universal and potentially severe adverse event with BOC and TPV therapy in LT recipients (4654). This is, in part, due to the impaired clearance of RBV in LT recipients with renal insufficiency as well as the bone marrow suppressive effects of PEG-IFN, BOC, and TPV (54,55). Despite a lower starting dose of RBV, aggressive RBV dose reductions have been needed and erythropoietin stimulating agents (ESA) have been used in 60 to 90% of treated patients (4652). Skin rashes have also been frequently noted but they have not been severe (4651). Mild to moderate renal insufficiency has also been reported during triple antiviral therapy, which may, in part, be due to drug-drug interactions with the CNI’s. (4449,52,56). However, recent studies in non-transplant patients have demonstrated significant but reversible reductions in renal function with TPV and BOC therapy attributed to renal tubular transporter effects (56,57). Due to these safety concerns, frequent therapeutic drug monitoring and assessment of renal function is recommended in LT recipients receiving these agents. Bacterial infections resulting in hospitalization or even death have also been reported in up to 33 % of LT recipients further highlighting the need for frequent and vigilant clinical assessment of all treated patients (46,47,59,50).

CNI and mTORi dosing during and after Telaprevir and Boceprevir therapy

Empiric adjustments of the CNI and mTORi dose and interval must be made at the time of initiation of BOC or TPV to minimize the risk of toxicity. Currently, prospective studies to provide safe and accurate estimates of the extent of CNI dose reduction are ongoing (43). However, since the severity of the CYP3A interaction is less with cyclosporine compared to tacrolimus, many centers have opted for conversion to cyclosporine prior to initiating BOC or TPV therapy in LT recipients. Regardless of the CNI or mTORi used, immunosuppressant blood levels should be stable and within therapeutic range for at least 1 month prior to starting antiviral therapy (Supplemental Table 3). Most studies have withheld CNI dosing after the initiation of TPV and then checked daily morning CNI blood levels to guide future doses (46,48,49). When using tacrolimus with TPV, it is suggested to use 10% of the initial total daily dose once the morning trough level goes below 3 or 4 ng/ml. In the ongoing REFRESH study, the reported dosing interval of tacrolimus ranged from once every 4 to 25 days. In contrast, the cyclosporine dose is usually 25% of the initial total daily dose and the dosing interval ranged from once every 1 to 7 days (48). There is less data available with BOC in LT recipients, but one study suggested that cyclosporine could be administered at 50% of the initial total daily dose and given once a day, while the tacrolimus dose should be started at approximately 25% of the initial dose and the interval guided by daily assessment of trough levels (46).

It is also critical to resume dosing of the CNI and mTORi to at least the pre-treatment dose within 1 to 2 days of BOC and TPV discontinuation and frequently monitor immunosuppressant blood levels for the first two weeks after BOC and TPV discontinuation. Since LT recipients with suppression of HCV replication have improved hepatic metabolic function, higher daily doses of the CNI’s and mTORi’s may be required early after discontinuation of BOC and TPV in up to 30% of patients (46,5860). Therefore, close monitoring of immunosuppressant blood levels is imperative throughout antiviral therapy as well as after discontinuation of BOC and TPV to prevent rejection.

Direct acting antivirals in the pipeline

Several DAA’s are in phase 3 development and may gain regulatory approval in the near future. Drugs that will likely reach the marketplace soon include the NS3 protease inhibitors, simeprevir and faldaprevir; the NS5A replication complex inhibitors daclatasvir; and the nucelos(t)ide NS5B polymerase inhibitor, sofosbuvir. In addition, an IFN-free regimen consisting of ritonavir boosted ABT-450, a protease inhibitor, ABT-267, a NS5A inhibitor, and ABT-333, a non-nucleoside polymerase inhibitor, is demonstrating promising efficacy results in both treatment naïve and experienced patients (61,62). However, IFN-free regimens may have reduced efficacy in LT recipients wherein the high frequency of HCV genotype 1a, advanced fibrosis, high levels of HCV replication, and altered drug pharmacokinetics pose substantial therapeutic challenges (63).

The new DAA’s offer several potential therapeutic advantages over the currently approved protease inhibitors, including improved antiviral efficacy, shorter duration of therapy, and fewer side effects. Studies of faldaprevir, simeprevir, daclatasivr and sofosbuvir in combination with PEG-IFN and RBV have demonstrated SVR rates, of ~70–90% in treatment naïve non-transplant, HCV genotype 1 patients treated for 12 to 48 weeks (6471). Furthermore, the addition of two DAA’s to PEG-IFN and RBV has demonstrated an almost 100% SVR even in historically difficult to treat populations (72,73). In addition, sofosbuvir combined with ribavirin alone for 12 weeks is associated with a 97% SVR in genotype 2 and 67% SVR in genotype 3 patients (74,75). The use of ledipsavir in combination with sofosbuvir and ribavirin may be particularly attractive in LT recipients with genotype 1 infection (76,77). Many of the new DAA’s also have improved bioavailability and longer half-lives requiring less frequent dosing and do not require administration with food.

Available data also suggest a lower likelihood of clinically significant DDI’s with some of the new DAA’s compared to BOC and TPV (Table 3) (32, 82,83, 8889). However, several are CYP3A and drug transporter substrates and inhibitors. For example, the AUC of tacrolimus decreased by 17% and that of cyclosporine increased by 19% with simeprevir co-administration (78). ABT-450 is an inhibitor of OATP1B1 that leads to unconjugated hyperbilirubinemia and the boosting of its bioavailability with ritonavir, a potent CYP3A4 substrate, may create difficulties in the LT population (79). Although, sofosbuvir does not undergo metabolism via CYP3A, dose adjustments are anticipated for patients with moderate or severe renal impairment. Faldaprevir can lead to unconjugated hyperbilirubinemia via inhibition of UGT1A1 (80). Lastly, simeprevir is a substrate of OATP1B1 and results in an increase in total bilirubin levels in subjects treated with ribavirin (81).

Table 3.

Pharmacokinetic and metabolic parameters of selected direct acting antiviral agents for hepatitis C

Drug and dose Metabolism / excretion route CYP inducer or inhibitor Transporter substrate or inhibitor Comments
NS3 Protease inhibitors
ABT-450/
Ritonavir (150 mg q day/100 mg q day)
Hepatic (CYP3A) Strong CYP3A inhibition by ritonavir Inhibitor of OATP1B1 Unconjugated hyperbilirubinemia
Boceprevir 800 mg tid Hepatic (CYP3A, aldoketoreductase) Moderate CYP3A inhibitor Weak P-gp inhibitor Significant DDI’s with other CYP3A substrate drugs
Faldaprevir 120 mg q day (BI 20335) Hepatic (CYP3A) Moderate CYP3A inhibitor; weak CYP2C9 inhibitor Inhibits OATP1B1, OATP1B2, OATP2B1; Substrate of P-gp and MRP2 Inhibition of UGTA1 results in unconjugated hyperbilirubinemia
Simeprevir 150 mg q day (TMC-435) Hepatic (CYP3A) Mild CYP1A2 inhibitor; mild intestinal/ hepatic CYP3A inhibitor Inhibitor of OATP1B1 and MRP2 Unconjugated hyperbilirubinemia commonly seen
Telaprevir 750 mg tid Hepatic (CYP3A) Strong CYP3A inhibitor Moderate P-gp inhibitor Significant DDI’s with other CYP3A and P-gp substrate drugs
NS5A replication complex inhibitors
ABT-267 25 mg q day No data No data No data AUC and CMax increased 62% and 67% by ritonavir, respectively
Daclatasvir 60 mg q day (BMS-790052) Hepatic (CYP3A) Not a inducer or inhibitor of CYP3A4 Moderate inhibitor of P-gp and OATP1B1
Ledipasvir 90 mg q day (GS-5885) Feces (major); hepatic and renal (minor) Not a CYP inhibitor or inducer Weak inhibitor of P-gp, OATP1B1
Nucleos(t)ide polymerase inhibitors
Sofosbuvir 400 mg q day (GS-7977) Renal No clinical evidence of CYP inhibition or induction Substrate of P-gp Dose reduction if moderate to severe renal impairment
Non-nucleoside polymerase inhibitors
ABT-333 400 mg BID Hepatic
CYP2C8 (60%); CYP3A4 (30%); CYP2D6 (10%)
Not a CYP3A inducer
Deleobuvir 600 mg BID (BI-207127) No data No data Substrate of P-gp, BCRP, OATP1BI, OATP1B3

Abbreviations: BCRP, breast cancer resistance protein; BID, two times a day; CYP, cytochrome P450; MRP, multiple drug resistance protein; OATP, organic anion transporting polypeptide; P-gp, P-glycoprotein; q, every day; UGT, uridine glucuronly transferase

Adapted from Kiser JJ, et al. Ref #32

Data regarding the safety and efficacy of the new DAA’s in various special patient populations will likely be lacking at the time of their approval. Therefore, careful scrutiny of available pharmacokinetic and clinical data will be essential for successful use of these new drugs in the transplant setting. Administration of daclatasvir with PEG-IFN and RBV for 24 weeks in an LT recipient with severe cholestatic HCV infection led to an SVR (82). In addition, the first ever successful use of an IFN-free regimen consisting of sofosbuvir and daclatasvir for 24 weeks in a LT recipient with FCH was recently reported (83). However, large, prospective, multicenter studies are needed to determine the optimal agent(s), duration of therapy, and safety profile in LT recipients (86).

Idiosyncratic drug induced liver injury in the transplant setting

Drug-induced liver injury (DILI) is an increasingly recognized cause of clinically significant acute and chronic liver disease in both children and adults (90,91). DILI is a leading cause of acute liver failure (ALF) in western countries and the most common reason for removal of approved medications from the marketplace (92, 93). However, most cases of DILI are “idiosyncratic” and not associated with the dose or duration of medication administered nor obvious clinical risk factors Furthermore, the protean clinical and laboratory presentations of liver injury due to a particular drug coupled with the lack of an objective and confirmatory diagnostic test frequently leads to a delay in diagnosis (94, 95).

DILI in the general population

The incidence of DILI in the general population is not well known. However, DILI accounts for < 1% of consecutive acute liver disease cases seen in referral centers with viral hepatitis, pancreaticobiliary disease, hepatic ischemia, and alcohol being much more common (96, 97). The incidence of DILI in a prospective cohort study from Northern France was 14 cases per 100,000 patient years (98). More recently, the incidence of DILI in the 250,000 adult inhabitants of Iceland was estimated to be 19.1 cases per 100,000 patient years (99). In western countries, the majority of DILI cases are attributed to antibiotics, anticonvulsants and psychiatric medications (Table 4) (100, 101). However herbal and dietary supplements (HDS) can also cause clinically significant liver injury (102). The Drug Induced Liver Injury Network (DILIN) demonstrated that 73% of DILI cases in the United States were attributed to a single prescription medication while 9% were attributed to a single or multiple HDS products and 18% were attributed to multiple medications (100).

Table 4.

Presentation and outcomes with DILI in the general population and LT recipients

Feature DILIN US
N=300
(ref #100)
Spain
N=446
(ref #101)
Mayo-Liver transplant
N= 29
(ref #118)
Study Design Prospective
Multicenter
(8 sites)
‘04 to ‘08
Prospective Multicenter
(32 sites)
’94-‘05
Retrospective LT center (1 site)
’85-‘10‘
Causality method DILIN
Expert opinion
RUCAM DILIN
Expert opinion
F/U duration (mon) 6 to 24 3 NA
Mean age (yrs) 48 53 52
% Female 60% 49% 52%
Race
 % Caucasian
 % African American
 % Asian
 % Other
79%
11%
4%
6%
100% NA
Liver injury type
 % Hepatocellular
 % Mixed/ Cholestatic
57%
20%/23%
58%
22%/20%
7%
4%/89%
% Jaundice 69% 71% 24%
% Liver biopsy 50% 25% 96%
% Hospitalized 60% 53% 8%
% Died or transplanted 10% 7% 0%
Median duration medication use (d) 42 105 57
Suspect drugs
 % Antibiotics
 % Psychotropic
 % HDS products
 % Hypolipidemic
% Immunosuppressants
45%
15%
9%
3%
1%
32%
17%
0%
3%
0%
58%
4%
4%
7%
14%

NA= Not available

The diagnosis of DILI rests on finding abnormalities in serum aspartate aminotransferase (AST), alanine aminotransferase (ALT), alkaline phosphatase, or total bilirubin levels while on the drug compared to pretreatment baseline values. Causality assessment is largely a diagnosis of exclusion that relies on 1) time from drug initiation to DILI onset, 2) clinical and laboratory features at presentation, 3) the time and course of recovery after drug discontinuation (i.e. de-challenge), 4) presence of established risk factors, 5) exclusion of competing causes of liver injury, and 6) previous reports on the hepatotoxicity of the implicated agent. Recently, a checklist of the essential elements to consider in investigating a possible DILI case was published (103). Causality assessment instruments can assist with DILI case recognition, but expert opinion appears to be a more reliable and accurate diagnostic method but is not widely available nor generalizable (104106). The LIverTox website was recently established by the NIH and National Library of Medicine to provide up-to-date and evidence based information on the hepatotoxicity profile of over 600 marketed drugs (107). The website also provides useful information on the proposed mechanism, risk factors, and overall likelihood of bonafide injury attributed to a particular drug.

The clinical course of DILI can be categorized as hepatocellular, cholestatic or mixed based upon the presenting laboratory profile and liver histology (90). The severity of a DILI episode can vary from asymptomatic to severe and life threatening. DILIN has established a 5-point system for grading severity based on symptoms, jaundice, need for hospitalization, and signs of hepatic failure (108). DILIN and other groups have demonstrated that subjects who present with severe hepatocellular injury that are jaundiced at the time of hospitalization may have as high as a 10% mortality rate validating “Hy’s law (100, 101).

Studies of DILI in the transplant setting

In the LT setting, exclusion of biliary, infectious, vascular, and immunological causes of allograft dysfunction is essential since they are more likely to cause liver injury than DILI (109). Furthermore, LT recipients may also develop recurrent disease in their allograft (110). In addition, idiopathic “alloimmune hepatitis” can develop at any time post-LT even in previously stable patients (111, 112). Finally, some solid organ transplant recipients may be chronically infected with hepatitis E virus and misdiagnosed as having DILI (113, 114). Therefore, a thorough evaluation for competing causes of liver injury using molecular diagnostic assays, liver imaging, and liver histology is required to exclude the myriad causes of allograft dysfunction in LT recipients.

Patients with liver disease may be at increased risk of developing DILI due to altered pharmacokinetics, up-regulated intrahepatic cytokine expression, and alterations in drug-metabolizing pathways (115, 116). For example, subjects with HIV and HCV or HBV co-infection are at greater risk of developing serum ALT elevations during anti-retroviral therapy compared to HIV mono-infected patients (116). However, it can be exceedingly difficult to reliably distinguish a flare in the underlying liver disease from a DILI episode.

Case series

Currently, there is a paucity of data on the frequency, etiologies, and outcomes with DILI in the LT setting. Recently, DILI was implicated in 131 Chinese LT recipients undergoing protocol liver biopsies at a single center over a 6-year period (117). Of note, 44% of the DILI cases occurred within the first 30 days of LT and antifungal agents were the leading suspect drug (29%). All of the patients survived and improved during follow-up. However, the criteria used to establish a diagnosis of DILI and the extent to which other causes of allograft dysfunction were excluded are unclear. In addition, many of the liver biopsy samples demonstrated evidence of hepatic steatosis and necrosis, which are commonly encountered in the early post-LT setting.

The frequency and risk factors for DILI in 1689 consecutive LT recipients from Mayo Clinic seen over a 15 year period were also recently reported (118). A diagnosis of “Definite DILI” was based on the presence of clinical criteria and a compatible liver biopsy after rigorous exclusion of competing causes using expert opinion for causality assessment (103). Of the 79 patients with suspected DILI based upon pathology records, there were only 28 individuals who met clinical criteria for “definite DILI” leading to an overall DILI incidence of 1.7%. The mean age of the DILI patients was 52 years and 52% were women. The major indications for LT in these 28 patients were primary sclerosing cholangitis (28%), cholangiocarcinoma (14%) and hepatocellular carcinoma (14%) with the former being over-represented compared to non-DILI LT recipients. The DILIN severity scores were mild (1) or moderate (2) in 92% of the patients (Table 4). The median duration of suspect medication use was 57 days and the most frequently identified drugs were antibiotics (48%), immunosuppressive agents (14%) and hypolipidemics (7%). Trimethoprim-sulfamethoxazole (TMP-SMZ) was the single most commonly implicated drug. The serum aminotransferase levels normalized during a median follow-up of 34 days after drug withdrawal. There was no clear relationship between donor characteristics nor time interval since LT and DILI diagnosis.

These intriguing data suggest that the incidence of DILI in LT recipients of 1.7% is substantially higher (i.e. 100 fold) than that reported in the general population (0.02%). Prior studies of immunosuppressed patients with HIV infection have also demonstrated that they are at increased risk of developing hepatotoxicity from TMP-SMZ and isoniazid (119). There are also prior case reports of LT recipients acquiring food allergies from the donor (120). Therefore, immunosuppressed LT recipients may be at increased risk of developing DILI.

Hepatotoxicity of frequently used drugs in LT recipients

Immunosuppressants

Azathioprine, a prodrug of mercaptopurine that inhibits T-cell maturation, has been a backbone of immunosuppressive regimens in LT recipients for several decades. Patients with low levels or deficiency in thiopurine methyltransferase, which affects ~10% of the population, have a higher rate of myelotoxicity with azathioprine use but do not appear to have a higher incidence of DILI. Azathioprine leads to hepatotoxicity in up to 1 to 5% of non-transplant patients treated for prolonged periods of time (121, 122). Many of these patients present with mild hepatocellular injury or cholestasis which resolves with drug discontinuation. Individual case reports have also described nodular regenerative hyperplasia with prolonged exposure to high dose azathioprine in LT recipients whom frequently present with a cholestatic laboratory profile (123, 124). The pathophysiology of this lesion is believed to be due to endothelial cell damage that leads to sinusoidal dilatation and obliterative pericentral veno-occlusive changes. Despite its widespread use, acute hepatocellular injury attributed to mycophenolate mofetil has been only rarely reported (133135).

Hepatotoxicity attributed to cyclosporine and tacrolimus also appears to be uncommon in light of their near universal use in hundreds of thousands of solid organ transplant recipients. Individual cases of cholestatic liver injury following the use of tacrolimus have been reported that usually improved with dose reductions or switching to an alternative agent (128131). Severe acute hepatocellular injury with jaundice was previously reported in kidney transplant patients receiving high doses of cyclosporine with histological features of cholestasis and pericholangitis (130, 131). The mechanism of this intrahepatic cholestasis may be due to inhibition of canalicular bile flow and inhibition of bile salt export pump (BSEP) (132). However, testing for HCV and other causes of viral infection were not routinely done in these early studies and many of the patients appeared to improve with cyclosporine dose reduction. Sirolimus has been reported to cause liver injury in HCV patients but clinically apparent DILI attributed to everolimus has not been reported (133135).

Antibiotics

Antibiotics are commonly used to prevent and treat bacterial and fungal infections post-transplant. Amoxicillin- clavulanate is a leading cause of DILI in the general population and has also been associated with DILI in a pediatric LT recipient (100, 136). TMP-SMZ can cause a cholestatic liver injury within a few days to weeks of drug initiation with prominent hypersensitivity features of skin rash, fever and eosinophilia (137). A minority of patients treated with TMP-SMZ may also develop life-threatening DRESS syndrome (Drug rash and eosinophilia and systemic symptoms) while others have mild biochemical liver injury and hepatic granulomas on biopsy (138). The presenting liver injury pattern is typically cholestatic or mixed and may be associated with prolonged jaundice. As with other sulfonamides, TMP-SMZ has also been linked to cases of severe acute hepatocellular injury that may be severe and even fatal.

The azole antifungals are frequently used to treat and prevent systemic and superficial fungal infections in LT recipients. In addition to being potent inhibitors of CYP3A4, fluconazole can cause mild to moderate serum aminotransferase elevations in up to 5% of treated patients. Fluconazole, as well as the other azole antifungals (itraconazole, voriconazole, ketoconazole), can also rarely lead to severe acute hepatocellular injury with jaundice (139, 140).

Isoniazid is a leading cause of severe acute DILI that may result in emergency LT (92). In these instances, anti-tuberculosis (TB) prophylaxis with an alternative regimen containing a quinolone, rifampin, or amikacin may be required in the early post-LT setting to prevent TB reactivation (141). The optimal time and duration of isoniazid therapy for LT recipients with latent TB remains unclear, but should generally be deferred until at least 6 months post-LT to reduce the risk of inadvertent hepatotoxicity (142, 143).

Antiviral agents

Ganciclovir and valganciclovir are frequently used to treat and prevent cytomegalovirus infection in the LT setting. Neither agent has been associated with clinically apparent liver injury, but intravenous administration of ganciclovir is associated with mild to moderate increases in serum ALT levels in ~ 2% of treated patients that are typically self-limited (144).

Other agents

Individuals who consume weight loss products that contain green tea extract with variable amounts of catechins may develop severe acute hepatocellular injury with jaundice including LT recipients (145, 146). Other drugs associated with DILI in LT recipients include sorafenib to treat recurrent liver cancer and intravenously administered amiodarone for peri-operative atrial fibrillation (147149).

Summary and conclusions

The introduction of potent and highly effective DAA’s has ushered in a new era in the management of both LT candidates and recipients with HCV infection. Knowledge of the metabolic pathways involved in the elimination of these agents will be critical for their optimal and safe use in the LT population. Clinically significant DDI’s have consistently been reported in LT recipients treated with TPV and BOC, which mandate empiric CNI dose reductions and intensive monitoring of immunosuppressant blood levels during and after their discontinuation. It is anticipated that several of the HCV polymerase inhibitors, NS5A replication complex inhibitors, and 2nd generation protease inhibitors will be associated with fewer DDI’s and adverse effects but prospective studies of these agents in LT recipients are needed. Finally, LT recipients appear to be at increased risk of developing DILI from various antibiotics, immunosuppressants, and hypolipidemics. An improved awareness of the potential for DILI in the LT setting will hopefully lead to earlier discontinuation of the suspect drug and help minimize allograft injury.

Supplementary Material

Supp 1
Supp 2
Supp 3

KEY POINTS.

  • Boceprevir and telaprevir based antiviral therapy are associated with improved response rates in liver transplant (LT) recipients with HCV infection compared to historical controls but side effects including anemia are also more frequent and potentially severe.

  • Clinically significant drug interactions of boceprevir and telaprevir with the calcineurin inhibitors mandate empiric dose reductions and frequent immunosuppressant blood level monitoring during and after treatment to prevent toxicity and subtherapeutic dosing/rejection, respectively.

  • The protease inhibitors, simeprevir and faldeprevir, as well as daclatasvir and sofosbuvir based antiviral therapy will likely be associated with improved antiviral response rates in LT recipients as well as fewer side effects; studies to identify the optimal agent(s) and duration of therapy are needed.

  • Liver transplant recipients appear to be at increased risk of developing drug-induced liver injury (DILI) from a multitude of agents with antibiotics, immunosuppressants and hypolipidemic agents most frequently implicated.

  • Differentiating DILI from other causes of allograft dysfunction is diagnostically challenging but important so that the suspect drug can be promptly discontinued.

Acknowledgments

Financial support: Dr. Fontana is a NIH funded investigator with research support as a principal investigator in the Drug Induced Liver Injury Network (2U01-DK065184-06) and the Acute Liver Failure Study Group (DK-U-01-58369).

Abbreviations

ALT

Alanine aminotransferase

AST

Aspartate aminotransferase

AUC

Area under the curve

BOC

Boceprevir

BSEP

Bile salt export pump

Cmax

Maximum concentration

CNI

Calcineurin inhibitors

CYP

Cytochrome- P450

DAA

Direct acting antivirals

DDI

Drug-drug interaction

DILI

Drug induced liver injury

DILIN

Drug induced liver injury network

FCH

Fibrosing cholestatic hepatitis

HCV

Hepatitis C virus

HDS

Herbal and dietary supplements

LT

Liver transplantation

mTORi

Mammalian target of rapamycin inhibitors

OATP

Organic anion transporting polypeptide

PEG-IFN

peg-interferon

P-gp

P-glycoprotein

RBV

Ribavirin

SVR

Sustained virological response

TB

Tuberculosis

TMP-SMZ

Trimethoprim-sulfamethoxasole

TPV

Telaprevir

Footnotes

Conflicts of interest: Dr Fontana has received research support from Gilead Sciences and Vertex pharmaceuticals. He has also served as a consultant to Tibotec, GlaxoSmithkline, and Merck in the past year.

References

  • 1.Zhou S, Yung Chan S, Cher Goh B, Chan E, Duan W, Huang M, et al. Mechanism-based inhibition of cytochrome P450 3A4 therapeutic drugs. Clin Pharmacokinet. 2005;44:279–304. doi: 10.2165/00003088-200544030-00005. [DOI] [PubMed] [Google Scholar]
  • 2.Zhou S, Chen E, Lim LY, Boelsterli UA, Li SC, Want J, et al. Therapeutic drugs that behave as mechanism-based inhibitor of cytochrome P450 3A4. Curr Drug Metabolism. 2004;5:415–442. doi: 10.2174/1389200043335450. [DOI] [PubMed] [Google Scholar]
  • 3.Flockart DA, Tanus-Santos JE. Implications of cytochrome P540 interactions when prescribing medications for hypertension. Arch Intern Med. 200262:1. 405–412. doi: 10.1001/archinte.162.4.405. [DOI] [PubMed] [Google Scholar]
  • 4.Knops K, Levthchenko E, van den Heuvel B, Kuypers D. From gut to kidney: Transporting and metabolizing calcineurin-inhibitors in solid organ transplantation. International Journal of pharmaceutics. 2013;452:14–35. doi: 10.1016/j.ijpharm.2013.05.033. [DOI] [PubMed] [Google Scholar]
  • 5.Srinivas TR, Meier-Kriesche Hu, Kaplan B. Pharmacokinetic principles of immunosuppressive drugs. Am J Transplant. 2005;5:207–217. doi: 10.1111/j.1600-6143.2005.00748.x. [DOI] [PubMed] [Google Scholar]
  • 6.Yu S, Wu L, Yan S, Jiang G, Xie H, Zheng S. Influence of CYP3A5 gene polymorphisms of donor rather than recipient to tacrolimus individual dose requirement in liver transplantation. Transplantation. 2006;81:46–51. doi: 10.1097/01.tp.0000188118.34633.bf. [DOI] [PubMed] [Google Scholar]
  • 7.Saad AH, DePestel DD, Carver PL. Factors influencing the magnitude and clinical significance of drug interactions between azole antifungals and select immunosuppressants. Pharmacotherapy. 2006;26:1730–44. doi: 10.1592/phco.26.12.1730. [DOI] [PubMed] [Google Scholar]
  • 8.Herbert MF, Park JM, Chen YL, Akhtar S, Larson AM. Effects of St. John’s wort (9hypericum perforatum) on tacrolimus pharmacokinetics in healthy volunteers. J Clin Pharmacol. 2004;44:89–94. doi: 10.1177/0091270003261078. [DOI] [PubMed] [Google Scholar]
  • 9.Burton JR, Everson GT. Management of the transplant recipient with hepatitis C. Clin Liver Dis. 2013;17:73–91. doi: 10.1016/j.cld.2012.09.013. [DOI] [PubMed] [Google Scholar]
  • 10.Gane EJ, Naoumov NV, Qian KP, Mondelli MU, Maertens G, Portmann BC, et al. A longitudinal analysis of hepatitis C virus replication following liver transplantation. Gastroenterology. 1996;110:167–177. doi: 10.1053/gast.1996.v110.pm8536853. [DOI] [PubMed] [Google Scholar]
  • 11.Berenguer M, Ferrell L, Watson J, Prieto M, Kim M, Rayon M, et al. HCV related fibrosis progression following liver transplantation: increase in recent years. J Hepatology. 2000;32:674–684. doi: 10.1016/s0168-8278(00)80231-7. [DOI] [PubMed] [Google Scholar]
  • 12.Guillouche P, Feray C. Systematic review: anti-viral therapy of recurrent hepatitis C after liver transplantation. Aliment Pharmacol Ther. 2011;33:163–174. doi: 10.1111/j.1365-2036.2010.04505.x. [DOI] [PubMed] [Google Scholar]
  • 13.Berenguer M, Aguilera V, Rubin A, Ortiz C, Jimenez M, Prieto M. Comparison of two non-contemperaneous HCV-liver transplant cohorts: strategies to improve the efficacy of antiviral therapy. J Hepatology. 2012;56:13101316. doi: 10.1016/j.jhep.2011.12.031. [DOI] [PubMed] [Google Scholar]
  • 14.Sharma P, Marrero JA, Fontana RJ, Greenson JK, Conjeevaram H, Su GL, et al. Sustained virologic response to therapy of recurrent hepatitis C after liver transplantation is related to early virologic response and dose adherence. Liver Transpl. 2007;13:1100–1108. doi: 10.1002/lt.21121. [DOI] [PubMed] [Google Scholar]
  • 15.Levitsky J, Fiel MI, Norvell JP, Wang E, Watt KD, Curry MP, et al. Risk for Immune-mediated Graft Dysfunction in liver transplant recipients with recurrent HCV infection treated with pegylated Interferon. Gastroenterology. 2012;142:1132–1139. doi: 10.1053/j.gastro.2012.01.030. [DOI] [PubMed] [Google Scholar]
  • 16.Selzner N, Guindi M, Renner EL, Berenguer M. Immune-mediated complications of the graft in interferon-treated hepatitis C positive liver transplant recipients. J Hepatol. 2011;55:207–217. doi: 10.1016/j.jhep.2010.11.012. [DOI] [PubMed] [Google Scholar]
  • 17.Sharma P, Hosmer A, Appelman H, McKenna B, Jafri M, Sullivan P, et al. Immunological dysfunction during or after antiviral therapy for Recurrent hepatitis C reduces graft survival. Hepatology International. doi: 10.1007/s12072-013-9436-1. accepted March 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Picciotto FP, Tritto G, Lanza AG, Addaario L, De Luca M, Di Costanzo GG, et al. Sustained virological response to antiviral therapy reduces mortality in HCV reinfection after liver transplantation. J Hepatol. 2007;46:459–465. doi: 10.1016/j.jhep.2006.10.017. [DOI] [PubMed] [Google Scholar]
  • 19.Veldt BJ, Poterucha JJ, Watt KDS, Wiesner RH, Hay JE, Kremers WK, et al. Impact of pegylated interferon and ribavirin treatment on graft survival in liver transplant patients with recurrent HCV infection. Am J Transpl. 2008;8:1–8. doi: 10.1111/j.1600-6143.2008.02362.x. [DOI] [PubMed] [Google Scholar]
  • 20.Jacobson IM, McHutchison JG, Dusheiko G, DiBisceglie AM, Reddy R, Bzowej NH, et al. Telaprevir for previously untreated chronic hepatitis C virus infection. N Engl J of Med. 2011;364:2405–2416. doi: 10.1056/NEJMoa1012912. [DOI] [PubMed] [Google Scholar]
  • 21.Poordad F, McCone J, Bacon BR, Bruno S, Manns MP, Sulkowski MS, et al. Boceprevir for untreated chronic HCV genotype 1 infection. N Engl J Med. 2011;364:1195–1206. doi: 10.1056/NEJMoa1010494. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Ghany MG, Nelson DR, Strader DB, Thomas DL, Seeff LB. An update on Treatment of Genotype 1 chronic Hepatitis C virus infection: 2011 Practice Guideline by the American Association for the Study of Liver Diseases. Hepatology. 2011;54:1434–1444. doi: 10.1002/hep.24641. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Sherman KE, Flamm SL, Afdhal NH, Nelson DR, Sulkowski MS, Everson GT, et al. Response-guided telaprevir combination treatment for hepatitis virus infection. N Engl J of Med. 2011;365:1014–1024. doi: 10.1056/NEJMoa1014463. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Bacon BR, Gordon SC, Lawitz E, Marcellin P, Vierling JM, Zeuzem S, et al. Boceprevir for previously treated chronic HCV genotype 1 infection. N Engl J of Med. 2011;364:1207–1217. doi: 10.1056/NEJMoa1009482. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Zeuzem S, Andreone P, Pol S, Lawitz E, Diago M, Roberts S, et al. Telaprevir for retreatment of HCV infection. N Engl J of Med. 2011;364:2417–2428. doi: 10.1056/NEJMoa1013086. [DOI] [PubMed] [Google Scholar]
  • 26.Boceprevir [package insert] Whaitehouse Station, NJ: Merck Laboratories; 2013. [Google Scholar]
  • 27.Telaprevir [package insert] Boston, MA: Vertex Pharmaceuticals; 2011. [Google Scholar]
  • 28.McCaughan GW. New Therapies against HCV: Expected risks and challenges associated with their use in the liver transplant setting. J Hepatology. 2012;57:1361–1367. doi: 10.1016/j.jhep.2012.07.035. [DOI] [PubMed] [Google Scholar]
  • 29.Wilby J, Grenya E, Ford, et al. A review of drug interaction with boceprevir and telaprevir: implications for HIV and transplant patients. Ann Hepatol. 2012;11:179–85. [PubMed] [Google Scholar]
  • 30.Kiser JJ, Burton JR, Anderson PL, Everson GT. Review and management of drug interaction with boceprevir and telaprevir. Hepatology. 2012;55:1620–1628. doi: 10.1002/hep.25653. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Burger D, Back D, Buggisch P, Buti M, Craxi A, Foster G, Klinker H, et al. Clinical management of drug-drug interactions in HCV therapy: challenges and solutions. J Hepatol. 2013;58:792–800. doi: 10.1016/j.jhep.2012.10.027. [DOI] [PubMed] [Google Scholar]
  • 32.Kiser JJ, Burton JR, Everson GT. Drug-drug interactions during antiviral therapy for chronic hepatitis C. Nat Rev Gastroenterol Hepatol. 2013 doi: 10.1038/nrgastro.2013.10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Lee JE, van Heeswijk R, Alves K, Smith F, Garg V. Effect of the hepatitis C virus protease inhibitor telaprevir on the pharmacokinetics of amlodipine and atorvastatin. Antimicrob Agents Chemothera. 2011;55:4569–4574. doi: 10.1128/AAC.00653-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Rangnekar AS, Fontana RJ. Managing Drug-drug interactions with Boceprevir and Telaprevir. Clin Liv Dis. 2012;1:35–40. doi: 10.1002/cld.10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Huloskotte EG, Feng HP, Xuan F, Gupta S, Van Zutven MG, O’Mara, Wagner JA, et al. Pharmacokinetic evaluation of the interaction between the hepatitis C virus protease inhibitor boceprevir and the 3-hydroxy-3-methylguaryl coenzyme A reductase inhibitors atorvastatin and pravastatin. Antimicrob Agents Chemother. 2013;57:2582–2588. doi: 10.1128/AAC.02347-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Garg V, Chandorkar G, Farmer F, Smith F, Alves K, Rolf PG, van Heeswijk R. Effect of telaprevir on the pharmacokinetics of midazolam and digoxin. J Clin Pharmcol. 2012;52:1566–1573. doi: 10.1177/0091270011419850. [DOI] [PubMed] [Google Scholar]
  • 37.Garg V, van Heeswijk R, Yang Y, Kaufman R, Smith F, Adda N. The pharmacokinetic interaction between oral contraceptives containing ethinyl estradiol and norethindrone and the HCV protease inhibitor telaprevir. J Clin Pharmacol. 2012;52:1574–83. doi: 10.1177/0091270011419855. [DOI] [PubMed] [Google Scholar]
  • 38.Reddy KR, Everson GT. Treatment of chronic hepatitis C with protease inhibitor-based therapy after liver transplantation. Hepatology. 2013;58:1181–1183. doi: 10.1002/hep.26612. [DOI] [PubMed] [Google Scholar]
  • 39.Hulskotte E, Gupta S, Xuan F, van Zutven M, O’Mara E, Feng HP, Wagner J, Butterton J. Pharmacokinetic interaction between the hepatitis C virus and protease inhibitor boceprevir and cyclosporine and tacrolimus in health volunteers. Hepatology. 2012;56:1622–1630. doi: 10.1002/hep.25831. [DOI] [PubMed] [Google Scholar]
  • 40.Garg V, van Heeswijk R, Lee JE, Alves K, Nakarini P, Luo X. Effect of telaprevir on the pharmacokinetics of cyclosporine and tacrolimus. Heptology. 2011;54:20–27. doi: 10.1002/hep.24443. [DOI] [PubMed] [Google Scholar]
  • 41.Hulskotte EG, Feng HP, Xuan F, Ling WH, Zhu T, Rasmussen S, Butteron RJ. Pharmacokinetics interactions between the HCV protease inhibitor boceprevir and sirolimus in healthy subject (Abstract 463) J Hepatology. 2013;58S:S190. [Google Scholar]
  • 42.Coilly A, Furlan V, Roche B, Barau C, Noel C, Bonhomme-Faivre, Antonini TM, et al. Practical management of boceprevir and immunosuppressive therapy in liver transplant recipients with hepatitis C virus recurrence. Antimicrob Agents Chemother. 2012;56:5728–34. doi: 10.1128/AAC.01151-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Vargas H, Dai Y, Brown K, Russo M, Yoshida E, Fontana R, Levitsky J, et al. Twice daily telaprevir in combination with peginterferon alfa-2a/ribavirin in HCV genotype 1 liver transplant recipients: interim pharmacokinetics of the REFERSH study (Abstract) Am J Transplant. 2013;S5:347. [Google Scholar]
  • 44.O’Leary JG, McKenna GJ, Klintlmalm GB, Davis GL. Effect of telaprevir on the pharmacokinetics of sirolimus in liver transplant recipients. Liver Transpl. 2013;19:463–5. doi: 10.1002/lt.23623. [DOI] [PubMed] [Google Scholar]
  • 45.Jumes P, Feng HP, Chatterjee M, Xuan F, Connolly SM, Wagner JA, Butterton JR. Pharmacokinetic interaction between the HCV protease inhibitor boceprevir and prednisone in healthy volunteers (Abstract) Hepatology. 2012;56:1076A. doi: 10.1002/hep.25831. [DOI] [PubMed] [Google Scholar]
  • 46.Pungpapong S, Aqel BA, Koning L, Murphy JL, Henry TM, Ryland KL, Yataco ML, et al. Multcenter experience using telaprevir or boceprevir with peginterferon and ribavirin to treat hepatitis C genotype 1 after liver transplantation. Liver Transpl. 2013;19:690–700. doi: 10.1002/lt.23669. [DOI] [PubMed] [Google Scholar]
  • 47.Coily A, Roche B, Dumortier J, Leroy V, Botta-Fridlund D, Sylvie R, et al. Safety and efficacy of protease inhibitors to treat hepatitis C after Liver transplantation: A multicenter experience. J Hepatology. 2013 doi: 10.1016/j.jhep.2013.08.018. (Accepted). doi: http://dx.doi.org/10.1016/j.jhep.2013.08.018. [DOI] [PubMed]
  • 48.Brown R, Russo M, Levitsky J, Bronw K, Fontana R, Vargas H, Yoshida E, Bsharat M, Rubrin R. Twice daily telaprevir in combination with peginterferon alfa-2a/ribavirin in HCV genotype 1 liver transplant recipients:interim safety/efficacy of the REFRESH study (Abstract) Am J Transplant. 2013;S5:145. [Google Scholar]
  • 49.Werner CR, Egetmeyr DP, Lauer UM, Nadalin S, Konigsrainer A, Malek NP, Berg CP. Telaprevir-based triple therapy in liver transplant patients with hepatitis C virus: A 12-week pilot study providing safety and efficacy data. Liver Transpl. 2012;18:1464–1470. doi: 10.1002/lt.23542. [DOI] [PubMed] [Google Scholar]
  • 50.De Oliverira Pereira AP, Shin HJ, Safdar A, Tobias H, Gelb B, Morgan G, Diflo A, Winnick L, et al. Post liver transplant therapy with telaprevir for recurrent hepatitis C (Abstract) Am J Transplant. 2012;12(s3):1369. [Google Scholar]
  • 51.Burton G, Everson T. Initial experience with telaprevir for treating hepatitis C virus in liver transplant recipients: Virologic response, safety and tolerability (Abstract) Am J Transplant. 2012;12(s3):LB01. [Google Scholar]
  • 52.O’Leary J, Verna E, Burton J, Lai J, Saxena VJ, Levistky J, Dodge J, et al. A high rate of eRVR with protease inhibitor-triple HCV therapy in liver transplant recipients: a multicenter study from CRUSH-C (Abstract) Am J Transplant. 2013;S5:32. [Google Scholar]
  • 53.Oo YH, Mutimer DJ. Rapid recovery of cytochrome P450 3A4 after protease inhibitor withdrawal in post-liver transplant patients. Liver Transpl. 2012;18:1264–5. doi: 10.1002/lt.23496. [DOI] [PubMed] [Google Scholar]
  • 54.Nair SP. Protease inhibitor therapy post-liver transplantation in the treatment of hepatitis c virus infection. Gastroenterol Hepatol. 2013;9:368–390. [PMC free article] [PubMed] [Google Scholar]
  • 55.Coilly A, Roche B, Samuel D. Current management and perspectives for HCV recurrent after liver transplantation. Liver International. 2013;33:56–62. doi: 10.1111/liv.12062. [DOI] [PubMed] [Google Scholar]
  • 56.Mauss S, Hueppe D, Alshuth U. Renal impairment is frequent in chronic hepatitis C patients under triple therapy with telaprevir or boceprevir. Hepatology. 2013 doi: 10.1001/hep26602. [DOI] [PubMed] [Google Scholar]
  • 57.Kunze A, Huwyler J, Camenisch G, Gutmann H. Interaction of the antiviral drug telaprevir with renal and hepatic drug transporters. Biochem Pharmacol. 2012;84:1096–1102. doi: 10.1016/j.bcp.2012.07.032. [DOI] [PubMed] [Google Scholar]
  • 58.Herold C. Quantitative testing of liver function in relation to fibrosis in patients with chronic hepatitis B and C. Liver. 212001:260–65. doi: 10.1034/j.1600-0676.2001.021004260.x. [DOI] [PubMed] [Google Scholar]
  • 59.Frey RF. Liver disease selectively modulates cytochrome P450-mediated metabolism. Clin Pharmacol Ther. 2006;80:235–245. doi: 10.1016/j.clpt.2006.05.006. [DOI] [PubMed] [Google Scholar]
  • 60.Kugelmas M, et al. Hepatitis C virus therapy, hepatocyte drug metabolism, and risk for acute cellular rejection. Liver Transpl. 2003;9:1159–1165. doi: 10.1053/jlts.2003.50233. [DOI] [PubMed] [Google Scholar]
  • 61.Poordad F, Lawitz E, Kowdley KV, Cohen DE, Podsadecki T, Siggelkow S, Heckaman M, et al. Exploratory study of oral combination antiviral therapy for hepatitis C. NEJM. 2013;368:45–53. doi: 10.1056/NEJMoa1208809. [DOI] [PubMed] [Google Scholar]
  • 62.Kowdley KV, Lawitz E, Poordad F, Cohen DE, Nelson DR, Zeuzem S, et al. A 12 week interferon-free treatment regimen with ABT-450/R, ABT-267, ABT-333, and ribavirin achieves SVR12 rates of 99% in treatment naïve patients and 93% in prior null-responders with HCV genotype 1 infection (Abstract) Hepatology. 2012;56:1515A. [Google Scholar]
  • 63.Lange CM, Zeuzem S. Perspectives and challenges of interferon-free therapy for chronic hepatitis C. J Hepatol. 2013;58:583–592. doi: 10.1016/j.jhep.2012.10.019. [DOI] [PubMed] [Google Scholar]
  • 64.Manns M, Marcellin P, Poordad F, Stanislau Alfonso de Araujo E, Buti M, Horsmans Y, Ewa J, et al. Simeprevir (TMC435) with peginterferon/ribavirin for treatment of chronic HCV genotype 1 infection in treatment-naive patients: results from QUEST-2 a phase III trial (Abstract) J Hepatol. 2013;58S:S568. [Google Scholar]
  • 65.Jacobson I, Dore GJ, Foster GR, Fried MN, Radi Mi, Rafalskiy VV, et al. Simeprevir (TMC435) with peginterferon/ribavirin for treatment of chronic HCV genotype 1 infection in treatment-naïve patients: results from QUEST-1 a phase III trial (Abstract) J Hepatol. 2013;58S:S574. [Google Scholar]
  • 66.Sulkowski MS, Asselah T, Lalezari J, Ferenci P, Fainbohm H, Leggett B, Bessone F, et al. Faldaprevir combined with peginterferon alfa-2a and ribavirin in chronic hepatitis C virus genotype1 HCV: SILEN-C1 trial. Hepatology. 2013;57:2143–2154. doi: 10.1002/hep.26276. [DOI] [PubMed] [Google Scholar]
  • 67.Sulkowski MS, Bourliere M, Bronowicki JP, Asselah T, Pawlostsky JM, Sharfran SD, Pol S, et al. Faldaprevir combined with peginterferon alfa-2a and ribavirin in chronic hepatitis C virus genotype-1 patients with prior nonresponse: SILEN-C2 Trial. Hepatology. 2013;57:2155–2163. doi: 10.1002/hep.26386. [DOI] [PubMed] [Google Scholar]
  • 68.Kowdley KV, Lawitz E, Crespo I, Hassanien T, Davis MN, DeMicco M, Bernstein D, et al. Sofosbuvir with pegylated interferon alfa-2 a and ribavirin for treatment-naïve patients with hepatitis C genotype 1 (ATOMIC): an open-label, randomized, multicenter phase 2 trial. Lancet. 2013:381-2100-07. doi: 10.1016/S0140-6736(13)60247-0. [DOI] [PubMed] [Google Scholar]
  • 69.Pol S, Ghalib RH, Rustgi VK, Martorell C, Everson GT, Tatum HA, Hezode C, et al. Daclatasvir for previously untreated chronic hepatitis C genotype-1 infection: a randomized, parallel-group, double-placebo, placebo-controlled, dose-finding, phase 2a trial. Lancet Infect Dis. 2012;12:671–7. doi: 10.1016/S1473-3099(12)70138-X. [DOI] [PubMed] [Google Scholar]
  • 70.Fried MW, Dore GJ, Flisiak R, Ferenci P, Jacobson I, Marcellin P, Manns M, et al. Once- daily simeprevir (TMC 435) with pegylated intereferon and ribavirin in treatment-naïve genoytipe I hepatitis C: The randomized PILLAR study [epub ahead of print] Hepatology. 2013 doi: 10.1002/hep.26641. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Zeuzem S, Soriano V, Asselah T, Bronowicki JP, Lohse AW, Mullhaupts B, et al. Faldaprevir and Deleobuvir for HCV genotype 1 infection. N Engl J Med. 2013;369:630–639. doi: 10.1056/NEJMoa1213557. [DOI] [PubMed] [Google Scholar]
  • 72.Lok AS, Gardiner DF, Lawitz E, Martorell C, Everson GT, Ghalib R, Reindollar R, et al. Preliminary study of two antiviral agents for hepatitis C Genotype 1. N Engl J Med. 2012;366:216–224. doi: 10.1056/NEJMoa1104430. [DOI] [PubMed] [Google Scholar]
  • 73.Feld JJ, et al. Up to 100% SVR4 rates with ritonavir-boosted danoprevir (DNRr), mercititabine (MCB), and ribavirin (R) ± peginterferon alfa-2a (40KD) (P) in HCV genotype 1-infected partial and null responders: results from MATTERHORN study (Abstract] Hepatology. 2012;56:231A. [Google Scholar]
  • 74.Lawitz E, Mangia A, Wyles D, Rodriguez-Torres M, Hassanein T, Gordon SC, Schultz M, et al. Sofosbuvir for previously untreated chronic hepatitis C infection. N Engl J Med. 2013;368:1878–1887. doi: 10.1056/NEJMoa1214853. [DOI] [PubMed] [Google Scholar]
  • 75.Jacobson IM, Gordon SC, Kowdley KV, et al. Sofosbuvir for hepatitis C genotype 2 or 3 in patients without treatment options. N Engl J Med. 2013;368:1867–1877. doi: 10.1056/NEJMoa1214854. [DOI] [PubMed] [Google Scholar]
  • 76.Thompson A, Han S, Shiffman ML, Rossaro L, Ghalib R, Beavers K, et al. GS-5885 (Ledipasvir) + GS-9451 + peginterferon and ribavirin for 6 or 12 weeks achieves high SVR12 rates in treatment naïve genotype 1 IL28B CC patients (Abstract) J Hepatol. 2013;58S:S29. [Google Scholar]
  • 77.Lawitz E, Gruener D, Hill JM, Marbury T, Komjathy S, DeMicco M, Murillo, et al. A phase I, randomized, placebo controlled, 3-day dose –ranging study of GS-5885: an NS5A inhibitor, in patients with genotype 1 hepatitis C. J Hepatol. 2012;57:24–31. doi: 10.1016/j.jhep.2011.12.029. [DOI] [PubMed] [Google Scholar]
  • 78.Ouwerkerk-Mahadevan S, Simion A, Mortier S, Peeters M, Beumont M. No clinically significant interaction between the investigational HCV protease inhibitor, TMC435 and the immunosuppressives cyclosporine and tacrolimus (Abstract) Hepatology. 2012;58:213A. [Google Scholar]
  • 79.Kowdley KV, Lawitz E, Poordad F, Cohen DE, Nelson DR, Zeuzem S, et al. A 12 week interferon-free treatment regimen with ABT-450/R, ABT-267, ABT-333, and ribavirin achieves SVR12 rates of 99% in treatment naïve patients and 93% in prior null-responders with HCV genotype 1 infection (Abstract) J Hepatology. 2012;56S:S478. [Google Scholar]
  • 80.Sane R, Podila L, Mathur A, Mease K, Taub M, et al. Mechanisms of isolated unconjugated hyperbilirubinemia induced by the HCV NS3/4A protease inhibitor, BI201335 (Abstract) J Hepatol. 2011;54(Suppl 1):S488. [Google Scholar]
  • 81.Huisman MT, Snoeys J, Monbaliu J, Martens M, Sekar V, Raoof A. In vitro studies investigating the mechanism of interaction between TMC435 and hepatic transporters (Abstract) Hepatology. 2010;52:461A. [Google Scholar]
  • 82.Fontana RJ, Hughes EA, Appelman H, Hindes R, Dimitrova D, Bifano M. Case report of successful peginterferon, ribavirin, and daclatasvir therapy for recurrent cholestatic hepatitis C after liver transplantation. Liver Transpl. 2012;18:1053–1059. doi: 10.1002/lt.23482. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Fontana RJ, Hughes EA, Bifano M, Appelman H, Dimitrova D, Hindes R, Symonds WT. Sofosbuvir and daclatasvir combination therapy in a liver transplant recipient with severe recurrent cholestatic hepatitis C. Am J Transpl. 2013;13:1601–1605. doi: 10.1111/ajt.12209. [DOI] [PubMed] [Google Scholar]
  • 84.Study to investigate GS-7977 and ribavirin for 24 weeks in subjects with recurrent chronic HCV post liver transplant. Clinicaltrials.gov: NCT01687270. Accessed September 1st, 2013. [Google Scholar]
  • 85.Sekar V, Verloes R, Meyvisch P, Spittaels K, Akuma SH, DeSmedt DG. TMC435 and Drug interactions: Evaluation of the metabolic interactions of TMC435 via cytochrome P450 (CYP) enzymes in health volunteers (abstract) J Hepatol. 2010;52:S416. [Google Scholar]
  • 86.Ouwerkerk-Mahadevan S, Simon A, Mortier S, Peeters M, Beumont M. No clinically significant interaction between the investigational HCV protease inhibitor, TMC435 and the immunosuppressives cyclosporine and tacrolimus (Abstract) Hepatology. 2012;56:213A. [Google Scholar]
  • 87.Manns MP, Bourliere M, Benhamou Y, et al. Potency, safety, and pharmacokinetics of the NS3/4A protease inhibitor BI210335 in patients with chronic HCV genotype 1 infection. J Hepatol. 2011;54:1114–1122. doi: 10.1016/j.jhep.2010.08.040. [DOI] [PubMed] [Google Scholar]
  • 88.Dumas E, Lawal A, Menon R, Podsadecki T, Awni W, Dutta S, Williams L. Pharmacokinetics, safety and tolerability of the HCV NS5A inhibitor ABT-267 following single and multiple doses in healthy adult volunteers (Abstract 1024) J Hepatol. 2011;54:S475. [Google Scholar]
  • 89.Maring C, Wagner R, Hutchinsom D, Flentge C, Kati W, et al. Preclinical potency and ADME characterization of ABT-333, a novel non-nucleoside HCV polymerase inhibitor (Abstract) J Hepatol. 2009;50:S346–S347. [Google Scholar]
  • 90.Fontana RJ, Seeff LB, Andrade RJ, Bjornsson E, Day CP, Serrano J, Hoofnagle JH. Standardization of nomenclature and causality assessment in Drug-Induced liver Injury: Summary of a clinical Research workshop. Hepatology. 2010;52:730–742. doi: 10.1002/hep.23696. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Molleston JP, Fontana RJ, Lopez MJ, Kleiner DE, Gu J, Chalasani N. Characteristics of idiosyncratic drug-induced liver injury in children: Results from the DILIN Prospective study. JPGN. 2011;53:182–189. doi: 10.1097/MPG.0b013e31821d6cfd. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Ostapowicz G, Fontana RJ, Schiodt FV, et al. Results of a prospective study of acute liver failure at 17 tertiary care centers in the United States. Ann Intern Med. 2002;137:947–954. doi: 10.7326/0003-4819-137-12-200212170-00007. [DOI] [PubMed] [Google Scholar]
  • 93.Reuben A, Koch DG, Lee WM. Drug induced acute liver failure: Results of a US multicenter, prospective study. Hepatology. 2010;52:2065–2076. doi: 10.1002/hep.23937. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Aithal PG, Day CP. The natural history of histologically proved drug induced liver disease. Gut. 1999;44:731–735. doi: 10.1136/gut.44.5.731. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Bjornsson E, Davidsdottir L. The long-term follow-up after idiosyncratic drug-induced liver injury with jaundice. J Hepatol. 2009;50:511–517. doi: 10.1016/j.jhep.2008.10.021. [DOI] [PubMed] [Google Scholar]
  • 96.De Valle MB, Klinteberg AV, Alem N, Olsson R, Björnsson E. Drug-induced liver injury in a Swedish University hospital out-patient hepatology clinic. Aliment Pharmacol Ther. 2006;24:1187–95. doi: 10.1111/j.1365-2036.2006.03117.x. [DOI] [PubMed] [Google Scholar]
  • 97.Galan MV, Potts JA, Silverman AL, Gordon SC. The burden of acute non-fulminant drug-induced hepatitis in a United States tertiary referral center. J Clin Gastro. 2005;39:64–67. [PubMed] [Google Scholar]
  • 98.Sgro C, Clinard F, Quazir K, Chanay H, Allard C, Guilleminet C, et al. Incidence of drug-induced hepatic injuries: A French population-based study. Hepatology. 2002;36:451–455. doi: 10.1053/jhep.2002.34857. [DOI] [PubMed] [Google Scholar]
  • 99.Bjornsson ES, Bergmann OM, Bjornsson HK, Kvaran RB, Olafsson S. Incidence, presentation, and outcomes in patients with drug-induced liver injury in the general population of Iceland. Gastroenterology. 2013;144:1419–1425. doi: 10.1053/j.gastro.2013.02.006. [DOI] [PubMed] [Google Scholar]
  • 100.Chalasani N, Fontana RJ, Bonkovsky HL, Watkins PB, Davern T, Serrano J, et al. Causes, clinical features, and outcomes from a prospective study of drug induced liver injury in the United States. Gastroenterology. 2008;135:1924–1934. doi: 10.1053/j.gastro.2008.09.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Andrade RJ, Lucena MI, Fernandez MC, et al. Drug-induced liver injury: an analysis of 461 incidences submitted to the Spanish registry over a 10-year period. Gastroenterology. 2005;129:512–521. doi: 10.1016/j.gastro.2005.05.006. [DOI] [PubMed] [Google Scholar]
  • 102.Takikawa H, Murata Y, Horiike N, Fukui H, Onji M. Drug-induced liver injury in Japan: an analysis of 1676 cases between 1997 and 2006. Hepatology Res. 2009;39:427–431. doi: 10.1111/j.1872-034X.2008.00486.x. [DOI] [PubMed] [Google Scholar]
  • 103.Agarwal VK, Mchutchison MG, Hoofnagle JH. Important elements for the diagnosis of drug induced liver injury. Clin Gastro and Hep. 2010;8:463–470. doi: 10.1016/j.cgh.2010.02.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Danan G, Benichou C. Causality assessment of adverse reactions to drugs-I. A novel method based on the conclusions of International Consensus Meetings: application to drug-induced liver injuries. J Clin Epidemiol. 1993;46:1323–30. doi: 10.1016/0895-4356(93)90101-6. [DOI] [PubMed] [Google Scholar]
  • 105.Rochon J, Protiva P, Seeff LB, Fontana RJ, Liangpunsakul S, Watkins PB, et al. Reliability of the RUCAM for assessing causality in drug-induced liver injury. Hepatology. 2008;48:1175–1183. doi: 10.1002/hep.22442. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Rockey DC, Seeff LB, Rochon J, Chalasani N, Bonacini M, Fontana RJ, et al. Comparison between expert opinion and RUCAM for assignment of causality in drug-induced liver injury. Hepatology. 2010;51:2117–2126. doi: 10.1002/hep.23577. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.LiverTox website accessed on September 30th, 2013. http://www.livertox.nih.gov/
  • 108.Fontana RJ, Watkins PB, Bonkovsky HL, Chalasani N, Davern T, Serrano J, et al. Rationale, design and conduct of the Drug Induced Liver Injury Network prospective study. Drug Saf. 2009;32:55–68. doi: 10.2165/00002018-200932010-00005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Wiesner RH, Demetris AJ, Belle SH, et al. Acute hepatic allograft rejection: incidence, risk factors, and impact on outcome. Hepatology. 1998;28:638–645. doi: 10.1002/hep.510280306. [DOI] [PubMed] [Google Scholar]
  • 110.Neuberger J. Recurrent primary biliary cirrhosis. Liver Transpl. 2003;9:539–544. doi: 10.1053/jlts.2003.50096. [DOI] [PubMed] [Google Scholar]
  • 111.Heneghan MA, Portmann BC, Norris SM, Williams R, Muiseauan P, Rela M, et al. Graft dysfunction mimicking autoimmune hepatitis following liver transplantation in adults. Hepatology. 2001;34:464–470. doi: 10.1053/jhep.2001.26756. [DOI] [PubMed] [Google Scholar]
  • 112.Guido M, Burra P. De novo autoimmune hepatitis after liver Transplantation. Sem Liv Dis. 2011;31:71–81. doi: 10.1055/s-0031-1272834. [DOI] [PubMed] [Google Scholar]
  • 113.Davern TJ, Chalasani N, Fontana RJ, Hayashi PH, Protiva P, Kleiner DE, Engle RE, Nguyen H, Emerson SU, Purcell RH, Tillmann HL, Gu J, Serrano J, Hoofnagle JH, for the Drug Induced Liver Injury Network Role of Acute hepatitis E in Suspected Drug-induced liver injury. Gastroenterology. 2011;141:1665–1672. doi: 10.1053/j.gastro.2011.07.051. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Kamar N, Garrouste C, Haagsma EB, Garrigue V, Pischke S, Chauvet C, eumortier J, et al. Factors associated with chronic hepatitis in patients with hepatitis E virus infection who have received solid organ transplants. Gastroenterology. 2011;140:1481–1489. doi: 10.1053/j.gastro.2011.02.050. [DOI] [PubMed] [Google Scholar]
  • 115.Wong WM, Wu PC, Yuen MF, Cheng CC, Yew WW, Wong PC, et al. Antituberculosis drug-related liver dysfunction in chronic hepatitis B infection. Hepatology. 2000;31:201–206. doi: 10.1002/hep.510310129. [DOI] [PubMed] [Google Scholar]
  • 116.Sulkowski MS, Thomas DL, Chaisson RE, Moore RD. Hepatotoxicity associated with antiretroviral therapy in adults infected with human immunodeficiency virus and the role of hepatitis C or B virus infection. JAMA. 2000;283:74–80. doi: 10.1001/jama.283.1.74. [DOI] [PubMed] [Google Scholar]
  • 117.Zhenglu W, Hui L, Shuying ZH, Wenjuan C, Zhongyang SH. A clinical-pathological analysis of drug-induced hepatic injury after liver transplantation. Trans Proceed. 2007;39:3287–3291. doi: 10.1016/j.transproceed.2007.08.096. [DOI] [PubMed] [Google Scholar]
  • 118.Sembera S, Lammert C, Talwalkar JA, Sanderson SO, Poterucha JJ, Hay JE, et al. Frequency and clinical presentation and outcomes of drug-induced liver injury after liver transplantation. Liver transpl. 2012;18:803–810. doi: 10.1002/lt.23424. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Hanses F, Zierhut S, Scholmerich J, Salzberger B, Wrede CE. Severe and long-lasting cholestasis after high dose co-trimoxazole treatment for pneumocystis pneumonia in HIV-infected paitnets- a report of two cases. Int J Infect Dis. 2009;13:e467–469. doi: 10.1016/j.ijid.2008.12.016. [DOI] [PubMed] [Google Scholar]
  • 120.Legendre C, Caillat-Zucman S, Samuel D, Morelon S, Bismuth H, Bach F, et al. Transfer of symptomatic peanut allergy to the recipient of a combined Liver-And-Kidney Transplant. N Eng J Med. 1997;337:822–825. doi: 10.1056/NEJM199709183371204. [DOI] [PubMed] [Google Scholar]
  • 121.Schwab M, Schaeffeler E, Marx C, Fischer C, Lang T, Behrens C, Gregor M, et al. Azathioprine therapy and adverse drug reactions in patients with inflammatory bowel disease: impact of thiopurine s-methyltransferase polymorphism. Pharmacogenetics. 2002;12:429–436. doi: 10.1097/00008571-200208000-00003. [DOI] [PubMed] [Google Scholar]
  • 122.Horsmans Y, Rahler J, Geubel AP. Reversible cholestasis with bile duct injury following azathioprine therapy. A case report Liver. 1991;11:89–93. doi: 10.1111/j.1600-0676.1991.tb00497.x. [DOI] [PubMed] [Google Scholar]
  • 123.Sterneck M, Wiesner R, Ascher N, Roberts J, Ferrell L, Ludwig J, Lake J. Azathioprine hepatotoxicity after liver transplantation. Hepatology. 1991;14:806–810. doi: 10.1002/hep.1840140511. [DOI] [PubMed] [Google Scholar]
  • 124.Gane E, Portmann B, Sazena R, Wong P, ramage J, Williams R. Nodular regenerative hyperplasia of the liver graft after liver transplantation. Hepatology. 1994;20(1):88–94. doi: 10.1016/0270-9139(94)90138-4. [DOI] [PubMed] [Google Scholar]
  • 125.Kamphues C, Bova R, Rocken C, Neuhaus R, pratschke J, Neuhaus P, Neumann UP. Safety of mycophenolate mofetil monotherapy in patients after liver transplantation. 2009;14:40–46. [PubMed] [Google Scholar]
  • 126.Dourakis SP, Boki K, Soultati A, Cherouvim E, Delladetsima I. Acute hepatitis following mycophenolate mofetil administration for ANCA-positive vasculitis. Scn J Rheumatol. 2007;36:237–9. doi: 10.1080/03009740600844274. [DOI] [PubMed] [Google Scholar]
  • 127.Loupy A, Anglicheau D, Mamzer-Bruneel MF, Martinez F, Thervet E, Legendre C, Serpaggi J, Pol S. Mycophenolate sodium-induced hepatotoxicity: first report. Transplantation. 2006;82:581. doi: 10.1097/01.tp.0000231873.86048.68. [DOI] [PubMed] [Google Scholar]
  • 128.Taniai N, Akimaru K, Ishikawas Y, Kanada T, Kakinuma D, Mizuguchi Y, et al. Hepatotoxicity caused by both tacrolimus and cyclosporine after living donor liver transplantation. J Nihon Med Sch. 2008;75:187–191. doi: 10.1272/jnms.75.187. [DOI] [PubMed] [Google Scholar]
  • 129.Klintmalm GB, Iwatsuki S, Starzl TE. Cyclosporine A hepatotoxicity in 66 renal allograft recipients. Transplantation. 1981;32:488–489. doi: 10.1097/00007890-198112000-00007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Oto T, Okazaki M, Takata K, Egi M, Yamane M, Toyooka S, Sano Y, et al. Calcineurin inhibitor-related cholestasis complicating lung transplantation. Ann Thorac Surg. 2010;89:1664–5. doi: 10.1016/j.athoracsur.2009.09.081. [DOI] [PubMed] [Google Scholar]
  • 131.Lorber ML, Van Buren CT, Flechner SM, Williams C, Kahan BD. Hepatobiliary and pancreatic complications of cyclosporine therapy in 466 renal transplant recipients. Transplantation. 1987;43:35–40. doi: 10.1097/00007890-198701000-00009. [DOI] [PubMed] [Google Scholar]
  • 132.Moran D, De Buitrago JM, Fernandez E, Galan AJ, Munoz ME, Jimenez R. Inhibition of biliary glutathione secretion of cyclosporine A in the rate possible mechanisms and role in the cholestasis induced by the drug. J Hepatol. 1998;29:68–77. doi: 10.1016/s0168-8278(98)80180-3. [DOI] [PubMed] [Google Scholar]
  • 133.Jacques J, Dickson Z, Carrier P, Essiq M, Guillaudeau A, Lacour C, et al. Severe sirolimus-induced acute hepatitis in a renal transplant recipient. Transpl Int. 2010;23:967–970. doi: 10.1111/j.1432-2277.2010.01100.x. [DOI] [PubMed] [Google Scholar]
  • 134.Neff GW, Ruiz P, Madaraiaga JR, Nishida S, Montalbano M, Meyer D, et al. Siroloimus-associated hepatotoxicity in liver transplantation. Ann Pharmacother. 2004;38:1593–1596. doi: 10.1345/aph.1E165. [DOI] [PubMed] [Google Scholar]
  • 135.Chang GJ, Mahanty HD, Quan D, Freise CE, Ascher NL, Roberts JP, et al. Experience with the use of sirolimus in liver transplantation- Use in patients for whom calcineurin inhibitors are contraindicated. Liver Transpl. 2000;6:734–740. doi: 10.1053/jlts.2000.19023. [DOI] [PubMed] [Google Scholar]
  • 136.Studniarz M, Czubkowski P, Cielecka-Kuszyk J, Jankowska I, Teisseyre M, Kaminska D, et al. Amoxicillin/clavulanic acid-induced cholestatic liver injury after pediatric liver transplantation. Ann Transplant. 2012;17:128–131. doi: 10.12659/aot.882646. [DOI] [PubMed] [Google Scholar]
  • 137.Mainra RR, Card SE. Trimethoprim-sulfamethoxasole associated hepatotoxicity- part of a hypersensitivity syndrome. Cn J Clin Pharmacol. 2003;10:175–178. [PubMed] [Google Scholar]
  • 138.Neuman MG, McKinney KK, Nanau RM, Kong V, Malkiewicz I, Mazulli T, et al. Drug induced severe adverse reaction enhanced by human herpes virus-6 reactivation. Transl Res. 2013;161:430–440. doi: 10.1016/j.trsl.2012.12.012. [DOI] [PubMed] [Google Scholar]
  • 139.Bronstein JA, Gros P, Hernandez E, Larroque P, Molinie C. Fatal acute hepatic necrosis due to dose-dependent fluconazole hepatotoxicity. Clin Infect Dis. 1997;25:1266–67. doi: 10.1086/516975. [DOI] [PubMed] [Google Scholar]
  • 140.Cruciani M, Mengoli C, Malena M, Bosco O, Serpelloni G, Grossi P. Antifungal prophylaxis in liver transplant patients: a systematic review and meta-analysis. Liver transpl. 2006;12L:850–855. doi: 10.1002/lt.20690. [DOI] [PubMed] [Google Scholar]
  • 141.Ichai P, Saliba F, Antoun F, Azoulay D, Sebagh M, Antonini TM, Escaut L, et al. Acute liver failure due to anti-tubercular therapy: Strategy for antitubercular treatment before and after liver transplantation. Liver Transplantation. 2010;16:1136–1146. doi: 10.1002/lt.22125. [DOI] [PubMed] [Google Scholar]
  • 142.Jafri M, Singal AG, Kaul D, Fontana RJ. Detection and management of latent tuberculosis in liver transplant patients. Liver Transplantation. 2011;17:306–314. doi: 10.1002/lt.22203. [DOI] [PubMed] [Google Scholar]
  • 143.Rubin RH. Management of tuberculosis in the transplant recipient. Am J Transplant. 2005;5:2599–2600. doi: 10.1111/j.1600-6143.2005.01087.x. [DOI] [PubMed] [Google Scholar]
  • 144.Cvetkovic RS, Wellington K. Valganciclovir: A review of its use in the management of CMV infectin and disease in immunocompromised patients. Drugs. 2005;65:859. doi: 10.2165/00003495-200565060-00012. [DOI] [PubMed] [Google Scholar]
  • 145.Teschke R, Glass X, Schulze J. Herbal hepatotoxicity by greater celandine (Chelidonium majus): Causality assessment of 22 spontaneous reports. Reg Toxicology and Pharm. 2011;61:282–291. doi: 10.1016/j.yrtph.2011.08.008. [DOI] [PubMed] [Google Scholar]
  • 146.Apestegui CA, Julliard O, Ciccarelli O, Duc DK, Lerut J. Energy Drinks: Another red flag for the liver allograft. Liver transplant. 2011;17:1117–1118. doi: 10.1002/lt.22360. [DOI] [PubMed] [Google Scholar]
  • 147.Herden U, Fischer L, Schafer H, Nashan B, Baehr V, Sterneck M. Sorafenib-induced severe acute hepatitis in a stable liver transplant recipient. Transplantation. 2010;90:98–99. doi: 10.1097/TP.0b013e3181daac69. [DOI] [PubMed] [Google Scholar]
  • 148.Von Vital JM, Karaschristos A, Singhal A, Thomas R, Jain A. Acute Amiodarone hepatotoxicity after liver transplantation. Transplantation. 2011;91(8):e62–e64. doi: 10.1097/TP.0b013e3182115bc1. [DOI] [PubMed] [Google Scholar]
  • 149.Ritz Bravo AE, Drewe J, Schlienger RG, et al. Hepatotoxicity during rapid intravenous loading with amiodarone: Description of three cases and review of the literature. Crit Care Med. 2005;33:128. doi: 10.1097/01.ccm.0000151048.72393.44. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supp 1
Supp 2
Supp 3

RESOURCES