7,498
Views
76
CrossRef citations to date
0
Altmetric
Reviews

Adenoviral vector-based strategies against infectious disease and cancer

&
Pages 2064-2074 | Received 30 Dec 2015, Accepted 10 Mar 2016, Published online: 20 May 2016

ABSTRACT

Adenoviral vectors are widely employed against infectious diseases or cancers, as they can elicit specific antibody responses and T cell responses when they are armed with foreign genes as vaccine carriers, and induce apoptosis of the cancer cells when they are genetically modified for cancer therapy. In this review, we summarize the biological characteristics of adenovirus (Ad) and the latest development of Ad vector-based strategies for the prevention and control of emerging infectious diseases or cancers. Strategies to circumvent the pre-existing neutralizing antibodies which dampen the immunogenicity of Ad-based vaccines are also discussed.

Introduction

Ad is non-enveloped, double stranded DNA virus with icosahedral capsids. It was first discovered by Rowe and his colleagues when they tried to culture the adenoid tissue in the laboratory in 1953. Ad infection is usually mild to human beings, but sometimes could be life threatening, especially to the immunocompromised individuals. In the 1970s, the United States army developed live Ad vaccines to prevent acute respiratory disease caused by AdHu4 and AdHu7.Citation1 In 1991, Rosenfeld et al developed the first in vivo gene transfer using an Ad vector, and demonstrated that human α 1-antitrypsin gene delivered by the E1-E3-deleted Ad could be detected in the lung of a cotton rat.Citation2 In 1993, the first human gene therapy study based on Ads was performed, a 23-year-old man with cystic fibrosis homozygous received the first in vivo gene therapy with administration of an E1-E3-deleted rAd vector expressing the normal human CFTR, and the subsequent clinical studies were then initiated. In recent two decades, Ads have been widely applied as vaccine carriers since they are capable of eliciting T and B cell responses. Furthermore, Ads can be genetically modified to induce the apoptosis of the cancer cells, which are known as the oncolytic Ads.Citation3,4 Ads are not only generally safe and can replicate in almost all the living cells, but also can be expanded easily in HEK293 cells and purified by CsCl gradient ultracentrifuge, and administered through oral, intranasal or intramuscular routes without adjuvants. Here, we review the Ads' potential in vaccine development against infectious pathogens or in cancer treatment, and address the latest advances in the field.

Biological characterization of Ad

Classification of the Ad

Ads are isolated from different mammalian species, such as human being, bovine and simian, among which the human Ads and chimpanzee Ads are widely used in the laboratory research or clinical study. Human Ads include more than 50 serotypes classified into subtype A to G, and chimpanzee Ads have more than 6 serotypes.Citation5-7 Human Ads are distributed widespreadly in the nature and most people have been infected, thus high neutralizing antibody titers were detected among the population.Citation8 Human subtype C Ads are the most common Ads which usually infect the children and cause upper respiratory tract infections or urinary tract infections. Human subtype B Ads sometimes cause severe eye or urinary tract infections. Some other serotypes, such as AdHu4 from subtype E, cause acute respiratory diseases.Citation9 However, most of the Ad infections are mild, which promotes Ad vectors into a new era as vaccine carriers.

Genome and structure of the Ad

Ads are DNA virus with icosahedral capsids of approximately 90 nm in diameter. Several studies have illustrated the structure of the Ads by cryo-electron microscopy.Citation10-12 The genomic DNA of Ads is about 26–45 kb, with two inverted terminal repeats of 100–140 bp flanking at both ends. The genes that express during the life cycle of Ads are generally divided into two types: the early genes and the late genes. The early genes include E1A, E1B, E2, E3 and E4, and they are mainly responsible for facilitating the replication of Ads by changing the expression levels of related host genes. The early genes can be further classified into two types: the immediate early genes (E1A) and the delayed early genes (E1B, E2, E3 and E4). E1A promotes the expression of the delayed early genes. The E1B protein generally suppresses the apoptosis of the host cells by binding to p53, Bak and Bax proteins.Citation13 The late genes are mainly responsible for the lysis of the host cells, assembly and release of the virions.

Ad virions mainly comprise two types of proteins: the capsid proteins and the core proteins. The core proteins mainly include proteins V, VII, X, and they mainly function as the DNA-associated proteins. The V protein mainly contacts with the nucleoli of the host cells and are involved in the viral assembly process.Citation14,15 The VII protein plays a key role in the manipulation of DNA, such as the DNA binding, the initiation of DNA replication and the viral genome's protection, et al.Citation16-18 The X protein is responsible for the viral chromosome condensation.Citation19 The capsid proteins comprise Hexon, Penton, fiber, IIIa, VIII and IX. There're 240 trimers of hexons on the surface of the Ad virions, and the hexons are the major structural protein on the capsids.Citation20 On the hexons, there're several hypervariable regions which are the major neutralization sites for the Ads, and the hypervariable regions can be replaced with other foreign antigens as potential vaccine carriers.Citation21-23 There have 12 pentamers of pentons on the top of the 12 icosahedral vertices, and they serve as the receptors for Ad internalization into the host cells,Citation24 and each vertex has 12 trimers of fibers protruding from the capsid surface which are mainly responsible for the interaction with the cellular receptors to initiate the viral entry.Citation10,25 The IIIa proteins are on the inner capsid surface, and are mainly responsible for the correct viral assembly, stabilization of the vertex region and the assembly of the packaged genome.Citation11,26 The VI proteins are inside the capsids, linking the core to the icosahedral shell, and are critical lytic factors of Ads during the endosome disruption.Citation27 The VIII proteins provide bonds between the peripentonal hexons and are involved in the stability of the capsids.Citation28 The IX proteins function to dampen the innate immune response, and affect the viral tropism and stability of the capsids.Citation12,29,30

Many Ads can be engineered for laboratory or clinical use. The recombinant Ads can be replication-incompetent or replication-competent. E1 gene is essential for the replication of the Ads, total or partial E1-deletion results that the vector can infect most of the living cells but cannot be expanded as it being replication-incompetent in normal cells. However, E1-deleted Ad can be propagated on dedicated helper cells, specialized cells that provide the E1 functions in trans, such as HEK 293 and PER.C6,Citation31 in which E1-deleted Ad is replication-competent.

Cellular receptors of the Ad

The entry of the Ads into host cells is initiated by the binding of fiber knob to the cell receptors. The CAR functions as the receptor for the fiber protein in subtype A and C-F Ads,Citation32-34 but in some cells, such as cancer cells and mature skeletal muscle cells, CAR is expressed in low levels.Citation35-37 In CAR-deficient lymphocytes, subtype C Ads can cause latent infection,Citation38 and the mutated CAR does not affect the tropism when Ads are administered systematically.Citation39 These results suggest that there might be other receptors for Ads except CAR. In fact, many other receptors for the entry of Ad have been found, for examples, CD46 or DSG2 for subtype B Ads,Citation40,41 and sialic acid and integrins, et al.Citation42-45

After the binding of the fiber knob to receptors, the virion internalization starts through endocytosis. Generally, the endocytosis is initiated by the binding of penton bases to the integrins,Citation43,46,47 but some reports revealed that the uptake of Ad virions could use lipid rafts or caveolae as entry route.Citation48 A review article suggested that when the Ad virions were coated with charged polymers, the entry routine might be changed accordingly.Citation49

Ad vector-based vaccine candidates for infectious diseases

Ad vectors are one of the most effective carriers for delivery of foreign antigens into the host cells. Compared with other viral vectors such as lentivirus, retrovirus and adeno-associated virus, etc, Ads are highly immunogenic and can induce both robust innate and adaptive immune responses in mammalian hosts. Ads have a large genome size, making the manipulation of the genetic DNA much more convenient. Unlike lentivirus or retrovirus, Ads do not integrate the viral genomic DNA into the hosts' genome, which reduces the risk of insertion mutagenesis. Adeno-associated virus is less pathogenic than Ads, but it is not yet suitable for mass production.Citation50 All above features make Ad a good vaccine carrier for the infectious diseases. and show a list of vaccine candidates based on Ad vectorsCitation51-88 against some certain infectious diseases.

Table 1. List of rAd vaccine candidates in clinical trials.

Table 2. List of rAd vaccine candidates in pre-clinical trials.

In the early stage, some Ads were modified as replicating-competent vectors with only E3 deletion which is not indispensible for the replication.Citation89 Nowadays, most of Ad vectors are replication-deficient with the deletion of E1 or both E1 and E3. E1-deficient vectors can only be rescued and expanded in the E1 trans-compensating cell lines. E3 deletion increases the packaging capacity of the Ad vectors, such E1-E3-deleted Ads can be incorporated with up to 7–8 kb foreign genes.Citation90 E4 can also be deleted, but the E4-deleted vectors only propagate in the E4-compensating cell lines.Citation91 The fully gutted Ad vectors were developed with containing the replication origins and packaging signals while most of the viral coding sequences were deleted. The fully gutted Ads can only be amplified with the appearance of helper virus.Citation90 Compared with the traditional Ad vectors, the fully gutted Ads have less toxicity caused by T cell responses, and the transgene products can be stably expressed.Citation92,93

Different Ad vectors can elicit different immune responses in various laboratory animals or species.Citation94 A study of SIV Gag-specific CD8(+) T cell responses in mice vaccinated with AdHu5, AdHu26 and AdHu35, respectively demonstrated that AdHu5 was more immunogenic than AdHu26 and AdHu35, but AdHu26 and AdHu35 generated long-lived memory T cells, whereas AdHu5 elicited more terminally differentiated phenotypes of T cells.Citation95 In another study, AdHu35, AdHu26 and AdHu48 were found to substantially produce higher levels of IFN-γ, IL-6 and 10-kDa gamma interferon-induced protein than AdHu5 in rhesus monkeys.Citation96 Based on their different immunogenicity profiles, certain serotype of Ads can be selected alternatively for specific researches. Ad vectors can be administered by injection and oral immunization, both of which elicit well immune responses, while oral administration can elicit mucosa immune response compared to injection, and greatly circumvent the pre-existing anti-vector immunity.Citation97,98

HIV vaccine based on Ad vector

Safe and efficient HIV vaccine is urgently needed since HIV still remains a severe public health threat. Several strategies have been developed for HIV vaccine design, of which Ad vectors are widely tested.Citation99 One of the most well-known clinical trials is AdHu5 based HIV vaccine which was developed by Merck, Inc.Citation51 In the clinical trial, the replication-incompetent AdHu5 vectors encoding gag, pol and nef genes were administered to 1494 participants at a dose of 3×1010 vp, while placebo administered to 1506 participants. This vaccine induced CD8+ T cell responses in homosexual men, but failed to prevent the HIV infection or reduce the early viral load. Further research revealed that the vaccine appeared to increase the risk of HIV infections in the AdHu5 serotype positive individuals.Citation56 To explain the phenomenon, several studies have been performed and suggested that one possible mechanism was that the immune complexes of AdHu5 and anti-AdHu5 antibodies could activate the dendritic cells and CD4+ T cellsCitation100 which might serve as the targets for HIV infection.Citation101

After the failure of the Merck HIV vaccine trial, other improved strategies have been tested, such as the regimen of DNA prime with AdHu5 boost. As reported by Churchyard GJ and Koup RA, et al,Citation53,102 a DNA plasmid encoding multiple HIV genes from multiple clades for priming at 0,1 and 2 month respectively at a dose of 4 mg, and AdHu5 expressing multiple HIV genes for boosting at 6 month induced polyfunctional CD4+ and CD8+ T cells as well as the anti-envelop binding antibodies, which revealed the heterologous prime-boost regimen was a potent immunization strategy for inducing both antibody and T cell responses. However, similar strategy used in another clinical trial with the regimen of priming 4 mg DNA encoding multiple HIV genes at week 0, 4 and 8, respectively, and boosting with 1010 pu rAdHu5 at week 24 reduced neither the rate of HIV-1 acquisition nor the viral-load set points in the participants.Citation103

As the AdHu5 based vaccine carriers are not suitable for HIV prevention, other serotype Ad based vaccines, such as AdHu26, AdHu35 or chimpanzee Ad vectors have been developed.Citation55,104,105 In a study of AdHu26 expressing HIV-1 envelop as a new vaccine candidate, both the AdHu26-serotype positive and negative participants received a single intramuscular immunization with 5×1010 vp rAdHu26.Citation106 The result revealed that rAdHu26 elicited both systemic and mucosal envelop-specific humoral and cellular immune responses, but interestingly the individuals with pre-existing AdHu26-specific neutralizing antibodies had comparable immune responses to the AdHu26-serotype negative ones. HIV vaccines based on the rAdHu35 expressing the HIV-1 envelop antigen have been studied.Citation107 In this phase 1b study, 192 healthy, HIV-uninfected participants were recruited and divided into one of following groups: rAdHu35/rAdHu5, DNA/rAdHu5, and DNA/rAdHu35 in AdHu5-seronegative persons, and DNA/rAdHu35 in AdHu5-seropositive persons, and a placebo group. The participants received three doses of 4 mg DNA or just one dose of 1010 pu rAd at the first 2 month, then were boosted with 1010 pu rAd at month 6. 4 weeks post boost, the immune responses were detected. The results indicated that all regimens were generally well tolerated and similarly immunogenic, and elicit cross-clade antibody responses including envelop V1/V2-specific IgG responses.

Presently, more novel Ad vectors are being discovered and developed for the HIV vaccine design, but none of them is ready for the market, thus more improvement is needed for the Ad vector-based HIV vaccine.

Influenza vaccine based on Ad vector

Ad vectors have been applied in the development of influenza vaccine. In most of Ad-based influenza vaccines, the influenza protein, such as HA, NP or M2 is expressed by the Ad vectors to induce neutralizing antibodies and T cell responses in the host. For example, HA protein of PR8 strain (H1N1) expressed by Ads can elicit HA-specific antibodies and cellular responses against the PR8 virus.Citation108 Besides Ad vectored vaccine against particular strain of influenza virus, the universal influenza vaccines based on Ads have been explored. In the multivalent influenza vaccines based on the replication-incompetent AdHu5, HAs from different subtypes and NP from one subtype were expressed on the rAds. The mice were immunized intramuscularly with 1010 PFU of rAds twice at 4-week interval. 4 weeks post the boost, high levels of humoral and cellular immune responses were well induced and the mice were protected from lethal challenge with H5, H7 and H9 avian influenza virus subtypes.Citation109 In another multivalent influenza vaccine based on AdHu4 and AdHu5,Citation110 HA genes from the H1, H3, H5 subtypes of influenza virus were expressed by the Ad vectors, and then immunized mice by rAdHu4-prime/rAdHu5-boost regimen at a doses of ranging from 107 to 1010 vp with a 4 week interval. The vaccination results revealed that the highest dose vaccine groups were 100% protected from the heterologous lethal challenge of different subtypes of influenza virus, indicating that Ad-based multivalent influenza vaccines had great potential in the prevention and control of the influenza virus.

Ad-based influenza vaccines have been tested in clinical trials. A non-replicating AdHu5 vector expressing HA from avian influenza and a TLR3 ligand were tested in humans.Citation60 Most of the participants received only one dose of Ads by capsule, with titers ranging from 108 to 1010 IU, but some were boosted with another dose of 109 IU Ad at 4 weeks post prime. The vaccination results revealed that the antigen specific cytotoxic and IFN-γ responses were induced in a dose dependent manner and cytotoxic responses increased after boost, which demonstrated that Ad-based vaccine administered orally could induce antigen specific immune responses and was safe as a vaccine candidate.Citation60 AdHu4 based avian influenza vaccine was developed and tested in a clinical trial. AdHu4 expressing HA of H5N1 was orally administered 3 times at a dose of 107 to 1010 vp within 56 days, and then boosted with 90 μg inactivated H5N1 viruses. The results demonstrated that cellular immune responses were well induced and oral administration of Ad might enhance the efficacy of poorly immunogenic vaccines such as H5N1, but the limitation of this study was that the HI titers were hardly measured.Citation61 By orally administered replication-competent AdHu4 vaccine, another improved strategy was reported.Citation111 In the study, the individuals were primed with AdHu4-H5-Vtn three times at dose of 107 to 1010 vp in 56 days, and then boosted with the 90 μg unadjuvanted licensed inactivated H5N1 subunit vaccine at 3.5 to 12 months. The new regimen induced high HI titers compared with unprimed individuals, which compensated for the disadvantages in the previous vaccination routine.

With the development of RNAi technology, Ad based RNAi strategies have been applied in prevention and control of influenza infection. For example, a novel chimpanzee Ad termed as AdC68 was used as the microRNAs expression vector and tested in mice. AdC68-expressing amiRNAs targeting M1, M2 or NP genes of influenza virus could efficiently suppress the viral replication and confer complete protection from the lethal challenge of H9N2 and H5N1.Citation112

Ebola vaccine based on Ad vector

Ebola virus was firstly discovered in 1976 with the outbreaks in Democratic Republic of Congo and Sudan,Citation113,114 but the outbreak in Africa in 2013 made it a public concern again.Citation115 There have been several strategies for the Ebola vaccine development, of which the Ad vectors are selected as a priority.

The NIH vaccine research center firstly developed a vaccine based on heterologous prime-boost regimen in 2000. In the research, the primates were firstly immunized with a DNA vaccine three times at 0, 4, 8 weeks at a dose of 4 mg, and then boosted with 1010 PFU AdHu5 which expressed GP protein of Ebola virus 20 weeks post prime. The results showed high antibody titers and CD4+T cell responses were induced in vaccinated animals, and the vaccinated groups had a higher survival rates than the control groups after challenged with Ebola virus.Citation116 Since this experiment took more than six months to complete the immunization flow chart, researchers developed an accelerated immunization method for the vaccination.Citation117 In the accelerated experiments, the animals were given AdHu5 expressing GP and NP of Ebola virus twice at doses of 2×1012 vp with a 9-week interval. The vaccinated animals were challenged with lethal Ebola virus and the protection was highly effective since the Ebola-specific CD8+ T cell and antibody responses were well induced. In the subsequent study, the animals were only primed with rAd–GP/NP and challenged 28 days later, but they still had high survival rates with either low or high doses of challenged virus. As the AdHu5-based Ebola vaccine showed a good prospect in the non-human primates, the first clinical trial based on AdHu5 was performed,Citation72 with a recombinant vaccine encoding the envelope GP from the Zaire and Sudan Ebola virus tested in a randomized, placebo-controlled, double-blinded, phase I human study. Thirty-one healthy adults received a single dose of the rAdHu5 at 2 × 109 vp (n = 12), or 2 × 1010 vp (n = 11) or placebo (n = 8). The results indicated the antibody responses to the two GPs in subjects were not well balanced. In the low dose group, antibody responses to Zaire GP were 50%, and 58% to Sudan GP. However, the antibody responses in the high dose group were 55% and 100%, respectively. In this study, the pre-existing neutralizing antibodies to AdHu5 was also noted, but it didn't appear to affect the T cell response to Ebola GP since 32% to 82% subjects responded with more CD4+ than CD8+ T cells.

Recently, Ebola vaccines based on the other serotypes are being developed, one of which is the chimpanzee Ad serotype 3 (cAd3). In a clinical trial of cAd3 based Ebola vaccine, 20 volunteers were recruited and divided into 2 groups. The volunteers in each group received the rAds expressing the GP protein from both Zaire and Sudan Ebola virus. One group received high titers of Ads with 1011 vp while another received 1010 vp, and two individuals in the high dose group experienced transient fever. The final results demonstrated that the rAds induced well specific antibody responses and T-cell responses, with higher levels of responses in the high dose group.Citation118 In another trial based on cAd3, 60 volunteers who were divided into 3 groups received a single dose of Ads ranging from 1×1010 vp to 5×1010 vp, and only 2 volunteers developed transient fever. After vaccination, specific antibody responses and T-cell responses were successfully elicited, but the levels were lower than those detected in the non-human primates.Citation119 In a recent phase I clinical trial which was performed between Oct 8, 2014, and Feb 16, 2015, 91 participants in Mali and 20 in the USA were recruited to receive cAd3 expressing GP of Ebola with a single dose ranging from 1010 to 1011 pu. After the prime of Ads, some Malians were boosted with vaccinia Ankara expressing GP of Zaire Ebola virus and filovirus antigens (MVA-BN-Phyllo). The vaccination results showed that 1 × 1011 pu single-dose rAds could suffice for a efficacy trials and the regimen of MVA-BN-Phyllo boosting could confer long-lived protection which might be needed for the health-care workers.Citation77

Other vaccines against infectious diseases based on Ad vector

Ad vectored vaccines have been developed for some other infectious diseases besides influenza virus, HIV and Ebola virus. A tetravalent dengue virus vaccine based on the rAds was tested in non-human primates. In the study, the prM and E gene from different subtypes of dengue virus were expressed by Ads. The vaccination included two doses of 109 IU rAds administration with a 57-day interval, and 85 days or 253 days post prime, the animals were challenged with dengue virus. The vaccination results revealed that the animals produced high-titer antibodies that could neutralize all four serotypes of dengue viruses in vitro. The challenge studies showed that significant protection from viremia was observed against all four dengue virus serotypes, but the protection efficacy was better in dengue-1 and dengue-3 challenges than in dengue-2 and dengue-4 challenges.Citation67

In addition to the Ad-based virus vaccine, Ads have been developed for the bacteria vaccine or protozoan vaccine. Mycobacterium tuberculosis causes serious bacterial infections in humans, and a vaccine based on AdHu5 expressing Ag85A has been tested in a phase 1 clinical trial. The results showed the polyfunctional CD4+ and CD8+ T cell responses were well stimulated, and the pre-existing neutralizing antibodies to AdHu5 had little influence on the potency of the vaccine.Citation82 Malaria, which is caused by Plasmodium falciparum, poses a serious threat to public health. An AdHu5 vector encoding the apical membrane antigen 1 and circumsporozoite protein of P. falciparum was evaluated in a clinical trial.Citation85 In the study, the DNA prime with Ad boost regimen was proved to be effective in eliciting specific T cell responses. Furthermore, some other serotypes of Ad, such as AdHu35 were developed for the malaria vaccines which listed in .

Human Ad serotypes such as AdHu5 have been extensively used for vaccine development mainly due to their excellent immunogenicity and safety. As the effect of pre-existing immunity on AdHu5-based vaccines, the clinical use of AdHu5 is greatly limited, while the rare human serotypes of Ads or non-human-originated Ads such as chimpanzee Ads have been extensively tested in both preclinical research and clinical trials.

Ad vector-based cancer therapy

Oncolytic Ads have shown great promise in cancer treatment since they exhibit distinct anti-cancer characteristics. During the life cycle of Ads, the Ad-infected cancer cells can be lysed in the end, and after the release of the Ad virions, they infect other cancer cells to initiate the next life cycle. Generally, two strategies are widely adopted for the modification of oncolytic Ads. The first one includes Ads expressing the therapeutic genes or combining RNAi technology to degrade the tumor promotion proteins. The second one mainly focuses on the capsids modification of the Ads, making Ads have specific tropism for the tumor cells or replicate to higher titers in the tumor tissues than in normal ones. These two strategies might be integrated to generate better anti-cancer effect.

One of the most well-known Ad-based anti-cancer drugs is the Advexin.Citation120 It is an E1-E3-deleted AdHu5 vector expressing p53 under the drive of CMV promoter in the E1 region. The Advexin has been applied in multiple cancer treatments, such as head and neck cancer, breast cancer and colon cancer, et al.Citation121-123 In the hand and neck squamous cell carcinoma, the Advexin was tested in a phase III clinical trial, patients were randomly treated with either Ad-p53 intratumorally on days 1 and 3 of each week at a daily dose of 2 × 1012 vp or methotrexate once weekly at a starting dose of 40 mg/m2, and each treatment cycle include 21 days. The results revealed that the vector was well tolerated and the anti-tumor activities were significant.Citation124 Another well-known drug against glioma is Sitimagene Ceradenovec. The drug is an E1-E3-deleted, AdHu5-based vaccine that expresses the herpes simplex virus' thymidine kinase at downstream of CMV promoter in E1 cassette. In the phase III trial, 250 patients were recruited with 124 in the AdHu5-treated group while 126 in the standard care group. Different groups received standard care plus injection of 1 × 1012 vp rAd or just standard care alone. Almost all the individuals experienced adverse events in the trial. The clinical results suggested that use of Sitimagene Ceradenovec increased the survival time or re-intervention in patients with newly diagnosed supratentorial glioblastoma multiforme.Citation125

Transduction of specific tumor antigen into dendritic cells is one of the most effective strategies against cancer. In an in vitro study, the DCs transduced with rAds expressing livin α induced strong specific cytotoxic T lymphocytes against different cancer cells.Citation126 Ads can be armed with immune modulator such as GMCSF or REIC to induced cytotoxic T lymphocytes against cancers. In a clinical trial, the patients received the combination treatment of Ad-GMCSF and alkylating agents had higher survival rates than the ones only treated with alkylating agents, possibly due to the activation of anti-tumor T cells.Citation127 In a E.G7 tumor-xenograft mouse model, Ad-REIC induced tumor-associated antigen specific cytotoxic T-lymphocytes, and the secreted REIC protein in the tumor generated a proper microenvironment for inducing of activated dendritic cells, resulting in decreased tumor size in the tumor-bearing mice receiving the Ad-REIC compared to the control groups.Citation128

RNAi technology is widely used in the downregulation of the specific gene's expression by sequence-specific degradation of the RISC complex.Citation129 The application of RNAi technology based on Ads may be extremely effective since the small RNA molecules can be steadily expressed, thus the targeted protein remains at low levels for long. One of the firstly used RNAi technologies based on Ads was the vascular endothelial growth factor (VEGF)-specific targeting small RNAs. To induce and maintain the long-lasting silencing of VEGF, the study constructed E1A-mutated, E1B-deleted Ads with shVEGF expressing at the E3 region under the drive of U6 promoter. After the vaccination of the Ad-shVEGF in tumor-bearing mice, potent anti-angiogenesis was induced and resulted in tumor suppression and survival benefits.Citation130 Recently, a study revealed that the amiRNAs based on the AdC68 vector could downregulate the survivin which was highly expressed in tumors, and the rAdC68 caused blockade of mitosis and cell cycle arrest at the G2/M phase. In the tumor-xenograft nude mice models, survivin-targeting amiRNAs expressed by rAdC68 effectively delayed growth of hepatocellular and cervical carcinomas.Citation131

Ads have great potential as anti-cancer vectors. However, the clinical use of Ads is limited due to their limited infectivity in some cancer cells. Modifying the tropism of the Ads is an alternative way to generate better anti-cancer effect. In a recent study, the epidermal growth factor-like domain of the human heregulin-α (HRG) was inserted into the HI loop of AdHu5 fiber without adverse effect on the Ad growth or yields. The fiber-modified Ad virions showed enhanced infection of cells expressing the cognate receptors HER3/ErbB3 and HER4/ErbB4, so the HER3-expressing Chinese hamster ovary (CHO) cells could be transduced by the HRG-modified virus, but not by wild type virus.Citation132 Other studies showed that the fiber-modified Ads had specific tropism to different cancer cells, which may provide a new strategy for Ad based cancer therapy.Citation133,134

Capsid-incorporation of foreign antigen into ad virions as vaccine candidate

The most common method for Ad vaccine development is the expression of the foreign antigens in the E1 or E3 region of Ad vector as previously described in and . However, pre-existing antibodies to the vectors may result in the failure of the vaccine. The “antigen capsid-incorporation” strategy has been developed to compensate for the drawbacks associated with the conventional antigen-expression system by the Ad vector to evade the pre-exisiting immunity. The Ad capsid proteins such as hexon, penton base, fiber, and pIX have variable sites for the antigen incorporation.Citation135,136

Hexon is the most abundant structural protein on the capsid and has several hypervariable regions which can be modified to display the foreign antigens without affecting the Ad's rescue and infectivity. For example, the AdC68 vectors were modified to express a linear B-cell epitope of the ectodomain of matrix 2 (M2e) of influenza virus within hypervariable regions 1 (HVR1) or HVR4 of the Ad hexon. Additional vectors with wild-type or M2e-modified hexon with influenza A virus NP as a transgene product in the E1-deleted region were also tested in the study. The vaccination regimen included priming with 1010 vp rAd and some mice boosted 2 months later. The pre-clinical study demonstrated that Ads expressing M2e within HVR1 of hexon induced higher magnitude and avidity of M2e-specific antibody responses than those carrying M2e within HVR4 or vectors expressing the M2e as part of a transgene product, and the M2e-specific antibody responses could be boosted by a second dose of the HVR1 hexon-modified vector but not by repeated immunization with the HVR4 hexon-modified vector.Citation23 Besides influenza virus vaccine, other studies reported that the insertion of the neutralizing epitopes of HFMD virus into hexon could elicit neutralizing antibodies against HFMD virus lethal challenge in the mice models.Citation22,137 As the HVRs of hexon contain the neutralizing epitopes of Ads,Citation20 the hexon-modified Ads might change the immunogenicity compared to the wild type Ads, and the anti-sera from the hexon-modified-Ad vaccinated animals cannot well neutralize the wild type Ads, which provides a good platform for the prime-boost regimens for Ad based vaccines.Citation22

Besides hexon, fiber can be engineered as an antigen-display system. Fiber modification makes Ads have specific tropism for cancer cells as above described, and can be incorporated with foreign antigens as vaccines against infectious diseases as well. A vector of AdHu5 expressing the 14-mer Pseudomonas aeruginosa immune-dominant outer membrane protein F (OprF) epitope 8 (Epi8) in five distinct sites of fiber was immunized in the mice. The results demonstrated that the FG-loop and HI-loop inserted sites were better than the other insertion sites in fiber since higher levels of protective immunity against P. aeruginosa were induced by FG-loop or HI-loop modified vectors.Citation138 The penton base and pIX were tested for antigen-incorporation in some studies.Citation139-142 However, compared with the other three proteins on the capsids, the penton base was rarely incorporated with foreign antigens perhaps due to the structural constraints.

Outlook and conclusions

Generally, Ad vectors are easy to be manipulated for genetic modification and capable of inducing potent antigen-specific immune responses. Most of the Ad species are rarely pathogenic to humans. Compared to the conventional vaccines, Ad vector-based vaccines can express a wide range of antigens from virus, bacteria or protozoan, and elicit long-term immune responses against infectious diseases. Despite the pre-existing neutralizing antibodies to the human Ads, the rare serotypes of Ads from different species have been developed to circumvent the disadvantages. All above advantages make Ads very attractive and potential vaccine candidates. Furthermore, Ad vectors show priority in anti-cancer research since they can be armed with therapeutic genes or modified to expand to higher titer in tumors than in the normal tissues. Many Ad vectors have been studied in animals against either infectious diseases or cancers, and revealed a good prospect of the further development.

Despite the incomplete success of Ad-based vaccines, Ad vectors still show great potential and are being extensively tested in the clinical trials recently. With more information obtained from Ad-related clinical trials, our understanding of the Ad vectors will be greatly enlarged, which will further promote the use of Ad vectors in the prevention and control of infectious diseases and cancer.

Abbreviations

Ad=

adenovirus

CAR=

coxsackie and adenovirus receptor

AdHu,=

human adenovirus

rAd=

recombinant Ad

DSG2=

desmoglein 2

HIV=

human immunodeficiency virus;

SIV=

simian immunodeficiency virus

MERS=

Middle East respiratory syndrome coronavirus

HA=

hemagglutinin

NP=

nucleoprotein

HI=

hemagglutination-inhibiting

GP,=

glycoprotein

amiRNA=

artificial microRNA

Disclosure of potential conflicts of interest

No potential conflicts of interest were disclosed.

Funding

This work was supported by grants from the “Knowledge Innovation Program” (No: Y014P31503) and the “100 Talent Program” (No: Y316P11503) from the Chinese Academy of Sciences and the Shanghai Pasteur Foundation.

References

  • Takafuji ET, Gaydos JC, Allen RG, Top FH, Jr. Simultaneous administration of live, enteric-coated adenovirus types 4, 7 and 21 vaccines: safety and immunogenicity. J Infect Dis 1979; 140:48-53; PMID:458200; http://dx.doi.org/10.1093/infdis/140.1.48
  • Rosenfeld MA, Siegfried W, Yoshimura K, Yoneyama K, Fukayama M, Stier LE, Pääkkö PK, Gilardi P, Stratford-Perricaudet LD, Perricaudet M. Adenovirus-mediated transfer of a recombinant α 1-antitrypsin gene to the lung epithelium in vivo. Science 1991; 252:431-4; PMID:2017680; http://dx.doi.org/10.1126/science.2017680
  • Khuri FR, Nemunaitis J, Ganly I, Arseneau J, Tannock IF, Romel L, Gore M, Ironside J, MacDougall RH, Heise C, et al. a controlled trial of intratumoral ONYX-015, a selectively-replicating adenovirus, in combination with cisplatin and 5-fluorouracil in patients with recurrent head and neck cancer. Nat Med 2000; 6:879-85; PMID:10932224; http://dx.doi.org/10.1038/78638
  • Bischoff JR, Kirn DH, Williams A, Heise C, Horn S, Muna M, Ng L, Nye JA, Sampson-Johannes A, Fattaey A, et al. An adenovirus mutant that replicates selectively in p53-deficient human tumor cells. Science 1996; 274:373-6; PMID:8832876; http://dx.doi.org/10.1126/science.274.5286.373
  • Lasaro MO, Ertl HC. New insights on adenovirus as vaccine vectors. Mol Therapy 2009; 17:1333-9; http://dx.doi.org/10.1038/mt.2009.130
  • Tatsis N, Ertl HC. Adenoviruses as vaccine vectors. Mol Therapy 2004; 10:616-29; http://dx.doi.org/10.1016/j.ymthe.2004.07.013
  • Walsh MP, Seto J, Liu EB, Dehghan S, Hudson NR, Lukashev AN, Ivanova O, Chodosh J, Dyer DW, Jones MS, et al. Computational analysis of two species C human adenoviruses provides evidence of a novel virus. J Clin Microbiol 2011; 49:3482-90; PMID:21849694; http://dx.doi.org/10.1128/JCM.00156-11
  • Wang X, Xing M, Zhang C, Yang Y, Chi Y, Tang X, Zhang H, Xiong S, Yu L, Zhou D. Neutralizing antibody responses to enterovirus and adenovirus in healthy adults in China. Emerg Microbes Infect 2014; 3:e30; PMID:26038738; http://dx.doi.org/10.1038/emi.2014.30
  • Hendrix RM, Lindner JL, Benton FR, Monteith SC, Tuchscherer MA, Gray GC, Gaydos JC. Large, persistent epidemic of adenovirus type 4-associated acute respiratory disease in US army trainees. Emerging Infect Dis 1999; 5:798-801; PMID:10603214; http://dx.doi.org/10.3201/eid0506.990609
  • Xia D, Henry LJ, Gerard RD, Deisenhofer J. Crystal structure of the receptor-binding domain of adenovirus type 5 fiber protein at 1.7 A resolution. Structure 1994; 2:1259-70; PMID:7704534; http://dx.doi.org/10.1016/S0969-2126(94)00126-X
  • Saban SD, Silvestry M, Nemerow GR, Stewart PL. Visualization of α-helices in a 6-angstrom resolution cryoelectron microscopy structure of adenovirus allows refinement of capsid protein assignments. J Virol 2006; 80:12049-59; PMID:17005667; http://dx.doi.org/10.1128/JVI.01652-06
  • Liu H, Jin L, Koh SB, Atanasov I, Schein S, Wu L, Zhou ZH. Atomic structure of human adenovirus by cryo-EM reveals interactions among protein networks. Science 2010; 329:1038-43; PMID:20798312; http://dx.doi.org/10.1126/science.1187433
  • Sundararajan R, Cuconati A, Nelson D, White E. Tumor necrosis factor-α induces Bax-Bak interaction and apoptosis, which is inhibited by adenovirus E1B 19K. J Biol Chem 2001; 276:45120-7; PMID:11571294; http://dx.doi.org/10.1074/jbc.M106386200
  • Ugai H, Dobbins GC, Wang M, Le LP, Matthews DA, Curiel DT. Adenoviral protein V promotes a process of viral assembly through nucleophosmin 1. Virology 2012; 432:283-95; PMID:22717133; http://dx.doi.org/10.1016/j.virol.2012.05.028
  • Matthews DA. Adenovirus protein V induces redistribution of nucleolin and B23 from nucleolus to cytoplasm. J Virol 2001; 75:1031-8; PMID:11134316; http://dx.doi.org/10.1128/JVI.75.2.1031-1038.2001
  • Johnson JS, Osheim YN, Xue Y, Emanuel MR, Lewis PW, Bankovich A, Beyer AL, Engel DA. Adenovirus protein VII condenses DNA, represses transcription, and associates with transcriptional activator E1A. J Virol 2004; 78:6459-68; PMID:15163739; http://dx.doi.org/10.1128/JVI.78.12.6459-6468.2004
  • Xue Y, Johnson JS, Ornelles DA, Lieberman J, Engel DA. Adenovirus protein VII functions throughout early phase and interacts with cellular proteins SET and pp32. J Virol 2005; 79:2474-83; PMID:15681448; http://dx.doi.org/10.1128/JVI.79.4.2474-2483.2005
  • Karen KA, Hearing P. Adenovirus core protein VII protects the viral genome from a DNA damage response at early times after infection. J Virol 2011; 85:4135-42; PMID:21345950; http://dx.doi.org/10.1128/JVI.02540-10
  • Anderson CW, Young ME, Flint SJ. Characterization of the adenovirus 2 virion protein, mu. Virology 1989; 172:506-12; PMID:2800334; http://dx.doi.org/10.1016/0042-6822(89)90193-1
  • Crawford-Miksza L, Schnurr DP. Analysis of 15 adenovirus hexon proteins reveals the location and structure of seven hypervariable regions containing serotype-specific residues. J Virol 1996; 70:1836-44; PMID:8627708
  • Pichla-Gollon SL, Drinker M, Zhou X, Xue F, Rux JJ, Gao GP, Wilson JM, Ertl HC, Burnett RM, Bergelson JM. Structure-based identification of a major neutralizing site in an adenovirus hexon. J Virol 2007; 81:1680-9; PMID:17108028; http://dx.doi.org/10.1128/JVI.02023-06
  • Zhang C, Yang Y, Chi Y, Yin J, Yan L, Ku Z, Liu Q, Huang Z, Zhou D. Hexon-modified recombinant E1-deleted adenoviral vectors as bivalent vaccine carriers for Coxsackievirus A16 and Enterovirus 71. Vaccine 2015; 33:5087-94; PMID:26296491; http://dx.doi.org/10.1016/j.vaccine.2015.08.016
  • Zhou D, Wu TL, Emmer KL, Kurupati R, Tuyishime S, Li Y, Giles-Davis W, Zhou X, Xiang Z, Liu Q, et al. Hexon-modified recombinant E1-deleted adenovirus vectors as dual specificity vaccine carriers for influenza virus. Mol Therapy 2013; 21:696-706; http://dx.doi.org/10.1038/mt.2012.248
  • Wickham TJ, Carrion ME, Kovesdi I. Targeting of adenovirus penton base to new receptors through replacement of its RGD motif with other receptor-specific peptide motifs. Gene Therapy 1995; 2:750-6; PMID:8750015
  • Henry LJ, Xia D, Wilke ME, Deisenhofer J, Gerard RD. Characterization of the knob domain of the adenovirus type 5 fiber protein expressed in Escherichia coli. J Virol 1994; 68:5239-46; PMID:8035520
  • San Martin C, Glasgow JN, Borovjagin A, Beatty MS, Kashentseva EA, Curiel DT, Marabini R, Dmitriev IP. Localization of the N-terminus of minor coat protein IIIa in the adenovirus capsid. J Mol Biol 2008; 383:923-34; PMID:18786542; http://dx.doi.org/10.1016/j.jmb.2008.08.054
  • Wodrich H, Henaff D, Jammart B, Segura-Morales C, Seelmeir S, Coux O, Ruzsics Z, Wiethoff CM, Kremer EJ. A capsid-encoded PPxY-motif facilitates adenovirus entry. PLoS Pathogens 2010; 6:e1000808; PMID:20333243; http://dx.doi.org/10.1371/journal.ppat.1000808
  • Fabry CM, Rosa-Calatrava M, Conway JF, Zubieta C, Cusack S, Ruigrok RW, Schoehn G. A quasi-atomic model of human adenovirus type 5 capsid. EMBO J 2005; 24:1645-54; PMID:15861131; http://dx.doi.org/10.1038/sj.emboj.7600653
  • Colby WW, Shenk T. Adenovirus type 5 virions can be assembled in vivo in the absence of detectable polypeptide IX. J Virol 1981; 39:977-80; PMID:7288921
  • de Vrij J, van den Hengel SK, Uil TG, Koppers-Lalic D, Dautzenberg IJ, Stassen OM, Bárcena M, Yamamoto M, de Ridder CM, Kraaij R, et al. Enhanced transduction of CAR-negative cells by protein IX-gene deleted adenovirus 5 vectors. Virol 2011; 410:192-200; http://dx.doi.org/10.1016/j.virol.2010.10.040
  • Havenga MJ, Holterman L, Melis I, Smits S, Kaspers J, Heemskerk E, van der Vlugt R, Koldijk M, Schouten GJ, Hateboer G, et al. Serum-free transient protein production system based on adenoviral vector and PER.C6 technology: high yield and preserved bioactivity. Biotechnol Bioengineering 2008; 100:273-83; http://dx.doi.org/10.1002/bit.21757
  • Bergelson JM, Cunningham JA, Droguett G, Kurt-Jones EA, Krithivas A, Hong JS, Horwitz MS, Crowell RL, Finberg RW. Isolation of a common receptor for Coxsackie B viruses and adenoviruses 2 and 5. Science 1997; 275:1320-3; PMID:9036860; http://dx.doi.org/10.1126/science.275.5304.1320
  • Kirby I, Lord R, Davison E, Wickham TJ, Roelvink PW, Kovesdi I, Sutton BJ, Santis G. Adenovirus type 9 fiber knob binds to the coxsackie B virus-adenovirus receptor (CAR) with lower affinity than fiber knobs of other CAR-binding adenovirus serotypes. J Virol 2001; 75:7210-4; PMID:11435605; http://dx.doi.org/10.1128/JVI.75.15.7210-7214.2001
  • Roelvink PW, Lizonova A, Lee JG, Li Y, Bergelson JM, Finberg RW, Brough DE, Kovesdi I, Wickham TJ. The coxsackievirus-adenovirus receptor protein can function as a cellular attachment protein for adenovirus serotypes from subgroups A, C, D, E, and F. J Virol 1998; 72:7909-15; PMID:9733828
  • Sachs MD, Rauen KA, Ramamurthy M, Dodson JL, De Marzo AM, Putzi MJ, Schoenberg MP, Rodriguez R. Integrin α(v) and coxsackie adenovirus receptor expression in clinical bladder cancer. Urology 2002; 60:531-6; PMID:12350512; http://dx.doi.org/10.1016/S0090-4295(02)01748-X
  • Fuxe J, Liu L, Malin S, Philipson L, Collins VP, Pettersson RF. Expression of the coxsackie and adenovirus receptor in human astrocytic tumors and xenografts. Int J Cancer J Int Du Cancer 2003; 103:723-9; http://dx.doi.org/10.1002/ijc.10891
  • Nalbantoglu J, Pari G, Karpati G, Holland PC. Expression of the primary coxsackie and adenovirus receptor is downregulated during skeletal muscle maturation and limits the efficacy of adenovirus-mediated gene delivery to muscle cells. Hum Gene Therapy 1999; 10:1009-19; http://dx.doi.org/10.1089/10430349950018409
  • Garnett CT, Talekar G, Mahr JA, Huang W, Zhang Y, Ornelles DA, Gooding LR. Latent species C adenoviruses in human tonsil tissues. J Virol 2009; 83:2417-28; PMID:19109384; http://dx.doi.org/10.1128/JVI.02392-08
  • Smith TA, Idamakanti N, Marshall-Neff J, Rollence ML, Wright P, Kaloss M, King L, Mech C, Dinges L, Iverson WO, et al. Receptor interactions involved in adenoviral-mediated gene delivery after systemic administration in non-human primates. Human Gene Therapy 2003; 14:1595-604; PMID:14633402; http://dx.doi.org/10.1089/104303403322542248
  • Gaggar A, Shayakhmetov DM, Lieber A. CD46 is a cellular receptor for group B adenoviruses. Nat Med 2003; 9:1408-12; PMID:14566335; http://dx.doi.org/10.1038/nm952
  • Wang H, Li ZY, Liu Y, Persson J, Beyer I, Moller T, Koyuncu D, Drescher MR, Strauss R, Zhang XB, et al. Desmoglein 2 is a receptor for adenovirus serotypes 3, 7, 11 and 14. Nat Med 2011; 17:96-104; PMID:21151137; http://dx.doi.org/10.1038/nm.2270
  • Nilsson EC, Storm RJ, Bauer J, Johansson SM, Lookene A, Angstrom J, Hedenström M, Eriksson TL, Frängsmyr L, Rinaldi S, et al. The GD1a glycan is a cellular receptor for adenoviruses causing epidemic keratoconjunctivitis. Nat Med 2011; 17:105-9; PMID:21151139; http://dx.doi.org/10.1038/nm.2267
  • Huang S, Kamata T, Takada Y, Ruggeri ZM, Nemerow GR. Adenovirus interaction with distinct integrins mediates separate events in cell entry and gene delivery to hematopoietic cells. J Virol 1996; 70:4502-8; PMID:8676475
  • Roelvink PW, Kovesdi I, Wickham TJ. Comparative analysis of adenovirus fiber-cell interaction: adenovirus type 2 (Ad2) and Ad9 utilize the same cellular fiber receptor but use different binding strategies for attachment. J Virol 1996; 70:7614-21; PMID:8892881
  • Arnberg N. Adenovirus receptors: implications for targeting of viral vectors. Trends Pharmacological Sci 2012; 33:442-8; http://dx.doi.org/10.1016/j.tips.2012.04.005
  • Davison E, Diaz RM, Hart IR, Santis G, Marshall JF. Integrin alpha5beta1-mediated adenovirus infection is enhanced by the integrin-activating antibody TS2/16. J Virol 1997; 71:6204-7; PMID:9223518
  • Davison E, Kirby I, Whitehouse J, Hart I, Marshall JF, Santis G. Adenovirus type 5 uptake by lung adenocarcinoma cells in culture correlates with Ad5 fibre binding is mediated by α(v)beta1 integrin and can be modulated by changes in beta1 integrin function. J Gene Med 2001; 3:550-9; PMID:11778901; http://dx.doi.org/10.1002/jgm.223
  • Colin M, Mailly L, Rogee S, D'Halluin JC. Efficient species C HAdV infectivity in plasmocytic cell lines using a clathrin-independent lipid raft/caveola endocytic route. Mol Therapy 2005; 11:224-36; http://dx.doi.org/10.1016/j.ymthe.2004.10.007
  • Choi JW, Lee JS, Kim SW, Yun CO. Evolution of oncolytic adenovirus for cancer treatment. Adv Drug Delivery Rev 2012; 64:720-9; http://dx.doi.org/10.1016/j.addr.2011.12.011
  • Ura T, Okuda K, Shimada M. Developments in Viral Vector-Based Vaccines. Vaccines 2014; 2:624-41; PMID:26344749; http://dx.doi.org/10.3390/vaccines2030624
  • Buchbinder SP, Mehrotra DV, Duerr A, Fitzgerald DW, Mogg R, Li D, Gilbert PB, Lama JR, Marmor M, Del Rio C, et al. Efficacy assessment of a cell-mediated immunity HIV-1 vaccine (the Step Study): a double-blind, randomised, placebo-controlled, test-of-concept trial. Lancet 2008; 372:1881-93; PMID:19012954; http://dx.doi.org/10.1016/S0140-6736(08)61591-3
  • Janes H, Frahm N, DeCamp A, Rolland M, Gabriel E, Wolfson J, Hertz T, Kallas E, Goepfert P, Friedrich DP, et al. MRKAd5 HIV-1 Gag/Pol/Nef vaccine-induced T-cell responses inadequately predict distance of breakthrough HIV-1 sequences to the vaccine or viral load. PloS One 2012; 7:e43396; http://dx.doi.org/10.1371/journal.pone.0043396
  • Churchyard GJ, Morgan C, Adams E, Hural J, Graham BS, Moodie Z, Grove D, Gray G, Bekker LG, McElrath MJ, et al. A phase IIA randomized clinical trial of a multiclade HIV-1 DNA prime followed by a multiclade rAd5 HIV-1 vaccine boost in healthy adults (HVTN204). PloS One 2011; 6:e21225
  • Baden LR, Walsh SR, Seaman MS, Tucker RP, Krause KH, Patel A, Johnson JA, Kleinjan J, Yanosick KE, Perry J, et al. First-in-human evaluation of the safety and immunogenicity of a recombinant adenovirus serotype 26 HIV-1 Env vaccine (IPCAVD 001). J Infect Dis 2013; 207:240-7; PMID:23125444; http://dx.doi.org/10.1093/infdis/jis670
  • Keefer MC, Gilmour J, Hayes P, Gill D, Kopycinski J, Cheeseman H, Cashin-Cox M, Naarding M, Clark L, Fernandez N, et al. A phase I double blind, placebo-controlled, randomized study of a multigenic HIV-1 adenovirus subtype 35 vector vaccine in healthy uninfected adults. PloS One 2012; 7:e41936; PMID:22870265; http://dx.doi.org/10.1371/journal.pone.0041936
  • Gray G, Buchbinder S, Duerr A. Overview of STEP and Phambili trial results: two phase IIb test-of-concept studies investigating the efficacy of MRK adenovirus type 5 gag/pol/nef subtype B HIV vaccine. Curr Opinion HIV AIDS 2010; 5:357-61; http://dx.doi.org/10.1097/COH.0b013e32833d2d2b
  • Frahm N, DeCamp AC, Friedrich DP, Carter DK, Defawe OD, Kublin JG, Casimiro DR, Duerr A, Robertson MN, Buchbinder SP, et al. Human adenovirus-specific T cells modulate HIV-specific T cell responses to an Ad5-vectored HIV-1 vaccine. J Clin Invest 2012; 122:359-67; PMID:22201684; http://dx.doi.org/10.1172/JCI60202
  • Barouch DH, Liu J, Peter L, Abbink P, Iampietro MJ, Cheung A, Alter G, Chung A, Dugast AS, Frahm N, et al. Characterization of humoral and cellular immune responses elicited by a recombinant adenovirus serotype 26 HIV-1 Env vaccine in healthy adults (IPCAVD 001). J Infect Dis 2013; 207:248-56
  • Van Kampen KR, Shi Z, Gao P, Zhang J, Foster KW, Chen DT, Marks D, Elmets CA, Tang DC. Safety and immunogenicity of adenovirus-vectored nasal and epicutaneous influenza vaccines in humans. Vaccine 2005; 23:1029-36; PMID:15620476; http://dx.doi.org/10.1016/j.vaccine.2004.07.043
  • Peters W, Brandl JR, Lindbloom JD, Martinez CJ, Scallan CD, Trager GR, Tingley DW, Kabongo ML, Tucker SN. Oral administration of an adenovirus vector encoding both an avian influenza A hemagglutinin and a TLR3 ligand induces antigen specific granzyme B and IFN-gamma T cell responses in humans. Vaccine 2013; 31:1752-8; PMID:23357198; http://dx.doi.org/10.1016/j.vaccine.2013.01.023
  • Gurwith M, Lock M, Taylor EM, Ishioka G, Alexander J, Mayall T, Ervin JE, Greenberg RN, Strout C, Treanor JJ, et al. Safety and immunogenicity of an oral, replicating adenovirus serotype 4 vector vaccine for H5N1 influenza: a randomised, double-blind, placebo-controlled, phase 1 study. Lancet Infect Dis 2013; 13:238-50; PMID:23369412; http://dx.doi.org/10.1016/S1473-3099(12)70345-6
  • Gao W, Soloff AC, Lu X, Montecalvo A, Nguyen DC, Matsuoka Y, Robbins PD, Swayne DE, Donis RO, Katz JM, et al. Protection of mice and poultry from lethal H5N1 avian influenza virus through adenovirus-based immunization. J Virol 2006; 80:1959-64; PMID:16439551; http://dx.doi.org/10.1128/JVI.80.4.1959-1964.2006
  • Prevec L, Campbell JB, Christie BS, Belbeck L, Graham FL. A recombinant human adenovirus vaccine against rabies. J Infect Dis 1990; 161:27-30; PMID:2295855; http://dx.doi.org/10.1093/infdis/161.1.27
  • Vos A, Neubert A, Pommerening E, Muller T, Dohner L, Neubert L, Hughes K. Immunogenicity of an E1-deleted recombinant human adenovirus against rabies by different routes of administration. J General Virol 2001; 82:2191-7; http://dx.doi.org/10.1099/0022-1317-82-9-2191
  • Bouet-Cararo C, Contreras V, Fournier A, Jallet C, Guibert JM, Dubois E, Thiery R, Bréard E, Tordo N, Richardson J, et al. Canine adenoviruses elicit both humoral and cell-mediated immune responses against rabies following immunisation of sheep. Vaccine 2011; 29:1304-10; PMID:21134446; http://dx.doi.org/10.1016/j.vaccine.2010.11.068
  • Xiang ZQ, Greenberg L, Ertl HC, Rupprecht CE. Protection of non-human primates against rabies with an adenovirus recombinant vaccine. Virol 2014; 450-451:243-9; http://dx.doi.org/10.1016/j.virol.2013.12.029
  • Raviprakash K, Wang D, Ewing D, Holman DH, Block K, Woraratanadharm J, Chen L, Hayes C, Dong JY, Porter K, et al. A tetravalent dengue vaccine based on a complex adenovirus vector provides significant protection in rhesus monkeys against all four serotypes of dengue virus. J Virol 2008; 82:6927-34; PMID:18480438; http://dx.doi.org/10.1128/JVI.02724-07
  • Jaiswal S, Khanna N, Swaminathan S. Replication-defective adenoviral vaccine vector for the induction of immune responses to dengue virus type 2. J Virol 2003; 77:12907-13; PMID:14610213; http://dx.doi.org/10.1128/JVI.77.23.12907-12913.2003
  • Khanam S, Pilankatta R, Khanna N, Swaminathan S. An adenovirus type 5 (AdV5) vector encoding an envelope domain III-based tetravalent antigen elicits immune responses against all four dengue viruses in the presence of prior AdV5 immunity. Vaccine 2009; 27:6011-21; PMID:19665609; http://dx.doi.org/10.1016/j.vaccine.2009.07.073
  • Guo X, Deng Y, Chen H, Lan J, Wang W, Zou X, Hung T, Lu Z, Tan W. Systemic and mucosal immunity in mice elicited by a single immunization with human adenovirus type 5 or 41 vector-based vaccines carrying the spike protein of Middle East respiratory syndrome coronavirus. Immunol 2015; 145:476-84; http://dx.doi.org/10.1111/imm.12462
  • Kim E, Okada K, Kenniston T, Raj VS, AlHajri MM, Farag EA, AlHajri F, Osterhaus AD, Haagmans BL, Gambotto A. Immunogenicity of an adenoviral-based Middle East Respiratory Syndrome coronavirus vaccine in BALB/c mice. Vaccine 2014; 32:5975-82; PMID:25192975; http://dx.doi.org/10.1016/j.vaccine.2014.08.058
  • Ledgerwood JE, Costner P, Desai N, Holman L, Enama ME, Yamshchikov G, Mulangu S, Hu Z, Andrews CA, Sheets RA, et al. A replication defective recombinant Ad5 vaccine expressing Ebola virus GP is safe and immunogenic in healthy adults. Vaccine 2010; 29:304-13; PMID:21034824; http://dx.doi.org/10.1016/j.vaccine.2010.10.037
  • Sullivan NJ, Hensley L, Asiedu C, Geisbert TW, Stanley D, Johnson J, Honko A, Olinger G, Bailey M, Geisbert JB, et al. CD8+ cellular immunity mediates rAd5 vaccine protection against Ebola virus infection of nonhuman primates. Nat Med 2011; 17:1128-31; PMID:21857654; http://dx.doi.org/10.1038/nm.2447
  • Geisbert TW, Bailey M, Hensley L, Asiedu C, Geisbert J, Stanley D, Honko A, Johnson J, Mulangu S, Pau MG, et al. Recombinant adenovirus serotype 26 (Ad26) and Ad35 vaccine vectors bypass immunity to Ad5 and protect nonhuman primates against ebolavirus challenge. J Virol 2011; 85:4222-33; PMID:21325402; http://dx.doi.org/10.1128/JVI.02407-10
  • Wong G, Richardson JS, Cutts T, Qiu X, Kobinger GP. Intranasal immunization with an adenovirus vaccine protects guinea pigs from Ebola virus transmission by infected animals. Anti Viral Res 2015; 116:17-9; http://dx.doi.org/10.1016/j.antiviral.2015.01.001
  • Wong G, Richardson JS, Pillet S, Racine T, Patel A, Soule G, Ennis J, Turner J, Qiu X, Kobinger GP. Adenovirus-Vectored Vaccine Provides Postexposure Protection to Ebola Virus-Infected Nonhuman Primates. J Infect Dis 2015; 212 Suppl 2:S379-83; PMID:25957963; http://dx.doi.org/10.1093/infdis/jiv102
  • Tapia MD, Sow SO, Lyke KE, Haidara FC, Diallo F, Doumbia M, et al. Use of ChAd3-EBO-Z Ebola virus vaccine in Malian and US adults, and boosting of Malian adults with MVA-BN-Filo: a phase 1, single-blind, randomised trial, a phase 1b, open-label and double-blind, dose-escalation trial, and a nested, randomised, double-blind, placebo-controlled trial. Lancet Infect dis 2015; 16:31-42; PMID:26546548; http://dx.doi.org/10.1016/S1473-3099(15)00362-X
  • Choi JH, Schafer SC, Zhang L, Kobinger GP, Juelich T, Freiberg AN, Croyle MA. A single sublingual dose of an adenovirus-based vaccine protects against lethal Ebola challenge in mice and guinea pigs. Mol Pharmaceutics 2012; 9:156-67; http://dx.doi.org/10.1021/mp200392g
  • Li W, Li M, Deng G, Zhao L, Liu X, Wang Y. Prime-boost vaccination with Bacillus Calmette Guerin and a recombinant adenovirus co-expressing CFP10, ESAT6, Ag85A and Ag85B of Mycobacterium tuberculosis induces robust antigen-specific immune responses in mice. Mol Med Reports 2015; 12:3073-80
  • Rahman S, Magalhaes I, Rahman J, Ahmed RK, Sizemore DR, Scanga CA, Weichold F, Verreck F, Kondova I, Sadoff J, et al. Prime-boost vaccination with rBCG/rAd35 enhances CD8(+) cytolytic T-cell responses in lesions from Mycobacterium tuberculosis-infected primates. Mol Med 2012; 18:647-58; PMID:22396020; http://dx.doi.org/10.2119/molmed.2011.00222
  • Hoft DF, Blazevic A, Stanley J, Landry B, Sizemore D, Kpamegan E, Gearhart J, Scott A, Kik S, Pau MG, et al. A recombinant adenovirus expressing immunodominant TB antigens can significantly enhance BCG-induced human immunity. Vaccine 2012; 30:2098-108; PMID:22296955; http://dx.doi.org/10.1016/j.vaccine.2012.01.048
  • Smaill F, Jeyanathan M, Smieja M, Medina MF, Thanthrige-Don N, Zganiacz A, Yin C, Heriazon A, Damjanovic D, Puri L, et al. A human type 5 adenovirus-based tuberculosis vaccine induces robust T cell responses in humans despite preexisting anti-adenovirus immunity. Sci Translational Med 2013; 5:205ra134; http://dx.doi.org/10.1126/scitranslmed.3006843
  • Tamminga C, Sedegah M, Maiolatesi S, Fedders C, Reyes S, Reyes A, Vasquez C, Alcorta Y, Chuang I, Spring M, et al. Human adenovirus 5-vectored Plasmodium falciparum NMRC-M3V-Ad-PfCA vaccine encoding CSP and AMA1 is safe, well-tolerated and immunogenic but does not protect against controlled human malaria infection. Hum Vaccines Immunotherapeutics 2013; 9:2165-77; http://dx.doi.org/10.4161/hv.24941
  • Schwenk R, Banania G, Epstein J, Kim Y, Peters B, Belmonte M, Ganeshan H, Huang J, Reyes S, Stryhn A, et al. Ex vivo tetramer staining and cell surface phenotyping for early activation markers CD38 and HLA-DR to enumerate and characterize malaria antigen-specific CD8+ T-cells induced in human volunteers immunized with a Plasmodium falciparum adenovirus-vectored malaria vaccine expressing AMA1. Malaria J 2013; 12:376; http://dx.doi.org/10.1186/1475-2875-12-376
  • Chuang I, Sedegah M, Cicatelli S, Spring M, Polhemus M, Tamminga C, Patterson N, Guerrero M, Bennett JW, McGrath S, et al. DNA prime/Adenovirus boost malaria vaccine encoding P. falciparum CSP and AMA1 induces sterile protection associated with cell-mediated immunity. PloS One 2013; 8:e55571; PMID:23457473; http://dx.doi.org/10.1371/journal.pone.0055571
  • Ouedraogo A, Tiono AB, Kargougou D, Yaro JB, Ouedraogo E, Kabore Y, Kangoye D, Bougouma EC, Gansane A, Henri N, et al. A phase 1b randomized, controlled, double-blinded dosage-escalation trial to evaluate the safety, reactogenicity and immunogenicity of an adenovirus type 35 based circumsporozoite malaria vaccine in Burkinabe healthy adults 18 to 45 years of age. PloS One 2013; 8:e78679; PMID:24244339; http://dx.doi.org/10.1371/journal.pone.0078679
  • Karen KA, Deal C, Adams RJ, Nielsen C, Ward C, Espinosa DA, Xie J, Zavala F, Ketner G. A replicating adenovirus capsid display recombinant elicits antibodies against Plasmodium falciparum sporozoites in Aotus nancymaae monkeys. Infect Immunity 2015; 83:268-75; http://dx.doi.org/10.1128/IAI.02626-14
  • Sheehy SH, Duncan CJ, Elias SC, Choudhary P, Biswas S, Halstead FD, Collins KA, Edwards NJ, Douglas AD, Anagnostou NA, et al. ChAd63-MVA-vectored blood-stage malaria vaccines targeting MSP1 and AMA1: assessment of efficacy against mosquito bite challenge in humans. Mol Therapy 2012; 20:2355-68; http://dx.doi.org/10.1038/mt.2012.223
  • Ginsberg HS. The life and times of adenoviruses. Adv Virus Res 1999; 54:1-13; PMID:10547672; http://dx.doi.org/10.1016/S0065-3527(08)60363-2
  • Kochanek S. High-capacity adenoviral vectors for gene transfer and somatic gene therapy. Hum Gene Therapy 1999; 10:2451-9; http://dx.doi.org/10.1089/10430349950016807
  • Yeh P, Dedieu JF, Orsini C, Vigne E, Denefle P, Perricaudet M. Efficient dual transcomplementation of adenovirus E1 and E4 regions from a 293-derived cell line expressing a minimal E4 functional unit. J Virol 1996; 70:559-65; PMID:8523570
  • Toietta G, Mane VP, Norona WS, Finegold MJ, Ng P, McDonagh AF, Beaudet AL, Lee B. Lifelong elimination of hyperbilirubinemia in the Gunn rat with a single injection of helper-dependent adenoviral vector. Proc Natl Acad Sci U S A 2005; 102:3930-5; PMID:15753292; http://dx.doi.org/10.1073/pnas.0500930102
  • Gilbert R, Dudley RW, Liu AB, Petrof BJ, Nalbantoglu J, Karpati G. Prolonged dystrophin expression and functional correction of mdx mouse muscle following gene transfer with a helper-dependent (gutted) adenovirus-encoding murine dystrophin. Hum Mol Genetics 2003; 12:1287-99; http://dx.doi.org/10.1093/hmg/ddg141
  • Dicks MD, Guzman E, Spencer AJ, Gilbert SC, Charleston B, Hill AV, Cottingham MG. The relative magnitude of transgene-specific adaptive immune responses induced by human and chimpanzee adenovirus vectors differs between laboratory animals and a target species. Vaccine 2015; 33:1121-8; PMID:25629523; http://dx.doi.org/10.1016/j.vaccine.2015.01.042
  • Tan WG, Jin HT, West EE, Penaloza-MacMaster P, Wieland A, Zilliox MJ, McElrath MJ, Barouch DH, Ahmed R. Comparative analysis of simian immunodeficiency virus gag-specific effector and memory CD8+ T cells induced by different adenovirus vectors. J Virol 2013; 87:1359-72; PMID:23175355; http://dx.doi.org/10.1128/JVI.02055-12
  • Teigler JE, Iampietro MJ, Barouch DH. Vaccination with adenovirus serotypes 35, 26, and 48 elicits higher levels of innate cytokine responses than adenovirus serotype 5 in rhesus monkeys. J Virol 2012; 86:9590-8; PMID:22787208; http://dx.doi.org/10.1128/JVI.00740-12
  • Xiang ZQ, Gao GP, Reyes-Sandoval A, Li Y, Wilson JM, Ertl HC. Oral vaccination of mice with adenoviral vectors is not impaired by preexisting immunity to the vaccine carrier. J Virol 2003; 77:10780-9; PMID:14512528; http://dx.doi.org/10.1128/JVI.77.20.10780-10789.2003
  • Liebowitz D, Lindbloom JD, Brandl JR, Garg SJ, Tucker SN. High titre neutralising antibodies to influenza after oral tablet immunisation: a phase 1, randomised, placebo-controlled trial. Lancet Infect Dis 2015; 15:1041-8; PMID:26333337; http://dx.doi.org/10.1016/S1473-3099(15)00266-2
  • Richert L, Lhomme E, Fagard C, Levy Y, Chene G, Thiebaut R. Recent developments in clinical trial designs for HIV vaccine research. Hum Vaccines Immunotherapeutics 2015; 11:1022-9; http://dx.doi.org/10.1080/21645515.2015.1011974
  • Perreau M, Pantaleo G, Kremer EJ. Activation of a dendritic cell-T cell axis by Ad5 immune complexes creates an improved environment for replication of HIV in T cells. J Exp Med 2008; 205:2717-25; PMID:18981239; http://dx.doi.org/10.1084/jem.20081786
  • Benlahrech A, Harris J, Meiser A, Papagatsias T, Hornig J, Hayes P, Lieber A, Athanasopoulos T, Bachy V, Csomor E, et al. Adenovirus vector vaccination induces expansion of memory CD4 T cells with a mucosal homing phenotype that are readily susceptible to HIV-1. Proc Natl Acad Sci U S A 2009; 106:19940-5; PMID:19918060; http://dx.doi.org/10.1073/pnas.0907898106
  • Koup RA, Roederer M, Lamoreaux L, Fischer J, Novik L, Nason MC, Larkin BD, Enama ME, Ledgerwood JE, Bailer RT, et al. Priming immunization with DNA augments immunogenicity of recombinant adenoviral vectors for both HIV-1 specific antibody and T-cell responses. PloS One 2010; 5:e9015; PMID:20126394; http://dx.doi.org/10.1371/journal.pone.0009015
  • Hammer SM, Sobieszczyk ME, Janes H, Karuna ST, Mulligan MJ, Grove D, Koblin BA, Buchbinder SP, Keefer MC, Tomaras GD, et al. Efficacy trial of a DNA/rAd5 HIV-1 preventive vaccine. N Eng J Med 2013; 369:2083-92; http://dx.doi.org/10.1056/NEJMoa1310566
  • Cheng C, Wang L, Ko SY, Kong WP, Schmidt SD, Gall JG, Colloca S, Seder RA, Mascola JR, Nabel GJ. Combination recombinant simian or chimpanzee adenoviral vectors for vaccine development. Vaccine 2015; 33:7344-51; PMID:26514419; http://dx.doi.org/10.1016/j.vaccine.2015.10.023
  • Baden LR, Liu J, Li H, Johnson JA, Walsh SR, Kleinjan JA, Engelson BA, Peter L, Abbink P, Milner DA, Jr, et al. Induction of HIV-1-specific mucosal immune responses following intramuscular recombinant adenovirus serotype 26 HIV-1 vaccination of humans. J Infect Dis 2015; 211:518-28; PMID:25165165; http://dx.doi.org/10.1093/infdis/jiu485
  • Baden LR, Liu JY, Li HL, Johnson JA, Walsh SR, Kleinjan JA, Engelson BA, Peter L, Abbink P, Milner DA, Jr, et al. Induction of HIV-1-specific mucosal immune responses following intramuscular recombinant adenovirus serotype 26 HIV-1 vaccination of humans. J Infect Dis 2015; 211:518-28; PMID:25165165 http://dx.doi.org/10.1093/infdis/jiu485
  • Fuchs JD, Bart PA, Frahm N, Morgan C, Gilbert PB, Kochar N, DeRosa SC, Tomaras GD, Wagner TM, Baden LR, et al. Safety and Immunogenicity of a Recombinant Adenovirus Serotype 35-Vectored HIV-1 Vaccine in Adenovirus Serotype 5 Seronegative and Seropositive Individuals. J AIDS Clin Res 2015; 6:23; http://dx.doi.org/10.4172/2155-6113.1000461
  • Van Kampen KR, Shi ZK, Gao P, Zhang JF, Foster KW, Chen DT, Marks D, Elmets CA, Tang DC. Safety and immunogenicity of adenovirus-vectored nasal and epicutaneous influenza vaccines in humans. Vaccine 2005; 23:1029-36; PMID:15620476; http://dx.doi.org/10.1016/j.vaccine.2004.07.043
  • Vemula SV, Ahi YS, Swaim AM, Katz JM, Donis R, Sambhara S, Mittal SK. Broadly protective adenovirus-based multivalent vaccines against highly pathogenic avian influenza viruses for pandemic preparedness. PloS One 2013; 8; http://dx.doi.org/10.1371/journal.pone.0062496
  • Webby RJ, Weaver EA. Centralized Consensus Hemagglutinin Genes Induce Protective Immunity against H1, H3 and H5 Influenza Viruses. PloS One 2015; 10:e0140702; PMID:26469190; http://dx.doi.org/10.1371/journal.pone.0140702
  • Khurana S, Coyle EM, Manischewitz J, King LR, Ishioka G, Alexander J, Smith J, Gurwith M, Golding H. Oral priming with replicating adenovirus serotype 4 followed by subunit H5N1 vaccine boost promotes antibody affinity maturation and expands H5N1 cross-clade neutralization. PloS One 2015; 10:e0115476; PMID: 25629161; http://dx.doi.org/10.1371/journal.pone.0115476
  • Zhang H, Tang X, Zhu C, Song Y, Yin J, Xu J, Ertl HC, Zhou D. Adenovirus-mediated artificial MicroRNAs targeting matrix or nucleoprotein genes protect mice against lethal influenza virus challenge. Gene therapy 2015; 22:653-62; PMID:25835311; http://dx.doi.org/10.1038/gt.2015.31
  • Feldmann H, Jones S, Klenk HD, Schnittler HJ. Ebola virus: from discovery to vaccine. Nat Rev Immunol 2003; 3:677-85; PMID:12974482; http://dx.doi.org/10.1038/nri1154
  • Feldmann H, Geisbert TW. Ebola haemorrhagic fever. Lancet 2011; 377:849-62; PMID:21084112; http://dx.doi.org/10.1016/S0140-6736(10)60667-8
  • Sacks JA, Zehe E, Redick C, Bah A, Cowger K, Camara M, Diallo A, Gigo AN, Dhillon RS, Liu A. Introduction of Mobile Health Tools to Support Ebola Surveillance and Contact Tracing in Guinea. Global Health Sci Practice 2015; 3:646-59; http://dx.doi.org/10.9745/GHSP-D-15-00207
  • Sullivan NJ, Sanchez A, Rollin PE, Yang ZY, Nabel GJ. Development of a preventive vaccine for Ebola virus infection in primates. Nature 2000; 408:605-9; PMID:11117750; http://dx.doi.org/10.1038/35046108
  • Sullivan NJ, Geisbert TW, Geisbert JB, Xu L, Yang ZY, Roederer M, Koup RA, Jahrling PB, Nabel GJ. Accelerated vaccination for Ebola virus haemorrhagic fever in non-human primates. Nature 2003; 424:681-4; PMID:12904795; http://dx.doi.org/10.1038/nature01876
  • Ledgerwood JE, DeZure AD, Stanley DA, Novik L, Enama ME, Berkowitz NM, Hu Z, Joshi G, Ploquin A, Sitar S, et al. Chimpanzee Adenovirus Vector Ebola Vaccine - Preliminary Report. N Eng J Med 2014; PMID: 25426834; http://dx.doi.org/10.1056/NEJMoa1410863
  • Rampling T, Ewer K, Bowyer G, Wright D, Imoukhuede EB, Payne R, Hartnell F, Gibani M, Bliss C, Minhinnick A, et al. A Monovalent Chimpanzee Adenovirus Ebola Vaccine - Preliminary Report. N Eng J Med 2016; 374:1635-46.
  • Wolf JK, Bodurka DC, Gano JB, Deavers M, Ramondetta L, Ramirez PT, Levenback C, Gershenson DM. A phase I study of Adp53 (INGN 201; ADVEXIN) for patients with platinum- and paclitaxel-resistant epithelial ovarian cancer. Gynecologic Oncol 2004; 94:442-8; http://dx.doi.org/10.1016/j.ygyno.2004.05.041
  • Wu J, Zhu Y, Xu C, Xu H, Zhou X, Yang J, Xie Y, Tao M. Adenovirus-mediated p53 and ING4 gene cotransfer elicits synergistic antitumor effects through enhancement of p53 acetylation in breast cancer. Oncol Reports 2016; 35:243-52
  • Nemunaitis J. Head and neck cancer: response to p53-based therapeutics. Head Neck 2011; 33:131-4; PMID:20222046; http://dx.doi.org/10.1002/hed.21364
  • Nie B, Shen Z, Wen JB, Wong OG, Hsueh WD, Huo LF, Kung HF, Jiang B, Lin MC. AAV-HGFK1 and Ad-p53 cocktail therapy prolongs survival of mice with colon cancer. Mol Cancer Therapeutics 2008; 7:2855-65; http://dx.doi.org/10.1158/1535-7163.MCT-08-0366
  • Nemunaitis J, Clayman G, Agarwala SS, Hrushesky W, Wells JR, Moore C, Hamm J, Yoo G, Baselga J, Murphy BA, et al. Biomarkers Predict p53 gene therapy efficacy in recurrent squamous cell carcinoma of the head and neck. Clin Cancer Res 2009; 15:7719-25; PMID:19996201; http://dx.doi.org/10.1158/1078-0432.CCR-09-1044
  • Westphal M, Yla-Herttuala S, Martin J, Warnke P, Menei P, Eckland D, Kinley J, Kay R, Ram Z. Adenovirus-mediated gene therapy with sitimagene ceradenovec followed by intravenous ganciclovir for patients with operable high-grade glioma (ASPECT): a randomised, open-label, phase 3 trial. Lancet Oncol 2013; 14:823-33; PMID:23850491; http://dx.doi.org/10.1016/S1470-2045(13)70274-2
  • Xie J, Guo X, Liu F, Luo J, Duan F, Tao X. In vitro antitumor immune response induced by dendritic cells transduced with human livin α recombinant adenovirus. Cell Immunol 2015; 297:46-52; PMID:26140980; http://dx.doi.org/10.1016/j.cellimm.2015.06.003
  • Liikanen I, Ahtiainen L, Hirvinen ML, Bramante S, Cerullo V, Nokisalmi P, Hemminki O, Diaconu I, Pesonen S, Koski A, et al. Oncolytic adenovirus with temozolomide induces autophagy and antitumor immune responses in cancer patients. Mol Therapy 2013; 21:1212-23; http://dx.doi.org/10.1038/mt.2013.51
  • Ariyoshi Y, Watanabe M, Eikawa S, Yamazaki C, Sadahira T, Hirata T, Araki M, Ebara S, Nasu Y, Udono H, et al. The induction of antigen-specific CTL by in situ Ad-REIC gene therapy. Gene Therapy 2016; PMID:26836118; http://dx.doi.org/10.1038/gt.2016.7
  • Yao C, Sasaki HM, Ueda T, Tomari Y, Tadakuma H. Single-Molecule Analysis of the Target Cleavage Reaction by the Drosophila RNAi Enzyme Complex. Mol Cell 2015; 59:125-32; PMID:26140368; http://dx.doi.org/10.1016/j.molcel.2015.05.015
  • Yoo JY, Kim JH, Kwon YG, Kim EC, Kim NK, Choi HJ, Yun CO. VEGF-specific short hairpin RNA-expressing oncolytic adenovirus elicits potent inhibition of angiogenesis and tumor growth. Mol Therapy 2007; 15:295-302; http://dx.doi.org/10.1038/sj.mt.6300023
  • Chi Y, Wang X, Yang Y, Zhang C, Ertl HC, Zhou D. Survivin-targeting Artificial MicroRNAs Mediated by Adenovirus Suppress Tumor Activity in Cancer Cells and Xenograft Models. Mol Therapy Nucleic Acids 2014; 3:e208; PMID:25368912; http://dx.doi.org/10.1038/mtna.2014.59
  • MacLeod SH, Elgadi MM, Bossi G, Sankar U, Pisio A, Agopsowicz K, Sharon D, Graham FL, Hitt MM. HER3 targeting of adenovirus by fiber modification increases infection of breast cancer cells in vitro, but not following intratumoral injection in mice. Cancer Gene Therapy 2012; 19:888-98; PMID:23099884; http://dx.doi.org/10.1038/cgt.2012.79
  • Zhan Y, Yu B, Wang Z, Zhang Y, Zhang HH, Wu H, Feng X, Geng RS, Kong W, Yu XH. A fiber-modified adenovirus co-expressing HSV-TK and Coli.NTR enhances antitumor activities in breast cancer cells. Int J Clin Exp Pathol 2014; 7:2850-60; PMID:25031704
  • Takagi-Kimura M, Yamano T, Tamamoto A, Okamura N, Okamura H, Hashimoto-Tamaoki T, Tagawa M, Kasahara N, Kubo S. Enhanced antitumor efficacy of fiber-modified, midkine promoter-regulated oncolytic adenovirus in human malignant mesothelioma. Cancer Sci 2013; 104:1433-9; PMID:23962292; http://dx.doi.org/10.1111/cas.12267
  • Rux JJ, Burnett RM. Adenovirus structure. Hum Gene Therapy 2004; 15:1167-76; http://dx.doi.org/10.1089/hum.2004.15.1167
  • Nemerow GR, Pache L, Reddy V, Stewart PL. Insights into adenovirus host cell interactions from structural studies. Virology 2009; 384:380-8; PMID:19019405; http://dx.doi.org/10.1016/j.virol.2008.10.016
  • Tian X, Su X, Li X, Li H, Li T, Zhou Z, Zhong T, Zhou R. Protection against enterovirus 71 with neutralizing epitope incorporation within adenovirus type 3 hexon. Plos One 2012; 7:e41381; PMID:22848478; http://dx.doi.org/10.1371/journal.pone.0041381
  • Sharma A, Krause A, Xu Y, Sung B, Wu W, Worgall S. Adenovirus-based vaccine with epitopes incorporated in novel fiber sites to induce protective immunity against Pseudomonas aeruginosa. PloS One 2013; 8:e56996; PMID:23437292; http://dx.doi.org/10.1371/journal.pone.0056996
  • Krause A, Joh JH, Hackett NR, Roelvink PW, Bruder JT, Wickham TJ, Kovesdi I, Crystal RG, Worgall S. Epitopes expressed in different adenovirus capsid proteins induce different levels of epitope-specific immunity. J Virol 2006; 80:5523-30; PMID:16699033; http://dx.doi.org/10.1128/JVI.02667-05
  • Vigne E, Mahfouz I, Dedieu JF, Brie A, Perricaudet M, Yeh P. RGD inclusion in the hexon monomer provides adenovirus type 5-based vectors with a fiber knob-independent pathway for infection. J Virol 1999; 73:5156-61; PMID:10233980
  • Dmitriev IP, Kashentseva EA, Curiel DT. Engineering of adenovirus vectors containing heterologous peptide sequences in the C terminus of capsid protein IX. J Virol 2002; 76:6893-9; PMID:12072490; http://dx.doi.org/10.1128/JVI.76.14.6893-6899.2002
  • Parks RJ. Adenovirus protein IX: a new look at an old protein. Mol Therapy 2005; 11:19-25; http://dx.doi.org/10.1016/j.ymthe.2004.09.018