Agarwal 2018

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

Research Article JOURNAL

Viral Hepatitis OF HEPATOLOGY

96 weeks treatment of tenofovir alafenamide vs. tenofovir


disoproxil fumarate for hepatitis B virus infection
Kosh Agarwal1,⇑, Maurizia Brunetto2, Wai Kay Seto3, Young-Suk Lim4, Scott Fung5,
Patrick Marcellin6, Sang Hoon Ahn7, Namiki Izumi8, Wan–Long Chuang9, Ho Bae10,
Manoj Sharma11, Harry L.A. Janssen12,13, Calvin Q. Pan14, Mustafa Kemal Çelen15,
Norihiro Furusyo16, Dr. Shalimar17, Ki Tae Yoon18, Huy Trinh19, John F. Flaherty20, Anuj Gaggar20,
Audrey H. Lau20, Andrea L. Cathcart20, Lanjia Lin20, Neeru Bhardwaj20, Vithika Suri20,
G. Mani Subramanian20, Edward J. Gane21, Maria Buti22, Henry L.Y. Chan23,⇑,
and the GS-US-320-0108 Investigators
1
Kings College Hospital, London, United Kingdom; 2University of Pisa, Pisa, Italy; 3University of Hong Kong, Hong Kong; 4Asan Medical Center,
University of Ulsan College of Medicine, Seoul, South Korea; 5Toronto General Hospital, Toronto, ON, Canada; 6Hôpital Beaujon, Clichy, France;
7
Yonsei University, Seoul, South Korea; 8Musashino Red Cross Hospital, Tokyo, Japan; 9Kaohsiung Medical University Hospital, Kaohsiung Medical
University, Kaohsiung, Taiwan; 10Asian Pacific Liver Center, St. Vincent Medical Center, Los Angeles, USA; 11Institute of Liver and Biliary
Sciences, New Delhi, India; 12Toronto Western Hospital, Toronto, ON, Canada; 13Erasmus Medical Center, Rotterdam, The Netherlands;
14
NYU Langone Medical Center, NYU School of Medicine, New York, USA; 15Dicle University Hospital Infectious Diseases, Diyarbakir, Turkey;
16
Kyushu University Hospital, Fukuoka, Japan; 17All India Institute of Medical Sciences, New Delhi, Delhi, India; 18Pusan National University
Yangsan Hospital, Yangsan, South Korea; 19San Jose Gastroenterology, San Jose, USA; 20Gilead Sciences, Foster City, CA, USA; 21Auckland Clinical
Studies, Auckland, New Zealand; 22Hospital Universitario Valle Hebron, Barcelona, Spain; 23The Chinese University of Hong Kong, Hong Kong

Background & Aims: Tenofovir alafenamide (TAF) is a new p <0.001) and lumbar spine (mean % change 0.75% vs.
prodrug of tenofovir developed to treat patients with chronic 2.57%; p <0.001), as well as a significantly smaller median
hepatitis B virus (HBV) infection at a lower dose than tenofovir change in estimated glomerular filtration rate by Cockcroft-
disoproxil fumarate (TDF) through more efficient delivery of Gault method (1.2 vs. 4.8 mg/dl; p <0.001).
tenofovir to hepatocytes. In 48-week results from two ongoing, Conclusion: In patients with HBV infection, TAF remained as
double-blind, randomized phase III trials, TAF was non-inferior effective as TDF, with continued improved renal and bone
to TDF in efficacy with improved renal and bone safety. We safety, two years after the initiation of treatment. Clinicaltrials.-
report 96-week outcomes for both trials. gov number: NCT01940471 and NCT01940341.
Methods: In two international trials, patients with chronic HBV Lay summary: At week 96 of two ongoing studies comparing
infection were randomized 2:1 to receive 25 mg TAF or 300 mg the efficacy and safety of tenofovir alafenamide (TAF) to teno-
TDF in a double-blinded fashion. One study enrolled HBeAg- fovir disoproxil fumarate (TDF) for the treatment of chronic
positive patients and the other HBeAg-negative patients. We hepatitis B patients, TAF continues to be as effective as TDF with
assessed efficacy in each study, and safety in the pooled population. continued improved renal and bone safety.
Results: At week 96, the differences in the rates of viral sup- Registration: Clinicaltrials.gov number: NCT01940471 and
pression were similar in HBeAg-positive patients receiving TAF NCT01940341.
and TDF (73% vs. 75%, respectively, adjusted difference 2.2% Ó 2017 European Association for the Study of the Liver. Published by
(95% CI 8.3 to 3.9%; p = 0.47), and in HBeAg-negative patients Elsevier B.V. All rights reserved.
receiving TAF and TDF (90% vs. 91%, respectively, adjusted dif-
ference 0.6% (95% CI 7.0 to 5.8%; p = 0.84). In both studies
the proportions of patients with alanine aminotransferase above Introduction
the upper limit of normal at baseline, who had normal alanine The World Health Organization estimates that approximately
aminotransferase at week 96 of treatment, were significantly 240 million people worldwide are chronically infected with
higher in patients receiving TAF than in those receiving TDF. the hepatitis B virus (HBV).1 Without treatment, chronic HBV
In the pooled safety population, patients receiving TAF had sig- infection can cause progressive liver fibrosis, which may lead
nificantly smaller decreases in bone mineral density than those to cirrhosis, decompensation, and hepatocellular carcinoma.2–4
receiving TDF in the hip (mean % change 0.33% vs. 2.51%; Suppressive antiviral treatment has been shown to reduce the
risk of liver-related complications, and can halt or even reverse
disease progression.5–7 However, since few patients achieve
Keywords: Chronic hepatitis B virus; Bone safety; Renal safety.
seroclearance of the hepatitis B surface antigen (HBsAg), which
Received 3 August 2017; received in revised form 13 October 2017; accepted 12
November 2017 is considered the hallmark of functional cure, treatment is gen-
⇑ Corresponding authors. Addresses: Kings College Hospital, London, United Kingdom erally life-long.6–9 In an aging population with comorbidities,
(K. Agarwal), or The Chinese University of Hong Kong, Hong Kong (H.L.Y. Chan). side effects of treatment such as renal and bone complications
E-mail addresses: kosh.agarwal@nhs.net (K. Agarwal), hlychan@cuhk.edu.hk
can be problematic with long-term treatment.9–13
(H.L.Y. Chan).

Journal of Hepatology 2018 vol. xxx j xxx–xxx


Please cite this article in press as: Agarwal K et al. 96 weeks treatment of tenofovir alafenamide vs. tenofovir disoproxil fumarate for hepatitis B virus infection. J Hepatol (2018), https://doi.org/
10.1016/j.jhep.2017.11.039
Research Article Viral Hepatitis

Tenofovir alafenamide (TAF) is an orally bioavailable prodrug ≥8 log10 IU/ml vs. <8 log10 IU/ml in Study GS-US-320-0110)
of tenofovir (TFV), a nucleotide analog that inhibits reverse tran- and prior oral antiviral treatment (naïve vs. previously treated).
scription of HIV and HBV.14–16 TAF was designed to have greater Before enrollment, written informed consent was obtained
plasma stability than tenofovir disoproxil fumarate (TDF) allow- from all patients. The study was approved by the institutional
ing delivery of the active metabolite, tenofovir diphosphate, to review board or independent ethics committees at all partici-
hepatocytes more efficiently than TDF, which must be dosed pating sites and was conducted in accordance with the princi-
at relatively high levels to achieve a therapeutic concentration ples of the Declaration of Helsinki and Good Clinical Practice.
in hepatic cells.17,18 Because of this high systemic exposure of All authors had access to the study data and had reviewed
tenofovir (TFV), the long-term use of TDF has been be associated and approved the final manuscript.
with bone and renal toxicity in some patients.10,13,19–21 When
TAF is administered at a dose of 25 mg to patients with HBV Procedures
or HIV infection, circulating levels of TFV were approximately Study visits occurred every four weeks starting at treatment
90% lower than levels with the standard 300 mg dose of week four until treatment week 48, after which study visits
TDF.22,23 occurred every eight weeks. Laboratory assessments included
GS-US-320-0110 and GS-US-320-0108 are large, ongoing, hematologic analysis, serum chemistry tests, fasting lipid
randomized, double-blind, international phase III trials parameters, and measures of renal function (serum creatinine,
designed to compare the efficacy and safety of TAF with that estimated glomerular filtration rate, proteinuria by dipstick),
of TDF in treatment-naïve and treatment-experienced patients as well as quantitative markers of proteinuria (protein to crea-
with chronic HBV infection, including those with compensated tinine ratio, albumin to creatinine ratio, retinol-binding protein
cirrhosis. The studies were identical in design except that to creatinine ratio, b2-microglobulin to creatinine ratio; Cov-
GS-US-320-0110 enrolled only patients with HBeAg-positive ance Laboratories, Indianapolis, IN, USA). Percentage change in
HBV infection and GS-US-320-0108 enrolled only patients with BMD was assessed in all patients by dual energy x-ray absorp-
HBeAg-negative HBV infection. After 48 weeks, TAF was shown tiometry scans of the lumbar spine and hip at screening, and
in both studies to be statistically non-inferior to TDF in antiviral every 24 weeks thereafter. Biomarkers of bone turnover were
efficacy, as measured by rates of suppression of HBV DNA to also assessed, including C-type collagen sequence, which is
<29 IU/ml.24,25 Moreover, patients receiving TAF in both trials associated with bone resorption, and bone-specific alkaline
had significantly smaller decreases in bone mineral density phosphatase, osteocalcin, and procollagen type 1 N-terminal
(BMD) in the lumbar spine and hip, smaller increases in serum propeptide, which are all associated with bone formation.
creatinine and smaller decreases in estimated creatinine clear-
ance, as well as other biomarkers of bone and renal safety than Outcomes
TDF. In the current report, we present 96-week results from Efficacy endpoints for this week 96 analysis were the proportion
both trials. of patients with HBV DNA <29 IU/ml, proportions of patients
with HBsAg loss and seroconversion to anti-HBs, and, in the
HBeAg-positive patients, proportions of patients HBeAg loss
Patients and methods and seroconversion to anti-HBe. Other prespecified week 96
Patients and study design efficacy endpoints include proportion of patients with ALT nor-
The designs of these two randomized, double-blind, active- malization (defined as ALT above the ULN at baseline but within
controlled international phase III trials have been described pre- the normal range at week 96) and the incidence of drug-
viously.24,25 Briefly, patients were at least 18 years of age with resistant mutations. We assessed ALT using normal ranges set
chronic HBV infection (with HBV DNA levels of at least 20,000 by the central laboratory (Covance), and those set forth in the
IU/ml), alanine transaminase (ALT) levels of >60 U/L in men or American Association for the Study of the Liver Diseases
>38 U/L in women, and estimated creatinine clearance of at least (AASLD) guidelines.8 Resistance analyses included population
50 ml/min (by the Cockcroft-Gault method). We excluded or deep sequencing of the HBV polymerase/reverse transcrip-
patients with platelet count ≤50,000 cells/ll, hemoglobin <10 tase region in all patients with HBV DNA ≥69 IU/ml at week
g/dl, albumin <3 g/dl, direct bilirubin of >2.5 times the upper 96 or those who discontinued the study early with viremia
limit of normal (ULN), and aspartate transaminase or ALT >10 (HBV DNA ≥69 IU/ml) after a minimum of 24 weeks of treat-
times the ULN. Patients with evidence of decompensation (i.e. ment. Safety endpoints at week 96 included percentage change
clinical ascites, encephalopathy, or variceal hemorrhage) and in hip BMD, percentage change in spine BMD, and changes in
those with hepatocellular carcinoma were not enrolled. Full eligi- renal function, as measured by serum creatinine and estimated
bility criteria are provided in the supplementary information. glomerular filtration rate (eGFRCG) determined by the
Patients in both trials were randomly assigned in a 2:1 ratio Cockcroft-Gault method.
to receive TAF 25 mg orally once daily or TDF 300 mg orally
once daily. All patients received placebo tablets matching the Statistical analysis
alternative treatment (i.e. patients assigned to receive TAF also For the primary efficacy analysis, the difference in proportions
received a matching TDF placebo tablet, and vice versa). Patients between treatment groups and its 95% CI was calculated based
and investigators were blinded to treatment assignment on the Mantel-Haenszel proportions adjusted by baseline HBV
throughout the 96 weeks of the double-blind phase. Members DNA categories and oral antiviral treatment status (naïve vs.
of the clinical research and biometrics departments of the spon- experienced) strata. P values were from the Cochran-Mantel-
sor were unblinded at the 48-week timepoint for the assess- Haenszel tests stratified by baseline HBV DNA categories and
ments related to the primary analysis. Randomization was oral antiviral treatment status strata.
stratified by screening HBV DNA levels (≥8 log10 IU/ml vs. 7 to For further details regarding the materials used, please refer
8 log10 IU/ml vs. <7 log10 IU/ml in Study GS-US-320-0108, and to the CTAT table and supplementary information.

2 Journal of Hepatology 2018 vol. xxx j xxx–xxx


Please cite this article in press as: Agarwal K et al. 96 weeks treatment of tenofovir alafenamide vs. tenofovir disoproxil fumarate for hepatitis B virus infection. J Hepatol (2018), https://doi.org/
10.1016/j.jhep.2017.11.039
JOURNAL
OF HEPATOLOGY
Role of the funding source (9%) receiving TAF and 32 patients (11%) receiving TDF; the rea-
The study sponsor oversaw trial management, data collection, sons for treatment failure (lack of efficacy, early discontinua-
and statistical analyses. The first draft of the report was pre- tions for an adverse event or death, other reasons [e.g.
pared by a medical writer employed by the sponsor. The corre- withdrawal of consent or lost to follow-up], or those having
sponding authors had full access to all data in the study and had missing data within the analysis window but remaining on
final responsibility for the decision to submit for publication. study) were similar between groups.
The proportion of HBeAg-negative patients receiving TAF
who had HBV DNA <29 IU/ml at week 96 was 90% (257 of 285
Results patients), compared to 91% (127 of 140 patients) of those receiv-
Patient disposition ing TDF (Fig. 1B and Table 2). The adjusted difference was 0.6%
Of the 873 HBeAg-positive patients originally randomized and (95% CI 7.0% to 5.8%; p = 0.84). At week 96, the lower bound of
treated in the GS-US-320-0110 study (581 to TAF and 292 to the two-sided 95% CI of the adjusted difference (TAF – TDF) in
receive TDF), 792 (91%; 530 TAF and 262 TDF) completed two the response rate was greater than the prespecified 10% mar-
years of double-blind treatment. Of the 425 HBeAg-negative gin used to determine noninferiority for the primary efficacy
patients originally randomized and treated in the GS-US-320- endpoint (proportion of patients with HBV DNA <29 IU/ml) at
0108 trial, 395 (93%; 266 TAF and 129 TDF) completed two week 48. In patients with HBV DNA <29 IU/ml, the proportion
years double-blind treatment at the time of this analysis. Fur- of patients with HBV DNA ‘‘target not detected” was 33% (93
ther details on patient disposition are provided (Figs. S1 and of 285 patients) in patients receiving TAF compared to 31%
S2), as well as details regarding treatment-emergent adverse (44 of 140 patients) of patients receiving TDF. Of the 425
events leading to treatment discontinuation (Table S1). patients randomized and treated, 41 were considered treatment
The baseline demographic characteristics of the patients for failures at week 96. Of these, eight had observed treatment fail-
the individual studies were described in the 48-week ure (HBV DNA ≥29 IU/ml) at the week-96 visit: five patients (2%)
reports.24,25 The patients characteristics of the pooled popula- receiving TAF and three patients (2%) receiving TDF. There were
tion are shown (Table 1). The characteristics of patients receiv- 33 patients who failed treatment at week 96 for nonvirologic
ing TAF and TDF were similar. Most patients were male (63%) reasons: 23 patients (8%) in the TAF group and 10 patients
and Asian (79%), with a mean age of 41 years. Mean HBV DNA (7%) in the TDF group; the reasons for nonvirologic failure were
and median ALT at baseline were 7.0 log10 IU/ml and 80 U/L, similar between treatment groups.
respectively. Two-thirds of patients were HBeAg-positive and
the most common HBV genotype was C, observed in about half ALT normalization
(48%) of patients, followed by genotype D (25%), genotype B The proportion of HBeAg-positive patients receiving TAF with
(19%), and genotype A (7%). Median estimated glomerular filtra- ALT above the ULN at baseline who had normal ALT at week
tion rate estimated by the Cockcroft-Gault method (eGFRCG) for 96 of treatment by central laboratory criteria was 75%, com-
patients at baseline was 106 ml/min. Comorbidities (hyperten- pared to 68% among patients receiving TDF; the difference of
sion, diabetes mellitus, cardiovascular disease, and hyperlipi- 8.0% was statistically significant (95% CI 1.2% to 14.7%; p =
demia) were present in a small percentage (<13%) of the 0.017) (Table 2). The same was true using laboratory criteria
overall study population with a similar distribution between recommended by the AASLD (≤30 U/L for men and ≤19 U/L for
the TAF and TDF groups. women), a significantly higher proportion of patients receiving
TAF achieved normalized ALT than those receiving TDF (52%
Efficacy vs. 42%, a difference in proportions of 10.6% (95% CI 3.6% to
Antiviral efficacy 17.6%; p = 0.003). Patients receiving TAF had higher rates of
The proportion of HBeAg-positive patients receiving TAF who ALT normalization than patients receiving TDF at every study
had HBV DNA <29 IU/ml at week 96 was 73% (423 of 581 visit after week 8 (Fig. 2).
patients), compared to 75% (218 of 292 patients) of those The proportion of HBeAg-negative patients receiving TAF,
receiving TDF (Fig. 1A and Table 2). The adjusted difference with ALT above the ULN at baseline, who had normal ALT at
was 2.2% (95% CI 8.3% to 3.9%; p = 0.47). At week 96, the week 96 of treatment by central laboratory criteria was 81%,
lower bound of the two-sided 95% CI of the adjusted difference compared to 71% among patients receiving TDF; the difference
(TAF – TDF) in the response rate was greater than the prespec- of 9.8% was statistically significant (95% CI 0.2% to 19.3%;
ified 10% margin used to determine noninferiority for the pri- p = 0.038) (Table 2). When assessed using laboratory criteria
mary efficacy endpoint (proportion of patients with HBV DNA recommended by the AASLD, a significantly higher proportion
<29 IU/ml) at week 48. In patients with HBV DNA <29 IU/ml, of patients receiving TAF than those receiving TDF achieved
the proportion of patients with HBV DNA ‘‘target not detected” normalized ALT (50% vs. 40%, a difference in proportions of
was 14% (81 of 581 patients) in patients receiving TAF compared 10.9% [95% CI 0.8% to 21.0%]; p = 0.035). Moreover, patients
to 9% (25 of 292 patients) of patients receiving TDF. Of the 873 receiving TAF had higher rates of ALT normalization than
patients randomized and treated, 232 did not have HBV DNA patients receiving TDF at every study visit after week 4 (Fig. 2).
<29 IU/ml at week 96. Of these, 148 had observed treatment
failure (HBV DNA ≥29 IU/ml) at the week-96 visit: 106 patients HBeAg and HBsAg loss and seroconversion
(18%) receiving TAF and 42 patients (14%) receiving TDF. Of The rate of HBeAg loss among HBeAg-positive patients receiving
those with observed treatment failure there was a higher pro- TAF was 22% (123/565) at week 96, which was not statistically
portion of patients in the TAF group (n = 28; 5%) compared with different from the rate of 18% (51/285) among patients receiv-
the TDF group (n = 6; 2%) with HBV DNA values ≥29 IU/ml and ing TDF; the rate of HBeAg seroconversion was also numerically
<69 IU/ml. There were 84 patients who were considered treat- higher among patients receiving TAF than among those
ment failures at week 96 for nonvirologic reasons: 52 patients receiving TDF, but the difference did not achieve statistical

Journal of Hepatology 2018 vol. xxx j xxx–xxx 3


Please cite this article in press as: Agarwal K et al. 96 weeks treatment of tenofovir alafenamide vs. tenofovir disoproxil fumarate for hepatitis B virus infection. J Hepatol (2018), https://doi.org/
10.1016/j.jhep.2017.11.039
Research Article Viral Hepatitis

Table 1. Patient characteristics in the pooled population.


TAF 25 mg TDF 300 mg Total
(n = 866) (n = 432) (N = 1,298)
Mean age, years (range) 40 (18–80) 41 (18–72) 41 (18–80)
Male sex, n (%) 544 (63) 275 (64) 819 (63)
Race
Asian 687 (79) 333 (77) 1,020 (79)
White 167 (19) 87 (20) 254 (20)
Black 7 (1) 6 (1) 13 (1)
Pacific Islander 3 (<1) 3 (1) 6 (<1)
Other 2 (<1) 3 (1) 5 (<1)
Mean HBV DNA, log10 IU/ml (range) 7.0 (1.8–9.9) 7.0 (1.4–9.9) 7.0 (1.4–9.9)
Median ALT (Q1, Q3) 80 (56, 123) 80 (53, 130) 80 (54, 125)
HBeAg status
Positive 569 (66) 290 (67) 859 (66)
Negative 297 (34) 142 (33) 439 (34)
HBV genotype
A 54 (6) 31 (7) 85 (7)
B 160 (18) 88 (20) 248 (19)
C 418 (48) 200 (46) 618 (48)
D 224 (26) 105 (24) 329 (25)
E 7 (1) 3 (1) 10 (1)
F 3 (<1) 2 (<1) 5 (<1)
H 0 2 (<1) 2 (<1)
Unknown 0 1 (<1) 1 (<1)
Patients with known cirrhosis 65/636 (10) 38/326 (12) 103/962 (11)
Mean Fibrotest score (range) 0.37 (0.04–0.98) 0.37 (0.03–0.99) 0.37 (0.03–0.99)
Fibrotest score ≥0.75 76/846 (9) 42/421 (10) 118/1267 (9)
Median eGFR by Cockcroft-Gault (Q1, Q3) 106 (91, 125) 105 (90, 124) 106 (91, 125)
Diabetes mellitus 57 (7) 29 (7) 86 (7)
Cardiovascular disease 28 (3) 14 (3) 42 (3)
Hypertension 98 (11) 61 (14) 159 (12)
Hyperlipidemia 76 (9) 42 (10) 118 (9)
Total hip bone mineral density clinical status
Normal (T-score ≥1.0) 570 (67) 285 (67) 855 (67)
Osteopenia (2.5 ≤T-score <1.0) 256 (30) 131 (31) 387 (30)
Osteoporosis (T-score <2.5) 12 (1) 2 (<1) 14 (1)
Not determined 13 (2) 8 (2) 21 (2)
Lumbar spine bone mineral density clinical status
Normal (T-score ≥1.0) 477 (56) 237 (56) 714 (56)
Osteopenia (2.5 ≤T-score <1.0) 309 (36) 152 (36) 461 (36)
Osteoporosis (T-score <2.5) 57 (7) 29 (7) 86 (7)
Not determined 13 (2) 8 (2) 21 (2)
25-hydroxy vitamin D, ng/ml n = 861 n = 429 n = 1,290
Median (Q1, Q3) 17.2 (12.8, 22.4) 17.2 (12.4, 22.4) 17.2 (12.8, 22.4)
<20 ng/ml 546 (63) 276 (64) 822 (64)
ALT, alanine aminotransferase; eGFR, estimated glomerular filtration rate; HBeAg, Hepatitis B e antigen; HBV, hepatitis B virus; TAF, tenofovir alafenamide; TDF, tenofovir
disoproxil fumarate.

significance (18% [99/565] vs. 12% [35/285], respectively, levels were observed at week 96 (0.14 and 0.10, for the
p = 0.05). In both groups, the rate of HBsAg loss at week 96 TAF and TDF groups, respectively; p = 0.50).
was 1%: 7 of 576 of patients receiving TAF, and 4 of 288 patients
receiving TDF. HBsAg seroconversion at week 96 was experi- Response in subgroups
enced by 6 of 576 patients (1%) receiving TAF and no patients Among patients with HBeAg-positive infection, differences in
receiving TDF. At baseline, mean levels of HBsAg were similar the proportion of patients with HBV DNA <29 IU/ml at week
between treatment groups (4.0 and 4.1 log10 IU/ml, for the 96 were not significantly different between treatment groups
TAF and TDF groups, respectively), and similar mean declines in predefined subgroups according to age (<50 years or ≥50
were observed at week 96 (0.51 and 0.64 log10 IU/ml for years), sex, race (Asian or non-Asian), baseline HBV DNA level
the TAF and TDF groups, respectively; p = 0.13). (<8 log10 IU/ml or ≥8 log10 IU/ml), treatment status
One patient in the study in HBeAg-negative patients experi- (treatment-experienced or treatment naïve), genotype (A/D or
enced loss of HBsAg: a 44-year-old Asian woman with genotype B/C), baseline ALT level (≤ULN or >ULN by central laboratory
A infection who was receiving TAF tested negative for HBsAg at normal range), and baseline FibroTest score (<0.75 or ≥0.75)
week 64 and achieved seroconversion to anti-HBs at week 80 (Fig. S3). In the small subgroup of patients with <95% adherence
which persisted through week 96. At baseline, mean HBsAg by pill count, a lower response rate was seen in the TAF group
levels were similar between treatment groups (3.4 log10 IU/ml (40% or 8/20 patients) compared with the TDF group (82% or 9
in both groups); small and similar mean declines in HBsAg of 11 patients); however, this difference is due in a large part

4 Journal of Hepatology 2018 vol. xxx j xxx–xxx


Please cite this article in press as: Agarwal K et al. 96 weeks treatment of tenofovir alafenamide vs. tenofovir disoproxil fumarate for hepatitis B virus infection. J Hepatol (2018), https://doi.org/
10.1016/j.jhep.2017.11.039
JOURNAL
OF HEPATOLOGY

Proportion of HBeAg-positive patients with breakthrough (i.e. persistent viremia above 69 IU/ml), with 36
A HBV DNA <29 IU/ml by study visit patients qualifying with virologic breakthrough (confirmed
Tenofovir alafenamide 25 mg
HBV DNA ≥69 IU/ml after achieving <69 IU/ml, or >1 log10 IU/
Tenofovir disoproxil fumarate 300 mg ml increase in HBV DNA from nadir). Of these, 11 of 36 (31%)
100 experienced virologic breakthrough associated with nonadher-
Proportions of patients (%)

ence to study medication. Overall, no HBV pol/RT amino acid


80 substitutions associated with resistance to tenofovir were
detected through week 96 in the TAF or TDF groups in either
60 study.
p = 0.47
40 Safety
The pooled safety analysis populations included 866 patients
20
receiving TAF and 432 receiving TDF. Both treatments were well
tolerated; the majority of patients experienced only adverse
0
events that were mild to moderate in severity (Table 3). The
0 16 32 48 64 80 96
Time (weeks) proportion of patients who discontinued treatment due to
adverse events was low in both groups: 13 patients (2%) receiv-
Proportion of HBeAg-negative patients with
B HBV DNA <29 IU/ml by study visit
ing TAF and four patients (1%) receiving TDF. The types and fre-
quencies of adverse events did not differ from those previously
Tenofovir alafenamide 25 mg
Tenofovir disoproxil fumarate 300 mg
reported at week 48.24,25 The most common adverse events
were headache, nasopharyngitis, and upper respiratory tract
100
Proportions of patients (%)

infection (Table 3). Seven percent of patients in both groups,


60 patients receiving TAF and 29 receiving TDF experienced
80
serious adverse events, none of which were deemed by the
p = 0.84
investigator to be related to study treatment. No patient died
60
during treatment, but five patients (two TAF and three TDF) died
while off treatment (two deaths were due to HCC, 1 TAF patient
40
and 1 TDF patient, at weeks 66 and 56, respectively; 1 TAF
20 patient due to H1NI influenza at week 14; 1 TDF patient due
to bilateral bronchopneumonia at week 59; 1 TDF patient due
0 to presumed cardiopulmonary arrest at week 64).
0 16 32 48 64 80 96 Similar percentages of patients receiving both drugs experi-
Time (weeks) enced grade 3 or 4 laboratory abnormalities, 36% of patients
receiving TAF and 34% of patients receiving TDF. The most com-
Fig. 1. Viral suppression by visit week. (A) shows proportion of HBeAg- mon grade 3 and 4 laboratory abnormalities were elevations in
positive patients with HBV DNA <29 IU/ml by study visit. (B) Shows
proportion of HBeAg-negative patients with HBV DNA <29 IU/ml by study
ALT (8% and 10% of patients receiving TAF and TDF, respectively)
visit. p value by Cochran-Mantel-Haenszel tests stratified by baseline HBV and AST (3% and 5% of patients, respectively). Five (1%) patients
DNA categories and oral antiviral treatment status. HBeAg, hepatitis B e receiving TAF and 4 (1%) patients receiving TDF experienced an
antigen; HBV, hepatitis B virus. ALT flare (elevation in serum ALT value of >2 x baseline and >10
x ULN and confirmed upon retesting) during treatment; these
to a high proportion of TAF patients (7 of the 12 treatment fail- events occurred early in treatment (within the first one to two
ures) having missing data (i.e. early study drug discontinuation) months) and all resolved without sequelae. In the TAF group,
at week 96. 50 patients (6%) experienced grade 3 elevations in fasting LDL
The proportion of HBeAg-negative patients with HBV DNA cholesterol (no patients had a grade 4 elevation) which were
<29 IU/ml at week 96 was not significantly different between mostly isolated events seen in individuals with a history of dys-
the two treatments in predefined subgroups, including age lipidemia, an elevated LDL level at baseline, or both. Three
(≥50 or <50), sex, race (Asian or non-Asian), HBV genotype patients (1%) receiving TDF had grade 3 elevations in fasting
(A/D or B/C), treatment status (naïve or experienced), baseline LDL cholesterol. For fasting glucose, 11 patients (1%) in the
HBV DNA (<7, or ≥7 log10 IU/ml), baseline ALT (> or ≤ULN by TAF group experienced grade 3 elevations compared to none
central laboratory), baseline FibroTest score (≥ or <0.75), and in the TDF group; no patients had a grade 4 elevation. These ele-
study drug adherence (<95% or ≥95%) (Fig. S3). vations were mostly isolated events seen in individuals with a
history of diabetes mellitus, an elevated fasting glucose level
Resistance surveillance at baseline, or both. No clinically meaningful differences were
Of the 1,298 patients who were randomized and treated in both detected in median (Q1, Q3) changes from baseline in fasting
studies, 1,242 entered year two of the study. Of these, 132 (11%) glucose levels at week 96 between TAF and TDF groups in both
met the criteria for resistance testing at week 96, 87 patients HBeAg-positive and HBeAg-negative patients.
receiving TAF and 45 receiving TDF. The types of sequence The mean percentage decrease in hip BMD from baseline to
changes observed were similar for the TAF and TDF groups, with week 96 was 0.33% (95% CI 0.51 to 0.14) for patients
the majority of patients having virus that was unable to receiving TAF, which was significantly less than the reduction
sequence, due to low viral load, or having no sequence changes of 2.51% (95% CI 2.82 to 2.21) for patients receiving TDF
from baseline at the consensus sequence level (59%). Most (p <0.001) (Fig. 3A). Similarly, the mean percentage decrease
patients who qualified had viremia in the absence of virologic in spine BMD from baseline to week 96 was 0.75% (95% CI

Journal of Hepatology 2018 vol. xxx j xxx–xxx 5


Please cite this article in press as: Agarwal K et al. 96 weeks treatment of tenofovir alafenamide vs. tenofovir disoproxil fumarate for hepatitis B virus infection. J Hepatol (2018), https://doi.org/
10.1016/j.jhep.2017.11.039
Research Article Viral Hepatitis

Table 2. Efficacy outcomes at week 96.


HBeAg-positive patients HBeAg-negative patients
TAF 25 mg TDF Difference in p value TAF 25 mg TDF Difference in p value
(n = 581) 300 mg proportions (n = 285) 300 mg proportions
(n = 292) (95% CI) (n = 140) (95% CI)
HBV DNA <29 IU/ml 423 (73%) 218 (75%) 2.2% 0.47 257 (90%) 127 (91%) 0.6% 0.84
(8.3% to 3.9%) (7.0% to 5.8%)
HBeAg loss, n/N (%)* 123/565 (22%) 51/285 (18%) 3.7% 0.20 — — — —
(1.9% to 9.4%)
HBeAg seroconversion, 99/565 (18%) 35/285 (12%) 5.1% 0.05 — — — —
n/N (%)* (0.2% to 10.1%)
HBsAg loss, n/N (%)y 7/576 (1%) 4/288 (1%) 0.1% 0.88 1/281 (<1%) 0 0.1% 0.72
(2.0% to 1.8%) (2.5% to 2.7%)
HBsAg seroconversion, 6/576 (1%) 0 1.1% 0.078 1/281 (<1%) 0 0.1% 0.72
n/N (%)y (0.3% to 2.4%) (2.5% to 2.7%)
Normalized ALT by 405/537 (75%) 181/268 (68%) 8.0% 0.017 191/236 (81%) 86/121 (71%) 9.8% 0.038
Central Lab (1.2% to 14.7%) (0.2% to 19.3%)
Normal Range, n/N (%)à
Normalized ALT by 299/572 (52%) 121/290 (42%) 10.6% 0.003 139/276 (50%) 55/138 (40%) 10.9% 0.035
AASLD Normal Range, n/N (%)§ (3.6% to 17.6%) (0.8% to 21.0%)
AASLD, American Association for the Study of Liver Diseases; ALT, alanine aminotransferase; HBeAg, hepatitis B e antigen; HBsAg, hepatitis B surface antigen; HBV, hepatitis
B virus; TAF, tenofovir alafenamide; TDF, tenofovir disoproxil fumarate.
*
Among patients who were seropositive for HBeAg and negative for anti-HBe at baseline.
y
Among patients who were seropositive for HBsAg and negative for anti-HBs at baseline.
à
Among patients with ALT at baseline above the central lab normal range.
§
Among patients with ALT at baseline above AASLD defined normal range.

1.01 to 0.49) for patients receiving TAF, which was signifi- larger median percentage decreases at week 96 in patients
cantly less than the mean percentage change of 2.57% (95% receiving TAF than those receiving TDF (Table S4). Fracture
CI 2.97 to 2.18) in patients receiving TDF (p <0.001) events were uncommon in both groups and were generally
(Fig. 3B). Moreover, the magnitude of the difference in BMD the result of trauma: nine patients (1%) receiving TAF and seven
decreases between the TAF and TDF groups was significantly patients (2%) receiving TDF experienced a fracture event. None
greater at week 96 compared to the difference in decline of the fractures were deemed related to the study drugs by
observed at week 48 (p <0.001; from mixed-model repeated the investigators, and none resulted in discontinuation of study
measures) when assessed at hip but not at spine drugs.
(Fig. 3A and B). These data, at least for hip, suggest that the Patients in both groups had small mean increases in serum
gap in difference in bone loss between the TDF and TAF groups creatinine from baseline to week 96; the mean increase of
continues to widen over time. 0.003 mg/dl in patients receiving TAF was significantly smaller
When categorical percentage changes in hip and spine BMD than the increase of 0.019 mg/dl in patients receiving TDF (p
are evaluated, decreases of 7% or greater for hip and 5% or = 0.001). Patients receiving TAF had a significantly smaller med-
greater for spine are considered clinically relevant. At week ian decrease in estimated glomerular filtration rate (by
96, 1.1% (8 of 740 patients) of the TAF group vs. 6% (21 of 369 Cockcroft-Gault equation; eGFRCG) than patients receiving TDF
patients) TDF patients demonstrated a ≥7% decrease in hip (1.2 ml/min vs. 4.8 ml/min, p <0.001) (Fig. 4). When assessed
BMD; for spine, 11% (82 of 746 patients) in the TAF group com- as the percentage of patients experiencing a 25% or greater
pared to 25% (93 of 371 patients) in the TDF group were decline in eGFRCG, a significantly smaller proportion of patients
observed to have a ≥5% decline in BMD (Table S2). Clinically, a receiving TAF compared with TDF met this endpoint (10% vs.
T-score is used to diagnose osteopenia (T-score ≥2.5 to 18%; p <0.001), and a smaller proportion of TAF patients experi-
<1.0) or osteoporosis (<2.5). At baseline, 570 patients in enced a confirmed decline in eGFRCG below 50 ml/min com-
the TAF group had a normal T-score (≥1.0) and 285 patients pared to TDF patients (0.1% TAF [1 patient] and 1.2% TDF [5
were normal in the TDF group with regard to hip BMD (Table 1). patients], respectively; p = 0.017). Independent predictors of
At week 96, in the TAF group, 6% (28 patients) with available eGFRCG decline of ≥25% from baseline were identified by multi-
data developed osteopenia whereas 16% (39 patients) devel- variate analysis and included the following characteristics: dia-
oped osteopenia in the TDF group; no patients with a normal betes mellitus, treatment with TDF, vitamin D level below the
T-score at baseline in either group developed osteoporosis. Of lower limit of the normal range, and baseline ALT value >5 times
the 256 TAF-treated patients with osteopenia at baseline, two ULN by AASLD criteria (Tables S5 and S6).
patients (1%) with available data at week 96 shifted to osteo- A similar proportion of patients in each group experienced a
porosis, compared with 4 of 131 (4%) of TDF-treated patients confirmed decrease in serum phosphorus level below 2.0 mg/dl
(Table S3). Biomarkers associated with bone resorption (C- (0.5% each group), and the median (Q1, Q3) change from base-
type collagen sequence), formation (procollagen type 1 N- line at week 96 in serum phosphorus levels was also similar
terminal propeptide, bone-specific alkaline phosphatase, and for the TAF and TDF groups (0.1 [0.4, 0.2] vs. 0.1 [0.4,
osteocalcin), and metabolism (parathyroid hormone) either 0.3]). The percentage of patients in each group with at least
showed significantly smaller median percentage increases or one graded event of proteinuria by dipstick during the study

6 Journal of Hepatology 2018 vol. xxx j xxx–xxx


Please cite this article in press as: Agarwal K et al. 96 weeks treatment of tenofovir alafenamide vs. tenofovir disoproxil fumarate for hepatitis B virus infection. J Hepatol (2018), https://doi.org/
10.1016/j.jhep.2017.11.039
JOURNAL
OF HEPATOLOGY

A Central Lab criteria B AASLD criteria

HBeAg-positive patients HBeAg-negative patients HBeAg-positive patients HBeAg-negative patients


Proportions of patients (%)

Proportions of patients (%)


100 Tenofovir alafenamide 25 mg 100 Tenofovir alafenamide 25 mg 100 Tenofovir alafenamide 25 mg 100 Tenofovir alafenamide 25 mg
Tenofovir disoproxil fumarate 300 mg Tenofovir disoproxil fumarate 300 mg Tenofovir disoproxil fumarate 300 mg Tenofovir disoproxil fumarate 300 mg

80 80 80 80
60 60 60 60
p = 0.017 p = 0.038
40 40 40 40
p = 0.003 p = 0.035
20 20 20 20

0 0 0 0
0 16 32 48 64 80 96 0 16 32 48 64 80 96 0 16 32 48 64 80 96 0 16 32 48 64 80 96
Time (weeks) Time (weeks) Time (weeks) Time (weeks)

Fig. 2. ALT Normalization by visit week. (A) shows proportion of patients achieving ALT normalization by central laboratory (Covance) criteria (≤34 U/L for
women <69 years or ≤32 for women ≥69 years; ≤43 U/L for men <69 years or ≤35 U/L for men ≥69 years) by study visit. (B) Shows proportion of patients
achieving ALT normalization by AASLD criteria (≤19 U/L for women and ≤30 U/L for men) by study visit. p value by Cochran-Mantel-Haenszel tests stratified by
baseline HBV DNA categories and oral antiviral treatment status. AASLD, American Association for the Study of Liver Diseases; ALT, alanine aminotransferase;
HBeAg, hepatitis B e antigen; HBV, hepatitis B virus.

Table 3. Safety data. increases from baseline to week 96 in the markers of proximal
tubular dysfunction urine retinol-binding protein to creatinine
TAF 25 mg TDF 300 mg
(n = 866) (n = 432) ratio and urine beta-2-microglobulin to creatinine ratio were
Patients with any adverse event 670 (77%) 327 (76%) significantly smaller among patients receiving TAF than among
Adverse event leading to study drug 13 (2%) 4 (1%) those receiving TDF (p <0.001 for the differences at every time-
discontinuation point through week 96) (Table S7). No patient in either group
Deaths 0 0 experienced a renal serious adverse event, a renal adverse event
Patients with grade 3 or 4 adverse events 58 (7%) 22 (5%) resulting in discontinuation of study drugs, an event of proximal
Patients with serious adverse event 60 (7%) 29 (7%) tubulopathy (including Fanconi syndrome), or an adverse event
Adverse events occurring in ≥5% of patients
of renal failure.
in any treatment group
Headache 104 (12%) 43 (10%)
Nasopharyngitis 105 (12%) 44 (10%)
Upper respiratory tract infection 98 (11%) 45 (10%) Discussion
Cough 70 (8%) 34 (8%) In reporting the 48-week outcomes for these large, randomized,
Back pain 54 (6%) 27 (6%)
phase III clinical trials, we noted that the interpretation of the
Fatigue 52 (6%) 23 (5%)
results was limited by the relatively short follow-up and that
Nausea 52 (6%) 24 (6%)
Dyspepsia 43 (5%) 22 (5%)
the durability of the favorable effects of TAF over TDF treatment
Diarrhea 47 (5%) 22 (5%) would need confirmation at a later timepoint. The present
Grade 3 or 4 laboratory abnormalities in ≥1% 96-week findings confirm these earlier results. Efficacy and
of patients in any treatment group* safety outcomes were consistent with those at week 48 for both
Alanine aminotransferase >5 ULN 72 (8%) 41 (10%) HBeAg-positive and HBeAg-negative patients, confirming that
Aspartate aminotransferase >5 ULN 30 (3%) 23 (5%) treatment with TAF resulted in a similar rate of viral suppres-
Amylase >2ULN 28 (3%) 13/427 (3%)
sion compared to that of TDF. Consistent with week 48 results,
Creatine kinase ≥10 ULN 28 (3%) 13 (3%)
treatment effects between TAF and TDF did not differ statisti-
Fasting cholesterol >300 mg/dl 10/857 0
(1%) cally in prespecified subgroups including age, race, region,
Fasting LDL cholesterol >190 mg/dl 50/843 3/418 (1%) HBV genotype, baseline HBV DNA level and prior oral antiviral
(6%) treatment status. Through 96 weeks of double-blind treatment,
Gamma glutamyl transferase >5x ULN 4 (<1%) 6 (1%) no resistance development was seen in either treatment group.
Hemoglobin <9.0 g/dl 7 (1%) 9/426 (2%) Further, the superior safety profile of TAF relative to TDF with
Segmented neutrophils <750/mm3 9 (1%) 3 (1%)
regard to bone and renal parameters seen at week 48 is con-
Fasting glucose >250 mg/dl 11 (1%) 0
firmed through 96 weeks of double-blind treatment.
Nonfasting glucose >250 mg/dl 31/856 7/426 (2%)
(4%) Another effect seen at both weeks 48 and 96 was that more
Occult blood 81 (9%) 39/426 (9%) patients in the TAF groups who had elevated ALT levels at base-
Urine erythrocytes 75/798 39/397 line achieved ALT normalization than those in the TDF groups.
(9%) (10%) At week 48, the difference was only significant when using
Urine glucose 44/859 7/426 (2%) the normal ranges for men and women proposed by AASLD. In
(5%)
the current week 96 analysis, the rate of ALT normalization
LDL, low-density lipoprotein; TAF, tenofovir alafenamide; TDF, tenofovir disoproxil was also significantly higher in both HBeAg-negative and
fumarate; ULN, upper limit of normal.
*
Laboratory results are based on 859 patients for TAF 25 mg, and 428 patients for HBeAg-positive patients receiving TAF when using the normal
TDF 300 mg, unless otherwise noted. ranges of the central laboratory which conducted the testing
(Covance). Given the consistency of this effect over time – the
was 72% (620 of 859) for patients receiving TAF and 75% (319 of rate of ALT normalization was higher for TAF than TDF patients
426) for patients receiving TDF; the difference was not statisti- at every visit after week 8 in both studies – there can be little
cally significant (p = 0.41). In contrast median percentage doubt that it is a treatment effect and not due to some

Journal of Hepatology 2018 vol. xxx j xxx–xxx 7


Please cite this article in press as: Agarwal K et al. 96 weeks treatment of tenofovir alafenamide vs. tenofovir disoproxil fumarate for hepatitis B virus infection. J Hepatol (2018), https://doi.org/
10.1016/j.jhep.2017.11.039
Research Article Viral Hepatitis

A HIP 10 Tenofovir alafenamide 25 mg

Median change from baseline,


Mean change from baseline, % (SD) Tenofovir alafenamide 25 mg Tenofovir disoproxil fumarate 300 mg
Tenofovir disoproxil fumarate 300 mg
4
p <0.001 5

ml/min (Q1, Q3)


2
0
-0.15 -1.2
0 -0.33 p <0.001
p <0.001
p <0.001 -5 -4.8
-2 -1.86 -2.51

-4 -10

-6
-15
24 48 72 96
-8 24 48 72 96 Time (weeks)
Time (weeks)
Fig. 4. Median change from baseline in eGFR (Cockcroft-Gault). The figure
SPINE shows median (Q1, Q3) change from baseline in estimated glomerular
B Tenofovir alafenamide 25 mg
Tenofovir disoproxil fumarate 300 mg
filtration rate (ml/min, by Cockcroft-Gault) by study visit. p value calculated
by two-sided Wilcoxon rank sum test to compare the two treatment groups.
Mean change from baseline, % (SD)

4 p = 0.80 eGFR, estimated glomerular filtration rate.


2
Serologic responses were similar for TAF compared with TDF,
0 -0.57 albeit among patients positive for HBeAg, the rates of HBeAg
-0.75
p <0.001 p <0.001 loss and anti-HBe seroconversion were numerically higher in
-2
-2.57
-2.38 patients receiving TAF than in those receiving TDF and in both
-4 groups these rates increased from week 48 to week 96 and
are consistent with results from previous studies of TDF, partic-
-6
ularly those in which treatment-experienced patients were
-8 24 48 72 96 included.26,29 Also consistent with prior studies the rates of
Time (weeks) HBsAg loss were low (approximately 1% in HBeAg-positive
patients) through two years of treatment, and mean declines
Fig. 3. Changes in bone mineral density. (A) shows mean percentage change
in HBsAg levels were small in both patient populations.
in hip bone mineral density at weeks 24, 48, 72, and 96 of treatment. (B)
shows mean percentage change in spine bone mineral density at weeks 24, Safety outcomes at week 96 were also consistent with those
48, 72, and 96 of treatment. p value to compare percent change from baseline of week 48. Overall, both treatments were well tolerated with
between treatment groups was calculated by ANOVA model including the same rate of serious adverse events (7%) and low rates of
treatment as a fixed effect. P value to compare the magnitude of the
discontinuations due to adverse events (1–2%). There were,
difference in percent BMD decline between treatment groups at week 48 vs.
Week 96 was calculated by mixed-model repeated measures. BMD, bone however, significant differences between the groups in favor
mineral density. of TAF in renal and bone safety. As noted, TAF was specifically
designed to substantially reduce systemic exposure to tenofovir,
undetected bias in the randomized populations. Several which was thought to be causing these known abnormalities
hypotheses about the cause of this unexpected effect have been associated with long-term TDF use.10,19–21 Regarding bone loss,
considered, and while exploratory investigations are currently over two years minimal declines in hip and spine BMD were
underway, the mechanism of this effect remains unknown. observed with TAF treatment compared with progressive decli-
TDF treatment is known to have a ‘‘lipid lowering effect” as nes in BMD with TDF. This difference is particularly evident at
previously described in HIV infected patients wherein levels of the hip, which continues to demonstrate ongoing significant
fasting total cholesterol, LDL cholesterol, and HDL cholesterol, BMD decline with TDF therapy compared to TAF from week
are all significantly reduced in patients who initiate or are 48 to week 96. While the overall clinical implications of these
switched to TDF-containing therapy.26,27 However, the ratio of data will require longer term studies and follow-up, preventing
total cholesterol to HDL is not significantly different following further BMD decline in patients is a meaningful clinical goal as it
TDF treatment; therefore, the clinical relevance of this lipid lower- is known that fracture risk increases exponentially as BMD
ing effect is unclear and a precise mechanism for these changes decreases.30 Similar to BMD findings, over two years of treat-
has not been ascertained. In the integrated analysis of these two ment, the significant differences in declines in estimated
phase III studies, median changes in fasting lipid parameters were glomerular filtration rate and smaller changes in biomarkers
small in the TAF group while declines in all three fasting lipid of proximal tubular function support that TAF has less of an
parameters and triglycerides were observed in the TDF group, con- impact on renal function than TDF. A limitation of this renal
sistent with results seen in HIV patients (Table S8). Thus, com- analysis is that the patients were all relatively healthy from a
pared to TDF, TAF seems to demonstrate a ‘‘lipid neutral” renal perspective – all patients enrolled had creatinine clear-
effect. As previously reported, at week 48, a higher proportion of ances ≥50 ml/min and most patients were ≤65 years old and
TAF-treated patients experienced grade 3 or higher levels of fast- without comorbidities. While longer term follow-up will be
ing LDL (≥300 mg/dl) compared with the TDF group.25,28 Inte- needed to fully characterize the renal safety profile of TAF, these
grated analysis at week 48 of available data from both studies findings of lower renal impact compared to TDF are important
for 1,254 patients (data on file, Gilead Sciences) showed rates of given the reports that link TDF use to kidney injury.19,31,32,20
≥grade 3 fasting LDL levels were 4% with TAF and 1% with TDF. This analysis has several limitations; while 96 weeks of treat-
At week 96 (Table 3), the rates were 6% with TAF and 1% with ment has yielded similar findings to the week 48 analysis, longer
TDF treatment, representing a small change after two years of term data are required to better determine differences in clinical
treatment. outcomes. In both of these studies, double-blind treatment will be

8 Journal of Hepatology 2018 vol. xxx j xxx–xxx


Please cite this article in press as: Agarwal K et al. 96 weeks treatment of tenofovir alafenamide vs. tenofovir disoproxil fumarate for hepatitis B virus infection. J Hepatol (2018), https://doi.org/
10.1016/j.jhep.2017.11.039
JOURNAL
OF HEPATOLOGY
continued through three years (week 144) in a substantial subset Board – Gilead, BMS, AbbVie; Research – Gilead, Merck. Employ-
of patients (approximately 58%) who had signed informed con- ees and stockholders of Gilead Sciences: John F. Flaherty, Anuj
sent for a recently enacted amendment to the study protocol Gaggar, Audrey H. Lau, Vithika Suri, Andrea L. Cathcart, Neeru
which allows another year of blinded therapy. Further, in both Bhardwaj, Lanjia Lin, and G. Mani Subramanian. Huy Trinh:
studies all patients who roll over to open-label TAF treatment Research—Gilead, Intercept; Advisory Board and Speaker—Gilead.
(at either week 96 or week 144) will have the ability to continue Edward J. Gane: Advisory Board – AbbVie, ALIOS, Gilead, Janssen,
this therapy through eight years (week 384). Across both study Roche; Speaker—Gilead, AbbVie. Maria Buti: Advisory Board and
populations the proportion of patients considered to be at higher Speaker – Gilead, MSD, BMS, Janssen, AbbVie. Henry L.Y. Chan:
risk of TDF-associated bone and renal complications (i.e. age over Advisor and speaker for AbbVie, BMS, Gilead and Roche; Advisor
60 years, history of clinically significant bone and/or renal dis- for Janssen; Speaker for MSD. No relevant conflicts of interest to
ease)9 is relatively small (estimated to be less than 20%) and addi- disclose: Scott Fung, Patrick Marcellin, Manoj Sharma, Mustafa
tional studies in these populations are warranted. Further, our Kemal Çelen, Norihiro Furusyo, Dr. Shalimar, Ki Tae Yoon.
studies enrolled only viremic patients with elevated levels of Please refer to the accompanying ICMJE disclosure forms for
serum ALT at screening who are considered candidates for initiat- further details.
ing antiviral treatment by all current guidelines.8,9,33 Whether the
safety advantages and comparable antiviral efficacy we observed
for TAF relative to TDF in these studies can be duplicated in virally Authors’ contributions
suppressed patients who switch treatment from TDF to TAF John F. Flaherty, Anuj Gaggar, G. and Mani Subramanian con-
remains to be determined. A preliminary analysis of 541 patients tributed to the study concept and design. Kosh Agarwal, Maur-
enrolled in these two trials who switched from double-blind TDF izia Brunetto, Young-Suk Lim, Calvin Q. Pan, Maria Buti, Wai Kay
to open-label TAF at week 96 has shown significant improve- Seto, Scott Fung, Patrick Marcellin, Sang Hoon Ahn, Namiki
ments in bone and renal parameters as early as 24 weeks follow- Izumi, Wan Long Chuang, Ho Bae, Manoj Sharma, Harry L.A.
ing switch.34 Of note, a large randomized, controlled trial to Janssen Mustafa Kemal Çelen, Norihiro Furusyo, Dr. Shalimar,
evaluate the efficacy and safety of switching chronic HBV patients Ki Tae Yoon, Huy Trinh, Edward Gane, and Henry L.Y. Chan con-
who are suppressed on long-term TDF 300 mg once daily to TAF tributed to the acquisition of data. Lanjia Lin contributed to the
25 mg once daily has been recently initiated (NCT02979613). statistical analysis. All authors contributed to the analysis and
In conclusion, after two years of treatment, TAF remained as interpretation of data, drafting of the manuscript, critical revi-
effective in suppressing HBV replication as TDF with no viro- sion of the manuscript for important intellectual content.
logic resistance, and was associated with significantly less bone
and renal toxicity. The mechanism behind the superior rates of Acknowledgements
ALT normalization for TAF over TDF remains to be elucidated. This study was sponsored by Gilead Sciences. Writing assistance
was provided by David McNeel of Gilead Sciences.
Financial support
The study sponsor (Gilead Sciences) was involved with the
study design, collection, analysis, and interpretation of data. Supplementary data
Supplementary data associated with this article can be found, in
the online version, at https://doi.org/10.1016/j.jhep.2017.11.
Conflict of interest 039.
Kosh Agarwal: Advisory Board and Speaker – AbbVie, Achillion,
BMS, GSK, Gilead, Intercept, Janssen, Merck, Novartis, Roche;
Consultant – AbbVie, Achillion, BMS, GSK, Gilead, Intercept, References
Janssen, Merck, Novartis, Roche. Grant – BMS, Gilead, Roche. [1] World Health Organization. Hepatitis B fact sheet. http://www.who.
Maurizia Brunetto: Advisory Board – Gilead, Merck, AbbVie, int/mediacentre/factsheets/fs204/en/.
Janssen; Speaker – AbbVie, BMS, Gilead, Janssen, Merck, Roche. [2] Ganem D, Prince AM. Hepatitis B virus infection—natural history and
clinical consequences. N Engl J Med 2004;350:1118–1129.
Wai Kay Seto: Advisory Board-Gilead, AbbVie, Bristol-Myers
[3] McMahon BJ. Chronic hepatitis B virus infection. Med Clin North Am
Squibb; Speaker’s Bureau-Gilead, AbbVie, Bristol-Myers Squibb, 2014;98:39–54.
Novartis, AstraZeneca, Alfa Wassermann. Young-Suk Lim: Advi- [4] Fattovich G, Bortolotti F, Donato F. Natural history of chronic hepatitis B:
sory Board – Bayer, Bristol-Myers Squibb, Gilead; Research – special emphasis on disease progression and prognostic factors. J Hepatol
Bayer, Bristol-Myers Squibb, Gilead Sciences, Novartis; Speaker 2008;48:335–352.
[5] Gish RG, Given BD, Lai C-L, Locarnini SA, Lau JY, Lewis DL, et al. Chronic
– Bayer, Gilead. Sang Hoon Ahn: Advisory Board – Bristol-Myers hepatitis B: virology, natural history, current management and a glimpse
Squibb, Gilead, AbbVie, MSD; Research – Bristol-Myers Squibb, at future opportunities. Antivir Res 2015;121:47–58.
Gilead Sciences, Roche. Namiki Izumi: Advisory Board-Gilead; [6] Marcellin P, Gane E, Buti M, Afdahl N, Sievert W, Jacobson IM, et al.
Speaker-Gilead, AbbVie, Shionogi, Otsuka, Bayer, MSD. Wan- Regression of cirrhosis during treatment with tenofovir disoproxil
fumarate for chronic hepatitis B: a 5-year open-label follow-up study.
Long Chuang: Advisory board–Gilead, AbbVie, BMS, MSD,
Lancet 2013;381:468–475.
PharmaEssentia; Speaker–Gilead, AbbVie, BMS, MSD, [7] Peng CY, Chien RN, Liaw YF. Hepatitis B virus-related decompensated
PharmaEssentia, Roche. Ho Bae: Speaker and research grant for liver cirrhosis: benefits of antiviral therapy. J Hepatol 2012;57:442–450.
Gilead. Harry L.A. Janssen: Grants – AbbVie, Bristol-Myers Squibb, [8] Terrault NA, Bzowej NH, Chang K-M, Hwang JP, Jonas MM, Murad MH.
Gilead Sciences, Innogenetics, Janssen, MedImmune, Medronic, AASLD guidelines for treatment of chronic hepatitis B. Hepatology
2016;63:261–283.
Merck, Roche; Consultant – AbbVie, Benitec, Bristol-Myers [9] European Association for the Study of the Liver. EASL 2017 clinical
Squibb, Gilead Sciences, Janssen, MedImmune, Merck, Roche, practice guidelines on the management of hepatitis B virus infection. J
Arbutus. Calvin Q. Pan: Consultant – Gilead, AbbVie; Advisory Hepatol 2017. https://doi.org/10.1016/j.jhep.2017.03.021.

Journal of Hepatology 2018 vol. xxx j xxx–xxx 9


Please cite this article in press as: Agarwal K et al. 96 weeks treatment of tenofovir alafenamide vs. tenofovir disoproxil fumarate for hepatitis B virus infection. J Hepatol (2018), https://doi.org/
10.1016/j.jhep.2017.11.039
Research Article Viral Hepatitis

[10] Gill US, Zissimopoulos A, Al-Shamma S, Burke K, McPhail MJ, Barr DA, of TAF as 10-day monotherapy in HIV-1-positive adults. J Acquir
et al. Assessment of bone mineral density in tenofovir-treated patients Immune Defic 2013;63:449–455.
with chronic hepatitis B: Can the fracture risk assessment tool identify [24] Buti M, Gane E, Seto WK, Chan HL, Chuang WL, Stepanova T, et al. TAF vs.
those at greatest risk? J Infect Dis 2015;211:374–382. tenofovir disoproxil fumarate for the treatment of patients with HBeAg-
[11] Chen CH, Lin CL, Kao CH. Association between chronic hepatitis B virus negative chronic hepatitis B virus infection: a randomised, double-blind,
infection and risk of osteoporosis: a nationwide population-based study. phase 3, non-inferiority trial. Lancet. Gastroenterol Hepatol
Medicine 2015;94:e2276. 2016;1:196–206.
[12] Chen YC, Su YC, Li CY, Hung SK. 13-year nationwide cohort study of [25] Chan HL, Fung S, Seto WK, Chuang WL, Chen CY, Kim HJ, et al. TAF vs.
chronic kidney disease risk among treatmen-naïve patients with chronic tenofovir disoproxil fumarate for the treatment of HBeAg-positive
hepatitis B in Taiwan. BMC Nephrology 2015;16:110. chronic hepatitis B virus infection: a randomised, double-blind, phase
[13] Maggi P, Montinaro V, Leone A, Fasano M, Volpe A, Bellacosa C, et al. 3, non-inferiority trial. Lancet. Gastroenterol Hepatol 2016;1:185–195.
Bone and kidney toxicity induced by nucleotide analogues in patients [26] Santos JR, Saumoy M, Curran A, Bravo I, Libre JM, Navarro J, et al. The
affected by HBV-related chronic hepatitis: a longitudinal study. J lipid-lowering effect of tenofovir/emtricitabine: a randomized, cross-
Antimicrob Chemother 2015;70:1150–1154. over, double-blind, placebo-controlled trial. Clin Infect Disease
[14] Lee WA, He GX, Eisenberg E, Cihlar T, Swaminathan S, Mulato A, et al. 2015;61:403–408.
Selective intracellular activation of a novel prodrug of the human [27] Tungsiripat M, Kitch D, Glesby MJ, Gupta SK, Mellors JW, Moran L, et al. A
immunodeficiency virus reverse transcriptase inhibitor tenofovir leads pilot study to determine the impact on dyslipidemia of adding tenofovir
to preferential distribution and accumulation in lymphatic tissue. to stable background antiretroviral therapy: ACTG 5206. AIDS
Antimicrob Agents Chemother 2005;49:1898–1906. 2010;24:1781–1784.
[15] De Clercq E. Tenofovir alafenamide (TAF) as the successor of tenofovir [28] Fung S, Kwan P, Fabri M, Horban A, Pelemis M, Hann HW, et al.
disoproxil fumarate (TDF). Biochem Pharmacol 2016;119:1–7. Randomized comparison of tenofovir disoproxil fumarate vs. emtric-
[16] Delaney 4th WE, Ray AS, Yang H, Qi X, Xiong S, Zhu Y, et al. Intracellular itabine and tenofovir disoproxil fumarate in patients with lamivudine-
metabolism and in vitro activity of tenofovir against hepatitis B virus. resistant chronic hepatitis B. Gastroenterology 2014;146:980–988.
Antimicrob Agents Chemother 2006;50:2471–2477. [29] Berg T, Zoulim F, Moeller B, Trinh H, Marcellin P, Chan S, et al. Long-term
[17] Babusis D, Phan TK, Lee WA, Watkins WJ, Ray AS, et al. Mechanism for efficacy and safety of emtricitabine plus tenofovir DF vs. tenofovir DF
effective lymphoid cell and tissue loading following oral administration monotherapy in adefovir-experienced chronic hepatitis B patients. J
of nucleotide prodrug GS-7340. Mol Pharmaceutics 2013;10:459–466. Hepatol 2014;60:715–722.
[18] Murakami E, Wang T, Park Y, Hao J, Lepist El, Babusis D, et al. [30] Cosman F, de Beur SJ, LeBoff MS, Lewiecki EM, Tanner B, Randall S, et al.
Implications of efficient hepatic delivery by TAF (GS-7340) for hepatitis Erratum to: Clinician’s guide to prevention and treatment of osteoporo-
B virus therapy. Antimicrob Agents Chemother 2015;59:3563–3569. sis. Osteoporos Int 2015;26:2045–2047. https://doi.org/10.1007/s00198-
[19] Hall AM, Hendry BM, Nitsch D, Connolly JO. Tenofovir-associated kidney 015-3037-x.
toxicity in HIV-infected patients: a review of the evidence. Am J Kidney [31] Viganò M, Brocchieri A, Spinetti A, Zaltron S, Mangia G, Facchetti F, et al.
Dis 2011;57:773–780. Tenofovir-induced Fanconi syndrome in chronic hepatitis B monoin-
[20] Gara N, Zhao X, Collins MT, Chong WH, Kleiner DE, Liang TJ, et al. Renal fected patients that reverted after tenofovir withdrawal. J Clin Virol
tubular dysfunction during long-term adefovir or tenofovir therapy in 2014;61:600–603.
chronic hepatitis B. Aliment Pharmacol Ther 2012;35:1317–1325. [32] Gracey DM, Snelling P, McKenzie P, Strasser SI. Tenofovir-associated
[21] Brown TT, Moser C, Currier JS, Ribaudo HJ, Rothenberg J, Kelesidis T, et al. Fanconi syndrome in patients with chronic hepatitis B monoinfection.
Changes in bone mineral density after initiation of antiretroviral Antivir Ther 2013;18:945–948.
treatment with tenofovir disoproxil fumarate/emtricitabine plus ataza- [33] Sarin SK, Kumar M, Lau GK, Abbas Z, Chan HL, Chen CJ, et al. Asian-Pacific
navir/ritonavir, darunavir/ritonavir, or raltegravir. J Infect Dis clinical practice guidelines on the management of hepatitis B: a 2015
2015;212:1241–1249. update. Hepatol Int 2016;10:1–98.
[22] Agarwal K, Fung SK, Nguyen TT, Cheng W, Sicard E, Ryder SD, et al. [34] Chan HL, Fung S, Seto WK, Gane E, Flaherty JF, Suri V, et al. Improved
Twenty-eight day safety, antiviral activity, and pharmacokinetics of TAF bone and renal safety of switching from tenofovir disoproxil fumarate
for treatment of chronic hepatitis B infection. J Hepatol 2015;62:533. (TDF) to tenofovir alafenamide (TAF): results from 2 Phase 3 studies in
[23] Ruane PJ, DeJesus E, Berger D, Markowitz M, Bredeek UF, Callebaut C, HBeAg-positive and HBeAg-negative patients with chronic hepatitis B. J
et al. Antiviral activity, safety, and pharmacokinetics/pharmacodynamics Hepatol 2017;66:#PS-041.

10 Journal of Hepatology 2018 vol. xxx j xxx–xxx


Please cite this article in press as: Agarwal K et al. 96 weeks treatment of tenofovir alafenamide vs. tenofovir disoproxil fumarate for hepatitis B virus infection. J Hepatol (2018), https://doi.org/
10.1016/j.jhep.2017.11.039

You might also like