32 Moreover, Pol and X have been shown to be able to counteract t

32 Moreover, Pol and X have been shown to be able to counteract the pattern recognition receptor signaling in hepatocytes.8, 33, 34 Therefore, we speculated that nonparenchymal liver cells may contribute to intrahepatic ISG expression during HBV infection, Selleck PLX-4720 though the mechanisms involved are yet to be determined. In addition, although HBsAg did not influence IFN signaling in vitro, we cannot neglect the role of it and

hepatitis B e antigen in contributing to the IFN response defect in vivo, as they were reported to suppress Toll-like receptor–induced IFN-β and ISG induction in both the parenchymal and nonparenchymal liver cells.35 In addition, the data obtained from liver biopsies revealed that the nuclear translocation of STAT1/2 was impaired in HBV-positive cells, but was still intact in many neighboring cells without HBV infection. Therefore, comprehensive analysis of the interaction between HBV and the IFN system in both hepatocytes and nonparenchymal cells is necessary, PF-562271 concentration and it will be interesting to compare the STAT activation pattern in different types of liver cells between the IFN responders and nonresponders to further explore factors affecting response to IFN-α therapy. Viruses have evolved various strategies to circumvent the IFN response, thus allowing them to escape the host defenses.36 HCV, for example, impairs type I

IFN response by blocking different levels of IFN-α signal pathway via its core, NS3 and NS5A Sorafenib purchase proteins. For HBV, we propose a two-part mechanism by which Pol inhibits the IFN-α–stimulated antiviral responses. These findings, together with our previous finding that Pol can inhibit the type I IFN induction,8 suggest that Pol is a multifunctional IFN antagonist. This knowledge not only helps us understand the mechanisms of resistance of HBV-infected patients to IFN treatment, it also clarifies the role of Pol in HBV persistence. Once viral replication reaches high levels, Pol may exert its anti-IFN activities to ensure the survival

of the virus. Notably, the sensitivity to IFN-α differs between HBV and HCV. It was reported that IFN treatment resulted in a rapid reduction in HCV but a moderate reduction in HBV.37 HBV seems to have a stronger ability to interfere with the IFN antiviral actions compared with HCV. However, the inhibitory effect of HBV on the IFN-α–mediated ISG induction was found to be modest.37 HBV, as a hepatotropic DNA virus, may have low sensitivity to IFN-induced ISGs and counteract the IFN actions at different levels, including the IFN signal transduction and antiviral functions of ISG products. Nevertheless, future studies are required to fully understand HBV resistance to IFN-α and precisely define the mechanisms by which IFN-α inhibits HBV replication.

Conclusions: The course of the bile ducts can be recognized on co

Conclusions: The course of the bile ducts can be recognized on conventional ultrasound by referencing virtual ultrasonography constructed by Gd-EOB-DTPAenhanced MRI. This imaging technology is useful

in avoiding bile duct injury during RFA. Disclosures: BTK inhibitor The following people have nothinq to disclose: Yohei Koizumi, Masashi Himooka, Hironori Ochi, Yoshio Tokumoto, Masanomi Abe, Fujimasa Tada, Atsushi Himaoka, Himoaki Tanaka, Takahamu Tsuda, Temuhito Mochizuki, Yoichi Hiasa Background and Aim Virtual Touch Quantification (VTQ) can be used to easily measure spleen stiffness (SS) by referring to the corresponding B-mode image without restricting the measurement distance. However, the usefulness and challenges associated with the measurement of SS for the prediction of liver fibrosis stage are not well documented. In the present study, we aimed to evaluate SS by VTQ for the prediction of liver fibrosis. Patients and Methods From December 2010 to February 2013, 352 patients (162 men and learn more 190 women) with chronic liver disease confirmed by liver biopsy were evaluated by VTQ for the measurement of liver stiffness (LS) and SS (average age 55.8 ± 13.5 years; 90 patients with hepatitis B, 179 with hepatitis C, and 1 with hepatitis B and C; 76 patients had non-B non-C hepatitis). The New Inuyama Classification was used to evaluate the degree

of hepatitis. The distribution of liver fibrosis stages was as follows: stage over F0 (n =15), F1(n =134), F2 (n = 66), F3 (n = 73), and F4 (n = 64). VTQ measurements were performed using the Siemens Acuson S2000 ultrasound system. SS values were compared with clinical parameters including measurements of LS; platelet count; levels of AST, ALT, bilirubin, hyaluronic acid, and albumin; prothrombin time; and APRI. Results The LS and SS values corresponding to each fibrosis stage were 1.16 and 2.40 for stage F0, 1.14 and 2.33 for stage F1, 1.34 and 2.44 for stage F2, 1.53 and 2.54 for stage F3, and 2.30 and 3.18 for stage F4, respectively. Significant differences between stages F3 and F4 were observed for both LS and SS values (P < 0.0001). SS values showed the highest correlation

with LS values (r = 0.595, P < 0.0001). The area under the receiver operating characteristic curve for SS to distinguish between fibrosis was the highest among all the parameters (SS = 0.918; LS = 0.905; hyaluronic acid = 0.830; APRI = 0.772; platelet count = 0.738; prothrombin time = 0.738). However, for SS measurements, 20% (n = 3) of F0 and 16% (n = 22) of F1 patients fell above the F4 cutoff levels; these rates were higher than those for LS (0% of F0; 3% of F1). All cases with high SS values and F0 and F1 stages had a small spleen except for 1 severely obese F1 patient. Conclusion SS measurements obtained using VTQ could be a good predictor of liver fibrosis stage, although the occurrence of false positive results should be carefully considered in cases with small spleens.

003 to 0 04 Methylation levels of the individual 26,486 autosoma

003 to 0.04. Methylation levels of the individual 26,486 autosomal CpG sites as well as the overall means were compared between the 62 pairs of tissues. There were 2,324 CpG sites that significantly differed in methylation level between tumor and nontumor tissues after Bonferroni’s adjustment (for a complete list, see Supporting Tables Osimertinib supplier 2 and 3). Among all significant CpG sites,

684 were significantly hypermethylated (covering 548 genes) and 1,640 were significantly hypomethylated (covering 1,290 genes) in tumor, compared to nontumor, tissues. Figure 1 displays mean DNA-methylation differences between the 62 paired tumor/adjacent tissues at all 26,486 CpG sites using a volcano plot. Both hyper- and hypomethylation alterations are common events in HCC tumor tissues. The top 20 hyper- or hypomethylated sites ranked by statistical significance are given in Table 2. Regardless of whether they were hypo- or hypermethylated, all significant CpG sites had similar mean methylation levels in tumor tissues

(42.2% versus 42.9%), whereas the mean methylation levels in nontumor tissues were dramatically different (26.0% for hypermethylated versus 58.4% for hypomethylated sites). Figure 2 shows Midostaurin purchase the heatmap of the top 1,000 CpG sites (based on statistical significance) distinguishing tumor from adjacent tissues. In general, good separation of tumor and adjacent tissues was observed, with a small amount of misclassification. A Manhattan plot was used MAPK inhibitor to display the −log10 (adjusted P value) for the differences in methylation by chromosome (Supporting Fig. 2) and indicates that aberrant methylation is spread across all chromosomes. Among the 2,324 significantly differentially methylated CpG sites, >80% (82.3% and 85.8% for hyper- and hypomethylated sites, respectively)

had a >10% absolute tumor/nontumor difference in percent methylation, and >50% had a >15% difference (Supporting Table 4). These data indicate that the methylation changes occurring during HCC development are robust and may provide useful biomarkers. The majority of the significantly differentially methylated CpG sites are located within the proximal promoter regions. Among the 2,324 significant CpG sites, the distances to the transcription start site (TSS) ranged from 0 to 1,498 bp (base pairs), with an average of 407 bp and an SD of 362 bp. Hypermethylated CpG sites are more common within a short distance of TSS (50.7% within 250 bp and 26.9% between 250 and 500 bp), compared to hypomethylated sites (41.6% and 23.3%, respectively) (Supporting Fig. 3). The average distance to the TSS was significantly shorter for hypermethylated (mean = 332 bp; SD = 312 bp), compared with hypomethylated, sites (mean = 437 bp; SD = 377 bp; P = 3.95 × 10−10). Within CpG islands, more sites were significantly hypermethylated in tumors, whereas within non-CpG island regions, more sites were significantly hypomethylated in tumors (Supporting Table 5; Supporting Fig. 4).

Consistent with its ability to

bind PARP1 for transcripti

Consistent with its ability to

bind PARP1 for transcriptional activation, the “ACTTCAAA” HBVCP PARP1 binding motif could also interfere with histone H1 ADP-ribosylation (Fig. 5A). This raises the possibility that HBVCP-PARP1 Tamoxifen manufacturer interaction not only supports HBV replication, but also impairs PARP1 enzyme-dependent functions, such as DNA repair in vivo. If this were true, the ability of cells to effectively carry out DNA strand-break repair when challenged by DNA-damaging agents would be compromised. To verify this, a construct bearing the HBV-PARP1 binding motif in three tandem copies (Fig. 5B) was tested for its capacity to inhibit cellular PARP1 enzymatic activity by determining the degree of DNA damage induced with etoposide (DNA single- and double-strand

break inducer) or bleomycin (DNA double-strand break inducer). Alkaline comet assays revealed that HepG2 cells transfected with the PARP1 motif had significantly more DNA in comet tails than cells treated with dimethyl sulfoxide (DMSO) or the control vector (Fig. 5C), ICG-001 concentration reflecting enhanced DNA damage. This suggests that the ability of PARP1 to ADP-ribosylate protein targets required in DNA damage-repair pathways was reduced, supporting the inhibitory role of HBVCP-PARP1 motif expression on nuclear PARP1 enzymatic activity. The effect of the PARP1 motif was further assessed for its ability to sensitize cells to induced cytotoxicity caused by DNA-damaging agents. Consistent with accumulation

of damaged DNA, etoposide or bleomycin treated HepG2 cells transfected with the PARP1 motif had a significantly larger population of Annexin V–positive cells (Fig. 5D). In contrast, DMSO treatment or vector control did not show significant changes in Annexin V staining. The enhanced cytotoxicity toward sublethal amounts of etoposide Leukotriene-A4 hydrolase and bleomycin in cells transfected with the motif is reminiscent of the hypersensitivity of PARP1 knockout and haploinsufficient mice toward DNA-damaging agents,18, 20 reflecting compromised DNA repair with the loss of PARP1 enzymatic function. The ability of the PARP1 motif to specifically disrupt cellular PARP1 function was also demonstrated by diminished HBs expression in HepG2 cells cotransfected with HBV-RFP (red fluorescent protein) (Supporting Fig. 7). To confirm that the effects of the HBVCP-PARP1 motif are specific to PARP1, rescue experiments were performed, in which PARP1 was overexpressed to compensate for the loss of DNA repair. Excess PARP1 cannot avert the accumulation of cytotoxic DNA lesions if alternative DNA repair pathways were instead compromised. Using apoptotic cell death as the end-point of extensive irreparable DNA damage, the effect of etoposide or bleomycin on HepG2 cells cotransfected with the HBVCP-PARP1 binding motif and PARP1 or RFP expression vectors was determined by apoptotic caspase-dependent cleavage of luminogenic substrates.

2) These results indicate that miR-7 may arrest cell-cycle progr

2). These results indicate that miR-7 may arrest cell-cycle progression by repressing p110δ expression. To verify our observations, we established relevant stable

subclones in QGY-7703, which were named QGY-null (mock), QGY-miR-NC (noneffective control), and QGY-miR-7, respectively. Ectopic expression of miR-7 was elevated by https://www.selleckchem.com/products/AC-220.html approximately 7-fold (Supporting Fig. 3A), which resulted in a 0.24-fold reduction of PIK3CD mRNA (Fig. 2B). Western blotting analysis showed that miR-7 specifically repressed p110δ protein expression (Fig. 2B), but did not affect the expression of the other two p110 catalytic subunits (p110α and p110β) or their corresponding regulatory subunit, p85 (Supporting Fig. 3B). We further investigated the effect of the stable expression of miR-7

on HCC cell growth in vitro. Using the cell-proliferation assay, we observed a significant decrease in cell number in QGY-miR-7 cells (538.8 ± 39.0 × 103, n = 3; P < 0.01) versus QGY-null cells (1,164 ± 34.1 × 103, n = 3; P < 0.01) or QGY-miR-NC cells (949 ± 48.1 × 103, n = 3; P < 0.01) on day 7 (Fig. 2C). No apoptosis was observed on day 4 (Supporting Fig. 3C) when miR-7 was stably expressed, indicating that the decrease in cell numbers might be caused by the arrest of cell-cycle progression (Fig. 2C). A similar inhibition in cell proliferation was observed in the PIK3CD siRNA#3 group, but not in the control siRNAs (Supporting see more Fig. 4). To further validate our results, we assayed for alterations in cell-cycle progression every 2 hours for 24 hours after 30 hours of serum starvation (Fig. 2D). A G0/G1 cell-cycle arrest that was detected in QGY-miR-7 cells was associated with miR-7 overexpression. It took QGY-miR-7 cells 8-9 hours to recover after serum starvation (G0/G1 ≤60%), whereas the controls recovered in approximately 5 hours, and the percentage Pyruvate dehydrogenase of cells in the G0/G1 phase remained over 50% and had no significantly periodic change

when miR-7 was stably expressed, which was obviously higher than those in S or G2/M phase (Fig. 2D, top). By analyzing changes in the cell proportion in S or G2/M phase, we found that QGY-miR-7 required 14 hours to complete a cell cycle after serum recovery, compared to approximately 12 hours for control cells (Fig. 2D, middle and bottom). All the results were consistent with those observed in transient transfection experiments. These data strongly suggest that miR-7 inhibits HCC cell growth by G0/G1 arrest, but not by triggering apoptosis. We further investigated whether overexpression of miR-7 could weaken the invasiveness and migratory capabilities in HCC. Using the wound-healing assay (Supporting Materials and Methods), we found that ectopic expression of miR-7 decreased cell motility in QGY-miR-7 cells, compared to QGY-null and QGY-miR-NC cells (Supporting Fig. 5).

Lastly, an important new study in the next review period, highlig

Lastly, an important new study in the next review period, highlighted only briefly here, has shown that the T4SS, independently of CagA signalling,

upregulates the cancer-related miRNA, miR-155. miR-155 has reported antiapoptotic effects in immune cells, and therefore, modulation of its expression by the T4SS as revealed by Koch et al. may have direct influence on the regulation of apoptosis BVD-523 order during H. pylori infection [42]. Considering the role of dysregulated protein kinase C (PKC) signalling in gastric cancer, a recent study has shown that H. pylori induces phosphorylation of PKC isoforms and their substrates [43]. Interestingly, H. pylori-mediated PKC activation upregulated matrix metalloproteinase-1 (MMP-1) expression. In turn, this increased the invasion of AGS cells suggesting a mechanism by which PKC activation promotes remodelling and destruction of gastric tissue in response to H. pylori infection independently of CagA signalling. Promotion of cell invasion is also indicated to occur via calpain protease-mediated disruption of adherens junctions in response to TLR2 stimulation by an as yet unidentified H. pylori outer membrane protein [44]. Other work Sorafenib nmr examining the secreted

HtrA protease has demonstrated functional conservation of its E-cadherin cleavage activity among a range of other gastrointestinal pathogens [45]. Cleavage specificity was shown to be a function of structural conservation within the active site of the protein, indicating that HtrA-mediated E-cadherin cleavage is a conserved mechanism underlying different pathogenic strategies. A virtual screening approach has also been successful in identifying several small

molecule inhibitors of H. pylori HtrA activity [46]. The vacuolating cytotoxin, VacA, is a major virulence factor of H. pylori and has pleiotropic effects in target host cells. Of these, the involvement of VacA in various mechanisms of programmed cell death including apoptosis and necrosis has attracted particular attention. Investigating VacA-targeting of mitochondria, Jain et al. show that VacA induces apoptosis through disruption of selleck chemicals llc mitochondrial morphological dynamics by inducing the activation of dynamin-related protein 1 (Drp1) [47]. Drp1 regulates mitochondrial fission and, once activated, locates to the mitochondrial outer membrane. VacA increases Drp1 localization indicating that the previously observed VacA-dependent fragmentation of the mitochondrial network involves the cellular fission machinery. The membrane channel activity of VacA was found to be important in this respect. VacA-induced cell death may therefore proceed via a mechanism of enhanced Drp1-dependent fission promoting activation of the proapoptotic Bcl-2 effector Bax and mitochondrial outer membrane permeabilization [47]. Examining morphological and biochemical markers of both necrotic and apoptotic cell death, Radin et al.

However, based on current knowledge, it cannot be excluded that a

However, based on current knowledge, it cannot be excluded that antibody responses against proteins such as FVIII could also occur in a T-cell independent way. Such antibodies should be of low affinity due to the lack of affinity maturation. “
“Summary.  Effective treatment with factor IX (FIX) requires a thorough consideration of the properties of the concentrate to be used as replacement therapy, to date, the only available treatment for haemophilia B. The aim of the study was to determine the pharmacokinetics, clinical efficacy and safety in routine clinical use of AlphaNine®, a high-purity human FIX concentrate. This open,

single-arm, multicentre, non-randomized trial included 25 subjects (age ≥ 12) with moderate/severe haemophilia B. Pharmacokinetics was assessed at baseline and after a 6-month follow-up. The degree of haemostasis control DAPT clinical trial achieved was evaluated during a 12-month follow-up. Safety was evaluated in terms of tolerance, thrombogenicity, immunogenicity and viral safety. Mean recovery was 1.01 ± 0.19 IU dL−1 per IU kg−1 at baseline and 1.23 ± 0.34 IU dL−1 per IU kg−1

6 months later. Terminal half-life was 34.5 ± 6.2 h and 33.7 ± 5.4 h, selleck inhibitor respectively. Ratios of each parameter between the two pharmacokinetic studies were all close to 1. A total of 1,576,890 IU AlphaNine® were administered in 889 infusions (mean dose per infusion: 1774 IU; 3.2 infusions per month per patient). The main reasons for infusion were mild/moderate bleeding (62.3%) and prophylaxis (20.5% continuous, 15.6% intermittent). Overall, 93.0% of the efficacy assessments were rated

as excellent/good and 88.8% of bleedings resolved after the first infusion. Twenty-one adverse events were reported in eight patients, none of which was considered related to the study medication. AlphaNine® showed a pharmacokinetic profile Roflumilast in agreement with that of other plasma-derived FIX concentrates and provides safe and clinically effective substitution therapy for patients with haemophilia B. “
“The temporary correction of the coagulation defect is the mainstay of treatment in hemophilia. However, the “ideal” dose of factor VIII (FVIII) or factor IX (FIX) that needs to be administered to invariably achieve hemostasis without “overtreating” is unknown. Dosing for hemophilia has been studied since the 1940s when initial data was based on the use of plasma in hemophilic dogs. These observations were fundamental for the understanding of dosing in prophylaxis. Years later, the use of plasma in hemophilia patients was instated followed by cryoprecipitate and a fraction of human plasma in the 1960s.

Q-RT-PCR was performed to detect ISGs 6 hours posttreatment HepG

Q-RT-PCR was performed to detect ISGs 6 hours posttreatment. HepG2 cells transfected with pEco63-1.3 (HBV 1.3x expression plasmid constructed using HBV sequence from pEco63) were treated with cTCR-L/IFNα ± 10 μg/mL HBc18-27 peptide, Roferon, or Peg-IFNα (Pegasys). After 72 hours the viral supernatant was collected and S-antigen was quantified using an HBsAg chemiluminescence Maraviroc immunoassay kit. HBV-specific CD8 T cells were cocultured with HBV peptide-pulsed or not pulsed HepG2 cells

with TCR-L/IFNα, fixed, and stained for IFNγ-PE. HepG2 were incubated with HBV-specific CD8 T cells alone or with TCR-L/IFNα overnight. Supernatants were collected after 18 hours and concentrations of CXCL-9 and CXCL-10 were measured using the Cytometric Bead Array System (BD Biosciences, San Jose, CA). In selected experiments, intracellular cytokine staining using fluorescent-conjugated anti-CXCL-10 antibodies was used. We recently reported the production and characterization click here of a murine IgG1 antibody specific for the surface HBs183-91/A*02:01 complex (sTCR-L).11 A second antibody specific for core HBc18-27/A*02:01 complex, a dominant HLA-A201 HBV-epitope, was produced using the same method. Figure 1 shows the specificity data of both cTCR-L (specific for HBc18-27/A*02:01) and sTCR-L (specific for HBs183-91/A*02:01). Both TCR-Ls selectively recognize

HLA-A*02:01+ targets pulsed with the respective specific peptides (Fig. 1A). In addition,

both TCR-Ls bound to HBV-producing HepG2 cells, but did not bind to HepG2 cells that had not been transfected with HBV (Supporting Fig. 1) or cells pulsed with other A*02:01 Avelestat (AZD9668) binding peptides. The specific recognition of HBc18-27 pulsed cells or HBV-producing cells by cTCR-L antibodies was not influenced by the presence of serum from CHB patients (data not shown), as demonstrated for sTCR-L.11 The two antibodies were tested for their ability to recognize naturally infected cells by immunohistochemistry on frozen liver biopsies from patients with CHB (Fig. 1B) or by staining of isolated hepatocytes purified from CHB patients biopsies (Fig. 1C). Both antibodies specifically recognized, with variable frequencies, the hepatocytes of HLA-A*02:01+ patients with CHB, but they did not bind to hepatocytes purified from HLA-A*02:01-negative subjects (Fig. 1B,C). The possible broadness of applicability of both cTCR-L and sTCR-L in patients of different ethnicities infected by different HBV genotypes was studied by analyzing the TCR-Ls ability to recognize the peptides of the respective HBc18-27 and HBs183-91 epitopes of HBV genotypes A, B, C, D, E, and F presented by different HLA-A*02 allotypes. Amino acid sequences of the corresponding peptides are shown in Fig. 1D with a description of the HLA-A02* subtypes present in distinct human populations listed in Fig. 1D.

23 A similar mechanism might also be applicable to HCC cells as w

23 A similar mechanism might also be applicable to HCC cells as well as in Alb/AEG-1 hepatocytes, and the monoubiquitination of overexpressed AEG-1 was confirmed (Supporting Fig. 9). The promiscuous accumulation of AEG-1 in the cytoplasm might facilitate an interaction with the translational machinery and loading of selective mRNAs to the polysome.

Indeed, ribosomal proteins as well as eukaryotic translation initiation factors were identified as potential AEG-1-interacting proteins, indicating a potential direct role of AEG-1 in regulating translation.8 It is intriguing that AEG-1 facilitates the translation of multiple members of the coagulation pathway, all of which are known mediators of tumor growth, metastasis, and angiogenesis, and this particular aspect of AEG-1 function might play a pivotal role in promoting tumor progression and metastasis. Plasma FXII analysis selleck screening library thus might be a potential biomarker for HCC. We observe that knocking down either FXII or TFF3 results in a marked inhibition of AEG-1-induced angiogenesis. Interestingly, both FXII and TFF3 interacts with EGFR on ECs to augment proliferation and differentiation, BAY 80-6946 hence angiogenesis.21, 24 Thus, there might be a key role of endothelial EGFR

in mediating AEG-1 function, a hypothesis that needs to be experimentally validated. One novel aspect of AEG-1 function is the induction of steatosis. Nonalcoholic fatty liver disease (NAFLD) is one of the precursors leading to nonalcoholic steatohepatitis and HCC.25 It will be interesting to check whether AEG-1 is also overexpressed in NAFLD patients, thus contributing to eventual hepatocarcinogenesis. Apart from significant increases in the expression of some components of fatty acid metabolism, our gene-expression network analysis did not identify the modulation of any major adipogenic or lipogenic pathway, such as the peroxisome proliferator-activated receptor

gamma, liver X receptor, Astemizole or pregnane X receptor pathways. This observation argues that rather than affecting a network, AEG-1 overexpression might lead to promiscuous increases in distinct regulators of fat metabolism resulting in steatosis. The significant increase in SCD2 expression by AEG-1 alone might contribute to steatosis. Induction in SCD2 has also been observed in the transforming growth factor alpha/c-myc TG mouse model of HCC.26 SCDs are crucial lipogenic enzymes for monounsaturated fatty acid biosynthesis. SCD1 expression is induced after weaning in mouse liver, whereas SCD2 expression is detected in livers of mouse embryos and neonates.27 There is a significant reduction in liver and plasma triglycerides in neonatal SCD2 KO mice.27 The increased SCD2 expression by AEG-1 suggests a shift toward embryonic gene-expression pattern, another hallmark of cancer. Crossing SCD2 KO mice with Alb/AEG-1 mice might provide insight into the importance of SCD2 in mediating the AEG-1-induced steatotic phenotype.

0mg As the rate of genotypic resistance to ETV (ETV-R) is report

0mg. As the rate of genotypic resistance to ETV (ETV-R) is reported high currently, HIF inhibitor changing ETV to the other agent may be needed even in patients whose resistance to ETV was not identified. However, appropriate indication and time point have not been proposed yet. This study was aimed to stratify ETV therapy in LAM-R patients. Methods: One hundred and nine CHB patients who developed LMV-R and then received ETV 1.0mg up to 5 years were evaluated prospectively. Virologic response (VR, HBV

DNA <20 IU/mL) and ETV-R during 5 years of treatment were evaluated as primary end points. We divided subjects into non-detection group (HBV DNA <20 IU/mL) and detection group (HBV DNA >20 IU/mL) at 6 months and 12 months of switching to ETV for prediction of long term response. Results: The mean age of the patients was 45±1 1 years,

the proportion of male and HBeAg-positive patient was Barasertib 72% (79/109) and 77% (84/109), respectively. VR rates were 0%, 20%, 24%, 32%, 37%, and 39% and the mean serum HBV DNA levels were 6.89±1.03, 3.26±1.81, 3.06±1.82, 2.49±1.53, 2.43±1.35 and 1.73±0.87 log 10 IU/ml at baseline, month 12, 24, 36, 48 and 60, respectively. Genotypic resistance to ETV occurred at 30±12 months (median 24 months, 12-66 months). Resistance rates were 4.6%, 19%, 32%, 36%, and 37% at baseline, month 12, 24, 36, 48 and 60, respectively. When we predicted long term responses according to delectability of HBV DNA at 6 months of treatment, VR (100% vs. 28%, p <0.001) was higher and ETV-R (0% vs. 45%, p = 0.001)

was lower in non-detection group than in detection group. Likewise, at 12 months of treatment, VR (96% vs. 21%, p <0.001) was higher and ETV-R (12% vs. 49%, p = 0.001) was lower in non-detection group than detection group. Multivariate analysis showed non-detection of HBV DNA at 6 months as well as 12 months were independent factors associated with VR. To evaluate predictive value for VR, area under the receiver operating characteristic Protein kinase N1 curve (AUROC) was used. AUROC (0.865; 95% confidence interval [CI], 0.789-0.940; P <0.001) of non-detection of HBV DNA at 12 months showed slightly better than that of 6 months (AUROC, 0.828; 95% confidence interval [CI], 0.742-0.914; P <0.001). Conclusion: Resistance rates were high in patients with detectable HBV DNA at 12 months of ETV therapy. Therefore, switching to or adding a potent nucleotide analogue (e.g. tenofovir) is warranted in LAM-R CHB patients whose HBV DNA is detected after 12 months of ETV therapy.