offprints and colour figure order form Credit Card

0 downloads 0 Views 952KB Size Report
Offprints Cost. Colour Figures. Total Cost. 3. PAYMENT. Credit Card Payment: Credit Card. Please charge my AMEX/VISA/MasterCard (delete as appropriate).
offprints and colour figure order form 1. OFFPRINTS You are entitled to order one set of 100 offprints at a discounted rate of: £270 / $530 / €450. These prices are only valid if the form is received before the issue goes to press.

Article details Manuscript ID: ____430912________________________ First author: ____Andrea Amalfitano _______ Title: _____________Overcoming pre-existing Adenovirus immunity by genetic_________________ ________________________engineering of Adenovirus-based vectors____________________________ Journal title: Expert Opinion on Biological Therapy

Volume and issue:____________9(12)__________

Please tick / delete as appropriate: ! I wish to order 100 additional copies of the above article. Please charge: £270 / $530 / €450 Delivery address: ________________________________________________________________________ ______________________________________________________________________________________ ______________________________________________________________________________________

2. COLOUR FIGURES I require the following number of figures to be printed in colour: __________________ Figure numbers: _______________________________________________________ Please charge: £100 / $150 / €120 per figure Colour Figures

Offprints Cost

Total Cost

3. PAYMENT

Credit Card Pa yment: Credit Card

Please charge my AMEX/VISA/MasterCard (delete as appropriate)

Card No

Exp.date

/

CVV number (last 3 or 4 digits on reverse of card): Name on credit card: Address where registered: Signature:

Date:

All orders are accepted subject to Informa Healthcare’s standard Terms and Conditions.

Please fax back completed order to: Jonathan Collard +44 (0)20 7017 7667 Material code: 5639 (ROW); 7943 (USA)

Journal: Expert Opinion on Biological Therapy (EOBT) Title: Overcoming pre-existing Adenovirus immunity by genetic engineering of Adenovirus-based vectors ID: 430912 Corresponding author: Andrea Amalfitano AUTHOR: Please complete the colour figure order form that accompanies your proof if you require figures to appear in colour in the print version of the journal; once you have completed your article and credit card details, email or fax the form back to the production editor. The following queries have arisen during the editing of your manuscript. Please answer the queries by making the necessary corrections on the CATS online corrections form. Once you have added all your corrections, please press the SUBMIT *button.

Page number 8

Line number

Query AQ1: Please confirm that the declaration of interest is reflective of the submitted form and relevant to this article. AQ2: Ref [99] may be available in print by the time this review is published. AQ3: Please provide volume, page range for ref [94]. AQ4: Please check the page range is ok for ref [59].

Review 1

Overcoming pre-existing Adenovirus immunity by genetic engineering of Adenovirus-based vectors

5

10

15

20

25

30

1.

Introduction

2.

Ad capsid modifications

3.

Alternative Ad serotypes

4.

Genome modified Ad5 vectors

5.

Expert opinion and overall summary

Sergey S Seregin & Andrea Amalfitano† †

Michigan State University, Department of Microbiology and Molecular Genetics, 4194 Biomedical and Physical Sciences Bldg, East Lansing, MI 48823, USA

Adenovirus (Ad)-based vectors offer several benefits showing their potential for use in a variety of vaccine applications. Recombinant Ad-based vaccines possess potent immunogenic potential, capable of generating humoral and cellular immune responses to a variety of pathogen-specific antigens expressed by the vectors. Ad5 vectors can be readily produced, allowing for usage in thousands of clinical trial subjects. This is now coupled with a history of safe clinical use in the vaccine setting. However, traditional Ad5-based vaccines may not be generating optimal antigen-specific immune responses, and generate diminished antigen-specific immune responses when pre-existing Ad5 immunity is present. These limitations have driven initiation of several approaches to improve the efficacy of Ad-based vaccines, and/or allow modified vaccines to overcome pre-existing Ad immunity. These include: generation of chemically modified Ad5 capsids; generation of chimeric Ads; complete replacement of Ad5-based vaccine platforms with alternative (human and non-human origin) Ad serotypes, and Ad5 genome modification approaches that attempt to retain the native Ad5 capsid, while simultaneously improving the efficacy of the platform as well as minimizing the effect of pre-existing Ad immunity. Here we discuss recent advances in- and limitations of each of these approaches, relative to their abilities to overcome pre-existing Ad immunity.

35 Keywords: adaptive immunity, cellular responses, humoral responses, innate immunity, neutralizing antibodies, recombinant Adenovirus Expert Opin. Biol. Ther. (2009) 9(12):1-11

40 1.

45

50

55

Introduction

Recombinant, Adenovirus (Ad)-based gene transfer vector platforms are heavily utilized in both gene therapy and vaccine applications. This utility is based on a number of positive attributes inherent to the use of recombinant Ads. Recombinant Ads are capable of transducing foreign genes into a wide range of cell types including antigen presenting cells; allow for efficient transgene expression, have large cloning capacities (8 – 36 kb); and can be easily produced to extremely high titers in a current good manufacturing practices (cGMP)-compliant fashion (up to 1 ! 1013 virus particles/ml). The ability to easily ‘scale-up’ traditional Ad vectors in a manner that is cGMP-compliant, has resulted in thousands of patients safely receiving recombinant, Ad5-based gene transfer vectors. In fact, it is the authors opinion that the proven ability to mass-produce Ad-based vectors makes them one of the most pragmatic of all gene transfer vectors currently proposed for direct in vivo human use. For example, the lack of simple, efficient, and large-scale cGMP production capabilities has limited the clinical utilization of a number of other 10.1517/14712590903307388 © 2009 Informa UK Ltd ISSN 1471-2598 All rights reserved: reproduction in whole or in part not permitted

1

Overcoming pre-existing Adenovirus immunity by genetic engineering of Adenovirus-based vectors

56

60

65

70

proposed gene transfer platforms (i.e., Adeno-associated virus (AAV) or lentivirus based systems) to a handful of human clinical trials (see: http://www.wiley.co.uk/wileychi/genmed/ clinical/). There are, however, several limitations to the use of recombinant Ad vectors as a vaccine platform, the primary one of which is the decreased efficacy of these platforms, especially in Ad-immune individuals, concerns that are the focus of this review. Recently, another concern has been raised, regarding the use of Ad-based vectors, namely the possibility that administration of HIV-specific Ad vaccines into individuals with pre-existing anti-Ad immunity might ‘enhance’ or predispose the vaccinees to infection by HIV. We will discuss and address this issue first, and use the observations to validate the efforts being undertaken by several groups to overcome problems associated with the diminished efficacy of Ad-based vaccines, especially in the context of pre-existing Ad immunity. The STEP trial Most recently, the Merck sponsored STEP trial revealed that a E1 deleted ([E1-]) Ad5-based HIV specific vaccine (consisting of a cocktail of three [E1-] Ad5 vectors respectively expressing the HIV-1 derived gag, pol and nef antigens) failed in preventing HIV infection in vaccinees. This disappointing outcome positively correlated with poor elicitation of antiGag-specific T cell responses in the Ad5 vaccinees, responses that were especially blunted in Ad5-immune individuals [1,2]. Simultaneously, it was widely reported that Ad5-immune vaccinees appeared to have higher rates of HIV infection than the Ad5-immune placebo group [1]. The STEP trial was designed so as to result in 50 trial participants getting infected by HIV per subgroup (Ad immune or Ad nonimmune groups, based on baseline Ad5 specific antibody titer analysis). In placebo-treated Ad5 immune participants, only 13 out of 528 (~ 2.3%) of these individuals in the Ad immune group of patients treated with a placebo subsequently went on to develop HIV-AIDS, while 29 out of 532 (~ 5.5%) of the Ad5 immune patients vaccinated with the Ad5 vaccine went on to develop HIV-AIDS. While in isolation, this result suggests ‘enhanced’ acquisition of HIV in Ad5-immune Ad5-HIV vaccine recipients, it must be noted that in Ad5 non-immune individuals the rates of HIV-AIDS in placebo and Ad5 vaccinated individuals were also ~ 5.1% and ~ 5.2% respectively [3-5]. Thus the Ad-immune Ad5-HIV vaccinees had a rate of HIV-AIDS infection identical to the rate of HIV-AIDS infection that occurred in both of the Ad5-naı¨ve treatment groups. As has been pointed out by others, if one is to conclude that being Ad5 immune ‘enhances’ the rate of HIV-AIDS infection in Ad5 vaccinees, one would be equally justified in concluding that being Ad5 immune and being treated with a placebo may prevent HIV-AIDS [5] Thus one caveat of the STEP trial is that for some reason the placebo-treated group of Ad5-immune clinical trial participants had a lower than expected HIV infection ‘event rate’ than all other trial subgroups, confounding the 1.1

75

80

85

90

95

100

105

110

2

data. Reasons for this are several, and have recently focused on circumcision rates being more closely correlated with HIV infection that Ad5 immune status [1]. Furthermore, the Ad5 immune status criteria used in the STEP trial may itself be at fault [6-8]. For example, the potential induction of T cell memory responses in Ad5 immune individuals by pre-existing Ad5 antibodies has been forwarded as a possible mechanism for enhanced HIV infection rates in Ad5 immune vaccinees [6-8]. However, STEP trial derived patient samples, supposedly Ad5 ‘antibody negative’ STEP trial participants (that did not get infected with HIV) did, in fact have preexisting T cell responses to Ad5 [6-8]. Although these latter studies have not assessed all potential immune responses present in the trial participants (i.e., pre-existing mucosal immune responses), the studies strongly suggest that preexisting Ad5 immunity did not correlate with acquisition of HIV in STEP trial vaccinees. Due to these and other considerations, we feel that the use of Ad5-based vaccines should not be limited due to this sole concern. However, many in the lay press and, in fact, the scientific theatre wrongly hold the view that Ad-based clinical trials are no longer being conducted or pursued due to this and/or other issues. Thus, it is very important to note the fact that Ad5-based vectors are currently, the most widely used gene transfer vector in human clinical trials (http://www.wiley. co.uk/wileychi/genmed/clinical/). More recently, a major advantage to the use of Ad-based vaccines has also become apparent, with the confirmation that integrating viral vectors (i.e., retrovirus, lentivirus, possibly AAVs) can cause insertional mutagenesis and cancer in animal models and in human clinical trial subjects, therefore vaccines based on such vectors may also be subject to similar risks [9-13]. The natural biology of the recombinant Ad genome, which does not integrate into the host chromosome, largely mitigates insertional mutagenesis risks [14]. Efficient administration of Ads has been reported utilizing: systemic intravenous delivery [15,16], intranasal administration [17] and intramuscular [18-20], footpad [21], and/or subcutaneous injections [22]. More relevant to the point of this review, Ad5-based vectors have been repeatedly proven to be a potent vaccine platform, capable of delivering and expressing significant amounts of Ad vaccine expressed target antigens to antigen-presenting cells, inducing rapid and robust antigen specific cellular as well as humoral immune responses [23-30]. Ad-based vaccines have been proven to generate not only systemic immunity, but can also induce robust mucosal immunity when enterically or intramuscularly administered [18,31]. This capability has now been verified in humans, as STEP trial subjects intramuscularly vaccinated with Ad5-based vaccines were found to have increased populations of HIV-antigen-specific cellular (T cell) immune responses, a feat not accomplished by previous use of DNA or poxvirus-based HIV vaccines expressing the same antigens [1,32-36]. These immune responses, were severely blunted in Ad5-immune individuals, and

Expert Opin. Biol. Ther. (2009) 9(12)

111

115

120

125

130

135

140

145

150

155

160

165

Seregin & Amalfitano

166

were also not potent enough to prevent HIV acquisition even in Ad-naı¨ve subjects. Pre-existing, Ad5 immunity Unfortunately, a majority of the human population has been infected by wild type Ads in childhood, and often these individuals develop pre-existing immunity to a number of common Ad serotypes (i.e., serotypes 2, 4, 5 and 7). The percentage of the human population with medium to high Ad-specific antibody titers largely depends upon geographic location, with 30 – 50% of the United States population, and up to 90% of the sub-Saharan African populations having significant Ad5 antibodies with an overall trend to having higher titers in developing countries [32,37,38]. Pre-existing immunity to Ad can include both neutralizing antibodies (NAb), as well as Ad-protein-specific T cell responses. Additionally, recent studies highlight a critical role for CD8+ T cells in pre-existing Ad immunity [39,40]. This latter point is very important, as these pre-existing responses can in fact still be harnessed when a host is exposed to a novel (previously unencountered) Ad serotype [41-44]. Therefore, in this review we discuss strategies that have been proposed to improve the efficacy or utility of Ad based vaccines. Sometimes these strategies can overcome specific aspects of the pre-existing Ad immune response, as well potentially allowing for improved vaccination of Ad-immune individuals with these same recombinant Ad-based vaccines [25,41,45]. 1.2

170

175

180

185

190

2.

Ad capsid modifications

2.1

Chemical modifications of the Ad capsid

195

200

205

210

215

220

Several groups have chemically modified the Ad5 capsid (i.e., in a non-covalent fashion) in an attempt to shield or hide highly antigenic epitopes on the viral capsid (mainly hexon protein) from the immune system of the Ad-immune host. This strategy primarily involves the addition of synthetic polymers onto the Ad capsid. These polymers can include PEG [46], polyactic glycolic acid (PLGA) [47] or other lipids [48]. PEGylation appears to be the most promising approach for non-covalent modification of Ad capsids. Ad5 PEGylation may allow for re-administration of such modified Ads in mice. For example, animals were given intravenous injections of an Ad5 vector expressing an irrelevant antigen, and then second injections of conventional or PEGylated Ad5 vectors each expressing the bacterial b-galactosidase gene (b-Gal) [16]. High levels of b-Gal expression were found only in Ad5-immune mice that received the the PEGylated Ad5 vector [16]. Moreover, it was shown that PEGylated Ad5 vectors also reduce the induction of Ad5-triggered innate toxicities, such as thrombocytopenia, plasma alanine aminotransferae (ALT) elevations, and release of pro-inflammatory cytokines such as IL-6 [16,49]. PEGylation with a smaller PEG (5 kDa) moiety increases transduction efficiency of the Ad5 capsid in Ad-immune mice [50]. Use of a 5 kDa PEGylation of Ad5

can also reduce Ad5-specific neutralizing antibody (Nab) titers generated after intranasal injection into Ad-naive BALB/c mice [17]. However, no significant differences in generating antigen-specific immune responses were found when different size PEGylated Ads (3 – 35 kDa PEG) and/or different injection routes were analyzed [17]. Several promising trends were noted, mainly highlighting that PEGylated Ads may be better as a ‘boosting’ vaccine relative to conventional Ad vaccines [17]. How PEGylation may affect the efficiency or tropism of Ad5-mediated gene transduction in vivo has not been fully delineated. For example, a dramatic reduction of liver transduction was reported after Ad PEGylation [51], while other studies, reported equal and/or increased liver transduction by PEGylated Ads [16,49]. In summary, characterization of how PEGylation affects the ability of so modified Ad vaccines relative to tropism changes, ability to transduce lymphoid tissues, ability to evade pre-existing Ad5 NAb and/or T cell immunity, is still required to determine if these strategies afford improved utility of PEGylated Ad vaccines, especially in the Ad-immune host.

221

225

230

235

240

Genetic modification of the Ad capsid Insertion of novel peptides into Ad capsid proteins 2.2

2.2.1

The non-enveloped Ad virion is composed of a large capsid containing nine proteins. The hexon (protein II), penton base (protein III) and fiber (protein IV) proteins are the most abundant, and are so termed the major capsid proteins, while proteins IIIa, VI, VIII and IX are referred to as the minor capsid proteins [52-54]. The fiber, penton, protein IX and hexon proteins have been exploited for genetic insertion of foreign peptides into these specific capsid proteins, either as ‘in-frame’ insertions within the proteins, or as ‘in-frame’ C-terminal fusions. If the insertion does not cause a deleterious change in the biology of the so-modified protein, (i.e., causes misfolding, lack of stability, lack of trimerization or lack of incorporation into the tertiary structure of the Ad capsid) novel, recombinant Ad vectors now ‘capsid-displaying’ the respective foreign peptide can be isolated. The penton and hexon proteins tolerate relatively small peptide insertions: up to 18 amino acids (aa) for penton base [55,56], and up to 36 aa within the hypervariable region 5 (HVR5) of hexon [19,57], whereas the HI loop of the fiber knob and C-terminus of protein IX allow for incorporation of peptides of substantial size: up to 83 aa for fiber [58] and up to 1018 aa for pIX [59]. Issues regarding capability of generating these recombinant viruses consistently, to very high titers, or in a cGMP-compliant fashion have many times not been addressed in studies published to date, but will need to be if clinical deployment of these vectors is to be achieved. The insertion of foreign peptides into capsid proteins in attempts to change the natural tropism of the Ad vector have been well-described [60,61]. However, in this review, we will shift our focus to studies using Ad ‘capsid-display’ as a method to introduce pathogen-derived antigens into a host to generate

Expert Opin. Biol. Ther. (2009) 9(12)

3

245

250

255

260

265

270

275

Overcoming pre-existing Adenovirus immunity by genetic engineering of Adenovirus-based vectors

276

280

285

290

295

300

305

310

315

immune responses to the respective antigens. For example, antigenic epitopes derived from the hemagglutinin (HA) protein of the influenza A virus were incorporated into and displayed from the Ad capsid as hexon, penton base, fiber knob or protein IX fusions [23]. The immunogenicity of these novel vaccines was then cross-compared. The fiberdisplaying Ads induced the highest levels of HA-specific immunity, as determined by measuring production of HA-specific IgM and IgG humoral responses, as well as HA-specific IL-4- or IFN-g-producing CD4+ T cellular immune responses [23]. In another example, the immunodominant portions of the Bacillus anthracis protective antigen (PA) were genetically inserted and displayed from the HVR5 site of the Ad5 hexon [19,57]. Intramuscular injections of the novel vector into BALB/c mice resulted in generation of PA-specific IgG1 and IgG2a antibodies, possibly indicating that TH1 and TH2 immunity to the antigen was generated, surpassing the efficacy of synthetic peptide based vaccination strategies [19]. The capabilities of these novel vaccine platforms in the context of pre-existing Ad5 immunity have not been extensively reported. However, in one study, the HVR5 site of hexon has also been exploited for display of Pseudomonas aeruginosa B cell epitope-encoding peptide. Footpad vaccinations with the novel vaccines induced antibody responses to P. aeruginosa 2 – 4 weeks post injection [21]. Both IFNg-positive CD4+ and CD8+ P. aeruginosa specific T cell responses were also generated. Most convincingly, mice vaccinated with the P. aeruginosa B cell epitope displaying Ad were protected against subsequent lethal pulmonary challenge with several P. aeruginosa strains. Subsequent studies with Ad vaccines simultaneously expressing (as a transgene encoded by the Ad vaccine genome) and displaying P. aeruginosa antigens suggested an improved ability to increase humoral responses to the bacterially derived antigens upon boosting injection relative to repeated vaccinations with a conventional Ad5 vaccine expressing the same antigen [21,62]. More extensive future studies are required to shed light on whether ‘capsid-display’ is a viable approach to overcome pre-existing Ad immunity. Ad vaccine chimeras – modifications of hexon and/or fiber

2.2.2

320

325

330

Classification of Ad serotypes have been historically based upon biological, biochemical and structural properties, including the lack of cross-neutralization between serotypes from different species. In fact, neutralization and hemagglutination tests are the most common clinical tests utilized to distinguish infections by different Ad serotypes. Subsequently such parameters as oncogenicity, length of fiber protein and genomic sequence similarity were taken into consideration to further sub-classify Ads [63,64]. The capsid proteins hexon and fiber are the major targets of Ad specific antibodies, many of which become neutralizing [40,65-69]. Some investigators have therefore begun to ‘swap’ portions of the Ad5 capsid with 4

homologous proteins from alternative serotype Ads, generating ‘chimeric’ vectors that have new properties as a result of the modifications. The Ad fiber and hexon proteins have been the two main targets for generation of chimeric Ads. Several fiber-chimeric Ads [70,71], including Ad5 chimeric Ads with fibers from non-human Ads [72] have been described. These novel vectors were primarily generated to alter the tissue tropism of the resultant vectors, since the fiber protein primarily interacts with specific cellular receptors, initiating virus transduction and thus directing vector tropism [72]. Subsequent studies revealed that Ad5 vectors lacking the fiber knob domain have reduced susceptibility to pre-existing Ad5 antibodies, supporting the notion that the fiber protein is one of the major targets of Ad antibodies in humans [69]. Incidentally, systemically administered Ad5/7 and Ad5/41 fiber chimeras had dramatically reduced activation of Ad triggered innate toxicities as compared to unmodified Ad5, but identical levels of transgene specific adaptive responses [15]. Although theoretically possible, the ability of fiberchimeric Ads to evade pre-existing Ad immunity has not been extensively reported. Initial studies suggested increased ability of a chimp AdC23/C24 fiber chimera to deliver and express a transgene in AdC23 pre-immune mice as compared with a AdC23 unmodified vector [73]. Subcutaneously injected Ad5/3 fiber knob chimeric vectors had an increased ability to evade pre-existing Ad5 immunity in immune-competent mice pre-immunized with unmodified Ad5 [22]. Hexon is the most abundant protein of the Ad capsid, and thus it stands to reason that hexon is also a major target for host neutralizing antibodies against Ads [40,65-68]. Therefore several reports have attempted to replace the Ad5 hexon, with hexon moieties derived from alternative Ad serotypes to evade pre-existing Ad5-neutralizing antibodies [74]. For example, chimpanzee derived AdC68-hexon-specific monoclonal antibodies predominantly recognize a small hexon loop region of the AdC68 hexon. Introduction of a mutation into this region allowed the modified virus to not only escape neutralization by the same monoclonal antibodies, but also reduced its neutralization by polyclonal sera [75]. While this study clearly provided evidence that the hexon R1 loop region is a major site for neutralizing antibodies, it did not directly address in vivo efficiency of the modified-hexon-containing vector, both in vector-naı¨ve and vector-immune hosts [75]. It was also recently shown that after intramuscular administration into Ad5 pre-immunized BALB/c mice, a modified Ad5 vaccine deleted for the HVR5 region of the Ad5 hexon was able to increase HIV env-specific humoral and cellular immune responses as compared with a conventional Ad5 vaccine expressing the same antigen [20]. More extensive modification of the hexon protein has also been described. In one study, hexon proteins, derived from 18 different serotypes were tested for their ability to replace the Ad5 hexon protein. However, only four serotypes yielded hexons capable of allowing for generation of viable chimeras [76]. Several groups now report the ability to produce

Expert Opin. Biol. Ther. (2009) 9(12)

331

335

340

345

350

355

360

365

370

375

380

385

Seregin & Amalfitano

386

390

395

Ad5 vectors with the Ad5 hexon replaced with hexons derived from Ad1 [76], Ad2 [66], Ad3 [68], Ad6 [76], and Ad12 [67]. Each of these chimeric vectors has shown an ability to evade pre-existing Ad5 immunity. While these studies provide proof of concept that hexonor fiber- swapped chimeric Ads can be viable, Ad5/48 or Ad5/26 hexon chimeric vaccines were also found to outperform Ad5-based conventional vaccine when administered into Ad5-immune animals. Most dramatically, a chimeric Ad5/48 with seven Ad5 hexon HVRs replaced with Ad48derived HVRs was constructed and produced, confirmed to be stable and infectious, and was shown to generate robust HIV-gag specific immune responses while evading pre-existing Ad5 immunity in a non-human primate model [77].

400 3.

405

410

415

420

425

430

435

440

Alternative Ad serotypes

Development of recombinant Ad vectors or vaccines, based entirely upon use of rarer alternative serotype Ads or nonhuman-derived Ads in lieu of Ad5, is a seemingly simple and straightforward alternative method for overcoming preexisting Ad5 immunity. Rare Ad serotypes can be selected based upon their minimal cross reactivity with Ad2- and Ad5-neutralizing antibodies, a feature, which by definition clinically delineated human Ad serotypes. Low seroprevalence for Ad serotypes from subgroups B (Ads 11, 35 and 50) and D (Ads 26, 48 and 49) justified their initial selection as possible alternatives to Ad5-based Ad vaccines, and testing for immunogenicity commenced in small animal models [32]. Interestingly, Ad5-based vaccines revealed the highest SIV-gag-specific T and B cell responses in Ad-naı¨ve mice, with Ad26 being the most potent of the alternative-Adserotype-derived recombinant vectors tested [32]. Subsequent studies, however, revealed that cross-reactive T cell responses between the various serotype-based vectors were still present and significant, suggesting that use of alternative-serotypederived Ad vectors will not substantially mitigate the problems associated with this form of pre-existing anti-Ad immunity [30,32,78,79]. Despite this reality, vectors based on Ad26 and Ad35, had the lowest cross-reactivity with Ad5 vectors and efficiently generated SIV-gag-specific immune responses in Ad5-pre-immune mice and non-human primates, whereas conventional Ad5 vaccines failed to generate substantial SIV-gag specific responses under the same conditions [30,78,79]. Recombinant-Ad26-based vaccines also elicited SIV-gag specific cellular immune responses with increased polyfunctionality (as measured by IFN-g/IL-2/TNF-a producing subpopulations of T cells), breadth and magnitude as well as increased humoral immune responses in Ad5 immune mice, as compared with use of an Ad5-based vaccine [30]. Importantly, utilization of priming vaccinations with an Ad26-based SIV-gag vaccine, followed by boosting immunization with the Ad5-based SIV-gag vaccine significantly improved survival of non-human primates subsequently

challenged with SIV infection, as compared with similar challenge studies utilizing Ad5 SIV-gag vaccine as both the priming and the boosting vaccine [30]. The improved capability of the Ad26/Ad5 combination also correlated with reduced SIV peak and setpoint viral loads, and a reduced rate of decline of the CD4 population in the SIV challenged animals [30]. Ad vectors derived from chimpanzee serotypes C68 and C1 expressing HIV-1 derived gag, pol, gp140 and Nef antigens were designed, produced, and tested in several animal models. AdC6-gag immunizations induced increased frequencies of HIV-gag specific CD8+ INFg + T cells in the Ad5-immune mice, as compared with the conventional Ad5-gag vaccine [33]. AdC68 was also shown to generate robust T cell responses in rhesus macaques even in the context of pre-existing Ad5 immunity, while the Ad5 vaccine was proven ineffective under these conditions [80]. These landmark studies not only confirmed that modification of Ad vectors can improve their efficacy in difficult vaccine applications (such as preventing SIV in SIV-challenge models) but also that these same maneuvers can overcome problems associated with pre-existing Ad5 immunity. 4.

441

445

450

455

460

Genome modified Ad5 vectors 465

Nearly all clinical trials utilizing Ad-based vectors have made use of Ad5-based platforms. This track record for widespread use is important to consider, as regulatory agencies can utilize this accumulated knowledge to assess the safety risks of future trials utilizing similar platforms. Herein lies a very important dilemma. If an investigator chooses to utilize a capsid chimeric Ad vaccine, or a novel Ad vaccine based entirely upon use of an alternative-serotype-based Ad, that investigator must now demonstrate that the altered capsid of the novel vector does not significantly alter the biological, and/or clinical characteristics of the novel vector. Use of novel Ad capsids have been confirmed to have significantly altered biological characteristics in vivo, inclusive of significantly altered biodistribution profiles, significantly different induction of innate and pro-inflammatory immune responses, as well as heightened ability to cause toxicity relative to Ad5-based vector platforms in vivo [32,81-83]. Furthermore, lack of in vitro cross neutralization by Ad5-serotype-specific antibodies may not guarantee efficient evasion of all forms of pre-existing Ad5 immunity, especially in vivo [84]. For example, T-cell-based cross reactivity between distinct Ad serotypes has been confirmed, and may be more responsible for the lack of efficacy of Ad5-based vaccines in the Ad-immune host [35,85-91]. As but one specific example, it has been recently reported that CD8+ T cells against the E2b-encoded polymerase protein were found in Ad-immune subjects at similar frequencies as CD8+ T cells against hexon, the latter is considered a major immuno-dominant Ad protein [92]. Thus strategies that attempt to retain the Ad5 capsid, but modify portions of the Ad genome to minimize expression of Ad5 immuno-dominant

Expert Opin. Biol. Ther. (2009) 9(12)

5

470

475

480

485

490

495

Overcoming pre-existing Adenovirus immunity by genetic engineering of Adenovirus-based vectors

496

500

505

510

515

520

525

530

535

540

545

550

proteins, have recently been described. E1-deleted Ad5 vectors (Ad5 [E1-]) are constructed in a manner such that a transgene replaces only the E1 region of genes; thus, 90% of the wildtype Ad5 genome is generally retained in the vector. In an effort to reduce immuno-dominant viral protein expression from Ad5 vaccines, multiply deleted Ad5 vectors, deleted for genes in addition to the E1 genes have been created [89,93-97]. Specifically, [E1-, E2b-] Ad5 vectors have the E1 region deleted, as well additional deletions in the E2b region, removing the Ad encoded DNA polymerase (pol) and the preterminal (pTP) protein genes [95]. These vectors can be grown to as high as titers as [E1-] Ads, utilizing identical procedures, and have an expanded cloning capacity, theoretically up to a 13 kb gene-carrying capacity as compared with the 8.0 kb capacity of Ad5 [E1-] vectors. A key aspect of the Ad5 [E1-, E2b-] vector is that expression of Ad5 proteins is greatly reduced as compared with [E1-] Ad5 vectors [90,95,98]. Several studies have demonstrated an improved safety of these genome-modified Ads in animal models [93]. We recently confirmed that the minimized ability of [E1-, E2b-] Ad5 vaccines to replicate or express late viral genes may result in evasion of pre-existing Ad5 cellular immunity, increasing the capability for the Ad5 [E1-, E2b-] vectors to induce beneficial immune responses in Ad5-immune animal models [89,94-97,99]. This confirmation included blunted induction of Ad-specific T cell proliferative immune responses by the [E1-, E2b-] Ad5 vaccines in the Ad5-immune hosts, results that were simultaneous with improved NK cell and antigen-specific CD8+ T cell responses to [E1-, E2b-] Ad5-expressed antigens, inclusive of HIV-gag and the tumor antigen CEA [94,97]. Upon immunization of Ad5-immune mice as well cynamalogous macaques made hyper-immune to Ad5, [E1-, E2b-] Ad5 vaccines induced HIV-gag-specific T cells producing IL-2, which can result in augmented CD4+ clonal expansion [97], and production of HIV-gag specific CD8+, IFN-g-secreting T cells. In a similar vain, ‘helper-virus-dependent’ Ads (HDAds) completely lack virus-encoded sequences, except for the viral ITRs, and allow for cloning capacities approaching 36 kb. These vectors can be grown to very high titers, and only require a helper virus in addition to specific packaging cell lines to allow for their large scale production. The absence of any Ad-encoded genes in HDAds has convincingly proven that many deleterious adaptive immune responses are directed to Ad genes [100]. In a recent study more relevant to this review, HDAds were also found to be capable of generating antigen-specific immune responses in Ad5-immune hosts [101]. Specifically, use of an HIV gp140 env-expressing HDAd5 yielded increased env-specific cellular and humoral immune responses in Ad5-pre-immune mice, and increased env specific antibody titers in Ad5 pre-immune macaques over fivefold, as compared with a traditional Ad vaccine [101]. These early studies suggests that Ad5-based vaccines can be improved upon to improve their efficacy in the face of pre-existing anti-Ad 6

immunity without requiring major alterations to their capsid 551 coat. Such vectors when used clinically will benefit from the long-term safety record of traditional Ad5-based vaccines, while potentially providing for improved efficacy in the 555 Ad5-immune host in general. 5.

Expert opinion and overall summary

The use of Ad5 vectors as a platform for vaccine development has been gaining steady momentum. Ad5-based vaccines have been repeatedly shown to outperform other vaccine platforms expressing identical antigens, including other Ad serotype vaccines [32-34], poxvirus based vectors [34,36], and DNA-based vaccines [35,36]. Additionally, the large number of patients safely treated with the Ad5 platform confirms its high likelihood of acceptance by regulatory bodies, relative to less well tested platforms, an important point when considering the considerable risks and costs involved in developing new therapeutics. Unfortunately, despite this tremendous track record for safe and potent induction of immune responses in vaccine applications, conventional, E1-deleted Ad5-based vaccines have several confirmed limitations. These include a sometimes less than optimal induction of antigen-specific adaptive immune responses, made worse in the context of pre-existing Ad immunity. These facts indicate a need for development of more potent Ad-based vaccines, vaccines that are efficacious even in the context of pre-existing Ad5 immunity. In this review, we have summarized how many groups have attempted to address the need to improve the efficacy of Ad-based vaccines in general, and/or avoid the problem of pre-existing Ad5 immunity specifically, as summarized in Figure 1. Some groups have proposed the utilization of alternative-human-Ad-serotype-based vaccines (some derived from non-human primates) as a relatively simple method to avoid pre-existing Ad5 immunity [30,82,102,103]. Use of these options has facilitated induction of improved T cell responses to Ad-expressed HIV antigens despite Ad5 immunity. These landmark studies also prove that improved induction of HIV-antigen-specific T cell responses can result in superior performance in very stringent SIV challenge studies in nonhuman primates, a feat that cannot be achieved with other vaccines, nor conventional [E1-] Ad5 vaccines [30,103-105]. Despite these achievements, use of alternative-Adserotype-based vaccines will also face neutralizing antibody induction after a single usage, and as we have detailed in the text, will probably be affected upon by crossreactive immune responses present in the Ad5-immune host [39,42-44,104]. More importantly, however, use of alternative-serotype-based Ad vaccines also poses several new serious problems including: i) lack of previous usage in humans; ii) significantly altered biodistribution profiles; iii) lack of induction of potent immune responses relative to Ad5 vaccines; and iv) induction of more deleterious inflammatory immune responses relative to Ad5, in some cases causing excessive morbidity and mortality in animal

Expert Opin. Biol. Ther. (2009) 9(12)

560

565

570

575

580

585

590

595

600

605

Seregin & Amalfitano

“HVR5 Hexon” displaying Ad5

Ad5 WT ITR E1 E2b

Alternative human Ad serotype

ITR

1

5

3 “Protein IX” displaying Ad

Alternative non-human Ad serotype

[E1-] Ad

6

3

4

“Penton” displaying Ad

-

-

[E1 , E2b ] Ad

Ad5/3 Fiber chimera

6 “Fiber” displaying Ad:

E1- region of Ad genome E2b- region of Ad genome Ad genome sequences Vector-encoded transgene Inverted terminal repeats

“Fully deleted” HDAd

Ad5/26 HVR5/7 chimera

2 “PEGylated” Ad vectors

Figure 1. Current approaches to design advanced Adenovirus based vaccines. Group 1. Starting with a wild-type Ad5 virus the E1 region is deleted to generate [E1-] Ad5 vectors; Group 2. Adenovirus vectors of any group can be PEGylated to improve evasion of pre-existing Ad immunity, see Section 2.1 for full descriptions; Group 3. Capsid ‘display’ of antigens at various locations on the Ad capsid where indicated, see Section 2.2.1 for full descriptions; Group 4. Chimeric Adenovirus vectors, that contain capsid proteins derived from two different Ad serotypes, see Section 2.2.2 for full descriptions; Group 5. Adenovirus vectors derived completely from alternative serotypes, see Section 3 for full descriptions; Group 6. Genome-modified Adenovirus vectors, that retain the parental serotype capsid, but are deleted for portions of the parental genome, to improve evasion of pre-existing cellular immunity, see Section 4 for full descriptions.

606

610

615

619

models [32,81-83]. All of these caveats may prevent/hinder regulatory approval of alternative-serotype-based Ad vaccines for widespread deployment. With this in mind, we have also reviewed efforts to improve the Ad5 platform in a fashion that retains major portions of the Ad5 capsid. We have reviewed how subtle modifications (such as amino acid substitution of immuno-dominant peptides present in the Ad5 capsid, or swapping of the HVR region of the Ad5 hexon) can improve the efficacy of these modified, Ad5-based vaccines, in the Ad5-naı¨ve or Ad5-immune host. Not so subtle modifications include complete incorporation of Ad5 hexon or fiber proteins derived from alternative-serotype Ads into the Ad5 capsid. While these novel vaccine platforms also improve performance in

the context of the Ad5-immune host, they also may face the same regulatory issues facing vaccines constructed entirely from alternative Ad serotypes. Strategies that do not alter the Ad5 capsid (such as use of [E1-, E2b-] Ads or HDAds) may avoid many of the regulatory (safety) issues associated with proposed use of alternativeserotype Ad-based vaccines. These strategies may also provide an improvement that allows for efficacy in the most difficult of applications, such as for use as a HIV prophylactic vaccine in the Ad-immune host. However, these modifications may not overcome certain aspects of pre-existing Ad5 immunity as well. As a result of these considerations, it is clear that each of the modified Ad vaccines described in this manuscript may be better utilized for some clinical applications than for

Expert Opin. Biol. Ther. (2009) 9(12)

7

620

625

630

633

Overcoming pre-existing Adenovirus immunity by genetic engineering of Adenovirus-based vectors

634 635

640

645

650 651

others. Despite this context-specific utility, one should also be impressed that the Ad vaccine platform appears to be highly ‘plastic’ and capable of tolerating a number of elegant molecular manipulations. In many instances, these modifications not only preserve important benefits inherent to the use of the traditional Ad5-based vaccine platform, but also provide for improved efficacy, even in the context of pre-existing Ad5 immunity. Future studies will expand upon these findings. Incorporation of some of the modifications summarized herein will probably improve the capabilities of this important vaccine platform for expanded use in a number of additional human and agricultural applications not targeted to date. One can also envision combining some of the approaches described in this review. For example, Ad5 vaccine platforms could be theoretically generated that are deleted for the E1 and E2b genes, as well having portions of the hexon HVR5 and HVR7 regions replaced with similar regions derived from Ad26.

PEGylation of such a novel vaccine could also then be con- 652 templated. Similarly, these multiple manoeuvres can be theoretically applied to any Ad serotype. However, theoretical contemplation of these multiply modified vaccines is not a 655 guarantee of reduction to practice, as nuances regarding compatibility of multiple manipulations to allow for viability and large-scale production of the resultant vaccine must be respected. We leave our readers with the view that despite these pragmatic considerations, the continued improvement 660 of Ad-based vectors is likely to yield several important vaccine platforms that will have high utility in a number of clinical and agricultural applications, even in the context of pre-existing Ad immunity. 665

Declaration of interest The authors state no conflict of interest and have received no 669 payment for preparation of this manuscript.

Bibliography 1.

Buchbinder SP, Mehrotra DV, Duerr A, 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(9653):1881-93

2.

Zak DE, Aderem A. Systems biology of innate immunity. Immunol Rev 2009;227(1):264-82

3.

4.

5.

Hacein-Bey-Abina S, Von Kalle C, Schmidt M, et al. LMO2-associated clonal T cell proliferation in two patients after gene therapy for SCID-X1. Science 2003;302(5644):415-9

Cohen J. AIDS research. Did Merck’s failed HIV vaccine cause harm? Science 2007;318(5853):1048-9

Seggewiss R, Dunbar CE. A new direction for gene therapy: intrathymic T cell-specific lentiviral gene transfer. J Clin Invest 2005;115(8):2064-7

11.

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(12):2717-25

Puck JM, Malech HL. Gene therapy for immune disorders: good news tempered by bad news. J Allergy Clin Immunol 2006;117(4):865-9

12.

Donsante A, Miller DG, Li Y, et al. AAV vector integration sites in mouse hepatocellular carcinoma. Science 2007;317(5837):477

Sekaly RP. The failed HIV Merck vaccine study: a step back or a launching point for future vaccine development? J Exp Med 2008;205(1):7-12 Hutnick NA, Carnathan DG, Dubey SA, et al. Baseline Ad5 serostatus does not predict Ad5 HIV vaccine-induced expansion of adenovirus-specific CD4+ T cells. Nat Med 2009;15(8):876-8

7.

O’Brien KL, Liu J, King SL, et al. Adenovirus-specific immunity after immunization with an Ad5 HIV-1 vaccine candidate in humans. Nat Med 2009;15(8):873-5

8

9.

10.

6.

8.

good news for an HIV-1 vaccine? Nat Med 2009;15(8):866-70

Stamatatos L, Morris L, Burton DR, Mascola JR. Neutralizing antibodies generated during natural HIV-1 infection:

13.

Dave UP, Akagi K, Tripathi R, et al. Murine leukemias with retroviral insertions at Lmo2 are predictive of the leukemias induced in SCID-X1 patients following retroviral gene therapy. PLoS Genet 2009;5(5):e1000491. Published online 22 May 2009, doi:10.1371/journal.pgen.1000491

14.

Harui A, Suzuki S, Kochanek S, Mitani K. Frequency and stability of chromosomal integration of adenovirus vectors. J Virol 1999;73(7):6141-6

15.

Schoggins JW, Nociari M, Philpott N, Falck-Pedersen E. Influence of fiber detargeting on adenovirus-mediated innate and adaptive immune activation. J Virol 2005;79(18):11627-37

Expert Opin. Biol. Ther. (2009) 9(12)

16.

Croyle MA, Le HT, Linse KD, et al. PEGylated helper-dependent adenoviral vectors: highly efficient vectors with an enhanced safety profile. Gene Ther 2005;12(7):579-87

17.

Weaver EA, Barry MA. Effects of shielding adenoviral vectors with polyethylene glycol (PEG) on Vector-specific and vaccine-mediated immune responses. Hum Gene Ther 2008;19(12):1369-82

18.

Lin SW, Cun AS, Harris-McCoy K, Ertl HC. Intramuscular rather than oral administration of replication-defective adenoviral vaccine vector induces specific CD8+ T cell responses in the gut. Vaccine 2007;25(12):2187-93

19.

McConnell MJ, Danthinne X, Imperiale MJ. Characterization of a permissive epitope insertion site in adenovirus hexon. J Virol 2006;80(11):5361-70

20.

Abe S, Okuda K, Ura T, et al. Adenovirus type 5 with modified hexons induces robust transgene-specific immune responses in mice with pre-existing immunity against adenovirus type 5. J Gene Med 2009;11(7):570-9

21.

Worgall S, Krause A, Rivara M, et al. Protection against P. aeruginosa with an adenovirus vector containing an OprF epitope in the capsid. J Clin Invest 2005;115(5):1281-9

22.

Sarkioja M, Pesonen S, Raki M, et al. Changing the adenovirus fiber for retaining gene delivery efficacy in the presence of

Seregin & Amalfitano

neutralizing antibodies. Gene Ther 2008;15(12):921-9 23.

24.

25.

26.

Krause A, Joh JH, Hackett NR, et al. Epitopes expressed in different adenovirus capsid proteins induce different levels of epitope-specific immunity. J Virol 2006;80(11):5523-30

34.

Reyes-Sandoval A, Sridhar S, Berthoud T, et al. Single-dose immunogenicity and protective efficacy of simian adenoviral vectors against Plasmodium berghei. Eur J Immunol 2008;38(3):732-41

35.

Schirmbeck R, Reimann J, Kochanek S, Kreppel F. The immunogenicity of adenovirus vectors limits the multispecificity of CD8 T-cell responses to vector-encoded transgenic antigens. Mol Ther 2008;16(9):1609-16

Yang TC, Dayball K, Wan YH, Bramson J. Detailed analysis of the CD8+ T-cell response following adenovirus vaccination. J Virol 2003;77(24):13407-11

46.

O’Riordan CR, Lachapelle A, Delgado C, et al. PEGylation of adenovirus with retention of infectivity and protection from neutralizing antibody in vitro and in vivo. Hum Gene Ther 1999;10(8):1349-58

47.

Matthews C, Jenkins G, Hilfinger J, Davidson B. Poly-L-lysine improves gene transfer with adenovirus formulated in PLGA microspheres. Gene Ther 1999;6(9):1558-64

48.

Lee SG, Yoon SJ, Kim CD, et al. Enhancement of adenoviral transduction with polycationic liposomes in vivo. Cancer Gene Ther 2000;7(10):1329-35

Hackett NR, Kaminsky SM, Sondhi D, Crystal RG. Antivector and antitransgene host responses in gene therapy. Curr Opin Mol Ther 2000;2(4):376-82

36.

Boyer JL, Kobinger G, Wilson JM, Crystal RG. Adenovirus-based genetic vaccines for biodefense. Hum Gene Ther 2005;16(2):157-68

37.

Lasaro MO, Ertl HC. New insights on adenovirus as vaccine vectors. Mol Ther 2009;17(8):1333-9

49.

38.

Cohen P. Immunity’s yin and yang. A successful vaccine must first avoid being eliminated by pre-existing immunity before it can promote a protective immune response. IAVI Rep 2006;10(1):1-5

Croyle MA, Chirmule N, Zhang Y, Wilson JM. PEGylation of E1-deleted adenovirus vectors allows significant gene expression on readministration to liver. Hum Gene Ther 2002;13(15):1887-900

50.

39.

Molinier-Frenkel V, Gahery-Segard H, Mehtali M, et al. Immune response to recombinant adenovirus in humans: capsid components from viral input are targets for vector-specific cytotoxic T lymphocytes. J Virol 2000;74(16):7678-82

Wortmann A, Vohringer S, Engler T, et al. Fully detargeted polyethylene glycol-coated adenovirus vectors are potent genetic vaccines and escape from pre-existing anti-adenovirus antibodies. Mol Ther 2008;16(1):154-62

51.

40.

Molinier-Frenkel V, Lengagne R, Gaden F, et al. Adenovirus hexon protein is a potent adjuvant for activation of a cellular immune response. J Virol 2002;76(1):127-35

Ogawara K, Rots MG, Kok RJ, et al. A novel strategy to modify adenovirus tropism and enhance transgene delivery to activated vascular endothelial cells in vitro and in vivo. Hum Gene Ther 2004;15(5):433-43

41.

Sumida SM, Truitt DM, Kishko MG, et al. Neutralizing antibodies and CD8+ T lymphocytes both contribute to immunity to adenovirus serotype 5 vaccine vectors. J Virol 2004;78(6):2666-73

52.

Parks RJ. Adenovirus protein IX: a new look at an old protein. Mol Ther 2005;11(1):19-25

53.

Rux JJ, Burnett RM. Adenovirus structure. Hum Gene Ther 2004;15(12):1167-76

42.

Flomenberg P, Piaskowski V, Truitt RL, Casper JT. Characterization of human proliferative T cell responses to adenovirus. J Infect Dis 1995;171(5):1090-6

54.

Vellinga J, Van der Heijdt S, Hoeben RC. The adenovirus capsid: major progress in minor proteins. J Gen Virol 2005;86(Pt 6):1581-8

43.

Tang J, Olive M, Champagne K, et al. Adenovirus hexon T-cell epitope is recognized by most adults and is restricted by HLA DP4, the most common class II allele. Gene Ther 2004;11(18):1408-15

55.

Einfeld DA, Brough DE, Roelvink PW, et al. Construction of a pseudoreceptor that mediates transduction by adenoviruses expressing a ligand in fiber or penton base. J Virol 1999;73(11):9130-6

44.

Tang J, Olive M, Pulmanausahakul R, et al. Human CD8+ cytotoxic T cell responses to adenovirus capsid proteins. Virology 2006;350(2):312-22

56.

45.

Hashimoto M, Boyer JL, Hackett NR, et al. Induction of protective immunity to anthrax lethal toxin with a nonhuman

Wickham TJ, Segal DM, Roelvink PW, et al. Targeted adenovirus gene transfer to endothelial and smooth muscle cells by using bispecific antibodies. J Virol 1996;70(10):6831-8

57.

McConnell MJ, Hanna PC, Imperiale MJ. Cytokine response and survival of mice

27.

Barouch DH, Nabel GJ. Adenovirus vector-based vaccines for human immunodeficiency virus type 1. Hum Gene Ther 2005;16(2):149-56

28.

Sullivan NJ, Sanchez A, Rollin PE, et al. Development of a preventive vaccine for Ebola virus infection in primates. Nature 2000;408(6812):605-9

29.

Millar J, Dissanayake D, Yang TC, et al. The magnitude of the CD8+ T cell response produced by recombinant virus vectors is a function of both the antigen and the vector. Cell Immunol 2007;250(1-2):55-67

30.

Liu J, O’Brien KL, Lynch DM, et al. Immune control of an SIV challenge by a T-cell-based vaccine in rhesus monkeys. Nature 2009;457(7225):87-91

31.

Mutwiri G, Bateman C, Baca-Estrada ME, et al. Induction of immune responses in newborn lambs following enteric immunization with a human adenovirus vaccine vector. Vaccine 2000;19(9-10):1284-93

32.

Abbink P, Lemckert AA, Ewald BA, et al. Comparative seroprevalence and immunogenicity of six rare serotype recombinant adenovirus vaccine vectors from subgroups B and D. J Virol 2007;81(9):4654-63

33.

Pinto AR, Fitzgerald JC, Giles-Davis W, et al. Induction of CD8+ T cells to an HIV-1 antigen through a prime boost regimen with heterologous E1-deleted adenoviral vaccine carriers. J Immunol 2003;171(12):6774-9

Shiver JW, Fu TM, Chen L, et al. Replication-incompetent adenoviral vaccine vector elicits effective anti-immunodeficiency-virus immunity. Nature 2002;415(6869):331-5

primate adenovirus-based vaccine in the presence of preexisting anti-human adenovirus immunity. Infect Immun 2005;73(10):6885-91

Expert Opin. Biol. Ther. (2009) 9(12)

9

Overcoming pre-existing Adenovirus immunity by genetic engineering of Adenovirus-based vectors

immunized with an adenovirus expressing Bacillus anthracis protective antigen domain 4. Infect Immun 2006;74(2):1009-15 58.

59.

60.

61.

Belousova N, Krendelchtchikova V, Curiel DT, Krasnykh V. Modulation of adenovirus vector tropism via incorporation of polypeptide ligands into the fiber protein. J Virol 2002;76(17):8621-31 Matthews QL, Sibley DA, Wu H, et al. Genetic incorporation of a herpes simplex virus type 1 thymidine kinase and firefly luciferase fusion into the adenovirus protein IX for functional display on the virion. Mol Imaging 2006;5(4):510-9 Worgall S, Busch A, Rivara M, et al. Modification to the capsid of the adenovirus vector that enhances dendritic cell infection and transgene-specific cellular immune responses. J Virol 2004;78(5):2572-80 Kurachi S, Koizumi N, Sakurai F, et al. Characterization of capsid-modified adenovirus vectors containing heterologous peptides in the fiber knob, protein IX, or hexon. Gene Ther 2007;14(3):266-74

62.

Worgall S, Krause A, Qiu J, et al. Protective immunity to Pseudomonas aeruginosa induced with a capsid-modified adenovirus expressing P. aeruginosa OprF. J Virol 2007;81(24):13801-8

63.

Bailey A, Mautner V. Phylogenetic relationships among adenovirus serotypes. Virology 1994;205(2):438-52

64.

Russell WC. Update on adenovirus and its vectors. J Gen Virol 2000;81(Pt 11):2573-604

65.

66.

Gall J, Kass-Eisler A, Leinwand L, Falck-Pedersen E. Adenovirus type 5 and 7 capsid chimera: fiber replacement alters receptor tropism without affecting primary immune neutralization epitopes. J Virol 1996;70(4):2116-23 Gall JG, Crystal RG, Falck-Pedersen E. Construction and characterization of hexon-chimeric adenoviruses: specification of adenovirus serotype. J Virol 1998;72(12):10260-4

67.

Roy S, Shirley PS, McClelland A, Kaleko M. Circumvention of immunity to the adenovirus major coat protein hexon. J Virol 1998;72(8):6875-9

68.

Wu H, Dmitriev I, Kashentseva E, et al. Construction and characterization of adenovirus serotype 5 packaged by

10

primates to vaccine regimens based on human- or chimpanzee-derived adenovirus vectors. J Virol 2007;81(12):6594-604

serotype 3 hexon. J Virol 2002;76(24):12775-82 69.

Myhre S, Henning P, Granio O, et al. Decreased immune reactivity towards a knobless, affibody-targeted adenovirus type 5 vector. Gene Ther 2007;14(4):376-81

70.

Wu H, Curiel DT. Fiber-modified adenoviruses for targeted gene therapy. Methods Mol Biol 2008;434:113-32

71.

Noureddini SC, Curiel DT. Genetic targeting strategies for adenovirus. Mol Pharm 2005;2(5):341-7

72.

Nakayama M, Both GW, Banizs B, et al. An adenovirus serotype 5 vector with fibers derived from ovine atadenovirus demonstrates CAR-independent tropism and unique biodistribution in mice. Virology 2006;350(1):103-15

73.

Roy S, Clawson DS, Calcedo R, et al. Use of chimeric adenoviral vectors to assess capsid neutralization determinants. Virology 2005;333(2):207-14

74.

Ritter T, Lehmann M, Volk HD. Improvements in gene therapy: averting the immune response to adenoviral vectors. BioDrugs 2002;16(1):3-10

75.

Pichla-Gollon SL, Drinker M, Zhou X, et al. Structure-based identification of a major neutralizing site in an adenovirus hexon. J Virol 2007;81(4):1680-9

76.

Youil R, Toner TJ, Su Q, et al. Hexon gene switch strategy for the generation of chimeric recombinant adenovirus. Hum Gene Ther 2002;13(2):311-20

77.

Roberts DM, Nanda A, Havenga MJ, et al. Hexon-chimaeric adenovirus serotype 5 vectors circumvent pre-existing anti-vector immunity. Nature 2006;441(7090):239-43

78.

79.

80.

Barouch DH, Pau MG, Custers JH, et al. Immunogenicity of recombinant adenovirus serotype 35 vaccine in the presence of pre-existing anti-Ad5 immunity. J Immunol 2004;172(10):6290-7 Liu J, Ewald BA, Lynch DM, et al. Magnitude and phenotype of cellular immune responses elicited by recombinant adenovirus vectors and heterologous prime-boost regimens in rhesus monkeys. J Virol 2008;82(10):4844-52 McCoy K, Tatsis N, Korioth-Schmitz B, et al. Effect of preexisting immunity to adenovirus human serotype 5 antigens on the immune responses of nonhuman

Expert Opin. Biol. Ther. (2009) 9(12)

81.

Hartman ZC, Appledorn DM, Serra D, et al. Replication-attenuated human adenoviral type 4 vectors elicit capsid dependent enhanced innate immune responses that are partially dependent upon interactions with the complement system. Virology 2008;374(2):453-67

82.

Hensley SE, Cun AS, Giles-Davis W, et al. Type I interferon inhibits antibody responses induced by a chimpanzee adenovirus vector. Mol Ther 2007;15(2):393-403

83.

Appledorn DM, Kiang A, McBride A, et al. Wild-type adenoviruses from groups A-F evoke unique innate immune responses, of which HAd3 and SAd23 are partially complement dependent. Gene Ther 2008;15(12):885-901

84.

Pichla-Gollon SL, Lin SW, Hensley SE, et al. Effect of preexisting immunity on an adenovirus vaccine vector: in vitro neutralization assays fail to predict inhibition by antiviral antibody in vivo. J Virol 2009;83(11):5567-73

85.

Smith CA, Woodruff LS, Rooney C, Kitchingman GR. Extensive cross-reactivity of adenovirus-specific cytotoxic T cells. Hum Gene Ther 1998;9(10):1419-27

86.

Jonuleit H, Tuting T, Steitz J, et al. Efficient transduction of mature CD83+ dendritic cells using recombinant adenovirus suppressed T cell stimulatory capacity. Gene Ther 2000;7(3):249-54

87.

Tuettenberg A, Jonuleit H, Tuting T, et al. Early adenoviral gene expression mediates immunosuppression by transduced dendritic cell (DC): implications for immunotherapy using genetically modified DC. J Immunol 2004;172(3):1524-30

88.

Hu H, Serra D, Amalfitano A. Persistence of an [E1-, polymerase-] adenovirus vector despite transduction of a neoantigen into immune-competent mice. Hum Gene Ther 1999;10(3):355-64

89.

Everett RS, Hodges BL, Ding EY, et al. Liver toxicities typically induced by first-generation adenoviral vectors can be reduced by use of E1, E2b-deleted adenoviral vectors. Hum Gene Ther 2003;14(18):1715-26

90.

Hodges BL, Serra D, Hu H, et al. Multiply deleted [E1, polymerase-, and pTP-]

Seregin & Amalfitano

97.

Schaack J. Induction and inhibition of innate inflammatory responses by adenovirus early region proteins. Viral Immunol 2005;18(1):79-88

Gabitzsch ES, Xu Y, Yoshida LH, et al. A preliminary and comparative evaluation of a novel Ad5 [E1-, E2b-] recombinant-based vaccine used to induce cell mediated immune responses. Immunol Lett 2009;122(1):44-51

98.

92.

Joshi A, Tang J, Kuzma M, et al. Adenovirus DNA polymerase is recognized by human CD8+ T cells. J Gen Virol 2009;90(Pt 1):84-94

Barjot C, Hartigan-O’Connor D, Salvatori G, et al. Gutted adenoviral vector growth using E1/E2b/E3-deleted helper viruses. J Gene Med 2002;4(5):480-9

99.

93.

Amalfitano A, Parks RJ. Separating fact from fiction: assessing the potential of modified adenovirus vectors for use in human gene therapy. Curr Gene Ther 2002;2(2):111-33

94.

Osada T, Yang XY, Hartman ZC, et al. Optimization of vaccine responses with an E1, E2b and E3-deleted Ad5 vector circumvents pre-existing anti-vector immunity. Cancer Gene Ther 2009

Gabitzsch ES, Xu Y, Yoshida LH, et al. Novel Adenovirus type 5 vaccine platform induces cellular immunity against HIV-1 Gag, Pol, Nef despite the presence of Ad5 immunity. Vaccine 2009, published online 24 June 2009, doi:10.1016/j.vaccine.2009.06.028

adenovirus vector persists despite deletion of the preterminal protein. J Gene Med 2000;2(4):250-9 91.

95.

Amalfitano A, Hauser MA, Hu H, et al. Production and characterization of improved adenovirus vectors with the E1, E2b, and E3 genes deleted. J Virol 1998;72(2):926-33

96.

Everett RS, Evans HK, Hodges BL, et al. Strain-specific rate of shutdown of CMV enhancer activity in murine liver confirmed by use of persistent [E1-, E2b-] adenoviral vectors. Virology 2004;325(1):96-105

100.

Palmer DJ, Ng P. Helper-dependent adenoviral vectors for gene therapy. Hum Gene Ther 2005;16(1):1-16

101.

Weaver EA, Nehete PN, Buchl SS, et al. Comparison of replication-competent, first generation, and helper-dependent adenoviral vaccines. PLoS One 2009;4(3): e5059. Published online 31 March 2009, doi:10.1371/journal.pone.0005059

102.

Farina SF, Gao GP, Xiang ZQ, et al. Replication-defective vector based on a chimpanzee adenovirus. J Virol 2001;75(23):11603-13

103.

Lemckert AA, Sumida SM, Holterman L, et al. Immunogenicity of heterologous

Expert Opin. Biol. Ther. (2009) 9(12)

prime-boost regimens involving recombinant adenovirus serotype 11 (Ad11) and Ad35 vaccine vectors in the presence of anti-ad5 immunity. J Virol 2005;79(15):9694-701 104.

Thorner AR, Lemckert AA, Goudsmit J, et al. Immunogenicity of heterologous recombinant adenovirus prime-boost vaccine regimens is enhanced by circumventing vector cross-reactivity. J Virol 2006;80(24):12009-16

105.

Casimiro DR, Wang F, Schleif WA, et al. Attenuation of simian immunodeficiency virus SIVmac239 infection by prophylactic immunization with dna and recombinant adenoviral vaccine vectors expressing Gag. J Virol 2005;79(24):15547-55

Affiliation Sergey S Seregin1 & Andrea Amalfitano†1,2 † Author for correspondence 1 Michigan State University, Department of Microbiology and Molecular Genetics, 4194 Biomedical and Physical Sciences Bldg, East Lansing, MI 48823, USA Tel: +1 517 884 5324; Fax: +1 517 353 8957; E-mail: [email protected] 2 Michigan State University, Department of Pediatrics, East Lansing, MI 48824, USA

11