Alongside anti-Ad immunity generated via natural Ad infection, anti-Ad immunity is also generated by the current use of Ad vectors in different therapeutic applications, such as recent mass vaccinations against SARS-CoV-2 using adenoviral platforms. Due to scale of production and high replicative ability, the use of these vectors is on the rise.
5.1. Widespread Ad Vector Development, Clinical Evaluation and Use
As a potential vector for cancer and gene therapies, Ads, especially HAdV-C5, have been well investigated. The immunogenic cell death of tumour cells induced by Ad based OVs and release of DAMPs attracts the immune system to the tumour. The ability of the OV to replicate will allow self-amplification within permissive transformed cells over time without further administration. Genetic changes can be introduced into prevent Ad replication in off-target tissues or prevent replication altogether. The deletion of large parts of the genome, which usually completely prevents replication, allows for the insertion of large transgenes which can serve different purposes [
124,
125].
A very common example of creating conditionally replicating Ads is deletions in the early phase gene region 1 (E1). Whilst deletion of the entire E1 region prevents replication entirely [
126], the deletion or loss of function of its genes E1B or E1A separately may be utilised in the context of OV cancer treatment [
124,
127]. In healthy cells, E1 gene product will repress host genes involved in preventing viral replication, such as p53 and pRB. Both of these are very commonly mutated in cancers, allowing E1-deleted viral platforms to replicate conditionally in cells deficient in these pathways [
124]. Additionally, replication as well as transgene expression can be placed under control of a tumour specific promoter [
128], whilst structural changes to capsid proteins can be incorporated to retarget Ads from their natural tropism [
8,
128], making the vector even more tumour selective.
Transgenes often incorporated into oncolytic Ad vectors include immunomodulatory genes to increase the patients anti-tumour immune response, or prodrug converting enzymes to reduce systemic toxicity of traditional chemotherapy drugs [
128]. There are a number of oncolytic Ads currently being investigated in clinical trials, most of which are based on HAdV-C5, some of which are summarised in
Table 1.
Where long term gene augmentation is required, such as with gene therapy for monogenic disorders, the use of helper dependent or “gutless” Ad vectors, which are devoid of all viral DNA (except for the inverted terminal repeats, ITRs) and require a helper virus for production, has been extensively explored [
141]. However, low level contamination of helper Ads presents a problem, leading to the development of better purification methods [
142,
143] and more recently, helper-free gutless Ads (HF-GLAds) [
144]. These HF-GLAds have shown promise in pre-clinical testing, but are yet to reach -human trials [
145].
Over the past decade, gene therapy applications have moved significantly towards using Adenovirus-Associated Virus (AAV) vectors instead of helper dependent Ads. AAVs have a smaller transgene capacity, but allow long term gene expression. They also tend to be less immunogenic than Ad vectors, which is desirable for long term gene correction [
146,
147], though HF-GLAds and replication deficient vectors also generate reduced anti-vector immune response compared to oncolytic or replication competent Ads [
145]. Some gene therapy applications of Ad vectors in clinical trials currently and in the past are listed in
Table 2.
As vaccine vectors, Ads are generally used in a replication deficient state. The historic vaccines against Ad, which worked extremely well, were live virus vaccines to protect military recruits against ARD caused by AdV-E4 and AdV-B7 [
66]. For vaccination for any other purpose than Ad immunity however, production of viral proteins is a disadvantage, as the aim is for the immune response to be directed against the antigen transgene.
An interesting middle ground has been proposed by Crosby and Barry in their development of “single-cycle” (SC) Ads. Their vaccine vector, based on HAdV-C6, has a deletion in protein IIIa which is essential for DNA packaging and virus assembly. The resulting vector, once it inside the cell, is able to replicate its genetic material and therefore its transgene, without producing viable progenitor virus. Some virus protein was still produced by infected cells [
156] still engages the immune system, however the transgene/vector protein ratio is still more favourable. In a later
in vivo experiment with Syrian hamsters to investigate the vector for influenza vaccinations, the single cycle replication vector required a 33x lower dosage compared to replication deficient vectors to achieve the same transgene levels, which is a drastically lower amount of vector protein patients would be exposed to [
157]. Furthermore, the added Ad antigen may act as an adjuvant to recruit the immune system.
The value of Ad-based vaccines was demonstrated during the COVID-19 pandemic. According to ourworldindata.org, an estimated 5.13 billion people worldwide were vaccinated against SARS-CoV-2. In the UK alone, 50.76 million primary doses were administered to >75% of the population. Within the European Union almost 100% of the vaccinated population was covered by 4 vaccine providers: Pfizer/BioNTech (Comirnaty), Moderna (Spikevax), Oxford/AstraZeneca (Covishield) and Janssen (Jcovden) in order of most administered [
158] (
Figure 5).
Both Pfizer (Comirnaty) and Moderna (Spikevax) are mRNA vaccines. The Janssen (Jcovden) vaccine is a HAdV-D26 vector based vaccine expressing the SARS-CoV-2 spike antigens [
159]. Similarly, the Oxford/AstraZeneca vaccine (Covishield) is based on their ChAdOx1 vector derived from ChAd-Y25, also with SARS-CoV-2 spike antigen transgene [
42,
160]. This means, that in the EU alone, over 85 million people have been immunised with Ad-based replication deficient vaccine vectors [
158].
Although the SARS-CoV-2 vaccines were developed for intramuscular administration, different routes of administration have since been investigated for SARS-CoV-2 [
161,
162], in addition to other pathogens, such as influenza [
163] and HIV [
164]. Current uses of and clinical trials into Ad based vaccines have been summarised in
Table 3.
As broad usage of Ad based vectors becomes increasingly common, we need a solid understanding of administered vectors with patient immune system to tailor the vector to be most effective in different applications.
5.2. Innate Immunity
Overall, due to the limited receptors available, we would expect sensing by the innate immune system to be very similar between natural Ad infection and Ad vector administration, though it is possible that there are tissue specific differences due to the differently localised or more systemic exposure to vectors, compared to natural infection.
In wild type Ads, the E3 protein enhances Ad immune evasion through several mechanisms, such prevention of intrinsic and extrinsic induction of apoptosis of virus infected cells, and the downregulation of MHC-I molecules on the cell surface [
176]. While this leads to decreased engagement of cytotoxic T cells, it also means increased susceptibility to killing by NK cells due to the missing-self hypothesis [
177]. Intranasal administration of HAdV-C2 and HAdV-C5 vectors with a complete E3 deletion to cotton rats led to an increased macrophage and polymorphonuclear leukocyte response. The same could be observed by only deleting the E3-19kDa region of the gene, making this the most important region in this response [
178]. The E3 region is commonly deleted in may Ad vector applications, either fully or partially, for various reasons. Large deletions of the whole E3 gene region increase transgene capacity, while deletions of the E3-19kDa gene specifically can be used to actively increase antigen exposure, both of viral and tumour antigens in oncolytic vectors [
124]. Examples of whole deletions include the oncolytic HAdV-C5 based Onyx-015 [
179] or the very similar Oncorine, which is approved for oncolytic virotherapy in China [
180]. Partial deletions have been made in several pre-clinical Ad vectors [
8,
181]. Though immune modulation may not always be the reason for an E3 deletion in vector development, it is important to take into consideration.
Fejer
et al. showed that, in intraperitoneal injections in mice, virus entry and endosomal escape trigger IFN expression
in vivo in myeloid DCs, in a TLR independent manner and heavily reliant on the positive feedback loop of IFN secretion described earlier (
Figure 3). They highlight the importance of splenic myeloid DCs in this interaction, as this is where the vast majority of IFNs were produced [
182]. Vectors used in this study were based on HAdV-B3, HAdV-C2 and HAdV-C5 with no modifications to structural proteins. It is likely that the interaction of the Ad penton base with integrin of the spleen is responsible for this strong response. Secretion if type I IFNs has been shown to induce the production of other anti-viral proteins
in vitro, which inhibit viral DNA-, RNA and protein synthesis [
183]. In a vector context, this means reduced expression of transgenes through the induction on type I IFNs by the innate immune system. Di Paolo
et al. also investigated accumulation of their HAdV-C5 based vector in the spleen after intravenous injection of mice. They found that uptake by tissue resident macrophages through integrin binding triggers the transcription of Interleukin (IL) 1α within 10 minutes of administration, with high levels of expression induced by 25 minutes after administration through positive feedback signalling. They also investigated the importance of integrin binding to the response and found a marked reduction of cytokine production if αvβ3 integrin cannot be engaged through the vector [
184]. In a later paper, they outlined how expression of IL1α leads to expression of more chemokines such as CXCL2 as well as complement proteins, which attract polymorphonuclear leukocytes to the area. They also observed death of splenic macrophages, which unlike the self-induced death of Kupffer cells in the liver, they were able to attribute to recruited targeted killing by recruited Neutrophils instead [
185]. This shows the importance of integrin binding in the very early innate responses to intravenously administered Ad vectors, as well as a potential mutation to bypass this mechanism through removal of the integrin binding RGD-motif.
Kupffer cells are tissue-resident liver macrophage. As natural Ad tropism will direct intravenously administered Ad vectors to the liver, they are an essential factor in the innate anti-Ad vector response: While tissue tropism remains, Kupffer cell depletion markedly increases Ad vector gene delivery in intravenous delivered HAdV-C5 based vectors in mice [
186]. Similarly, in vectors where Kupffer cell recognition is reduced, FX dependent transduction of hepatocytes is markedly increased, as shown be Khare
et al. in both intravenously injected mouse and Syrian hamster models [
187]. Only the prevention of both FX binding and Kupffer cell uptake leads to an increase in systemic Ad delivery
in vivo [
186]. Interestingly, in repeat administrations within 72 hours without prior Kupffer cell depletion, transduction to the liver and Kupffer cells is still reduced, instead higher Ad gene expression is observed in the marginal zone of the spleen. Changes in biodistribution were attributed to Kupffer cell depletion after the initial dose due to immunogenic cell death. While the authors suggest Kupffer cells will have replenished by 72 hours, these newly bone marrow derived cells will have slightly different receptor expression. Their impaired ability to clear Ad vectors leads to increased transduction to the spleen. At later time points of administration, Kupffer cells regain their scavenger receptors and biodistribution is more similar to the initial administration [
188]. This may be an important observation that may change considerations when designing repeat administration schedules, such as for oncolytic virotherapies.
Interactions with components of the complement system further activate the anti-vector immune responses. Ad vectors can directly interact with C3. In the absence of C3, intravenously administered HAdV-C5 vectors cannot induce thrombocytopenia in mice, indicating complement is involved in this response. Overall cytokine release and systemic inflammation was also reduced in C3 knockout mice [
189], which can be expected as C3 is involved in positive feedback loops activating the innate immune system (
Figure 3). Tian
et al. observed that
in vivo in mice after intravenous application, complement mainly senses Ad vectors when they have already infected cells, as HAdV-C5 vector defective in cell entry had a much reduced complement response.
In vitro, complement was able to interact with vector particles directly, causing a much larger response [
190]. This is important to note, as it demonstrates that data on complement will likely have to be collected using
in vivo experiments to provide accurate results. For example, the Ad hexon has also been implicated in binding IgG, which may further facilitate complement binding and neutralisation via the complement cascade.
In vitro, this mechanism reduces Ad transduction via destruction by the classical complement cascade.
In vivo, there appear to be other mechanisms involved as well [
190]: As described earlier signalling of activated cells of the innate immune system can increase complement activity (
Figure 3), which would not be picked up by pure
in vitro experiments.
As with natural Ad infection, vectors from different serotypes and species will engage the immune system in different ways, which may make some serotypes more suitable than others. As touched upon before different species activate the innate immune system to different levels due to differences in tricellular trafficking in natural infection. This is also true for Ad vectors. In a rhesus monkeys vaccination study by Teigler
et al., low seroprevalence vectors based on HAdV-B35, HAdV-D26 and HAdV-D48, induced a much stronger innate cytokine response that the HAdV-C5 vector [
191].
Though the innate immune system presents challenges for the immediate systemic administration of Ad based vectors, the innate and adaptive immune response are intrinsically linked. Differences between vectors in the innate response will likely also translate to differences in the adaptive response.
5.3. Adaptive Immunity
The development of anti-vector NAbs is a concern for Ad use in any vector context. Neutralisation occurs instantly when viral vectors are administered. There are several mechanisms through which anti-vector antibodies can neutralise an administered Ad based therapy. Anti-vector antibodies can redirect Ads to phagocytes, such as macrophages, and inhibit endosomal escape, leading to Ad degradation rather than propagation in mice [
103]. Aggregation has been suggested as the main mechanism of anti-hexon neutralisation in intramuscularly vaccinated rabbits, though the same article also confirms post-entry neutralisation of Ads [
100]. This is very likely through a similar mechanism as described above, involving intracellular actions of antibodies. For vector specific anti-Ad antibodies, the intracellular receptor TRIM21, which is antibody activated, has been shown to be a major facilitator of neutralisation through recruitment of proteases to digest intracellular vector particles [
192].
Using the same HAdV-C5 vaccine vector encoding different transgenes does not give a satisfactory anti-transgene response, as demonstrated by Steffensen
et al. which they were able to attribute mainly to the development of anti-vector NAbs. However, priming and boosting against the same antigen using the same vector is possible, likely because a much lower concentration of antigen required to activate the recall response [
193].
Intratumoural injection of replication deficient Ad vectors into subcutaneous tumours in mice is still generates a strong systemic anti-vector NAb response, which is an important consideration in potential Ad based cancer therapies [
194]. In 1998, Gahéry-Ségard
et al. identified in intratumoural injection of replication deficient HAdV-C5 based vectors in 4 lung cancer patients that antibodies against fiber are expressed first, followed by anti-penton and anti-hexon antibodies. While half the patients developed a strong anti-penton IgG response, the other half developed a strong anti-hexon IgG response. They also found that the presence of anti-penton antibodies was indicative of a strongly neutralising anti-vector response [
95]. More recent studies into oncolytic vectors based on HAdV-C5 found that, while replication deficient vectors were supressed more by NAbs (in both human and mouse serum), oncolytic vectors still had diminished transduction in cancer cell lines. Cell killing
in vitro was also markedly reduced by the presence of high titer NAbs. In the presence of low titer NAbs, cell killing was possible through overcoming of NAbs, though it was inhibited at earlier time points. Consequently, they found that the higher NAb titer, the later (if at all) decrease in cell viability was detected. In subsequent
in vivo studies with tumour xenografts in mice, transgene expression was reduced after administration of anti-vector NAbs to nude mice. Tumour shrinkage was still observed in mice with low dose of transferred anti-Ad NAbs, but significantly reduced in mice transferred with high dose of NAbs [
195]. Since for xenograft experiments mice need to be immunosuppressed, the authors transplanted high or low amounts of NAb serum from other mice induced by intravenous challenge, then injected Ad therapies intratumorally. This excludes the presence of cellular immunity, so any effect will be due to NAbs only. While informative, that means these results are likely not representative of a full
in vivo system.
Intravenously administered replication deficient vectors induce both anti-hexon and anti-fiber antibodies in mice, though anti-hexon antibodies have a much stronger neutralising effect [
93]. The main target for this neutralising response are the HVRs of the hexon [
98].
A vaccination study in healthy volunteers by Cheng
et al. showed that intramuscular vaccination induced NAbs against the capsid proteins other than the fiber [
196]. Previous
in vitro reports indicate that this may be due to an anti-hexon response [
100,
197]. In intramuscular vaccinations with chimeric vectors based on HAdV-C5 and HAdV-B35, which previously showed to be non-crossreactive, NAbs against both parent vectors were detected. However, vector-specific anti-hexon NAbs were more common and effective in adoptive transfer studies, using purified antibodies from previously vaccinated mouse serum [
97]. Following a Phase II intramuscular vaccination trial using a HAdV-C5 based vector, NAbs to vector pre- and post-vaccinations were measured, only distinguishing between fiber and not fiber. Vaccine induced NAbs were directed against the capsid, and in some cases both capsid and fiber, while fiber NAbs were mostly detected in volunteers that were seropositive for HAdV-C5 pre-vaccination [
198].
Steffensen
et al. compared subcutaneous and intramuscular routes of administration using the same replication deficient vector expressing different transgenes and found that the route of administration generating the neutralising response did not matter [
193]. More recently, the ChAd36 based vaccine vector BB154 (
Table 3) showed that after intranasal vaccination in Phase III clinical trials, vector specific neutralising antibody levels were low [
162]. On the other hand, a Phase I clinical trial utilising HAdV-C6 and ChAd3 vectors as intramuscular Hepatitis C vaccines in the UK found heterologous prime-boost schedules to be less effective than expected. They attributed this to cross-reactive NAbs, effective against both vectors [
199]. This highlights how differences in route of administration and different serotypes can alter immune response to the vector, which if harnessed correctly can work to our advantage.
Another Phase I clinical trial using a ChAd63 vaccine vector intradermally and intramuscularly for malaria measured anti-ChAd63 NAbs pre- and post-vaccination. They concluded that, since participants with pre-existing anti-ChAd63 NAbs were not observed to mount a reduced transgene response, this will likely not prevent future applications [
200]. However, antibody titers pre-vaccination were not published and may therefore be much lower than post-vaccination. Furthermore, re-boosting against the same antigen, which has proven successful before, does not prove that the vector can be re-used against other antigens in the presence of heightened anti-vector immunity. Ewer et a.l and Kimani et al. reported on the same ChAd63 based vector in the UK and African cohorts of a PhaseI/II trial. Both measured low levels of pre-existing NAbs to ChAd63 [
201,
202]. Interestingly while Ewer et al. noted a minor correlation between low levels of anti-vector antibodies and increased anti-transgene T cell responses [
201], indicating the Ad vector might work as an adjuvant, Kimani et al. observed the opposite: Higher anti-vector NAbs correlated with a slight reduction in T cell anti-transgene response [
202].
Vaccinations with the ChAdOx1 vector described earlier have put the spotlight on reusability of the vector. A Phase2/3 covid vaccine trial intramuscularly administrated ChAdOx1, anti-vector antibodies were induced with the first vaccination, however boosting did not increase NAbs further. Presence of anti-vector NAbs pre-vaccination had a minor negative impact on anti-transgene NAb titers. Anti-vector NAbs generated after boost vaccination however did not negatively impact the cellular anti-transgene response [
203]. As mentioned previously, boosting against the same antigen is possible despite the presence of anti-vector immunity. It is likely that repeated boosters using the ChAdOx1 vector against the same SARS-CoV2 antigen will be successful. However comparing the same metrics (prime vs boost NAbs against transgene or prime vs boost cellular response against transgene) might have been more informative. In 68 patients in Korea, no pre-existing anti-Ad NAbs were detected in only 19. This is not surprising, as pre-existing anti-Ad immunity is very commonly found in populations. The transgene response was observed to be much higher overall in patients without pre-existing anti-Ad NAbs, especially after booster vaccine [
204]. This is very surprising, given it is unlikely that many people will have encountered a ChAd before. Authors state that this indicates the presence of cross-reactive NAbs. Using a different vector based on HAdV-C5, 346 Mexican subjects were vaccinated intramuscularly with Ad5-nCoV. Hernandez-Bello observed a trend that pre-existing anti-HAdV-C5 NAbs lead to a less effective transgene response, but this result was not statistically significant [
205]. This again defies expectation, as anti-HAdV-C5 NAbs are usually very common. HIV vaccine studies tested intramuscularly in rhesus macaques based on HAdV-C5, ChAd6, ChAd7 showed no anti-Ad antibodies before the first dose. Low titer NAbs to ChAd7 were found after the first dose, with a slight increase over time, while titers of NAbs to HAdV-C5 were high even within short time after first administration. Booster with a different Ad serotype did not only produce vector specific NAbs, but also increased the amount of anti-prime vector NAbs [
206]. This is a good example of how different types of Ad can be more or less immunogenic.
Overall, anti-vector NAbs pose a problem to both vaccine and OV applications, especially upon repeat administrations, as NAbs often show the strongest activity against vector specific regions such as the hexon HVRs. However, there is also some evidence for NAbs cross-reacting even across species. It is possible that, by only looking at antibody responses, some studies attributed cross-reactivity to NAbs when cellular immunity may have played a part in unexpected responses as well, especially in human subjects with pre-existing anti-Ad immunity.
Trials for a HAdV-C5 based SARS-CoV2 vaccine in China results showed that vaccine efficacy is limited by pre-existing anti-HAdV-C immunity [
207], though authors do not expand on whether his effect is largely due to T cell immunity or neutralizing antibody responses. Likely, both play a role. It has been shown, that ross-reactivity of T cells between Ad serotypes can impair generation of target antibodies even in rare Ad based vaccine vectors [
208].
Anti-Ad-vector T cell responses and epitopes are not often studied
in vivo. Instead, most articles focus on differences in anti-transgene responses as a measure of efficacy for new vaccine vectors [
209,
210,
211,
212,
213]. Whilst responses to transgene are important, the development of anti-Ad vector responses and how they differ according to route of administration needs to be studied in more detail.
Heemskerk
et al. investigated anti-Ad T cells in PBMCs from 25 healthy donors. In response to stimulation with inactivated HAdV-C5, 76% of donor T cells reacted. Newly stimulated T cells were found to be cross reactive to varying degrees: 3 clones were cross-reactive within species, 4 clones between some species, and 5 cross-reacted between all species tested [
214]. While T cells can often be widely cross-reactive, this shows that vector specific T cells are also raised in response to vector administration. Schirmbeck
et al., similar to other studies, found no response against a new transgene in intramuscularly HAdV-C5 vector vaccinated mice induced with pre-existing Anti-HAdV-C5 immunity. Their experiment showed that viral replication was not required for the development of an anti-vector CD8+ T-cell response, so minimal amounts of antigen are required for an effective response, which was determined to be targeted to highly conserved regions of the hexon [
215].
Investigating the low seroprevalence ChAd63 as a vector for intramuscular malaria vaccines in Phase I clinical trial, pre-existing T cell responses to the ChAd63 vector hexon were found in 8 out of 12 volunteers tested, and increased in all participants after vaccination. Pre-vaccination anti-vector T cell responses led to lower anti-transgene responses, with lower final anti-transgene antibody titers. Both CD4+ and CD8+ T cells were found to contribute to this response [
208]. As stated previously, it is unlikely that participants encountered ChAd63 through infection, hence the observed response likely resulted from cross-reactive T cells mounted in response to other Ad infections.
Another phase I clinical trial in the UK investigated using HAdV-C6 and ChAd3 vectors as heterologous HepC vaccine vectors. The study excluded volunteers with anti-vector antibody titers of >200, to prevent NAbs interfering with vector efficacy. They also measured anti-vector T cell response against the hexon. Responses were found even in non-primed subjects, and were increased substantially after vector administration. Booster vaccinations induced less transgene response than predicted. The study excluded participants with >200 anti-Ad antibody titer, so author concluded that anti-Hexon T cell responses were however not negatively correlated with transgene response, and instead attributed less-than-expected booster responses to cross-reactive NAbs developed after first administration [
199]. Despite measuring a substantial increase in vector targeted T-cells, authors attribute low booster responses to NAbs. It is extremely unlikely that T cells are completely uninvolved in the anti-vector response presented.
A large amount of anti-Ad vector data is available from the STEP trial (NCT00125970) and follow up data analyses. A replication deficient HAdV-C5 vector was used in a phase II clinical trial as a HIV vaccine vector. 480 participants were vaccinated intramuscularly. The trial failed to protect vaccinees from HIV infection, and actually made some recipients of the HAdV-C5 based vector (uncircumcised men) more susceptible to HIV. While this was a devastating result, it led to several more in-depth studies on anti-Ad vector responses, and how these may affect vector applications. Using PBMCs from trial participants, the HAdV-C5 vector was found to induce receptor expression favourable to HIV in CD4+ T cells, as well as inducing a stronger anti-vector CD4+ than CD8+ T cell response, making them more susceptible to HIV infection [
216]. In a follow up from STEP trial, Frahm et al. investigated cellular immunity to Ad vectors. Ad specific T cells could commonly be detected even in the absence of anti-Ad NAbs (in 54% of the seronegative/placebo vaccinated group). Pre-existing CD4+ and CD8+ T cell immunity to Ads induced impaired anti-transgene responses in vaccine vectors, regardless of the presence of NAbs. T cell target epitopes map to conserved region of the vector Ads. For CD4+ and CD8+ most these are commonly hexon, but other, non-structural epitopes are also recognised, such as protein (p) V, pVII, E2 terminal protein (E2pTP) for CD4+ and E3 glycoprotein 19 (E3gp19) and E4 open reading frame 6 (E4ORF6) for CD8+. Interestingly, E3gp19 and E4ORF6 were only targeted in the vaccine group, though results were not statistically significant due to the small sample size [
217]. Hutnick et al. identified pre-existing anti-Ad CD8+ T cells in 95% of unvaccinated volunteers of the STEP trial. They attribute these high numbers to cross-reactivity rather than natural occurrence rate of HAdV-C5. Differences in CD8+ response were observed between different NAb titer subjects, though anti-vector response overall increased regardless of seropositivity before vaccination, even with replication deficient Ads [
218]. Overall, these results emphasize that serotype specific immunity, though important more immediately upon administration, cannot predict later cross-reactivity due to T cells, and resulting loss of efficacy of vector applications. Furthermore, seropositivity as defined by Nab titer is not an indicator that a person is completely naïve to Ad infection. More research into anti-Ad vector responses is required to more accurately predict how a vector is likely to perform in clinic. Furthermore, it is impossible to regard only one side of the anti-vector response, as T cell and B cell responses (as well as the innate immune system) are intrinsically linked.
For example early gene therapy studies for the treatment of cystic fibrosis in mice using an intranasally administered HAdV-C5 based vector highlighted the importance of CD8+ T cell anti-vector responses, as mice that were challenged pre-treatment and only able to mount CD4+ T cell responses still saw some transgene expression. Additionally, the development of NAbs to the vector they made subsequent administrations much less effective, even in the absence of T cells [
219]. Sumida et al. studied intramuscular immunisations with HAdV-C5 based vectors. After immunisation, they transferred splenocytes and serum to naïve mice, to separately test the effects of T cells and NAbs on subsequent vector administration. Both resulted in drastically lower responses to the transgene. Transfer of high levels of NAbs resulted in immediate, but short lived vector inhibition. 53% of mice were still able to induce a transgene response after transfer of splenocytes from double vaccinated mice, though anti-vector antibodies were developed after 2 weeks, either through transferred B or CD4+ T cells. This elegantly shows the synergistic effect of antibodies and T cells: while NAbs confer immediate immunity, long term immunity requires CD4+ T cell help. The immediate vector suppression from transfer of splenocytes was attributed to CD8+ T cells [
220]. Similarly, the adaptive immune system will always interact with the innate immune system: For example, complement protein C3 is required to mount a neutralising ant-Ad vector antibody response, as fond by Appledorn et al. [
221]. The complement system is an important in the bridge between innate and adaptive immunity, and a lot of the cytokines secreted due to complement activation are essential for the maturation of T and B cells. In the absence of these pro-inflammatory cytokines, it is possible to generate a tolerating anti-Ad response instead of a neutralising one.
As briefly touched upon before, different species and serotypes of Ads are considered to have different immunogenicity. Johnson
et al. showed that in PBMCs from healthy donors, HAdV-D28 and HAdV-B35 lead to increased death of APCs. Consequently, less antigen was presented to T cells and response to the transgene was significantly lower than an equivalent HAdV-C5 based vaccine vectors [
83]. While in a vaccine context this is not ideal, it also means decreased presentation of vector antigens, leading to reduced anti-vector responses and increasing the potential for effective repeated administration. Similarly, in a study investigating a broad range of ChAd vectors as well as a selection of rare HAdVs for their suitability as vaccine vectors
in vivo, found lower doses of species C Ads were required to induce a robust anti-transgene response compared to other human Ads in mice and rhesus macaques [
222]. This could be attributed to a similar mechanism as Johnson
et al. In the same study, ChAd vectors showed surprisingly effective at inducing anti-transgene responses and additionally were found to be minimally affected by cross-serotype and cross-species anti-vector immunity in mice [
222], which indicates no cross-reactive neutralising antibodies are produced. Responses to the same ChAd vector carrying different transgenes however were very limited [
222], indicating a strong vector specific response was still induced. For some examples, Ad hexon PF4 binding has been linked to very rare instances of thrombotic thrombocytopenia [
43,
223,
224], which was only apparent due to the wide rollout of the ChAdOx1 nCov-19 and Ad26.COV2.S vaccines during the coronavirus pandemic. Vaccine-induced thrombotic thrombocytopenia (VITT) is hypothesised to driven by autoantibodies against PF4 [
224]. It is extremely rare, with roughly one case occurring in 67,000 primary dose vaccinations and one in 500,000 booster dose vaccinations with ChAdOx1 nCoV-19 in the UK according to the National Institute for Care Excellence (NICE) [
225,
226]. Interestingly, incidence of VITT in the US after administration of the Ad26.COV2.S vaccine was estimated to be 1 in 263,000 administered doses, which is lower than was found with the ChAdOx1 vector [
227]. The generation of autoantibodies to PF4 induce aggregation, leading to platelet activation resulting in a thrombotic event [
43,
228,
229].Binding to PF4 to the viral vector, which may well be critical in the generation of anti-PF4 autoantibodies, has been suggested to occur via HVR1[
230] and involving interactions at the inter-hexon spaces [
231], though neither of these theories have been confirmed. Confirming this mechanism would also allow for predictions of other Ad serotypes to bind PF4, and their consequent prevention.