key: cord-0882923-xw3n0fhi authors: Zhao, Ping; Ma, Dao-Xin; Yu, Shuang; Xue, Fu-Zhong; Zhu, Wei-Wei; Shao, Na; Zhang, Jing-Ru; Ji, Chun-Yan title: The development of Chinese specific human cytomegalovirus polyepitope recombinant vaccine date: 2011-12-13 journal: Antiviral Res DOI: 10.1016/j.antiviral.2011.12.005 sha: 3769fc772f7e3fb0d0822bb2887ae12c0dbaf0d2 doc_id: 882923 cord_uid: xw3n0fhi Human cytomegalovirus (HCMV) infection is a major cause of morbidity in the recipients of organ transplants and in the congenitally infected infants. HCMV vaccine has emerged as an effective approach to prevent HCMV infection particularly for the development of multiple viral antigens vaccination and human leukocyte antigen (HLA)-restricted polyepitope technology. As the Chinese population makes up more than one fifth of the population worldwide, it is important to develop HCMV vaccines more specific for the Chinese population by targeting Chinese-restricted HLA alleles and antigens. In the present study, we designed a novel chimeric polyepitope vaccine based on the replication-deficient adenovirus Ad5F35, which encodes 83 HCMV T cell epitopes from 15 different HCMV antigens, restricted to 14 HLA I and 7 HLA II alleles that cover 92% of the Chinese population. Our results show that the recombinant adenovirus vaccine Ad5F35-CTL·Th can be efficiently transfected and expressed in peripheral blood mononuclear cells (PBMCs) with little cytopathic activity. Ad5F35-CTL·Th can also be endogenously processed and presented by PBMCs. Ad5F35-CTL·Th-stimulated HCMV-specific cytotoxic T lymphocytes (CTLs) showed strong cytolytic activity against HCMV polyepitope-sensitized target cells. The CTL activity was accompanied by high levels of IFN-γ production after Ad5F35-CTL·Th stimulation. The specificity and vigorous response to the recombinant adenovirus vaccine in vitro makes it a potential candidate to be used for transplantation recipients or congenitally infected infants. Human cytomegalovirus (HCMV) is found universally throughout all geographic locations and socioeconomic groups, with seroprevalence ranging from 45% to 100% . HCMV can cause clinical disease in the fetus, neonate and immunocompromised individuals such as transplant recipients and HIVinfected patients. Congenital HCMV infection is an important cause of hearing, cognitive, and motor impairments in newborns (Dollard et al., 2007; Hyde et al., 2010) . In the allograft recipient, viremic dissemination can cause end-organ disease, such as hepatitis, pneumonitis, gastroenteritis and retinitis, and can predispose for transplant rejection. Developing a vaccine in the two populations has been rated as high priority. Recently, two HCMV phase 2 clinical trials have now been completed with positive results from the induction of humoral immunity, which showed that HCMV glycoprotein B (gB) vaccine has the potential to decrease incident cases of maternal and congenital CMV infection (Griffiths et al., 2011; Pass et al., 2009) . We have also generated vaccine Ad5F35-AD-1 that expressed the conserved conformational epitope AD-1 from gB protein (Zhao et al., 2009) . Moreover, cellular immunity provided by the CD8+ and CD4+ T lymphocytes has shown to play a key role in resolving the primary infection, providing long-term immune surveillance, and the clearance of replicating virus (Crough et al., 2007; Fornara et al., 2011; Nebbia et al., 2008) . Therefore, it is of great importance to develop an effective HCMV vaccine based on viral antigens that activate cellular immunity to prevent HCMV-associated disease (Bunde et al., 2005; Einsele et al., 2002) . Over the past 30 years, various strategies of cellular immunity, including the use of subunit vaccines, its DNA or dense bodies to vaccinate, have been developed and promising results in preclinical studies have been demonstrated (Khanna and Diamond, 2006; Plotkin, 1999; Schleiss, 2005 Schleiss, , 2007 of the strategies is hindered by the limited range of epitopes and HLA alleles that could be included to develop the HCMV vaccine. Ideally, a formulation should cover most viral antigens, which could induce a broad repertoire of HCMV-specific cellular immune responses. Recently developed polyepitope technology allows the presentation of multiple HLA class I-restricted CTL (cytotoxic T lymphocyte) epitopes derived from different antigens within a single reading frame and thus provides a new platform to achieve this goal. The epitope-based vaccine should also contain a minimal number of epitopes which cover a vast majority of the human population (Buteau et al., 2002; Fischer et al., 2007; Khan et al., 2006) . T-cell epitopes that bind to multiple alleles of HLA are prime targets for vaccine and immunotherapy development due to their relevance to a large proportion of the human population, for example, SARS coronavirus nucleocapsid (Gupta et al., 2006) , HIV-1 proteins (Berzofsky et al., 1991; Brown et al., 2003; Surman et al., 2001) , and Chlamydia trachomatis outer membrane protein (Kim and De-Mars, 2001) . Epitope-based vaccines can effectively induce cellular immunity by selecting a small number of hotspots that can elicit most of the required T-cell functions. To date, many studies have focused on HLA-restricted HCMV epitope vaccines and progress has been made (Elkington et al., 2003; Rist et al., 2005) . However, these investigations were mainly aimed at the Caucasian population or for a general population with limited coverage of HLA alleles or antigens. Since Chinese population with more than 50 different ethnic groups makes up one fifth of the world population, it is essential to develop HCMV epitope vaccines more specific to the Chinese population by targeting on more Chinese-restricted HLA alleles and broader antigens. To develop HCMV vaccines specific for Chinese population, we designed a novel chimeric polyepitope vaccine based on the replication-deficient adenovirus Ad5F35 and evaluated their immunological effects in vitro. The prediction and selection of Chinese HLA-restricted HCMV polyepitope were performed according to our forecasting system previously published on spatial coverage of cumulative phenotypic frequency (CPF) of HLA-I (Xue et al., 2005a,b) . The CPF algorithm allows the identification of alleles that represent a desired percentage of a population, and also allows prediction of theoretical responder status in any given population where HLA gene frequencies are known. Briefly, the CPF of 14 HLA allele sites (A2, A24, A1, A3, A11, A68, B44, B7, A23, A26, B35, B38, B8 and B27) in the Chinese population was forecasted and the immunoactivity of HLA-restricted HCMV polyepitope nucleotide vaccine in different geographical locations was evaluated. Utilizing literature data on HLA-restricted HCMV epitopes that have been published, we selected 76 CTL epitopes and 7 Th epitopes derived from different HCMV protein antigens that can induce cellular immunologic response and stably bind corresponding class I and class II HLA molecules. Through SYFPEITHY (Rammensee et al., 1999) combined with MAPPP algorithms (Hakenberg et al., 2003) , the selected Fig. 1 . Schematic view of the construction of a recombinant adenovirus that expresses a synthetic DNA encoding for a polyepitope protein which contains 83 HCMV T-cell epitopes. The DNA sequence encoding this polyepitope protein was synthesized using overlapping epitope sequence specific primers as described in ''Section 2''. This synthetic insert was cloned into a T plasmid prior to cloning into the pHMCMV5 vector. After amplification in E. coli, the expression cassette from pHMCMV5 was excised and ligated into the pAd5F35 expression vector. Following linearization of the DNA using Pac I restriction enzyme, the recombinant pAd5F35 vector was packaged into infectious adenovirus by transfecting HEK 293 cells, and recombinant adenovirus (referred to as Ad5F35-CTLÁTh) was harvested from transfected cells by repeated freeze-thawing cycles. epitopes and polyepitope were predicted for the specific HLA-binding value and proteasomal cleavage site. The design and construction of a recombinant adenovirus vaccine expressing CTLÁTh polyepitope of HCMV is shown in Fig. 1 . The amino acid sequence of the 83 HLA restricted CTLÁTh epitopes was translated into the nucleotide sequence using human universal codon usage, and the polyepitope DNA sequence representing the 83 epitopes has been synthesized using Splicing by Overlap Extension and stepwise asymmetric Polymerase Chain Reaction (PCR) (Thomson et al., 1995) . The final DNA sequence was coded with a Kozak sequence, a start codon, 83 contiguous minimal HCMV CTLÁTh epitopes (Table 1) , and a hexahistidine tag. After being cloned into a T vector and sequenced, the CTLÁTh sequence was inserted into shuttle plasmid pHMCMV5 (kindly provided by Hiroyuki Mizuguchi, Osaka University, Osaka, Japan). The expression cassette was then cloned into Ad5F35 vector (provided by Hiroyuki Mizuguchi) and verified by restriction digestion and sequencing. The recombinant Ad5F35 was linearized and transfected into human embryonic kidney (HEK) 293 cells for 7 days until maximal cytopathic effect (CPE) was observed, pAd5F35-GFP was included as a vector control. The recombinant adenoviruses (Ad5F35-CTLÁTh) were harvested from the transfected cells by successive freeze-thawing cycles. The titer of the large-scale Ad5F35-CTLÁTh virus preparation was measured by plaque assay. To confirm the successful development of the recombinant adenovirus, PCR was used to amplify the DNA extracted from Ad5F35-CTLÁTh virions. Reverse Transcription (RT)-PCR and Western blotting were used to confirm the mRNA and protein expression of CTLÁTh in HEK293-CTLÁTh cells, while HEK293-GFP and parental HEK293 cells served as negative controls. PBMCs from healthy donors were cultured in 6-well plates, and then infected with Ad5F35-CTLÁTh or Ad5F35-GFP for 6 h. On day 1, 2, 6, and 10 post-infection, CTLÁTh expression was analyzed by immunocytochemistry as follows. Briefly, PBMCs placed on slides were fixed with 4% paraformaldehyde. Non-specific antibody sites were blocked with 10% goat serum, then the cells were incubated with anti-6ÂHis tag antibody (Abcam, Cambridge, England) or relevant isotype control antibody at 4°C overnight. Slides were washed with PBS and then incubated with HRP-conjugated secondary antibody. All slides were processed by the Streptavidin-Peroxidase (SP) method (Zhongshan Co. Ltd., Beijing, China) for 30 min. Following staining with diaminobenzidone, slides were visualized using an OLYMPUS-BX51 microscope to observe the expression and localization of recombinant polyepitope or its proteasomal cleaved epitopes. The viability of infected PBMCs was determined by trypan blue staining. Epstein Barr virus-transformed lymphoblastoid cell lines (LCLs) were established from HCMV-seropositive donors by exogenous virus transformation of peripheral B cells using the B95.8 virus isolates. Autologous LCLs, pulsed with HCMV peptides and Ad5F35-CTLÁTh, were used to stimulate T cell lines and clones on a weekly basis or as target cells in cytotoxicity assays (see below). All cell lines except HEK293 were routinely maintained in RPMI1640 medium supplemented with 100 units/ml penicillin G sodium, 100 lg/ ml streptomycin, 4 mmol/L L-glutamine and 10% FCS. The HEK293 cell line was maintained in DMEM containing 10% FCS. The HCMV experimental strain AD169 was cultured on fibroblasts. Peptides, synthesized by the Merrifield solid phase method, were purchased from Shanghai Sangon Biological Engineering Company. They were dissolved in 10% dimethyl sulphoxide, and diluted in serum-free RPMI1640 medium. Purity of these peptides were examined by HPLC and proven to be >94%. Peripheral blood was collected from 10 donors. DNA was extracted to test HLA alleles by PCR-SSP (sequence-specific primers), and serum was used to detect HCMV status by ELISA. Informed consent was obtained from each donor, and ethical approval for the study was obtained from the Ethical Committee of Qilu Hospital, Shandong University. Polyclonal CTL lines specific for HCMV epitopes were established according to methods previously published (Elkington et al., 2003; Khanna, 1998) . Briefly, 2 Â 10 6 PBMCs from healthy virus carriers were cocultured with autologous x-irradiated PBMCs (1 Â 10 6 ) pulsed with 20 lmol/L peptide for 16 h. On day 3, growth medium with rIL-2 was added, and the cells were further expanded. On day 7, the cultures were restimulated with peptidesensitized, x-irradiated autologous LCLs. After 10 days in culture medium, the cells were used as polyclonal effectors in an in vitro cytotoxicity assay against peptide-sensitized autologous LCLs. HCMV specific CTLs were generated from the healthy virus carriers by stimulating PBMCs with Ad5F35-CTLÁTh. For adenovirus polyepitope stimulation, 2 Â 10 6 PBMCs from healthy virus carriers were cocultured with autologous x-irradiated PBMCs (1 Â 10 6 ) which were infected with Ad5F35-CTLÁTh (MOI of 100:1, 50:1, or 25:1) at a responder to stimulator ratio of 2:1. On day 3, the cultures were supplemented with growth medium containing rIL-2. On day 7, the cultures were restimulated with adenovirus-infected (MOI 50:1), x-irradiated autologous LCLs and supplemented with growth medium containing rIL-2. HCMV-specific T-cell reactivity (stimulated once or twice) was sequentially assessed on day 14 using an in vitro cytotoxicity assays and IFN-c production. The LDH method was used to detect the cytotoxicity of HCMVspecific CTLs. Briefly, the effector cells (E) and the target cells (T) were seeded in a 96-well plate at different E:T ratios. After 4 h of incubation, 100 ll cell-free supernatant was transferred to a new 96-well plate and 100 ll reagent was added into each well. Thirty minutes later, the LDH release was detected by spectrophotometer at 490 nm. Maximal or spontaneous LDH release was obtained by adding 1% Triton X-100 or complete medium to the target cells, respectively. The percentage of specific lysis was calculated by the following formula: % specific lysis = 100 Â (sample release À spontaneous release)/(maximum release À spontaneous release). PBMCs that were stimulated with Ad5F35-CTLÁTh (MOI of 100:1, 50:1), or CTLs that were restimulated with Ad5F35-CTLÁTh (MOI 50:1) were detected with ELISPOT to measure the production of IFN-c according to the protocol (R&D, Minneapolis, MN). Cytokine-producing cells were detected as purple spots, and the developed microplate was analyzed by counting spots using a specialized ELISPOT reader. Quantitation of results was done by calculating the number of spot forming cells (SFC) per number of cells added into the well. T cell precursor frequencies for each epitope were based on the total number of cells and the number of SFC per well (average of triplicate wells). Ad5F35-CTLÁTh-specific spots were calculated after subtraction of the number of spots in negative control wells. Predominant Chinese-restricted alleles (A2, A24, A1, A3, and A11) were put into the algorithm of CPF, and the CPF of the set of alleles was 88.1% in the Chinese population ( Fig. 2A) . Then, other alleles (A68, B44, B7, A23, A26, B35, B38, B8, and B27) were added to the algorithm, and the total CPF of these 14 HLA alleles reached 92.1% in Chinese population (Fig. 2B) . Based on the selected set of HLA alleles, we selected 76 CTL epitopes and 7 Th epitopes in different HCMV antigens that could induce wide-coverage immune response and stably bind class I and class II HLA molecules. Then the selected epitopes were put into the SYFPEITHI algorithm and the predicted scores of the selected epitopes has reached above 14, most between 20 and 30, indicating high HLA-binding affinity. MAPPP could predict both the proteasomal cleavage and peptide anchoring to HLA I molecules. The selected polyepitope sequence was put into the MAPPP algorithm. Most of the predicted proteasomal cleavage values reached a score above 0.5, suggesting the high possibility for single epitope production and presentation ( Table 1) . The epitopes included in this polyepitope were derived from 15 different viral proteins (pp28, pp50, pp65, pp150, pp71, gH, gB, IE-1, IE-2, US2, US3, US6, US11, UL16 and UL18) that were involved in virus attachment, replication, assembly, reactivation and immune escape from the latent phase, all of which are crucial stages in the progression of HCMV infection. The full-length CTLÁTh nucleotide sequence has been synthesized, sequenced, and cloned into pHMCMV5. Double digestion of pHMCMV5-CTLÁTh with NheI/NotI or I-CeuI/PI-SceI produced expected fragments confirming the construction of pHMCMV5-CTLÁTh. The 3602 bp CTLÁTh expression cassette from pHMCMV5-CTLÁTh was then cloned into the pAd5F35 vector to produce pAd5F35-CTLÁTh. Double-digestion of pAd5F35-CTLÁTh with I-CeuI/PI-SceI produced the two expected fragments. Digestion of pAd5F35-CTLÁTh with XhoI also generated seven expected fragments ( Fig. 3A and B) . The correct insertion of the CTLÁTh gene was further confirmed by sequencing. The recombinant pAd5F35-CTLÁTh vector, and the control pAd5F35-GFP, were both packaged into HEK293 cells, and emerging recombinant adenoviruses Ad5F35-CTLÁTh and Ad5F35-GFP were cultivated. In both cases a normal CPE was observed compared with uninfected HEK293 cells. Green fluorescence was observed in HEK293-GFP cells, indicating the efficient transfection and gene expression (Fig. 4A-F) . Recombinant adenovirus was harvested from the transfected cells, with the Ad5F35-CTLÁTh titer measured as 2.5 Â 10 9 PFU. Successful PCR amplification of recombinant virions DNA confirmed the inclusion of CTLÁTh sequence into recombinant adenovirus virions. CTLÁTh expression in HEK293-CTLÁTh was further analyzed by RT-PCR and the amplified fragment was expected (Fig. 3C) . CTLÁTh protein expression was analyzed by Western blotting using antibody directed against the 6ÂHis tag. The band at 90 kDa, corresponding to the predicted size of the recombinant CTLÁTh protein, was observed in extracts of HEK293-CTLÁTh cells, but not in parental HEK293 cells or HEK293-GFP cells (Fig. 3D) . Strong expression of CTLÁTh antigen was found in PBMCs cells infected with Ad5F35-CTLÁTh, while no expression was detected in PBMCs infected with control Ad5F35-GFP. The typical pattern of staining indicated that the recombinant CTLÁTh polyepitope or its proteasomal cleaved epitopes might both be localized in the cell membrane and cytoplasm (Fig. 4G-I) . Expression of CTLÁTh was time-dependent, and strongest expression was observed on day 6. No significant cytopathic activity was detected in infected cells, as cell viability was not significantly affected by Ad5F35-CTLÁTh infection, with 90% of cells remaining viable. 1) with either Ad5F35-CTLÁTh or control adenovirus (referred to as Ad5F35-control) for 16-18 h and then exposed to HCMV-specific CTL lines. LCLs infected with adenovirus vectors were used as targets for NLV, VLE, VTE, and GPI -specific CTL lines. An E:T ratio of 2.5:1 was used for both assays. HCMV-specific CTL lines were generated from healthy virus carriers and their specificity was confirmed by their ability to lyse HLA-matched target cells coated with the respective epitopes (Fig. 5A) . HLA-matched LCLs infected with Ad5F35-CTLÁTh (MOI 25:1) were also efficiently recognized by individual HCMV-specific CTL lines, whereas target cells infected with control Ad5F35 were not recognized (Fig. 5B) . were co-cultured with autologous PBMCs infected with Ad5F35-CTLÁTh (MOI 50:1 or 25:1) at a responder to stimulator ratio of 2:1. On day 7, the cultures were restimulated with Ad5F35-CTLÁTh -infected (MOI 50:1), x-irradiated autologous LCLs, and the growth medium was supplemented with rIL-2 (20 U/ml). Epitope-specific T cell reactivity was subsequently assessed on day 10 using in vitro cytotoxicity assays. Autologous LCLs coated with individual peptide epitopes (20 lg/ml) or uncoated were used as targets for these assays with an E:T ratio of 10: 3.6. Activated HCMV-specific CTLs stimulated with Ad5F35-CTLÁTh show strong cytolytic activity PBMCs from healthy virus carriers were stimulated with autologous PBMCs infected with Ad5F35-CTLÁTh. It showed that a single stimulation was highly efficient to rapidly expand T cells, and these cells exhibited a strong HCMV epitope-specific cytolytic activity. CTLs expanded from two different donors showed reactivity to more than one epitope derived from different antigens. For example, T cell cultures from donor JXX showed strong cytolytic activity against 3 epitopes coming from two antigens (IE-1, pp65); while donor ZNN demonstrated cytolytic activity against 1 epitope coming from pp50 antigen (Fig. 6A) . Moreover, the two different MOI (50:1 and 25:1) showed comparable levels of T cell stimulation. The efficacy of Ad5F35-CTLÁTh was also confirmed by testing the expanded T cell populations at different effector to target (E:T) ratios. Ad5F35-CTLÁTh was highly efficient in stimulating multiple CTL responses, with strong cytolytic activity against four different epitopes (VTE, HLA A1-restricted; NLV and VLE, HLA A2restricted; and GPI, HLA A11-restricted) observed at an E:T ratio as low as 1:1 ( Fig. 6B and C) . Moreover, the cytolytic activity was also elevated with the increase of E:T ratio. PBMCs stimulated with Ad5F35-CTLÁTh produced higher IFN-c amounts than controls. Ad5F35-CTLÁTh efficiently sensibilized PBMCs to antigen-specific CTLs. In addition, the two different MOI (100:1 and 50:1) showed comparable levels of IFN-c production, with higher IFN-c production observed at MOI 100:1 than MOI 50:1 especially for the donor JXX (Fig. 6D) . Furthermore, IFN-c production cells in PBMCs stimulated with Ad5F35-CTLÁTh (stimulated once) was lower than that in CTLs restimulated with Ad5F35-CTLÁTh (stimulated twice), indicating stronger HCMV epitope-specific cytolytic activity upon twofold stimulation (Fig. 6E ). HCMV disease remains a major obstacle for allogeneic BMT and prevention of HCMV infection by a vaccine has became a highpriority goal (Schleiss, 2008) . Adoptive transfer of HCMV-specific CTL cells into transplant recipients has demonstrated therapeutic benefits in relation to HCMV diseases (Hebart and Einsele, 2004; Peggs et al., 2003) . A vaccine that can induce a broad repertoire of HCMV-specific immune responses in different ethnic populations with various HLA alleles is likely to provide more effective protection against HCMV-associated pathogenesis. Since the Chinese population constitutes approximately one fifth of the world population and has extremely polymorphic HLA alleles, it is essential to develop HCMV epitope vaccines more specific to the Chinese to prevent worldwide HCMV infection. In this context and based on our forecasting system, we identified the combination of 14 HLA allele sites (A2, A24, A1, A3, A11, A68, B44, B7, A23, A26, B35, B38, B8 and B27), and the CPF of 14 HLA allele sites in the Chinese population reached 92.1%. Then, according to these selected HLAs and literature data on HLA-restricted HCMV epitopes, we designed the novel chimeric vaccine which encodes 76 HCMV CTL epitopes and 7 Th epitopes as a polyepitope. Optimized selection of epitopes can precisely direct the evoked immune response at conserved and highly immunogenic regions of several antigens. Therefore, in addition to the coverage of HLA alleles in a given population, a good epitope-based vaccine also takes into account coverage of epitopes, coverage of HLA/antigen combinations, and the probability of each epitope to be properly cleaved in the sequence. HCMV has 165 open reading frames (ORFs), and accumulating evidence has shown that HCMV specific immune responses are not only restricted to gB, pp65 and IE-1 antigens, but also are directed towards more than 70% of the HCMV reading frames (Elkington et al., 2004; Manley et al., 2004; Schleiss, 2008; Sylwester et al., 2005) which includes ''non-essential'' genes encoding modulators of adaptive immune responses, such as US2, US3, US6, US11. In this study, computational methods such as CPF algorithm, SYFPEITHI algorithm and MAPPP algorithm for predicted as well as experimentally determined epitopes were incorporated into the selection of the optimal polyepitope. These epitopes included in this polyepitope were derived from 15 different viral proteins involved in different stages of viral infection, all of which significantly reduced any potential threat of immune escape from the selective expression of antigens. As described above, vaccines incorporating various antigens expressed in all reproduction phase of virus would provide a strategy to immunize against HCMV infection clinically. To circumvent allele specificity for the required Th epitope, a series of Th sequences that promiscuously bind to HLA II alleles have been evaluated in combination with the polyepitope. In our study, the HCMV epitopes encoded by this polyepitope were endogenously expressed and processed by human cells. We found that stimulation with Ad5F35-CTLÁTh rapidly expanded multiple antigen-specific human CTL cells from healthy virus carriers in vitro, and these CTL cells exhibited a strong HCMV epitope-specific cytolytic activity against HLA restricted target cells. IFN-c-producing cells were largely induced from PBMCs stimulated by Ad5F35-CTLÁTh or expanded antigen-specific CTL, which indicated T cell reactivation. The data presented here emphasize the strength and adaptability of the polyepitope-based approach for clinical translation and may lead to significant advances in the application of adoptive immunotherapy to a wide range of diseases. It will be useful for HCMV vaccine development for the BMT patients. In the development of a new HCMV vaccine, it is important that the vaccine has high immunogenicity, low production cost, and low pathogenicity. Compared with traditional adenoviral vectors, Ad5F35 used in this study is being increasingly recognized for its high efficiency and low toxicity and has been used in multiple human gene therapy clinical trials. The Ad5F35 vector contains a chimeric fiber gene encoding the fiber tail of Ad5 in combination with the fiber shaft and knob domains of Ad35. Cell entry of Ad5F35 is thought to be mediated by CD46, a receptor expressed on most human cells (Gaggar et al., 2003) . Liver tropism and hepatoxicity are significantly lower for Ad5F35 than for Ad5 (Xin et al., 2005) . Importantly, Ad5F35 infection does not induce acute responses or the production of proinflammatory cytokines. Ad5F35 has been used as a vaccine delivery vehicle in other infectious disease models including hepatitis C (Thammanichanond et al., 2008) and HIV (Xin et al., 2007) , where potent transgene product-specific antibody and T-cell responses were reported. In terms of efficacy, Ad5F35 appears to be more effective than other comparable vectors for recombinant protein expression in hematopoietic cells (Yotnda et al., 2001) . In this study we successfully constructed a novel HCMV vaccine based on the replication-deficient Ad5F35 that encodes HLA-specific HCMV polyepitope nucleotide vaccine. Ex vivo infecting PBMCs with Ad5F35-CTLÁTh consistently demonstrated high expression of CTLÁTh polyepitope antigen and low toxicity, which in turn resulted in strong stimulation of antigen specific CTLs. Thus, combination of the Ad5F35 expression system with polyepitope technology provides a potential strategy for the expansion of T cells. In summary, we have developed a recombinant adenovirus vaccine against HCMV based on Ad5F35 expression system and polyepitope technology, which covers 92% of the Chinese population. Stimulation with this adenovirus vector expressing HCMV polyepitope is capable of inducing multiple, independent HLA-restricted CTL responses. Therefore, this vaccine may be a promising candidate for clinical trials and in prime-boost strategies in combination with other HCMV vaccine formulations, and we might also use this approach to develop suitable vaccines for the non-Chinese population. There is no conflict of interests for each author. Construction of peptides encompassing multideterminant clusters of human immunodeficiency virus envelope to induce in vitro T cell responses in mice and humans of multiple MHC types Clustering of Th cell epitopes on exposed regions of HIV envelope despite defects in antibody activity Protection from cytomegalovirus after transplantation is correlated with immediate early 1-specific CD8 T cells Challenges in the development of effective peptide vaccines for cancer Review of cytomegalovirus seroprevalence and demographic characteristics associated with infection Symptomatic and asymptomatic viral recrudescence in solid-organ transplant recipients and its relationship with the antigen-specific CD8(+) T-cell response New estimates of the prevalence of neurological and sensory sequelae and mortality associated with congenital cytomegalovirus infection Infusion of cytomegalovirus (CMV)-specific T cells for the treatment of CMV infection not responding to antiviral chemotherapy Ex vivo profiling of CD8+-T-cell responses to human cytomegalovirus reveals broad and multispecific reactivities in healthy virus carriers Cross-reactive recognition of human and primate cytomegalovirus sequences by human CD4 cytotoxic T lymphocytes specific for glycoprotein B and H Polyvalent vaccines for optimal coverage of potential T-cell epitopes in global HIV-1 variants Kinetics of effector functions and phenotype of virus-specific and cd T lymphocytes in primary human cytomegalovirus infection during pregnancy CD46 is a cellular receptor for group B adenoviruses Cytomegalovirus glycoprotein-B vaccine with MF59 adjuvant in transplant recipients SARS coronavirus nucleocapsid immunodominant T-cell epitope cluster is common to both exogenous recombinant and endogenous DNA-encoded immunogens MAPPP: MHC class I antigenic peptide processing prediction Clinical aspects of CMV infection after stem cell transplantation Cytomegalovirus seroconversion rates and risk factors: implications for congenital CMV Large-scale analysis of antigenic diversity of T-cell epitopes in dengue virus Tumour surveillance: missing peptides and MHC molecules Human cytomegalovirus vaccine: time to look for alternative options Epitope clusters in the major outer membrane protein of Chlamydia trachomatis Immune evasion proteins of human cytomegalovirus do not prevent a diverse CD8+ cytotoxic T-cell response in natural infection Polyfunctional cytomegalovirus-specific CD4+ and pp65 CD8+ T cells protect against highlevel replication after liver transplantation Vaccine prevention of maternal cytomegalovirus infection Adoptive cellular therapy for early cytomegalovirus infection after allogeneic stem-cell transplantation with virus-specific T-cell lines Cytomegalovirus vaccine SYFPEITHI: database for MHC ligands and peptide motifs Ex vivo expansion of human cytomegalovirus-specific cytotoxic T cells by recombinant polyepitope: implications for HCMV immunotherapy Progress in cytomegalovirus vaccine development Prospects for development and potential impact of a vaccine against congenital cytomegalovirus (CMV) infection Cytomegalovirus vaccine development Localization of CD4+ T cell epitope hotspots to exposed strands of HIV envelope glycoprotein suggests structural influences on antigen processing Broadly targeted human cytomegalovirus-specific CD4+ and CD8+ T cells dominate the memory compartments of exposed subjects Fibermodified recombinant adenoviral constructs encoding hepatitis C virus proteins induce potent HCV-specific T cell response Minimal epitopes expressed in a recombinant polyepitope protein are processed and presented to CD8+ cytotoxic T cells: implications for vaccine design Prime-boost vaccination with plasmid DNA and a chimeric adenovirus type 5 vector with type 35 fiber induces protective immunity against HIV Chimeric adenovirus 5/35 vector containing the clade C HIV gag gene induces a cross-reactive immune response against HIV Identification of spatial genetic boundaries using a multifractal model in human population genetics Forecasting system on spatial coverage of cumulative phenotypic frequency of HLA class I for designing HLA-based vaccines in China Efficient infection of primitive hematopoietic stem cells by modified adenovirus Towards a novel vaccine against human cytomegalovirus based on a chimeric Ad5F35 adenovirus vector expressing the immunodominant antigenic domain 1 epitope This study was partially supported by research funding from the National Natural Science Foundation (30600680, 81070407, 81170515), the Shandong Technological Development Project (2005BS03022, Q2008C07, BS2009SW014 and BS2010YY004), the SRF for ROCS, SEM and IIFSDU (2009TS063, 2009TS071).