1,421
Views
0
CrossRef citations to date
0
Altmetric
Review

Locked and loaded: engineering and arming oncolytic adenoviruses to enhance anti-tumor immune responses

&
Pages 1359-1378 | Received 19 Aug 2022, Accepted 20 Oct 2022, Published online: 09 Nov 2022

ABSTRACT

Introduction

Oncolytic adenoviruses (Ads) are promising therapeutics to enhance anti-tumor immune responses and modulate the immune-suppressive tumor microenvironment (TME). Due to their potent ability to deliver genes in vivo, oncolytic Ads have been armed with a variety of immune stimulatory payloads to boost tumor immunotherapy.

Areas covered

We describe current knowledge about engineering oncolytic Ads to insert transgene payloads, including methods to increase its genome capacity for transgene insertion. We also review several categories of immune stimulatory payloads that have been used in oncolytic Ads to combat different barriers to effective immunotherapy.

Expert opinion

We anticipate that multi-armed oncolytic Ads alone or in combination with other types of immunotherapies will greatly improve the efficacy of oncolytic virotherapy in the future by targeting several immune barriers. However, the production and testing of multiple payload-armed Ads can be complex and time-consuming due to the limitations of current tools. Given this, we should develop new tools for rapid construction of armed Ads, improve animal models or new systems to compare the efficacy of multiple payloads, and carefully monitor the immunological toxicity induced by payloads or by systemic delivery. Most importantly, we should pursue payload-arming approaches with great care to ensure safety.

1. Introduction

Oncolytic viruses are a novel class of self-amplifying therapeutic agents that have the natural ability to replicate in and kill cancer cells (reviewed in [Citation1,Citation2]). The original concept of oncolytic viruses is that their efficacy would come from killing primary and disseminated cancers by direct cell lysis. While this does occur, preclinical and clinical data suggest that much of the efficacy of virotherapy may actually arise from their ability to ‘make tumors hot’ or activate the immune system against cancer cells [Citation3,Citation4]. This ability to increase inflammation in tumors comes in part from viral replication of DNA, RNA, and proteins that can be recognized as pathogen-associated molecular patterns (PAMPs) to activate immune cells [Citation3]. Direct viral lysis can also induce immunogenic cell death (ICD) through the release of cellular contents that be recognized as damage-associated molecular patterns (DAMPs) to activate immune cells [Citation3]. Both PAMPs and DAMPs can trigger the secretion of pro-inflammatory cytokines and establish an anti-tumor immunological environment in the tumors. Direct viral lysis can also assist in exposing cancer antigens to antigen-presenting cells (APCs) like macrophages and dendritic cells (DCs) to trigger anti-tumor immunity [Citation3].

A large number of viruses are being developed as oncolytics (reviewed in [Citation1]). While each has its unique strengths and weaknesses, this review focuses on adenoviruses (Ads) as an exemplar for using viruses to amplify immune responses against cancer.

In this review, we will focus on (1) approaches to genetically engineering armed oncolytic Ads and (2) types of payloads to arm oncolytic Ads to overcome current barriers to effective cancer immunotherapy.

2. Adenoviruses (Ads)

Ads are non-enveloped double-stranded DNA viruses with icosahedrons that are about 90 nm in diameter and genomes of approximately 36 kilobase pairs (kbp) [Citation5,Citation6] (). This relatively large genome size allows Ads to carry large payloads. Unlike certain pleiomorphic enveloped viruses, mature Ads form nearly identical particles in the nucleus of cells. This property makes them ideal to be produced at high concentrations from cell lysates, be purified on density gradients or chromatography columns, and ideal for scalable production and relatively simpler GMP production when compared to the purification of enveloped viruses [Citation7]. The high stability of Ad viral proteins and DNA can be lyophilized and stored at relatively high temperatures for months or years so that adenoviral therapeutics can be shipped or stored without the need for low-temperature freezers [Citation8–10]. This is important in remote areas that do not have −80°C freezers, but it is also important even in pharmacies in even well-developed countries [Citation11].

Figure 1. Adenovirus virion. Human Ad26 EM image of the icosahedrons is shown overlaid with cartoons representing short and long fibers that may be present on different Ad serotypes.

Figure 1. Adenovirus virion. Human Ad26 EM image of the icosahedrons is shown overlaid with cartoons representing short and long fibers that may be present on different Ad serotypes.

Figure 2. Adenovirus genome and transcriptome. The E1A gene labeled yellow is an immediately early gene. Green labels are early and intermediate genes, while blue labels are late genes belonging to the major late transcript unit (MLTU). Fragments in the gray shadow show the protein products generated by alternative spliced transcripts. Scissors sign indicates the gene can partially or fully be deleted in oncolytic Ads. Brown diamonds point to the insertion sites for autonomous expression cassettes. Purple circles point to the insertion sites for exogenous splicing elements, or the viral ORFs can be replaced by a transgene. The red club sign indicated reported sites for IRES or 2A peptide insertion.

Figure 2. Adenovirus genome and transcriptome. The E1A gene labeled yellow is an immediately early gene. Green labels are early and intermediate genes, while blue labels are late genes belonging to the major late transcript unit (MLTU). Fragments in the gray shadow show the protein products generated by alternative spliced transcripts. Scissors sign indicates the gene can partially or fully be deleted in oncolytic Ads. Brown diamonds point to the insertion sites for autonomous expression cassettes. Purple circles point to the insertion sites for exogenous splicing elements, or the viral ORFs can be replaced by a transgene. The red club sign indicated reported sites for IRES or 2A peptide insertion.

Ads are well documented as one of the most potent vectors for in vivo gene delivery of gene therapy or genetic vaccines. For example, a replication-defective human Ad5 (RD-Ad5) vector can generate higher anti-poxvirus antigen B5 antibody titers than modified vaccinia Ankara (MVA), Venezuelan equine encephalitis replicons (VEE-VRP), or even a replication-competent rVSV vector [Citation12]. This makes Ads less attractive for gene therapy but makes Ads useful as genetic vaccines or in our case as oncolytic immunotherapies.

There are, in fact, many serotypes of Ads that infect humans, mammals, avian species, reptiles, and even fishes [Citation13]. At the moment, there are more than 100 genetically distinct Ads that infect humans. The viruses in the human Ad virome can be up to 40% different at the DNA sequence level [Citation13–16]. These differences translate into the ability for Ads to naturally target different receptors [Citation14,Citation15], to bind or avoid the binding of host factors in the blood [Citation17–20]. The diverse pools of Ad also allow ‘serotype-switching,’ a shell game by switching Ad capsids with different serotypes to avoid neutralizing antibodies [Citation21–23]. These features make Ads a ‘palette’ of different genetic oncolytic genetic platforms to carry immunomodulatory transgenes for cancer therapy.

3. The adenovirus genetic program

Following receptor binding, Ads enter cells by receptor-mediated endocytosis [Citation5,Citation24–26]. This is followed by endosome escape, trafficking to the nucleus, and delivery of viral DNA into the nucleus usually within 1 hour of virus binding to the cells [Citation24]. Once viral DNA is delivered into the nucleus, host transcription factors bind rapidly to the promoters of immediate early genes to activate virus life cycle.

Ad gene activation and transcription are regulated in a stepwise manner. Viral genes are categorized into four groups based on the timing of expression: (1) immediate early gene: E1A, (2) early genes: E1B, E2A, E2B, E3, L1-52/55 K, (3) intermediate genes: IX, Iva2, and (4) late genes: L1-L5, or major late transcription unit (MLTU) (). The early-late gene switch is tightly regulated by essential host factors and viral proteins expressed during each phase that activate viral gene promoters for the next stage as well as control RNA splicing site preferences [Citation27,Citation28].

E1A is the master regulator of the viral life cycle. E1A encodes the first viral proteins expressed after infection. Upon DNA entry into the nucleus, typically abundant and ubiquitous host transcription factors bind to the E1A enhancer and promoter to activate its expression without any input from viral proteins. E1A proteins then activate the expression of downstream early genes E1B, E2, E3, and E4 that are essential for viral replication, repression of host cell transcription and translation, and inhibition of host cell antiviral responses [Citation29]. The transition from the intermediate phase to the late phase is dictated by the activation of the major late promoter and the expression of L1-L5 regions of the genome via the MLTU. The expression of these late proteins is mediated by L4-33 K, an RNA splicing factor whose expression is driven by the L4 promoter activated by E1A, E4-ORF3, and IVa2 [Citation30,Citation31]. This stepwise early to late gene expression pattern is evolutionarily developed in most large DNA viruses. It allows efficient virus production by first inhibiting host antiviral responses, amplifying viral DNA, and then producing viral structural proteins for the virion assembly [Citation29].

In human cells with intact p300 and pRB pathways (e.g. A549 human lung cells), early genes are activated within 6 hours of infection and late genes are activated generally by 12 hours [Citation32]. E1, E2, and E3 expression peaks early, but can persist throughout the life cycle. In contrast, E4 transcripts peak early and are largely quenched within 12 hours of infection [Citation32,Citation33]. DNA synthesis commences in A549 cells within 12 hours of infection and plateaus by 36 hours. Despite these early events in the viral life cycle, cells may not lose membrane integrity and die in culture until 2 to 3 days after infection.

This time course is similar when comparing human species C Ad6 and human species D Ad26 which are 34% genetically different when they infect A549 cells [Citation32]. In other cells, like human bone marrow and B cell myeloma cells that have differential sensitivity to different Ads, some serotypes of Ads replicate their DNA over days (i.e. species C Ad5 and Ad6 and species D Ad26 and Ad48), and others fail to replicate and having their DNA degraded over that same period of time (i.e. species B Ad11 and Ad35) [Citation34]. The observation from this report is contrary to other literature stating that the Ad life cycle completes within 24 hours. While short Ad life cycles can occur, some of these may be observed during infection of cells that are hyper-activated and that are already being driven into the cell cycle by other viral proteins in the cells like HeLa cells [Citation35].

4. Complex transcription and splicing of mRNA during the adenovirus life cycle

Ads have evolved the ability to generate many mRNA transcripts and viral proteins from a relatively limited genome size. Ads use both strands of their double-stranded DNA genome to code proteins. They have also evolved a complicated mRNA transcription and splicing system to deploy many proteins at different stages of the life cycle off both strands of the genome. The E2B, E3, E4, and MLTU regions heavily use alternative splicing machinery to generate various viral proteins (). With modern next-generation sequencing technologies [Citation32,Citation36] and long-read sequence technologies [Citation33,Citation37], the temporal distributions of different serotype Ad RNA transcripts are now better able to be dissected and used to help engineer Ads.

For example, there are over 900 mRNA patterns from species C Ad2 and more than 11,000 unique transcripts from species C Ad5 with different bioinformatics data processing pipelines [Citation33,Citation37]. In the late infection phase, L1-L5 genes and E3 genes (MLTU) are expressed under the control of the major late promoter. The viral transcripts from the MLTU are the most diverse and account for 50% of total spliced mRNA variants arising from the Ad genome [Citation33]. Transcripts from the MLTU make up 80% of viral transcripts and are so abundant that they account for up to 40% of total RNA transcripts from cells during the late phase of the viral life cycle [Citation37]. This detailed mRNA variant information from long-read next-generation sequencing is extremely valuable for determining payload insertion sites and designing transgene payload expression in the context of oncolytic Ads.

5. Strategies to increase genome capacity for transgenes

To arm an oncolytic Ad, foreign DNA sequences have to be inserted somewhere in the viral genome. Ad is an icosahedral virus with fixed genome packaging capacity (). Wild-type human adenovirus type 5 (Ad5) has about 36 kbp in genome size and can package up to 105% of its normal genome size without destabilizing the viral genome [Citation38]. This means that only about 2 kbp of foreign sequences can be added to an intact Ad genome without deleting any other DNA viral sequences. Because of this limitation, almost all recombinant Ads also have deletions somewhere else in their genomes to make more space for inserting transgenes.

5.1. E3 region deletions

Most researchers delete the E3 region of the virus to make space since this region encodes immune escape proteins that are dispensable for the replication of the virus in vitro [Citation39,Citation40] (). Mastadenoviruses have genetically divergent E3 genes [Citation39–41]. Some open reading frames (ORFs) are highly conserved in all Ads whereas other E3 ORFs may be unique to different serotypes or Ad species (). For example, species C Ad6 and species D Ad26 are 34% genetically divergent with genomes of 35,760 and 35,152 bp, respectively [Citation32]. Both express E3 12.5 K, 19 K, RIDa, RIDb, and 14.7k ORFs. However, Ad6 expresses 6.7 K and the adenovirus death protein (ADP) but does not express 23 K, 49 K, and 31 K. These differences in E3 ORF content translate into quite different sizes of E3 regions: 2,949 bp in Ad6 and 4,479 bp in Ad26 (). The substantially larger size in Ad26 is compensated in part by having a shorter 8 repeat fiber protein than Ad6ʹs 18 repeat fiber. Therefore, depending on the Ad serotype, approximately 3 to 4.5 kbp of DNA capacity can be used for inserting transgenes after deleting E3 genes. By using a short-shafted fiber gene rather than a long species C flexible fiber gene, an extra 450 bp of DNA capacity can also be available (), but the tropism and infectivity of Ads may also be changed by swapping the fibers from different serotypes.

Figure 3. Adenovirus genome, E3, and fiber regions. Cartoon comparing the sizes of Ad6 and Ad26 genome regions and ORFs.

Figure 3. Adenovirus genome, E3, and fiber regions. Cartoon comparing the sizes of Ad6 and Ad26 genome regions and ORFs.

In the E3 genes of species C Ads, the function of the E3-12.5 K ORF is still unknown, but its sequences are highly conserved between human species C Ads [Citation42]. Deleting 12.5 K can be problematic and reduce virus potency in part because there is a putative poly adenylation sequence for MLP transcripts at the C-terminus of the ORF. Species C E3-CR1-α (6.7 K) is a membrane glycoprotein that localizes to the ER. 6.7 K has been shown to inhibit cell apoptosis by regulating Ca2+ homeostasis [Citation43] and is involved in the degradation of TRAIL receptor 2 with E3-RID complex [Citation44]. The E3-glycoprotein 19 K (gp19K) inhibits MHC-class-I and MIC molecules from being transported from the ER to the cell membrane to inhibit the presentation of viral peptides to cytotoxic T cells and detection by NK cells [Citation45]; E3-ADP is known to enhance nuclear envelope rupture for efficient virus releases and cell death [Citation46]. ADP over-expression accelerates cell death but does not necessarily increase the number of cells that are killed. Accelerating cell death may be useful if all efficacy is derived by direct oncolysis. In contrast, if the virus gains efficacy by expressing transgenes, accelerating death may reduce the duration of transgene expression and potency [Citation47]. E3-RID-α/β suppresses cell apoptosis by internalizing Fas or TRAIL death receptors from the cell surface to protect the cell from death signals from immune cells [Citation48–50]; E3-14.7 K is known to inhibit NF-κB signaling by blocking TNF receptor 1 (TNFR1) internalization and prevent TNF-induced cytotoxicity [Citation51,Citation52].

Previous studies have demonstrated that deletion of partial E3 (1.88 kb) gene or full E3 (2.69 kb) gene in Ad5, which deletes immune evasion ORFs, does not interfere with viral replication in vitro [Citation38]. Up to 4.5 kbp of the E3 gene can also be deleted to make a total of 6.5 kbp capacity, but 3 kbp deletions are more common as bigger deletions generate less robust viruses [Citation39,Citation53]. Some studies also demonstrate that deletion of E3 genes may reduce Ad persistence in vivo [Citation54,Citation55].

For example, Wang et al. compared wildtype Ad5, Ad5(E3-RID-α/β + E3-14.7 K deletion), and Ad5(E3-gp19K deletion) in both immunodeficient and immunocompetent mouse tumor models [Citation54]. They found that E3-RID-α/β + E3-14.7 K deleted Ad5 was cleared more rapidly in mice and had less anti-tumor responses. The tumors injected with Ad5(E3-RID-α/β + E3-14.7 K deletion) had elevated macrophage infiltration/low CD8+ T cell infiltration, and higher TNF and interferon-γ expression compared to wild-type Ad5 or E3-gp19K. While E3-gp19K deletion reduced MHC I display, this deleted virus generated similar immune profiles and potentially better tumor control in the CMT-93 immunocompetent mouse model. A caveat here is that Ad does not replicate well in mouse cells, so amplification of E3 genes via genome replication is weak in these models. In another study, Bortolanza et al. used an immunocompetent and human Ad permissive Syrian hamster tumor model to compare the persistence of wild-type Ad5 and Ad5(E3-CR1-α /gp19K deletion) [Citation55]. They found E3-CR1-α and E3-gp19K deleted Ad5 was cleared much faster than wild-type Ad. However, the anti-tumor effect and overall survival were not reported in this study.

Hence, the deletion of E3 genes can increase insertion capacity, but some E3 gene deletions may cause faster Ad clearance in vivo. Some proteins from E3 genes may also be involved in immunogenic cell death for oncolytic efficacy by inhibiting normal cellular apoptosis and death to release DAMPs [Citation3]. However, whether the deletion of immune evasion E3 genes is beneficial or detrimental for overall anti-tumor efficacy still requires more investigation since the removal of immune evasion viral genes could potentially enhance anti-tumor immune responses.

5.2. E1B deletion

E1B gene encodes two protein products: (1) E1B-19 K and (2) E1B-55 K. The E1B-22S mRNA is a polycistronic mRNA that encodes both E1B-19 K and E1B-55 K [Citation56]. E1B-19 K is a potent inhibitor for cell apoptosis by mimicking anti-apoptotic Bcl-2 protein and binds to pro-apoptotic protein Bax [Citation57]. Interestingly, E1B appears to be better able to protect cells from TNF-mediated apoptosis than E3 proteins, since E3 deletion has only mild effects on protection against TRAIL [Citation32]. E1B-55 K inhibits p53 function by direct binding, regulates viral mRNA biogenesis, and inhibits cellular mRNA accumulation [Citation58]. A dead cell will not support viral replication, so protecting the cell from apoptosis is a critical function of E1B proteins.

Both E1B-19 K and E1B-55 K can be deleted without overtly disturbing the viral life cycle [Citation59]. Full deletion of E1B yields 1.8 kbp of extra payload capacity. The deletion of E1B-55 K is known to make Ad into a conditional-replicative adenovirus (CRAd) that decreases viral replications in several cell types based on three proposed mechanisms: (1) p53 deficiency-dependent, (2) late viral RNA export-dependent, and (3) Cyclin E expression-dependent [Citation60], while the deletion of E1B-19 K is known to increase viral oncolytic property and viral release [Citation61,Citation62].

5.3. pIIIa cement protein gene deletion: an oncolytic Ad that does not generate progeny viruses

1.5 kbp more insertion space can also be gained by deleting the pIIIa gene that encodes a viral cement protein [Citation63]. This generates a single-cycle adenovirus (SC-Ad) that replicates its DNA and completes the life cycle but does not generate infectious virions [Citation63]. SC-Ad has advanced to human testing as a COVID-19 vaccine (NCT04839042). While SC-Ads do not generate progeny to spread infectious viruses to the second wave of cells, they do kill the cells that they infect [Citation64]. Although SC-Ads do not generate infectious progeny that sets aside the spreading feature of oncolytic Ads to kill cancer cells, it trades with another feature: safety.

6. Types of transgene insertions and their locations

Five types of transgene insertion approaches and their insertion locations are summarized below ():

6.1. Locations for transgene insertions

Theoretically, transgene cassettes can be inserted anywhere in the Ad genome. However, the complex expression, transcription, and mRNA splicing of Ads mean that not all locations will tolerate insertions. Transgenes are generally inserted in the place of E1 (only for RD-Ad vectors), in between E1A and B, in E3, between fiber and E4 at the end of both transcripts for the MLPU and E4, and occasionally in other sites (). Farrera-Sal et al. provided an excellent discussion on different locations for transgene insertions into Ads [Citation65].

Terry Hermistons group also provided interesting guidance on insertion sites in Ad5 by randomly inserting autonomous expression cassettes or exogenous splicing elements in the viral genome with a transposon system [Citation66,Citation67]. In E3-deleted Ad5, they found that the autonomous expression cassettes could be tolerated in E4 untranslated regions, between E1A and E1B, and within E1B [Citation66]. For the exogenous splicing elements, they found different versions could be inserted in front of or into E3 ORF regions, into the polypeptide V ORF, at the ends of the MLPU or E4, between the ITR and E4 ORF, or within E4 ORFs () [Citation67].

Data from long-read next-generation sequencing show that the E3 region is heavily spliced. These transcripts are derived not just from the E3 promoter, but also from the MLPU [Citation33]. Although autonomous expression cassettes are commonly used in the E3 region in an E3-deleted Ad backbone, insertion in the E3 region can generate aberrant mRNA splicing and accidentally decrease transgene expression and viral titers [Citation33]. Suzuki et al. also demonstrated that transgene insertion in E1 in the opposite transcriptional orientation or before E4 in the opposite transcriptional orientation gives the best viral titer and transgene expression level.

As a rule of thumb, it is better to avoid heavily spliced regions for inserting autonomous expression cassettes. It is also critical to check transgene sequences to avoid cryptic splicing donor (AGGT/AAGT), acceptor (CAG/G) sites, and exonic splicing enhancers (ESEs) within transgenes to prevent unexpected splicing. Several online software tools can help predict cis-splicing elements [Citation68,Citation69].

6.2. Types of transgene insertions

6.2.1. Replacing existing viral ORFs

Replacing a viral ORF with a transgene ORF utilizes the intrinsic viral expression program to express transgenes. It can make transgenes express as if they were early, intermediate, or late genes for programmed expression. It also entrains transgene expression into the viral program such that if the viral genes are not activated, the transgenes might not be expressed. This ORF substitution strategy has advantages in minimizing transgene cassette space over other approaches that require external regulatory sequence elements. As we reviewed, E1B and E3 genes are non-essential for viral replication and virion production, thus making those genes potential candidates for replacement with therapeutic transgenes. Terry Hermistons group has shown that E3-CR1-α(6.7 K)/gp19K, E3-ADP, and E3-RID-α/β/E3-14.7 K regions can be replaced by a transgene or multiple transgenes () [Citation70–73]. Reporter and IFN genes also have been inserted in E3B regions [Citation74]. Notably, the choice of exact insertion sites in the complex E3 region can affect neighboring E3 ORFs expression and the overall viral fitness [Citation72]. This approach can save some space; however, human Ads do not replicate well in most mouse cells and other animal cells. Therefore, transgene cassettes that piggyback their expression on Ads’ transcripts will probably not be expressed well in some animal models of cancer.

6.2.2. Autonomous expression cassettes

An autonomous expression cassette is composed of an enhancer/promoter region to activate transcription, a transgene (usually a cDNA, sometimes a gene), and a polyadenylation (polyA) signal. Many times, these enhancers/promoters are promiscuous strong viral or cellular elements that will activate in nearly any cell (e.g. from cytomegaloviruses (CMV), Rous sarcoma virus (RSV), EF-1a, etc.). In other cases, these may be cell-specific or ‘cancer-specific’ enhancers/promoters (telomerase, surviving, probasin, etc.). Natural or artificial introns may be inserted between the transcriptional start site and the start methionine to increase mRNA trafficking out of the nucleus to increase the expression [Citation75]. If introns are used, it should be careful not to disrupt normal Ad splicing in the insertion region as described above.

Notably, using an autonomous expression cassette allows immunostimulatory transgenes to be expressed in non-permissive, immunocompetent mouse cancer models. The primary negative effect of using expression cassettes is that they take up more space in the viral genome. A second negative effect is that they may always be ‘on’ in all cells, both cancer and non-target tissues. This can decrease specificity and run the risk of increasing side effects to the oncolytic immunotherapy particularly when they are given systemically.

6.2.3. Internal ribosome entry sequences (IRESs)

In mammalian cells, ribosomes engage the 5’ cap on mRNAs and slide until they find a good start codon to start protein translation. If that ribosome hits a stop codon or falls off, it will generally not re-engage downstream on the same mRNA. Because of this, if two cDNAs are located under one enhancer/promoter, generally only the first cDNA will have robust expression.

To circumvent this problem and conserve space, internal ribosome entry sites (IRESs) from RNA viruses can be utilized. IRESs are 300–600 bp sequences derived from RNA viruses that will allow ribosomes to be recruited and initiate protein translation without 5ʹcap recognition. Placing IRES upstream of the second cDNA will allow polycistronic gene expressions [Citation76]. IRESs have been used to co-express transgenes on the same transcripts as E1A, E2B, and fiber [Citation77]. Head-to-head comparisons of IRESs in these locations showed that fiber-linked IRES transcripts had the strongest expression, although this expression was delayed [Citation77]. Intermediate levels of expression were observed with E2B-linked IRES transcripts, while E1A-linked IRES transcripts have the lowest luciferase expression level [Citation77]. Although IRES elements eliminate the need for a second set of enhancer/promoters and polyA sequences, they are still relatively long and consume precious viral genome space. In addition, typically weaker expression of the second cDNA in an IRES cassette is observed. This can vary from gene to gene, so the expression of genes in IRES cassettes has to be measured empirically.

6.2.4. Translational truncation sequences

Translational truncation sequences have been co-opted from viruses like Foot and Mouth Disease Virus (FMDV) (reviewed in [Citation78]). These so-called ‘2A’ peptide sequences are usually 18–22 amino acids (50–70 bp). When they are inserted in-frame between two ORFs, they induce ribosomal stalling and skipping during translation to yield two separate proteins from one mRNA [Citation79]. 2A elements have the primary advantage of drastically reducing the size of two cDNA cassettes when compared to autonomous cassettes and IRES elements since 2A elements are only 50–70 bp in length. While 2A elements can work, there will always be a 2A sequence ‘scar’ on the c-terminus of the first protein and a proline added to the second protein. 2A elements have been used to link many transgene proteins together (i.e. IL-12 alpha and beta subunits), but also to fuse transgenes to E1A, pIX, or fiber genes [Citation80–82]. However, the fusion of 2A proteins to the pIX capsid cement protein can cause instability of capsids [Citation80]. Fiber-linked-2A peptide can cause repression of the E1A expression [Citation81]. Thus, the use of 2A peptides in Ads should be empirically evaluated.

6.2.5. Insertion of exogenous splicing elements

Insertion of splice acceptor (SA) sites hijack intrinsic viral RNA splicing machinery to stitch foreign cDNAs into viral transcripts to allow transgene expression. This approach is similar to replacing a viral ORF but avoids deleting what may be an important viral protein. This approach has the great advantage of minimizing insert size because SA sequences can be as small as 30–50 bp. This strategy has been reported by using SA sites derived from Ad5 pIIIa(IIIaSA) [Citation83], Ad40 long fiber (40SA) [Citation84], Ad41 long fiber (41SA) [Citation85], SV40 large T antigen [Citation86] and beta globulin gene [Citation87]. It is critical to check and optimize the transgene sequence to avoid potential inhibitory splicing cis-elements within transgene as they can interfere with viral protein splicing and transgene expression [Citation81]. These elements usually need to be placed after stop codons but before polyA elements. These approaches are best made at the end of transcriptional units to avoid deranging normal viral splicing.

Terry Hermistons transposon-based approach identified several potential SA-transgene insertion sites within pV ORF, between E3-14.7 K and fiber, between fiber and E4, between E4-ORF2 and E4-ORF4, within E4-ORF6/7 and between E4 and ITR [Citation67]. Robinson et al. also reported an insertion position before or after the hexon gene () [Citation88]. The most popular insertion site is between the L5 gene (fiber) and the E4 region, expanding a new splicing transgene region, the so-called ‘L6’ region [Citation85]. Robinson et al. compared insertion sites for both within E3 and after L3 genes and showed that L3 insertion has stronger transgene expression than SA insertion in E3. Farrera-Sal et al. compared the insertion location in the L6 region and between E4 and ITR regions, as well as using two different SA sites: IIIASA and 40SA [Citation89]. They found insertion in the L6 region with 40SA gives the highest transgene expression. It should be noted that the types of SA sites and each transgene sequence may affect virus fitness.

7. Barriers to generating anti-tumor immune responses

Tumorigenesis is a slow process during which normal cells accumulate oncogenic mutations. Immunogenic cancer cells are more likely to be recognized and cleared by the immune system, so there is a natural selection to accumulate mutations that avoid immune surveillance [Citation90]. These mutations can manifest into generating at least four major barriers to effective immune control of cancer ():

Figure 4. Barriers to immunotherapy. (1) Insufficient immune signaling and priming, (2) tumor cell-intrinsic immune evasion, (3) barriers to immune cell infiltration, and (4) immunosuppressive tumor microenvironment.

Figure 4. Barriers to immunotherapy. (1) Insufficient immune signaling and priming, (2) tumor cell-intrinsic immune evasion, (3) barriers to immune cell infiltration, and (4) immunosuppressive tumor microenvironment.
  1. Insufficient immune cell activation against self or tumor neoantigens

  2. Tumor cell-intrinsic immune evasion

  3. Physical barriers to immune cell infiltration into tumor

  4. Development of an immunosuppressive tumor microenvironment (TME)

In this review, we will briefly focus on selected barriers to anti-cancer immunity and how Ad oncolytics might combat these challenges. We refer readers to several excellent reviews on arming oncolytic viruses to combat tumor immune evasion [Citation91–93] for more detailed discussions on these topics.

7.1. Insufficient immune signaling and priming

Immune tolerance can be imposed during B and T cell ontogeny by central tolerance. However, the balance between immune activation and immune tolerance is also modulated later by innate immune cells, antigen-presenting cells (APCs), and other types of immune cells in the peripheral [Citation94]. Innate immune cells can be activated by receptors of PAMPs and DAMPs to recognize pathogens [Citation3]. These pattern recognition systems are good at detecting strange external invaders but less useful in detecting neoplastic cells as they lack these patterns. In the absence of innate immune triggers, long-term exposure of tumor antigens to T cells can gradually convert T cells into an anergic or dysfunctional state () [Citation90]. Thus, slow growth in the absence of innate cell danger signals can allow tumor cells to evade T cell recognition. In other cases, inflammation itself can assist in the tumorigenesis [Citation95], but this is a different discussion.

7.2. Tumor cell-intrinsic immune evasion

Immune pressure can drive cancer cells to be selected to evade cytotoxic T cells and other immune effects. For example, tumor cells that down-regulate MHC class I molecules (MHC-I) on their surface avoid being killed by cytotoxic CD8 + T cells [Citation96]. While down-regulation of MHC-I would normally set up the cancer cell to be recognized by natural killer (NK) cells, tumor cells that can also up-regulate non-classical MHC class I molecules can also be selected to evade NK cell-mediated killing [Citation96].

Cancer cells can also evade immune surveillance by constantly mutating their genomes as they grow. There may be an assumption that a higher mutation burden gives higher neo-antigen loads to be recognized by the immune system. However, these mutations can also increase tumor heterogeneity to generate escape clones and dilute potent tumor antigens to immunologically ‘cool down’ the tumor [Citation97]. Cancer cells can also be selected for their ability to up-regulate immune checkpoint molecules like CD47, CD200, PD-L1, PD-L2, CTLA-4, LAG-3, TIGIT, and TIM-3 that inhibit dendritic cells (DCs), T cells, and natural killer (NK) cells [Citation98]. Interestingly, overexpression of surface glycans by tumor cells, like sialic acids, can also be selected to suppress NK and DC cell functions by binding to inhibitory receptors Siglec-7 and Siglec-9, respectively [Citation98,Citation99].

7.3. Barrier for immune cell infiltration

The aberrant vasculature in some solid tumors causes high tumor interstitial fluid pressure (IFP) that can compress lymphatics and vessels reducing the ability of immune cells to enter tumors [Citation100,Citation101]. Tumor cells can also recruit non-cancerous stroma cells to form solid physical barriers that prevent immune cells from penetrating beyond blood vessels and lymphatics [Citation102]. Cancer cells recruit cancer-associated fibroblasts (CAFs). These are a particular problem as they secrete extracellular matrix (ECM) materials, including collagen and fibronectin to form a tight extracellular physical barrier that prevents immune cells from migrating within solid tumors [Citation102]. The added ECM also induces mechanical stress in the tumor to induce the production of transforming growth factor-beta (TGF-β) from these stroma cells. TGF-β secretion further increases collagen production by CAFs and inhibits T cell infiltration [Citation101]. Beyond these physical barriers to immune cell infiltration, tumors frequently lack the expression of chemokines and proper adhesion molecules for T cells to home to the tumor and infiltrate into its core [Citation103].

7.4. Immunosuppressive tumor microenvironment (TME)

TME formation is mediated by communications between tumor cells, stroma cells (endothelial cells and fibroblasts), and immune cells [Citation104]. As mentioned above, CAFs can secrete ECM materials to confine the tumor space physically. In this confined space, uncontrolled growth of tumor cells not only depletes nutrients and oxygen that are essential for survival but also produces floods of toxic metabolites that suppress the ability of immune cells to activate and indeed even survive [Citation105,Citation106]. These metabolic signals affect immune cells in TME and can transform them into immune suppressor cells like regulatory T cells (Tregs), M2 tumor‐associated macrophages (TAM), and myeloid-derived suppressor cells (MDSCs) [Citation98,Citation104]. These immune suppressive cells can secrete immune suppressive cytokines, like TGF-β and IL-10, and express immune inhibitory ligands to inhibit new immune responses or to render infiltrating T cells or NK cells dysfunctional [Citation98,Citation104].

8. Arming oncolytic Ads to break down barriers to the immune system

Oncolytic Ads themselves can be potent immune adjuvants to make tumors immunologically hot to allow immune checkpoint inhibitors to be effective [Citation3]. While Ads intrinsic immune-stimulatory functions are useful, their utility can be improved by arming them with immune-modulating payloads. A survey of the literature for immune payloads in Ads gives an overwhelming array of choices that are beyond the scope of this review to capture. Instead, we will touch upon a subset that can act as examples. We summarize several published immune modulating payloads expressed by oncolytic Ads for either single-armed oncolytic Ads () or multi-armed oncolytic Ads () for targeting each barrier of immunotherapy ().

Figure 5. Strategic targeting by arming oncolytic Ads with immune modulation payloads. (1) Enhancing T cell priming, polarization, and memory T cell generation. (2) Inhibiting cancer immune evasion by cytokines and blocking immune inhibitory ligands. (3) Disrupting physical barriers and increasing immune cell infiltration. (4) Inhibiting immune suppressive cells.

Figure 5. Strategic targeting by arming oncolytic Ads with immune modulation payloads. (1) Enhancing T cell priming, polarization, and memory T cell generation. (2) Inhibiting cancer immune evasion by cytokines and blocking immune inhibitory ligands. (3) Disrupting physical barriers and increasing immune cell infiltration. (4) Inhibiting immune suppressive cells.

Table 1. Payloads used in single-armed oncolytic adenoviruses.

Table 2. Payloads used in multi-armed oncolytic adenoviruses.

Cytokines play key immunoregulatory roles. However, not surprisingly, some cytokines can provoke either anti-tumor or pro-tumor immune responses, depending on the doses and existing cytokines in the environment. For example, granulocyte-macrophage colony-stimulating factor (GM-CSF) is one of the most frequently used cytokine transgenes for arming oncolytic viruses. Indeed, GM-CSF is the payload in T-VEC, the first FDA-approved oncolytic virus in the U.S. [Citation111]. GM-CSF stimulates the differentiation and maturation of APCs and DCs and therefore could be useful in improving immune responses against tumors. GM-CSF has been shown to enhance anti-tumor effects [Citation112]. However, GM-CSF can also recruit immunosuppressive TAMs into tumors. Depending on the balance of other immune responses, GM-CSF can control the tumor or make it worse [Citation112].

On the other hand, overexpression of a potent pro-inflammatory cytokine can also lead to the upregulation of immune inhibitory ligands and immune cell exhaustion as it may induce intrinsic negative feedback loops to prevent autoimmunity. For example, a high concentration of IFNγ expression may induce overexpression of PD-L1 on tumor cells to suppress anti-tumor immune responses [Citation113]. It may be possible to balance these effects if an oncolytic virus can be carefully crafted to express multiple immune-modulating payloads to activate anti-tumor immune responses while also combating negative feedback immune-suppressive responses.

It should be noted that secreted cytokines can spread throughout the body. Therefore, they may risk broader side effects than immunostimulatory proteins that are embedded in the cell membrane. Conversely, an embedded protein trapped only in tumor cells may never be able to reach out and touch immune cells in tumor-draining lymph nodes to amplify their activity [Citation114].

8.1. Enhancing T cell priming, polarization, and memory T cell generation

Oncolytic Ad itself can trigger strong innate immune activation. However, promoting DC maturation can more specifically activate T cells against tumor antigens with low immunogenicity [Citation115]. DC maturation can be enhanced through ligation of CD40 ligand (CD40L) with CD40 that is expressed on DCs [Citation116]. Alternately, DCs can be activated by stimulation with cytokines like TNF-α, IFN-α, IFN-γ, and GM-CSF (with the caveats noted earlier). Several oncolytic Ads have been armed with these factors that potentially target DCs. These have been reported to enhance anti-tumor immune responses by promoting Th1 cytokine secretions and expansion of anti-tumor T cells [Citation117–133]. Interestingly, an Ad expressing the virulence factor neutrophil-activating protein from Helicobacter pylori (HP-NAP) can also enhance DC maturation and amplify priming of anti-cancer T cell responses [Citation134].

T cell activation is another target to enhance anti-tumor responses. It has been shown that immune co-stimulatory factors 4–1BBL, OX40L, GITRL, LIGHT, and ICOSL can provide potent activation of strong T cell responses [Citation135]. Several Ads have been armed with these and other co-stimulatory factors to enhance anti-tumor T cell responses [Citation136–143].

Cytokine stimulation for T cell activation is another important target for armed oncolytic Ads. Different types of cytokines dictate the expansion lifetime of T cells, and cytokines also determine the differentiation of T cells into either pro-tumor or anti-tumor responses. IL-2, IL-15, and IL-7 allow T cell expansion and memory T cell generation [Citation144]. IL-12, IL-18, and IFN-γ polarize T helper cells into the Th1 anti-tumor phenotype [Citation145]. Those immune payloads have been shown to have positive effects on anti-tumor responses for oncolytic Ad therapy in animal models or patients [Citation146–156].

8.2. Inhibiting cancer immune evasion

IFN-γ stimulation can induce MHC-I molecule expression on tumor cells to be recognized by CD8+ T cells [Citation96]. For targeting immune checkpoints, adenoviral expression of anti-CTLA-4 [Citation126], anti-PD-1/anti-PD-L1 or PD-1 decoy [Citation150,Citation157–163], and anti-CD47 or SIRPa decoy [Citation164,Citation165] has been shown to enhance anti-tumor immune responses. Anti-CTLA-4 and anti-PD1/PD-L1/PD-1 decoy target inhibitory signal to T cell responses, while anti-CD47/SIRPa decoy blocks ‘don’t eat me’ signal from tumor cells to macrophage or DCs [Citation166].

8.3. Disrupting physical barriers and increasing immune cell infiltration

Several components of ECM have to be broken down to enhance immune cell infiltration into tumors: collagen matrix, hyaluronan, tight junction, and extracellular DNA.

Decorin is a small proteoglycan that can interact with collagen to interrupt fibrogenesis [Citation167]. Oncolytic Ads expressing decorin demonstrated the ability to remodel tumor ECM and inhibit angiogenesis and tumor metastasis [Citation167–169]. Another payload, relaxin, is a peptide hormone that can degrade collagen matrix by up-regulating matrix metalloproteinases (MMPs) [Citation170]. Ad-relaxin has been shown to remodel tumor ECM to allow efficient viral spread, immune cell infiltration, and potentiate immune checkpoint inhibitor treatment [Citation170,Citation171]. Ad-MMP8 also has been used to break down ECM [Citation172].

Hyaluronan is an abundant glycosaminoglycan that supports the structure of ECM [Citation173]. Oncolytic Ad-hyaluronidase was shown to degrade hyaluronan in tumor ECM, promote viral spread and immune cell infiltration, and increase anti-tumor efficacy [Citation174–178].

Tight junctions of epithelial cells also serve as a barrier to effective infiltration of immune cells and viral infection [Citation179]. Yumul et al. demonstrated that oncolytic Ad5 expressing junction opener derived from Ad3 significantly improve anti-tumor efficacy compared to control Ad5 [Citation179].

Extracellular DNA in the tumor is released from the dead cells which can be physical barriers to the infiltration of immune cells. Tedcastle et al. compared CRAd encoding DNase I or hyaluronidase. They found that CRAd-DNase has better viral spread in the tumor compared to the empty CRAd control [Citation180].

Last, to actively increase immune cell infiltration, Ad expressing chemokines, such as CCL5, can be used to increase the T cell infiltration [Citation181].

It should be noted that one step toward the development of metastatic cancer is gaining the ability to break down ECM to spread. Along these lines, relaxin has been reported to have both pro-tumor and anti-tumor effects because MMPs can facilitate angiogenesis and tumor metastasis [Citation182]. To counter the effect of angiogenesis from remodeling ECM, Ad-encoded shVEGF can be used to down-regulated VEGF expression [Citation183].

Great care should be used in arming replication-competent viruses with potent proteases and other enzymes. If such viruses were not stringently restricted to cancer cells, they could produce considerable damage to other tissues as they could begin degrading structures that are used naturally to hold tissues together.

8.4. Inhibiting immune suppressive cells

Oncolytic Ads can also be used to combat immune suppressive cells or cytokines. TGF-β is a major suppressive cytokine in the TME to induce pro-tumor immune cells (MDSCs, Tregs, M2 TAMs, and CAFs). TGF-β traps are recombinant TGF-β binding proteins that can neutralize secreted TGF-β [Citation184]. Ads expressing TGF-β trap, decorin, or shRNATGF-β have been used to neutralize or inhibit TGF-β expression in TME [Citation185–188]. Ad encoding Bi-specific T-cell engager (BiTE) to deplete CAFs and M2 TAMs has been demonstrated to change tumor suppressive TME [Citation189,Citation190], while sCD200R1-IgG is an immune checkpoint inhibitor to block CD200 inhibitory function of CD200R1 on macrophages [Citation191].

Oncolytic viruses can also be used to combat metabolic pathways that help remodel the TME [Citation192]. Although we did not find a specific payload-armed oncolytic Ad that targets metabolic suppression in TME, Rivadeneira et al. have demonstrated that an oncolytic vaccinia virus expressing leptin, a metabolic hormone, can enhance the oxidative activities of tumor-infiltrating T cells in TME and thus retain the anti-tumor activity of T cells in the metabolic insufficient environments [Citation193]. Ads encoding leptin have been evaluated in the context of gene therapy against leptin deficiency-induced obesity [Citation194], but oncolytic Ads armed with leptin have not been studied.

9. Multiple-armed oncolytic Ads for targeting several immunotherapy barriers

Arming oncolytic Ads with one immune-stimulatory payload to activate immune cells can greatly boost anti-tumor immune responses. However, targeting only one set of immune receptors/signaling pathways may induce negative feedbacks that squelch robust immunotherapy [Citation113]. Given this, several groups have begun arming oncolytic Ads with multiple immune stimulatory payloads ().

9.1. The categories of immune payloads in multiple-armed onclytic Ads

By a literature survey (), the most common arming approach is always started with one payload from the ‘immune activation’ category and then in combination with payloads from either another factor of the ‘immune activation’ category that targets a different signaling pathway [Citation143,Citation156,Citation195–199], ‘blocking inhibitory signals’ [Citation188,Citation200–202], ‘promoting cell infiltration’ [Citation133,Citation171,Citation187,Citation203,Citation204] or ‘modulation of TME’ [Citation171,Citation183,Citation187,Citation188,Citation202,Citation205–208]. Although some multiple-armed oncolytic Ads are only armed with the payloads from the ‘immune activation’ category, the downstream effects of the activation of immune cells can actually increase the overall secretion of anti-tumor cytokines and chemokines and thus boost immune cell infiltration and change TME.

9.2. Approaches to increase transgene capacity for multiple-armed oncolytic adenoviruses

There is generally enough space in the Ad genome to allow one to express two transgenes in an E1-intact Ad. However, there is a limit to how many cDNAs can be squeezed into the fixed genome capacity of Ad vectors and still keep them replication competent. The construction of multi-armed oncolytic Ads can be challenging. Below are a few novel strategies to overcome Ad capacity limits.

9.2.1. Co-infection of replication-competent oncolytic Ad with helper-dependent adenovirus (HD-Ad) carrying multiple payloads

Helper-dependent Ads have all viral ORFs deleted [Citation194,Citation209,Citation210]. This makes HD-Ads less likely to provoke anti-Ad T cells allowing them to be used for gene therapy lasting at least 7 years in non-human primates [Citation211]. HD-Ads have also been used by many groups as gene-based vaccine platforms [Citation212–215] demonstrating their ability to not only hide themselves from the immune system but to also stimulate the immune system. While HD-Ads can be stealthy, ablation of all their genes means that they themselves cannot be oncolytic viruses. While this is true, the genomes of HD-Ads (and RD-Ads) can replicate in cells if they are partnered with another replicating helper Ad. For example, Masataka Suzukis group uses HD-Ad to carry multiple immunomodulatory molecules for targeting different aspects of immune pathways: chemokine, cytokine, immune checkpoint inhibitors, and T cell or NK cell engager molecules [Citation216]. When they co-infect HD-Ad with oncolytic Ad, HD-Ad can replicate and amplify transgenes with the viral protein from oncolytic Ads [Citation216].

9.2.2. Using microRNA (miRNA) or short hairpin RNA (shRNA) expression cassettes instead of protein-encoded transgenes to target inhibitory ligands

The application of RNA interference (RNAi) approaches for arming oncolytic Ads for immune modulation is still underdeveloped. miRNA or shRNA can also be either delivered directly or from infected cells to other non-infected cells through extracellular vesicles [Citation217]. miRNA or shRNA expression cassette also minimizes the insert size (100–200 bp) compared to a few thousand base pairs of genes encoding protein antagonists. For example, the shRNA TGF-beta2 expression cassette carried by oncolytic Ad is much smaller than the 1.2kb TGF-β trap protein [Citation188], though their functional efficacy was not directly compared.

9.2.3. Fusion of two immune payloads to reduce transgene size

Another approach to minimize transgene size is to fuse two functional transgenes into a smaller, bifunctional gene. For example, there are many immune checkpoints decoy and co-stimulatory gene fusion proteins, such as PD-1-4-1BBL, PD1-Fc-OX40L, SIRPa-Fc-CD40L fusion, or bispecific antibody that stimulates multiple co-stimulatory factors [Citation200,Citation218–220].

10. Expert opinion

Oncolytic Ads armed with immune stimulatory payloads are advancing the field of immunotherapy by overcoming immune suppressive TME with the combination of immune checkpoint inhibitors or CAR-T therapy [Citation143,Citation201,Citation205,Citation221–226]. Several promising oncolytic Ad candidates are currently evaluated in clinical trials (reviewed in [Citation227,Citation228]). CG0070, a human serotype 5 oncolytic Ad armed with GM-CSF, has been advanced to Phase III clinical trials for treating bladder cancer (anonymized). We expect more and more interesting combinations of immune modulation payloads to appear in the future along with the advances in cancer immunotherapy. We will see more strategic targeting of oncolytic Ad on multiple immunotherapy barriers: immune signaling priming and activation, tumor cell immune evasion mechanisms, immune cell infiltration barriers, and TME. Until now, the field still has great potential since the payload choices and their combination can be unlimited.

However, it could be technically challenging to evaluate the syngeneic effects of multiple payloads when more and more payloads (variables) are used in oncolytic Ads. It is because those immunological effects have to be tested in immunocompetent animal models. For example, to properly control the effect of each payload and their interactions, for 2 payloads armed Ad, it takes 5 groups of animals to compare (no treatment, Ad vector only, payload A, payload B, and payload A + B). For 4 payloads, the total number of animal groups is 17. Moreover, although local administration of oncolytic Ad and engineered transgene greatly reduce the systemic toxicity of payloads, such as IL-12 [Citation151,Citation153], with more and more immune stimulatory factors arming, we would expect a higher degree of toxicity caused by enhanced inflammation in the tumors. Therefore, an effective screening approach or model system would be required to properly evaluate both the syngeneic effect of payloads and immunological toxicities.

Not surprisingly, the choice of animal model can also affect the efficacy prediction of oncolytic Ad for human patients. The most available syngeneic tumor models are murine tumor models, but mouse models are usually not permissive for Ad [Citation229]. Thus, immunodeficient mouse xenograft models are usually used to evaluate oncolytic properties, while immunocompetent mouse tumor models are used to measure immune responses. That creates a problem that we cannot measure the interaction of oncolysis and immune stimulation at the same time in mouse models. Now, there are other immunocompetent animal models, such as the Syrian hamster tumor models that have some degree of permissiveness for Ad [Citation230] or humanized mouse models for oncolytic Ad evaluation [Citation231]. There were also several attempts to identify restriction factors for Ads in murine cells [Citation229,Citation232,Citation233]. Young et al. investigated and excluded the viral infection step, gene transcriptions, mRNA splicing, and viral replication as the restricted points for Ad to complete their viral life cycle. Instead, they identified that significant reduction of late viral mRNA translation in murine cell lines compared to human cell lines. And the expression of Ad L4-100 K protein in trans can partially restore the Ad late protein translation, although not fully recovered when compared to the level in human cell lines [Citation229]. If an Ad fully permissive, immunocompetent mouse model can be developed, it will greatly advance the field with better prediction of oncolytic Ad efficacy and with more available immunological tools on hand.

To discover novel immune payloads, the current method to construct recombinant Ads is also not practical for high-throughput screening. The traditional Ad cloning methods require a few months of work, including tedious recombination steps of plasmid in bacteria, a long time rescuing period, and a long viral amplification period [Citation234]. Gibson DNA assembly technology offers a faster cloning process for Ad DNA backbone in bacteria [Citation235,Citation236], but 1–2 week rescuing period after transfection of viral DNA is still needed probably because of the lack of terminal protein on linearized Ad backbone [Citation237]. A faster and simple Ad construction method for high-throughput screening will allow us to test variants of payloads quickly and facilitate new drug development.

Although not covered in this review, in order to systemically deliver oncolytic Ads into the majority of metastatic tumors in the body, there are several approaches for modifying Ad surface to reduce innate immune responses and to evade neutralizing antibodies [Citation238–240], as well as de-targeting and re-targeting approaches to enhance tumor-specificity [Citation241,Citation242]. We expect to see this angle of research further advancing the field of oncolytic adenoviral therapy.

Finally, it is important that these concepts be pursued in a safe and responsible manner. Arming replicating viruses with potent immunostimulatory proteins or ECM-degrading enzymes can help patients. However, it is very important to ensure that these viruses are tightly restricted to cancer cells and are leashed to prevent them from spreading to off-target tissues or even other people during the treatment. DNA-based oncolytics have extra control features that are unavailable to RNA viruses, including controlling transcriptional activation by promoter selections and the ability of the virus to push the cell into the S phase. Ads’ DNA polymerases also have higher fidelity than RNA polymerases, making them less likely to mutate the safety control features. Safety switches that are unique in Ads include making them conditionally-replication competent in cancer cells. This can be done by removing promiscuous viral E1 promoters or using cancer-specific promoters or by mutating early proteins like E1 protein (reviewed in [Citation1]). These restraints can be somewhat leaky if viruses are present at high multiplicities of infection. For example, even E1-deleted RD-Ads can replicate at high MOIs [Citation243]. However, not all cells can host Ad infection, and most cannot enter the S phase, so this leakiness may be manifested most strongly in cancer cells. A different type of safety switch for Ads is making them single-cycle adenoviruses (SC-Ad) [Citation63].

The immune system is another safety control and containment feature. Deleting E3 immune evasion genes makes the virus much less able to hide from the immune system. It may also be possible to use Ad with high seroprevalence in patients against certain Ad serotypes as a safety feature rather than a burden as demonstrated by William Wolds labs use of anti-Ad5 immunity to limit oncolytic Ad5 [Citation244]. In conclusion, great care is needed in arming Ad and other oncolytics to maximize efficacy while also maintaining safety.

Article highlights

  • Oncolytic adenoviruses (Ads) are highly immunogenic and potent vectors to deliver transgenes.

  • Adenoviral mRNA splicing is highly complex. Intrinsic viral splicing events should be considered when determining the location of transgene insertion.

  • Ad E3 and E1B genes can be partially or fully deleted to increase genome capacity, with consequences that need to be carefully evaluated.

  • Transgenes can be expressed by replacing existing viral ORFs, inserting autonomous expression cassettes, using internal ribosome entry sites (IRESs), translational truncation sequences (2A peptides), and/or by the insertion of exogenous splicing elements.

  • At least four tumor immunotherapy barriers need to be addressed:

    1. Insufficient immune signaling and priming

    2. Tumor cell-intrinsic immune evasion mechanisms

    3. Physical barriers to immune cell infiltration

    4. Immunosuppressive tumor microenvironments (TMEs).

  • Multiple immune stimulatory payloads can be loaded into oncolytic Ads to overcome each barrier.

This box summarizes key points contained in the article.

Declaration interest

M Barry is the Chief Scientific Officer for Adze Biotechnology, he does not own stock in this company. M Barry is co-inventor of single-cycle adenovirus which has been licensed to Tetherex Pharmaceuticals, and he does not own stock in this company. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

Reviewer disclosures

Peer reviewers on this manuscript have no relevant financial or other relationships to disclose.

Additional information

Funding

This project was supported by the Congressionally Directed Medical Research Program (W81XWH-19-1-0756), the Mayo Graduate School of Biomedical Sciences, and the Walter & Lucille Rubin Fund in Infectious Diseases Honoring Michael Camilleri, M.D. at Mayo Clinic.

References

  • Cattaneo R, Miest T, Shashkova EV, et al. Reprogrammed viruses as cancer therapeutics: targeted, armed and shielded. Nat Rev Microbiol. 2008 Jul;6(7):529–540.
  • Kelly E, Russell SJ. History of oncolytic viruses: genesis to genetic engineering. Mol Ther. 2007 Apr;15(4):651–659.
  • Tian Y, Xie D, Yang L. Engineering strategies to enhance oncolytic viruses in cancer immunotherapy. Signal Transduct Target Ther. 2022 Apr 6;7(1):117.
  • Davola ME, Mossman KL. Oncolytic viruses: how “lytic” must they be for therapeutic efficacy? Oncoimmunology. 2019;8(6):e1581528.
  • Greber UF, Flatt JW. Adenovirus entry: from infection to immunity. Annu Rev Virol. 2019 Sep 29; 6(1):177–197.
  • Reddy VS, Yu X, Barry MA. Refined capsid structure of human adenovirus D26 at 3.4 A resolution. Viruses. 2022 Feb 17; 14(2):414.
  • Lusky M. Good manufacturing practice production of adenoviral vectors for clinical trials. Hum Gene Ther. 2005 Mar;16(3):281–291.
  • Croyle MA, Cheng X, Sandhu A, et al. Development of novel formulations that enhance adenoviral-mediated gene expression in the lung in vitro and in vivo. Mol Ther. 2001 Jul;4(1):22–28.
  • Evans RK, Nawrocki DK, Isopi LA, et al. Development of stable liquid formulations for adenovirus-based vaccines. J Pharm Sci. 2004 Oct;93(10):2458–2475.
  • Bajrovic I, Schafer SC, Romanovicz DK, et al. Novel technology for storage and distribution of live vaccines and other biological medicines at ambient temperature. Sci Adv. 2020 Mar;6(10):eaau4819.
  • Kumru OS, Joshi SB, Smith DE, et al. Vaccine instability in the cold chain: mechanisms, analysis and formulation strategies. Biologicals. 2014 Sep;42(5):237–259.
  • Barefoot B, Thornburg NJ, Barouch DH, et al. Comparison of multiple vaccine vectors in a single heterologous prime-boost trial. Vaccine. 2008 Nov 11;26(48):6108–6118.
  • Davison AJ, Benko M, Harrach B. Genetic content and evolution of adenoviruses. J Gen Virol. 2003 Nov;84(Pt 11):2895–2908.
  • Nicklin SA, Wu E, Nemerow GR, et al. The influence of adenovirus fiber structure and function on vector development for gene therapy. Mol Ther. 2005 Sep;12(3):384–393.
  • Arnberg N. Adenovirus receptors: implications for tropism, treatment and targeting. Rev Med Virol. 2009 May;19(3):165–178.
  • Barry MA, Weaver EA, Chen CY. Mining the adenovirus “virome” for systemic oncolytics. Curr Pharm Biotechnol. 2012 Jul;13(9):1804–1808.
  • Parker AL, Waddington SN, Nicol CG, et al. Multiple vitamin K-dependent coagulation zymogens promote adenovirus-mediated gene delivery to hepatocytes. Blood. 2006 Oct 15;108(8):2554–2561.
  • Shayakhmetov DM, Gaggar A, Ni S, et al. Adenovirus binding to blood factors results in liver cell infection and hepatotoxicity. J Virol. 2005 Jun;79(12):7478–7491.
  • Xu Z, Tian J, Smith JS, et al. Clearance of adenovirus by Kupffer cells is mediated by scavenger receptors, natural antibodies, and complement. J Virol. 2008 Dec;82(23):11705–11713.
  • Xu Z, Qiu Q, Tian J, et al. Coagulation factor X shields adenovirus type 5 from attack by natural antibodies and complement. Nat Med. 2013 Apr;19(4):452–457.
  • Santra S, Sun Y, Korioth-Schmitz B, et al. Heterologous prime/boost immunizations of rhesus monkeys using chimpanzee adenovirus vectors. Vaccine. 2009 Sep 25;27(42):5837–5845.
  • Lemckert AA, Sumida SM, Holterman L, et al. Immunogenicity of heterologous prime-boost regimens involving recombinant adenovirus serotype 11 (Ad11) and Ad35 vaccine vectors in the presence of anti-ad5 immunity. J Virol. 2005 Aug;79(15):9694–9701.
  • Roberts DM, Nanda A, Havenga MJ, et al. Hexon-chimaeric adenovirus serotype 5 vectors circumvent pre-existing anti-vector immunity. Nature. 2006 May 11;441(7090):239–243.
  • Greber UF, Willetts M, Webster P, et al. Stepwise dismantling of adenovirus 2 during entry into cells. Cell. 1993 Nov 5;75(3):477–486.
  • Wickham TJ, Mathias P, Cheresh DA, et al. Integrins avb3 or avb5 promote adenovirus internalization but not virus attachment. Cell. 1993;73(2):309–319.
  • Huang S, Kamata T, Takada Y, et al. Adenovirus interaction with distinct integrins mediates separate events in cell entry and gene delivery to hematopoietic cells. J Virol. 1996;70(7):4502–4508.
  • Farley DC, Brown JL, Leppard KN. Activation of the early-late switch in adenovirus type 5 major late transcription unit expression by L4 gene products. J Virol. 2004 Feb;78(4):1782–1791.
  • Fessler SP, Young CS. Control of adenovirus early gene expression during the late phase of infection. J Virol. 1998 May;72(5):4049–4056.
  • Crisostomo L, Soriano AM, Mendez M, et al. Temporal dynamics of adenovirus 5 gene expression in normal human cells. PLoS One. 2019;14(1):e0211192.
  • Tormanen H, Backstrom E, Carlsson A, et al. L4-33K, an adenovirus-encoded alternative RNA splicing factor. J Biol Chem. 2006 Dec 1;281(48):36510–36517
  • Morris SJ, Scott GE, Leppard KN. Adenovirus late-phase infection is controlled by a novel L4 promoter. J Virol. 2010 Jul;84(14):7096–7104.
  • Turner MA, Middha S, Hofherr SE, et al. Comparison of the life cycles of genetically distant species C and species D human adenoviruses Ad6 and Ad26 in human cells. J Virol. 2015 Dec;89(24):12401–12417.
  • Jakobsson AW, Segerman B, Wallerman O, et al. The human Adenovirus 2 transcriptome: an amazing complexity of alternatively spliced mRNAs. J Virol. 2021;95(4):e01869–20. ** An important study demonstrating the complexity of mRNA splicing of adenoviruses by long-read next-generation sequencing.
  • Senac JS, Doronin K, Russell SJ, et al. Infection and killing of multiple myeloma by adenoviruses. Hum Gene Ther. 2010 Feb;21(2):179–190.
  • Zhao H, Granberg F, Pettersson U. How adenovirus strives to control cellular gene expression. Virology. 2007 Jul 5; 363(2):357–375.
  • Wu L, Zhou P, Ge X, et al. Deep RNA sequencing reveals complex transcriptional landscape of a bat adenovirus. J Virol. 2013 Jan;87(1):503–511.
  • Donovan-Banfield I, Turnell AS, Hiscox JA, et al. Deep splicing plasticity of the human adenovirus type 5 transcriptome drives virus evolution. Commun Biol. 2020 Mar 13;3(1):124.
  • Bett AJ, Prevec L, Graham FL. Packaging capacity and stability of human adenovirus type 5 vectors. J Virol. 1993 Oct;67(10):5911–5921.
  • Lichtenstein DL, Toth K, Doronin K, et al. Functions and mechanisms of action of the adenovirus E3 proteins. Int Rev Immunol. 2004 Jan-Apr;23(1–2):75–111.
  • Horwitz MS. Function of adenovirus E3 proteins and their interactions with immunoregulatory cell proteins. J Gene Med. 2004 Feb;6(1):S172–83.
  • Robinson CM, Singh G, Lee JY, et al. Molecular evolution of human adenoviruses. Sci Rep. 2013;3(1):1812.
  • Hawkins LK, Wold WSM. Wold WS. A 12,500 MW protein is coded by region E3 of adenovirus. Virology. 1992 Jun;188(2):486–494.
  • Moise AR, Grant JR, Vitalis TZ, et al. Adenovirus E3-6.7K maintains calcium homeostasis and prevents apoptosis and arachidonic acid release. J Virol. 2002 Feb;76(4):1578–1587.
  • Lichtenstein DL, Doronin K, Toth K, et al. Adenovirus E3-6.7K protein is required in conjunction with the E3-RID protein complex for the internalization and degradation of TRAIL receptor 2. J Virol. 2004 Nov;78(22):12297–12307.
  • Burgert HG, Maryanski JL, Kvist S. “E3/19K” protein of adenovirus type 2 inhibits lysis of cytolytic T lymphocytes by blocking cell-surface expression of histocompatibility class I antigens. Proc. Natl. Acad. Sci. USA. 1987;84(5):1356–1360.
  • Georgi F, Greber UF. The adenovirus death protein - a small membrane protein controls cell lysis and disease. FEBS Lett. 2020 Jun;594(12):1861–1878.
  • Robertson MG, Eidenschink BB, Iguchi E, et al. Cancer imaging and therapy utilizing a novel NIS-expressing adenovirus: the role of adenovirus death protein deletion. Mol Ther Oncolytics. 2021 Mar 26;20:659–668. 10.1016/j.omto.2021.03.002.
  • Shisler J, Yang C, Walter B, et al. The adenovirus E3-10.4K/14.5K complex mediates loss of cell surface Fas (CD95) and resistance to Fas-induced apoptosis. J Virol. 1997 Nov;71(11):8299–8306.
  • Tollefson AE, Hermiston TW, Lichtenstein DL, et al. Forced degradation of Fas inhibits apoptosis in adenovirus-infected cells. Nature. 1998 Apr 16;392(6677):726–730.
  • Lichtenstein DL, Krajcsi P, Esteban DJ, et al. Adenovirus RIDbeta subunit contains a tyrosine residue that is critical for RID-mediated receptor internalization and inhibition of Fas- and TRAIL-induced apoptosis. J Virol. 2002 Nov;76(22):11329–11342.
  • Horton TM, Ranheim TS, Aquino L, et al. Adenovirus E3 14.7K protein functions in the absence of other adenovirus proteins to protect transfected cells from tumor necrosis factor cytolysis. J Virol. 1991 May;65(5):2629–2639.
  • Schneider-Brachert W, Tchikov V, Merkel O, et al. Inhibition of TNF receptor 1 internalization by adenovirus 14.7K as a novel immune escape mechanism. J Clin Invest. 2006 Nov;116(11):2901–2913.
  • Tollefson AE, Ying B, Doronin K, et al. Identification of a new human adenovirus protein encoded by a novel late l-strand transcription unit. J Virol. 2007 Dec;81(23):12918–12926.
  • Wang Y, Hallden G, Hill R, et al. E3 gene manipulations affect oncolytic adenovirus activity in immunocompetent tumor models. Nat Biotechnol. 2003 Nov;21(11):1328–1335.
  • Bortolanza S, Bunuales M, Alzuguren P, et al. Deletion of the E3-6.7K/gp19K region reduces the persistence of wild-type adenovirus in a permissive tumor model in Syrian hamsters. Cancer Gene Ther. 2009 Sep;16(9):703–712.
  • Bos JL, Polder LJ, Bernards R, et al. The 2.2 kb E1b mRNA of human Ad12 and Ad5 codes for two tumor antigens starting at different AUG triplets. Cell. 1981 Nov;27(1 Pt 2):121–131.
  • Han J, Sabbatini P, Perez D, et al. The E1B 19K protein blocks apoptosis by interacting with and inhibiting the p53-inducible and death-promoting Bax protein. Genes Dev. 1996 Feb 15;10(4):461–477.
  • Hidalgo P, Ip WH, Dobner T, et al. The biology of the adenovirus E1B 55K protein. FEBS Lett. 2019 Dec;593(24):3504–3517.
  • Doloff JC, Waxman DJ, Jounaidi Y. Human telomerase reverse transcriptase promoter-driven oncolytic adenovirus with E1B-19 kDa and E1B-55 kDa Gene deletions. Hum Gene Ther. 2008 Dec;19(12):1383–1399.
  • Cheng PH, Wechman SL, McMasters KM, et al. Oncolytic replication of E1b-deleted adenoviruses. Viruses. 2015 Nov 6;7(11):5767–5779.
  • Sauthoff H, Heitner S, Rom WN, et al. Deletion of the adenoviral E1b-19kD gene enhances tumor cell killing of a replicating adenoviral vector. Hum Gene Ther. 2000 Feb 10;11(3):379–388.
  • Harrison D, Sauthoff H, Heitner S, et al. Wild-type adenovirus decreases tumor xenograft growth, but despite viral persistence complete tumor responses are rarely achieved–deletion of the viral E1b-19-kD gene increases the viral oncolytic effect. Hum Gene Ther. 2001 Jul 1;12(10):1323–1332.
  • Crosby CM, Barry MA. IIIa deleted adenovirus as a single-cycle genome replicating vector. Virology. 2014 Aug;462-463:158–165.
  • Crosby CM, Barry MA. Transgene expression and host cell responses to replication-defective, single-cycle, and replication-competent adenovirus vectors. Genes (Basel). 2017 Feb 18; 8(2):79.
  • Farrera-Sal M, Fillat C, Alemany R. Effect of transgene location, transcriptional control elements and transgene features in armed oncolytic adenoviruses. Cancers (Basel). 2020 Apr 23; 12(4):1034.
  • Kretschmer PJ, Jin F, Chartier C, et al. Development of a transposon-based approach for identifying novel transgene insertion sites within the replicating adenovirus. Mol Ther. 2005 Jul;12(1):118–127.
  • Jin F, Kretschmer PJ, Hermiston TW. Identification of novel insertion sites in the Ad5 genome that utilize the Ad splicing machinery for therapeutic gene expression. Mol Ther. 2005 Dec;12(6):1052–1063.
  • Wang M, Marin A. Characterization and prediction of alternative splice sites. Gene. 2006 Feb 1; 366(2):219–227.
  • Cartegni L, Wang J, Zhu Z, et al. ESEfinder: a web resource to identify exonic splicing enhancers. Nucleic Acids Res. 2003 Jul 1;31(13):3568–3571.
  • Hawkins LK, Hermiston T. Gene delivery from the E3 region of replicating human adenovirus: evaluation of the E3B region. Gene Ther. 2001 Aug;8(15):1142–1148.
  • Hawkins LK, Hermiston TW. Gene delivery from the E3 region of replicating human adenovirus: evaluation of the ADP region. Gene Ther. 2001 Aug;8(15):1132–1141.
  • Hawkins LK, Johnson L, Bauzon M, et al. Gene delivery from the E3 region of replicating human adenovirus: evaluation of the 6.7 K/gp19 K region. Gene Ther. 2001 Aug;8(15):1123–1131.
  • Bauzon M, Castro D, Karr M, et al. Multigene expression from a replicating adenovirus using native viral promoters. Mol Ther. 2003 Apr;7(4):526–534.
  • Shashkova EV, Spencer JF, Wold WS, et al. Targeting interferon-alpha increases antitumor efficacy and reduces hepatotoxicity of E1A-mutated spread-enhanced oncolytic adenovirus. Mol Ther. 2007 Mar;15(3):598–607.
  • Shaul O. How introns enhance gene expression. Int J Biochem Cell Biol. 2017 Oct;91(Pt B):145–155.
  • Martinez-Salas E, Francisco-Velilla R, Fernandez-Chamorro J, et al. Insights into structural and mechanistic features of viral IRES elements. Front Microbiol. 2017;8:2629.
  • Rivera AA, Wang M, Suzuki K, et al. Mode of transgene expression after fusion to early or late viral genes of a conditionally replicating adenovirus via an optimized internal ribosome entry site in vitro and in vivo. Virology. 2004 Mar 1;320(1):121–134.
  • Sharma P, Yan F, Doronina VA, et al. 2A peptides provide distinct solutions to driving stop-carry on translational recoding. Nucleic Acids Res. 2012 Apr;40(7):3143–3151.
  • Liu Z, Chen O, Wall JBJ, et al. Systematic comparison of 2A peptides for cloning multi-genes in a polycistronic vector. Sci Rep. 2017 May 19;7(1):2193.
  • Funston GM, Kallioinen SE, de Felipe P, et al. Expression of heterologous genes in oncolytic adenoviruses using picornaviral 2A sequences that trigger ribosome skipping. J Gen Virol. 2008 Feb;89(Pt 2):389–396.
  • Quirin C, Rohmer S, Fernandez-Ulibarri I, et al. Selectivity and efficiency of late transgene expression by transcriptionally targeted oncolytic adenoviruses are dependent on the transgene insertion strategy. Hum Gene Ther. 2011 Apr;22(4):389–404.
  • Hagihara Y, Sakamoto A, Tokuda T, et al. Photoactivatable oncolytic adenovirus for optogenetic cancer therapy. Cell Death Dis. 2020 Jul 23;11(7):570.
  • Muhlemann O, Yue BG, Petersen-Mahrt S, et al. A novel type of splicing enhancer regulating adenovirus pre-mRNA splicing. Mol Cell Biol. 2000 Apr;20(7):2317–2325.
  • Carette JE, Graat HC, Schagen FH, et al. Replication-dependent transgene expression from a conditionally replicating adenovirus via alternative splicing to a heterologous splice-acceptor site. J Gene Med. 2005 Aug;7(8):1053–1062.
  • Fuerer C, Iggo R. 5-Fluorocytosine increases the toxicity of Wnt-targeting replicating adenoviruses that express cytosine deaminase as a late gene. Gene Ther. 2004 Jan;11(2):142–151.
  • Berg M, Difatta J, Hoiczyk E, et al. Viable adenovirus vaccine prototypes: high-level production of a papillomavirus capsid antigen from the major late transcriptional unit. Proc Natl Acad Sci USA. 2005 Mar 22;102(12):4590–4595.
  • Fernandez-Ulibarri I, Hammer K, Arndt MA, et al. Genetic delivery of an immunoRNase by an oncolytic adenovirus enhances anticancer activity. Int J Cancer. 2015 May 1;136(9):2228–2240.
  • Robinson M, Ge Y, Ko D, et al. Comparison of the E3 and L3 regions for arming oncolytic adenoviruses to achieve a high level of tumor-specific transgene expression [Comparative Study]. Cancer Gene Ther. 2008 Jan;15(1):9–17.
  • Farrera-Sal M, de Sostoa J, Nunez-Manchon E, et al. Arming oncolytic adenoviruses: effect of insertion site and splice acceptor on transgene expression and viral fitness. Int J Mol Sci. 2020 Jul 21;21(14):5158.
  • Philip M, Schietinger A. CD8(+) T cell differentiation and dysfunction in cancer. Nat Rev Immunol. 2022 Apr;22(4):209–223.
  • Wan PK, Ryan AJ, Seymour LW. Beyond cancer cells: targeting the tumor microenvironment with gene therapy and armed oncolytic virus. Mol Ther. 2021 May 5; 29(5):1668–1682.
  • Boagni DA, Ravirala D, Zhang SX. Current strategies in engaging oncolytic viruses with antitumor immunity. Mol Ther Oncolytics. 2021 Sep 24; 22:98–113.
  • Rahman MM, McFadden G. Oncolytic viruses: newest frontier for cancer immunotherapy. Cancers (Basel). 2021 Oct 29; 13(21):5452.
  • Xing Y, Hogquist KA. T-cell tolerance: central and peripheral. Cold Spring Harb Perspect Biol. 2012 Jun 1; 4(6):a006957.
  • Greten FR, Grivennikov SI. Inflammation and cancer: triggers, mechanisms, and consequences. Immunity. 2019 Jul 16; 51(1):27–41.
  • Dhatchinamoorthy K, Colbert JD, Rock KL. Cancer immune evasion through loss of MHC class I antigen presentation. Front Immunol. 2021;12:636568.
  • Wolf Y, Samuels Y. Intratumor heterogeneity and antitumor immunity shape one another bidirectionally. Clin Cancer Res. 2022 Jul 15; 28(14):2994–3001.
  • Archilla-Ortega A, Domuro C, Martin-Liberal J, et al. Blockade of novel immune checkpoints and new therapeutic combinations to boost antitumor immunity. J Exp Clin Cancer Res. 2022 Feb 14;41(1):62.
  • Lubbers J, Rodriguez E, van Kooyk Y. Modulation of immune tolerance via siglec-sialic acid interactions. Front Immunol. 2018;9:2807.
  • Follain G, Herrmann D, Harlepp S, et al. Fluids and their mechanics in tumour transit: shaping metastasis. Nat Rev Cancer. 2020 Feb;20(2):107–124.
  • Simsek H, Klotzsch E. The solid tumor microenvironment-breaking the barrier for T cells: how the solid tumor microenvironment influences T cells: how the solid tumor microenvironment influences T cells. Bioessays. 2022 Jun;44(6):e2100285.
  • Turley SJ, Cremasco V, Astarita JL. Immunological hallmarks of stromal cells in the tumour microenvironment. Nat Rev Immunol. 2015 Nov;15(11):669–682.
  • Sackstein R, Schatton T, Barthel SR. T-lymphocyte homing: an underappreciated yet critical hurdle for successful cancer immunotherapy. Lab Invest. 2017 Jun;97(6):669–697.
  • Anderson NM, Simon MC. The tumor microenvironment. Curr Biol. 2020 Aug 17; 30(16):R921–R925.
  • DePeaux K, Delgoffe GM. Metabolic barriers to cancer immunotherapy. Nat Rev Immunol. 2021 Dec;21(12):785–797.
  • Watson MJ, Delgoffe GM. Fighting in a wasteland: deleterious metabolites and antitumor immunity. J Clin Invest. 2022 Jan 18; 132(2):e148549.
  • Aldhamen YA, Appledorn DM, Seregin SS, et al. Expression of the SLAM family of receptors adapter EAT-2 as a novel strategy for enhancing beneficial immune responses to vaccine antigens. J Immunol. 2011 Jan 15;186(2):722–732.
  • O’Connell P, Blake MK, Pepelyayeva Y, et al. Adenoviral delivery of an immunomodulatory protein to the tumor microenvironment controls tumor growth. Mol Ther Oncolytics. 2022 Mar 17;24:180–193.
  • Ramachandran M, Yu D, Wanders A, et al. An infection-enhanced oncolytic adenovirus secreting H. pylori neutrophil-activating protein with therapeutic effects on neuroendocrine tumors. Mol Ther. 2013 Nov;21(11):2008–2018.
  • Liikanen I, Basnet S, Quixabeira DCA, et al. Oncolytic adenovirus decreases the proportion of TIM-3(+) subset of tumor-infiltrating CD8(+) T cells with correlation to improved survival in patients with cancer. J Immunother Cancer. 2022 Feb;10(2):e003490.
  • Raman SS, Hecht JR, Chan E. Talimogene laherparepvec: review of its mechanism of action and clinical efficacy and safety. Immunotherapy. 2019 Jun;11(8):705–723.
  • Kumar A, Taghi Khani A, Sanchez Ortiz A, et al. GM-CSF: a double-edged sword in cancer immunotherapy. Front Immunol. 2022;13:901277.
  • Mandai M, Hamanishi J, Abiko K, et al. Dual Faces of IFNgamma in cancer progression: a role of PD-L1 induction in the determination of pro- and antitumor immunity. Clin Cancer Res. 2016 May 15;22(10):2329–2334.
  • Ge Y, Wang H, Ren J, et al. Oncolytic vaccinia virus delivering tethered IL-12 enhances antitumor effects with improved safety. J Immunother Cancer. 2020 Mar;8(1):e000710.
  • Blair TC, Bambina S, Alice AF, et al. Dendritic cell maturation defines immunological responsiveness of tumors to radiation therapy. J Immunol. 2020 Jun 15;204(12):3416–3424.
  • Patente TA, Pinho MP, Oliveira AA, et al. Human dendritic cells: their heterogeneity and clinical application potential in cancer immunotherapy. Front Immunol. 2018;9:3176.
  • Diaconu I, Cerullo V, Hirvinen ML, et al. Immune response is an important aspect of the antitumor effect produced by a CD40L-encoding oncolytic adenovirus. Cancer Res. 2012 May 1;72(9):2327–2338.
  • Pesonen S, Diaconu I, Kangasniemi L, et al. Oncolytic immunotherapy of advanced solid tumors with a CD40L-expressing replicating adenovirus: assessment of safety and immunologic responses in patients. Cancer Res. 2012 Apr 1;72(7):1621–1631.
  • Yang YF, Xue SY, Lu ZZ, et al. Antitumor effects of oncolytic adenovirus armed with PSA-IZ-CD40L fusion gene against prostate cancer. Gene Ther. 2014 Aug;21(8):723–731.
  • Eriksson E, Moreno R, Milenova I, et al. Activation of myeloid and endothelial cells by CD40L gene therapy supports T-cell expansion and migration into the tumor microenvironment. Gene Ther. 2017 Feb;24(2):92–103.
  • Hirvinen M, Rajecki M, Kapanen M, et al. Immunological effects of a tumor necrosis factor alpha-armed oncolytic adenovirus. Hum Gene Ther. 2015 Mar;26(3):134–144.
  • LaRocca CJ, Han J, Gavrikova T, et al. Oncolytic adenovirus expressing interferon alpha in a syngeneic Syrian hamster model for the treatment of pancreatic cancer. Surgery. 2015 May;157(5):888–898.
  • LaRocca CJ, Salzwedel AO, Sato-Dahlman M, et al. Interferon alpha-expressing oncolytic adenovirus for treatment of esophageal adenocarcinoma. Ann Surg Oncol. 2021 Dec;28(13):8556–8564.
  • Burke JM, Lamm DL, Meng MV, et al. A first in human phase 1 study of CG0070, a GM-CSF expressing oncolytic adenovirus, for the treatment of nonmuscle invasive bladder cancer. J Urol. 2012 Dec;188(6):2391–2397.
  • Kanerva A, Nokisalmi P, Diaconu I, et al. Antiviral and antitumor T-cell immunity in patients treated with GM-CSF-coding oncolytic adenovirus. Clin Cancer Res. 2013 May 15;19(10):2734–2744.
  • Du T, Shi G, Li YM, et al. Tumor-specific oncolytic adenoviruses expressing granulocyte macrophage colony-stimulating factor or anti-CTLA4 antibody for the treatment of cancers. Cancer Gene Ther. 2014 Aug;21(8):340–348.
  • Dobbins GC, Ugai H, Curiel DT, et al. A multi targeting conditionally replicating adenovirus displays enhanced oncolysis while maintaining expression of Immunotherapeutic agents. PLoS One. 2015;10(12):e0145272.
  • Vassilev L, Ranki T, Joensuu T, et al. Repeated intratumoral administration of ONCOS-102 leads to systemic antitumor CD8(+) T-cell response and robust cellular and transcriptional immune activation at tumor site in a patient with ovarian cancer. Oncoimmunology. 2015 Jul;4(7):e1017702.
  • Wang L, Zheng J, He Q, et al. TOA02, a recombinant adenovirus with tumor-specific granulocyte macrophage colony-stimulating factor expression, has limited biodistribution and low toxicity in rhesus monkeys. Hum Gene Ther Methods. 2015 Apr;26(2):62–70.
  • Bramante S, Koski A, Liikanen I, et al. Oncolytic virotherapy for treatment of breast cancer, including triple-negative breast cancer. Oncoimmunology. 2016 Feb;5(2):e1078057.
  • Moesta AK, Cooke K, Piasecki J, et al. Local Delivery of OncoVEX(mGM-CSF) generates systemic antitumor immune responses enhanced by cytotoxic T-lymphocyte-associated protein blockade. Clin Cancer Res. 2017 Oct 15;23(20):6190–6202.
  • Kuryk L, Moller AW, Garofalo M, et al. Antitumor-specific T-cell responses induced by oncolytic adenovirus ONCOS-102 (AdV5/3-D24-GM-CSF) in peritoneal mesothelioma mouse model. J Med Virol. 2018 Oct;90(10):1669–1673.
  • Su Y, Li J, Ji W, et al. Triple-serotype chimeric oncolytic adenovirus exerts multiple synergistic mechanisms against solid tumors. J Immunother Cancer. 2022 May;10(5):e004691.
  • Ramachandran M, Jin C, Yu D, et al. Vector-encoded Helicobacter pylori neutrophil-activating protein promotes maturation of dendritic cells with Th1 polarization and improved migration. J Immunol. 2014 Sep 1;193(5):2287–2296.
  • Kober J, Leitner J, Klauser C, et al. The capacity of the TNF family members 4-1BBL, OX40L, CD70, GITRL, CD30L and LIGHT to costimulate human T cells. Eur J Immunol. 2008 Oct;38(10):2678–2688.
  • Martinez-Perez AG, Perez-Trujillo JJ, Garza-Morales R, et al. An oncolytic adenovirus encoding SA-4-1BBL adjuvant fused to HPV-16 E7 antigen produces a specific antitumor effect in a cancer mouse Model. Vaccines (Basel). 2021 Feb 12;9(2):149.
  • Laspidea V, Puigdelloses M, Labiano S, et al. Exploiting 4-1BB immune checkpoint to enhance the efficacy of oncolytic virotherapy for diffuse intrinsic pontine gliomas. JCI Insight. 2022 Apr 8;7(7):e154812.
  • Martinez-Velez N, Laspidea V, Zalacain M, et al. Local treatment of a pediatric osteosarcoma model with a 4-1BBL armed oncolytic adenovirus results in an antitumor effect and leads to immune memory. Mol Cancer Ther. 2022 Mar 1;21(3):471–480.
  • Jiang H, Rivera-Molina Y, Gomez-Manzano C, et al. Oncolytic adenovirus and tumor-targeting immune modulatory therapy improve autologous cancer vaccination. Cancer Res. 2017 Jul 15;77(14):3894–3907.
  • Andarini S, Kikuchi T, Nukiwa M, et al. Adenovirus vector-mediated in vivo gene transfer of OX40 ligand to tumor cells enhances antitumor immunity of tumor-bearing hosts. Cancer Res. 2004 May 1;64(9):3281–3287.
  • Rivera-Molina Y, Jiang H, Fueyo J, et al. GITRL-armed Delta-24-RGD oncolytic adenovirus prolongs survival and induces anti-glioma immune memory. Neurooncol Adv. 2019 May-Dec;1(1):vdz009.
  • Dai S, Lv Y, Xu W, et al. Oncolytic adenovirus encoding LIGHT (TNFSF14) inhibits tumor growth via activating anti-tumor immune responses in 4T1 mouse mammary tumor model in immune competent syngeneic mice. Cancer Gene Ther. 2020 Dec;27(12):923–933.
  • Garofalo M, Bertinato L, Staniszewska M, et al. Combination therapy of novel oncolytic adenovirus with anti-PD1 resulted in enhanced anti-cancer effect in syngeneic immunocompetent melanoma mouse model. Pharmaceutics. 2021 Apr 14;13(4):547.
  • Schluns KS, Lefrancois L. Cytokine control of memory T-cell development and survival. Nat Rev Immunol. 2003 Apr;3(4):269–279.
  • Yoshimoto T, Takeda K, Tanaka T, et al. IL-12 up-regulates IL-18 receptor expression on T cells, Th1 cells, and B cells: synergism with IL-18 for IFN-gamma production. J Immunol. [1998 Oct 1];161(7):3400–3407.
  • Santos JM, Havunen R, Siurala M, et al. Adenoviral production of interleukin-2 at the tumor site removes the need for systemic postconditioning in adoptive cell therapy. Int J Cancer. 2017 Oct 1;141(7):1458–1468.
  • Quixabeira DCA, Zafar S, Santos JM, et al. Oncolytic adenovirus coding for a variant interleukin 2 (vIL-2) cytokine re-programs the tumor microenvironment and confers enhanced tumor control. Front Immunol. 2021;12:674400.
  • Huang J, Zheng M, Zhang Z, et al. Interleukin-7-loaded oncolytic adenovirus improves CAR-T cell therapy for glioblastoma. Cancer Immunol Immunother. 2021 Sep;70(9):2453–2465.
  • Poutou J, Bunuales M, Gonzalez-Aparicio M, et al. Safety and antitumor effect of oncolytic and helper-dependent adenoviruses expressing interleukin-12 variants in a hamster pancreatic cancer model. Gene Ther. 2015 Sep;22(9):696–706.
  • Rosewell Shaw A, Porter CE, Watanabe N, et al. Adenovirotherapy delivering cytokine and checkpoint inhibitor augments CAR T cells against metastatic head and neck cancer. Mol Ther. 2017 Nov 1;25(11):2440–2451.
  • Wang P, Li X, Wang J, et al. Re-designing Interleukin-12 to enhance its safety and potential as an anti-tumor immunotherapeutic agent. Nat Commun. 2017 Nov 9;8(1):1395.
  • Zhang Z, Zhang C, Miao J, et al. A tumor-targeted replicating oncolytic adenovirus ad-TD-nsIL12 as a promising therapeutic agent for human esophageal squamous cell carcinoma. Cells. 2020 Nov 10;9(11):2438.
  • Zalacain M, Bunuales M, Marrodan L, et al. Local administration of IL-12 with an HC vector results in local and metastatic tumor control in pediatric osteosarcoma. Mol Ther Oncolytics. 2021 Mar 26;20:23–33.
  • Yan Y, Li S, Jia T, et al. Combined therapy with CTL cells and oncolytic adenovirus expressing IL-15-induced enhanced antitumor activity. Tumour Biol. 2015 Jun;36(6):4535–4543.
  • Zhang Q, Zhang J, Tian Y, et al. Efficacy of a novel double-controlled oncolytic adenovirus driven by the Ki67 core promoter and armed with IL-15 against glioblastoma cells. Cell Biosci. 2020;10(1):124.
  • Choi IK, Lee JS, Zhang SN, et al. Oncolytic adenovirus co-expressing IL-12 and IL-18 improves tumor-specific immunity via differentiation of T cells expressing IL-12Rbeta2 or IL-18Ralpha. Gene Ther. 2011 Sep;18(9):898–909.
  • Bunuales M, Ballesteros-Briones MC, Gonzalez-Aparicio M, et al. Adenovirus-mediated inducible expression of a PD-L1 blocking antibody in combination with macrophage depletion improves survival in a mouse model of peritoneal carcinomatosis. Int J Mol Sci. 2021 Apr 17;22(8):4176.
  • Hamdan F, Ylosmaki E, Chiaro J, et al. Novel oncolytic adenovirus expressing enhanced cross-hybrid IgGA Fc PD-L1 inhibitor activates multiple immune effector populations leading to enhanced tumor killing in vitro, in vivo and with patient-derived tumor organoids. J Immunother Cancer. 2021 Aug;9(8):e003000.
  • Rosewell Shaw A, Porter CE, Yip T, et al. Oncolytic adeno-immunotherapy modulates the immune system enabling CAR T-cells to cure pancreatic tumors. Commun Biol. 2021 Mar 19;4(1):368.
  • Zhang H, Zhang Y, Dong J, et al. Recombinant adenovirus expressing the fusion protein PD1PVR improves CD8(+) T cell-mediated antitumor efficacy with long-term tumor-specific immune surveillance in hepatocellular carcinoma. Cell Oncol (Dordr). 2021 Dec;44(6):1243–1255.
  • Vitale M, Scialo F, Passariello M, et al. Oncolytic adenoviral vector-mediated expression of an anti-PD-L1-scFv improves anti-tumoral efficacy in a melanoma mouse model. Front Oncol. 2022;12:902190.
  • Zhou P, Wang X, Xing M, et al. Intratumoral delivery of a novel oncolytic adenovirus encoding human antibody against PD-1 elicits enhanced antitumor efficacy. Mol Ther Oncolytics. 2022 Jun 16;25:236–248. 10.1016/j.omto.2022.04.007.
  • Tanoue K, Rosewell Shaw A, Watanabe N, et al. Armed oncolytic adenovirus-expressing PD-L1 mini-body enhances antitumor effects of chimeric antigen receptor T cells in solid tumors. Cancer Res. 2017 Apr 15;77(8):2040–2051.
  • Huang Y, Lv SQ, Liu PY, et al. A SIRPalpha-Fc fusion protein enhances the antitumor effect of oncolytic adenovirus against ovarian cancer. Mol Oncol. 2020 Mar;14(3):657–668.
  • Zhang B, Shu Y, Hu S, et al. In situ tumor vaccine expressing anti-CD47 antibody enhances antitumor immunity. Front Oncol. 2022;12:897561.
  • Takimoto CH, Chao MP, Gibbs C, et al. The macrophage ‘do not eat me’ signal, CD47, is a clinically validated cancer immunotherapy target. Ann Oncol. 2019 Mar 1;30(3):486–489.
  • Choi IK, Lee YS, Yoo JY, et al. Effect of decorin on overcoming the extracellular matrix barrier for oncolytic virotherapy. Gene Ther. 2010 Feb;17(2):190–201.
  • Xu W, Neill T, Yang Y, et al. The systemic delivery of an oncolytic adenovirus expressing decorin inhibits bone metastasis in a mouse model of human prostate cancer. Gene Ther. 2015 Mar;22(3):247–256.
  • Li Y, Hong J, Oh JE, et al. Potent antitumor effect of tumor microenvironment-targeted oncolytic adenovirus against desmoplastic pancreatic cancer. Int J Cancer. 2018 Jan 15;142(2):392–413.
  • Kim JH, Lee YS, Kim H, et al. Relaxin expression from tumor-targeting adenoviruses and its intratumoral spread, apoptosis induction, and efficacy. J Natl Cancer Inst. 2006 Oct 18;98(20):1482–1493.
  • Jung BK, Ko HY, Kang H, et al. Relaxin-expressing oncolytic adenovirus induces remodeling of physical and immunological aspects of cold tumor to potentiate PD-1 blockade. J Immunother Cancer. 2020 Aug;8(2):e000763.
  • Cheng J, Sauthoff H, Huang Y, et al. Human matrix metalloproteinase-8 gene delivery increases the oncolytic activity of a replicating adenovirus. Mol Ther. 2007 Nov;15(11):1982–1990.
  • Garantziotis S, Savani RC. Hyaluronan biology: a complex balancing act of structure, function, location and context. Matrix Biol. 2019 May;78-79:1–10.
  • Rodriguez-Garcia A, Gimenez-Alejandre M, Rojas JJ, et al. Safety and efficacy of VCN-01, an oncolytic adenovirus combining fiber HSG-binding domain replacement with RGD and hyaluronidase expression. Clin Cancer Res. 2015 Mar 15;21(6):1406–1418.
  • Bazan-Peregrino M, Garcia-Carbonero R, Laquente B, et al. VCN-01 disrupts pancreatic cancer stroma and exerts antitumor effects. J Immunother Cancer. 2021 Nov;9(11):e003254.
  • Farrera-Sal M, Moreno R, Mato-Berciano A, et al. Hyaluronidase expression within tumors increases virotherapy efficacy and T cell accumulation. Mol Ther Oncolytics. 2021 Sep 24;22:27–35. 10.1016/j.omto.2021.05.009.
  • Kiyokawa J, Kawamura Y, Ghouse SM, et al. Modification of extracellular matrix enhances oncolytic adenovirus immunotherapy in glioblastoma. Clin Cancer Res. 2021 Feb 1;27(3):889–902.
  • Mato-Berciano A, Morgado S, Maliandi MV, et al. Oncolytic adenovirus with hyaluronidase activity that evades neutralizing antibodies: VCN-11. J Control Release. 2021 Apr 10;332:517–528. 10.1016/j.jconrel.2021.02.035.
  • Yumul R, Richter M, Lu ZZ, et al. Epithelial junction opener improves oncolytic adenovirus therapy in mouse tumor models. Hum Gene Ther. 2016 Apr;27(4):325–337.
  • Tedcastle A, Illingworth S, Brown A, et al. Actin-resistant DNAse I expression from oncolytic adenovirus enadenotucirev enhances its intratumoral Spread and reduces tumor growth. Mol Ther. 2016 Apr;24(4):796–804.
  • Li J, Liu H, Li L, et al. The combination of an oxygen-dependent degradation domain-regulated adenovirus expressing the chemokine RANTES/CCL5 and NK-92 cells exerts enhanced antitumor activity in hepatocellular carcinoma. Oncol Rep. 2013 Mar;29(3):895–902.
  • Rizvi S, Gores GJ, Corey KE. The two faces of relaxin in cancer: antitumor or protumor? Hepatology. 2020 Mar;71(3):1117–1119.
  • Ahn HM, Hong J, Yun CO. Oncolytic adenovirus coexpressing interleukin-12 and shVEGF restores antitumor immune function and enhances antitumor efficacy. Oncotarget. 2016 Dec 20; 7(51):84965–84980.
  • Kim BG, Malek E, Choi SH, et al. Novel therapies emerging in oncology to target the TGF-beta pathway. J Hematol Oncol. 2021 Apr 6;14(1):55.
  • Hu Z, Zhang Z, Guise T, et al. Systemic delivery of an oncolytic adenovirus expressing soluble transforming growth factor-beta receptor II-Fc fusion protein can inhibit breast cancer bone metastasis in a mouse model. Hum Gene Ther. 2010 Nov;21(11):1623–1629.
  • Larson C, Oronsky B, Abrouk NE, et al. Toxicology and biodistribution of AdAPT-001, a replication-competent type 5 adenovirus with a trap for the immunosuppressive cytokine, TGF-beta. Am J Cancer Res. 2021;11(10):5184–5189.
  • Liu Z, Yang Y, Zhang X, et al. An Oncolytic adenovirus encoding decorin and granulocyte macrophage colony stimulating factor inhibits tumor growth in a colorectal tumor model by targeting pro-tumorigenic signals and via immune activation. Hum Gene Ther. 2017 Aug;28(8):667–680.
  • Kim SY, Kang D, Choi HJ, et al. Prime-boost immunization by both DNA vaccine and oncolytic adenovirus expressing GM-CSF and shRNA of TGF-beta2 induces anti-tumor immune activation. Oncotarget. 2017 Feb 28;8(9):15858–15877.
  • Freedman JD, Duffy MR, Lei-Rossmann J, et al. An oncolytic virus expressing a T-cell engager simultaneously targets cancer and immunosuppressive stromal cells. Cancer Res. 2018 Dec 15;78(24):6852–6865.
  • Scott EM, Jacobus EJ, Lyons B, et al. Bi- and tri-valent T cell engagers deplete tumour-associated macrophages in cancer patient samples. J Immunother Cancer. 2019 Nov 21;7(1):320.
  • Shin SP, Goh AR, Ju JM, et al. Local adenoviral delivery of soluble CD200R-Ig enhances antitumor immunity by inhibiting CD200-beta-catenin-driven M2 macrophage. Mol Ther Oncolytics. 2021 Dec 17;23:138–150.
  • Kennedy BE, Sadek M, Gujar SA. Targeted metabolic reprogramming to improve the efficacy of oncolytic virus therapy. Mol Ther. 2020 Jun 3; 28(6):1417–1421.
  • Rivadeneira DB, DePeaux K, Wang Y, et al. Oncolytic viruses engineered to enforce leptin expression reprogram tumor-infiltrating T cell metabolism and promote tumor clearance. Immunity. 2019 Sep 17;51(3):548–560 e4.
  • Morsy MA, Gu M, Motzel S , et al. An adenoviral vector deleted for all viral coding sequences results in enhanced safety and extended expression of a leptin transgene. Proc. Natl. Acad. Sci. USA. 1998 Jul 7;95(14):7866–7871.
  • Eriksson E, Milenova I, Wenthe J, et al. Shaping the tumor stroma and sparking immune activation by CD40 and 4-1BB signaling induced by an armed oncolytic virus. Clin Cancer Res. 2017 Oct 1;23(19):5846–5857.
  • Lu SC, Hansen MJ, Hemsath JR, et al. Modulating oncolytic adenovirus immunotherapy by driving two axes of the immune system by expressing 4-1BBL and CD40L. Hum Gene Ther. 2022 Mar;33(5–6):250–261.
  • Ylosmaki E, Ylosmaki L, Fusciello M, et al. Characterization of a novel OX40 ligand and CD40 ligand-expressing oncolytic adenovirus used in the PeptiCRAd cancer vaccine platform. Mol Ther Oncolytics. 2021 Mar 26;20:459–469.
  • Choi IK, Li Y, Oh E, et al. Oncolytic adenovirus expressing IL-23 and p35 elicits IFN-gamma- and TNF-alpha-co-producing T cell-mediated antitumor immunity. PLoS One. 2013;8(7):e67512.
  • Du YN, Wei Q, Zhao LJ, et al. Hydrogel-based co-delivery of CIK cells and oncolytic adenovirus armed with IL12 and IL15 for cancer immunotherapy. Biomed Pharmacother. 2022 Jul;151:113110.
  • Zhang Y, Zhang H, Wei M, et al. Recombinant adenovirus expressing a soluble fusion protein PD-1/CD137L subverts the suppression of CD8(+) T cells in HCC. Mol Ther. 2019 Nov 6;27(11):1906–1918.
  • Porter CE, Rosewell Shaw A, Jung Y, et al. Oncolytic adenovirus armed with bite, cytokine, and checkpoint inhibitor enables CAR T cells to control the growth of heterogeneous tumors. Mol Ther. 2020 May 6;28(5):1251–1262.
  • Eriksson E, Milenova I, Wenthe J, et al. IL-6 Signaling blockade during CD40-mediated immune activation favors antitumor factors by reducing TGF-beta, collagen type I, and PD-L1/PD-1. J Immunol. 2019 Feb 1;202(3):787–798.
  • Oh E, Choi IK, Hong J, et al. Oncolytic adenovirus coexpressing interleukin-12 and decorin overcomes Treg-mediated immunosuppression inducing potent antitumor effects in a weakly immunogenic tumor model. Oncotarget. 2017 Jan 17;8(3):4730–4746.
  • Nishio N, Diaconu I, Liu H, et al. Armed oncolytic virus enhances immune functions of chimeric antigen receptor-modified T cells in solid tumors. Cancer Res. 2014 Sep 15;74(18):5195–5205.
  • Cervera-Carrascon V, Siurala M, Santos JM, et al. TNFa and IL-2 armed adenoviruses enable complete responses by anti-PD-1 checkpoint blockade. Oncoimmunology. 2018;7(5):e1412902.
  • Havunen R, Siurala M, Sorsa S, et al. Oncolytic adenoviruses armed with tumor necrosis factor alpha and Interleukin-2 enable successful adoptive cell therapy. Mol Ther Oncolytics. 2017 Mar 17;4:77–86.
  • Quixabeira DCA, Cervera-Carrascon V, Santos JM, et al. Local therapy with an engineered oncolytic adenovirus enables antitumor response in non-injected melanoma tumors in mice treated with aPD-1. Oncoimmunology. 2022;11(1):2028960.
  • Kim W, Seong J, Oh HJ, et al. A novel combination treatment of armed oncolytic adenovirus expressing IL-12 and GM-CSF with radiotherapy in murine hepatocarcinoma. J Radiat Res. 2011;52(5):646–654.
  • Parks RJ, Chen L, Anton M, et al. A helper-dependent adenovirus vector system: removal of helper virus by Cre-mediated excision of the viral packaging signal. Proc Natl Acad Sci USA. 1996;93(24):13565–13570.
  • Morral N, Parks RJ, Zhou H, et al. High doses of a helper-dependent adenoviral vector yield supraphysiological levels of α 1 -antitrypsin with Negligible toxicity. Hum Gene Ther. 1998;9(18):2709–2716.
  • Brunetti-Pierri N, Ng T, Iannitti D, et al. Transgene expression up to 7 years in nonhuman primates following hepatic transduction with helper-dependent adenoviral vectors. Hum Gene Ther. 2013 Aug;24(8):761–765.
  • 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.
  • Weaver EA, Nehete PN, Nehete BP, et al. Comparison of systemic and mucosal immunization with helper-dependent adenoviruses for vaccination against mucosal challenge with SHIV. PLoS One. 2013;8(7):e67574.
  • Harui A, Roth MD, Kiertscher SM, et al. Vaccination with helper-dependent adenovirus enhances the generation of transgene-specific CTL. Gene Ther. 2004 Nov;11(22):1617–1626.
  • Fu YH, He JS, Zheng XX, et al. Intranasal vaccination with a helper-dependent adenoviral vector enhances transgene-specific immune responses in BALB/c mice. Biochem Biophys Res Commun. 2010 Jan 1;391(1):857–861.
  • Rosewell Shaw A, Porter C, Biegert G, et al. HydrAd: a helper-dependent adenovirus targeting multiple immune pathways for cancer immunotherapy. Cancers (Basel). 2022 Jun 2;14(11):2769.
  • Valadi H, Ekstrom K, Bossios A, et al. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat Cell Biol. 2007 Jun;9(6):654–659.
  • Pandey MS, Wang C, Umlauf S, et al. Simultaneous inhibition of PD-1 and stimulation of CD40 signaling pathways by anti-PD-L1/CD40L bispecific fusion protein synergistically activate target and effector Cells. Int J Mol Sci. 2021 Oct 21;22(21):11302.
  • de Silva S, Fromm G, Shuptrine CW, et al. CD40 enhances type I interferon responses downstream of CD47 blockade, bridging innate and adaptive immunity. Cancer Immunol Res. 2020 Feb;8(2):230–245.
  • Fromm G, de Silva S, Johannes K, et al. Agonist redirected checkpoint, PD1-Fc-OX40L, for cancer immunotherapy. J Immunother Cancer. 2018 Dec 18;6(1):149.
  • Guedan S, Alemany R. CAR-T cells and oncolytic viruses: joining forces to overcome the solid tumor challenge. Front Immunol. 2018;9:2460.
  • Kuryk L, Moller AW, Jaderberg M. Combination of immunogenic oncolytic adenovirus ONCOS-102 with anti-PD-1 pembrolizumab exhibits synergistic antitumor effect in humanized A2058 melanoma huNOG mouse model. Oncoimmunology. 2019;8(2):e1532763.
  • Yang Y, Xu W, Peng D, et al. An oncolytic adenovirus targeting transforming growth factor beta inhibits protumorigenic signals and produces immune activation: a novel approach to enhance anti-PD-1 and anti-CTLA-4 therapy. Hum Gene Ther. 2019 Sep;30(9):1117–1132.
  • Hu HJ, Liang X, Li HL, et al. Enhanced anti-melanoma efficacy through a combination of the armed oncolytic adenovirus ZD55-IL-24 and immune checkpoint blockade in B16-bearing immunocompetent mouse model. Cancer Immunol Immunother. 2021 Dec;70(12):3541–3555.
  • Wenthe J, Naseri S, Hellstrom AC, et al. Immune priming using DC- and T cell-targeting gene therapy sensitizes both treated and distant B16 tumors to checkpoint inhibition. Mol Ther Oncolytics. 2022 Mar 17;24:429–442.
  • Zhang H, Xie W, Zhang Y, et al. Oncolytic adenoviruses synergistically enhance anti-PD-L1 and anti-CTLA-4 immunotherapy by modulating the tumour microenvironment in a 4T1 orthotopic mouse model. Cancer Gene Ther. 2022 May;29(5):456–465.
  • Farrera-Sal M, Moya-Borrego L, Bazan-Peregrino M, et al. Evolving status of clinical immunotherapy with oncolytic adenovirus. Clin Cancer Res. 2021 Jun 1;27(11):2979–2988.
  • Mantwill K, Klein FG, Wang D, et al. Concepts in oncolytic adenovirus therapy. Int J Mol Sci. 2021 Sep 29;22(19):10522.
  • Young AM, Archibald KM, Tookman LA, et al. Failure of translation of human adenovirus mRNA in murine cancer cells can be partially overcome by L4-100K expression in vitro and in vivo. Mol Ther. 2012 Sep;20(9):1676–1688.
  • Siurala M, Vaha-Koskela M, Havunen R, et al. Syngeneic Syrian hamster tumors feature tumor-infiltrating lymphocytes allowing adoptive cell therapy enhanced by oncolytic adenovirus in a replication permissive setting. Oncoimmunology. 2016 May;5(5):e1136046.
  • McKenna MK, Rosewell-Shaw A, Suzuki M. Modeling the efficacy of oncolytic adenoviruses in vitro and in vivo: current and future perspectives. Cancers (Basel). 2020 Mar 7; 12(3):619.
  • Saruuldalai E, Park J, Kang D, et al. A host non-coding RNA, nc886, plays a pro-viral role by promoting virus trafficking to the nucleus. Mol Ther Oncolytics. 2022 Mar 17;24:683–694. .
  • Lei J, Jacobus EJ, Taverner WK, et al. Expression of human CD46 and trans-complementation by murine adenovirus 1 fails to allow productive infection by a group B oncolytic adenovirus in murine cancer cells. J Immunother Cancer. 2018 Jun 13;6(1):55.
  • Chartier C, Degryse E, Gantzer M, et al. Efficient generation of recombinant adenovirus vectors by homologous recombination in Escherichia coli. J Virol. 1996 Jul;70(7):4805–4810.
  • Hamdan F, Martins B, Feodoroff M, et al. GAMER-Ad: a novel and rapid method for generating recombinant adenoviruses. Mol Ther Methods Clin Dev. 2021 Mar 12;20:625–634.
  • Ni N, Deng F, He F, et al. A one-step construction of adenovirus (OSCA) system using the Gibson DNA assembly technology. Mol Ther Oncolytics. 2021 Dec 17;23:602–611. 10.1016/j.omto.2021.11.011.
  • Hatanaka K, Ohnami S, Yoshida K, et al. A simple and efficient method for constructing an adenoviral cDNA expression library. Mol Ther. 2003 Jul;8(1):158–166.
  • Rojas LA, Condezo GN, Moreno R, et al. Albumin-binding adenoviruses circumvent pre-existing neutralizing antibodies upon systemic delivery. J Control Release. 2016 Sep 10;237:78–88. 10.1016/j.jconrel.2016.07.004.
  • Hofherr SE, Shashkova EV, Weaver EA, et al. Modification of adenoviral vectors with polyethylene glycol modulates in vivo tissue tropism and gene expression. Mol Ther. 2008 Jul;16(7):1276–1282.
  • Schmid M, Ernst P, Honegger A, et al. Adenoviral vector with shield and adapter increases tumor specificity and escapes liver and immune control. Nat Commun. 2018 Jan 31;9(1):450.
  • Yoon AR, Hong J, Kim SW, et al. Redirecting adenovirus tropism by genetic, chemical, and mechanical modification of the adenovirus surface for cancer gene therapy. Expert Opin Drug Deliv. 2016 Jun;13(6):843–858.
  • Barry MA, Rubin JD, Lu S-C. Retargeting adenoviruses for therapeutic applications and vaccines. FEBS Lett. 2020 Jun;594(12):1918–1946.
  • Saha B, Parks RJ, Nevels M. Human adenovirus type 5 vectors deleted of early region 1 (E1) undergo limited expression of early replicative E2 proteins and DNA replication in non-permissive cells. PLoS One. 2017;12(7):e0181012.
  • Dhar D, Spencer JF, Toth K, et al. Pre-existing immunity and passive immunity to adenovirus 5 prevents toxicity caused by an oncolytic adenovirus vector in the Syrian hamster model. Mol Ther. 2009 Oct;17(10):1724–1732. DOI:10.1038/mt.2009.156.