Next Article in Journal
IL-6 Cytokine Family: A Putative Target for Breast Cancer Prevention and Treatment
Next Article in Special Issue
Friends with Benefits: Chemokines, Glioblastoma-Associated Microglia/Macrophages, and Tumor Microenvironment
Previous Article in Journal
Up and Down γ-Synuclein Transcription in Dopamine Neurons Translates into Changes in Dopamine Neurotransmission and Behavioral Performance in Mice
Previous Article in Special Issue
The Eclectic Nature of Glioma-Infiltrating Macrophages and Microglia
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Tumor-Associated Macrophages/Microglia in Glioblastoma Oncolytic Virotherapy: A Double-Edged Sword

1
Harvard Medical School, Boston, MA 02115, USA
2
Department of Neurosurgery, Brigham and Women’s Hospital, Boston, MA 02115, USA
3
Department of Neurosurgery, University Medicine Rostock, 18057 Rostock, Germany
4
Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(3), 1808; https://doi.org/10.3390/ijms23031808
Submission received: 1 January 2022 / Revised: 29 January 2022 / Accepted: 1 February 2022 / Published: 4 February 2022
(This article belongs to the Special Issue Macrophages in the Glioblastoma Tumor Microenvironment)

Abstract

:
Oncolytic virotherapy is a rapidly progressing field that uses oncolytic viruses (OVs) to selectively infect malignant cells and cause an antitumor response through direct oncolysis and stimulation of the immune system. Despite demonstrated pre-clinical efficacy of OVs in many cancer types and some favorable clinical results in glioblastoma (GBM) trials, durable increases in overall survival have remained elusive. Recent evidence has emerged that tumor-associated macrophage/microglia (TAM) involvement is likely an important factor contributing to OV treatment failure. It is prudent to note that the relationship between TAMs and OV therapy failures is complex. Canonically activated TAMs (i.e., M1) drive an antitumor response while also inhibiting OV replication and spread. Meanwhile, M2 activated TAMs facilitate an immunosuppressive microenvironment thereby indirectly promoting tumor growth. In this focused review, we discuss the complicated interplay between TAMs and OV therapies in GBM. We review past studies that aimed to maximize effectiveness through immune system modulation—both immunostimulatory and immunosuppressant—and suggest future directions to maximize OV efficacy.

1. Introduction

1.1. Oncolytic Viruses

Oncolytic virotherapy centers on engineered viruses that target neoplastic cells. Oncolytic viruses (OVs) have shown great promise in the treatment of various cancer types due to their selective replication in cancer cells and the subsequent induction of tumor cell death [1,2]. This is possibly secondary to tumor cells’ impaired mechanisms of viral clearance [3,4,5,6] and/or through the genetic modification of OVs which enhance malignant cell selectivity [7]. Once in the tumor cells, OVs work through a mixed mechanism of induction of oncolysis and stimulation of antitumor immune activity [1,8]. Direct destruction of cells occurs, and lysis leads to release of viral particles, cytokines, and other cellular contents, and ultimately the induction of an immune response [1]. OVs can also be engineered to enhance direct tumor lysis through the delivery of suicide genes [9,10,11]. Furthermore, viral infection stimulates antitumor immune activity; after cell lysis, danger-associated molecular patterns (DAMPs), pathogen-associated molecular patterns (PAMPs), and cytokines are released that simulate the innate immune system [1]. Antigen presenting cells (APCs) recruit CD4+ and CD8+ T cells to create an adaptive immune response against infected tumor cells [12]. Immunogenic cell death (ICD) is a more recently discovered cell death modality that involves specific, timed changes to the cell surface (CRT expression) followed by release of soluble DAMPs (e.g., HMGB1, HSP). Importantly, this increases the immunogenicity of dying/dead cancer cells, and has the ability to potentiate adaptive immune responses and target residual cancer cells/tissues [13,14].
Almost all oncolytic viruses have some preference for infecting tumor cells over normal cells [15]. However, genetic engineering can be used to enhance OVs’ natural preference for cancer cells. For example, viruses can be re-targeted (genetically altered for tumor-specific viral entry) by preventing replication in non-dividing cells [7]. OVs can also be engineered to be armed (containing therapeutic transgene variants) to increase immunogenic reactions [16]. For example, arming HSV with interleukin-12 (IL-12) [17] or IL-4 [18] has demonstrated increased potency.
For the treatment of glioma, three main viruses have been studied extensively: HSV, poliovirus, and adenovirus. HSV is the most broadly studied, with clinical trials in many cancer types including GBM [16]. HSV an attractive OV candidate for many reasons, including a large and highly stable genome, its potent cytolytic capability, high immunogenicity, and a convenient genome for genetic engineering; the availability of effective anti-herpetic drugs to treat adverse reactions is an added benefit [16,19]. Another common virus studied for glioma virotherapy is poliovirus. Therapeutic poliovirus (PVSRIPO) was created by combining a nonpathogenic poliovirus with a rhinovirus [20,21]. PVSRIPO demonstrated successful innate immune stimulation and cytotoxic cell recruitment in GBM [22,23]. Finally, adenoviruses have also been exploited for use in glioma. Adenovirus can function as a nonintegrating vector with relatively high capacity for gene delivery, including delivery of suicide genes [24,25,26,27]. It demonstrated successful induction of immune responses in GBM and caused direct oncolysis and autophagy [28].
Viral infection by any OV creates both an antiviral and antitumor response. In addition to direct oncolysis, OVs stimulate a host immune response. While tumor inflammation and immune cell recruitment is known to create an antitumor effect, it also prevents viral replication and distribution through its antiviral properties. The relative impact of these forces varies by cancer type [29]. Immune cell evasion is a classic characteristic of GBM [30], and OVs aim to reverse this by exposing the tumor to the innate and ultimately adaptive immune system.

1.2. Glioma-Associated Macrophages and Their Dichotomy

Tumor-associated macrophages/microglia (TAMs) are the most abundant non-neoplastic cell in the GBM tumor microenvironment (TME) and have been investigated as potential causes of OV therapy failure [31]. TAMs, which consist of both brain-resident microglia and bone marrow-derived myeloid cells from the periphery, constitute about 40% of the tumor mass in GBMs [32]. TAMs in the TME enhance tumor cell migration and invasion through secretion of chemotactic factors, enzymes, and cytokines [33,34]. As a result, there is a positive correlation between the number of TAMs and the malignancy of the brain tumor [35].
Classically, TAMs have been described in the M1/M2 dichotomy [36]. Although TAM phenotypes exist on a spectrum and are not truly dichotomous [37,38], this simplified classification provides a framework through which to understand their competing actions. Figure 1 shows a schematic representation of TAM polarization as well as the interaction with tumor growth and OVs. The classically activated M1 phenotype is activated by interferon gamma (IFN-γ) and lipopolysaccharides (LPS) through signal transducer and activator of transcription 1 (STAT1) activity [12,39]. M1 TAMs promote strong IL-12 mediated T helper 1 (Th1) responses and activate natural killer (NK) cells through pro-inflammatory cytokine production, including tumor-necrosis factor alpha (TNF-α), IL-β, IL-6, IL-8, IL-12, and IL-23, regulated in part by the nuclear factor kappa B (NF-κB) pathway [12,37,40,41,42]. They are also capable of phagocytosis and antigen processing and presentation, making them a bridge between innate and adaptive immune systems [12,39,43]. This creates a systemic and durable immunity to tumor cells [8].
M2 phenotype TAMs are alternatively activated by peroxisome proliferator-activated receptor-γ (PPARγ) and STAT6, which suppress the NF-κB pathway [39]. They promote strong Th2-associated effector functions and induce regulatory T cells (Tregs). Through the production of IL-1RA, IL-10, vascular endothelial growth factor (VEGF), and transforming growth factor beta (TGF-β), M2s stimulate tissue remodeling and tumor development [37,39,44,45,46,47,48]. They also lead to the resolution of inflammation through high endocytic clearance capacities and trophic factor synthesis, angiogenesis, and tumor proliferation and progression [12,33]. Overall, the phenotypes are primarily regulated through either interferon regulatory factor 4 (IRF4) or IRF5, competing through Toll-like receptor (TLR) signaling, and polarizing cells towards the M2 or M1 phenotype, respectively [49,50]. This demonstrates that the in vivo TAM phenotype is based on the dominant cytokines, trophic factors, and proteins in the microenvironment.

1.3. Glioma-Associated Macrophages and Immunosuppression

In GBM, there is general immunosuppression in the TME. Glioma cells and TAMs work symbiotically—glioma cells attract TAM infiltration and TAMs promote glioma growth and invasiveness [41,51,52]. TAMs are recruited to the TME by chemoattractants from glioma cells, such as CC chemokine ligand 2 (CCL2)[53,54] and soluble colony-stimulating factor 1 (sCSF-1) [55,56]. Polarization to M2 occurs through a variety of cytokines, including IL-10, IL-4, IL-6, macrophage colony stimulating factor (M-CSF), TGF-β, and prostaglandin E2, and the TAMs become immunosuppressive [37,51,52,57,58,59]. Interactions between TAMs and other immune cells in the TME provides additional mechanisms of immunosuppression. TAMs produce chemokines that recruit Tregs, and they both secrete IL-10, which interferes with IFN-γ production and impairs infiltrating T cells [60,61]. TAMs also upregulate several surface molecules that inhibit T cell activation and induce T cell apoptosis including cluster of differentiation 95 (CD95), CD70, and programmed cell death ligand 1 (PD-L1) [30,62]. This leads to fewer tumor-infiltrating immune effector cells (Teffector) and prevents an immune attack against the glioma [30,62].

2. Discussion

2.1. Polarizing TAMs towards the M1 Phenotype

Polarizing TAMs towards the M1 phenotype and creating a pro-inflammatory TME is a key mechanism of the OV-induced antitumor effect. This polarization and increased inflammatory response are seen in a variety of viruses in GBM, including HSV [63,64,65,66], adenoviruses [67,68,69], parvoviruses [70,71,72], and vaccinia viruses [73]. This coincides with an increased Teffector to Treg ratio, likely related to an IFN-γ influx [65]. Previous studies have utilized this mechanism in altering viruses to enhance the M1 phenotypic shift. IL-12 is the most commonly used cytokine to enhance antitumor efficacy of OVs [74,75]. As seen in Figure 1, IL-12 plays a role in the antitumor response, through induction of Th1 differentiation, stimulation of NK growth and cytotoxicity, IFN-γ release, and angiogenesis inhibition [75,76]. Arming HSV with IL-12 has demonstrated increase in IFN-γ and reduction in Tregs in tumors, along with increased survival in murine models [17,76,77,78,79]. Other cytokines have also been used, including FMS-like tyrosine kinase 3 ligand (Flt3L), which is associated with increases in intratumoral dendritic cells and CD8+ T cells [80,81] and improves survival in glioma-bearing mice [82]. The addition of IL-4 to HSV also increased infiltration of macrophages and CD4+ and CD8+ T cells into murine models of intracranial glioma and prolonged survival [18]. In contrast however, transfection of HSV expressing IL-10, which is anti-inflammatory, did not produce a significant survival advantage compared to saline-treated controls in the same animal model and did not result in increased infiltration of CD8+ T cells [18].

2.2. Combination Therapies with Checkpoint Inhibitors

Combination therapies have also been explored to enhance immunostimulation with OVs. Checkpoint inhibitors, such as anti-CTLA-4 (cytotoxic T-lymphocyte-associated protein 4) and anti-PD-1, have demonstrated efficacy in many cancer types. They enhance activation of T helper cells and effector cells while suppressing Tregs [83]. Anti-CTLA-4 and anti-PD-1 antibodies work through distinct and non-redundant inhibitory pathways on immune cells [65,83]. Unfortunately, they have not resulted in significant benefits in GBM clinical trials [84,85]. This is related in part to the immunologically “cold” and highly immunosuppressive TME [86]. However, triple combination of HSV armed with IL-12 along with anti-CTLA-4 and anti-PD-L1 increased influx of macrophages and M1 polarization and was very effective in curing both murine glioma models including aggressive carcinogen-induced tumors [65,87]. Immune checkpoint modulation has also been used in adenovirus armed with the immune costimulatory OX40 ligand, which activates lymphocytes and leads to proliferation of CD8+ T cells [88]. Adding anti-PD-L1 antibody with this OV also demonstrated significantly increased survival in mice with gliomas [88]. Adenovirus armed with costimulatory ligand glucocorticoid-induced tumor necrosis factor receptor (TNFR) family-related ligand (GITRL) also increased CD8+ T cell infiltration and increased survival in mice with gliomas [89]. Clearly, the inflammatory role of M1 TAMs, which go hand in hand with the various mechanisms of armed viruses, is crucial to effectiveness of OVs.

2.3. Combination Therapies with Immunosuppressive Medications

However, TAMs and M1 polarization also play a detrimental role in OV efficacy through their antiviral activity, which is not virus-specific [31]. Gliomas treated with HSV have shown intratumoral clearance of over 80% of viral particles shortly after delivery, which is associated with up-regulation and infiltration of TAMs [90,91]. TAMs can directly uptake viruses through endocytosis or reduce replication through secretion of antiviral cytokines [64,66,92]. Upon injection, there is an immediate M1 antiviral reaction competing with glioma cells for virus uptake [93,94]. Within 72 h of HSV injection, there is a significant decrease in the volume of tumor cells that contains HSV-mediated gene expression [91]. However, depletion of the innate immune response including peripheral phagocytic cells and brain microglia via immunosuppressants can enhance intratumoral uptake and spread of OV [31,91]. TAMs are also able to create a non-permissive barrier that prevents OV replication and dissemination, which leads to viral gene silencing [64]. This detrimental TAM-induced antiviral effect has been supported in studies that demonstrated that blocking STAT1/3 activity rescues OV replication and enhances the therapeutic efficacy of oncolytic virotherapy [64]. In addition, chemical ablation of TAMs in glioma-bearing rodent models enhanced the antitumoral effects of HSV [31]. Decreasing TAMs, despite their antitumoral proinflammatory effects, can increase OV uptake, replication, and efficacy.
Due to the known deleterious effects of TAMs related to early viral clearance, many studies have aimed to suppress recruitment of TAMs with OVs. For example, cyclophosphamide inhibits the production of IFN-γ by natural killer (NK) cells and reduces the concentration of TAMs [91]. Even residual TAMs demonstrated suppressed expression of antiviral cytokines [95,96]. This led to a 10-fold increase in intratumoral OV gene expression [91]. Preadministration of cyclophosphamide in athymic mouse models of human glioma, caused increased OV uptake and intratumor distribution allowing for reduced OV doses, reduced tumor burden, and increased survival [97]. Other ways to decrease IFN signaling and pro-inflammatory cytokine induction in tumor cells, therefore limiting TAM recruitment, includes administration of rapamycin to block integrin beta 1 receptors [98], which are expressed on the cell surfaces of macrophages [99]; pretreatment with the histone deacetylase inhibitor valproic acid [100]; or administration of cellular communication network factor 1 (CCN1) antibodies [101,102,103]. The addition of TGF-β can also suppress TAM recruitment by dampening the innate immune response through cell growth inhibition and apoptosis via transcriptional induction of genes, such as cyclin-dependent kinase inhibitors (CDKIs) [104,105]. This inhibits NK and TAM recruitment, activation, and function, thereby enhancing OV replication [105].
Other techniques have focused more on inhibiting TAMs that are present in the TME. Clodronate encapsulated in liposomes is taken up by phagocytic cells and results in intracellular accumulation and TAM apoptosis, thereby depleting the TAM population [106,107,108]. In murine GBM, this led to a five-fold increase in viral replication [31]. M1 TAM mechanisms can also be restricted, such as blocking brain angiogenesis inhibitor [63,109] or STAT1/3 activity [43,64,110]. In addition, depleting NK cells, which coordinate TAM activation in response to OV, has a beneficial effect [105,111].
Finally, some studies have investigated ways to bypass the antagonizing effects TAMs have on virotherapy regardless of their presence. OVs can be transported with carrier cells, protecting them from neutralization and opsonization and assisting with homing to the tumor site in studies with systemic injection [112,113,114,115,116]. Overall, finding ways preclinically to restrict TAM function, whether through reduced recruitment, reduced activation and function, or preventing interactions with OVs demonstrates that inhibiting TAMs has potential to benefit OV efficacy.

3. Future Directions

One of the key next steps is to investigate the combined mechanisms that prevent initial antiviral TAM actions, while still allowing for later TAM-directed antitumor responses. The balance between these two opposing functions in part determines the efficacy of OV therapy. However, modulating these responses, which often encompass overlapping immunological pathways, is challenging and poorly understood [8,117]. It is possible that many of the previously mentioned tactics can be combined to create both antiviral and antitumor responses. For example, selective and transient immunomodulation with immune-inhibiting therapeutics may still allow for sufficient tumoral infection to induce a robust antitumor response.

4. Conclusions

Understanding the mechanisms that inhibit and potentiate oncolytic virotherapy is necessary for this promising therapy to reach its full potential. Overall, TAMs function in OV therapy as a double-edged sword. They play a crucial role in the immune stimulation that creates the antitumor response generated by OVs. Initial innate immune responses orchestrate subsequent lasting adaptive immune responses. However, TAMs also inhibit efficient intratumoral viral distribution. Both immunostimulatory and immunosuppressant adjuvants have shown benefits in OV research. More research on combination therapies is necessary to find cooperative tactics. Harnessing TAMs to promote both antiviral and antitumor effects will optimize OV efficacy in the future.

Author Contributions

Conceptualization, J.D.B. and G.K.F.; writing—original draft preparation, S.E.B., J.D.B. and A.D.K.; writing—review and editing, F.A.G., N.V.K., O.A., Y.L., P.P.P., T.R.S., E.A.C., G.F.K. and J.D.B.; project administration, O.A., T.R.S., E.A.C., G.F.K. and J.D.B.; All authors have read and agreed to the published version of the manuscript.

Funding

GKF supported by Food and Drug Administration (R01FD006368 and R01FD005379), Cannonball Kids can-cer Foundation, and Hyundai Hope on Wheels. GKF and JDB supported by the Rally Foundation for Child-hood Cancer Research, CureSearch for Children’s Cancer, The V Foundation, Andrew McDonough B+ Foundation, the National Pediatric Cancer Foundation, and the Pediatric Cancer Research Foundation.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

J.D.B. has an equity position in Avidea Technologies, Inc., which is commercializing polymer-based drug delivery technologies for immunotherapeutic applications and has an equity position in Treovir LLC, an oHSV clinical stage company and is a member of the POCKiT Diagnostics Board of Scientific Advisors. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

References

  1. Cook, M.; Chauhan, A. Clinical Application of Oncolytic Viruses: A Systematic Review. Int. J. Mol. Sci. 2020, 21, E7505. [Google Scholar] [CrossRef] [PubMed]
  2. Russell, S.J.; Peng, K.-W.; Bell, J.C. Oncolytic Virotherapy. Nat. Biotechnol. 2012, 30, 658–670. [Google Scholar] [CrossRef] [Green Version]
  3. Clemens, M.J. Targets and Mechanisms for the Regulation of Translation in Malignant Transformation. Oncogene 2004, 23, 3180–3188. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Yu, Z.; Chan, M.-K.; O-charoenrat, P.; Eisenberg, D.P.; Shah, J.P.; Singh, B.; Fong, Y.; Wong, R.J. Enhanced Nectin-1 Expression and Herpes Oncolytic Sensitivity in Highly Migratory and Invasive Carcinoma. Clin. Cancer Res. 2005, 11, 4889–4897. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Anderson, B.D.; Nakamura, T.; Russell, S.J.; Peng, K.-W. High CD46 Receptor Density Determines Preferential Killing of Tumor Cells by Oncolytic Measles Virus. Cancer Res. 2004, 64, 4919–4926. [Google Scholar] [CrossRef] [Green Version]
  6. Aghi, M.; Visted, T.; Depinho, R.A.; Chiocca, E.A. Oncolytic Herpes Virus with Defective ICP6 Specifically Replicates in Quiescent Cells with Homozygous Genetic Mutations in P16. Oncogene 2008, 27, 4249–4254. [Google Scholar] [CrossRef] [Green Version]
  7. Martuza, R.L.; Malick, A.; Markert, J.M.; Ruffner, K.L.; Coen, D.M. Experimental Therapy of Human Glioma by Means of a Genetically Engineered Virus Mutant. Science 1991, 252, 854–856. [Google Scholar] [CrossRef]
  8. Hofman, L.; Lawler, S.E.; Lamfers, M.L.M. The Multifaceted Role of Macrophages in Oncolytic Virotherapy. Viruses 2021, 13, 1570. [Google Scholar] [CrossRef]
  9. Naik, S.; Russell, S.J. Engineering Oncolytic Viruses to Exploit Tumor Specific Defects in Innate Immune Signaling Pathways. Expert Opin. Biol. Ther. 2009, 9, 1163–1176. [Google Scholar] [CrossRef]
  10. New, P.Z.; Baskin, D.; Trask, T.; Cavaliere, R.; Chaudhury, A.R.; Bell, S.; Aguilar, L.K.; Aguilar-Cordova, E.; Chiocca, A.; Wong, K. Radiographic and Immunologic Responses to Adjuvant Immunotherapy for Malignant Gliomas. J. Clin. Oncol. 2008, 26, 2039. [Google Scholar] [CrossRef]
  11. Rainov, N.G. A Phase III Clinical Evaluation of Herpes Simplex Virus Type 1 Thymidine Kinase and Ganciclovir Gene Therapy as an Adjuvant to Surgical Resection and Radiation in Adults with Previously Untreated Glioblastoma Multiforme. Hum. Gene Ther. 2000, 11, 2389–2401. [Google Scholar] [CrossRef] [PubMed]
  12. Martinez, F.O.; Sica, A.; Mantovani, A.; Locati, M. Macrophage Activation and Polarization. Front. Biosci. J. Virtual Libr. 2008, 13, 453–461. [Google Scholar] [CrossRef] [Green Version]
  13. Ma, J.; Ramachandran, M.; Jin, C.; Quijano-Rubio, C.; Martikainen, M.; Yu, D.; Essand, M. Characterization of Virus-Mediated Immunogenic Cancer Cell Death and the Consequences for Oncolytic Virus-Based Immunotherapy of Cancer. Cell Death Dis. 2020, 11, 48. [Google Scholar] [CrossRef] [Green Version]
  14. Decraene, B.; Yang, Y.; De Smet, F.; Garg, A.D.; Agostinis, P.; De Vleeschouwer, S. Immunogenic Cell Death and Its Therapeutic or Prognostic Potential in High-Grade Glioma. Genes Immun. 2022, 1–11. [Google Scholar] [CrossRef] [PubMed]
  15. Rius-Rocabert, S.; García-Romero, N.; García, A.; Ayuso-Sacido, A.; Nistal-Villan, E. Oncolytic Virotherapy in Glioma Tumors. Int. J. Mol. Sci. 2020, 21, 7604. [Google Scholar] [CrossRef] [PubMed]
  16. Nguyen, H.-M.; Saha, D. The Current State of Oncolytic Herpes Simplex Virus for Glioblastoma Treatment. Oncolytic Virother. 2021, 10, 1–27. [Google Scholar] [CrossRef]
  17. Parker, J.N.; Gillespie, G.Y.; Love, C.E.; Randall, S.; Whitley, R.J.; Markert, J.M. Engineered Herpes Simplex Virus Expressing IL-12 in the Treatment of Experimental Murine Brain Tumors. Proc. Natl. Acad. Sci. USA 2000, 97, 2208–2213. [Google Scholar] [CrossRef] [Green Version]
  18. Andreansky, S.; He, B.; van Cott, J.; McGhee, J.; Markert, J.M.; Gillespie, G.Y.; Roizman, B.; Whitley, R.J. Treatment of Intracranial Gliomas in Immunocompetent Mice Using Herpes Simplex Viruses That Express Murine Interleukins. Gene Ther. 1998, 5, 121–130. [Google Scholar] [CrossRef] [Green Version]
  19. Glorioso, J.C.; Cohen, J.B.; Goins, W.F.; Hall, B.; Jackson, J.W.; Kohanbash, G.; Amankulor, N.; Kaur, B.; Caligiuri, M.A.; Chiocca, E.A.; et al. Oncolytic HSV Vectors and Anti-Tumor Immunity. Curr. Issues Mol. Biol. 2021, 41, 381–468. [Google Scholar] [CrossRef]
  20. Gromeier, M.; Lachmann, S.; Rosenfeld, M.R.; Gutin, P.H.; Wimmer, E. Intergeneric Poliovirus Recombinants for the Treatment of Malignant Glioma. Proc. Natl. Acad. Sci. USA 2000, 97, 6803–6808. [Google Scholar] [CrossRef] [Green Version]
  21. Desjardins, A.; Gromeier, M.; Herndon, J.E.; Beaubier, N.; Bolognesi, D.P.; Friedman, A.H.; Friedman, H.S.; McSherry, F.; Muscat, A.M.; Nair, S.; et al. Recurrent Glioblastoma Treated with Recombinant Poliovirus. N. Engl. J. Med. 2018, 379, 150–161. [Google Scholar] [CrossRef] [PubMed]
  22. Brown, M.C.; Holl, E.K.; Boczkowski, D.; Dobrikova, E.; Mosaheb, M.; Chandramohan, V.; Bigner, D.D.; Gromeier, M.; Nair, S.K. Cancer Immunotherapy with Recombinant Poliovirus Induces IFN-Dominant Activation of Dendritic Cells and Tumor Antigen-Specific CTLs. Sci. Transl. Med. 2017, 9, eaan4220. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Walton, R.W.; Brown, M.C.; Sacco, M.T.; Gromeier, M. Engineered Oncolytic Poliovirus PVSRIPO Subverts MDA5-Dependent Innate Immune Responses in Cancer Cells. J. Virol. 2018, 92, e00879-18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Liu, P.; Wang, Y.; Wang, Y.; Kong, Z.; Chen, W.; Li, J.; Chen, W.; Tong, Y.; Ma, W.; Wang, Y. Effects of Oncolytic Viruses and Viral Vectors on Immunity in Glioblastoma. Gene Ther. 2020, 1–12. [Google Scholar] [CrossRef] [PubMed]
  25. Ji, N.; Weng, D.; Liu, C.; Gu, Z.; Chen, S.; Guo, Y.; Fan, Z.; Wang, X.; Chen, J.; Zhao, Y.; et al. Adenovirus-Mediated Delivery of Herpes Simplex Virus Thymidine Kinase Administration Improves Outcome of Recurrent High-Grade Glioma. Oncotarget 2015, 7, 4369–4378. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Varda-Bloom, N.; Shaish, A.; Gonen, A.; Levanon, K.; Greenbereger, S.; Ferber, S.; Levkovitz, H.; Castel, D.; Goldberg, I.; Afek, A.; et al. Tissue-Specific Gene Therapy Directed to Tumor Angiogenesis. Gene Ther. 2001, 8, 819–827. [Google Scholar] [CrossRef] [Green Version]
  27. Brenner, A.J.; Peters, K.B.; Vredenburgh, J.; Bokstein, F.; Blumenthal, D.T.; Yust-Katz, S.; Peretz, I.; Oberman, B.; Freedman, L.S.; Ellingson, B.M.; et al. Safety and Efficacy of VB-111, an Anticancer Gene Therapy, in Patients with Recurrent Glioblastoma: Results of a Phase I/II Study. Neuro-Oncology 2020, 22, 694–704. [Google Scholar] [CrossRef]
  28. Lang, F.F.; Conrad, C.; Gomez-Manzano, C.; Yung, W.K.A.; Sawaya, R.; Weinberg, J.S.; Prabhu, S.S.; Rao, G.; Fuller, G.N.; Aldape, K.D.; et al. Phase I Study of DNX-2401 (Delta-24-RGD) Oncolytic Adenovirus: Replication and Immunotherapeutic Effects in Recurrent Malignant Glioma. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2018, 36, 1419–1427. [Google Scholar] [CrossRef]
  29. Denton, N.L.; Chen, C.-Y.; Scott, T.R.; Cripe, T.P. Tumor-Associated Macrophages in Oncolytic Virotherapy: Friend or Foe? Biomedicines 2016, 4, 13. [Google Scholar] [CrossRef] [Green Version]
  30. Razavi, S.-M.; Lee, K.E.; Jin, B.E.; Aujla, P.S.; Gholamin, S.; Li, G. Immune Evasion Strategies of Glioblastoma. Front. Surg. 2016, 3, 11. [Google Scholar] [CrossRef]
  31. Fulci, G.; Dmitrieva, N.; Gianni, D.; Fontana, E.J.; Pan, X.; Lu, Y.; Kaufman, C.S.; Kaur, B.; Lawler, S.E.; Lee, R.J.; et al. Depletion of Peripheral Macrophages and Brain Microglia Increases Brain Tumor Titers of Oncolytic Viruses. Cancer Res. 2007, 67, 9398–9406. [Google Scholar] [CrossRef] [Green Version]
  32. Buonfiglioli, A.; Hambardzumyan, D. Macrophages and Microglia: The Cerberus of Glioblastoma. Acta Neuropathol. Commun. 2021, 9, 54. [Google Scholar] [CrossRef]
  33. Kamińska, B.; Gabrusiewicz, K.; Sielska, M. Characteristics of Phenotype and Pro-Tumorigenic Roles of Glioma Infiltrating Microglia/Macrophages. J. Neurol. Neurophysiol. 2011, s5, 1–6. [Google Scholar] [CrossRef] [Green Version]
  34. Wesolowska, A.; Kwiatkowska, A.; Slomnicki, L.; Dembinski, M.; Master, A.; Sliwa, M.; Franciszkiewicz, K.; Chouaib, S.; Kaminska, B. Microglia-Derived TGF-Beta as an Important Regulator of Glioblastoma Invasion--an Inhibition of TGF-Beta-Dependent Effects by ShRNA against Human TGF-Beta Type II Receptor. Oncogene 2008, 27, 918–930. [Google Scholar] [CrossRef] [Green Version]
  35. Roggendorf, W.; Strupp, S.; Paulus, W. Distribution and Characterization of Microglia/Macrophages in Human Brain Tumors. Acta Neuropathol. 1996, 92, 288–293. [Google Scholar] [CrossRef]
  36. Mills, C.D.; Kincaid, K.; Alt, J.M.; Heilman, M.J.; Hill, A.M. M-1/M-2 Macrophages and the Th1/Th2 Paradigm. J. Immunol. 2000, 164, 6166–6173. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Mantovani, A.; Biswas, S.K.; Galdiero, M.R.; Sica, A.; Locati, M. Macrophage Plasticity and Polarization in Tissue Repair and Remodelling. J. Pathol. 2013, 229, 176–185. [Google Scholar] [CrossRef] [PubMed]
  38. Choi, J.; Mai, N.; Jackson, C.; Belcaid, Z.; Lim, M. It Takes Two: Potential Therapies and Insights Involving Microglia and Macrophages in Glioblastoma. Neuroimmunol. Neuroinflamm. 2018, 5, 42. [Google Scholar] [CrossRef]
  39. Lawrence, T.; Natoli, G. Transcriptional Regulation of Macrophage Polarization: Enabling Diversity with Identity. Nat. Rev. Immunol. 2011, 11, 750–761. [Google Scholar] [CrossRef]
  40. Rider, P.; Carmi, Y.; Guttman, O.; Braiman, A.; Cohen, I.; Voronov, E.; White, M.R.; Dinarello, C.A.; Apte, R.N. IL-1α and IL-1β Recruit Different Myeloid Cells and Promote Different Stages of Sterile Inflammation. J. Immunol. Baltim. Md 1950 2011, 187, 4835–4843. [Google Scholar] [CrossRef] [Green Version]
  41. Hambardzumyan, D.; Gutmann, D.H.; Kettenmann, H. The Role of Microglia and Macrophages in Glioma Maintenance and Progression. Nat. Neurosci. 2016, 19, 20–27. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Cooper, M.A.; Fehniger, T.A.; Turner, S.C.; Chen, K.S.; Ghaheri, B.A.; Ghayur, T.; Carson, W.E.; Caligiuri, M.A. Human Natural Killer Cells: A Unique Innate Immunoregulatory Role for the CD56 (Bright) Subset. Blood 2001, 97, 3146–3151. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Wu, A.; Wei, J.; Kong, L.-Y.; Wang, Y.; Priebe, W.; Qiao, W.; Sawaya, R.; Heimberger, A.B. Glioma Cancer Stem Cells Induce Immunosuppressive Macrophages/Microglia. Neuro-Oncology 2010, 12, 1113–1125. [Google Scholar] [CrossRef]
  44. Honda, K.; Taniguchi, T. IRFs: Master Regulators of Signalling by Toll-like Receptors and Cytosolic Pattern-Recognition Receptors. Nat. Rev. Immunol. 2006, 6, 644–658. [Google Scholar] [CrossRef] [PubMed]
  45. Kang, K.; Reilly, S.M.; Karabacak, V.; Gangl, M.R.; Fitzgerald, K.; Hatano, B.; Lee, C.-H. Adipocyte-Derived Th2 Cytokines and Myeloid PPARdelta Regulate Macrophage Polarization and Insulin Sensitivity. Cell Metab. 2008, 7, 485–495. [Google Scholar] [CrossRef] [Green Version]
  46. Odegaard, J.I.; Ricardo-Gonzalez, R.R.; Red Eagle, A.; Vats, D.; Morel, C.R.; Goforth, M.H.; Subramanian, V.; Mukundan, L.; Ferrante, A.W.; Chawla, A. Alternative M2 Activation of Kupffer Cells by PPARdelta Ameliorates Obesity-Induced Insulin Resistance. Cell Metab. 2008, 7, 496–507. [Google Scholar] [CrossRef] [Green Version]
  47. Marigo, I.; Dolcetti, L.; Serafini, P.; Zanovello, P.; Bronte, V. Tumor-Induced Tolerance and Immune Suppression by Myeloid Derived Suppressor Cells. Immunol. Rev. 2008, 222, 162–179. [Google Scholar] [CrossRef]
  48. Murray, P.J.; Allen, J.E.; Biswas, S.K.; Fisher, E.A.; Gilroy, D.W.; Goerdt, S.; Gordon, S.; Hamilton, J.A.; Ivashkiv, L.B.; Lawrence, T.; et al. Macrophage Activation and Polarization: Nomenclature and Experimental Guidelines. Immunity 2014, 41, 14–20. [Google Scholar] [CrossRef] [Green Version]
  49. Negishi, H.; Ohba, Y.; Yanai, H.; Takaoka, A.; Honma, K.; Yui, K.; Matsuyama, T.; Taniguchi, T.; Honda, K. Negative Regulation of Toll-like-Receptor Signaling by IRF-4. Proc. Natl. Acad. Sci. USA 2005, 102, 15989–15994. [Google Scholar] [CrossRef] [Green Version]
  50. Takaoka, A.; Yanai, H.; Kondo, S.; Duncan, G.; Negishi, H.; Mizutani, T.; Kano, S.-I.; Honda, K.; Ohba, Y.; Mak, T.W.; et al. Integral Role of IRF-5 in the Gene Induction Programme Activated by Toll-like Receptors. Nature 2005, 434, 243–249. [Google Scholar] [CrossRef]
  51. Li, W.; Graeber, M.B. The Molecular Profile of Microglia under the Influence of Glioma. Neuro-Oncol. 2012, 14, 958–978. [Google Scholar] [CrossRef] [Green Version]
  52. Charles, N.A.; Holland, E.C.; Gilbertson, R.; Glass, R.; Kettenmann, H. The Brain Tumor Microenvironment. Glia 2011, 59, 1169–1180. [Google Scholar] [CrossRef]
  53. Brault, M.S.; Kurt, R.A. Impact of Tumor-Derived CCL2 on Macrophage Effector Function. J. Biomed. Biotechnol. 2005, 2005, 37–43. [Google Scholar] [CrossRef] [PubMed]
  54. Conti, I.; Rollins, B.J. CCL2 (Monocyte Chemoattractant Protein-1) and Cancer. Semin. Cancer Biol. 2004, 14, 149–154. [Google Scholar] [CrossRef]
  55. Lamagna, C.; Aurrand-Lions, M.; Imhof, B.A. Dual Role of Macrophages in Tumor Growth and Angiogenesis. J. Leukoc. Biol. 2006, 80, 705–713. [Google Scholar] [CrossRef]
  56. Pixley, F.J.; Stanley, E.R. CSF-1 Regulation of the Wandering Macrophage: Complexity in Action. Trends Cell Biol. 2004, 14, 628–638. [Google Scholar] [CrossRef] [PubMed]
  57. Lin, E.Y.; Nguyen, A.V.; Russell, R.G.; Pollard, J.W. Colony-Stimulating Factor 1 Promotes Progression of Mammary Tumors to Malignancy. J. Exp. Med. 2001, 193, 727–740. [Google Scholar] [CrossRef] [Green Version]
  58. Pollard, J.W. Tumour-Educated Macrophages Promote Tumour Progression and Metastasis. Nat. Rev. Cancer 2004, 4, 71–78. [Google Scholar] [CrossRef]
  59. Wei, J.; Gabrusiewicz, K.; Heimberger, A. The Controversial Role of Microglia in Malignant Gliomas. Clin. Dev. Immunol. 2013, 2013, 285246. [Google Scholar] [CrossRef] [PubMed]
  60. D’Andrea, A.; Aste-Amezaga, M.; Valiante, N.M.; Ma, X.; Kubin, M.; Trinchieri, G. Interleukin 10 (IL-10) Inhibits Human Lymphocyte Interferon Gamma-Production by Suppressing Natural Killer Cell Stimulatory Factor/IL-12 Synthesis in Accessory Cells. J. Exp. Med. 1993, 178, 1041–1048. [Google Scholar] [CrossRef] [PubMed]
  61. Biswas, S.K.; Mantovani, A. Macrophage Plasticity and Interaction with Lymphocyte Subsets: Cancer as a Paradigm. Nat. Immunol. 2010, 11, 889–896. [Google Scholar] [CrossRef]
  62. Nduom, E.K.; Weller, M.; Heimberger, A.B. Immunosuppressive Mechanisms in Glioblastoma. Neuro-Oncol. 2015, 17 (Suppl. 7), vii9–vii14. [Google Scholar] [CrossRef] [PubMed]
  63. Bolyard, C.; Meisen, W.H.; Banasavadi-Siddegowda, Y.; Hardcastle, J.; Yoo, J.Y.; Wohleb, E.S.; Wojton, J.; Yu, J.-G.; Dubin, S.; Khosla, M.; et al. BAI1 Orchestrates Macrophage Inflammatory Response to HSV Infection—Implications for Oncolytic Viral Therapy. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2017, 23, 1809–1819. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Delwar, Z.M.; Kuo, Y.; Wen, Y.H.; Rennie, P.S.; Jia, W. Oncolytic Virotherapy Blockade by Microglia and Macrophages Requires STAT1/3. Cancer Res. 2018, 78, 718–730. [Google Scholar] [CrossRef] [Green Version]
  65. Saha, D.; Martuza, R.L.; Rabkin, S.D. Macrophage Polarization Contributes to Glioblastoma Eradication by Combination Immunovirotherapy and Immune Checkpoint Blockade. Cancer Cell 2017, 32, 253–267.e5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Wakimoto, H.; Johnson, P.R.; Knipe, D.M.; Chiocca, E.A. Effects of Innate Immunity on Herpes Simplex Virus and Its Ability to Kill Tumor Cells. Gene Ther. 2003, 10, 983–990. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. van den Bossche, W.B.L.; Kleijn, A.; Teunissen, C.E.; Voerman, J.S.A.; Teodosio, C.; Noske, D.P.; van Dongen, J.J.M.; Dirven, C.M.F.; Lamfers, M.L.M. Oncolytic Virotherapy in Glioblastoma Patients Induces a Tumor Macrophage Phenotypic Shift Leading to an Altered Glioblastoma Microenvironment. Neuro-Oncology 2018, 20, 1494–1504. [Google Scholar] [CrossRef]
  68. Kleijn, A.; Kloezeman, J.; Treffers-Westerlaken, E.; Fulci, G.; Leenstra, S.; Dirven, C.; Debets, R.; Lamfers, M. The in Vivo Therapeutic Efficacy of the Oncolytic Adenovirus Delta24-RGD Is Mediated by Tumor-Specific Immunity. PLoS ONE 2014, 9, e97495. [Google Scholar] [CrossRef] [Green Version]
  69. Kiyokawa, J.; Kawamura, Y.; Ghouse, S.M.; Acar, S.; Barçın, E.; Martínez-Quintanilla, J.; Martuza, R.L.; Alemany, R.; Rabkin, S.D.; Shah, K.; et al. Modification of Extracellular Matrix Enhances Oncolytic Adenovirus Immunotherapy in Glioblastoma. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2021, 27, 889–902. [Google Scholar] [CrossRef] [PubMed]
  70. Angelova, A.L.; Barf, M.; Geletneky, K.; Unterberg, A.; Rommelaere, J. Immunotherapeutic Potential of Oncolytic H-1 Parvovirus: Hints of Glioblastoma Microenvironment Conversion towards Immunogenicity. Viruses 2017, 9, 382. [Google Scholar] [CrossRef] [Green Version]
  71. Grekova, S.P.; Raykov, Z.; Zawatzky, R.; Rommelaere, J.; Koch, U. Activation of a Glioma-Specific Immune Response by Oncolytic Parvovirus Minute Virus of Mice Infection. Cancer Gene Ther. 2012, 19, 468–475. [Google Scholar] [CrossRef] [PubMed]
  72. Geletneky, K.; Hajda, J.; Angelova, A.L.; Leuchs, B.; Capper, D.; Bartsch, A.J.; Neumann, J.-O.; Schöning, T.; Hüsing, J.; Beelte, B.; et al. Oncolytic H-1 Parvovirus Shows Safety and Signs of Immunogenic Activity in a First Phase I/IIa Glioblastoma Trial. Mol. Ther. J. Am. Soc. Gene Ther. 2017, 25, 2620–2634. [Google Scholar] [CrossRef] [Green Version]
  73. Kober, C.; Rohn, S.; Weibel, S.; Geissinger, U.; Chen, N.G.; Szalay, A.A. Microglia and Astrocytes Attenuate the Replication of the Oncolytic Vaccinia Virus LIVP 1.1.1 in Murine GL261 Gliomas by Acting as Vaccinia Virus Traps. J. Transl. Med. 2015, 13, 216. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Nguyen, H.-M.; Guz-Montgomery, K.; Saha, D. Oncolytic Virus Encoding a Master Pro-Inflammatory Cytokine Interleukin 12 in Cancer Immunotherapy. Cells 2020, 9, 400. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Nguyen, K.G.; Vrabel, M.R.; Mantooth, S.M.; Hopkins, J.J.; Wagner, E.S.; Gabaldon, T.A.; Zaharoff, D.A. Localized Interleukin-12 for Cancer Immunotherapy. Front. Immunol. 2020, 11, 575597. [Google Scholar] [CrossRef]
  76. Cheema, T.A.; Wakimoto, H.; Fecci, P.E.; Ning, J.; Kuroda, T.; Jeyaretna, D.S.; Martuza, R.L.; Rabkin, S.D. Multifaceted Oncolytic Virus Therapy for Glioblastoma in an Immunocompetent Cancer Stem Cell Model. Proc. Natl. Acad. Sci. USA 2013, 110, 12006–12011. [Google Scholar] [CrossRef] [Green Version]
  77. Hellums, E.K.; Markert, J.M.; Parker, J.N.; He, B.; Perbal, B.; Roizman, B.; Whitley, R.J.; Langford, C.P.; Bharara, S.; Gillespie, G.Y. Increased Efficacy of an Interleukin-12-Secreting Herpes Simplex Virus in a Syngeneic Intracranial Murine Glioma Model. Neuro-Oncol. 2005, 7, 213–224. [Google Scholar] [CrossRef]
  78. Ino, Y.; Saeki, Y.; Fukuhara, H.; Todo, T. Triple Combination of Oncolytic Herpes Simplex Virus-1 Vectors Armed with Interleukin-12, Interleukin-18, or Soluble B7-1 Results in Enhanced Antitumor Efficacy. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2006, 12, 643–652. [Google Scholar] [CrossRef] [Green Version]
  79. Markert, J.M.; Cody, J.J.; Parker, J.N.; Coleman, J.M.; Price, K.H.; Kern, E.R.; Quenelle, D.C.; Lakeman, A.D.; Schoeb, T.R.; Palmer, C.A.; et al. Preclinical Evaluation of a Genetically Engineered Herpes Simplex Virus Expressing Interleukin-12. J. Virol. 2012, 86, 5304–5313. [Google Scholar] [CrossRef] [Green Version]
  80. Lynch, D.H.; Andreasen, A.; Maraskovsky, E.; Whitmore, J.; Miller, R.E.; Schuh, J.C. Flt3 Ligand Induces Tumor Regression and Antitumor Immune Responses in Vivo. Nat. Med. 1997, 3, 625–631. [Google Scholar] [CrossRef]
  81. Chakravarty, P.K.; Alfieri, A.; Thomas, E.K.; Beri, V.; Tanaka, K.E.; Vikram, B.; Guha, C. Flt3-Ligand Administration after Radiation Therapy Prolongs Survival in a Murine Model of Metastatic Lung Cancer. Cancer Res. 1999, 59, 6028–6032. [Google Scholar] [PubMed]
  82. Barnard, Z.; Wakimoto, H.; Zaupa, C.; Jeyeretna, D.S.; Patel, A.P.; Kle, J.; Martuza, R.L.; Rabkin, S.D.; Curry, W.T. Expression of FMS-like Tyrosine Kinase 3 Ligand by Oncolytic Herpes Simplex Virus Type I Prolongs Survival in Mice Bearing Established Syngeneic Intracranial Malignant Glioma. Neurosurgery 2012, 71, 741–748. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Topalian, S.L.; Taube, J.M.; Anders, R.A.; Pardoll, D.M. Mechanism-Driven Biomarkers to Guide Immune Checkpoint Blockade in Cancer Therapy. Nat. Rev. Cancer 2016, 16, 275–287. [Google Scholar] [CrossRef]
  84. Filley, A.C.; Henriquez, M.; Dey, M. Recurrent Glioma Clinical Trial, CheckMate-143: The Game Is Not over Yet. Oncotarget 2017, 8, 91779–91794. [Google Scholar] [CrossRef] [Green Version]
  85. Reardon, D.A.; Freeman, G.; Wu, C.; Chiocca, E.A.; Wucherpfennig, K.W.; Wen, P.Y.; Fritsch, E.F.; Curry, W.T.; Sampson, J.H.; Dranoff, G. Immunotherapy Advances for Glioblastoma. Neuro Oncol. 2014, 16, 1441–1458. [Google Scholar] [CrossRef]
  86. Khasraw, M.; Reardon, D.A.; Weller, M.; Sampson, J.H. PD-1 Inhibitors: Do They Have a Future in the Treatment of Glioblastoma? Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2020, 26, 5287–5296. [Google Scholar] [CrossRef]
  87. Oh, T.; Fakurnejad, S.; Sayegh, E.T.; Clark, A.J.; Ivan, M.E.; Sun, M.Z.; Safaee, M.; Bloch, O.; James, C.D.; Parsa, A.T. Immunocompetent Murine Models for the Study of Glioblastoma Immunotherapy. J. Transl. Med. 2014, 12, 107. [Google Scholar] [CrossRef] [Green Version]
  88. Jiang, H.; Rivera-Molina, Y.; Gomez-Manzano, C.; Clise-Dwyer, K.; Bover, L.; Vence, L.M.; Yuan, Y.; Lang, F.F.; Toniatti, C.; Hossain, M.B.; et al. Oncolytic Adenovirus and Tumor-Targeting Immune Modulatory Therapy Improve Autologous Cancer Vaccination. Cancer Res. 2017, 77, 3894–3907. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Rivera-Molina, Y.; Jiang, H.; Fueyo, J.; Nguyen, T.; Shin, D.H.; Youssef, G.; Fan, X.; Gumin, J.; Alonso, M.M.; Phadnis, S.; et al. GITRL-Armed Delta-24-RGD Oncolytic Adenovirus Prolongs Survival and Induces Anti-Glioma Immune Memory. Neuro-Oncol. Adv. 2019, 1, vdz009. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Lamfers, M.L.M.; Fulci, G.; Gianni, D.; Tang, Y.; Kurozumi, K.; Kaur, B.; Moeniralm, S.; Saeki, Y.; Carette, J.E.; Weissleder, R.; et al. Cyclophosphamide Increases Transgene Expression Mediated by an Oncolytic Adenovirus in Glioma-Bearing Mice Monitored by Bioluminescence Imaging. Mol. Ther. J. Am. Soc. Gene Ther. 2006, 14, 779–788. [Google Scholar] [CrossRef]
  91. Fulci, G.; Breymann, L.; Gianni, D.; Kurozomi, K.; Rhee, S.S.; Yu, J.; Kaur, B.; Louis, D.N.; Weissleder, R.; Caligiuri, M.A.; et al. Cyclophosphamide Enhances Glioma Virotherapy by Inhibiting Innate Immune Responses. Proc. Natl. Acad. Sci. 2006, 103, 12873–12878. [Google Scholar] [CrossRef] [Green Version]
  92. Meisen, W.H.; Wohleb, E.S.; Jaime-Ramirez, A.C.; Bolyard, C.; Yoo, J.Y.; Russell, L.; Hardcastle, J.; Dubin, S.; Muili, K.; Yu, J.; et al. The Impact of Macrophage- and Microglia-Secreted TNFα on Oncolytic HSV-1 Therapy in the Glioblastoma Tumor Microenvironment. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2015, 21, 3274–3285. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Ikeda, K.; Ichikawa, T.; Wakimoto, H.; Silver, J.S.; Deisboeck, T.S.; Finkelstein, D.; Harsh, G.R.; Louis, D.N.; Bartus, R.T.; Hochberg, F.H.; et al. Oncolytic Virus Therapy of Multiple Tumors in the Brain Requires Suppression of Innate and Elicited Antiviral Responses. Nat. Med. 1999, 5, 881–887. [Google Scholar] [CrossRef]
  94. Kurozumi, K.; Hardcastle, J.; Thakur, R.; Yang, M.; Christoforidis, G.; Fulci, G.; Hochberg, F.H.; Weissleder, R.; Carson, W.; Chiocca, E.A.; et al. Effect of Tumor Microenvironment Modulation on the Efficacy of Oncolytic Virus Therapy. J. Natl. Cancer Inst. 2007, 99, 1768–1781. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Wakimoto, H.; Fulci, G.; Tyminski, E.; Chiocca, E.A. Altered Expression of Antiviral Cytokine MRNAs Associated with Cyclophosphamide’s Enhancement of Viral Oncolysis. Gene Ther. 2004, 11, 214–223. [Google Scholar] [CrossRef] [Green Version]
  96. Zemp, F.J.; McKenzie, B.A.; Lun, X.; Reilly, K.M.; McFadden, G.; Yong, V.W.; Forsyth, P.A. Cellular Factors Promoting Resistance to Effective Treatment of Glioma with Oncolytic Myxoma Virus. Cancer Res. 2014, 74, 7260–7273. [Google Scholar] [CrossRef] [Green Version]
  97. Kambara, H.; Saeki, Y.; Chiocca, E.A. Cyclophosphamide Allows for In Vivo Dose Reduction of a Potent Oncolytic Virus. Cancer Res. 2005, 65, 11255–11258. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Lun, X.; Alain, T.; Zemp, F.J.; Zhou, H.; Rahman, M.M.; Hamilton, M.G.; McFadden, G.; Bell, J.; Senger, D.L.; Forsyth, P.A. Myxoma Virus Virotherapy for Glioma in Immunocompetent Animal Models: Optimizing Administration Routes and Synergy with Rapamycin. Cancer Res. 2010, 70, 598–608. [Google Scholar] [CrossRef] [Green Version]
  99. Lee, T.J.; Nair, M.; Banasavadi-Siddegowda, Y.; Liu, J.; Nallanagulagari, T.; Jaime-Ramirez, A.C.; Guo, J.Y.; Quadri, H.; Zhang, J.; Bockhorst, K.H.; et al. Enhancing Therapeutic Efficacy of Oncolytic Herpes Simplex Virus-1 with Integrin Β1 Blocking Antibody OS2966. Mol. Cancer Ther. 2019, 18, 1127–1136. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  100. Alvarez-Breckenridge, C.A.; Yu, J.; Price, R.; Wei, M.; Wang, Y.; Nowicki, M.O.; Ha, Y.P.; Bergin, S.; Hwang, C.; Fernandez, S.A.; et al. The Histone Deacetylase Inhibitor Valproic Acid Lessens NK Cell Action against Oncolytic Virus-Infected Glioblastoma Cells by Inhibition of STAT5/T-BET Signaling and Generation of Gamma Interferon. J. Virol. 2012, 86, 4566–4577. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  101. Thorne, A.H.; Meisen, W.H.; Russell, L.; Yoo, J.Y.; Bolyard, C.M.; Lathia, J.D.; Rich, J.; Puduvalli, V.K.; Mao, H.; Yu, J.; et al. Role of Cysteine-Rich 61 Protein (CCN1) in Macrophage-Mediated Oncolytic Herpes Simplex Virus Clearance. Mol. Ther. 2014, 22, 1678–1687. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Haseley, A.; Boone, S.; Wojton, J.; Yu, L.; Yoo, J.Y.; Yu, J.; Kurozumi, K.; Glorioso, J.C.; Caligiuri, M.A.; Kaur, B. Extracellular Matrix Protein CCN1 Limits Oncolytic Efficacy in Glioma. Cancer Res. 2012, 72, 1353–1362. [Google Scholar] [CrossRef] [Green Version]
  103. Jacobsen, K.; Russell, L.; Kaur, B.; Friedman, A. Effects of CCN1 and Macrophage Content on Glioma Virotherapy: A Mathematical Model. Bull. Math. Biol. 2015, 77, 984–1012. [Google Scholar] [CrossRef] [PubMed]
  104. Robson, C.N.; Gnanapragasam, V.; Byrne, R.L.; Collins, A.T.; Neal, D.E. Transforming Growth Factor-Beta1 up-Regulates P15, P21 and P27 and Blocks Cell Cycling in G1 in Human Prostate Epithelium. J. Endocrinol. 1999, 160, 257–266. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Han, J.; Chen, X.; Chu, J.; Xu, B.; Meisen, W.H.; Chen, L.; Zhang, L.; Zhang, J.; He, X.; Wang, Q.-E.; et al. TGFβ Treatment Enhances Glioblastoma Virotherapy by Inhibiting the Innate Immune Response. Cancer Res. 2015, 75, 5273–5282. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Buiting, A.M.; Zhou, F.; Bakker, J.A.; van Rooijen, N.; Huang, L. Biodistribution of Clodronate and Liposomes Used in the Liposome Mediated Macrophage “suicide” Approach. J. Immunol. Methods 1996, 192, 55–62. [Google Scholar] [CrossRef]
  107. Wang, J.; Shen, F.; Yao, Y.; Wang, L.-L.; Zhu, Y.; Hu, J. Adoptive Cell Therapy: A Novel and Potential Immunotherapy for Glioblastoma. Front. Oncol. 2020, 10, 59. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Dogrammatzis, C.; Waisner, H.; Kalamvoki, M. Cloaked Viruses and Viral Factors in Cutting Edge Exosome-Based Therapies. Front. Cell Dev. Biol. 2020, 8, 376. [Google Scholar] [CrossRef]
  109. Park, D.; Tosello-Trampont, A.-C.; Elliott, M.R.; Lu, M.; Haney, L.B.; Ma, Z.; Klibanov, A.L.; Mandell, J.W.; Ravichandran, K.S. BAI1 Is an Engulfment Receptor for Apoptotic Cells Upstream of the ELMO/Dock180/Rac Module. Nature 2007, 450, 430–434. [Google Scholar] [CrossRef] [Green Version]
  110. Zhang, L.; Alizadeh, D.; Van Handel, M.; Kortylewski, M.; Yu, H.; Badie, B. Stat3 Inhibition Activates Tumor Macrophages and Abrogates Glioma Growth in Mice. Glia 2009, 57, 1458–1467. [Google Scholar] [CrossRef]
  111. Alvarez-Breckenridge, C.A.; Yu, J.; Price, R.; Wojton, J.; Pradarelli, J.; Mao, H.; Wei, M.; Wang, Y.; He, S.; Hardcastle, J.; et al. NK Cells Impede Glioblastoma Virotherapy through NKp30 and NKp46 Natural Cytotoxicity Receptors. Nat. Med. 2012, 18, 1827–1834. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Lamfers, M.; Idema, S.; van Milligen, F.; Schouten, T.; van der Valk, P.; Vandertop, P.; Dirven, C.; Noske, D. Homing Properties of Adipose-Derived Stem Cells to Intracerebral Glioma and the Effects of Adenovirus Infection. Cancer Lett. 2009, 274, 78–87. [Google Scholar] [CrossRef]
  113. Herrlinger, U.; Woiciechowski, C.; Sena-Esteves, M.; Aboody, K.S.; Jacobs, A.H.; Rainov, N.G.; Snyder, E.Y.; Breakefield, X.O. Neural Precursor Cells for Delivery of Replication-Conditional HSV-1 Vectors to Intracerebral Gliomas. Mol. Ther. J. Am. Soc. Gene Ther. 2000, 1, 347–357. [Google Scholar] [CrossRef]
  114. Dey, M.; Yu, D.; Kanojia, D.; Li, G.; Sukhanova, M.; Spencer, D.A.; Pituch, K.C.; Zhang, L.; Han, Y.; Ahmed, A.U.; et al. Intranasal Oncolytic Virotherapy with CXCR4-Enhanced Stem Cells Extends Survival in Mouse Model of Glioma. Stem Cell Rep. 2016, 7, 471–482. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Morshed, R.A.; Gutova, M.; Juliano, J.; Barish, M.E.; Hawkins-Daarud, A.; Oganesyan, D.; Vazgen, K.; Yang, T.; Annala, A.; Ahmed, A.U.; et al. Analysis of Glioblastoma Tumor Coverage by Oncolytic Virus-Loaded Neural Stem Cells Using MRI-Based Tracking and Histological Reconstruction. Cancer Gene Ther. 2015, 22, 55–61. [Google Scholar] [CrossRef] [Green Version]
  116. Yong, R.L.; Shinojima, N.; Fueyo, J.; Gumin, J.; Vecil, G.G.; Marini, F.C.; Bogler, O.; Andreeff, M.; Lang, F.F. Human Bone Marrow-Derived Mesenchymal Stem Cells for Intravascular Delivery of Oncolytic Adenovirus Delta24-RGD to Human Gliomas. Cancer Res. 2009, 69, 8932–8940. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Iannacone, M.; Moseman, E.A.; Tonti, E.; Bosurgi, L.; Junt, T.; Henrickson, S.E.; Whelan, S.P.; Guidotti, L.G.; von Andrian, U.H. Subcapsular Sinus Macrophages Prevent CNS Invasion on Peripheral Infection with a Neurotropic Virus. Nature 2010, 465, 1079–1083. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Schematic representation of the interaction between M1/M2 tumor-associated macrophage/microglia (TAM) polarization with oncolytic viral distribution and tumor growth in glioblastoma multiforme. Elements of the microenvironment influence polarization between TAM subsets of immuno-suppressive M2 and immune-stimulatory M1. Tumors polarize TAMs towards the M2 phenotype, which support tumor growth. Oncolytic virotherapy (OV) inhibits tumor growth through two main mechanisms: direct oncolysis and the antitumor response. The latter is a result of virus-induced polarization towards the M1 phenotype, which creates an immune response against tumor growth. However, this also stimulates an antiviral response, limiting the beneficial effects of OVs. IL interleukin, M-CSF macrophage colony stimulating factor, TGF-β transforming growth factor beta, PGE2 prostaglandin E2, IFN-γ interferon gamma, LPS lipopolysaccharide, Th T helper, Treg T regulatory, VEGF vascular endothelial growth factor, CD cluster of differentiation, PD-L1 programmed cell death ligand 1, and TNF-α tumor necrosis factor alpha.
Figure 1. Schematic representation of the interaction between M1/M2 tumor-associated macrophage/microglia (TAM) polarization with oncolytic viral distribution and tumor growth in glioblastoma multiforme. Elements of the microenvironment influence polarization between TAM subsets of immuno-suppressive M2 and immune-stimulatory M1. Tumors polarize TAMs towards the M2 phenotype, which support tumor growth. Oncolytic virotherapy (OV) inhibits tumor growth through two main mechanisms: direct oncolysis and the antitumor response. The latter is a result of virus-induced polarization towards the M1 phenotype, which creates an immune response against tumor growth. However, this also stimulates an antiviral response, limiting the beneficial effects of OVs. IL interleukin, M-CSF macrophage colony stimulating factor, TGF-β transforming growth factor beta, PGE2 prostaglandin E2, IFN-γ interferon gamma, LPS lipopolysaccharide, Th T helper, Treg T regulatory, VEGF vascular endothelial growth factor, CD cluster of differentiation, PD-L1 programmed cell death ligand 1, and TNF-α tumor necrosis factor alpha.
Ijms 23 01808 g001
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Blitz, S.E.; Kappel, A.D.; Gessler, F.A.; Klinger, N.V.; Arnaout, O.; Lu, Y.; Peruzzi, P.P.; Smith, T.R.; Chiocca, E.A.; Friedman, G.K.; et al. Tumor-Associated Macrophages/Microglia in Glioblastoma Oncolytic Virotherapy: A Double-Edged Sword. Int. J. Mol. Sci. 2022, 23, 1808. https://doi.org/10.3390/ijms23031808

AMA Style

Blitz SE, Kappel AD, Gessler FA, Klinger NV, Arnaout O, Lu Y, Peruzzi PP, Smith TR, Chiocca EA, Friedman GK, et al. Tumor-Associated Macrophages/Microglia in Glioblastoma Oncolytic Virotherapy: A Double-Edged Sword. International Journal of Molecular Sciences. 2022; 23(3):1808. https://doi.org/10.3390/ijms23031808

Chicago/Turabian Style

Blitz, Sarah E., Ari D. Kappel, Florian A. Gessler, Neil V. Klinger, Omar Arnaout, Yi Lu, Pier Paolo Peruzzi, Timothy R. Smith, Ennio A. Chiocca, Gregory K. Friedman, and et al. 2022. "Tumor-Associated Macrophages/Microglia in Glioblastoma Oncolytic Virotherapy: A Double-Edged Sword" International Journal of Molecular Sciences 23, no. 3: 1808. https://doi.org/10.3390/ijms23031808

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop