Role of Adenoviruses in Cancer Therapy
Received Date: January 26, 2022 Accepted Date: February 26, 2022 Published Date: February 28, 2022
doi: 10.17303/jcrto.2022.10.103
Citation:Sintayehu Tsegaye Tseha (2022) Role of Adenoviruses in Cancer Therapy. J Vir Res Adv Vac 10: 1-18.
Abstract
Cancer is one of the leading causes of death in the world, which is the second after heart diseases. Adenoviruses (Ads) have become the promise of new therapeutic strategy for cancer treatment. The objective of this review is to discuss current advances in the applications of adenoviral vectors in cancer therapy. Adenoviral vectors can be engineered in different ways so as to change the tumor microenvironment from cold tumor to hot tumor, including; 1. by modifying Ads to deliver transgenes that codes for tumor suppressor gene (p53) and other proteins whose expression result in cell cycle arrest 2. Ads can also be modified to express tumor specific antigens, cytokines, and other immune-modulatory molecules. The other strategy to use Ads in cancer therapy is to use oncolytic adenoviruses, which directly kills tumor cells. Gendicine and Advexin are replication-defective recombinant human p53 adenoviral vectors that have been shown to be effective against several types of cancer. Gendicine was approved for treatment of squamous cell carcinoma of the head and neck by the Chinese Food and Drug Administration (FDA) agency in 2003 as a first-ever gene therapy product. Oncorine and ONYX-015 are oncolytic adenoviral vectors that have been shown to be effective against some types of cancer. The Chiness FDA agency has also approved Oncorin for the treatment of head and neck cancer. Ads that were engineered to express immune-stimulatory cytokines and other immune-modulatory molecules such as TNF-α, IL-2, BiTE, CD40L, 4-1BBL, GM-CSF, and IFN have shown promising outcome in treatment of cancer. Ads can also improve therapeutic efficacy of immune checkpoint inhibitors and adoptive cell therapy (Chimeric Antigen Receptor T Cells). In addition, different replication-deficient adenoviral vectors (Ad5-CEA, Ad5-PSA, Ad-E6E7, ChAdOx1–MVA and Ad-transduced Dendritic cells) that were tested as anticancer vaccines have been demonstrated to induce strong antitumor immune response. However, the use of adenoviral vectors in gene therapy is limited by several factors such as pre-existing immunity to adenoviral vectors and high immunogenicity of the viruses. Thus, innovative strategies must be continually developed so as to overcome the obstacles of using adenoviral vectors in gene therapy.
Keywords:Adenoviruses; Cancer; Cancer Therapy; Gene Therapy
Background
In the past few decades, gene therapy for diseases such as cancer using adenoviral vectors has been significantly advanced. Adenoviruses (Ads) can be used either as replication-competent Ads or replication-defective adenoviral vectors for gene therapy [1]. The following are the characteristics that made Ads one of the most favorable viruses for gene therapy:-1. Ads have unique ability to infect wide range of cell types (broad cell tropism) and have the capacity to induce strong cell mediated immunity and humoral response [2, 3]; 2. The genetics of Ads has been well known and Ads have stable genome [4]; 3. Ads have lytic replication cycle. This lytic replication cycle causes lysis of tumor cells [5]; 4. Adenoviral vectors have low pathogenicity and relatively safe and well-tolerated [4, 6]; 5. Ads have large transgene carrying capacity about and can transduce both in dividing and non- dividing cells [4, 7, 8] and 6. Ads can infect professional antigen-presenting cells that are very effective in presenting antigens to T-cells [9].
There are three generations of adenoviral vectors that have been developed for gene therapy. In the first generation, two genes are deleted (E1 and E3) so as to make the adenoviral vector replication defective, but keeping them to transduce host cells without killing them and liberating nearly 8 kb of space in the genome for the transgene and regulatory sequences [10, 11]. In the second-generation adenoviral vectors, in addition to the E1/E3 genes, E2 or E4 regions are also deleted, providing additional space for cargo sequences (nearly 10.5 kb). The third-generation adenoviral vectors is also called gutless or high capacity adenoviral vectors (HCAds) because they can accept cargo sequences up to 36 Kb, which were generated after deletion of almost all viral sequences except the sequences required for genome replication and encapsulation during vector production [12, 13]. The advantages of the third-generation adenoviral vectors are that HCAds can simultaneously encode multiple transgene cassettes. The other benefits of the third-generation adenoviral vectors is that the HCAds have less cellular toxicity and reduced immunogenicity, that efficiently transduces host cells because of the reduced stimulation of anti-adenoviral neutralizing antibodies [14, 15]. The objective of this review is to discuss current advances in applications of Ads in cancer therapy.
General Information on Adenoviruses
Ads are non-enveloped viruses with double stranded deoxy ribonucleic acid (DNA) genome. The genome of Ads ranges in size from 26 kb to 45 kb that is encompassed within icosahedral capsid [16, 17]. The Ads virion size ranges from 90-100 nm in diameter. There are six kinds of proteins that constitute the adenoviral capsid: penton, fiber, hexon, IX, VIII, and IIIa. The IIIa involves in the assembly of the viral structure. The fiber and penton proteins involve in the attachment and entry of the Ads into host cells and the hexon constitutes most of the viral capsid [18]. The proteins IIIa, VIII, and IX make up the virion core, which are associated with the DNA genome. The VIII is important for the stability of the viral capsid [19].
The genes of Ads can be categorized into two classes: as early genes (five early genes) and late genes (five late genes). The Ads bind to host cell through its receptors, which includes scavenger receptors, CD46, integrin αvβ5 heparin sulfate proteoglycans, sialic acid etc) and gains entry into the cytoplasm [20, 21].
After entry into the target host cell by micropinocytosis [22], Ads first express the five early proteins that are coded by the five early genes (E1A, E1B, E2, E3 and E4 which are involved in protein synthesis and DNA replication. Structural proteins (L1-L5) are coded by the five late genes [23]. Following replication, the adenoviral virion leaves the host cell by killing the host cell (lytic cycle). Newly produced Ads can infect wide range of host cells including antigen presenting cells and quiescent cells.
Ads belong to the family adenoviridae that is consisted of five genera: Mastadenoviruses, Avidadenoviruses, Siadenoviruses, Atadenoviruss and Ichtadenoviruse. The genus Mastadenovirus is consisted of Ads that infects humans (human adenoviruses) and non-human primates [24]. The genus Aviadenovirus is consisted of Ads that are isolated from birds. Viruses that are isolated from birds, ovine, bovine, deer and possum belong to the genus Atadenoviruses. Ads that are isolated from fish belong to the genus Ichtadenoviruses. The genus Siadenovirus includes viruses that are isolated from invertebrates [24].
Human adenoviruses (HAds) are categorized into seven species (A-G) that are further divided into 57 serotypes (Ad1-Ad57) [25]. Adenoviral serotyping is based on capsid proteins (VIII, hexon), phylogenetic distance (≥10%) in adenoviral genes that codes for protease, viral surface antigen neutralizing antibodies, and DNA polymerase [26].
HAds have worldwide distribution. Humans can be infected with more than one serotype or species of Ads that are usually acquired in early childhood, which leads to lifelong immunity [27]. Ads account for 5% of common cold cases. Wild type Ads often cause mild illness in immunocomptetent individuals, which mainly affects the respiratory tract, eyes and digestive system [28]. However, HAds causes severe illness in immunosuppressed individuals [29].
Ads induce diverse innate immune signaling pathways that result in the secretion of a number of proinflammatory cytokines. These proinflammatory cytokines result in the induction of robust adaptive humoral and cellular immune responses. The adaptive immune responses that develop against Ads include both T cells and neutralizing antibodies against the viral surface antigens such as hexon, penton, and fiber proteins [30].
Humans can be infected with different kinds of non-human Ads because of their broad tissue tropism and the structural similarity that they have with that of HAds. These characteristics subjected to use the non-human Ads as a vector for gene therapy and recombinant vaccine development so as to overcome the pre-existing antibodies that exists against human adenoviral vectors. There are numerous non-human adenoviral vectors that have used for recombinant vaccine development and gene therapy such as chimpanzee adenoviral vectors (ChAdOx1 nCoV-19, ChAd1, ChAd2, ChAd3, ChAd5, ChAd6, ChAd7, and ChAd68); bovine adenoviral vectors; fowl adenoviral vectors; canine adenoviral vectors, ovine adenoviral vectors; porcine adenoviral vectors [31, 32].
Application of adenoviruses in cancer therapy
Cancer is one of the leading causes of death in the world, which is the second after heart diseases. Globally, each year cancer causes about 10 million deaths and about 1 in 6 deaths are due to cancer [33]. Adenoviruses have become the promise of new therapeutic strategy for cancer treatment. Adenoviral vectors can be engineered in different ways so as to change the tumor microenvironment (TME) from cold tumor to hot tumor, including; 1. By modifying Ads to express cytokines, and other immune-modulatory molecules;2.By modifying Ads to deliver tumor suppressor gene and code for tumor specific antigen. The other way to use Ads in cancer therapy is to use oncolytic adenoviruses, which directly kill tumor cells after replication [34-36].
Adenoviral vectors coding for tumor suppressor protein (p53)
One of the strategies that have been developed to use Ads in cancer therapy is to use replication-deficient adenoviral vectors to carry transgenes that codes for a tumor suppressor protein (p53) or proteins that induce apoptosis or cell cycle arrest [36]. Wild-type p53 prevents development of cancer by inhibiting the activation of oncogenes and inducing programmed cell death (apoptosis) when the cell’s DNA repair functions are insufficient to repair DNA damage [37]. Suppression of p53 function is common in human cancers and 50% of cancers have mutations in the gene that codes for p53 protein [38]. For this reason, the p53 gene has become one of the target genes for transformation research of cancer gene therapy.
Advexin is a replication-defective recombinant human p53 adenoviral vector with a deletion on E3 and E1 genes that expresses a functional p53 protein from a Cytomegalovirus promoter [38, 39]. Advexin was proved efficacious against bladder cancer, ovarian cancer, prostate cancer, breast cancer, squamous cell carcinoma of the head and neck, hepatocellular carcinoma, colorectal cancer, squamous cell carcinoma of the oral cavity, oropharynx, hypopharynx, and larynx and non-small cell lung cancer (NSCLC) [39, 40].
Likewise, Gendicine is also a replication- defective recombinant human p53 Ads vector (rAd-p53) expressing p53 proteins which inhibits the uncontrolled division of cancer cells and induces apoptosis of cancerous cells [25]. Gendicine is very similar to advexin except that the p53 in gendicine is expressed from Rous Sarcoma Virus promoter [25]. Gendicine was approved for treatment of squamous cell carcinoma of the head and neck by the Chinese Food and Drug Administration agency in 2003 as a first-ever gene therapy product to be used in combination with chemotherapy and have been in use for more than 15 years [41, 42]. Gendecine has been also shown to be effective for treatment of different kinds of cancer in China including malignant glioma, epithelial ovarian carcinoma, HCC, and NSCLC [41, 42].
Oncolytic adenoviruses as anticancer virotherapy
Oncolytic viruses are viruses that that specifically infect and replicate in a tumor cells and kill the cancer cells by their lytic replication [35, 43]. Oncolytic Ads, particularly oncolytic HAds are one of the leading candidate viruses for cancer virotherapy because of their good safety profile and high immunogenicity [3]. Oncolytic Ads are genetically engineered Ads which acquired traits that enables them to infect and preferentially replicate in tumor cells [44]. Oncolytic adenoviral vector technologies have been approved in some countries for treatment of cancer in humans [25, 35]. As compared with normal and quiescent cells, generally, tumor cells are more permissive to Ads [45], because of different reasons. The first reason is that the entire pattern of gene expression in cancer cells is conducive for Ad replication [46]. The second reason is the fact that specific viral entry receptor is highly expressed in tumor cells. The other reason is the higher cell division and metabolic rate that take place in cancerous cells than that of normal and quiescent cells [47, 48]. The advantage of an oncolytic Ads is not only to specifically replicate in and lyse tumor cells, but oncolytic adenoviruses can also stimulate potent anti-viral and anti-tumor immune responses for tumor-specific antigens that are released following lysis of Ads infected tumor cells [49, 50].
Adenoviral vectors have been engineered to efficiently undergo oncolytic replication in cancer cells without replicating in healthy cells [51, 52]. For example, ONYX-015 with a partial E1B gene deficiency is oncolytic adenoviral vector that infects and replicate in tumor cells that lacks p53 but unable to replicate in healthy cells expressing p53 [51]. ONYX-015 has been demonstrated to be effective and well-tolerated oncolytic adenoviral vector that is reported to be more effective when given in combination with different cancer chemotherapies [52]. Oncorine (H101) is also a genetically modified oncolytic adenoviral vector expressing p53 gene. The Chinese food and drug administration agency has approved Oncorin for the treatment of head and neck cancer in combination with chemotherapy [39, 53].
Adenoviruses expressing Immunomodulatory Molecules
Adenoviruses can be used in cancer therapy by modifying the viruses to stimulate antitumor immune response in different ways including by expressing cytokines, and other immune-modulatory molecules [34, 54].
Interferon armed adenoviruses
Interferon (IFN) has a strong antitumor effect and has been used in the treatment of pancreatic cancer. Some studies showed that IFN-α significantly prolongs survival rate (by 2 to 5 years) [55, 56]. However, there are limitations in using IFN-based therapies, including dose-limiting systemic toxicities and low intratumoral concentration of IFN because of its short half-life in the bloodstream [57]. In response to this, oncolytic adenoviruses have been engineered to express IFN, which showed positive outcomes in treatment of cancer.
Armstrong et al. (2012) has reported oncolytic adenoviruses expressing human IFN-α as a promising platform for selective, long-term expression of IFN in human pancreatic cancer tissues [58, 59]. They used the oncloytic adenovirus Ad5/Ad3-Cox2-ΔE3-ADP-IFN in their study, which was developed to selectively replicate within cancer cells expressing cyclooxygenase 2 (Cox2). In order to improve the infectivity and oncolysis of the oncolytic adenoviruses, they made genetic modification in the virus to include an Ad5/Ad3 chimeric fiber and overexpress the adenovirus death protein.
Similarly, other researchers have also reported as oncolytic adenoviruses expressing IFN-α have promising outcomes in treatment of cancer. The researchers reported that an oncolytic adenovirus (OAd-hamIFN) that was investigated in an immunocompetent Syrian hamster model of pancreatic ductal adenocarcinoma showed efficient viral replication in tumor, significant inhibition of tumor growth, and enhanced survival when used in combination with chemo and radiation therapies [60]. Likewise, studies conducted by Tao et al. (2006) have shown that significant tumor regression of bladder cancers occurred following administration of an adenovirus expressing human interferon α (Ad-IFNα) using a mouse superficial bladder cancer model in which human bladder tumors are growing [61].
GM-CSF expressing adenoviruses (ONCOS-102)
Granulocyte macrophage colony-stimulating factor (GM-CSF) promotes activation of T cells and maturation of dendritic cells [62, 63]. ONCOS-102 is an oncolytic adenovirus that contains GM-CSF transgene [64]. A clinical study (phase-I) in patients with advanced solid tumors including colon, lung, and ovarian cancers demonstrated a strong immune cell infiltrate into tumors without dose-limiting toxicities [64].
LOAd703 expressing 4-1BBL and trimerized CD40L
The other oncolytic adenovirus that expresses immunostimulatoy cytokine is LOAd703. LOAd703 is armed with 4-1BBL and trimerized CD40L that was shown to replicate and kill pancreatic cancer cells via oncolysis in both in vitro and in vivo assays [65].
TILT-123 (Expressing TNF- α)
TILT-123 is an oncolytic adenovirus that incorporates transgenes for human tumor necrosis factor alpha (TNF-α) and interleukin-2 (IL-2). TNF-α and IL-2 were shown to be promising T cell stimulating factors when used in combination with adoptive cell therapy [66].
IL-12 and other cytokines armed adenoviruses
IL-12 represents the ideal candidate for tumor immunotherapy because of the fact that IL-12 induces a potent antitumor effect by promoting natural killer cells and cytotoxic T cell activities [67]. Several preclinical studies demonstrated promising antitumor effects of IL-12 in mice having solid tumor and hematologic malignancies [68, 69]. However, findings from clinical trials indicated severe side effects of systemic administration of IL-12 that markedly dampened hopes of the successful use of this cytokine in cancer patients [70]. But, the adverse effects associated with systemic administration of IL-12 can be reduced by using adenoviruses that express IL-12.
Ads that were engineered to express IL-12 have been shown to enhance immune stimulation and antitumor effect in a clinical trial [71] and pre-clinical studies [72]. Likewise, a study by Wang et al showed that oncolytic adenovirus (Ad-TD-nsIL-12) expressing IL-12 induces strong antitumor immune response against pancreatic cancer in Syrian hamster models without toxicity [73]. Similarly, a replication-deficient adenoviral vector encoding human IL-12 p70 transgene (Ad-RTS-hIL-12) was also shown to have no toxic effect in phase one clinical trial [74]. A recent trial found that intratumoral injection of Ad-RTS-hIL-12 was safe in patients with recurrent glioblastoma [74].
In addition to IL-12, there are also other cytokines, such as IL-24 and IL-13 that have been used to arm adenoviruses and have shown promising immune-activating properties in multiple preclinical cancer models [75, 76]. RANTES is another cytokine engineered in Ad that has been shown to enhance anticancer effect. In murine models of mammary adenocarcinoma and lymphoma, Ad-RANTES-E1A eradicated established tumors and inhibited metastases by recruiting DCs, macrophages, NK cells, and CD8+ T cells into the immunologically cold tumors [77].
Adenoviruses armed with Bispecific T cell engager (BiTE)
Bispecific T cell engager (BiTE) is a kind of artificial antibody that represents an innovative immunotherapy approach which enhances patients’ immune response to tumors. BiTE has dual antigen specificity, allowing them to bind to two unique antigens at the same time, i.e. the BiTE bind simultaneously to both tumor associated antigen and T cell (usually CD3), ultimately stimulating T-cell activation, tumor killing and cytokine production [78]. BiTE has been shown to be promising immunotherapy for the treatment of cancer in preclinical and clinical studies [79, 80]. The therapeutic efficacy of BiTE can be improved by using BiTE in conjunction with adenoviruses.
Alemany group engineered an oncolytic adenovirus expressing an EGFR-targeting BiTE that showed improved T cell-mediated killing of cancer cells both in vivo and in vitro [81]. They also demonstrated that anti-EGFR BiTE-armed OAd in combination with adoptive CAR-T cell therapy results in improved antitumor efficacy and prolonged survival of mice as a result of intratumoral T cell activation by BiTE [82].
Similarly, another research group (Fisher group) that developed BiTE armed oncolytic adenovirus (EnAd-SA-EpCAM) reported promising results in use of BiTE expressing adenoviruses against cancer [83]. The BiTE of EnAd-SA-EpCAM binds to epithelial cell adhesion molecule (EpCAM) in cancer cells. The Fisher group reported that the EnAd-SA-EpCAM effectively activate endogenous T cells within the immune-suppressive microenvironment and exhibited killing of endogenous tumor cells without the addition of exogenous T cells [83].
Recently, the Fisher group engineered another BiTE armed oncolytic adenovirus (EnAd-FAP-BiTE), which is targeted fibroblast activation protein (FAP) in cancer-associated fibroblasts (CAFs). CAFs are the main cellular component of solid tumor TME. The EnAd-FAP-BiTE induced the activation of tumor-infiltrating T cells that target and kill CAFs [84]. Likewise, another BiTE armed adenovirus (ICO15K-FBiTE) that was developed by the Fisher research group was shown to enhance overall antitumor efficacy without increasing the toxicity in mouse model [84].
ADV/HSV-TK
ADV/HSV-TK is an adenoviral vector expressing the herpes simplex virus (HSV) thymidine kinase (TK) gene. The HSV-TK protein has two principal functions, including 1. TK is a superantigen that stimulates a potent immune reaction and 2. A nucleotide analog product of prodrug phosphorylation lead to the death of dividing cancer cells [85]. Herman et al. studied ADV/HSV-TK in combination with ganciclovir for the treatment of human prostate cancer [86]. They reported that injection of ADV/HSV-TK into the prostate gland in the region with the greatest concentration of tumor cells resulted in significant reduction in tumor burden. Herman et al also showed that ADV/HSV-TK was proven safe, with minimal toxicity [86]. Likewise, other researchers also made similar observation on the efficacy and toxicology of ADV/HSV-TK against glioma, retinoblastoma, and mesothelioma [87, 88].
Combination therapy using adenoviruses and Chimeric Antigen Receptor (CAR) T Cells
One of the strategies that have been used for the immunotherapy of cancer is adoptive cell therapy, that includes chimeric antigen receptor (CAR) T cells, tumor-infiltrating lymphocytes, and T cell receptor modified T cells [89]. TCR-T cells are designed to encode receptors that specifically recognize cancer-specific antigens, and function through Major Histocompatibility Complex (MHC)-dependent mechanism, that limits their use [90]. Whereas CAR-T cell therapy functions through MHC-independent mechanism. CAR-T cell has been effective in the treatment of different types of cancer, which includes chronic lymphocytic leukemia and non-Hodgkin’s lymphoma [91, 92]. However, the use of CAR-T cells as a monotherapy has not demonstrated much success in solid tumors because of immunosuppressive tumor microenvironment (TME) and poor tumor infiltration of CAR-T cells [93]. Combination therapy with adenoviruses viruses provides one potential strategy for improvement of CAR-T therapy in solid tumors.
A study conducted by Watanabe and his colleagues demonstrated that the combination of oncolytic adenoviruses (expressing TNF-α and/or IL-2) and mesothelin-redirected CAR-T cells (meso-CAR-T) overcomes the immunosuppressive nature of the pancreatic cancer TME [94] Watanabe et al demonstrated that tumors treated with the combination of the virus expressing TNF-α and IL-2 (Ad5/3-E2F-d24-TNF-α-IRES-IL-2 (OAd-TNFα-IL2) and meso-CAR-T cells were infiltrated with significantly more CD4+ and CD8+ T cells compared to monotherapy with meso-CAR-T cells, or a combination with meso-CAR-T cells and the parent adenovirus lacking cytokine expression [94]. They also showed that meso-CAR-T cells in combination with OAd-TNFα-IL2 resulted in significantly higher accumulation of CAR-T cells at the tumor site when compared to meso-CAR-T monotherapy.
Likewise, a study that was done by a Suzuki group demonstrated better efficacy of CAR-T cell in treatment of prostate cancer when used in combination with oncolytic adenoviruses. In order to improve the efficacy of the CAR-T cell therapy, this research group (Suzuki group) used a combinatorial adenovirus vector (oncolytic adenovirus (Ad5Δ24) and helper-dependent adenovirus expressing a mini anti-PD-L1 antibody (HDAdPD-L1) collectively termed CAd-VECPDL1) in combination with human epidermal growth factor receptor 2 (HER2)-specific CAR-T cells [95]. The findings showed that using this combination in an NSG mouse model was more effective in reducing tumor size and prolong survival of mice with prostate cancer [95].
The Suzuki group further modified the CAd-VECPDL1 vector by incorporating IL-12 (CAdVECIL12_PDL1) and tested it in a head and neck squamous cell carcinoma (HNSCC) model [96, 97]. In a xenograft model of NSG mice (NOD scid gamma mouse), the combination of HER2-CAR-T cells and CAdVECIL12_PDL1 virus significantly prolonged survival of treated animals to more than 100 days as compared to 21–24 days in the control groups, and HER2-CAR-T cells were detected in the tumors of surviving mice over 100 days after initial therapy [97]. The research group also used an orthotopic HNSCC model, establishing both primary tumors and lymphatic metastases, to test the aforementioned combination therapy. Mice that received both HER2-CAR-T cells and CAdVECIL12_PDL1 had improved tumor growth control at both primary and metastatic sites, maintained body weight, and had prolonged survival when compared to untreated and monotherapy groups [97]. Taken together, combination therapy with oncolytic viruses provides one potential strategy for improvement of CAR-T therapy in solid tumors.
Combination therapy using adenoviruses and antibodies against Immune checkpoint proteins
Immune checkpoint inhibitors therapy is a kind of immunotherapy that work by blocking the binding of checkpoint proteins (PD-1, PD-L and CTLA-4) with partner proteins so that T-cell became free and active to attack cancer cells. Ipilimumab is an anti-CTLA-4 antibody (that blocks CTLA-4 ligand and prevents inhibition T-cells) that was approved by the United States food and drug administrations (U.S. FDA) in 2010 for the treatment of advanced melanoma [98]. However, the systemic administrations of immune checkpoint inhibitors have been shown to cause severe immune-related adverse events [99, 100]. One of the strategies that can be employed to overcome these obstacles of using immune checkpoint inhibitors (such as anti-PD-L1, anti-PD-1 and anti-CTLA-4) in cancer therapy is to utilize immune checkpoint inhibitors in combination with oncolytic adenoviruses.
Ad5/3-Δ24: adenovirus expressing anti-CTLA-4 antibodies
Ad5/3-Δ24 is an oncolytic adenovirus that was engineered to code for anti-CTLA4 antibody [101]. Promising results have been achieved with the oncolytic adenovirus armed with anti-CTLA-4 antibodies (Ad5/3-Δ24) in mouse model. The local expression of anti-CTLA-4 antibody following administration of Ad5/3-Δ24-CTLA4 resulted in activation of T cells [101]. In addition, a significantly higher concentration of antitumor antibody was produced, while plasma levels remained at safe concentrations. The anti-CTLA-4 antibodies also showed direct proapoptic effect both in vivo and in vitro [101].
Ad5-CMV-mIL2 and Ad5-CMV-mTNF- α in combination with anti-PD-1 antibodies
Cervera-Carrascon and his colleagues that tested nonreplicating vectors expressing IL-2 (Ad5-CMV-mIL2) and TNF-α (Ad5-CMV-mTNF- α) in combination with programmed cell-death protein 1 (PD-1) blocking antibodies in a mouse model demonstrated complete regression of murine melanoma tumors, and prolonged survival of mice [102]. Furthermore, they showed that the viral infection shifted the cytokine profile of the tumor microenvironment towards T-helper type 1, indicating that nonreplicating adenoviral vectors significantly improve antitumor immunity [102].
DNX-2401 (Tasadenoturev) in combination with pembrolizumab
DNX-2401 is a replication-competent oncolytic adenovirus that selectively infects cancer cells lacking the normal retinoblastoma protein signaling pathway [103]. Aiken et al tested DNX-2401 in combination with intravenous pembrolizumab (PD-1 immune checkpoint inhibitor) in patients with recurrent glioma and the findings showed that treatment of glioma with combination of DNX-2401 and pembrolizumab significantly improves the disease burden [104].
ONCOS102 in combination with pembrolizumab
Ternyila (2020) investigated the recombinant oncolytic adenovirus ONCOS-102 in combination with the antibody pembrolizumab (PD-1 immune checkpoint inhibitor) in patients with malignant melanoma [105]. They reported that combination therapy with ONCOS-102 and pembrolizumab resulted in regression of the disease and increases in circulating proinflammatory cytokines, and tumor specific T cells without dose limiting toxicities [105].
Adenoviral vectors as recombinant anticancer vaccines
Genetic vaccine is a third-generation vaccine platform that is designed to induce an immune response against an antigen that is encoded by a gene delivered into a vector or nucleic acid instead of the antigen itself. There are three main classes of genetic vaccines, which include viral or bacterial vectored vaccines, ribonucleic acid (RNA) vaccines, and DNA vaccine. The most important advantage of genetic vaccine over the traditional vaccine strategies like killed or inactivated microorganisms and subunit vaccines is the fact that genetic vaccines have capacity to induce T-cell responses; especially cytotoxic CD8 T cells responses, in addition to the humoral responses [3]. Viral vectored vaccines are one of the important forms of genetic vaccines that have been shown to induce robust cell mediated immunity (transgene-specific T cell) and humoral (antibody) response [3]. Adenoviral vectors are one of the most extensively investigated vaccine vectors. Since the 1980s, adenoviral vectors have shown great promise as vaccine vectors [106-108].
Adenoviral vectors can also be used as a platform for anticancer vaccine development. This is based on the fact that adenoviral vectors can be engineered to stimulate antitumor immune response by expressing tumor-antigens. Replication-deficient adenoviral vectors are one of the viral vectors that have been extensively used as recombinant cancer vaccines as they cause potent cell mediated and humoral immune response against transgenes expressed by the adenoviral vectors [25].
ETBX-011(Ad5-CEA)
ETBX-011(also called Ad5-CEA) is an adenoviral vector-based cancer vaccine that is engineered to express a modified carcinoembryonic antigen (CEA) which contains the highly immunogenic epitope CAP1-6D. CEA is found in different kinds of cancer cells. ETBX-011 induces potent CEA-specific cell-mediated immune responses with antitumor activity [109]. ETBX-011 is well-tolerated in metastatic colorectal cancer patients and has potential survival benefit [110].
In order to overcome the challenge posed by tumor heterogeneity, such as the diversity of tumor associated antigens (TAA), a Tri-Ad vaccine (a combination of ETBX-011 with three different human TAA-expressing Ad vector vaccines (ETBX-011, ETBX-061, and ETBX-051) have been developed and tested in phase I clinical trial. The Tri-Ad vaccine regimen induces antitumor cytotoxic T cell responses and was proven safe and well tolerated in treatment of advanced cancer [111].
Ad5-PSA
Ad5-PSA is replication-deficient adenoviral vector that is engineered to express human prostate specific antigen (PSA). Ad5-PSA stimulates potent anti-PSA T cell responses and causes the destruction of PSA-secreting tumor cells both in preclinical [112] and clinical trials [113, 114]. Furthermore, Ad5-PSA prolongs survival and was demonstrated safe in patients with recurrent and hormone refractory prostate cancer [113, 114].
Ad-E6E7 in combination with and Ad-MAGEA3
Ad-E6E7 is another replication-deficient adenovirus-based anti-cancer vaccine, which expresses human papillomavirus (HPV) genes E6 and E7 [115, 116]. MG1-E6E7 is an oncolytic maraba virus strain which also expresses the HPV genes E6 and E7. The combination of Ad-E6E7 and MG1-E6E7 induced potent tumor-specific responses in various mouse cancer models [117]. In addition, the combination of Ad-E6E7 with MG1-E6E7 was proven to significantly prolong survival of mice with HPV-associated cancer [117].
ChAdOx1–MVA
Cappuccini et al investigated the immunogenicity and efficacy of ChAdOx1–MVA against prostate cancer in mouse model [118]. They reported that the ChAdOx1–MVA induced tumor specific cell mediated immune response. Furthermore, the ChAdOx1–MVA was proven to prolong the survival of the mice when used in combination with anti-PD-1 antibody [118].
Adenovirus-transduced Dendritic cells
The other approach to use adenoviral vectors for anticancer vaccine is the use Dendritic cells (DC) -based adenoviral vaccines. In this approach, DCs are transduced ex vivo with Ads encoding cancer specific antigens. The advantageous of transducing DC ex vivo instead of injecting Ads in vivo include: 1. Ex vivo transduction of DC overcomes pre-existing anti-viral immunity and induce effective anti-tumor responses [119, 120] and 2. Ex vivo transduced DC induces lower anti-viral antibody responses than that of Ads injected in vivo [119].
Ad-transduced DCs have shown positive outcomes against both solid tumor and hematologic cancers [121, 122]. Butterfield et al. (2008) tested an autologous DCs transduced ex vivo with Ads encoding the full-length melanoma antigen MART-1/Melan-A. They reported that the autologous DC-based adenoviral vaccine significantly induced cell mediated immune response (CD+8-T cells response) in metastatic melanoma patients [121]. Similarly, in patients with advanced NSCLC, injections of autologous DCs resulted in induction of systemic tumor antigen-specific immune responses with enhanced CD8+T-cell infiltration [4].
Challenges Of Using Adenoviral Vectors in Gene Therapy
The application of Adenoviral vectors in gene therapy and vaccine development is severely hampered by the widely prevalent pre-existing immunity to the most common Ad vectors infecting the human population [135, 136]. As Adenoviral vectors have high immunogenicity, their first use for vaccine can also leads to development of potent humoral and cellular immunity [137], which prevents the subsequent administration of adenoviral vector-based vaccines that requires repeated administration. Preexisting HAds immunity causes considerable reduction in immunogenicity of HAd5 vector vaccines in both humans and animals [137, 138].
In addition to preexisting immunity, there are also other obstacles in the use of adenoviral vectors in gene therapy and vaccine development, which include the possibility of the adenoviral vectors to reestablish replication competence, non-specificity, immunodominance of adenoviral antigens over the vaccine transgene antigen(s), and heterologous immunity with other pathogens such as hepatitis C virus [139, 140].
Several strategies have been employed in order to avoid the challenges of using adenoviral vectors, which includes the use of non-human adenoviral vectors [141], mucosal delivery of HAd5 vectors [142], engineering fiber- or hexon-chimeric HAd5 vectors [143], priming with recombinant DNA prior to boosting with a HAds vector [144], microencapsulation of HAds vectors in inert polymers [145], and use of vectors derived from HAd serotypes that rarely infects humans [135] and the use of gutless (helper-dependent Ad (HD-Ad) vectors [146].
Conclusions
Adenoviruses viruses have become the promise of new therapeutic strategy for cancer treatment. Adenoviral vectors can be engineered in different ways so as to change the tumor microenvironment from cold tumor to hot tumor, including; 1. By modifying adenoviruses (Ads) to deliver transgenes that codes for tumor suppressor gene (p53) and other proteins whose expression result in cell cycle arrest 2. Ads can also be modified to express tumor specific antigens, cytokines, and other immune-modulatory molecules. The other strategy to use Ads in cancer therapy is to use oncolytic adenoviruses, which directly kills tumor cells after replication. Gendicine is a replication-defective recombinant human p53 adenoviral vector that was approved for treatment of squamous cell carcinoma of the head and neck by the Chinese FDA. The Chiness FDA agency has also approved Oncorin (oncolytic adenoviral vectors) for the treatment of head and neck cancer. Ads that were engineered to express immune-stimulatory cytokines and other immune-modulatory molecules have shown promising outcome in treatment of cancer. Adenoviruses can also improve therapeutic efficacy of adoptive cell therapy when used in combination with CAR T Cells. Adenoviruses can also improve therapeutic efficacy of immune checkpoint inhibitors when used in combination with anti-PD-L1, anti-PD-1 and anti-CTLA-4 antibodies. Furthermore, different replication-deficient adenoviral vectors that were used as anticancer vaccines have been shown to stimulate potent antitumor immune response. However, the use of adenoviral vectors in gene therapy is limited by several factors such as pre-existing immunity to adenoviral vectors; high immunogenicity of the viruses; the possibility of the adenoviral vectors to reestablish replication competence; non-specificity; immunodominance of adenoviral antigens over the vaccine transgene antigen(s); and heterologous immunity with other pathogens. Thus, innovative strategies must be continually developed so as to overcome the obstacles of using adenoviral vectors in gene therapy.
List of Abbreviations
Ads: Adenoviruses: BiTE: Bispecific T cell engager: CAR: Chimeric Antigen Receptor: ChAd: chimpanzee adenoviral vectors: CTLA-4: cytotoxic T-lymphocyte-associated protein 4: DC: Dendritic cells: DNA: deoxy ribonucleic acid: GM-CSF: Granulocyte macrophage colony-stimulating factor: HAds : Human adenoviruses: HCAds: high capacity adenoviral vectors: HPV: human papillomavirus: HSV: herpes simplex virus: IFN: Interferon: IL-2: interleukin-2: MHC: Major Histocompatibility Complex: NSCLC: non-small cell lung cancer: PD1: Programmed Death 1 Receptor: PD-L1: Programmed death ligand-1: PSA: prostate specific antigen; TK: thymidine kinase gene; TME: tumor microenvironment; TNF-α: Tumor necrosis factor alpha; U.S. FDA: United States food and drug administrations
Declarations
Ethics approval and consent to participate
Not applicable
Consent for publication
Not applicable
Availability of data and material
Not applicable
Competing interests
There is no conflict of interest in this work
Funding
No fund was obtained for this study.
Authors’ contributions
Not applicable
- Sato-Dahlman, M., and Yamamoto, M. (2018). The development of oncoltyic adenovirus therapy in the past and future—For the case of pancreatic cancer. Curr. Cancer Drug Targets. 18:153–161.
- Yang, ZY., Wyatt, LS., Kong, WP., Moodie, Z., Moss, B., Nabel, GJ. (2003). Overcoming immunity to a viral vaccine by DNA priming before vector boosting. J Virol.77 (1): 799–803.
- Sullivan, N.J., Hensley, L., Asiedu, C., Geisbert, T.W., Stanley, D., Johnson, J., et al. (2011). CD8+ cellular immunity mediates rAd5 vaccine protection against Ebola virus infection of nonhuman primates. Nat. Med. 17: 1128–1131.
- Lee, C.S., Bishop, E.S., Zhang, R., Yu, X., Farina, E.M., Yan, S., et al. (2017). Adenovirus-mediated gene delivery: Potential applications for gene and cell-based therapies in the new era of personalized medicine. Genes Dis. 4: 43–63.
- Shenk, T. (1996). Adenoviridae: The viruses and their replication. In Virology; Fields, B., Knipe, D., Howley, P., Eds.; Lipponcott-Raven: Philadelphia, PA, USA, 1996; Volume 2, pp. 2111–2148.
- Thacker, E., Timares, L., and Matthews, Q. (2009) Strategies to Overcome Host Immunity to Adenovirus Vectors in Vaccine Development. Expert Rev. Vaccines. 8(6): 761–777.
- Crystal, R.G.(2014). Adenovirus: The First Effective In Vivo Gene Delivery Vector. Hum. Gene Ther. 25: 3–11.
- Gao, J., Mese, K., and Bunz, O. (2019). Ehrhardt, A. State-of-the-art Human Adenovirus Vectorology for Therapeutic Approaches. FEBS Lett. 593: 3609–3622.
- Robert-Guroff, M. (2007). Replicating and non-replicating viral vectors for vaccine development. Curr. Opin. Biotechnol.18: 546–556.
- Bett, A.J., Haddara, W., Prevec, L., and Graham, F.L.(1994). An Efficient and Flexible System for Construction of Adenovirus Vectors with Insertions or Deletions in Early Regions 1 and 3. Proc. Natl. Acad. Sci. USA, 91: 8802–8806.
- McGrory, W.J., Bautista, D.S., and Graham, F.L. (1988). A Simple Technique for the Rescue of Early Region I Mutations into Infectious Human Adenovirus Type 5. Virology. 163: 614–617.
- Kochanek, S., Clemens, P., Mitani, K., Chen, H., Chan, S., and Caskey, C. (1996). A new adenoviral vector: Replacement of all viral coding sequences with 28 kb of DNA independently expressing both full-length dystrophin and beta-galactosidase. Proceedings of the National Academy of Sciences. 93(12): 5731-5736.
- Lusky, M., Christ, M., Rittner, K., Dieterle, A., Dreyer, D., Mourot, B.et al (1998). In Vitro and In Vivo Biology of Recombinant Adenovirus Vectors with E1, E1/E2A, or E1/E4 Deleted. J. Virol. 72: 2022–2032.
- Morral, N., Parks, R., Zhou, H., Langston, C., Schiedner, G., Quinones, J., et al (1998). High doses of a helper-dependent adenoviral vector yield supraphysiological levels of alpha1-antitrypsin with negligible toxicity. Human Gene Therapy. 9(18): 2709-2716.
- Schiedner, G., Morral, N., Parks, RJ., Wu, Y., Koopmans, SC., Langston, C, et al (1998). Genomic DNA transfer with a high-capacity adenovirus vector results in improved in vivo gene expression and decreased toxicity. Nature Genetics.18(2):180-183.
- Breyer, B., Jiang, W., Cheng, H., Zhou, L., Paul, R., Feng,T., et al. (2001). Adenoviral vector-mediated gene transfer for human gene therapy. Curr Gene Ther. 2: 149-162.
- Berk, AJ.(2013) Adenoviridae. In: Knipe, DM., Howley, PM., editors. Fields Virology. Philadelphia, PA: Lippincott Williams & Wilkins; 2013. p. 1704-1731.
- Baker, A.T., Greenshields-Watson, A., Coughlan, L., Davies, J.A., Uusi-Kerttula, H., Cole, D.K., et al. (2019). Diversity within the adenovirus fiber knob hypervariable loops influences primary receptor interactions. Nat. Commun. 10: 741.
- Wickham, T., Carrion, M., and Kovesdi, I. (1995). Targeting of adenovirus penton base to new receptors through replacement of its RGD motif with other receptor-specific peptide motifs. Gene Therapy.2: 750-756.
- Amberg, N. (2012). Adenovirus receptors: Implications for targeting of viral vectors. Trends in Pharmacological Sciences.33: 442-448.
- Lenman, A., Liaci, MA., Liu, Y., Årdahl, C., Rajan, A., Nilsson, E., et al. (2015). Human adenovirus 52 uses sialic acid-containing glycoproteins and the coxsackie and adenovirus receptor for binding to target cells. PLOS Pathogens.11(2): e1004657.
- Gaggar, A., Shayakhmetov, DM., and Lieber, A. (2003). CD46 is a cellular receptor for group B adeno- viruses. Nature Medicine. 9: 1408-1412.
- Crawford-Miksza, L., and Schnurr, D.(1996). Analysis of 15 adenovirus hexon proteins reveals the location and structure of seven hypervariable regions containing serotype-specific residues. Journal of Virology.70(3): 1836-1844.
- Davison, AJ., Benko, M., and Harrach, B. (2003). Genetic content and evolution of adenoviruses. J Gen Virol. 84: 2895–908.
- Wold, W.S.M., and Toth, K.(2013). Adenovirus vectors for gene therapy, vaccination and cancer gene therapy. Curr. Gene Ther.13: 421–433.
- Wadell, G. (1998). Adenoviridae: The adenoviruses. In: Laboratory Diagnosis of Infectious Diseases Principles and Practice. New York, NY: Springer.
- Yu, B., Wang, Z., Dong, J., Wang, C., Gu, L., Sun, C. et al. (2012). A serological survey of human adenovirus serotype 2 and 5 circulating pediatric populations in Changchun, China. Virology Journal.9: 287
- Khanal, S., Ghimire, P., Dhamoon, A. (2018). The Repertoire of Adenovirus in Human Disease: The Innocuous to the Deadly. Biomedicines. 6: 30
- Ephros, M., Friedman, B., Elhasid, R., Kra-Oz, Z., Shaked-Mishan, P., Sattinger, J. et al. (2009). Incidence and Clinical Manifestations of Adenoviral Infection among Children Undergoing Allogeneic Stem Cell Transplantation. IMAJ. 11: 744–748.
- Zhang, S., Huang, W., Zhou, X., Zhao, Q., Wang, Q., and Jia, B. (2013). Seroprevalence of neutralizing anti- bodies to human adenoviruses type-5 and type-26 and chimpanzee adenovirus type- 68 in healthy Chinese adults. Journal of Medical Virology.85: 1077-1084
- Colloca, S., Barnes, E., Folgori, A., Ammendola, V., Capone, S., Cirillo, A., et al. (2012). Vaccine vectors derived from a large collection of simian adenoviruses induce potent cellular immunity across multiple species. Science Translational Medicine.4: 115ra2-115ra2.
- Ewer, K., Sebastian, S., Spencer, A., Gilbert, S., Hill, A., Lambe, T.(2017). Chimpanzee adenoviral vectors as vaccines for outbreak pathogens. Human Vaccines & Immunotherapeutics.13: 3020-3032
- Ferlay, J., Ervik M., Lam, F., Colombet, M., Mery, L., Piñeros, M., et al. (2020). Global Cancer Observatory: Cancer Today. Lyon: International Agency for Research on Cancer. https://gco.iarc.fr/today, accessed 15 March 2021.
- Seth, P. (2005). Vector-Mediated Cancer Gene Therapy: An Overview. Cancer Biol. Ther. 4: 512–517.
- Shaw, A., and Suzuki, M. (2016). Recent advances in oncolytic adenovirus therapies for cancer. Current Opinion in Virology.21: 9-15.
- Saito, H., Kitagawa, K., Yoneda, T., Fukui, Y., Fujsawa, M., Bautista, D., et al. (2017). Combination of p53-DC vaccine and rAd-p53 gene therapy induced CTLs cytotoxic against p53-deleted human prostate cancer cells in vitro. Cancer Gene Therapy.24: 289-296.
- Brown, CJ., Lain, S., Verma, CS., Fersht, AR., and Lane, DP (2009). Awakening guardian angels: drugging the p53 pathway. Nat Rev Cancer. 9(12): 862-73.
- Isobe, T., Hiyama, K., Yoshida, Y., Fujiwara, Y., and Yamakido, M. (1994). Prognostic significance of p53 and ras gene abnormalities in lung adenocarcinoma patients with stage I disease after curative resection. Jpn J Cancer Res.85: 1240-6.
- Räty, J., Pikkarainen, J., Wirth, T., and Ylä-Herttuala, S. (2008). Gene therapy: The first approved gene-based medicines, molecular mechanisms, and clinical indications. Current Molecular Pharmacology.1:13-23.
- Yoo, G.H., Moon, J., LeBlanc, M., Lonardo, F., Urba, S., Kim, H., et al (2009). A Phase 2 Trial of Surgery with Perioperative INGN 201 (Ad5CMV-P53) Gene Therapy Followed by Chemoradiotherapy for Advanced, Resectable Squamous Cell Carcinoma of the Oral Cavity, Oropharynx, Hypopharynx, and Larynx: Report of the Southwest Oncology Group. Arch. Otolaryngol. Neck Surg. 135: 869.
- Chen, G., Zhang, S., He, X., Liu, S., Ma, C., and Zou, X-P. (2014). Clinical utility of recombinant ade- noviral human p53 gene therapy: Current perspectives. Onco Targets and Therapy.7:1901-1909.
- Zhang, W., Li, L., Li, D., Liu, J., Li, X., Li, W., et al. (2018). The first approved gene therapy product for cancer Ad-p53 (gendicine): 12 years in the clinic. Human Gene Therapy. 29(2): 160-179.
- Curiel, D., and Yamamoto, M. (2005). Cancer Gene Therapy. Technology in Cancer Research & Treatment. 4: 315-330.
- Dong, F., Wang, L., John, J., Davis, JJ., Hu, W., Zhang, L., et al (2006). Eliminating Established Tumor in nu/nu Nude Mice by a Tumor Necrosis Factor-A-Related Apoptosis-Inducing Ligand Armed Oncolytic Adenovirus. Clin Cancer Res.12: 5224-5230.
- Vaillancourt, M., Atencio, I., Quijano, E., Atencio, I., Quijano, E., Howe, J., and Ramachandra, M., (2005). Inefficient killing of quiescent human epithelial cells by replicating adenoviruses: potential implications for their use as oncolytic agents. Cancer Gene Ther. 12: 691–698.
- Ferrari, R., Pellegrini, M., Horwitz, GA., Xie, W., Berk, A., and Kurdistan, S. (2008). Epigenetic reprogramming by adenovirus e1a. Science. 321: 1086–1088.
- Coffey, MC., Strong, JE., Forsyth, PA., and Lee, PW. (1998). Reovirus therapy of tumors with activated Ras pathway. Science. 282: 1332-1334.
- Aghi, M., Visted, T., Depinho, R.A., and Chiocca, E.A.(2008). Oncolytic herpes virus with defective ICP6 specifically replicates in quiescent cells with homozygous genetic mutations in p16. Oncogene. 27: 4249-4254.
- Andtbacka, R.H., Kaufman, H.L., Collichio, F., Amatruda, T., Senzer, N., Chesney, J., et al. (2015). Talimogene laherparepvec improves durable response rate in patients with advanced melanoma. J. Clin. Oncol. 33: 2780–2788.
- Marelli, G., Howells, A., Lemoine, N.R., and Wang, Y. (2018). Oncolytic viral ther-apy and the immune system: a double-edged sword against cancer. Front. Immunol. 9: 866.
- Bischoff, J., Kirn, D., Williams, A., Heise, C., Horn, S., Muna, M., et al. (1996). An adenovirus mutant that replicates selectively in p53-deficient human tumor cells. Science.274: 373-376.
- Heise, C., Williams, A., Xue, S., Propst, M., and Kirn, D. (1999). Intravenous administration of ONYX- 015, a selectively replicating adenovirus, induces antitumoral efficacy. Cancer Research. 59: 2623-2628.
- Guo, W., and Song, H. (2018). Development of gene therapeutics for head and neck cancer in China: From bench to bedside. Human Gene Therapy. 29: 180-187.
- Cerullo, V., Capasso, C., Vähä-Koskela, M., Hemminki, O., and Hemminki, A. (2018). Cancer-targeted oncolytic adenoviruses for modulation of the immune system. Current Cancer Drug Targets. 8: 124-138.
- Nukui, Y., Picozzi, V.J., and Traverso, L.W (2000). Interferon-based adjuvant chemoradiation therapy improves survival after pancreaticoduodenectomy for pancreatic adenocarcinoma. Am. J. Surg. 179: 367–371.
- Picozzi, V.J.; Kozarek, R.A.; Traverso, L.W.W (2003). Interferon-based adjuvant chemoradiation therapy after pancreaticoduodenectomy for pancreatic adenocarcinoma. Am. J. Surg. 185: 476–480.
- Tagliaferri, P., Caraglia, M., Budillon, A., Marra, M., Vitale, G., and Viscomi, C. (2005). New pharmacokinetic and pharmacodynamic tools for interferon-alpha (IFN-α) treatment of human cancer. Cancer Immunol. Immunother. 54, 1–10.
- Armstrong, L., Arrington, A., Han, J., Gavrikova, T., Brown, E., Yamamoto, M., et al.(2012A). Generation of a novel, cyclooxygenase-2-targeted, interferon-expressing, conditionally replicative adenovirus for pancreatic cancer therapy. Am. J. Surg. 204: 741–750.
- Armstrong, L., Davydova, J., Brown, E., Han, J., Yamamoto, M., Vickers, S.M. (2012B). Delivery of interferon alpha using a novel Cox2-controlled adenovirus for pancreatic cancer therapy. Surgery. 152: 114–122.
- LaRocca, C.J.; Han, J.; Gavrikova, T.; Armstrong, L.; Oliveira, A.R.; Shanley, R. et al (2015). Oncolytic adenovirus expressing interferon alpha in a syngeneic Syrian hamster model for the treatment of pancreatic cancer. Surgery. 157: 888–898.
- Tao, Z., Connor, R., Ashoori, F., Dinney, C., Munsell, M., JA Philopena, JA. (2006). Efficacy of a single intravesical treatment with Ad-IFN/Syn 3 is dependent on dose and urine IFN concentration obtained: implications for clinical investigation. Cancer Gene Therapy. 13: 125–130.
- Lee, S., and Margolin, K (2011). Cytokines in Cancer Immunotherapy. Cancers (Basel). 3: 3856–3893.
- Hamid, O., Ismail, R., and Puzanov, I (2020). Intratumoral Immunotherapy—Update 2019. Oncologist. 25 (3):e423-e438.
- Ranki, T., Pesonen, S., Hemminki, A., Partanen, K., Kairemo, K., Alanko, T., et al (2016). Phase I study with ONCOS-102 for the treatment of solid tumors - an evaluation of clinical response and exploratory analyses of immune markers. J. Immunother. Cancer. 4: 17.
- Eriksson, E.; Milenova, I.; Wenthe, J.; Ståhle, M.; Leja-Jarblad, J.; Ullenhag, G. et al (2017). Shaping the Tumor Stroma and Sparking Immune Activation by CD40 and 4-1BB Signaling Induced by an Armed Oncolytic Virus. Clin. Cancer Res. 23: 5846–5857.
- Tähtinen, S., Kaikkonen, S., Merisalo-Soikkeli, M., Grönberg-Vänberg-Vähä-Koskela, S., Kanerva, A., Parviainen, S., et al (2015). Favorable Alteration of Tumor Microenvironment by Immunomodulatory Cytokines for Efficient T-Cell Therapy in Solid Tumors. PLoS ONE. 10: e0131242.
- Del Vecchio, M., Bajetta, E., Canova, S., Lotze, M.T., Wesa, A., and Parmiani, G., (2007). Interleukin-12: Biological properties and clinical application. Clin. Cancer Res. 13: 4677–4685.
- Teicher BA, Ara G, Buxton D, Leonard J, Schaub RG (1997). Optimal scheduling of interleukin 12 and chemotherapy in the murine MB-49 bladder carcinoma and B16 melanoma. Clin Cancer Res. 3: 1661–1667.
- Kozar, K., Kaminski, R., Switaj, T., Ołdak, T., Machaj, E., Wysocki, PJ., et al (2003). Interleukin 12-based immunotherapy improves the antitumor effectiveness of a low-dose 5-Aza-2-deoxycitidine treatment in L1210 leukemia and B16F10 melanoma models in mice. Clin Cancer Res. 9: 3124–3133.
- Lasek, W., Zagoz˙dz˙on, R., Jakobisiak, M(2014). Interleukin 12: still a promising candidate for tumor immunotherapy? Cancer immunol Immunother. 63: 419-435.
- Sangro, B., Mazzolini, G., Ruiz, J., Herraiz, M., Quiroga, J., Herrero, I., et al. (2004). Phase I trial of intratumoral injectio of an adenovirus encoding interleukin-12 for advanced digestive tumors. J. Clin. Oncol. 22: 1389–1397.
- Bortolanza, S., Bunuales, M., Otano, I., Gonzalez-Aseguinolaza, G., Ortiz-de-Solorzano, C., Perez, D., et al., (2009). Treatment of pancreatic cancer with an oncolytic adenovirus expressing interleukin-12 in Syrian hamsters. Mol. Ther. 17: 614–622.
- Wang, P., Li, X., Wang, J., Gao, D., Li, Y., Li, H., et al (2017). Re-designing Interleukin-12 to enhance its safety and potential as an anti-tumor immunotherapeutic agent. Nat. Commun. 8, 1395
- Chiocca, E.A., Yu, J.S., Lukas, R.V., Solomon, I.H., Ligon, K.L., Nakashima, H. et al. (2019). Regulatable interleukin-12 gene therapy in patients with recurrent high-grade glioma: Results of a phase 1 trial. Sci. Transl. Med. 11: 5680.
- Santos, J.M.; Cervera-Carrascon, V.; Havunen, R.; Zafar, S.; Siurala, M.; Sorsa, S.; Anttila, M.; Kanerva, A.; Hemminki, A. (2018). Adenovirus coding for Interleukin-2 and tumor necrosis factor alpha replaces lymphodepleting chemotherapy in Adoptive T cell therapy. Mol. Ther. 26: 2243–2254.
- Liu, L.; Ma, J.; Qin, L.; Shi, X.; Si, H.; Wei, Y. (2019). Interleukin-24 enhancing antitumor activity of chimeric oncolytic adenovirus for treating acute promyelocytic leukemia cell. Medicine (Baltimore). 98: e15875.
- Lapteva, N., Aldrich, M., Weksberg, D., Rollins, L., Goltsova, T., Chen, S.Y., (2009). Targeting the intratumoral dendritic cells by the oncolytic adenoviral vaccine expressing RANTES elicits potent antitumor immunity. J. Immunother. 32: 145–156
- Huehls, A., Coupet, T., and Sentman, C (2014). Bispecific T cell engager for cancer immunotherapy. Immunology and cell biology. 93: 290-296.
- Brischwein, K., Schlereth, B., Guller, B., Steiger, C., Wolf, A., Lutterbuese, R. et al (2006). MT110: a novel bispecfic single chain antibody construct with high efficacy in eradicating established tumors. Mol immunol. 43: 1129-1143.
- Bargou, R., Leo, E., Zugmaier, G., Klinger, M., Goebeler, M., Knop, S. et al. (2008). Tumor regression in cancer patients by very low doses of a T-cell engaging antibody. Science. 321: 974-977.
- Fajardo, C.A., Guedan, S., Rojas, L.A., Moreno, R., Arias-Badia, M., de Sostoa, J., et al (2017). Oncolytic adenoviral delivery of an egfr-targeting t-cell engager improves antitumor efficacy. Cancer Res. 77: 2052–2063.
- Wing, A., Fajardo, C.A., Posey, A.D., Shaw, C., Da, T., Young, R.M., et al (2018). Improving CART-Cell Therapy of Solid Tumors with Oncolytic Virus-Driven Production of a Bispecific T-cell Engager. Cancer Immunol. Res. 6: 605–616.
- Freedman, J.D., Hagel, J., Scott, E.M., Psallidas, I., Gupta, A., Spiers, L., et al (2017). Oncolytic adenovirus expressing bispecific antibody targets T-cell cytotoxicity in cancer biopsies. EMBO Mol. Med. 9: 1067–1087
- Freedman, J.D., Duffy, M.R., Lei-Rossmann, J., Muntzer, A., Scott, E.M., Hagel, J., et al (2018). An Oncolytic Virus Expressing a T-cell Engager Simultaneously Targets Cancer and Immunosuppressive Stromal Cells. Cancer Res. 78: 6852–6865.
- Aguilar, L.K., Shirley, L.A., Chung, V.M., Marsh, C.L., Walker, J., Coyle, W., et al. (2015). Gene-Mediated Cytotoxic Immunotherapy as Adjuvant to Surgery or Chemoradiation for Pancreatic Adenocarcinoma. Cancer Immunol. Immunother. 64: 727–736.
- Herman, J.R., Adler, H.L., Aguilar-Cordova, E., Rojas-Martinez, A., Woo, S., Timme, T.L., et al (1999). In Situ Gene Therapy for Adenocarcinoma of the Prostate: A Phase I Clinical Trial. Hum. Gene Ther. 10: 1239–1250.
- Sterman, D.H., Recio, A., Vachani, A., Sun, J., Cheung, L., and DeLong, P. et al. (2005). Long-Term Follow-up of Patients with Malignant Pleural Mesothelioma Receiving High-Dose Adenovirus Herpes Simplex Thymidine Kinase/Ganciclovir Suicide Gene Therapy. Clin. Cancer Res. 11: 7444–7453.
- Maatta, A.-M., Samaranayake, H., Pikkarainen, J., Wirth, T., and Yla-Herttuala, S. (2009). Adenovirus Mediated Herpes Simplex Virus-Thymidine Kinase/Ganciclovir Gene Therapy for Resectable Malignant Glioma. Curr. Gene Ther. 9: 356–367.
- Guedan, S.; Ruella, M.; June, C.H (2019). Emerging Cellular Therapies for Cancer. Annu. Rev. Immunol. 37: 145–171.
- Li, D., Li, X., Zhou, W.L., Huang, Y., Liang, X., Jiang, L., et al (2019). Genetically engineered T cells for cancer immunotherapy. Signal Transduct. Target. Ther. 4: 35
- Kochenderfer, J.N.; Wilson, W.H.; Janik, J.E.; Dudley, M.E.; Stetler-Stevenson, M.; Feldman, S.A..J.; et al. (2010). Eradication of B-lineage cells and regression of lymphoma in a patient treated with autologous T cells genetically engineered to recognize CD19. Blood. 116: 4099–4102.
- Porter, D.L.; Levine, B.L.; Kalos, M.; Bagg, A.; June, C.H (2011). Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. N. Engl. J. Med. 365: 725–733
- Newick, K., Moon, E. and Albelda, S.M (2016). Chimeric antigen receptor T-cell therapy for solid tumors. Mol. Ther. Oncolytics. 3: 16006.
- Watanabe, K., Luo, Y., Da, T., Guedan, S., Ruella, M., Scholler, J., et al (2018). Pancreatic cancer therapy with combined mesothelin-redirected chimeric antigen receptor T cells and cytokine-armed oncolytic adenoviruses. JCI Insight. 3: e99573.
- Tanoue, K.; Rosewell, A.; Watanabe, N.; Porter, C.; Rana, B.; Gottschalk, S.; et al. (2017). Armed oncolytic adenovirus-expressing PD-L1 Mini-Body enhances antitumor effects of chimeric antigen receptor T Cells in Solid Tumors. Cancer Res. 77: 2040–2051.
- Nishio, N.; Diaconu, I.; Liu, H.; Cerullo, V.; Caruana, I.; Hoyos, V.; et al., (2014). Armed oncolytic virus enhances immune functions of chimeric antigen receptor-modified T cells in solid tumors. Cancer Res. 74: 5195–5205.
- Rosewell, A.; Porter, C.E.; Watanabe, N.; Tanoue, K.; Sikora, A.; Gottschalk, S.; et al. (2017). Adenovirotherapy delivering cytokine and checkpoint inhibitor augments CAR T Cells against Metastatic Head and Neck Cancer. Mol. Ther. 25: 2440–2451.
- Wolchok, J.D., Hodi, F.S., Weber, J.S., Allison, J.P., Urba, W.J., Robert, C., et al (2013). Development of ipilimumab: A novel immunotherapeutic approach for the treatment of advanced melanoma. Ann. N. Y. Acad. Sci. 1291: 1–13.
- Ribas, A., Hanson, D.C., Noe, D.A., Millham, R., Guyot, D.J., Bernstein, S.H., et al (2007). Tremelimumab (CP-675,206),a Cytotoxic T Lymphocyte–Associated Antigen 4 Blocking Monoclonal Antibody in Clinical Development for Patients with Cancer. Oncologist. 12: 873–883.
- Hodi, F.S., O’Day, S.J., McDermott, D.F., Weber, R.W., Sosman, J.A., Haanen, J.B et al. (2010). Improved Survival with Ipilimumab in Patients with Metastatic Melanoma. N. Engl. J. Med. 363: 711–723.
- Dias, J.D., Hemminki, O., Diaconu, I., Hirvinen, M., Bonetti, A., Guse, K., et al (2012). Targeted cancer immunotherapy with oncolytic adenovirus coding for a fully human monoclonal antibody specific for CTLA-4. Gene Ther. 19: 988–998.
- Cervera-Carrascon, V., Siurala, M., Santos, J.M., Havunen, R., Tähtinen, S., Karell, P., et al (2018). TNFa and IL-2 armed adenoviruses enable complete responses by anti-PD-1 checkpoint blockade. Oncoimmunology. 7: e1412902.
- Fueyo, J., Gomez-Manzano, C., Alemany, R., Lee, P.S., McDonnell, T.J., Mitlianga, P., et al (2000). A mutant oncolytic adenovirus targeting the Rb pathway produces anti-glioma effect in vivo. Oncogene. 19: 2–12.
- Aiken, R., Chen, C., Cloughesy, T., Colman, H., Daras, M., Groves, M., et al (2019). Atim-33. interim results of a phase ii multi-center study of oncolytic adenovirus dnx-2401 with pembrolizumab for recurrent glioblastoma; captive study (keynote-192). Neuro-Oncology. 21, vi8–vi9.
- Ternyila, D (2020). ONCOS-102 Plus Pembrolizumab Induces Impressive Responses in Anti-PD1–Refractory Melanoma. Targeted Oncology. https://www.targetedonc.com/view/oncos-102-plus-pembrolizumab-induces-impressive-responses-in-anti-pd1-refractory-melanoma
- Graham, FL (1990). Adenoviruses as expression vectors and recombinant vaccines. Trends Biotechnol. 8: 85–7
- Imler, JL. (1995). Adenovirus vectors as recombinant viral vaccines. Vaccine: 13(13): 1143–51.
- Yarosh, OK., Wandeler, AI., Graham, FL., Campbell, JB., and Prevec, L. (1996). Human adenovirus type 5 vectors expressing rabies glycoprotein. Vaccine.14(13): 1257–64.
- Gabitzsch, E.S., Morse, M.A., Lyerly, H.K., Balint, J., and Jones, F. (2014). Immunotherapeutic treatment of metastatic colorectal cancer using ETBX-011. J. Clin. Oncol. 32: 3093.
- Morse, M.A., Chaudhry, A., Gabitzsch, E.S., Hobeika, A.C., Osada, T., Clay, T.M., et al. (2013). Novel adenoviral vector induces T-cell responses despite anti-adenoviral neutralizing antibodies in colorectal cancer patients. Cancer Immunol. Immunother. 62: 1293–1301.
- Gatti-Mays, M.E., Redman, J.M., Donahue, R.N., Palena, C., Madan, R.A., Karzai, F., et al (2019). A Phase I Trial Using a Multitargeted Recombinant Adenovirus 5 (CEA/MUC1/Brachyury)-Based Immunotherapy Vaccine Regimen in Patients with Advanced Cancer. Oncologist 2019.
- Elzey, B.D., Siemens, D.R., Ratliff, T.L., and Lubaroff, D.M. (2001). Immunization with type 5 adenovirus recombinant for a tumor antigen in combination with recombinant canarypox virus (alvac) cytokine gene delivery induces destruction of established prostate tumors. Int. J. Cancer. 94: 842–849.
- Lubaroff, D.M., Konety, B., Link, B.K., Ratliff, T.L., Madsen, T., Shannon, M., et al (2006). Clinical Protocol: Phase I Study of an Adenovirus/Prostate-Specific Antigen Vaccine in Men with Metastatic Prostate Cancer. Hum. Gene Ther. 17: 220–229
- Lubaroff, D.M., Williams, R.D., Vaena, D., Joudi, F., Brown, J., Smith, M., et al. (2012). An ongoing Phase II trial of an adenovirus/PSA vaccine for prostate cancer. Cancer Res. 72: 2692.
- Wieking, B.G., Vermeer, D.W., Spanos, W.C., Lee, K.M., Vermeer, P., Lee, W.T et al. (2012). A non-oncogenic HPV 16 E6/E7 vaccine enhances treatment of HPV expressing tumors. Cancer Gene Ther. 19: 667–674.
- Atherton, M.J., Stephenson, K.B., Nikota, J.K., Hu, Q.N., Nguyen, A., Wan, Y., et al. (2018). Preclinical development of peptide vaccination combined with oncolytic MG1-E6E7 for HPV- associated cancer. Vaccine. 36: 2181–2192.
- Pol, J.G., Atherton, M.J., Bridle, B.W., Stephenson, K.B., Le Boeuf, F., Hummel, J.L., et al. (2018). Development and applications of oncolytic Maraba virus vaccines. Oncolytic Virotherapy. 7: 117–128.
- Cappuccini, F., Stribbling, S., Pollock, E., Hill, A.V.S., and Redchenko, I (2016). Immunogenicity and Efficacy of the Novel Cancer Vaccine Based on Simian Adenovirus and MVA Vectors Alone and in Combination with PD-1 MAb in a Mouse Model of Prostate Cancer. Cancer Immunol. Immunother. 65: 701–713.
- Wan, Y., Emtage, P., Foley, R., Carter, R., Gauldie, J., et al. (1999). Murine dendritic cells transduced with an adenoviral vector expressing a defined tumor antigen can overcome anti-adenovirus neutralizing immunity and induce effective tumor regression. Int. J. Oncol. 14: 771–776.
- Nikitina, E.Y., Chada, S., Muro-Cacho, C., Fang, B., Zhang, R., Roth, JA. et al. (2002). An effective immunization and cancer treatment with activated dendritic cells transduced with full-length wild-type p53. Gene Ther. 9: 345–352.
- Butterfield, L.H., Comin-Anduix, B., Vujanovic, L,, Lee, Y., Dissette, V.B., Yang, J.-Q., et al (2008). Adenovirus MART-1–Engineered Autologous Dendritic Cell Vaccine for Metastatic Melanoma: J. Immunother. 31: 294–309.
- Lee, J.M., Lee, M.-H., Garon, E., Goldman, J.W., Salehi-Rad, R., Baratelli, F.E., et al. (2017). Phase I Trial of Intratumoral Injection of CCL21 Gene–Modified Dendritic Cells in Lung Cancer Elicits Tumor-Specific Immune Responses and CD8 + T-Cell Infiltration. Clin. Cancer Res. 23: 4556–4568.
- Kumon, H., Ariyoshi, Y., Sasaki, K., Sadahira, T., Araki, M., Ebara, S., et al. (2016). Adenovirus Vector Carrying REIC/DKK-3 Gene: Neoadjuvant Intraprostatic Injection for High-Risk Localized Prostate Cancer Undergoing Radical Prostatectomy. Cancer Gene Ther. 23: 400–409.
- Goto, Y., Ohe, Y., Kuribayashi, K., Nakano, T., Okada, M., Toyooka, S et al (2018). P2.06-11 A Phase I/II Study of Intrapleural Ad-SGE-REIC Administration in Patients with Refractory Malignant Pleural Mesothelioma. J. Thorac. Oncol. 13: S746.
- Lowenstein, P.R., Orringer, D.A., Sagher, O., Heth, J., Hervey-Jumper, S.L., Mammoser, A.G. et al (2019). First-in-Human Phase I Trial of the Combination of Two Adenoviral Vectors Expressing HSV1-TK and FLT3L for the Treatment of Newly Diagnosed Resectable Malignant Glioma: Initial Results from the Therapeutic Reprogramming of the Brain Immune System. J. Clin. Oncol. 37: 2019.
- Balint, J.P., Gabitzsch, E.S., Rice, A., Latchman, Y., Xu, Y., Messerschmidt, G.L et al., (2015). Extended evaluation of a phase 1/2 trial on dosing, safety, immunogenicity, and overall survival after immunizations with an advanced-generation Ad5 [E1-, E2b-]-CEA(6D) vaccine in late-stage colorectal cancer. Cancer Immunol. Immunother. 64: 977–987.
- Zhu, R., Weng, D., Lu, S., Lin, D., Wang, M., Chen, D et al (2018). Double-Dose Adenovirus-Mediated Adjuvant Gene Therapy Improves Liver Transplantation Outcomes in Patients with Advanced Hepatocellular Carcinoma. Hum. Gene Ther. 29: 251–258.
- Buller, R.E., Runnebaum, I.B., Karlan, B.Y., Horowitz, J.A., Shahin, M.;, Buekers, T. M et al., (2002). A Phase I/II Trial of RAd/P53 (SCH 58500) Gene Replacement in Recurrent Ovarian Cancer. Cancer Gene Ther. 9: 553–566.
- Kieran, M.W.. Goumnerova, L.. Manley, P., Chi, S.N., Marcus, K.J., Manzanera, A.G., et al (2019). Phase I Study of Gene-Mediated Cytotoxic Immunotherapy with AdV-Tk as Adjuvant to Surgery and Radiation for Pediatric Malignant Glioma and Recurrent Ependymoma. Neuro Oncol. 21: 537–546.
- Shore, N.D., Boorjian, S.A., Canter, D.J., Ogan, K., Karsh, L.I., Downs, T.M. et al (2017). Intravesical RAd–IFNα/Syn3 for Patients with High-Grade, Bacillus Calmette-Guerin–Refractory or Relapsed Non–Muscle-Invasive Bladder Cancer: A Phase II Randomized Study. J. Clin. Oncol.35: 3410–3416.
- Chiocca, E.A., Yu, J.S., Lukas, R.V., Solomon, I.H., Ligon, K.L., Nakashima, H. et al. (2019). Regulatable interleukin-12 gene therapy in patients with recurrent high-grade glioma: Results of a phase 1 trial. Sci. Transl. Med. 11: 5680.
- Behbahani, T.E., Rosenthal, E.L., Parker, W.B., and Sorscher, E.J.(2019). Intratumoral Generation of 2-fluoroadenine to Treat Solid Malignancies of the Head and Neck. Head Neck, 41: 1979–1983.
- Gabrilovich, DI. (2006). INGN 201 (Advexin): Adenoviral p53 gene therapy for cancer. Expert Opinion on Biological Therapy.6: 823-832
- Zhang, W., Li, L., Li, D., Liu, J., Li, X., Li, W., et al. (2018). The first approved gene therapy product for cancer Ad-p53 (gendicine): 12 years in the clinic. Human Gene Therapy. 29(2): 160-179.
- Vogels, R., Zuijdgeest, D., van, RR., Hartkoorn, E., Damen, I., de Bethune, MP., et al (2003). Replication-deficient human adenovirus type 35 vectors for gene transfer and vaccination: efficient human cell infection and bypass of preexisting adenovirus immunity. J Virol. 77(15): 8263–71.
- Nwanegbo E, Vardas E, Gao W, Whittle H, Sun H, Rowe D, et al (2004). Prevalence of neutralizing antibodies to adenoviral serotypes 5 and 35 in the adult populations of The Gambia, South Africa, and the United States. Clin Diagn Lab Immunol.11(2): 351–7.
- Yang, Y., Li, Q., Ertl, HC., and Wilson, JM. (1995). Cellular and humoral immune responses to viral antigens create barriers to lung-directed gene therapy with recombinant adenoviruses. J Virol. 69(4): 2004–15.
- Casimiro, DR., Chen, L., Fu, TM., Evans, RK., Caulfield, MJ., Davies, ME., et al (2003). Comparative immunogenicity in rhesus monkeys of DNA plasmid, recombinant vaccinia virus, and replication-defective adenovirus vectors expressing a human immunodeficiency virus type 1 gag gene. J Virol.77(11): 6305–13.
- Singh, S., Vedi, S., Samrat, S., Li, W., Kumar, R., and Agrawal, B. (2016). Heterologous immunity between adenoviruses and hepatitis C virus: A new paradigm in HCV immunity and vaccines. PLoS One.11(1): e0146404.
- Agrawal, B., Singh, S., Gupta, N., Li, W., Vedi, S., and Kumar, R. (2017). Unsolved puzzles surround-ing HCV immunity: Heterologous immunity adds another dimension. International Journal of Molecular Sciences. 18(8): 1626-1639.
- Hofmann C, Loser P, Cichon G, Arnold W, Both GW, Strauss M (1999). Ovine adenovirus vectors overcome preexisting humoral immunity against human adenoviruses in vivo. J Virol. 73(8): 6930–6.
- Xiang, Z., Li, Y., Gao, G., Wilson, JM., and Ertl, HC. (2003). Mucosally delivered E1-deleted adenoviral vaccine carriers induce transgene product-specific antibody responses in neonatal mice. J Immunol.171(8): 4287–93.
- Gall JG, Crystal RG, Falck-Pedersen E (1998). Construction and characterization of hexon-chimeric adenoviruses: specification of adenovirus serotype. J Virol. 72(12): 10260–4.
- Barouch, DH., McKay, PF., Sumida, SM., Santra, S., Jackson, SS., Gorgone, DA., et al (2003). Plasmid chemokines and colony-stimulating factors enhance the immunogenicity of DNA priming-viral vector boosting human immunodeficiency virus type 1 vaccines. J Virol. 77(16): 8729–35.
- Croyle, MA., Chirmule, N., Zhang, Y., and Wilson, JM (2002). PEGylation of E1-deleted adenovirus vectors allows significant gene expression on re-administration to liver. Hum Gene Ther.13(15): 1887–900.
- Brown, BD., Shi, CX., Rawle, FE., Tinlin, S., A McKinven, A., Hough, C et al (2004). Factors influencing therapeutic efficacy and the host immune response to helper-dependent adenoviral gene therapy in hemophilia A mice. J. Thromb. Haemost. 2(1): 111–118.
Tables at a glance