You're using an outdated browser. Please upgrade to a modern browser for the best experience.
Carrier Cells for Oncolytic Virotherapy: Comparison
Please note this is a comparison between Version 4 by Jessie Wu and Version 3 by Jessie Wu.

Oncolytic viruses (OVs) are an emerging class of therapeutics which combine multiple mechanisms of action, including direct cancer cell-killing, immunotherapy and gene therapy. To target metastatic diseases and tumors that cannot be directly accessed, it is of great interest to develop effective approaches for the systemic delivery of OVs, such as the use of carrier cells, which can be loaded with virus ex vivo and administered intravenously. In general, the ideal carrier cell should have a tropism towards the tumor microenvironment (TME), and it must be susceptible to OV infection but remain viable long enough to allow migration and finally release of the OV within the tumor bed.

  • oncolytic virus
  • carrier cells

1. Background

 A growing number of clinical trials have indicated that OVs have an excellent safety profile and provide some degree of efficacy, but to date only a single OV drug, HSV-1 talimogene laherparepvec (T-Vec), has achieved marketing approval in the US and Europe. An important issue to consider in order to accelerate the clinical advancement of OV agents is the development of an effective delivery system. Currently, the most commonly employed OV delivery route is intratumoral; however, to target metastatic diseases and tumors that cannot be directly accessed, it is of great interest to develop effective approaches for the systemic delivery of OVs, such as the use of carrier cells

2. Carrier Cells for Oncolytic Virus Delivery

While the use of carrier cells is an attractive approach to potentially improve the pharmacokinetics and biodistribution of OVs, the ideal cell type to employ for this function is a matter for debate. The optimal carrier cells for the delivery of oncolytic viruses should possess three essential features: (1) they must have a tropism towards tumors; (2) they must be able to either internalize the virus or allow virions to stably attach to their cell membrane; (3) they must maintain viability for a sufficient time to allow distribution in the bloodstream and delivery of the viral cargo to the tumor site(s). Furthermore, although somewhat an issue of debate, there is a strong rationale for the use of autologous cells in order to avoid any issues of rejection. Beyond this, a lot of uncertainty arises, and there is no consensus in the field. What is the ideal kind of carrier cell to achieve the efficient systemic delivery of OVs? Traditionally, it has been assumed that an antiviral immune response should be considered as a negative factor because it induces viral clearance before all cancer cells within the mass have been killed. This is also a factor in the choice of mesenchymal stem cells (MSCs), which have immunosuppressive properties, in many studies on carrier cells and OVs [1] (see paragraph below). Although the inhibition of antiviral immune responses could be beneficial for enhancing the therapeutic effect of the OV, a general immune suppression could dampen the virus-mediated immune-stimulatory effects against the cancer, which would be counterproductive and severely hamper an important mechanism of the therapy. Therefore, the use of immune-suppressive MSCs as part of an oncolytic virus regimen should be carefully considered and tested. It is likely desirable to achieve a fine tuning that allows the virus to replicate in the tumor without dampening the downstream adaptive antitumoral immune response [2].
As already mentioned, most preclinical studies, and the few clinical trials which employed carrier cells as a means of OV delivery so far (Table 1), have employed MSCs [1]. Nevertheless, other cells, including neural stem cells (NSCs) [3], monocytes [4] and T lymphocytes [5], have also been investigated as viral carriers and show potential in this function. These non-MSC cell types and their potential as carrier cells for OV delivery will be the focus of the following sections.
Table 1. Clinical trials involving mesenchymal stem cells (MSCs) as carriers for oncolytic viruses (as of December 2021). Abbreviations: AdV Adenovirus. CELYVIR: bone marrow-derived autologous MSCs infected with ICOVIR5 (oncolytic AdV). AloCELYVIR: bone marrow-derived allogeneic MSCs infected with ICOVIR5 (oncolytic AdV).
Trial Target Disease Oncolytic Virus Results
EudraCT Number: 2008-000364-16 Pediatric solid tumors ICOVIR5 (AdV) CELYVIR Trial ended prematurely
NCT 02068794 Ovarian cancer Measles virus Recruiting
NCT 01844661 Miscellaneous metastatic tumors ICOVIR5 CELYVIR Completed in 2016—results not available
EudraCT Number: 2019-001154-26 Extracranial solid tumors ICOVIR5 AloCELYVIR Ongoing
NCT03896568 Recurrent high-grade glioma AdV, DNX-2401 Recruiting
NCT05047276 (phase I/II) Metastatic Uveal Melanoma ICOVIR5 AloCELYVIR Trial not yet recruiting

2.1. T Lymphocytes

T lymphocytes represent attractive candidates as carrier cells for OV delivery due to their ability to circulate freely in the bloodstream and home to their tumor targets, as well as their potential to provide synergistic therapeutic effects via their cytotoxic effector functions. Even naïve activated T cells have been shown to successfully shield OVs from neutralization and nonspecific uptake while delivering them to the tumor bed. In the case of oncolytic measles virus (MV) therapy, it was shown that a virus loaded onto activated T cells could be transferred to tumor cells, even in the presence of neutralizing anti-MV serum in vitro and in vivo, indicating successful shielding conferred by the approach [5]. Interestingly, the expression of the fusogenic envelope proteins of MV on the surface of infected T cells allowed for the heterofusion of the lymphocytes with the tumor cells, which facilitated the transfer of infectious virions and subsequent lysis of the tumor cells. Similarly, oncolytic reovirus was shown to be protected from pre-existing antiviral immunity when loaded onto T cells, which effectively delivered the virus to B16 melanoma tumors in vivo and mediated an anti-tumor immune response and long-term protection against the tumor [6].
Although Newcastle disease virus (NDV) may be unable to replicate within T lymphocytes, it can bind to the T cell surface via its hemagglutinin-neuraminidase attachment protein, which recognizes cell surface sialic acid-containing receptors that are present on glycoproteins or glycolipids, followed by membrane fusion via the viral F protein. In this way, it was speculated that NDV can “hitchhike” on intravenously applied T cells for delivery and transfer to tumor cells. Proof-of-concept was demonstrated in vitro, whereby oncolytic NDV was attached to the surface of activated peripheral blood-derived T cells and shown to be transferred to human MCF-7 breast carcinoma cells, leading to subsequent oncolysis [7].
In a further demonstration of the utility of antigen-nonspecific T cells as OV carriers, Qiao and colleagues investigated the use of naïve T cells to chaperone oncolytic vesicular stomatitis virus (VSV) to lymphoid organs and, thereby, eradicate metastases [8]. In this interesting approach, it was shown that VSV loaded onto naïve T cells could effectively eliminate primary B16 melanoma lesions, as well as lymph node and spleen metastases, even in virus-immune mice [8]. The group later went on to show that the efficacy of the adoptive transfer of antigen-specific OT-I T cells could be enhanced by loading them with oncolytic VSV in the same B16 tumor model [9]. Here, it was demonstrated that tumor-specific T-cell activation and tumor trafficking was even enhanced by the pre-infection of the T cells with VSV prior to delivery, leading to an effective combinatorial approach.
Antigen-specific T lymphocytes have been utilized for the shielding of a variety of oncolytic virus vectors, including VSV, herpes simplex virus type 1 (HSV-1), adenovirus and vaccinia virus (VV) [10][11][12][13]. Tumor-infiltrating lymphocytes (TILs) are a particularly interesting subset, as they have a natural propensity to infiltrate the tumor bed and do not require additional transduction with a T cell receptor or CAR for tumor targeting. Furthermore, the use of autologous TILs provides a personalized approach, which can potentially target a variety of specific tumor-associated antigens expressed within the patient’s own tumor. Santos and colleagues have recently reported on the systemic delivery of an armed adenovirus vector (TILT-123) using TILs isolated from human ovarian cancer and hamster pancreatic cancer as carriers [13]. They hypothesized that loading an optimized OV vector onto TILs would allow for the delivery of the viral therapeutic to the tumor site, and that the strategy would provide a means for the administration of both therapeutic components in one self-amplifying product, which was then demonstrated in various in vivo models [13].
Yotnda et al. utilized a novel approach, in which tumor-specific cytotoxic T lymphocytes (CTLs) were modified to express the adenoviral E1 gene under the control of the activation-dependent CD40 ligand in order to induce infectious adenovirus production specifically when the CTLs were exposed to HLA-matched tumor antigen-expressing target tumor cells [14]. This not only represents an effective combination therapy and OV delivery approach, but it also provides a clever mechanism for restricted viral dissemination specifically at the target tumor site. The benefit of antigen-specific lymphocytes over naïve T cells was further demonstrated using the oncolytic strain F HSV1 mutant, R3616, which carries a deletion of both copies of the virulence factor γ34.5, which is essential for viral replication in neurons [15], loaded onto lymphocytes harvested from mice that had developed an antitumor immunity. R3616 adsorbed onto antigen-specific lymphocytes mediated in significantly improved responses in mice bearing peritoneally-disseminated tumors, compared to R3616 adsorbed onto naïve lymphocytes or with either monotherapy [16].
In addition to the isolation of autologous antigen-specific T lymphocytes, T cells can also be engineered ex vivo to express a T cell receptor (TCR) or chimeric antigen receptor (CAR) in order to confer tumor-specific cytotoxic effector functions. The recent marketing approval of CAR T cells makes this a particularly attractive approach for OV delivery. In fact, it was recently demonstrated that loading HER2-specific CAR T cells with low doses of VSV or oncolytic vaccinia virus (vvDD) does not interfere with receptor expression or function, and either the RNA or DNA virus could be transferred to target tumor cells using either mouse or human T cells [12]. As an alternative to the CAR T cell approach, we have recently reported on the use of TCR-transduced CD8+ central memory T cells as a delivery vehicle for oncolytic VSV, resulting in an effective combination therapy in a xenograft model of acute myeloid leukemia (AML) in mice. Interestingly, in addition to the viral delivery and potent cytotoxic effector functions provided by the central memory subset of TCR T cells, it was shown that the approach substantially improved the safety of intravenously applied VSV compared to the use of a naked virus [11].
Despite the growing body of evidence in support of the use of lymphocytes as OV carriers, one important factor to consider is that various immunosuppressive mechanisms within the tumor microenvironment can mean exclusion of T lymphocytes from the tumor [17]. By design, the function of OV carrier cells is dependent on the ability of those cells to extravasate from the tumor vasculature and infiltrate into the tumor mass. Therefore, the utility of T cells as OV carriers may be limited to those tumors which are not characterized as immune deserts. Additionally, various approaches that are under development to improve lymphocyte trafficking into tumors [18][19] could be applied in OV-loaded T cell regimes in order to pre-sensitize the tumor to the adoptive T cell transfer and concomitantly improve OV delivery. Furthermore, the potential cytotoxic effects of the OV on the lymphocyte would need to be characterized for each viral vector to be utilized in order to avoid the lymphocytes being lysed and releasing their viral cargo before reaching the tumor.
In summary, T lymphocytes, whether naïve, TILs or engineered CAR or TCR T cells, represent promising cell carriers of oncolytic viruses that can shield against antiviral neutralizing antibodies and deliver to the tumor bed, potentially also providing cytotoxic effector functions to synergize with the viral oncolysis. Additional evidence that virus replication within the tumor further enhances T cell-mediated therapeutic effects provides an added rationale for the combination approach. We are likely to see increasing numbers of examples of these approaches in the next few years, as adoptive T cell therapies and oncolytic viruses make their way into routine clinical practice and as new approaches to enhance lymphocyte extravasation and infiltration into tumors are developed.

2.2. Myeloid Cells

An assortment of myeloid cells infiltrate the TME of most solid tumors [20]. These include tumor-associated macrophages (TAMs) [21], tumor-associated neutrophils (TANs) [22], dendritic cells and immature myeloid-derived suppressor cells (MDSCs) [23]. Myeloid cells are actually a functional and important part of the TME, where they are actively recruited by chemokines and cytokines and where they play a role in secreting tumorigenic growth factors and dampening the adaptive immune response.
This explains why infiltration by myeloid cells is a conserved feature in many solid tumors, even those with scarce or no lymphocytic infiltrate [17]. For example, the presence of TAMs is so fundamental that new therapeutic strategies have been designed to target these cells to increase the sensitivity of tumors to both immunotherapy and chemotherapy [22]. As a consequence, TAMs or their circulating precursors (monocytes) are attractive carrier cells for OVs because it is unlikely that cancer cells can develop a resistance to treatment by excluding TAMs from the TME. Furthermore, autologous monocytes can be easily recovered in large numbers from peripheral blood and are amenable to differentiation in vitro into macrophages or dendritic cells. Therefore, it seems that some myeloid cells (with the possible exception of neutrophils, which are difficult to maintain ex vivo for OV infection) have almost ideal characteristics as carrier cells. Nevertheless, there remain few reports of monocytes or other myeloid cells being utilized as OV carriers.
Buñuales et al. investigated primary human monocytes and a Syrian hamster monocyte/macrophage cell line (HM-1) [24] as carrier cells for an oncolytic adenovirus (oAdV) in nude mice with human tumors (HuH hepatocarcinoma cell line) and immunocompetent hamsters with hamster pancreatic tumors, respectively [4]. Freshly isolated human monocytes improved the biodistribution of oAdV in nude mice by intravenous injection, while the injection of HM-1 hamster cells resulted in the accumulation of oAdV in the liver. Biodistribution in vivo was measured with a luciferase-expressing adenoviral vector. The authors concluded that the selected carrier cells were not suitable for systemic delivery in an immunocompetent animal model and chose to use HM-1 cells as carriers for an intratumoral delivery of oAdV. In this setting, they showed that intratumoral HM-1-mediated OV administration allowed for repeated therapeutic gene expression after multiple injections, which was more efficient than after injections of naked virus. However, a limitation of this study is that reportedly, even uninfected macrophages from the HM-1 cell line failed to infiltrate the pancreatic tumor, thus not mimicking the real biological behavior of TAMs. Utilizing primary monocytes instead of a macrophage cell line could have potentially led to an improved infiltration.
Peng et al. [25] used dendritic cells to deliver measles virus to an immunocompromised murine model of human myeloma. Immature dendritic cells (iDCs) were selected due to their efficiency in the transmission of oncolytic measles viruses to myeloma cells. Briefly, primary human monocytes were collected and differentiated into iDCs in vitro, infected with an oncolytic measles virus expressing luciferase and injected in the tail vein of SCID mice harboring subcutaneous KAS 6/1 tumors. The authors showed that luciferase activity could be detected in tumors 48 h after the injection. By loading carrier cells with measles viruses expressing a red fluorescent protein (RFP), it was possible to demonstrate that infection was transmitted to tumor cells in vivo. Furthermore, in a systemic myeloma model (SCID mice intravenously injected with KAS 6/1 cells) iDCs loaded with measles virus extended survival but were not curative. This study provided proof-of-concept that monocyte-derived cells can specifically deliver an OV to a tumor, though it did not use an immunocompetent animal model.
A second approach employing DCs as carriers for OV therapy was reported by Ilett et al. [6][26]. Here, DCs were shown to internalize oncolytic reovirus and shield it from inactivation by neutralizing antibodies. Furthermore, it was demonstrated that reovirus-loaded DCs retained their functionality with regards to the phagocytic and T-cell priming potential.

References

  1. Hadryś, A.; Sochanik, A.; McFadden, G.; Jazowiecka-Rakus, J. Mesenchymal Stem Cells as Carriers for Systemic Delivery of Oncolytic Viruses. Eur. J. Pharmacol. 2020, 874, 172991.
  2. Kaufman, H.L.; Kohlhapp, F.J.; Zloza, A. Oncolytic Viruses: A New Class of Immunotherapy Drugs. Nat. Rev. Drug Discov. 2015, 14, 642–662.
  3. Hammad, M.; Cornejo, Y.R.; Batalla-Covello, J.; Majid, A.A.; Burke, C.; Liu, Z.; Yuan, Y.C.; Li, M.; Dellinger, T.H.; Lu, J.; et al. Neural Stem Cells Improve the Delivery of Oncolytic Chimeric Orthopoxvirus in a Metastatic Ovarian Cancer Model. Mol. Ther. Oncolytics 2020, 18, 326–334.
  4. Bunuales, M.; Garcia-Aragoncillo, E.; Casado, R.; Quetglas, J.I.; Hervas-Stubbs, S.; Bortolanza, S.; Benavides-Vallve, C.; Ortiz-De-Solorzano, C.; Prieto, J.; Hernandez-Alcoceba, R. Evaluation of Monocytes as Carriers for Armed Oncolytic Adenoviruses in Murine and Syrian Hamster Models of Cancer. Hum. Gene Ther. 2012, 23, 1258–1268.
  5. Ong, H.; Hasegawa, K.; Dietz, A.; Russell, S.; Peng, K.-W. Evaluation of T Cells as Carriers for Systemic Measles Virotherapy in the Presence of Antiviral Antibodies. Gene Ther. 2007, 14, 324–333.
  6. Ilett, E.; Prestwich, R.; Kottke, T.; Errington, F.; Thompson, J.M.; Harrington, K.J.K.; Pandha, H.A.; Coffey, M.; Selby, P.J.; Vile, R.G.; et al. Dendritic Cells and T Cells Deliver Oncolytic Reovirus for Tumour Killing despite Pre-Existing Anti-Viral Immunity. Gene Ther. 2009, 16, 689–699.
  7. Pfirschke, C.; Schirrmacher, V. Cross-infection of tumor cells by contact with T lymphocytes loaded with Newcastle disease virus. Int. J. Oncol. 2009, 34, 951–962.
  8. Qiao, J.; Kottke, T.; Willmon, C.; Galivo, F.; Wongthida, P.; Diaz, R.M.; Thompson, J.; Ryno, P.; Barber, G.N.; Chester, J.; et al. Purging Metastases in Lymphoid Organs Using a Combination of Antigen-Nonspecific Adoptive T Cell Therapy, Oncolytic Virotherapy and Immunotherapy. Nat. Med. 2008, 14, 37–44.
  9. Qiao, J.; Wang, H.; Kottke, T.; Diaz, R.M.; Willmon, C.; Hudacek, A.; Thompson, J.; Parato, K.; Bell, J.; Naik, J.; et al. Loading of Oncolytic Vesicular Stomatitis Virus onto Antigen-Specific T Cells Enhances the Efficacy of Adoptive T-Cell Therapy of Tumors. Gene Ther. 2008, 15, 604–616.
  10. Cole, C.; Qiao, J.; Kottke, T.; Diaz, R.M.; Ahmed, A.; Sanchez-Perez, L.; Brunn, G.; Thompson, J.; Chester, J.; Vile, R.G. Tumor-Targeted, Systemic Delivery of Therapeutic Viral Vectors Using Hitchhiking on Antigen-Specific T Cells. Nat. Med. 2005, 11, 1073–1081.
  11. Melzer, M.K.; Zeitlinger, L.; Mall, S.; Steiger, K.; Schmid, R.M.; Ebert, O.; Krackhardt, A.; Altomonte, J. Enhanced Safety and Efficacy of Oncolytic VSV Therapy by Combination with T Cell Receptor Transgenic T Cells as Carriers. Mol. Ther. Oncolytics 2019, 12, 26–40.
  12. VanSeggelen, H.; Tantalo, D.; Afsahi, A.-M.T. Chimeric Antigen Receptor-Engineered T Cells as Oncolytic Virus Carriers; Elsevier: Amsterdam, The Netherlands, 2015.
  13. Santos, J.; Heiniö, C.; Quixabeira, D.; Zafar, S.; Cells, J.C. Systemic Delivery of Oncolytic Adenovirus to Tumors Using Tumor-Infiltrating Lymphocytes as Carriers. Cells 2021, 10, 978.
  14. Yotnda, P.; Savoldo, B.; Charlet-Berguerand, N.; Blood, C.R. Targeted Delivery of Adenoviral Vectors by Cytotoxic T Cells. Blood 2004, 104, 2272–2280.
  15. Lasner, T.M.; Tal-Singer, R.; Kesari, S.; Lee, V.M.-Y.; Trojanowski, J.Q.; Fraser, N.W. Toxicity and Neuronal Infection of a HSV-1 ICP34.5 Mutant in Nude Mice. J. Neurovirol. 1998, 4, 100–105.
  16. Kanzaki, A.; Kasuya, H.; Yamamura, K.; Sahin, T.T.; Nomura, N.; Shikano, T.; Shirota, T.; Tan, G.; Fukuda, S.; Misawa, M.; et al. Antitumor efficacy of oncolytic herpes simplex virus adsorbed onto antigen-specific lymphocytes. Cancer Gene Ther 2012, 19, 292–298.
  17. Joyce, J.A.; Fearon, D.T. T Cell Exclusion, Immune Privilege, and the Tumor Microenvironment. Science 2015, 348, 74–80.
  18. Bellone, M.; Calcinotto, A. Ways to Enhance Lymphocyte Trafficking into Tumors and Fitness of Tumor Infiltrating Lymphocytes. Front. Oncol. 2013, 3, 231.
  19. Calcinotto, A.; Grioni, M.; Jachetti, E.; Curnis, F.; Mondino, A.; Parmiani, G.; Corti, A.; Bellone, M. Targeting TNF-α to Neoangiogenic Vessels Enhances Lymphocyte Infiltration in Tumors and Increases the Therapeutic Potential of Immunotherapy. J. Immunol. 2012, 188, 2687–2694.
  20. Lanitis, E.; Irving, M.; Coukos, G. Targeting the Tumor Vasculature to Enhance T Cell Activity; Elsevier: Amsterdam, The Netherlands, 2015.
  21. Mantovani, A.; Marchesi, F.; Jaillon, S.; Garlanda, C.; Allavena, P. Tumor-Associated Myeloid Cells: Diversity and Therapeutic Targeting. Cell. Mol. Immunol. 2021, 18, 566–578.
  22. Tan, Y.; Wang, M.; Zhang, Y.; Ge, S.; Zhong, F.; Xia, G.; Sun, C. Tumor-Associated Macrophages: A Potential Target for Cancer Therapy. Front. Oncol. 2021, 11, 3517.
  23. Cerezo-Wallis, D.; Ballesteros, I. Neutrophils in Cancer, a Love–Hate Affair. FEBS J. 2021, 1–12.
  24. Vanhaver, C.; van der Bruggen, P.; Bruger, A. MDSC in Mice and Men: Mechanisms of Immunosuppression in Cancer. J. Clin. Med. 2021, 10, 2872.
  25. Miyoshi, I.; Hiraki, S.; Kubonishi, I.; Matsuda, Y.; Nakayama, T.; Kishimoto, H.; Masuji, H.; Kimura, I. Establishment and Characterization of Two Hamster Macrophage Cell Lines. Cancer Lett. 1978, 4, 253–257.
  26. Peng, K.W.; Dogan, A.; Vrana, J.; Liu, C.; Ong, H.T.; Kumar, S.; Dispenzieri, A.; Dietz, A.B.; Russell, S.J. Tumor-Associated Macrophages Infiltrate Plasmacytomas and Can Serve as Cell Carriers for Oncolytic Measles Virotherapy of Disseminated Myeloma. Am. J. Hematol. 2009, 84, 401–407.
More
Academic Video Service