Open Access

Oncolytic adenovirus: A tool for reversing the tumor microenvironment and promoting cancer treatment (Review)

  • Authors:
    • Xiaoxi Wang
    • Liping Zhong
    • Yongxiang Zhao
  • View Affiliations

  • Published online on: March 2, 2021     https://doi.org/10.3892/or.2021.8000
  • Article Number: 49
  • Copyright: © Wang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Immunogene therapy can enhance the antitumor immune effect by introducing genes encoding co‑stimulation molecules, cytokines, chemokines and tumor‑associated antigens into treatment cells or human cells through genetic engineering techniques. Oncolytic viruses can specifically target tumor cells and replicate indefinitely until they kill tumor cells. If combined with immunogene therapy, oncolytic viruses can play a more powerful antitumor role. The high pressure, hypoxia and acidity in the tumor microenvironment (TME) provide suitable conditions for tumor cells to survive. To maximize the potency of oncolytic viruses, various methods are being developed to promote the reversal of the TME, thereby maximizing transmission of replication and immunogenicity. The aim of the present review was to discuss the basic mechanisms underlying the effects of oncolytic adenoviruses on the TME, and suggest how to combine the modification of the adenovirus with the TME to further combat malignant tumors.

Introduction

Solid tumors are a major cause of mortality in humans (1). Due to the advanced proliferative, invasive and migratory abilities of tumor cells, the prognosis for patients with cancer is extremely poor (2). Effective tumor therapy is disrupted immunosuppression of immune cells in the tumor microenvironment (TME) and tumor specificity of stromal cells (3). For example, chemotherapy, while killing tumor cells, stimulates other cells in the TME to release signals that promote tumor growth, ultimately resulting in treatment tolerance (4). Drug development with specific targeting of tumor cells and the TME will be a promising approach to tumor therapy. Thus, several studies have focused on the transformation of oncolytic adenoviruses (5,6), which can specifically target tumor cells and retain the efficacy of the drug at the tumor site. Gene therapy is a novel approach to cancer treatment (7), which aimed to target any aspect of tumor occurrence (8). Thus, in terms of genetic modification, several strategies have been adopted to overcome obstacles and reverse the TME.

Oncolytic adenovirus

An adenovirus is a non-enveloped double-stranded DNA virus with a symmetrical icosahedral structure (9). The genome is ~36 kb in length and can encode >40 gene products (10). These gene products are divided into three subtypes based on their transcription start time, including early, middle and late stages (11). Early gene products are predominantly responsible for coding gene regulation, including the E region, while late gene products are predominantly responsible for coding structural proteins, including the L region (12,13). Among adenovirus subtypes, adenovirus serotype 3 (Ad.3) and adenovirus serotype 5 (Ad.5) are the most commonly studied subtypes, and Ad.5 is the most commonly used subtype (14). Following infection, the oncolytic adenovirus initially recognizes specific receptors on the surface of tumor cells and triggers their internalization (15). Subsequently, it enters tumor cells, viral genomes migrate to the nucleus through microtubules, and early viral proteins in the E1 region immediately begin to be transcribed (16). The protein binds to Rb to release the transcription factor, E2F, which also activates the cell cycle, allowing oncolytic adenovirus-infected cells to enter the S phase (6,17). Concurrently, the E1A protein maintains p53 stability and inhibits tumor growth by relying on the p53 pathway (18). The release of E2F also triggers the coordinated activation of viral genes, which results in the production of new virions, the lysis of infected cells and the spread of viral offspring (19). The oncolytic adenovirus continuously replicates in tumor cells, eventually lysing tumor cells and infecting other tumor cells via the same mechanism of action (2023). Due to the large loading capacity of the oncolytic adenovirus vector, therapeutic genes are commonly inserted into the adenovirus vector (24,25). Due to the continuous replication and accumulation of adenoviruses in tumor cells, therapeutic genes are expressed and thus spread, playing a synergistic antitumor role (26).

TME of solid tumors

The TME is the internal environment for the growth of tumors, which includes tumor cells (27,28), stromal cells (tumor-associated vascular endothelial cells and tumor-associated fibroblasts), immune cells [T lymphocytes and B lymphocytes, tumor-associated macrophages (TAMs), dendritic cells (DCs) and natural killer (NK) cells], the extracellular matrix (ECM) and signaling molecules such as IL-4 and IL-10 (2931). The ECM includes various proteins, glycoproteins, proteoglycans and other biochemical substances, which regulate vascular endothelial cells and fibroblasts, and promote tumor growth and cell migration (32) (Fig. 1). In the TME, tumor blood vessels are constantly supplying oxygen and nutrients to support tumor growth (28,3234). When the tumor is exposed to hypoxic conditions locally, tumor blood vessels receive signal stimulation and generate branches from existing blood vessels (35,36). However, the structure of these tumor vessels differs from that of normal vessels, with absence of basement membranes, uneven diameter and size of the tubes, and short circuit of arteries and veins, resulting in tumor interstitial hypertension (37,38). Under hypoxic conditions, tumor cells undergo glycolysis and produce more lactic acid, which lowers the pH of the TME (39). Proton transport channels exist in all parts of tumor tissues, which transfer the metabolized H+ out of tumor tissues and maintain the pH in the TME (4042). However, pH reduction in normal tissues results in necrosis, which is more conducive to tumor metastasis and growth (43). Among the myeloid progenitors cells located in the TME, myeloid-derived suppressor cells (MDSCs), mast cells and most TAMs play key roles in promoting tumor development (44). MDSCs are immunosuppressive precursors of DCs, macrophages and granulocytes (35,45). MDSCs maintain a normal tissue dynamic balance in response to a variety of systemic infections and injuries (33). Several animal models have demonstrated that MDSCs can promote tumor angiogenesis and disrupt the main mechanisms of immune surveillance by interfering with antigen presentation, T-cell activation and NK cell killing of DCs (46,47). It has also been reported that mast cells are recruited into the tumor, where they release factors that promote endothelial cell proliferation to promote tumor angiogenesis (4850). Increasing evidence suggests that microenvironment-mediated external stimulation plays a key role in tumor cell survival and drug resistance (28,51). The complexity of the TME makes it difficult for the traditional oncolytic virus to reverse the conditions set by the TME while targeting tumor cells (27,52). The traditional oncolytic virus can only inhibit the growth of tumor cells to a certain extent.

Owing to the constant improvement of genetic engineering techniques, it is getting easier to develop oncolytic adenovirus constructs with required properties. Preclinical trials involve wild-type and recombinant oncolytic adenovirus (Table I), aimed to reverse the TME while suppressing tumor cells (6,53) (Fig. 2). Several oncolytic adenoviruses are currently undergoing clinical trials as antitumor agents, and notably some progress has been made in reversing the TME (Table II). An ongoing clinical trial is testing RGD (Delta-24-rgd), a genetically modified oncolytic adenovirus, as an agent against glioma. The first results obtained in the phase I trials indicate that 20% of patients showed durable responses and CD8+ T cells infiltrated the tumor in large quantities (54). TILT-123 in preclinical studies altered the cytokine balance in the TME towards Th1 and resulted in a significant increase of the survival rate in severe combined immunodeficiency (SCID) mice with human tumors (55,56). The currently ongoing phase I trial is recruiting patients with solid tumors to evaluate the safety.

Table I.

Partial oncolytic adenovirus trials to reverse the tumor microenvironment.

Table I.

Partial oncolytic adenovirus trials to reverse the tumor microenvironment.

Oncolytic adenovirusGene modificationTarget cells in TMETarget tumor cells(Refs.)
Ad3-hTERT-CMV-hCD40LCD40LDCsA549(64)
LOAd703CD40LDCsA549(65)
Ad5 [i/ppt-sNAP]HP-NAPDCsLNCaP(69)
ONCOS-102GM-CSFMacrophages, CD8+ T cellsAB12(75)
Ad-CD-GMCSFGM-CSFMacrophages, CD8+ T cellsColon cancer cell line(5)
SG635-SFA signal regulatory protein-α (SIRPα)-IgG1 Fc fusion geneMacrophagesSK-OV3, HO8910(77)
MMAD-IL-13IL-13MacrophagesCal-27, SCC-4, Tca8113(78)
EnAdBiTEs/TriTEsMacrophagesDLD-1(79)
Ad-CCL21-IL21CCL21DCsPC-3M, THP-1, HeLa, Caco-2(82)
Ad-CCL21-IL21IL-21NK cellsPC-3M, THP-1, HeLa, Caco-2(82)
Ad-E2F/IL15IL-15NK cells, CD8+ T cellsU87MG, BGC823, SW620, HCT116(84)
ICO15K-FBiTEFBiTECAFs, T cellsHT1080, A549(53)
AdCEAp-miR126/34amiR-126, miR-34aVascular endothelial cellsPancreatic adenocarcinoma(96)
VEGF-CRAdVEGFVascular endothelial cellsNCI-H28, NCI-H226, NCI-H20, NCI-H2452, MSTO-211H(97)
SKL002CTLA4T cellsHepG2, A549, Lovo, HeLa, HCT116, SW780(106)
Ad5-PCPD-1CD8+ T cellsHCC-LM3, H22, Hepa1-6, A549, B16-F10, LLC1(105)

[i] TME, tumor microenvironment; DCs, dendritic cells; NK, natural killer; CAFs, cancer-associated fibroblasts.

Table II.

Clinical trials of oncolytic adenoviruses affecting the tumor microenvironment (Jan. 2021, ClinicalTrials.gov).

Table II.

Clinical trials of oncolytic adenoviruses affecting the tumor microenvironment (Jan. 2021, ClinicalTrials.gov).

Trial numberOncolytic adenovirusStudy titleTargetPhase
NCT01582516Delta-24-rgdSafety Study of Replication-competent Adenovirus (Delta-24-rgd) in Patients With Recurrent GlioblastomaRecurrent glioblastomaI, II
NCT04695327TILT-123TNFα and IL-2 Coding Oncolytic Adenovirus TILT-123 Monotherapy (TUNIMO)Solid tumorI
NCT04217473TILT-123TNFalpha and Interleukin 2 Coding Oncolytic Adenovirus TILT-123 During TIL Treatment of Advanced Melanoma (TUNINTIL)Advanced melanomaI
NCT02705196LOAd703LOAd703 Oncolytic Virus Therapy for Pancreatic CancerPancreatic cancerI, II
NCT04123470LOAd703A Phase I/II Trial Investigating LOAd703 in Combination With Atezolizumab in Malignant MelanomaMalignant melanomaI, II
NCT03514836ONCOS-102A Phase I/II, Safety Clinical Trial of DCVAC/PCa and ONCOS-102 in Men With Metastatic Castration-resistant Prostate CancerProstate cancerI, II
NCT02963831ONCOS-102A Phase 1/2 Study to Investigate the Safety, Biologic and Anti-tumor Activity of ONCOS-102 in Combination With Durvalumab in Subjects With Advanced Peritoneal MalignanciesAdvanced peritoneal malignanciesI, II
NCT04053283NG-641First in Human Study With NG-641, an Oncolytic Transgene Expressing Adenoviral VectorMetastatic or advanced epithelial tumoursI
NCT03852511NG-350AFirst in Human Study of NG-350A (an Oncolytic Adenoviral Vector Which Expresses an Anti-CD40 Antibody) (FORTITUDE)Metastatic or advanced epithelial cancerI
NCT04097002ORCA-010First in Man Clinical Study to Evaluate Safety and Tolerability of an Oncolytic Adenovirus in Prostate Cancer Patients.Prostate cancerI, II

Adenovirus modification combined with immune cells in the TME

DCs

DCs are derived from the bone marrow and play a key role in inducing and maintaining antitumor immunity (57,58). Infiltration of mature DCs into the tumor can enhance immune activation and increase the recruitment of antitumor immune effector cells and pathways (58,59). However, in the TME, the antigen-presenting function of DCs may be lost or inefficient (60). Tumor cells can inhibit the function of DCs or change the TME by recruiting immunosuppressive DCs (61). CD40 is a member of the tumor necrosis factor receptor family and is expressed in DCs, which is a target for infiltrating T cells (62). CD40L is instantaneously expressed in T cells, which activates the maturation of DCs and triggers the immune response. Adenovirus delivery of CD40L induces DC activation, thereby inducing a Th1 immune response (63). The oncolytic virus restricts CD40L expression in cancer cells, thus decreasing systemic exposure and weakening the systemic immune response (64,65). Currently, there are already two phase I/II clinical trials involving LOAd703 that are recruiting patients. One of the studies is recruiting patients with pancreatic cancer to evaluate whether it supports the current treatment standards for pancreatic cancer and whether it can improve the survival rate of patients. Another study recruited patients with malignant melanoma and monitored their tumor response, immune response, virus shedding and survival rate. One of the major virulence factors of bacteria was Helicobacter pylori neutrophil-activating protein (HP-NAP), which is a TLR-2 agonist capable of chemotaxis of neutrophils, and monocytes and stimulates them to produce reactive oxygen species (66,67). HP-NAP also induced Th1-polarized immune responses by stimulating the secretion of interleukin (IL)-12 and IL-23 and other pro-inflammatory cytokines such as tumor necrosis factor (TNF)-α and IL-8 (68). Ag-presenting-HP-NAP-activated DCs effectively amplified Ag specific T cells, an important characteristic of mature DCs (69). HP-NAP-activated DCs resulted in Th1 cytokine secretion, with high IL-12 expression, relatively low IL-10 secretion and migrated to CCL19 (69).

Macrophages

TAMs are divided into specific M1-like macrophage subsets and specific M2-like macrophage subsets, and M1-like macrophage subsets are activated by the classical pathway and exert notable antitumor effects (70,71). In the TME, specific M2-like macrophage subsets are the most common, and their cytokines IL-6, TNF, IL-1 and IL-23 promote tumor growth and metastasis and silence T-cell function (72,73). Selective removal of specific M2-like macrophage subsets has become a research hotspot. Granulocyte-macrophage colony-stimulating factor (GM-CSF) can affect macrophages, promote their rapid differentiation to mature macrophages, prolong the life of mature macrophages, and enhance their cellular immune function (74). By inserting GM-CSF into the Ad5 vector, ONCS-102 induced notable antitumor immunity (75). ONCOS-102 is currently being assessed in two phase I clinical trials in advanced peritoneal malignancies and malignant pleural mesothelioma. Ad-CD-GMCSF is an adenovirus carrying cytomegalovirus promoter cytosine deaminase (CD) and GM-CSF (75). Adenovirus vectors expressing CD and GM-CSF are well tolerated in refractory tumors (5). CD47 is a cell surface transmembrane protein present in normal tissues (76). It is highly expressed in malignant tumor cells and binds to signaling regulatory protein-α (SIRPα) expressed on macrophages to inhibit macrophage phagocytosis, resulting in immune escape. SIRPα-FC fusion protein inserted into the oncolytic adenovirus vector blocks the binding of CD47 to macrophages, leading to a large increase in macrophage infiltration in tumor tissues, thus enhancing the antitumor effect (77). MMAD-IL13 loaded with IL13 demonstrated enhanced antitumor effects by inducing apoptosis in the TME in vivo, and decreased the percentage of specific M2-like macrophages (78). Scott et al (79) constructed a set of bivalent and trivalent T-cell adapters (BiTEs/TriTEs), which can specifically recognize CD3ε on T cells and the folate receptor or CD206 on specific M2-like macrophages. T-cell adapters were used to specifically direct the cytotoxicity of endogenous T cells to M2-like macrophages and deplete M2-like macrophages in tumor tissues. There was a significant increase in specific M1-like macrophage fraction among surviving macrophages, indicating a reversal of macrophage type in the TME (79).

NKs

NK cells have a notable antitumor effect in the initial stages of tumors, which can eliminate tumor cells (30). However, at the advanced tumor stage, NK cells gradually lose their antitumor ability and become dysfunctional (80). IL-21 is involved in NK cell differentiation (81), and the oncolytic adenovirus equipped with IL-21 exerts an obvious inhibitory effect on the proliferation of tumor cells (82). Similarly, NK cells can be activated by IL-15 (83), and oncolytic adenovirus (Ad-E2F/IL15), which expresses IL-15, can lyse tumor cells and coordinate with immune cells to enhance the antitumor response (84).

Adenovirus modification combined with stromal cells in the TME

Cancer-associated fibroblasts (CAFs)

CAFs are a major component of the tumor stroma, which regulate the TME and influence the behavior of tumor cells, and play crucial roles in the occurrence, development, invasion and metastasis of tumors (85,86). Fibroblasts in the TME secrete growth factors such as hepatocyte growth factor, fibroblast growth factor and CXCL12 (87), which promote the growth and survival of malignant cells, and act as chemokines to induce the migration of other cells into the TME (88,89). Concurrently, CAFs form a barrier in tumors and prevent the effective penetration and transmission of oncolytic virus, thus limiting its efficacy (90,91). By modifying oncolytic adenovirus, the effects of tumor cells and CAFs will be inhibited at the same time (92). Fibroblast activation protein-α (FAP) is highly expressed in CAFs. The FAP single-chain antibody is linked to an anti-human CD3 single-chain variable region (scFv) and loaded into oncolytic adenovirus. FAP scFv, while specifically recognizing and targeting CAFs, activates T cells and enhances T-cell-mediated cytotoxic effects on tumor-associated fibroblasts, thus weakening the cell barrier caused by CAFs and enhancing oncolytic activity (24,53).

Vascular endothelial cells

Blood vessels play a vital role in the development of tumors, providing nutrition and metastasis channels for tumor cells (93). The phenotype of vascular endothelial cells changes in the TME. Tumor cells secrete vascular endothelial growth factor (VEGF) and other endothelial growth factors to promote the generation of tumor neovascularization (94). Given that the downstream target gene of microRNA (miRNA/miR)-126 is VEGF (95), miR-126 is loaded into the oncolytic adenovirus, namely ADCEAP-miR126/34A, which decreases the generation of tumor blood vessels (96). Concurrently, the VEGF promoter is inserted into the adenovirus vector, targeting the tumor vascular endothelial cells via the same mechanism of action enhancing the oncolysis of adenovirus (97). IL-24 is a tumor suppressor molecule with broad-spectrum antitumor activity (98). It inhibits the growth of tumor cells by inhibiting tumor angiogenesis (99). A previous study has demonstrated that while expressing IL24, CRAd-IL24 significantly increased the release of virus particles and enhanced their antitumor effect (6).

Adenovirus modification combined with immune checkpoints in the TME

Checkpoint molecules are regulatory molecules that play an inhibitory role in the immune system and are critical to maintain tolerance, prevent an autoimmune response and minimize tissue damage by controlling the timing and intensity of the immune response (100,101). The expression of immunological checkpoint molecules on immune cells suppresses the immune cell function as the host fails to produce an effective antitumor immune response. There are numerous receptors on tumorigenic immune escape T cells, including co-stimulatory signal receptors that can stimulate T-cell proliferation, and co-inhibitory signal receptors that can inhibit T-cell proliferation (102). Immune checkpoint molecules are predominantly inhibitory molecules, in which the immune checkpoint on T cells suppresses the immune function of T cells, causing tumor escape (103). However, the clinical benefits of monotherapy with immune checkpoint inhibitors, such as anti-programmed death-1 antibody, are limited to small populations (104). Zhang et al (105) designed an adenovirus (Ad5-PC) to express a soluble fusion protein (programmed cell death protein 1/CD137L), which significantly increased the number of T lymphocytes in the TME and effectively improved the survival rate of tumor-bearing mice (105). After loading anti-cytotoxic T lymphocyte-associated antigen-4 (CTLA-4) antibody into the adenovirus vector, tumor cells were infected. CTLA-4 antibody was significantly expressed in tumor cells and its antitumor activity was significantly enhanced (106). Ad5/3-Δ24aCTLA4 can express CTLA-4 human intact monoclonal antibody, and in the normal donors and patients with advanced solid tumors in the peripheral blood mononuclear cells that were tested (107). Ad5/3-Δ24aCTLA4 significantly enhanced the immune response and activated the pro-apoptotic effect of T cells (107).

Outlook

Recently, the transformation technology of oncolytic adenoviruses has significantly progressed. However, increasing evidence suggest concerns about the tumor-promoting effect of the TME. Thus, the transformation of oncolytic viruses into TME has become a research hot spot; however, the in vitro simulation of the TME does not accurately reflect the human microenvironment. Cytokines and growth factors were added to the cell culture, either co-cultured with tumor cells and other cells, or with three-dimensional scaffoldings to simulate the TME. However, due to the complexity of the TME, it remains difficult to completely simulate the human TME completely in vitro. Currently, xenotransplantation of immunodeficient mice is the most commonly used animal experimental method for studying human tumors. Briefly, human tumor cells are inserted into mice; however, this fails to fully reflect the human TME. Several factors affect the oncolytic effect of an oncolytic virus. The strong immune response and severe cytokine storm when the virus first enters the host may be fatal. Subsequent challenges arise at later stages during elimination of oncolytic virus by the immune system of the host. It is hypothesized that the selection of oncolytic viruses will be the focus of future research, and it will be individualized, with the intent that the oncolytic virus most suitable for each patient can be selected, based on the characteristics of the tumor cells and the TME.

Acknowledgements

Not applicable.

Funding

This review was funded by Programs for the National Natural Scientific Foundation of China (grant no. 81430035), the Major National Science and Technology Projects-Major New Drug Creation (grant no. 2019ZX09301-132), the Changjiang Scholars and Innovative Research Team in University (grant no. IRT_15R13), and the Guangxi Science and Technology Base and Talent Special Project (grant no. AD17129003).

Availability of data and materials

Not applicable.

Authors' contributions

XW and YZ conceived the subject of the review. LZ designed the review. XW wrote the manuscript, performed the literature research as well as interpreted the relevant literature, and prepared the figures. LZ analyzed the review critically for important intellectual content. XW and LZ edited and revised the manuscript. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Giraldo NA, Sanchez-Salas R, Peske JD, Vano Y, Becht E, Petitprez F, Validire P, Ingels A, Cathelineau X, Fridman WH and Sautès-Fridman C: The clinical role of the TME in solid cancer. Br J Cancer. 120:45–53. 2019. View Article : Google Scholar : PubMed/NCBI

2 

Chen Y, Liu J, Wang W, Xiang L, Wang J, Liu S, Zhou H and Guo Z: High expression of hnRNPA1 promotes cell invasion by inducing EMT in gastric cancer. Oncol Rep. 39:1693–1701. 2018.PubMed/NCBI

3 

Choi J, Gyamfi J, Jang H and Koo JS: The role of tumor-associated macrophage in breast cancer biology. Histol Histopathol. 33:133–145. 2018.PubMed/NCBI

4 

Sun Y: Tumor microenvironment and cancer therapy resistance. Cancer Lett. 380:205–215. 2016. View Article : Google Scholar : PubMed/NCBI

5 

Akbulut H, Coleri A, Sahin G, Tang Y and Icli F: A bicistronic adenoviral vector carrying cytosine deaminase and granulocyte-macrophage colony-stimulating factor increases the therapeutic efficacy of cancer gene therapy. Hum Gene Ther. 30:999–1007. 2019. View Article : Google Scholar : PubMed/NCBI

6 

Ashshi AM, El-Shemi AG, Dmitriev IP, Kashentseva EA and Curiel DT: Combinatorial strategies based on CRAd-IL24 and CRAd-ING4 virotherapy with anti-angiogenesis treatment for ovarian cancer. J Ovarian Res. 9:382016. View Article : Google Scholar : PubMed/NCBI

7 

Athanasopoulos T, Munye MM and Yanez-Munoz RJ: Nonintegrating gene therapy vectors. Hematol Oncol Clin North Am. 31:753–770. 2017. View Article : Google Scholar : PubMed/NCBI

8 

Salzman R, Cook F, Hunt T, Malech HL, Reilly P, Foss-Campbell B and Barrett D: Addressing the value of gene therapy and enhancing patient access to transformative treatments. Mol Ther. 26:2717–2726. 2018. View Article : Google Scholar : PubMed/NCBI

9 

Stasiak AC and Stehle T: Human adenovirus binding to host cell receptors: A structural view. Med Microbiol Immunol. 209:325–333. 2020. View Article : Google Scholar : PubMed/NCBI

10 

Greber UF and Flatt JW: Adenovirus entry: From infection to immunity. Annu Rev Virol. 6:177–197. 2019. View Article : Google Scholar : PubMed/NCBI

11 

Hoeben RC and Uil TG: Adenovirus DNA replication. Cold Spring Harb Perspect Biol. 5:a0130032013. View Article : Google Scholar : PubMed/NCBI

12 

Gao J, Zhang W and Ehrhardt A: Expanding the spectrum of adenoviral vectors for cancer therapy. Cancers (Basel). 12:11392020. View Article : Google Scholar

13 

Ip WH and Dobner T: Cell transformation by the adenovirus oncogenes E1 and E4. FEBS Lett. 594:1848–1860. 2020. View Article : Google Scholar : PubMed/NCBI

14 

Bradley RR, Maxfield LF, Lynch DM, Iampietro MJ, Borducchi EN and Barouch DH: Adenovirus serotype 5-specific neutralizing antibodies target multiple hexon hypervariable regions. J Virol. 86:1267–1272. 2012. View Article : Google Scholar : PubMed/NCBI

15 

Niemann J and Kuhnel F: Oncolytic viruses: Adenoviruses. Virus Genes. 53:700–706. 2017. View Article : Google Scholar : PubMed/NCBI

16 

Machitani M, Katayama K, Sakurai F, Matsui H, Yamaguchi T, Suzuki T, Miyoshi H, Kawabata K and Mizuguchi H: Development of an adenovirus vector lacking the expression of virus-associated RNAs. J Control Release. 154:285–289. 2011. View Article : Google Scholar : PubMed/NCBI

17 

Zheng Y, Stamminger T and Hearing P: E2F/Rb family proteins mediate interferon induced repression of adenovirus immediate early transcription to promote persistent viral infection. PLoS Pathog. 12:e10054152016. View Article : Google Scholar : PubMed/NCBI

18 

Yamauchi S, Zhong B, Kawamura K, Yang S, Kubo S, Shingyoji M, Sekine I, Tada Y, Tatsumi K, Shimada H, et al: Cytotoxicity of replication-competent adenoviruses powered by an exogenous regulatory region is not linearly correlated with the viral infectivity/gene expression or with the E1A-activating ability but is associated with the p53 genotypes. BMC Cancer. 17:6222017. View Article : Google Scholar : PubMed/NCBI

19 

Cervera-Carrascon V, Quixabeira DCA, Havunen R, Santos JM, Kutvonen E, Clubb JHA, Siurala M, Heiniö C, Zafar S, Koivula T, et al: Comparison of clinically relevant oncolytic virus platforms for enhancing T cell therapy of solid tumors. Mol Ther Oncolytics. 17:47–60. 2020. View Article : Google Scholar : PubMed/NCBI

20 

Huang H, Liu Y, Liao W, Cao Y, Liu Q, Guo Y, Lu Y and Xie Z: Oncolytic adenovirus programmed by synthetic gene circuit for cancer immunotherapy. Nat Commun. 10:48012019. View Article : Google Scholar : PubMed/NCBI

21 

Rosewell Shaw A and Suzuki M: Recent advances in oncolytic adenovirus therapies for cancer. Curr Opin Virol. 21:9–15. 2016. View Article : Google Scholar : PubMed/NCBI

22 

Ran L, Tan X, Li Y, Zhang H, Ma R, Ji T, Dong W, Tong T, Liu Y, Chen D, et al: Delivery of oncolytic adenovirus into the nucleus of tumorigenic cells by tumor microparticles for virotherapy. Biomaterials. 89:56–66. 2016. View Article : Google Scholar : PubMed/NCBI

23 

Tazawaa H, Kagawab S and Fujiwarab T: Oncolytic adenovirus-induced autophagy: Tumor-suppressive effect and molecular basis. Acta Med Okayama. 67:333–342. 2013.PubMed/NCBI

24 

Freedman JD, Duffy MR, Lei-Rossmann J, Muntzer A, Scott EM, Hagel J, Campo L, Bryant RJ, Verrill C, Lambert A, et al: An oncolytic virus expressing a T-cell engager simultaneously targets cancer and immunosuppressive stromal cells. Cancer Res. 78:6852–6865. 2018. View Article : Google Scholar : PubMed/NCBI

25 

Jung KH, Choi IK, Lee HS, Yan HH, Son MK, Ahn HM, Hong J, Yun CO and Hong SS: Oncolytic adenovirus expressing relaxin (YDC002) enhances therapeutic efficacy of gemcitabine against pancreatic cancer. Cancer Lett. 396:155–166. 2017. View Article : Google Scholar : PubMed/NCBI

26 

Yokoda RT, Nagalo BM and Borad MJ: Oncolytic adenoviruses in gastrointestinal cancers. Biomedicines. 6:332018. View Article : Google Scholar

27 

Quail DF and Joyce JA: Microenvironmental regulation of tumor progression and metastasis. Nat Med. 19:1423–1437. 2013. View Article : Google Scholar : PubMed/NCBI

28 

Belli C, Trapani D, Viale G, D'Amico P, Duso BA, Della Vigna P, Orsi F and Curigliano G: Targeting the microenvironment in solid tumors. Cancer Treat Rev. 65:22–32. 2018. View Article : Google Scholar : PubMed/NCBI

29 

Zhong S, Jeong JH, Chen Z, Chen Z and Luo JL: Targeting tumor microenvironment by small-molecule inhibitors. Transl Oncol. 13:57–69. 2020. View Article : Google Scholar : PubMed/NCBI

30 

Terren I, Orrantia A, Vitalle J, Zenarruzabeitia O and Borrego F: NK cell metabolism and tumor microenvironment. Front Immunol. 10:22782019. View Article : Google Scholar : PubMed/NCBI

31 

Maimela NR, Liu S and Zhang Y: Fates of CD8+ T cells in tumor microenvironment. Comput Struct Biotechnol J. 17:1–13. 2018. View Article : Google Scholar : PubMed/NCBI

32 

Najafi M, Farhood B and Mortezaee K: Extracellular matrix (ECM) stiffness and degradation as cancer drivers. J Cell Biochem. 120:2782–2790. 2019. View Article : Google Scholar : PubMed/NCBI

33 

Guo S and Deng CX: Effect of stromal cells in tumor microenvironment on metastasis initiation. Int J Biol Sci. 14:2083–2093. 2018. View Article : Google Scholar : PubMed/NCBI

34 

Jang I and Beningo KA: Integrins, CAFs and mechanical forces in the progression of cancer. Cancers (Basel). 11:7212019. View Article : Google Scholar

35 

Jarosz-Biej M, Smolarczyk R, Cichon T and Kulach N: Tumor microenvironment as A ‘Game Changer’ in cancer radiotherapy. Int J Mol Sci. 20:32122019. View Article : Google Scholar

36 

Nishide S, Uchida J, Matsunaga S, Tokudome K, Yamaguchi T, Kabei K, Moriya T, Miura K, Nakatani T and Tomita S: Prolyl-hydroxylase inhibitors reconstitute tumor blood vessels in mice. J Pharmacol Sci. 143:122–126. 2020. View Article : Google Scholar : PubMed/NCBI

37 

Carretero R, Sektioglu IM, Garbi N, Salgado OC, Beckhove P and Hammerling GJ: Eosinophils orchestrate cancer rejection by normalizing tumor vessels and enhancing infiltration of CD8(+) T cells. Nat Immunol. 16:609–617. 2015. View Article : Google Scholar : PubMed/NCBI

38 

Zhao H, Tian X, He L, Li Y, Pu W, Liu Q, Tang J, Wu J, Cheng X, Liu Y, et al: Apj+ vessels drive tumor growth and represent a tractable therapeutic target. Cell Rep. 25:1241–1254.e5. 2018. View Article : Google Scholar : PubMed/NCBI

39 

Brand A, Singer K, Koehl GE, Kolitzus M, Schoenhammer G, Thiel A, Matos C, Bruss C, Klobuch S, Peter K, et al: LDHA-Associated lactic acid production blunts tumor immunosurveillance by T and NK cells. Cell Metab. 24:657–671. 2016. View Article : Google Scholar : PubMed/NCBI

40 

Fischer K, Hoffmann P, Voelkl S, Meidenbauer N, Ammer J, Edinger M, Gottfried E, Schwarz S, Rothe G, Hoves S, et al: Inhibitory effect of tumor cell-derived lactic acid on human T cells. Blood. 109:3812–3819. 2007. View Article : Google Scholar : PubMed/NCBI

41 

Colegio OR, Chu NQ, Szabo AL, Chu T, Rhebergen AM, Jairam V, Cyrus N, Brokowski CE, Eisenbarth SC, Phillips GM, et al: Functional polarization of tumour-associated macrophages by tumour-derived lactic acid. Nature. 513:559–563. 2014. View Article : Google Scholar : PubMed/NCBI

42 

Ohashi T, Aoki M, Tomita H, Akazawa T, Sato K, Kuze B, Mizuta K, Hara A, Nagaoka H, Inoue N and Ito Y: M2-like macrophage polarization in high lactic acid-producing head and neck cancer. Cancer Sci. 108:1128–1134. 2017. View Article : Google Scholar : PubMed/NCBI

43 

Urbanska K and Orzechowski A: Unappreciated role of LDHA and LDHB to control apoptosis and autophagy in tumor cells. Int J Mol Sci. 20:20852019. View Article : Google Scholar

44 

Fleming V, Hu X, Weber R, Nagibin V, Groth C, Altevogt P, Utikal J and Umansky V: Targeting myeloid-derived suppressor cells to bypass tumor-induced immunosuppression. Front Immunol. 9:3982018. View Article : Google Scholar : PubMed/NCBI

45 

Hinshaw DC and Shevde LA: The tumor microenvironment innately modulates cancer progression. Cancer Res. 79:4557–4566. 2019. View Article : Google Scholar : PubMed/NCBI

46 

Safarzadeh E, Orangi M, Mohammadi H, Babaie F and Baradaran B: Myeloid-derived suppressor cells: Important contributors to tumor progression and metastasis. J Cell Physiol. 233:3024–3036. 2018. View Article : Google Scholar : PubMed/NCBI

47 

Dysthe M and Parihar R: Myeloid-Derived suppressor cells in the tumor microenvironment. Adv Exp Med Biol. 1224:117–140. 2020. View Article : Google Scholar : PubMed/NCBI

48 

Ribatti D, Tamma R and Crivellato E: Cross talk between natural killer cells and mast cells in tumor angiogenesis. Inflamm Res. 68:19–23. 2019. View Article : Google Scholar : PubMed/NCBI

49 

Albini A, Bruno A, Noonan DM and Mortara L: Contribution to tumor angiogenesis from innate immune cells within the tumor microenvironment: Implications for immunotherapy. Front Immunol. 9:5272018. View Article : Google Scholar : PubMed/NCBI

50 

Kabiraj A, Jaiswal R, Singh A, Gupta J, Singh A and Samadi FM: Immunohistochemical evaluation of tumor angiogenesis and the role of mast cells in oral squamous cell carcinoma. J Cancer Res Ther. 14:495–502. 2018. View Article : Google Scholar : PubMed/NCBI

51 

Shee K, Yang W, Hinds JW, Hampsch RA, Varn FS, Traphagen NA, Patel K, Cheng C, Jenkins NP, Kettenbach AN, et al: Therapeutically targeting tumor microenvironment-mediated drug resistance in estrogen receptor-positive breast cancer. J Exp Med. 215:895–910. 2018. View Article : Google Scholar : PubMed/NCBI

52 

Binnewies M, Roberts EW, Kersten K, Chan V, Fearon DF, Merad M, Coussens LM, Gabrilovich DI, Ostrand-Rosenberg S, Hedrick CC, et al: Understanding the tumor immune microenvironment (TIME) for effective therapy. Nat Med. 24:541–550. 2018. View Article : Google Scholar : PubMed/NCBI

53 

de Sostoa J, Fajardo CA, Moreno R, Ramos MD, Farrera-Sal M and Alemany R: Targeting the tumor stroma with an oncolytic adenovirus secreting a fibroblast activation protein-targeted bispecific T-cell engager. J Immunother Cancer. 7:192019. View Article : Google Scholar : PubMed/NCBI

54 

Lang FF, Conrad C, Gomez-Manzano C, Yung WKA, Sawaya R, Weinberg JS, Prabhu SS, Rao G, Fuller GN, Aldape KD, et al: Phase I Study of DNX-2401 (Delta-24-RGD) Oncolytic Adenovirus: Replication and immunotherapeutic effects in recurrent malignant glioma. J Clin Oncol. 36:1419–1427. 2018. View Article : Google Scholar : PubMed/NCBI

55 

Havunen R, Siurala M, Sorsa S, Grönberg-Vähä-Koskela S, Behr M, Tähtinen S, Santos JM, Karell P, Rusanen J, Nettelbeck DM, et al: Oncolytic adenoviruses armed with tumor necrosis factor alpha and interleukin-2 enable successful adoptive cell therapy. Mol Ther Oncolytics. 4:77–86. 2016. View Article : Google Scholar : PubMed/NCBI

56 

Santos JM, Cervera-Carrascon V, Havunen R, Zafar S, Siurala M, Sorsa S, Anttila M, Kanerva A and Hemminki A: Adenovirus coding for interleukin-2 and tumor necrosis factor alpha replaces lymphodepleting chemotherapy in adoptive T cell therapy. Mol Ther. 26:2243–2254. 2018. View Article : Google Scholar : PubMed/NCBI

57 

Lee YS and Radford KJ: The role of dendritic cells in cancer. Int Rev Cell Mol Biol. 348:123–178. 2019. View Article : Google Scholar : PubMed/NCBI

58 

Palucka K and Banchereau J: Cancer immunotherapy via dendritic cells. Nat Rev Cancer. 12:265–277. 2012. View Article : Google Scholar : PubMed/NCBI

59 

Parnas O, Jovanovic M, Eisenhaure TM, Herbst RH, Dixit A, Ye CJ, Przybylski D, Platt RJ, Tirosh I, Sanjana NE, et al: A genome-wide CRISPR screen in primary immune cells to dissect regulatory networks. Cell. 162:675–686. 2015. View Article : Google Scholar : PubMed/NCBI

60 

Oh DS and Lee HK: Autophagy protein ATG5 regulates CD36 expression and anti-tumor MHC class II antigen presentation in dendritic cells. Autophagy. 15:2091–2106. 2019. View Article : Google Scholar : PubMed/NCBI

61 

Chen L, Hasni MS, Jondal M and Yakimchuk K: Modification of anti-tumor immunity by tolerogenic dendritic cells. Autoimmunity. 50:370–376. 2017. View Article : Google Scholar : PubMed/NCBI

62 

Karnell JL, Rieder SA, Ettinger R and Kolbeck R: Targeting the CD40-CD40L pathway in autoimmune diseases: Humoral immunity and beyond. Adv Drug Deliv Rev. 141:92–103. 2019. View Article : Google Scholar : PubMed/NCBI

63 

Vitale LA, Thomas LJ, He LZ, O'Neill T, Widger J, Crocker A, Sundarapandiyan K, Storey JR, Forsberg EM, Weidlick J, et al: Development of CDX-1140, an agonist CD40 antibody for cancer immunotherapy. Cancer Immunol Immunother. 68:233–245. 2019. View Article : Google Scholar : PubMed/NCBI

64 

Zafar S, Sorsa S, Siurala M, Hemminki O, Havunen R, Cervera-Carrascon V, Santos JM, Wang H, Lieber A, De Gruijl T, et al: CD40L coding oncolytic adenovirus allows long-term survival of humanized mice receiving dendritic cell therapy. Oncoimmunology. 7:e14908562018. View Article : Google Scholar : PubMed/NCBI

65 

Eriksson E, Milenova I, Wenthe J, Moreno R, Alemany R and Loskog A: IL-6 signaling blockade during CD40-mediated immune activation favors antitumor factors by reducing TGF-β, collagen type I, and PD-L1/PD-1. J Immunol. 202:787–798. 2019. View Article : Google Scholar : PubMed/NCBI

66 

Guo X, Ding C, Lu J, Zhou T, Liang T, Ji Z, Xie P, Liu X and Kang Q: HP-NAP ameliorates OXA-induced atopic dermatitis symptoms in mice. Immunopharmacol Immunotoxicol. 42:416–422. 2020. View Article : Google Scholar : PubMed/NCBI

67 

Codolo G, Fassan M, Munari F, Volpe A, Bassi P, Rugge M, Pagano F, D'Elios MM and de Bernard M: HP-NAP inhibits the growth of bladder cancer in mice by activating a cytotoxic Th1 response. Cancer Immunol Immunother. 61:31–40. 2012. View Article : Google Scholar : PubMed/NCBI

68 

D'Elios MM, Amedei A, Cappon A, Del Prete G and de Bernard M: The neutrophil-activating protein of Helicobacter pylori (HP-NAP) as an immune modulating agent. FEMS Immunol Med Microbiol. 50:157–164. 2007. View Article : Google Scholar : PubMed/NCBI

69 

Ramachandran M, Jin C, Yu D, Eriksson F and Essand M: Vector-encoded Helicobacter pylori neutrophil-activating protein promotes maturation of dendritic cells with Th1 polarization and improved migration. J Immunol. 193:2287–2296. 2014. View Article : Google Scholar : PubMed/NCBI

70 

Mantovani A, Marchesi F, Malesci A, Laghi L and Allavena P: Tumour-associated macrophages as treatment targets in oncology. Nat Rev Clin Oncol. 14:399–416. 2017. View Article : Google Scholar : PubMed/NCBI

71 

Ubil E, Caskey L, Holtzhausen A, Hunter D, Story C and Earp HS: Tumor-secreted Pros1 inhibits macrophage M1 polarization to reduce antitumor immune response. J Clin Invest. 128:2356–2369. 2018. View Article : Google Scholar : PubMed/NCBI

72 

Myers KV, Amend SR and Pienta KJ: Targeting Tyro3, Axl and MerTK (TAM receptors): Implications for macrophages in the tumor microenvironment. Mol Cancer. 18:942019. View Article : Google Scholar : PubMed/NCBI

73 

Huang YJ, Yang CK, Wei PL, Huynh TT, Whang-Peng J, Meng TC, Hsiao M, Tzeng YM, Wu AT and Yen Y: Ovatodiolide suppresses colon tumorigenesis and prevents polarization of M2 tumor-associated macrophages through YAP oncogenic pathways. J Hematol Oncol. 10:602017. View Article : Google Scholar : PubMed/NCBI

74 

Cho H, Seo Y, Loke KM, Kim SW, Oh SM, Kim JH, Soh J, Kim HS, Lee H, Kim J, et al: Cancer-Stimulated CAFs enhance monocyte differentiation and protumoral TAM activation via IL6 and GM-CSF secretion. Clin Cancer Res. 24:5407–5421. 2018. View Article : Google Scholar : PubMed/NCBI

75 

Kuryk L, Moller AW, Garofalo M, Cerullo V, Pesonen S, Alemany R and Jaderberg M: Antitumor-specific T-cell responses induced by oncolytic adenovirus ONCOS-102 (AdV5/3-D24-GM-CSF) in peritoneal mesothelioma mouse model. J Med Virol. 90:1669–1673. 2018. View Article : Google Scholar : PubMed/NCBI

76 

Hayat SMG, Biancon V, Pirro M, Jaafari MR, Hatamipour M and Sahebkar A: CD47 role in the immune system and application to cancer therapy. Cell Oncol (Dordr.). 43:19–30. 2020. View Article : Google Scholar : PubMed/NCBI

77 

Huang Y, Lv SQ, Liu PY, Ye ZL, Yang H, Li LF, Zhu HL, Wang Y, Cui LZ, Jiang DQ, et al: A SIRPα-Fc fusion protein enhances the antitumor effect of oncolytic adenovirus against ovarian cancer. Mol Oncol. 14:657–668. 2020. View Article : Google Scholar : PubMed/NCBI

78 

Zhang KL, Li RP, Zhang BP, Gao ST, Li B, Huang CJ, Cao R, Cheng JY, Xie XD, Yu ZH and Feng XY: Efficacy of a new oncolytic adenovirus armed with IL-13 against oral carcinoma models. Onco Targets Ther. 12:6515–6523. 2019. View Article : Google Scholar : PubMed/NCBI

79 

Scott EM, Jacobus EJ, Lyons B, Frost S, Freedman JD, Dyer A, Khalique H, Taverner WK, Carr A, Champion BR, et al: Bi- and tri-valent T cell engagers deplete tumour-associated macrophages in cancer patient samples. J Immunother Cancer. 7:3202019. View Article : Google Scholar : PubMed/NCBI

80 

Yao C, Ni Z, Gong C, Zhu X, Wang L, Xu Z, Zhou C, Li S, Zhou W, Zou C and Zhu S: Rocaglamide enhances NK cell-mediated killing of non-small cell lung cancer cells by inhibiting autophagy. Autophagy. 14:1831–1844. 2018. View Article : Google Scholar : PubMed/NCBI

81 

Davis MR, Zhu Z, Hansen DM, Bai Q and Fang Y: The role of IL-21 in immunity and cancer. Cancer Lett. 358:107–114. 2015. View Article : Google Scholar : PubMed/NCBI

82 

Li Y, Li YF, Si CZ, Zhu YH, Jin Y, Zhu TT, Liu MY and Liu GY: CCL21/IL21-armed oncolytic adenovirus enhances antitumor activity against TERT-positive tumor cells. Virus Res. 220:172–178. 2016. View Article : Google Scholar : PubMed/NCBI

83 

Rhode PR, Egan JO, Xu W, Hong H, Webb GM, Chen X, Liu B, Zhu X, Wen J, You L, et al: Comparison of the superagonist complex, ALT-803, to IL15 as cancer immunotherapeutics in animal models. Cancer Immunol Res. 4:49–60. 2016. View Article : Google Scholar : PubMed/NCBI

84 

Yan Y, Li S, Jia T, Du X, Xu Y, Zhao Y, Li L, Liang K, Liang W, Sun H and Li R: Combined therapy with CTL cells and oncolytic adenovirus expressing IL-15-induced enhanced antitumor activity. Tumour Biol. 36:4535–4543. 2015. View Article : Google Scholar : PubMed/NCBI

85 

Kubo N, Araki K, Kuwano H and Shirabe K: Cancer-associated fibroblasts in hepatocellular carcinoma. World J Gastroenterol. 22:6841–6850. 2016. View Article : Google Scholar : PubMed/NCBI

86 

Chen X and Song E: Turning foes to friends: Targeting cancer-associated fibroblasts. Nat Rev Drug Discov. 18:99–115. 2019. View Article : Google Scholar : PubMed/NCBI

87 

Feig C, Jones JO, Kraman M, Wells RJ, Deonarine A, Chan DS, Connell CM, Roberts EW, Zhao Q, Caballero OL, et al: Targeting CXCL12 from FAP-expressing carcinoma-associated fibroblasts synergizes with anti-PD-L1 immunotherapy in pancreatic cancer. Proc Natl Acad Sci USA. 110:20212–20217. 2013. View Article : Google Scholar : PubMed/NCBI

88 

Erdogan B, Ao M, White LM, Means AL, Brewer BM, Yang L, Washington MK, Shi C, Franco OE, Weaver AM, et al: Cancer-associated fibroblasts promote directional cancer cell migration by aligning fibronectin. J Cell Biol. 216:3799–3816. 2017. View Article : Google Scholar : PubMed/NCBI

89 

Erdogan B and Webb DJ: Cancer-associated fibroblasts modulate growth factor signaling and extracellular matrix remodeling to regulate tumor metastasis. Biochem Soc Trans. 45:229–236. 2017. View Article : Google Scholar : PubMed/NCBI

90 

Arwert EN, Milford EL, Rullan A, Derzsi S, Hooper S, Kato T, Mansfield D, Melcher A, Harrington KJ and Sahai E: STING and IRF3 in stromal fibroblasts enable sensing of genomic stress in cancer cells to undermine oncolytic viral therapy. Nat Cell Biol. 22:758–766. 2020. View Article : Google Scholar : PubMed/NCBI

91 

Puig-Saus C, Gros A, Alemany R and Cascallo M: Adenovirus i-leader truncation bioselected against cancer-associated fibroblasts to overcome tumor stromal barriers. Mol Ther. 20:54–62. 2012. View Article : Google Scholar : PubMed/NCBI

92 

Jing Y, Chavez V, Ban Y, Acquavella N, El-Ashry D, Pronin A, Chen X and Merchan JR: Molecular effects of stromal-selective targeting by uPAR-retargeted oncolytic virus in breast cancer. Mol Cancer Res. 15:1410–1420. 2017. View Article : Google Scholar : PubMed/NCBI

93 

Czekierdowska S, Stachowicz N, Chrosciel M and Czekierdowski A: Proliferation and maturation of intratumoral blood vessels in women with malignant ovarian tumors assessed with cancer stem cells marker nestin and platelet derived growth factor PDGF-B. Ginekol Pol. 88:120–128. 2017. View Article : Google Scholar : PubMed/NCBI

94 

Wang R, Chadalavada K, Wilshire J, Kowalik U, Hovinga KE, Geber A, Fligelman B, Leversha M, Brennan C and Tabar V: Glioblastoma stem-like cells give rise to tumour endothelium. Nature. 468:829–833. 2010. View Article : Google Scholar : PubMed/NCBI

95 

Wang S, Aurora AB, Johnson BA, Qi X, McAnally J, Hill JA, Richardson JA, Bassel-Duby R and Olson EN: The endothelial-specific microRNA miR-126 governs vascular integrity and angiogenesis. Dev Cell. 15:261–271. 2008. View Article : Google Scholar : PubMed/NCBI

96 

Feng SD, Mao Z, Liu C, Nie YS, Sun B, Guo M and Su C: Simultaneous overexpression of miR-126 and miR-34a induces a superior antitumor efficacy in pancreatic adenocarcinoma. Onco Targets Ther. 10:5591–5604. 2017. View Article : Google Scholar : PubMed/NCBI

97 

Harada A, Uchino J, Harada T, Nakagaki N, Hisasue J, Fujita M and Takayama K: Vascular endothelial growth factor promoter-based conditionally replicative adenoviruses effectively suppress growth of malignant pleural mesothelioma. Cancer Sci. 108:116–123. 2017. View Article : Google Scholar : PubMed/NCBI

98 

Chen QN, Chen X, Chen ZY, Nie FQ, Wei CC, Ma HW, Wan L, Yan S, Ren SN and Wang ZX: Long intergenic non-coding RNA 00152 promotes lung adenocarcinoma proliferation via interacting with EZH2 and repressing IL24 expression. Mol Cancer. 16:172017. View Article : Google Scholar : PubMed/NCBI

99 

Zhuo B, Wang R, Yin Y, Zhang H, Ma T, Liu F, Cao H and Shi Y: Adenovirus arming human IL-24 inhibits neuroblastoma cell proliferation in vitro and xenograft tumor growth in vivo. Tumour Biol. 34:2419–2426. 2013. View Article : Google Scholar : PubMed/NCBI

100 

Zhang Y and Zheng J: Functions of immune checkpoint molecules beyond immune evasion. Adv Exp Med Biol. 1248:201–226. 2020. View Article : Google Scholar : PubMed/NCBI

101 

Li K and Tian H: Development of small-molecule immune checkpoint inhibitors of PD-1/PD-L1 as a new therapeutic strategy for tumour immunotherapy. J Drug Target. 27:244–256. 2019. View Article : Google Scholar : PubMed/NCBI

102 

Kurachi M, Barnitz RA, Yosef N, Odorizzi PM, DiIorio MA, Lemieux ME, Yates K, Godec J, Klatt MG, Regev A, et al: The transcription factor BATF operates as an essential differentiation checkpoint in early effector CD8+ T cells. Nat Immunol. 15:373–383. 2014. View Article : Google Scholar : PubMed/NCBI

103 

Deng J, Wang ES, Jenkins RW, Li S, Dries R, Yates K, Chhabra S, Huang W, Liu H, Aref AR, et al: CDK4/6 inhibition augments antitumor immunity by enhancing T-cell activation. Cancer Discov. 8:216–233. 2018. View Article : Google Scholar : PubMed/NCBI

104 

Johnson DB, Sullivan RJ and Menzies AM: Immune checkpoint inhibitors in challenging populations. Cancer. 123:1904–1911. 2017. View Article : Google Scholar : PubMed/NCBI

105 

Zhang Y, Zhang H, Wei M, Mou T, Shi T, Ma Y, Cai X, Li Y, Dong J and Wei J: Recombinant adenovirus expressing a soluble fusion protein PD-1/CD137L subverts the suppression of CD8+ T cells in HCC. Mol Ther. 27:1906–1918. 2019. View Article : Google Scholar : PubMed/NCBI

106 

Du T, Shi G, Li YM, Zhang JF, Tian HW, Wei YQ, Deng H and Yu DC: Tumor-specific oncolytic adenoviruses expressing granulocyte macrophage colony-stimulating factor or anti-CTLA4 antibody for the treatment of cancers. Cancer Gene Ther. 21:340–348. 2014. View Article : Google Scholar : PubMed/NCBI

107 

Dias JD, Hemminki O, Diaconu I, Hirvinen M, Bonetti A, Guse K, Escutenaire S, Kanerva A, Pesonen S, Löskog A, et al: Targeted cancer immunotherapy with oncolytic adenovirus coding for a fully human monoclonal antibody specific for CTLA-4. Gene Ther. 19:988–998. 2012. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

April-2021
Volume 45 Issue 4

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wang X, Zhong L and Zhao Y: Oncolytic adenovirus: A tool for reversing the tumor microenvironment and promoting cancer treatment (Review). Oncol Rep 45: 49, 2021
APA
Wang, X., Zhong, L., & Zhao, Y. (2021). Oncolytic adenovirus: A tool for reversing the tumor microenvironment and promoting cancer treatment (Review). Oncology Reports, 45, 49. https://doi.org/10.3892/or.2021.8000
MLA
Wang, X., Zhong, L., Zhao, Y."Oncolytic adenovirus: A tool for reversing the tumor microenvironment and promoting cancer treatment (Review)". Oncology Reports 45.4 (2021): 49.
Chicago
Wang, X., Zhong, L., Zhao, Y."Oncolytic adenovirus: A tool for reversing the tumor microenvironment and promoting cancer treatment (Review)". Oncology Reports 45, no. 4 (2021): 49. https://doi.org/10.3892/or.2021.8000