40
Elsevier Editorial System(tm) for Antiviral Research Manuscript Draft Manuscript Number: Title: NOVEL HBSAG MARKERS TIGHTLY CORRELATE WITH OCCULT HBV INFECTION AND STRONGLY AFFECT HBSAG DETECTION Article Type: Research Paper Section/Category: Host - Virus Interactions Keywords: HBV, Occult infection, HBsAg structure, HBsAg detection Corresponding Author: Dr. C.F. Perno, Corresponding Author's Institution: Universita' degli Studi di Roma First Author: Valentina Svicher Order of Authors: Valentina Svicher; Valeria Cento; Martina Bernassola; Maria Neumann-Fraune; Formijn Van Hemert; Mengjie Chen; Romina Salpini; Chang Liu; Roberta Longo; Michela Visca; Sara Romano; Valeria Micheli; Ada Bertoli; Caterina Gori; Francesca Ceccherini-Silberstein; Cesare Sarrecchia; Massimo Andreoni; Mario Angelico; Antonella Ursitti; Alberto Spanò; Jing Maria Zhang; Jens Verheyen; Giuseppina Cappiello; C.F. Perno Manuscript Region of Origin: ITALY Abstract: Occult HBV infection (OBI) is a threat for the safety of blood-supply, and has been associated with the onset of HBV-related hepatocellular carcinoma and lymphomagenesis. Nevertheless, genetic markers in HBsAg (particularly in D-genotype, the most common in Europe) significantly associated with OBI in vivo are missing. Thus, the goal of this study is to define: i) prevalence and clinical profile of OBI among blood-donors; ii) HBsAg-mutations associated with OBI; iii) their impact on HBsAg- detection. OBI was searched among 422,278 blood-donors screened by Nucleic-Acid-Testing. Following Taormina-OBI-definition, 26 (0.006%) OBI-patients were identified. Despite viremia<50IU/ml, HBsAg-sequences were obtained for 25/26 patients (24/25 Genotype-D). OBI- associated mutations were identified by comparing OBI-HBsAg with that of 82 chronically-infected (genotype-D) patients as control. Twenty HBsAg-mutations significantly correlated for the first time with OBI. By structural analysis, they localized in the major HBV B-cell-epitope, and in HBsAg-capsid interaction region. 14/24 OBI-patients (58.8%) carried in median 3 such mutations (IQR:2.0-6.0) against 0 in chronically-infected patients. By co-variation analysis, correlations were observed for R122P+S167L (phi=0.68,P=0.01), T116N+S143L (phi=0.53,P=0.03), and Y100S+S143L (phi=0.67,P<0.001). Mutants (obtained by site-directed mutagenesis) carrying T116N, T116N+S143L, R122P, R122P+Q101R, or R122P+S167L strongly decreased HBsAg-reactivity (54.9±22.6S/CO, 31.2±12.0S/CO, 6.1±2.4S/CO, 3.0±1.0S/CO and 3.9±1.3S/CO, respectively) compared to wild-type (306.8±64.1S/CO). Even more, Y100S and Y100S+S143L supernatants show no detectable-HBsAg (experiments in quadruplicate). In conclusions, unique HBsAg-mutations in genotype-D, different than those described in genotypes B/C (rarely found in western countries), tightly correlate with OBI, and strongly affect HBsAg-

Novel HBsAg markers tightly correlate with occult HBV infection and strongly affect HBsAg detection

Embed Size (px)

Citation preview

Elsevier Editorial System(tm) for Antiviral Research Manuscript Draft Manuscript Number: Title: NOVEL HBSAG MARKERS TIGHTLY CORRELATE WITH OCCULT HBV INFECTION AND STRONGLY AFFECT HBSAG DETECTION Article Type: Research Paper Section/Category: Host - Virus Interactions Keywords: HBV, Occult infection, HBsAg structure, HBsAg detection Corresponding Author: Dr. C.F. Perno, Corresponding Author's Institution: Universita' degli Studi di Roma First Author: Valentina Svicher Order of Authors: Valentina Svicher; Valeria Cento; Martina Bernassola; Maria Neumann-Fraune; Formijn Van Hemert; Mengjie Chen; Romina Salpini; Chang Liu; Roberta Longo; Michela Visca; Sara Romano; Valeria Micheli; Ada Bertoli; Caterina Gori; Francesca Ceccherini-Silberstein; Cesare Sarrecchia; Massimo Andreoni; Mario Angelico; Antonella Ursitti; Alberto Spanò; Jing Maria Zhang; Jens Verheyen; Giuseppina Cappiello; C.F. Perno Manuscript Region of Origin: ITALY Abstract: Occult HBV infection (OBI) is a threat for the safety of blood-supply, and has been associated with the onset of HBV-related hepatocellular carcinoma and lymphomagenesis. Nevertheless, genetic markers in HBsAg (particularly in D-genotype, the most common in Europe) significantly associated with OBI in vivo are missing. Thus, the goal of this study is to define: i) prevalence and clinical profile of OBI among blood-donors; ii) HBsAg-mutations associated with OBI; iii) their impact on HBsAg-detection. OBI was searched among 422,278 blood-donors screened by Nucleic-Acid-Testing. Following Taormina-OBI-definition, 26 (0.006%) OBI-patients were identified. Despite viremia<50IU/ml, HBsAg-sequences were obtained for 25/26 patients (24/25 Genotype-D). OBI-associated mutations were identified by comparing OBI-HBsAg with that of 82 chronically-infected (genotype-D) patients as control. Twenty HBsAg-mutations significantly correlated for the first time with OBI. By structural analysis, they localized in the major HBV B-cell-epitope, and in HBsAg-capsid interaction region. 14/24 OBI-patients (58.8%) carried in median 3 such mutations (IQR:2.0-6.0) against 0 in chronically-infected patients. By co-variation analysis, correlations were observed for R122P+S167L (phi=0.68,P=0.01), T116N+S143L (phi=0.53,P=0.03), and Y100S+S143L (phi=0.67,P<0.001). Mutants (obtained by site-directed mutagenesis) carrying T116N, T116N+S143L, R122P, R122P+Q101R, or R122P+S167L strongly decreased HBsAg-reactivity (54.9±22.6S/CO, 31.2±12.0S/CO, 6.1±2.4S/CO, 3.0±1.0S/CO and 3.9±1.3S/CO, respectively) compared to wild-type (306.8±64.1S/CO). Even more, Y100S and Y100S+S143L supernatants show no detectable-HBsAg (experiments in quadruplicate). In conclusions, unique HBsAg-mutations in genotype-D, different than those described in genotypes B/C (rarely found in western countries), tightly correlate with OBI, and strongly affect HBsAg-

detection. By altering HBV-antigenicity and/or viral-particle maturation, they may affect full-reliability of universal diagnostic-assays for HBsAg-detection. Suggested Reviewers: Vincent Calvez Department of Virology, Hospital Pitiè-Salpetriere, Paris, France [email protected] Charles Boucher Department of Virology, Erasmus University Rotterdam, the Netherlands [email protected] Hans Zaaijer Department of Clinical Virology, University of Amsterdam [email protected] Daniel Douek Department of Immunology, Vaccine Research Center, Bethesda [email protected]

Prof. Carlo-Federico Perno

Dr. Valentina Svicher

University of Rome Tor Vergata

e-mail: [email protected]

[email protected]

Dr Erik De Clercq

Editor, Antiviral Research

August 18th, 2011

Dear Erik,

On behalf of our co-authors, we submit the enclosed manuscript entitled “Novel HBsAg

markers tightly correlate with occult HBV infection and strongly affect HBsAg detection"

by V Svicher, V Cento, M Bernassola, M Neumann-Fraune, F Van Hemert, M Chen, R

Salpini, C Liu, R Longo, M Visca, S Romano, V Micheli, G Gubertini, A Bertoli, N

Marino, F Mazzotta, C Gori, V Micheli, C Sarrecchia, M Andreoni, M Angelico, A Ursitti,

A Spanò, F Ceccherini-Silberstein, JM Zhang, J Verheyen, G Cappiello, CF Perno for

potential publication in Antiviral Research.

The manuscript has not been published in this or a substantially similar form (in print or

electronically, including on a web site), nor accepted for publication elsewhere, nor it is

under consideration by another publication.

The philosophy of this study is based on a strong interdisciplinary approach joining

together a vertical network of researchers with particular and diverse skills, including

virologists, molecular biologists, biochemists, clinicians, and mathematicians. Therefore,

all people listed as authors actively participated in this study and each gave substantial

contribution at different levels in the conception, design and performance of the work.

Occult HBV infection (characterized by the persistence of HBV-DNA in the serum or liver

tissu in absence of HBsAg) is so far of clinical and pathological importance due to i) its

association with the progression of hepatitis B towards cirrhosis and hepatocellular

carcinoma despite the apparently low HBV-replication rate, ii) HBV-related

lymphomagenesis, iii) its potential risk of transmission in cases of transfusion of blood

products, and of orthotopic liver transplantation.

Several host and viral factors have been hypothesized for the OBI pathogenesis.

Nevertheless, conclusive studies on HBV genetic markers (in particular in the HBsAg)

responsible for Occult HBV infection in vivo are lacking.

In this light, this study is aimed at:

1. Defining the prevalence of occult HBV infection in a large cohort (N=422,278) of

blood donors tested by Nucleic Acid Testing procedure between 2004 and 2008 in

an important reference center for blood transfusion in Central Italy.

2. Providing a systematic and comprehensive characterization of HBsAg genetic

markers underlying occult D-genotype infection in-vivo by using an integrated

clinical, structural and mathematical approach in one of the largest cohort of

patients with OBI so far analyzed for this purpose. Twenty unique HBsAg-

UNIVERSITA' DEGLI STUDI DI ROMA "TOR VERGATA"

DIPARTIMENTO DI MEDICINA SPERIMENTALE E SCIENZE BIOCHIMICHE

Cover Letter

mutations in HBV D genotype were found for the first time significantly correlated

with OBI in-vivo. This is the first study showing such correlation of OBI with the

HBV genotype most commonly found in western countries. Of note, these HBsAg

genetic markers are different from those recently identified in HBV B and C

genotype in far East Asia (Yuan et al., 2010), thus supporting the concept that the

natural variability of HBV genotypes makes difficult the definition of universal

diagnostic markers of HBV.

3. Investigating the impact of such markers on the detection of HBsAg in the

supernatants of cell culture experiments, by means of a clinically-validated test

with a known sensitivity >99%. These in vitro experiments showed that these OBI-

related HBsAg mutations strongly decrease (up to abrogate) the amount of detected

HBsAg in the supernatants, thus potentially altering the full reliability of

diagnostic-assays for HBsAg-detection in blood-samples.

Overall, this information can be used for planning and implementation of screening of this

disease in transfusion services, and to further improve the rate of true-positive results by

standard assay for HBsAg detection in diagnostic practice

These data have been presented in part at the 46th

Annual Meeting of the European

Association for the Study of the Liver (EASL), held in Berlin, Germany, March 30-April 3,

2011, where the abstract was scored among the TOP 10%

For all these reasons we propose our paper to the attention of Antiviral Research.

Among scientists renowned for their expertise in the field, may we suggest the following

names:

Dr Vincent Calvez

Department of Virology,

Hospital Pitiè-Salpetriere, Paris, France

83 Boulevard de l’Ospital, F-75013 Paris

Tel: +33-1-42177416

E-mail: [email protected]

Dr Charles Boucher

Department of Virology,

Erasmus University Rotterdam, the Netherlands

Tel: +30 250 6526

E-mail: [email protected]

Dr Hans L Zaaijer

Department of Clinical Virology,

Academic Medical Center (AMC), Clinical Virology (L1-104),

University of Amsterdam, P.O.Box 22660, 1100DD Amsterdam, The Netherlands

Tel: +31-20-5669111

E-mail: [email protected]

Dr Daniel Douek,

Vaccine Research Center, Bethesda

Bldg 40

Room 3504

MSC 3022

Bethesda, MD 20892-3005

Tel: +1 301-594-8484

E-mail: [email protected]

Looking forward to hearing from you.

Yours sincerely,

Prof. Carlo Federico Perno, MD, PhD

and Dr. Valentina Svicher, PhD

Department of Experimental Medicine

University of Rome Tor Vergata

Via Montpellier 1, 00133 Rome, Italy

Tel: +39-06-72596566, 06-72596551

Fax: +39-06-72596039

E-mail: [email protected],

[email protected]

Highlights

HBsAg genetic markers correlated with occult HBV D genotype infection in vivo were

studied

Twenty HBsAg-mutations significantly correlated for the first time with OBI were found.

They localized in the major HBV B-cell-epitope, and in HBsAg-capsid interaction region.

They strongly affect, up to abrogate, HBsAg detection,

Thus, they can affect full-reliability of diagnostic-assays for HBsAg-detection.

*Highlights

1

NOVEL HBSAG MARKERS TIGHTLY CORRELATE WITH OCCULT HBV 1

INFECTION AND STRONGLY AFFECT HBSAG DETECTION 2

Running Title: HBsAg Markers in Occult HBV infection 3

Valentina Svicher1#

, Valeria Cento1#

, Martina Bernassola2, Maria Neumann-Fraune

3, 4

Formijn Van Hemert4, Mengjie Chen

5, Romina Salpini

1, Chang Liu

5, Roberta Longo

2, 5

Michela Visca2, Sara Romano

2, Valeria Micheli

6, Ada Bertoli

7, Caterina Gori

8, 6

Francesca Ceccherini-Silberstein1, Cesare Sarrecchia

7, Massimo Andreoni

7, Mario 7

Angelico7, Antonella Ursitti

2, Alberto Spanò

2, Jing Maria Zhang

5, Jens Verheyen

3, 8

Giuseppina Cappiello2, Carlo Federico Perno

1,7* 9

1 University of Rome “Tor Vergata” Rome, Italy; 2 “S. Pertini” Hospital, Rome, Italy; 10

3 University of Cologne, Cologne, Germany; 4 Center for Infection and Immunity 11

Amsterdam (CINIMA), Academic Medical Center, University of Amsterdam, 12

Amsterdam, the Netherlands; 5 Yale University, USA; 6 “L. Sacco” Hospital, Milan, 13

Italy; 7 University Hospital of Rome “Tor Vergata” Rome, Italy; 8 “L. Spallanzani” 14

Hospital, Rome, Italy 15

# equally contributed to the manuscript 16

Corresponds to: Carlo-Federico Perno, PhD, MD; University of Rome Tor Vergata; 17

Via Montpellier 1, 00133 Rome, Italy. Tel: +39 06-72596551; Fax: +39 06-72596039. 18

E-mail: [email protected]. 19

20

These data have been presented in part at the 46th Annual Meeting of the European 21

Association for the Study of the Liver (EASL), held in Berlin, Germany, March 30-April 22

3, 2011 where the abstract was scored among the 10%, and at the 2nd

International HIV 23

and Viral Hepatitis Drug Resistance Workshop held in Los Cabos, Mexico, June 8-11, 24

2011. 25

26

27

*ManuscriptClick here to view linked References

2

ABSTRACT 28

Occult HBV infection (OBI) is a threat for the safety of blood-supply, and has been 29

associated with the onset of HBV-related hepatocellular carcinoma and 30

lymphomagenesis. Nevertheless, genetic markers in HBsAg (particularly in D-31

genotype, the most common in Europe) significantly associated with OBI in vivo are 32

missing. Thus, the goal of this study is to define: i) prevalence and clinical profile of 33

OBI among blood-donors; ii) HBsAg-mutations associated with OBI; iii) their impact 34

on HBsAg-detection. OBI was searched among 422,278 blood-donors screened by 35

Nucleic-Acid-Testing. Following Taormina-OBI-definition, 26 (0.006%) OBI-patients 36

were identified. Despite viremia<50IU/ml, HBsAg-sequences were obtained for 25/26 37

patients (24/25 Genotype-D). OBI-associated mutations were identified by comparing 38

OBI-HBsAg with that of 82 chronically-infected (genotype-D) patients as control. 39

Twenty HBsAg-mutations significantly correlated for the first time with OBI. By 40

structural analysis, they localized in the major HBV B-cell-epitope, and in HBsAg-41

capsid interaction region. 14/24 OBI-patients (58.8%) carried in median 3 such 42

mutations (IQR:2.0-6.0) against 0 in chronically-infected patients. By co-variation 43

analysis, correlations were observed for R122P+S167L (phi=0.68,P=0.01), 44

T116N+S143L (phi=0.53,P=0.03), and Y100S+S143L (phi=0.67,P<0.001). 45

Mutants (obtained by site-directed mutagenesis) carrying T116N, T116N+S143L, 46

R122P, R122P+Q101R, or R122P+S167L strongly decreased HBsAg-reactivity 47

(54.9±22.6S/CO, 31.2±12.0S/CO, 6.1±2.4S/CO, 3.0±1.0S/CO and 3.9±1.3S/CO, 48

respectively) compared to wild-type (306.8±64.1S/CO). Even more, Y100S and 49

Y100S+S143L supernatants show no detectable-HBsAg (experiments in quadruplicate). 50

3

In conclusions, unique HBsAg-mutations in genotype-D, different than those described 51

in genotypes B/C (rarely found in western countries), tightly correlate with OBI, and 52

strongly affect HBsAg-detection. By altering HBV-antigenicity and/or viral-particle 53

maturation, they may affect full-reliability of universal diagnostic-assays for HBsAg-54

detection. 55

Keywords: HBV, Occult infection, HBsAg structure, HBsAg detection 56

1. INTRODUCTION 57

Chronic overt hepatitis B is characterized by detectable hepatitis B surface 58

antigen (HBsAg) in the serum, while occult HBV infection (OBI) is defined by the 59

persistence of HBV-DNA in the liver tissue (and in some cases also in the serum) in 60

absence of HBsAg (Raimondo et al., 2010). OBI is one of the most challenging topics 61

in the field of viral hepatitis. Indeed, OBI can be transmitted in cases of transfusion of 62

blood products, as well as during orthotopic liver transplantation, and it has been 63

associated with the progression of chronic liver damage towards cirrhosis and 64

hepatocellular carcinoma despite the apparently low HBV-replication rate (Raimondo et 65

al., 2008). Indeed, OBI has been shown to maintain the pro-oncogenic properties typical 66

of the chronic infection, and has been identified in up to 70.4% of HBsAg-negative 67

patients with hepatocellular carcinoma (Fang et al., 2009). In addition, it has been 68

shown that lymphotropism is a major feature of OBI, and that a relationship may exist 69

between OBI and lymphomagenesis (Engels et al., 2010). Finally, patients with OBI are 70

at potential risk of HBV reactivation during immune-suppressive therapy in the future, 71

especially in regimens containing rituximab (Hoofnagle et al., 2009). This supports the 72

need to set up effort for a correct detection of OBI. 73

4

OBI is also a potential threat for the safety of blood supply. The prevalence of OBI in 74

blood donors has been reported in some European Countries such as Poland, Germany, 75

and France (Broyer et al., 2006; Katsoulidou et al., 2009; Levast et al., 2010), while in 76

others, such Italy, recent data are limited (Velati et al., 2008; Dettori et al., 2009). This 77

information is critical in order to set up optimized transfusion services. 78

Several host and viral factors have been hypothesized for the OBI pathogenesis 79

(Blum et al., 1991; Chaudhuri et al., 2004; Zaaijer et al., 2008; Vivekanandan et al., 2008; 80

El Chaar et al., 2010). Nevertheless, conclusive studies on HBV genetic markers (in 81

particular in the HBsAg) responsible for OBI in vivo are lacking. This can be due to a 82

number of factors: 1. The number and type of patients studied: the majority of reports 83

included a limited number of patients infected with different HBV genotypes, thus 84

making difficult to extrapolate the effect of mutations from the different genetic 85

background; 2. The technology used for viral genome sequencing: not widely available 86

for low level viremia; 3. The analysis of different fragments of HBV genome and the 87

methodologies used to characterize HBV genetic variability: the majority of studies 88

reported single mutations correlated with occult HBV infection without providing a 89

more complex level of information regarding the cluster of mutations underlying occult 90

HBV infection in-vivo. 91

Thus, and despite its clinical relevance, the genetic characteristics in HBsAg 92

from patients with occult HBV is still poorly known. This is particularly true for HBV, 93

D-genotype infection, that is the most prevalent in Mediterranean Countries including 94

Southern Europe. 95

In this light, using extensive sequence analyses, unique computational methods, 96

and phenotypic characterization, this study is aimed i) at defining the extent of occult 97

5

HBV infection in asymptomatic blood donors in a Country with low HBV endemicity, 98

ii) at identifying HBsAg-genetic markers specifying occult HBV infection in-vivo in 99

one of the largest homogenous cohort of OBI-patients so far analyzed, iii) as proof of 100

concept, at investigating the impact of such markers on the detection of HBsAg in the 101

supernatants of cell culture experiments. 102

Overall, this information can provide new insights regarding the pathogenesis of OBI, 103

and can be used to further improve the rate of true-positive results by standard assay for 104

HBsAg detection in diagnostic practice. 105

106

2. METHODS 107

2.1 Patients. The present study was carried out in two cohorts involving a total of 108

422,278 regular and occasional blood donors, screened for HCV, HIV-1/2 and HBV 109

infection by Nucleic Acid Testing (NAT) at a reference center for transfusion security in 110

Lazio region. The first cohort involved blood donors who donated blood between 111

December 2004 and April 2007 These samples were tested using polymerase chain 112

reaction (PCR)-based HIV-1/HCV/HBV test (COBAS Ampliscreen, Roche 113

Diagnostics, Branchburg, NJ; lower limit of detection [LOD]: 5 IU/mL, 95% 114

confidence interval: 3-13 IU/mL) in pools of 20 samples, generated with Tecan Genesis 115

pipetting machine (Tecan Schweiz AG, Männedorf, Switzerland). HBV-DNA reactive 116

pools were resolved by minipools of 5 samples testing. Donations of minipool, found to 117

be NAT-reactive, were tested individually to identify the positive unit. 118

The second cohort of donors, who donated blood between March 2007 and December 119

2008, were screened in minipools of 6 samples using the COBAS Taqscreen MPX 120

(Roche Molecular Systems, Branchburg, NJ; LOD: 3.1 IU/mL, 95% confidence 121

6

interval: 1.7-7.8 IU/mL) test on the COBAS s201 system for the direct detection of 122

HIV1-2/HCV/HBV (Roche Instrument Center, Rotkreuz, Switzerland). When a 123

minipool was reported as reactive, the 6 donation were individually tested with the 124

COBAS Taqscreen MPX test on the COBAS s201 system as a part of the resolution 125

process to identify the reactive and non-reactive donor sample. 126

Any individual specimens found to be reactive was tested using HIV1/HCV/HBV 127

COBAS Ampliscreen (Roche Diagnostics, Branchburg, NJ) for viral discrimination. 128

HBV-DNA was quantified by quantitative PCR COBAS Taqman High Pure System 129

HBV (Roche Molecular Systems, Branchburg, NJ; LOD: 6 IU/mL) between December 130

2004 and December 2005 and by COBAS Ampliprep/COBAS Taqman HBV test 131

(Roche Molecular Systems, Branchburg, NJ; LOD: 12 IU/mL) between November 2005 132

and December 2008. 133

Confirmed HBV-DNA positive samples were retested for serological markers of HBV 134

infection (HBsAg, anti-HBc, anti-HBc-IgM, HBeAg, anti-HBe, anti-HBs) using 135

specific tests on the Roche Elecsys 2010 immunoassay analyzer (Roche Diagnostics, 136

Branchburg, NJ). 137

2.2 HBsAg-DNA sequencing. HBsAg-DNA sequencing was performed following the 138

methodology reported in (Weinberger et al., 1999). Specific primers and amplification 139

protocols are reported in detail in the supplementary text 1. Translated HBsAg-140

sequences were generated and aligned using Bioedit 7.0 software and CLUSTALW1.8. 141

All sequences are in process to be submitted to GenBank. A phylogenetic analysis was 142

performed to exclude possible contaminations. 143

2.3 Genetic variability analysis. 144

7

Shannon entropy (Sn = -Σi(pi lnpi)/lnN) was calculated, where pi was the frequency of 145

each distinct nucleotide sequence and N was the total number of sequences analyzed. 146

Statistical significance was assessed trough Wilcoxon rank test. Evolutionary 147

divergence of HBsAg-sequences was estimated as the extent of nucleotide substitutions 148

per site determined by the HKY model of MEGAv5.0 (Tamura et al., 2011), applying a 149

gamma distribution with shape parameter = 0.8. The nested codon models 7 and 8 of 150

PAML 3.15 (Yang et al., 2007) were used to calculate dN/dS ratio values. The nwk user 151

tree was generated in MEGA5 (amino acid alignment, JTT replacement model), 152

CodonFreq F3x4 was chosen and gaps were treated as “missing data” (cleandata=0). 153

Likelihood Ratio Tests were performed to verify a better fit of the data with model 8 154

(extra size class (dN/dS >1) with respect to the null-hypothesis (model 7, 10 size 155

classes, dN/dS =<1). 156

2.4 Statistical analysis. 157

(i) Mutation prevalence. To assess the association of HBsAg-mutations with occult 158

infection, we calculated the prevalence of all amino acids (including the wild-type 159

amino acids) found at each HBsAg-position in the 24 sequences from occult infected 160

patients, and 82 sequences from chronically infected patients. We then performed chi-161

squared tests of independence with a Benjamini-Hochberg correction (false discovery 162

rate, FDR=0.05) to identify statistically significant differences in frequency between the 163

two groups of patients. 164

(ii) Mutation covariation. Binomial-correlation coefficient (phi) was calculated to 165

assess covariation among mutations associated with occult infection (Svicher et al., 166

2009). Statistically significant pairs of mutations were identified by Fisher’s exact test, 167

corrected for multiple-testing by Benjamini-Hochberg method (FDR=0.05). 168

8

(iii) Cluster analysis. In order to identify and summarize higher-order interactions of 169

mutations, we transformed the pairwise phi correlation coefficients into dissimilarity 170

values. A dendrogram was then computed by hierarchical clustering, and its stability 171

was assessed from 100 bootstrap replicates. The details of this explorative data analysis 172

procedure have been described elsewhere (Svicher et al., 2009). 173

2.5 Bayesian Variable Partition Model and Recursive Model Selection. Bayesian 174

Variable Partition Model and Recursive Model Selection were carried out according to 175

the methodology described in (Zhang et al., 2010). A detailed description of these 176

models is reported in supplementary text 2. 177

2.6 HBsAg three-dimensional structure prediction. An in silico generated tertiary 178

structure of HBV S-protein was available including the amino acid residues comprising 179

the interface composition of a surface-capsid complex (Roy et al., 2010). The amino 180

acid replacements described to be associated with occult HBV infection (this paper) 181

were manually introduced into the sequence. To generate a 3D model of surface protein 182

carrying these mutations, the file was submitted to I_TASSER for homology modeling 183

with the original "D-consensus" structure as a custom modeling template. Residues are 184

colored according to membrane topology, A-determinant size and surface-capsid 185

interface composition as described in (Roy et al., 2010). Hydrophobicity profiles were 186

constructed using values normalized between 0 and 1 (Black et al., 1991). A sliding 187

window of 5 residues with a step size of 1 was applied. 188

2.7 Site-specific mutagenesis and HBsAg detection assay. The expression vector 189

pCHsAg (genotype D) under the control of cytomegalovirus immediate early promoter-190

enhancer was used as wild type control and as backbone vector for the expression of 191

HBsAg. Single or double HBsAg-mutations were introduced by site directed 192

9

mutagenesis in the expression vector and confirmed by sequence reactions. Wild type 193

and HBsAg mutants were tested by transfection of human HuH7 cells with pCHsAg 194

plasmids using FuGene6 (Roche diagnostics GmbH, Mannheim, Germany). The 195

supernatant was collected after 72 hours and tested using the ABBOTT Architect 196

HBsAg (Qual) assay. HBsAg was tested in cell-supernatant obtained from four 197

independent experiments for each mutant and wild type. These results were used to 198

calculate the mean value and standard deviation for each HBsAg mutant. 199

200

3. RESULTS 201

3.1 Prevalence of Occult infection and Description of the Study Population 202

The frequency of HBV-DNA positive/HBsAg negative donations was assessed in a 203

large cohort of blood donors recruited in an important reference center for transfusion 204

security in Central Italy. 205

Among 422,278 blood donors tested, 269 (0.06%) were positive for serum HBV-DNA 206

by NAT procedure. After HBsAg testing, 243/269 (90.3%) patients were HBV-DNA-207

positive and HBsAg-positive (thus with diagnosis of overt chronic HBV hepatitis), 208

while the other 26 patients (0.006%) were HBV-DNA-positive and HBsAg-negative. 209

No significant difference in the prevalence of HBV-DNA-positive and HBsAg-negative 210

patients was observed in the two cohorts of blood donors (0.005% in the first cohort 211

screened between 2004-2007 versus 0.008% in the second cohort screened between 212

2007-2008). Following the position paper issued by the recent meeting of experts in 213

Taormina (Raimondo et al., 2008), OBI is associated with undetectable or low level 214

HBV-DNA (<200IU/ml) in the serum of HBsAg negative individuals. For 22/26 215

patients (84.6%), serum HBV-DNA quantification was <12IU/ml, while for the 216

10

remaining 4 patients, viremia ranged from 30 to 50IU/ml (Table 1). ALT values were in 217

the normal range. Therefore, these 26 HBV-DNA-positive and HBsAg-negative patients 218

were considered as occult HBV-infected patients. 219

Demographic and serological characteristics of the 26 blood donors with OBI are 220

summarized in Table 1. The majority were Italian (25/26, 96.2%), and male (22/26, 221

84.6%). All blood donors with OBI were anti-HBc positive, and anti-HBc IgM 222

negative, thus indicating that they were not in the window phase of acute hepatitis B 223

infection. Eleven out of 26 (42.3%) patients also showed low level of protective anti-224

HBs (>10mUI/ml) (Median [IQR]= 28.0 [20.0-54.0]). 225

Despite HBV-DNA<50IU/ml, HBsAg-DNA sequences were obtained for 25/26 (96%). 226

Direct HBsAg sequencing showed that genotype-D was dominant (24/25, 96.0%), 227

while genotype-A was present only in 1 patient. 228

3.2 HBsAg genetic markers underlying OBI in-vivo. 229

HBsAg genetic characteristics underlying OBI in-vivo were investigated. In order to 230

make data more clear and readable, the analysis was focused on 24 patients with occult 231

HBV D-genotype infection (thus excluding the only genotype-A patient). A group of 82 232

randomly-selected, chronically HBV D-genotype infected patients was used as control. 233

All chronically infected patients were HBsAg-positive, anti-HBc positive with a median 234

(IQR) serum HBV-DNA of 5.9 (3.3-6.2) logIU/ml (Table S1). All of them were naïve 235

to any anti-HBV drug-treatment. 236

A higher degree of genetic variability was found in HBsAg-sequences from OBI 237

compared to patients with chronic HBV infection. Indeed, both the median Shannon 238

Entropy, the median evolutionary rate, and the dN/dS ratio were remarkably higher in 239

patients with OBI than in patients with chronic HBV infection (median Shannon 240

11

Entropy (IQR): 0.29 (0.00-0.54) versus 0.07 (0.00-0.18), P=0.000068; median 241

evolutionary divergence (IQR): 0.06 (0.03-0.08) versus 0.02 (0.02-0.04), P=0.0003; 242

dN/dS: 0.84 (0.74-2.36) versus 0.32 (0.15-0.59), P=4.863∙10-29

) (Fig.1). 243

This result prompted us to investigate the existence of HBsAg markers specifying OBI 244

in-vivo. 245

Overall, 20 HBsAg-mutations were found for the first time significantly correlated with 246

occult HBV D-genotype infection in-vivo (Fig.2A) (P-values from 10-2

to 10-4

after 247

correction for multiple comparison). Fourteen out of 24 (58.8%) patients with OBI (vs. 248

only 1/82 chronic infected patients) carried >1 of these mutations (median number 249

[IQR]: 3.0 [2.0-6.0]). Conversely, in our cohort of 82 chronically-infected patients, such 250

mutations were completely absent (0/82 for Y100S, P105R, T116N, P120L, R122P, 251

T126I, Y134C, S143L, S167L, R169H) or nearly absent (1/82 for T115N, P127H, 252

S174N, V177A, 2/82 for Q129P, and 3/82 for Q101R, P127L, L175S, one mutation per 253

patient). Their prevalence ranged from 8.3 to 20.8% in OBI patients, compared to 0-254

3.7% in the 82 chronically-infected patients. 255

A Bayesian variable partition model (Zhang et al., 2010) was then applied to further 256

confirm whether HBsAg-positions are associated with OBI. All HBsAg-mutations 257

identified in the above-mentioned analysis were fully confirmed by this model. Indeed, 258

all mutations showed a posterior probability ranging from 0.75 to 1, indicating a strong 259

association with occult infection (data not shown). 260

3.3 Localization of HBsAg residues in the predicted HBsAg three-dimensional 261

structure. 262

To gain insight into effects of OBI-associated amino acid replacements on HBsAg, we 263

introduced these replacements in the sequence and built a 3D-model of mutant HBsAg 264

12

for comparison with D-consensus HBsAg. Fig.2B shows that OBI-associated residues 265

are mainly located at positions involved in antibody recognition and capsid interaction. 266

In the 3D-structure of mutant HBsAg (Fig.2C), the luminal loop including the A-267

determinant region displays a "collapse" towards the hydrophobic center of the 268

molecule. The architecture at the capsid-interaction regions is less modified. A similar 269

view is presented by the hydrophobicity profiles (Fig.2D) showing a replacement of 270

hydrophilic into more hydrophobic residues particularly at the A-determinant region 271

(110-140). 272

3.4 Specific associations among HBsAg markers. 273

A further step of this study was to investigate the complex interaction patterns of 274

HBsAg-mutations associated with OBI in-vivo. This is the first study addressing this 275

point. 276

Associations among HBsAg-mutations. To identify significant patterns of pairwise 277

correlations between mutations at the above mentioned positions, we calculated the 278

binomial correlation coefficient (phi) and its statistical significance for each pair of 279

mutations. A positive and statistically significant correlation between mutations at two 280

specific positions (0<phi<1, P< 0.05) indicates that these two positions co-evolve; thus 281

it indicates that the co-occurrence of mutations is not due by chance. A strong and 282

significant positive association of S143L with either T116N (phi=0.68) or Y100S 283

(phi=0.66), and of R122P with either T127S (phi=0.84), Q101R (phi=0.56), or S167L 284

(phi=0.56) was found. A significant correlation was also observed between L175S and 285

V177A, both localized in the HBsAg hydrophobic C-terminus (phi=0.56). 286

Clusters of correlated mutations. Because pairwise analysis suggested that some novel 287

mutations are associated with specific evolutionary pathways, an average linkage 288

13

hierarchical agglomerative cluster analysis was performed to investigate this hypothesis 289

in more detail. 290

The topology of the dendrogram (Fig.S1) highlighted the existence of two distinct 291

clusters of HBsAg-mutations in OBI. The first cluster included the mutations T116N 292

and S143L (bootstrap value=0.51), that were linked to Y100S (bootstrap=0.54). The 293

second pattern included the mutations S167L, and Q101R that were linked to T127S 294

and R122P (bootstrap value=0.50). 295

3.5 Phenotypic Analysis of HBsAg-mutation’s Patterns in Occult Infection 296

Cluster analysis highlighted the existence of specific cluster underlying OBI in-vivo. As 297

proof of concept, the ability of these mutation clusters to hamper HBsAg detection was 298

investigated in cell-culture using the ABBOTT Architect HBsAg (Qual) assay. 299

The detection rates of expressed HBsAg mutants significantly differed according to the 300

pattern of mutations analyzed (Fig. 3). The supernatants of mutants Y100S and 301

Y100S+S143L showed no detectable HBsAg (<1 sample/cutoff-ratio [S/CO]). A strong 302

decrease in the detection was also observed for HBsAg-mutants carrying T116N, 303

T116N+S143L, R122P, R122P+Q101R, or R122P+S167L. These mutations were 304

associated with remarkable decrease of HBsAg-reactivity from 306.8±64.1S/CO for 305

wild-type to 54.9±22.6S/CO, 31.2±12.0S/CO, 6.1±2.4S/CO, 3.0±1.0S/CO and 306

3.9±1.3S/CO, respectively (p<0.01 for all tests). By contrast, HBsAg-results for Q101R, 307

S167L and S143L were comparable to wild-type. 308

309

4. DISCUSSION 310

This study documented a prevalence of 0.057% of HBsAg-positive HBV-DNA 311

positive patients, and of 0.006% of OBI in a large cohort of blood donors tested by 312

14

NAT procedure, resolved by minipools of 5/6 samples, between 2004 and 2008. This is 313

in the range of prevalence of OBI estimated in Italy in previous studies (0.013%-314

0.006%) (Dettori et al., 2009; Velati et al., 2008) and in other European Countries, such 315

as Greece (0.0002%) and Poland (0.084%) (Brojer et al., 2006; Katsoulidou et al, 316

2009). 317

Several possible mechanisms have been hypothesized for the pathogenesis of 318

OBI and the condition is probably multi-factorial (Raimondo et al., 2008). Due to the 319

very small group of OBIs so far analyzed, HBsAg genetic markers in D-genotype 320

significantly associated with OBI in vivo are still missing. 321

By an integrated clinical, structural and mathematical approach, 20 mutations 322

(18 not previously correlated, and two previously associated with HBV immune-escape, 323

T123N and M133T) (Hou et al., 2001; Torresi et al., 2002) were found to be 324

significantly associated with occult HBV D-genotype infection in vivo, mainly located 325

in the “a-determinant” region. This is the first study showing such correlation of OBI 326

with an HBV genotype most commonly found in western countries. 327

The majority of these OBI-associated HBsAg genetic-markers were completely 328

absent in patients with chronic hepatitis. In addition, they occurred with a median 329

number [IQR] of 3.0 [2.0-6.0] in patients with OBI. The route towards the occult state 330

may be therefore characterized by an accumulation of variants, followed by a step-by-331

step loss of antigen-antibody reactivity (that is the basis of HBsAg detection test in 332

clinical practice). 333

In-vitro experiments showed that specific patterns of HBsAg-mutations (Y100S; 334

Y100S+S143L; T116N+S143L; R122P; R122P+Q101R and R122P+167L) strongly 335

15

decreased the amount of detected HBsAg in the supernatants, suggesting an impairment 336

in HBsAg-detection and/or changes in the antigenicity. 337

The impact of such mutation-patterns on HBsAg detection has not yet been 338

evaluated before. An important HBsAg function is the anchorage in the endoplasmatic-339

reticulum followed by assembly and secretion of mature virions (also HBsAg-mediated 340

processes). Notably, transmembrane helices and the hydrophobic C-terminus are largely 341

saved from extensive mutation in OBI. Conversely, we observed a linkage between 342

amino acid replacement near or at positions involved in capsid interaction (100, 101, 343

116, 122, 169, 174, 175, 177), suggesting a potential ability to impair viral particle 344

maturation and/or to hamper viral infectivity (van Hemert et al., 2008). Consistent with 345

this hypothesis, a recent in-vitro study showed the ability of mutations at position 169 346

(such as the R169H) to drastically hamper virion secretion, and to have a dominant 347

negative effect when it is co-expressed with wild-type envelope proteins (Ito et al., 348

2010). 349

For mutations T115N , T116N and T123N, the correlation with OBI can be 350

explained by the fact that these mutations introduce a novel N-liked glycosylation site, 351

that may impair virion secretion, along with reduction of HBsAg immunogenicity; 352

indeed T123N has been recently shown to strongly reduce virion assembly by leading to 353

the production of a new N-glycosylated form of HBsAg (Wu et al., 2010). 354

Residues associated with occult HBV D-genotype infection at positions 105, 355

120, 122, and 123 have been recently shown to be critical conformation-dependent 356

features for HBV entry into the target cells (Salisse et al., 2009). Mutations at these 357

residues have been found to drastically hamper (or even abrogate) in-vitro viral 358

infectivity (Salisse et al., 2009), thus supporting their association with OBI in-vivo. 359

16

Notably, while mutations in the “a-determinant” often resulted in non-360

synonymous mutations in the reverse transcriptase (RT) protein, the majority of the 361

amino acid substitutions involving capsid-interacting residues were silent in the RT. 362

From the evolutionary point of view, this observation points towards an accumulation of 363

mutations due to a relaxation of selective constraints and may indicate a contribution of 364

these regions to the evolutionary transition from a chronic into an OBI, observation also 365

supported by previous studies (van Hemert et al., 2008) and by the higher values of 366

evolutionary divergence and dN/dS found in occult infection respecting to chronic. 367

Functionally, a diminished interaction between core and surface proteins due to the 368

mutations introduced at these regions may support this process, rendering the transition 369

irreversible. 370

A recent study showed an association between OBI and immune-escape (El 371

Chaar et al., 2010), postulating that patients with OBI carrying both anti-HBc and anti-372

HBs, are under strong humoral immune-pressure. This selecting-pressure may lead to 373

the selection of viral strains with highly mutated HBsAg, in particular in the HBsAg 374

regions 125-131 and 158-169, crucial for HBsAg recognition and to reduce the 375

exposure of HBsAg immunogenic surface (Wu et al., 2010). These mutations may be 376

responsible for HBV evasion from the immune-system, and, at the same time, may 377

hamper viral replication, justifying the very low serum HBV-DNA levels classically 378

observed in OBI. Due to the quasispecies nature of HBV, the reactivation of viral 379

replication under immune-suppression may be mainly sustained by the re-emergence of 380

the wild-type strain. 381

OBI has been associated with the onset of HBV-related hepatocarcinogenesis 382

and lymphomagenesis (Fang et al., 2009; Engels et al., 2010). Further studies are thus 383

17

necessary to verify the ability of these mutations to modulate HBV oncogenic potential. 384

In addition, recent studies have highlighted new insights regarding HBV pathogenesis 385

(Neumann et al., 2010; Tian et al., 2010). In particular, a study shows that expression 386

and secretion of HBsAg is a novel mode of inducing secretion of Cyclophilin A 387

(CypA), that has been involved in the pathogenesis of HBV infection (Tian et al., 2010).

388

Another study identifies a novel antiviral mechanism of antibodies to HBsAg involving 389

prolonged blocking of the HBV and HBsAg subviral particles release from infected 390

cells (Neumann et al., 2010). The impact of HBsAg mutations correlated with OBI on 391

these two novel mechanisms underlying HBV pathogenesis should be matter of further 392

investigation. 393

A new Abbott ARCHITECT® assay for the detection of HBsAg has been 394

recently proposed to specifically enhance the detection of some HBsAg mutants at 395

position 122 and 123 (Lou et al., 2011). The impact of the other genetic markers we 396

identified on the performance of this new assay has not yet been defined. 397

It should be noted that such study has been conducted in OBIs all carrying D-398

genotype HBV. Therefore, the conclusions driven by these results may not be 399

applicable to other HBV-genotypes. Consistent with this hypothesis, a recent study, led 400

in China, identified specific HBsAg mutations (in the regions from amino acids 117 to 401

121 and amino acids 144 to 147) potentially associated with OBI-infection in HBV B- 402

and C-genotype. These mutations are completely different from those we identified in 403

HBV D-genotype (none among those described in genotype D are reported in genotype 404

B and C, and viceversa) (Yuan et al., 2010), thus supporting the concept that immune 405

escape and OBI can be driven by HBV-associated mutations different for each HBV 406

18

genotype. As a consequence, the natural variability of HBV genotypes may make 407

difficult the definition of universal diagnostic markers of HBV. 408

In conclusions, unique HBsAg-mutations in genotype-D, different than those 409

described in genotypes B/C (rarely found in Western-Countries), tightly correlate with 410

OBI, and strongly affect HBsAg-detection. These mutations can alter HBV-antigenicity 411

and/or viral-particle maturation, and can affect full-reliability of diagnostic-assays for 412

HBsAg-detection. Thus, the incorporation of these mutants in reagent development can 413

further improve the rate of true-positive results. 414

415

416

417

418

419

420

421

422

423

424

425

426

19

REFERENCES 427

1. Black, S.D., Mould, D.R., 1991. Development of hydrophobicity parameters to 428

analyze proteins which bear post- or cotranslational modifications. Anal. Biochem. 429

193, 72-82. 430

2. Blum, H.E., Galun, E., Liang, T.J., Wands, J.R., 1991. Naturally occurring missense 431

mutation in the polymerase gene terminating hepatitis B virus replication. J. Virol. 432

65, 1836-1842. 433

3. Brojer, E., Grabarczyk, P., Liszewski, G., Mikulska, M., Allain, J.P., Letowska, M., 434

2006. Characterization of HBV DNA+/HBsAg- blood donors in Poland identified by 435

triplex NAT. Hepatology 44, 1666-1674. 436

4. Chaudhuri, V., Tayal, R., Nayak, B., Acharya, S.K., Panda, S.K., 2004. Occult 437

hepatitis B virus infection in chronic liver disease: full-length genome and analysis 438

of mutant surface promoter. Gastroenterology 127, 1356-1371. 439

5. Dettori, S., Candido, A., Kondili, L.A., Chionne, P., Taffon, S., Genovese, D., 440

Iudicone, P., Miceli, M., Rapicetta, M., 2009. Identification of low HBV-DNA levels 441

by nucleic acid amplification test (NAT) in blood donors. J. Infect. 59, 128-133. 442

6. El Chaar, Candotti, D., Crowther, R.A., Allain, J.P., 2005. Impact of hepatitis B virus 443

surface protein mutations on the diagnosis of occult hepatitis B virus infection. 444

Hepatology 52, 1600-1610. 445

7. Engels, E.A., Cho, E.R., Jee, S.H., 2010. Hepatitis B virus infection and risk of non-446

Hodgkin lymphoma in South Korea: a cohort study. Lancet Oncol. 11, 827-834. 447

20

8. Fang, Y., Shang, Q.L., Liu, J.Y., Li, D., Xu, W.Z., Teng, X., Zhao, H.W., Fu, L.J., 448

Zhang, F.M., Gu, H.X., 2009. Prevalence of occult hepatitis B virus infection 449

among hepatopathy patients and healthy people in China. J. Infect. 58, 383-388. 450

9. Hoofnagle, J.H., 2009. Reactivation of hepatitis B. Hepatology 49, S156-S165 451

10. Hou, J., Wang, Z., Cheng, J., Lin, Y., Lau, G.K., Sun, J., Zhou, F., Waters, J., 452

Karayiannis, P., Luo, K., 2001. Prevalence of naturally occurring surface gene 453

variants of hepatitis B virus in nonimmunized surface antigen-negative Chinese 454

carriers. Hepatology 34, 1027-1034. 455

11. Ito, K., Qin, Y., Guarnieri, M., Garcia, T., Kwei, K., Mizokami, M., Zhang, J., Li, J., 456

Wands, J.R., Tong, S., 2010. Impairment of hepatitis B virus virion secretion by 457

single-amino-acid substitutions in the small envelope protein and rescue by a novel 458

glycosylation site. J. Virol. 84, 12850-12861. 459

12. Katsoulidou, A., Paraskevis, D., Magiorkinis, E., Moschidis, Z., Haida, C., 460

Hatzitheodorou, E., Varaklioti, A., Karafoulidou, A., Hatzitaki, M., Kavallierou, L., 461

Mouzaki, A., Andrioti, E., Veneti, C., Kaperoni, A., Zervou, E., Politis, C., 462

Hatzakis, A., 2009. Molecular characterization of occult hepatitis B cases in Greek 463

blood donors. J. Med. Virol. 81, 815-825. 464

13. Levast, M., Larrat, S., Thelu, M.A., Nicod, S., Plages, A., Cheveau, A., Zarski, J.P., 465

Seigneurin, J.M., Morand, P., Leroy, V., 2010. Prevalence and impact of occult 466

hepatitis B infection in chronic hepatitis C patients treated with pegylated interferon 467

and ribavirin. J. Med. Virol. 82, 747-754. 468

21

14. Lou, S.C., Pearce, S.K., Lukaszewska, T.X., Taylor, R.E., Williams, G.T., Leary, 469

T.P., 2011. An improved Abbott ARCHITECT((R)) assay for the detection of 470

hepatitis B virus surface antigen (HBsAg). J. Clin. Virol. 51, 59-63. 471

15. Neumann, A.U., Phillips, S., Levine, I., Ijaz, S., Dahari, H., Eren, R., Dagan, S., 472

Naoumov, N.V., 2010. Novel mechanism of antibodies to hepatitis B virus in 473

blocking viral particle release from cells. Hepatology 52, 875-885. 474

16. Raimondo, G., Pollicino, T., Romano, L., Zanetti, A.R., 2010. A 2010 update on 475

occult hepatitis B infection. Pathol. Biol. (Paris) 58, 254-257. 476

17. Raimondo, G., Allain, J.P., Brunetto, M.R., Buendia, M.A., Chen, D.S., Colombo, 477

M., Craxì, A., Donato, F., Ferrari, C., Gaeta, G.B., Gerlich, W.H., Levrero, M., 478

Locarnini, S., Michalak, T., Mondelli, M.U., Pawlotsky, J.M., Pollicino, T., Prati, 479

D., Puoti, M., Samuel, D., Shouval, D., Smedile, A., Squadrito, G., Trépo, C., Villa, 480

E., Will, H., Zanetti, A.R., Zoulim, F., 2008. Statements from the Taormina expert 481

meeting on occult hepatitis B virus infection. J. Hepatol. 49, 652-657. 482

18. Salisse, J., Sureau, C., 2009. A function essential to viral entry underlies the 483

hepatitis B virus "a" determinant. J. Virol. 83, 9321-9328. 484

19. Roy, A., Kucukural, A., Zhang, Y. I-TASSER, 2010. A unified platform for 485

automated protein structure and function prediction. Nat. Protoc. 5, 725-738. 486

20. Svicher, V., Gori, C., Trignetti, M., Visca, M., Micheli, V., Bernassola, M., Salpini, 487

R., Gubertini, G., Longo, R., Niero, F., Ceccherini-Silberstein, F., De Sanctis, 488

G.M., Spanò, A., Cappiello, G., Perno, C.F., 2009. The profile of mutational 489

22

clusters associated with lamivudine resistance can be constrained by HBV 490

genotypes. J. Hepatol. 50, 461-470. 491

21. Tamura, K,, Peterson, D,, Peterson, N., Stecher, G., Nei, M., Kumar, S., 2011. 492

MEGA5 2011: Molecular Evolutionary Genetics Analysis using Maximum 493

Likelihood, Evolutionary Distance, and Maximum Parsimony Methods. Molecular 494

Biology and Evolution. 495

22. Tian, X., Zhao, C., Zhu, H., She, W., Zhang, J., Liu, J., Li, L., Zheng, S., Wen, 496

Y.M., Xie, Y., 2010. Hepatitis B virus (HBV) surface antigen interacts with and 497

promotes cyclophilin a secretion: possible link to pathogenesis of HBV infection. J. 498

Virol. 84, 3373-3381. 499

23. Torresi, J., 2002. The virological and clinical significance of mutations in the 500

overlapping envelope and polymerase genes of hepatitis B virus. J. Clin. Virol. 25, 501

97-106. 502

24. van Hemert, F.J., Zaaijer, H.L., Berkhout, B., Lukashov, V.V., 2008. Occult 503

hepatitis B infection: an evolutionary scenario. Virol. J. 5, 146. 1743-1746. 504

25. Velati, C., Romano, L., Fomiatti, L., Baruffi, L., Zanetti, A.R., 2008. Impact of 505

nucleic acid testing for hepatitis B virus, hepatitis C virus, and human 506

immunodeficiency virus on the safety of blood supply in Italy: a 6-year survey. 507

Transfusion 48, 2205-2213. 508

26. Wu, C., Zhang, X., Tian, Y., Song, J., Yang, D., Roggendorf, M., Lu, M., Chen, X., 509

2010. Biological significance of amino acid substitutions in hepatitis B surface 510

23

antigen (HBsAg) for glycosylation, secretion, antigenicity and immunogenicity of 511

HBsAg and hepatitis B virus replication. J. Gen. Virol. 91, 483-492. 512

27. Weinberger, K.M., Zoulek, G., Bauer, T., Bohm, S., Jilg, W., 1999. A novel 513

deletion mutant of hepatitis B virus surface antigen. J. Med. Virol. 58, 105-110. 514

28. Yang, Z., 2007. PAML 4: phylogenetic analysis by maximum likelihood. Mol. Biol. 515

Evol. 24, 1586-1591. 516

29. Yuan, Q., Ou, S.H., Chen, C.R., Ge, S.X., Pei, B., Chen, Q.R., Yan, Q., Lin, Y.C., 517

Ni, H.Y., Huang, C.H., Yeo, A.E., Shih, J.W., Zhang, J., Xia, N.S., 2010. 518

Molecular characteristics of occult hepatitis B virus from blood donors in southeast 519

China. J. Clin. Microbiol. 48, 357-62. 520

30. Zaaijer, H.L., Torres, P., Ontañón, A., Ponte, L.G., Koppelman, M.H., Lelie, P.N., 521

van Hemert, F.J., Boot, H.J., 2008. Multiple surface antigen mutations in five blood 522

donors with occult hepatitis B virus infection. J. Med. Virol. 80, 1344-1349. 523

31. Zhang, J., Hou, T., Wang, W., Liu, J.S., 2010. Detecting and understanding 524

combinatorial mutation patterns responsible for HIV drug resistance. Proc. Natl. 525

Acad. Sci. U. S. A. 107, 1321-1326 526

527

528

529

530

531

532

24

LEGENDS TO FIGURES 533

Figure 1. Nucleotidic and amino acidic variability in HBsAg-gene and protein. The 534

three box-plots represent Shannon Entropy (A), Evolutionary Divergence (B) and the 535

ratio of non-synonymous (dN) and synonymous (dS) substitutions rates (C) among 536

patients with chronic infection and OBI. P-Values were calculated by Mann-Whitney 537

Test. 538

Figure 2. Amino acidic HBsAg-protein mutations associated with OBI. (A) prevalence 539

of HBsAg-mutations associated with OBI. Mutations reported in bold can introduce a 540

new N-linked glycosylation site in HBsAg-protein. P-values were determined by Fisher 541

exact test applying a Benjamini-Hochberg correction for multiple comparison at a false 542

positive rate of 0.05. * p<0.05; ** p<0.01; *** p<0.001. (B) Localization of amino acid 543

residues associated with HBV occult infection in 3D models of "D-consensus" and 544

mutant HBsAg-protein. Functional HBsAg domains are color-coded in the "D-545

consensus" protein: amino acids not assigned (1-7, gray), transmembrane signals (8-22, 546

80-98, red) and membrane-embedded C-terminus (169-226, red), cytosolic loop (23-79, 547

dark-red), luminal loop (99-169, dark-blue), the a-determinant region (110-140, blue) 548

and capsid interaction (96, 100, 103, 104, 113, 116, 117, 118, 121, 122, 169, 173, 174, 549

175, 177, 181, 182, 191, 192, 195, green). OBI-related residues are marked by 550

spacefilling in the "D-consensus" 3D structure. (C) The 3D-modeled structure of mutant 551

HBsAg with similar color coded and spacefilled residues shows to what extent the 552

HBsAg architecture becomes affected by amino acid replacements associated with 553

occult HBV infection. (D) Superposition of normalized hydrophobicity profiles of D-554

consensus (blue) and mutant (red) HBsAg-proteins (window 5 and step-size 1 residues) 555

25

displays these alterations particularly in the A-determinant region (110-140) and also at 556

capsid interacting residues (97-105, 160-177). 557

Figure 3. Impact of HBsAg-mutations associated with OBI on HBsAg reactivity. The 558

graph reports the mean of 4 independent experiments of the HBsAg amount detected in 559

presence of the with wild-type and mutated HBV clones. HBsAg has been quantified by 560

the Abbott Architect HBsAg assay. Specimens <1.0 S/CO are considered not reactive. 561

562

563

564

565

566

567

568

569

570

571

572

573

574

575

576

577

578

579

26

ACKNOWLEDGMENTS 580

We thank Laura Scipioni, and Andrea Biddittu for data-management.This work was 581

financially supported by grants from the Italian National Institute of Health, the 582

Ministry of University and Scientific Research, Current and Finalized Research of the 583

Italian Ministry of Health. The funders had no role in study design, data collection and 584

analysis, decision to publish, or preparation of the manuscript 585

586

587

TRANSPARENCY DECLARATION 588

C.-F. Perno have received funds for attending symposia, speaking, organizing 589

educational activities, grant research support, consultancy and advisory board 590

membership, from Abbott, Boehringer Ingelheim, Bristol Myers Squibb, Gilead, Merck 591

Sharp & Dohme, Janssen Cilag, Pfizer, Tibotec, Roche, and ViiV. F. Ceccherini-592

Silberstein has received funds for attending symposia, speaking and organizing 593

educational activities from Abbott, Merck Sharp & Dohme, Janssen Cilag, and Virco. V 594

Svicher has received funds for attending symposia, speaking and organizing educational 595

activities from VIIV. The other authors declare that no competing interests exist. 596

597

598

599

600

601

602

603

604

27

Table 1. Demographic and serological characteristics of the study population. 605

Characteristic Patients with Occult HBV infection

Patients, N 26

Age (years), Median (IQR) 56.0 (51.1-62.6)

Male, N (%) 22 (84.6)

Italian Nationality, N (%) 25 (96.2)

HBV Genotype

D 24 (92.3)

A 1 (3.8)

Undetermined 1 (3.8)

Type of Blood Donation

First time donors 10 (38.5)

Habitual donors 16 (61.5)

N (%) of patients with serum HBV-DNA

< 12 UI/ml 22 (84.6)

> 12 UI/ml 4 (15.4)b

ALT (IU/ml), Median (IQR) 21.5 (19.0-29.3)

Serological characteristics of patients

HBcAb IgG+, N (%) 26 (100)

HBcAb IgM+, N (%) 0

HBeAg+, N (%) 0

HBeAb+, N (%) 19 (73.1)

HBsAg (S/CO), Median, (IQR) 0.46 (0.38-0.59)

HBsAb+, N (%) 11 (42.3)

HBsAb (mIU/ml), Median, (IQR)a 28.0 (20.0-54.0)

a Serum HBV-DNA was determined by COBAS TaqMan High Pure System and by COBAS AmpliPrep/TaqMan 606

HBV. 607

b Viremia ranges from 30IU/ml to 50IU/ml. 608

IQR, interquartile range. 609

610

611

612

28

613

10

8

6

4

2

0

Patients with chronic HBV

infection (N=82)

Patients with occult HBV

infection (N=24)

dN

/dS

2.0

1.5

1.0

0.5

0.0

Patients with chronic HBV

infection (N=82)

Patients with occult HBV

infection (N=24)

Sh

ann

on

En

trop

y

A

P = 6.855e-05 0.2

0.15

0.1

0.05

0.0

Patients with chronic HBV

infection (N=82)

Patients with occult HBV

infection (N=24)

Aver

age

Evolu

tion

ary D

iver

gen

ce (

Nsu

bs/

Sit

e)

B

P = 0.0003

C

P = 4.863e-29

Fig.1

Figure(s)

0

5

10

15

20

25

Prevale

nce (

%)

Occult HBV Infection, N=24 Chronic HBV Infection, N=82

*

Y100S Q101R P105R T115N T116N P120L R122P T123N T126I P127H P127L Q129P M133T Y134C S143L S167L R169H S174N L175S V177A

*

*

**

**

**

**

**

**

**

** **

**

** ** **

***

**

**

**

Luminal loop "a" determinant Luminal loop Membrane-embedded

C-terminus

A

S143

TMH1

TMH2

TMH4

TMH3

S174&L175&V177

S167&R169

Y100&Q101

P105

T115&T116

P120&R122&T123

T126&P127&129M133&Y134

CB

Rel

ati

ve

hy

dro

ph

ob

icit

y

0,3

0,4

0,5

0,6

0,7

0,8

0,9

1

1 7 13 19 25 31 37 43 49 55 61 67 73 79 85 91 97 103 109 115 121 127 133 139 145 151 157 163 169 175 181 187 193 199 205 211 217

Surf_Dcons

Surf_Mutant

Amino acid sequence position

D

Fig.2Figure(s)

S/CO WT 100S 116N 143L100S,

143L

116N,

143L122P 101R 167L

122P,

167L

101R,

122P

Mean 306.86 0.55 54.91 229.97 0.47 31.20 6.12 169.04 303.42 3.89 3.02

SE 64.10 0.05 22.57 90.30 0.02 12.05 2.44 91.27 74.71 1.33 1.00

0

50

100

150

200

250

300

350

400

1 S/CO

Am

ou

nt

of

HB

sAg

det

ecte

d (

S/C

O)

Fig.3

Figure(s)

e-componentsClick here to download e-components: SI_text for AVR.doc

e-componentsClick here to download e-components: Table S1.docx

e-componentsClick here to download e-components: Figura S1.ppt