Research Article
J Immun Res. 2021; 7(2): 1041.
CAR T Cell-Packaged Oncolytic Vaccinia Virus Displays Enhanced Antitumor Efficacy
Kevin Song¹* and Xingbing Wang²*
1Icell Kealex Therapeutic, Houston, JLABS, 2450 Holcombe Blvd, Houston, TX, USA
2Center for Cell and Gene Therapy, Baylor College of Medicine, One Baylor Plaza, Houston, TX, USA
*Corresponding author: Kevin Song, Icell Kealex Therapeutic, Houston, JLABS, 2450 Holcombe Blvd, Houston, TX 77021, USA
Xingbing Wang, Center for Cell and Gene Therapy, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
Received: August 31, 2021; Accepted: September 09, 2021; Published: September 10, 2021
Abstract
Background: Oncolytic vaccinia virus is a promising cancer therapeutic modality. However, the effectiveness of oncolytic viruses is limited by several factors. Systemic or intratumoral delivery of vaccinia viruses with the subsequent quick clearance of the viruses from the tumor site and the body by the strong immune responses induced by the virus are among the key challenges. In this study, we explored CAR T cell-packaged oncolytic vaccinia virus as a combinational therapy strategy in order to overcome current limitations for oncolytic virotherapy.
Materials and Methods: We generated human HER2-CAR T cells and infected the HER2-CAR T cells with a EphA2-CD3 T cell engager-armed oncolytic vaccinia virus and evaluated the virus infectivity and replication within the T cells by flow analysis and virus tittering. T cell activation and cytotoxicity were determined by ELISA and 51Cr release assay.
Results: We demonstrated that oncolytic vaccinia virus infected human HER2-CAR T cells effectively and virus particle in the activated human T cells increased >1000 fold in 3 days. In addition, EphA2-CD3 T cell engager effectively activated HER2-CAR T cells in the presence of HER2dimEphA2high NSCLC A549 cell lines, indicated by the elevated expression level of IFNγ and IL2. Importantly, in vitro studies showed that HER2-CAR T cell-packaged EphA2-TEA-VV displayed enhanced cytotoxicity against HER2dimEphA2high NSCLC A549 cell lines compared to HER2-CAR T cells or EphA2-TEA-VV alone.
Conclusion: HER2-CAR T cell-packaged EphA2-TEA-VV is a promising therapeutic candidate with the ability to overcome the virus’s high immunogenicity and tumor heterogeneity, resulting in enhanced antitumor effects.
Keywords: CAR T cell therapy; Oncolytic vaccinia virus; Immunotherapy; T cell engager
Introduction
Oncolytic viruses have emerged as a promising cancer therapeutic modality since they specifically infect, replicate in, and lyse tumor cells without damaging normal cells. The tumor selectivity pegs oncolytic viruses as ideal delivery tools for the development of novel cancer immunotherapies [1-6]. Various oncolytic virotherapies have been developed by arming the virus with therapeutics genes, such as GM-CSF, IL-12, CD40-L, 4-1 BB-L, OX-40L, IL-2, TNF-a, anti- CTLA-4 antibody or anti-PD1/PD-L1 antibody [7-13]. Oncolytic virus has also been engineered to express Tumor Associate Antigens (TAA) in order to induce TAA-specific immune responses, providing long-term protection against tumor relapse [14-16]. In addition, oncolytic vaccinia virus has also been armed by T-cell engager (TEAVV), directing endogenous T cells to recognize and kill the tumor cells regardless of the oncolytic virus infection of the tumor cells, increasing the efficiency of oncolytic virotherapy [17-21].
Although the anti-tumor efficacy of oncolytic viruses has been proved by the animal and human studies, it has been realized that the effectiveness of oncolytic viruses is significantly hindered by several factors [22]. Rapid and efficient clearance of the virus by host immune system is among the key challenges. Immune factors such as antibodies neutralize the virus by binding to it directly and preventing a successful infection of the cells or by marking it for destruction either by complement or by immune cells [23-29]. With each subsequent administration of the oncolytic virus, the immune responses induced by the virus become faster and stronger, preventing the virus reaching the tumor cells. Intratumoral or regional injection of the oncolytic virus can certainly overcome this limitation by delivering all the viral particles directly to tumor or the surrounding tissues. However, this approach may be only suitable for some surficial tumors and does not take into the account cases in which the tumors are located at deep in the tissues or have metastasized to other locations. Thus, systemic administration of the virus is desirable since it potentially allows the virus to reach all the tumor cells.
Various strategies have been developed to overcome the antivirus neutralization antibody and deliver the virus systemically. For example, Cytokine-Induced Killer (CIK) cells have been utilized to package the oncolytic vaccinia virus and deliver the virus to the tumor cells via systemic injection route [30-32]. The studies have shown that systemic administration of CIK-packaged WR strain double deleted vaccinia virus (vvDD) successfully delivered the virus to tumor tissues and achieved regression of tumors in both immunocompetent and immunodeficient mouse models. Administration of CIKpackaged vvDD resulted in a prolonged persistence of the virus within the tumor tissues compared to administration of vvDD. And the vvDD-infection didn’t affect CIK cell’s ability of tumor homing and infiltrating. These results indicated the potential of immune cell-packaged oncolytic vaccinia virus for systemic therapy of solid tumors. Adoptive immune cellular therapy such as Chimeric Antigen Receptor (CAR) T cell therapy has demonstrated tremendous success in eradicating hematological malignancies resulting in FDA’s approval of four CAR T cell therapies for leukemia, lymphoma, and multiple myeloma [33-37]. However, the clinical results of CAR T cell therapy for solid tumors so far have been suboptimal. The limitations for the CAR T cell therapy for solid tumors might be due to tumor heterogeneity/antigen loss variants (ALV), limited tumor infiltration due to surrounding matrix, and the immune suppressive environment [38-43]. In this study, we developed a HER2-CAR T cell-packaged EphA2-TEA-VV strategy and evaluated VV’s infectivity of human T cells and its efficacy against tumor cells in vitro.
Materials and Methods
Cell line and blood donors
The breast cancer cell line SK-BR-3 that expresses HER2 and nonsmall- cell lung cancer cell (NSCLC) A549 that is HER2dimEphA2high [44,45] were purchased from the American Type Culture Collection (ATCC, Manassas, VA). All cell lines were grown in Dulbecco’s modified Eagle’s medium (Invitrogen, Carlsbad, CA) with 10% Fetal bovine serum (FBS, HyClone, Logan, UT), supplemented with 2mmol/l GlutaMAX-I, 1.5g/l sodium bicarbonate, 0.1mmol/l nonessential amino acids, and 1.0mmol/l sodium pyruvate (Invitrogen). Human Peripheral Blood Mononuclear Cells (PBMC) were used in this study and purchased from Gulf Coase Regional Blood Center in Houston, Texas. The individually identifiable private information was not collected for the study. Human PBMCs were maintained in RPMI 1640 with 10% FBS containing 2mmol/l GlutaMAX-I.
Production of retroviral vectors
The HER2-specific CAR vector was a gift from Dr. Stephen Gottschalk [40,41]. The HER2-specific CAR contains the HER2- specific single chain variable fragment derived from the anti-HER2 antibody FRP5 clone, a hinger region from human IgG1, a human CD28 transmembrane domain and a CD28.ζ signaling domain. To produce HER2-specifc retroviral supernatant, 293T cells were co-transfected with HER2-specific CAR vector, Peg-Pam-e plasmid containing the sequence for MoMLV gag-pol, and plasmid pMEVSVg encoding the sequence for vesicular stomatitis virus G, using GeneJuice transfection reagent (EMD Biosciences, San Diego, CA) following the manufacturer’s instructions. The 293T cell culture supernatants that contains the HER2-CAR retroviral particles were collected at 48 and 72 hours post plasmid transfection and subjected to retroviral transduction of human T cells.
Retroviral transduction of human T cells
Human PBMC were activated with anti-CD3 (OKT clone) and anti-CD28 antibodies followed by retroviral transduction as previously described [40,41]. Briefly, human PBMC were washed with phosphate-buffered saline (PBS; Sigma, St Louis, MO) for three times and seed in 24-well plate that was pre-coated with anti- CD3 antibody OKT3 (OrthoBiotech, Raritan, NJ) and anti-CD28 antibodie (BD Biosciences, Palo Alto, CA) at a final concentration of 1μg/ml for each antibody. 24 hours later, the cells were harvested for retroviral transduction. A non-tissue culture treated 24-well plate was pre-coated with a recombinant fibronectin fragment (FN CH-296; Retronectin; Takara Bio USA, Madison, WI) following the manufacturer’s instructions. Wells were then washed with PBS and incubated twice for 30 minutes with the 293T cell culture supernatants that contains the HER2-specifc retroviral particles. And then 3x105 anti-CD3/CD28 antibody activated human T cells were added to each well in the presence of 100 U IL-2/ml. 48 hours later, cells were harvested and cultured in the presence of 100 U/IL2/ml for 5 days prior to use.
VV transduction of human T cells
Double-deleted VVs (vvDD, Western Reserve strain) expressing EphA2-scFv-CD3-scFv T cell engager (EphA2-TEA-VV) was generated as previously described [20]. The EphA2-TEA-VV also expresses Red Fluorescent Protein (RFP) allowing flow analysis of the VV infectivity. To infect the human T cells, HER2-CAR T cells or control T cells were infected with EphA2-TEA-VV or control VVs at increasing MOIs (i.e. 0.1, 1, or 5) in minimum essential medium with 2.5% FBS for 2 hours at 37°C. Then the medium was replaced with fresh RPMI medium with 10% FBS, and T cells were incubated until harvesting at 1, 2, 3, 4, 5, and 7 days after infection. After three freeze-thaw cycles, virus was quantified by plaque tittering on CV-1 cells as described previously.
Flow cytometry
To determine the HER2-specfic CAR expression on the surface of human T cells, the T cells were first incubated with a recombinant HER2-Fc fusion protein (R&D Systems, Minneapolis, MN) for 30 minutes followed by staining with a goat anti-Fc FITC secondary antibody (Chemicon, Temecula, CA). T cells were washed three times with PBS containing 2% FBS and 0.1% sodium azide (Sigma; FACS buffer) prior to addition of HER2-Fc or antibodies. To determine the VV infectivity, human T cells were infected with EphA2-TEAVV expressing RFP and the RFP expression in T cells was used to determine the VV infectivity. The cells were analyzed using a FACScalibur instrument (Becton Dickinson, Mountain View, CA) and CellQuest software (Becton Dickinson).
Chromium-51 release assay
Chromium-51 release assay was performed as previously described. Briefly, tumor cells were labeled with 51Cr as described and mixed with decreasing numbers of HER2-specific CAR T cells to give T cells to tumor cells ratios of 40:1, 20:1, 10:1, 5:1, and 0:1. 51Cr labeled tumor cells were incubated in cell culture medium alone or in 1% Triton X-100 as negative or positive control to determine spontaneous or maximum 51Cr release respectively. 51Cr labeled tumor cells and HER2-CAR T cells were co-cultured for 4 hours, and supernatants were collected and assessed as described.
Co-culture of tumor cells and HER2-CAR T cells and analysis of cytokine production
HER2-specfic CAR T cells from healthy donors were co-cultured with HER2-positive tumor cell lines at a 2:1 T cell to tumor cell ratio in 24-well cell culture plate in the presence of 100 U IL-2/ml. After 48 hours incubation, culture supernatants were harvested and subjected to enzyme-linked immunosorbent assay (ELISA) to measure the presence of IFN-γ and IL-2 following the manufacturer’s instruction (R&D Systems).
Statistical analysis
All in vitro experiments were performed in duplicate or in triplicate. The data were presented as mean ± SD. The differences between means were tested by student t-test. The significance level used was P < 0.05.
Results
Generation of HER2-specific CAR modified human T cells
We generated a second-generation CAR specific for human HER2 using the single chain variable fragment scFv derived from FRP5 antibody (HER2-CAR). Human T cells were activated by anti-CD3/CD28 antibodies and transduced with a retroviral vector encoding HER2-CAR to generate HER2-specific T cells (HER2-T cells). Seven days after transduction, the T cells were cultured with HER2-Fc fusion protein for 30 minutes followed by staining with FITC conjugated anti-Fc antibody. HER2-specific CAR expression was then measured by flow cytometry. Over 90% of the T cells were HER2-CAR positive (Figure 1A), indicating the susceptibility of anti- CD3/CD28 antibodies activated human T cell to retroviral infection.
Figure 1: Construction of HER2-CAR T cells. OKT3/CD28 activated human T cells were transduced with the retrovirus encoding HER-2 specific CAR. The HER2- specific CAR expression on the surface of the T cells were measured by flow cytometry (A). HER2-positive SK-BR-5 breast tumor cells were labeled with 51Cr and 51Crrelease assay was conducted to determine the tumor lysis by the HER2-CAR T cells (B).
We next investigated the cytotoxicity of HER2-specific T cells. SK-BR-5 is a HER2 overexpressed human breast tumor cell line and has been widely used by other investigators for the development of HER2-targeted therapies. A standard 51Cr release cytotoxicity assay was conducted to determine the cytotoxicity of HER2-speicifc T cells against SK-BR-5. HER2-specific T cells as well as non-transduced (NT) T cells were incubated with SK-BR-5. The results suggested that HER2-CAR T cells induced SK-BR-5 tumor cell lysis effectively, whereas non-transduced T cells (NT-T) didn’t kill SK-BR-5 cell lines due to the lack of HER2 CAR expression on the surface of the T cells (Figure 1B). These results indicated that the T cell-mediated killing of HER2-positive breast tumor cell line depended on the expression of HER2-specific CARs on the T cells.
Oncolytic VV infected HER2-CAR T cells effectively
We next investigated EphA2-TEA-VV’s infectivity in HER2- CAR T cells. EphA2-TEA-VV was produced as described previously. EphA2-TEA-VV also encodes RFP, allowing us to identify the virus infected cells. Human PBMCs were activated by anti-CD3/CD28 antibodies followed by transduction of retroviral vector encoding HER2-specific CAR. The HER2-specfic T cells were cultured in the presence of 100U IL2/ml for 7 days prior to the VV infection. Nontransduced T cells and CIKs were produced as control. CIK cells are cytokine-induced cell population with phenotypic markers of NK and T-cells. Human PBMC was activated by interferon-γ and anti- CD3 antibody and expansion in IL-2 as described previously. HER2- CAR T cells (CAR T), CIK and non-transduced T cells (NT-T) were infected with EphA2-TEA-VV at increasing MOI of 0, 0.1, 1, to 5. At different time point, the cells were harvested and subjected to flow analysis of EphA2-TEA-VV’s infectivity. The results showed that EphA2-TEA-VV infected HER2-CAR T cells (CAR T), CIK, or NT-T cells effectively. We observed that 27.79% - 36.56% of HER2-CAR T cells (CAR T), CIK, or NT-T cells were infected with EphA2-TEAVV at day 7 (Figure 2A). Comparison of MOIs revealed that EphA2-TEA-VV infection at MOI 1 resulted in an optimal infectivity in HER2-CAR T cells (CAR T), CIK, or NT-T cells (Figure 2A and 2B). Thus, we infected T cells with EphA2-TEA-VV or GFP-VV at MOI 1 in our following experiments. In addition, the cells were collected at different time point, and flow analysis demonstrated that the significant RFP expression was detected from day 3 to day 14 (Figure 2B). Taken together, these data suggested that EphA2-TEA-VV could infect CAR T cells, CIK, and NT-T cells effectively.
Figure 2: EphA2-TEA-VV effectively infected HER2-CAR T cells in vitro. HER2-CAR T cells (CAR T), CIK and non-transduced T cells (NT-T) were infected with EphA2-TEA-VV that encodes RFP at an MOI of 0.1 to 5 PFU/cell. CIK cells are cytokine-induced cell population with phenotypic markers of NK and T-cells. Human PBMC was activated interferon-γ and anti-CD3 antibody and expansion in interleukin-2, and has been described previously. (A) At day 7 post virus infection, the cells were collected and subjected to the flow analysis of viral-derived RFP expression by the cells. The viral-derived RFP expression by EphA2-TEA-VV loaded HER2-CAR T cells (B), non-transduced T cells (C), or CIK cells (D) at day 0, 1, 2, 3, 4, 7, 10, or 14 were analyzed and shown in a dot plot figure.
Oncolytic VV replicated in HER2-CAR T cells effectively
We investigated if the VVs can replicate effectively in CAR T cells. EphA2-TEA-VV or GFP-VV was transduced to HER2-CAR T cells at MOI 1. At different time point post infection, the T cells (CAR T) as well as cell culture medium (CM) were collected and subjected to the virus tittering in order to determine if the virus replicated effectively in CAR T cells or released in the culture medium. We observed that either EphA2-TEA-VV or GFP-VV particle in HER2-CAR T cells (Figure 3A) or NT-T cells (Figure 3B) increased > 1000-fold in 3 days. And up to 80% of the virus particles were located in T cells, while up to 20% of the virus particles were released to the culture medium of HER2-CAR T cells (Figure 3C) or NT-T cells (Figure 3D). These results indicated that oncolytic vaccinia virus replicated in HER2- CAR T cells effectively.
Figure 3: EphA2-TEA-VV replicated significantly in HER2-CAR T cells. HER2-CAR T cells non-transduced T cells were transduced with GFP-VV (CAR T + GFPVV or NT-T + GFP-VV) or EphA2-TEA-VV (CAR T + EphA2-vv, or NT-T + EphA2-vv) at MOI 1. At day 0, 1, 2, 3, 5 or 7-post VV transduction, the EphA2-TEA-VV or GFP-VV loaded HER2-CAR T cells (A) or non-transduced T cells (B) were collected and subjected to the virus tittering using CV-1 plaque assay. To determine if the virus particles are in the cytoplasm or released to the cell culture medium, the cells (CAR T) or cell culture medium (CM) of EphA2-TEA-VV loaded HER2-CAR T cells (C) or non-transuded T cells (D) were collected and subjected to the virus tittering using CV-1 plaque assay.
HER2-CAR T cell-packaged EphA2-TEA-VV displayed enhanced tumor killing ability
We next investigated if HER2-CAR T cell-packaged EphA2- TEA-VV displayed enhanced tumor killing ability since it co-targets HER2 and EphA2 in tumor cells. To do this, a HER2dimEphA2high NSCLC A549 cell line was used as the target. A standard 51Cr release assay was conducted to determine the cytotoxicity of HER2-CAR T cell-packaged EphA2-TEA-VV against A549. The results showed that HER2-CAR T cells that were infected with EphA2-TEA-VV at either MOI 0.1, 1, or 5-killed A549 tumor cells effectively. In contrast, no killing of A549 tumor cells by GFP-VV-loaded HER2-CAR T cells was observed (Figure 4A). This is likely due to the low HER2 expression density on the surface of A549 tumor cells and the suboptimal T cell function caused by the virus. The observed A549 killing by EphA2- TEA-VV loaded HER2-CAR T cells indicated that co-targeting HER2 and EphA2 on the surface of A549 resulted in the effective killing of A549 tumor cells. In addition, we compared the efficacy of EphA2- TEA-VV loaded HER2-CAR T cells (TEA-VV/T) with either VVs alone or HER2-CAR T cells alone and found that TEA-VV/T is superior to HER2-CAR T cells alone (T only), EphA2-TEA-VV alone (TEA-VV) or GFP-VV alone (Figure 4B). These results suggested that HER2-CAR T cell-packaged EphA2-TEA-VV represents a unique dual-targeting strategy with the ability of targeting both HER2 and EphA2 on the surface of the tumor cells, resulting in effective killing of tumor cells that express both HER2 and EphA2.
Figure 4: HER2-CAR T cell-packaged EphA2-TEA-VV displayed enhanced anti-tumor activity in vitro. A549 cells were labeled with 51Cr and mixed with decreasing numbers of T cells to give T (effector) cells to tumor (target) cells (E:T) ratios of 40:1, 20:1, 10:1, 5:1, and 0:1. 51Cr labeled A549 tumor cells and T cells were cocultured for 4 hours, and supernatants were collected and assessed as described. (A) Data was analyzed for the co-culture of A549 with EphA2-TEA-VV loaded HER2-CAR T cells (EphA2-TEA-VV) or GFP-VV loaded HER2-CAR T cells (GFP-VV). *P value <0.05: A549 cell lysis rate by HER2-CAR T cells loaded with EphA2-TEA-VV at MOI of 0.1, 1, or 5 (EphA2-TEA-VV) vs. that of HER2-CAR T cells loaded with GFP-VV at MOI of 0.1, 1, or 5 (GFP-VV). (B) Data was analyzed for the culture of A549 tumor cells alone (Tumor only), GFP-VV infected A549 tumor cells (GFP-VV), EphA2-TEA-VV infected A549 tumor cells (TEA-VV), nontransduced T cells (T only), A549 tumor cells with GFP-VV loaded HER2-CAR T cells (GFP-VV/T), and A549 tumor cells with EphA2-TEA-VV loaded HER2-CAR T cells (TEA-VV/T). *P value <0.05: GFP-VV, TEA-VV, or T only vs. GFP-VV/T. **P value <0.05: TEA-VV/T vs. GFP-VV/T.
EphA2-TEA-VV activated HER2-CAR T cells in the presence of EphA2-positive A549 cells
We also investigated if EphA2-CD3 engager activated HER2-T cells in the presence of EphA2-positive A549 tumor cells. EphA2- TEA-VV loaded HER2-specfic CAR T cells (CAR T + EphA2-VV), GFP-VV loaded HER2-specfic CAR T cells (CAR T + GFP-VV), HER2-specific CAR T cells (CAR T), or non-transduced T cells (NT-T) from healthy donors were co-cultured with A549 tumor cells. After 48 hours incubation, interferon (IFN) - γ and interleukin (IL)-2 production in the cell culture medium was determined by enzyme-linked immunosorbent assay (ELISA). EphA2-TEA-VV loaded HER2-specific T cells produced elevated level of IFN-γ and IL-2 compared to GFP-VV loaded HER2-specific T cells, indicating that elevated cytokine production was dependent on stimulation of EphA2 antigen. These results showed that tumor antigen dual targeting strategy could enhance T cell activation.
Figure 5: EphA2-TEA-VV activated HER2-CAR T cells in the presence of EphA2-positive A549 cells in vitro. 2x105 of EphA2-TEA-VV loaded HER2- specfic CAR T cells (CAR T + EphA2-VV), GFP-VV loaded HER2-specfic CAR T cells (CAR T + GFP-VV), HER2-specific CAR T cells (CAR T), or non-transduced T cells (NT-T) from healthy donors were co-cultured with 1x105 of HER2- and EphA2- double positive tumor cell A549 at a 2:1 T cell to tumor cell ratio in 24-well cell culture plate in the presence of 100 U of IL2/ ml. The cells were cultured in triplicate. After 48 hours incubation, culture supernatants were collected and subjected to enzyme-linked immunosorbent assay (ELISA) to measure the presence of IFN-γ and IL-2 following the manufacturer’s instructions (R&D Systems). The data were presented as average + SD. *P value <0.05: IFN-r and IL2 production of EphA2-TEA-VV loaded HER2-CAR T cells (CAR T + EphA2-VV) vs. that of GFP-VV loaded HER2-CAR T cells (CAR T + GFP-VV).
Discussion
In this study, we developed the HER2-CAR T cell-packaged EphA2-TEA-VV and assessed its activity in vitro. We first generated a second generation of anti-HER2 antibody FRP5 based HER2-CAR and constructed the HER2-CAR T cells. In vitro cell culture assays demonstrated the HER2-CAR T cells killed HER2-positive SK-BR-3 human breast tumor cell line effectively. We then assessed the EphA2- TEA-VV’s infectivity in HER2-CAR T cells and more than 30% of the T cells were infected with VVs at 3 days post infection. We found the VVs replicated effectively in the T cell culture. The virus particles increased > 1000-fold in 3 days. And the majority of the virus was found in the T cells, while less than 20% of the viruses were released to the cell culture medium. The co-culture of HER2dimEphA2high NSCLC line A549 and the EphA2-TEA-VV loaded HER2-CAR T cells suggested that HER2-CAR T cell-packaged EphA2-TEA-VV displayed enhanced tumor killing ability compared to HER2-CAR T cells or EphA2-TEA-VV alone. In addition, EphA2-TEA-VV activated HER2-CAR T cells in the presence of EphA2-positive tumor cells, indicated by the elevated expression level of cytokines such as IFNγ and IL-2 by HER2-CAR T cells.
While some patients with HER2-positive breast tumors have benefited from anti-HER2 monoclonal antibody therapy, one arising challenge in the treatment of breast cancer is that some patients with HER2-positive breast tumors are resistant to the treatment of trastuzumab, an anti-HER2 monoclonal Ab [46-49]. Studies have shown that the resistance to trastuzumab is largely due to the elevated level of EphA2 in the breast tumors [50-52]. The elevated level of EphA2 in breast tumors is also correlated with the decrease in the overall and disease-free survival of the patients with HER2-positive breast tumors. Studies also showed that inhibiting EphA2 using anti- EphA2 antibody successfully restored the efficacy of trastuzumab treatment for HER2-positive breast tumor in mouse models. Thus, co-targeting EphA2 and HER2 provides a potentially effective strategy to treat the patients with trastuzumab-resistant breast tumors. In addition, both EphA2 and HER2 are overexpressed in other types of cancers at various level. For example, NSCLC has been reported to express both EphA2 and HER2. In this study, we have assessed the efficacy of our strategy to treat a HER2dimEphA2high NSCLC line A549 cells. Our results demonstrated that co-targeting EphA2 and HER2 enhanced anti-tumor activity compared to targeting EphA2 by EphA2-TEA-VV alone or targeting HER2 by HER2-CAR T cells alone. Thus, such a strategy that co-targets both EphA2 and HER2 might be suitable to the treatment of the patients with EphA2 and HER2 double positive tumors by overcoming the existing or potential resistance to HER2 targeted therapies.
In addition, such a bispecific therapeutic strategy that co-targets two TAAs has the potential to overcome the limitation of the therapies that target single target and have been proved to induce antigen loss variant (ALV). For example, clinical studies of CD19-CAR T cell therapies in B-ALL reported CD19 ALV in 30-95% of relapses [53]. The reported CD19 ALV is resulted from splice mutations and retained intracellular CD19 [54-57]. To overcome this limitation, current efforts have been focused on the development of bispecific CAR T cells with dual targeting of CD19 and CD22 [58-61]. Similar cases with ALV associated treatment resistance have been reported with a monospecific CD22-CAR or BCMA-CAR T cell therapy [62,63]. In addition, several reports have suggested that effective immunotherapies are often correlated with high target antigen expression density [64-68]. Thus, it remains an important unmet goal to develop novel strategies to overcome the resistance caused by monotherapy-induced ALV or the low target antigen expression density.
Conclusion
In conclusion, we designed and assessed the HER2-CAR T cellpackaged EphA2-TEA-VV and provided the preclinical evidence for the therapeutic potential of such a combinational strategy for patients with trastuzumab-resistant breast tumors or other cancers (i.e. EphA2- and HER2- positive NSCLC). In addition, our CAR T cell-packaged T-cell engager armed VV strategy may be applicable to a broad range of CAR T cells and TEA-VVs currently under investigation.
Declaration
Author’s contributions: Conceptualization: X.W; Methodology: K.S and X.W; Validation: X.W; Formal analysis: K.S, X.W; Investigation: K.S, X.W; Resources: X.W; Data curation: X.W; Writing-original draft preparation: K.S; Writing-review and editing: K.S; Visualization: K.S, X.W; Supervision: X.W; Project administration: X.W. All authors have read and agreed to the published version of the manuscript.
Funding: This research was funded by NIH R01 R01CA148748 to Baylor College of Medicine.
Conflicts of interest and current contact information: The authors declare no conflict of interest. Kevin Song’s current contact in-formation is Debakey High School, Houston, TX, USA; Xingbing Wang’s current contact information is The First Affiliated Hospital of USTC, P.R. China, Department of Hematology, 17 Lujiang Road, Hefei, Anhui.
References
- Chaurasiya S, Fong Y, Warner SG. Oncolytic Virotherapy for Cancer: Clinical Experience. Biomedicines. 2021; 9: 419.
- Moaven O, W Mangieri C, A Stauffer J, Anastasiadis PZ, Borad MJ. Evolving Role of Oncolytic Virotherapy: Challenges and Prospects in Clinical Practice. JCO Precis Oncol. 2021; 5.
- Ehrlich M, Bacharach E. Oncolytic Virotherapy: The Cancer Cell Side. Cancers (Basel). 2021; 13: 939.
- Martinez-Quintanilla J, Seah I, Chua M, Shah K. Oncolytic viruses: Overcoming translational challenges. J. Clin. Investig. 2019; 129: 1407-1418.
- Twumasi-Boateng K, Pettigrew JL, Kwok YYE, Bell JC, Nelson BH. Oncolytic viruses as engineering platforms for combination immunotherapy. Nat. Rev. Cancer. 2018; 18: 419-432.
- Bommareddy PK, Shettigar M, Kaufman HL. Integrating oncolytic viruses in combination cancer immunotherapy. Nat. Rev. Immunol. 2018; 18: 498-513.
- Harrington K, Freeman DJ, Kelly B, Harper J, Soria JC. Optimizing oncolytic virotherapy in cancer treatment. Nat Rev Drug Discov. 2019; 18: 689-706.
- Meyers DE, Wang AA, Thirukkumaran CM, Morris DG. Current Immunotherapeutic Strategies to Enhance Oncolytic Virotherapy. Front Oncol. 2017; 7: 114.
- Moesta AK, et al. Local delivery of OncoVEX (mGM-CSF) generates systemic antitumor immune responses enhanced by cytotoxic T-lymphocyteassociated protein blockade. Clin. Cancer Res. 2017; 23: 6190-6202.
- Cockle JV, Rajani K, Zaidi S, Kottke T, Thompson J, Diaz RM, et al. Combination viroimmunotherapy with checkpoint inhibition to treat glioma, based on location-specific tumor profiling. Neuro Oncol. 2016; 18: 518-527.
- LaRocca CJ & Warner SG. Oncolytic viruses and checkpoint inhibitors: combination therapy in clinical trials. Clin. Transl Med. 2018; 7: 35.
- Harrington KJ, et al. Safety and preliminary efficacy of talimogene laherparepvec (T-VEC) in combination (combo) with pembrolizumab (pembro) in patients (pts) with recurrent or metastatic squamous cell carcinoma of the head and neck (R/M HNSCC): a multicenter, phase 1b study (MASTERKEY-232). J. Clin. Oncol. 2018; 36: 6036.
- Chesney J, et al. Randomized, open-label phase II study evaluating the efficacy and safety of talimogene laherparepvec in combination with ipilimumab versus ipilimumab alone in patients with advanced, unresectable melanoma. J. Clin. Oncol. 2018; 36: 1658-1667.
- Pol JG, Bridle BW, Lichty BD. Detection of Tumor Antigen-Specific T-Cell Responses After Oncolytic Vaccination. Methods Mol Biol. 2020; 2058: 191- 211
- Sharp DW & Lattime EC. Recombinant poxvirus and the tumor microenvironment: oncolysis, immune regulation and immunization. Biomedicines. 2016; 4: 19.
- Pol JG, et al. Preclinical evaluation of a MAGE-A3 vaccination utilizing the oncolytic Maraba virus currently in first-in-human trials. Oncoimmunology. 2018; 8: e1512329.
- Fajardo CA, et al. Oncolytic adenoviral delivery of an EGFR-targeting T cell engager improves antitumor efficacy. Cancer Res. 2017; 77: 2052-2063.
- Freedman JD, et al. Oncolytic adenovirus expressing bispecific antibody targets T cell cytotoxicity in cancer biopsies. EMBO Mol. Med. 2017; 9: 1067- 1087.
- Guo ZS, Lotze MT, Zhu Z, Storkus WJ, Song XT. Bi- and Tri-Specific T Cell Engager-Armed Oncolytic Viruses: Next-Generation Cancer Immunotherapy. Biomedicines. 2020; 8: 204.
- Yu F, Wang X, Guo ZS, Bartlett DL, Gottschalk SM, Song XT. T-cell engagerarmed oncolytic vaccinia virus significantly enhances antitumor therapy. Mol Ther. 2014; 22: 102-111.
- Song XT. Combination of virotherapy and T-cell therapy: arming oncolytic virus with T-cell engagers. Discov Med. 2013; 16: 261-266.
- Ferguson MS, Lemoine NR, Wang Y. Systemic delivery of oncolytic viruses: Hopes and hurdles. Adv. Virol. 2012; 2012: 805629.
- Singh K, Gittis AG, Gitti RK, Ostazeski SA, Su HP, Garboczi DN. The vaccinia virus H3 envelope protein, a major target of neutralizing antibodies, exhibits a glycosyltransferase fold and binds UDP-glucose. J. Virol. 2016; 90: 5020- 5030.
- Matho MH, Maybeno M, Benhnia MR, Becker D, Meng X, Xiang Y, et al. Structural and biochemical characterization of the vaccinia virus envelope protein D8 and its recognition by the antibody LA5. J. Virol. 2012; 86: 8050- 8058.
- Kaever T, Matho MH, Meng X, Crickard L, Schlossman A, Xiang Y, et al. Linear epitopes in vaccinia virus A27 are targets of protective antibodies induced by vaccination against smallpox. J. Virol. 2016; 90: 4334-4345.
- Kaever T, Meng X, Matho MH, Schlossman A, Li S, Sela-Culang I, et al. Potent neutralization of vaccinia virus by divergent murine antibodies targeting a common site of vulnerability in L1 protein. J. Virol. 2014; 88: 11339-11355.
- Matho MH, de Val N, Miller GM, Brown J, Schlossman A, Meng X, et al. Murine anti-vaccinia virus D8 antibodies target different epitopes and differ in their ability to block D8 binding to CS-E. PLoS Pathogens. 2014; 10: e1004495.
- Moss B. Smallpox vaccines: Targets of protective immunity. Immunol. Rev. 2011; 239: 8-26.
- Ichihashi Y, Oie M. Neutralizing epitope on penetration protein of vaccinia virus. Virology. 1996; 220: 491-494.
- Thorne SH, Negrin RS, Contag CH. Synergistic antitumor effects of immune cell-viral biotherapy. Science. 2006; 311: 1780-1784.
- Thorne SH, Contag CH. Integrating the biological characteristics of oncolytic viruses and immune cells can optimize therapeutic benefits of cell-based delivery. Gene Ther. 2008; 15: 753-758.
- Sampath P, Li J, Hou W, Chen H, Bartlett DL, Thorne SH. Crosstalk between immune cell and oncolytic vaccinia therapy enhances tumor trafficking and antitumor effects. Mol Ther. 2013; 21: 620-628.
- Maude SL, Frey N, Shaw PA, Aplenc R, Barrett DM, Bunin NJ, et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. N Engl J Med. 2014; 371: 1507-1517.
- Davila ML, Riviere I, Wang X, Bartido S, Park J, Curran K, et al. Efficacy and toxicity management of 19-28z CAR T cell therapy in B cell acute lymphoblastic leukemia. Sci Transl Med. 2014; 6: 224ra25.
- Turtle CJ, Hanafi LA, Berger C, Gooley TA, Cherian S, Hudecek M, et al. CD19 CAR T cells of defined CD4+:CD8+ composition in adult B cell ALL patients. J Clin Invest. 2016; 126: 2123-2138.
- Turtle CJ, Hanafi LA, Berger C, Hudecek M, Pender B, Robinson E, et al. Immunotherapy of non-Hodgkin’s lymphoma with a defined ratio of CD8+ and CD4+ CD19-specific chimeric antigen receptor-modified T cells. Sci Transl Med. 2016; 8: 355ra116.
- Lee DW, Kochenderfer JN, Stetler-Stevenson M, Cui YK, Delbrook C, Feldman SA, et al. T cells expressing CD19 chimeric antigen receptors for acute lymphoblastic leukaemia in children and young adults: a phase 1 doseescalation trial. Lancet. 2015; 385: 517-528.
- Zhai B, Shi D, Gao H, Qi X, Jiang H, Zhang Y, et al. A phase I study of anti- GPC3 chimeric antigen receptor modified T cells (GPC3 CAR T) in Chinese patients with refractory or relapsed GPC3+ hepatocellular carcinoma (r/r GPC3+ HCC). J Clin Oncol. 2017; 35.
- Heczey A, Louis CU, Savoldo B, Dakhova O, Durett A, Grilley B, et al. CAR T cells administered in combination with lymphodepletion and PD-1 inhibition to patients with neuroblastoma. Mol Ther. 2017; 25: 2214-2224.
- Ahmed N, Brawley V, Hegde M, Bielamowicz K, Kalra M, Landi D, et al. HER2-specific chimeric antigen receptor-modified virus-specific T cells for progressive glioblastoma: a phase 1 dose-escalation trial. JAMA Oncol. 2017; 3: 1094-1101.
- Ahmed N, Brawley VS, Hegde M, Robertson C, Ghazi A, Gerken C, et al. Human epidermal growth factor receptor 2 (HER2)-specific chimeric antigen receptor-modified T cells for the immunotherapy of HER2-positive sarcoma. J Clin Oncol. 2015; 33: 1688-1696.
- Park JR, Digiusto DL, Slovak M, Wright C, Naranjo A, Wagner J, et al. Adoptive transfer of chimeric antigen receptor re-directed cytolytic T lymphocyte clones in patients with neuroblastoma. Mol Ther. 2007; 15: 825-833.
- Louis CU, Savoldo B, Dotti G, Pule M, Yvon E, Myers GD, et al. Antitumor activity and long-term fate of chimeric antigen receptor-positive T cells in patients with neuroblastoma. Blood. 2011; 118: 6050-6056.
- Subik K, Lee JF, Baxter L, Strzepek T, Costello D, Crowley P, et al. The Expression Patterns of ER, PR, HER2, CK5/6, EGFR, Ki-67 and AR by Immunohistochemical Analysis in Breast Cancer Cell Lines. Breast cancer: basic and clinical research. 2010; 4: 35-41.
- Paul A. Bunn Jr, Barbara Helfrich, Ariel F. Soriano, Wilbur A. Franklin, Marileila Varella-Garcia, Fred R. Hirsch, et al. Chan. Expression of Her- 2/neu in Human Lung Cancer Cell Lines by Immunohistochemistry and Fluorescence in Situ Hybridization and its Relationship to in Vitro Cytotoxicity by Trastuzumab and Chemotherapeutic Agents. Clin Cancer Res. 2001; 7: 3239-3250.
- Vogel CL, Cobleigh MA, Tripathy D, et al. Efficacy and safety of trastuzumab as a single agent in first-line treatment of HER2- overexpressing metastatic breast cancer. J Clin Oncol. 2002; 20: 719-726.
- Slamon DJ, Leyland-Jones B, Shak S, et al. Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast can- cer that overexpresses HER2. N Engl J Med. 2001; 344: 783-792.
- Valabrega G, Capellero S, Cavalloni G, Zaccarello G, Petrelli A, Migliardi G, et al. HER2-positive breast cancer cells resistant to trastuzumab and lapatinib lose reliance upon HER2 and are sensitive to the multi targeted kinase inhibitor sorafenib. Breast Cancer Res Treat. 2011; 130: 29-40.
- Joshi JP, Brown NE, Griner SE, Nahta R. Growth differentiation factor 15 (GDF15)-mediated HER2 phosphorylation reduces trastuzumab sensitivity of HER2-overexpressing breast cancer cells. Biochem Pharmacol. 2011; 82: 1090-1099.
- Brantley-Sieders DM, Zhuang G, Hicks D, et al. The receptor tyrosine kinase EphA2 promotes mammary adenocarcinoma tumorigenesis and metastatic progression in mice by amplifying ErbB2 signaling. J Clin Invest. 2008; 118: 64-78.
- Zhuang G, Brantley-Sieders DM, Vaught D, Yu J, Xie L, Wells S, et al. Elevation of Receptor Tyrosine Kinase EphA2 Mediates Resistance to Trastuzumab Therapy. Cancer Res. 2010; 70: 299-308.
- Youngblood VM, Kim LC, Edwards DN, Hwang Y, Santapuram PR, Stirdivant SM, et al. The Ephrin-A1/EPHA2 Signaling Axis Regulates Glutamine Metabolism in HER2-Positive Breast Cancer. Cancer Res. 2016; 76: 1825- 1836.
- Majzner RG & Mackall CL. Tumor antigen escape from CAR T-cell therapy. Cancer Discov. 2018; 8: 1219-1226.
- Bagashev A, et al. CD19 alterations emerging after CD19-directed immunotherapy cause retention of the misfolded protein in the endoplasmic reticulum. Mol. Cell. Biol. 2018; 38: e00383-18.
- Asnani M, et al. Retention of CD19 intron 2 contributes to CART-19 resistance in leukemias with subclonal frameshift mutations in CD19. Leukemia. 2020; 34: 1202-1207.
- Orlando EJ, et al. Genetic mechanisms of target antigen loss in CAR19 therapy of acute lymphoblastic leukemia. Nat. Med. 2018; 24: 1504-1506.
- Sotillo E, et al. Convergence of acquired mutations and alternative splicing of CD19 enables Resistance to CART-19 Immunotherapy. Cancer Discov. 2015; 5: 1282-1295.
- Spiegel JY, Patel S, Muffly L, Hossain NM, Oak J, Baird JH, et al. CAR T cells with dual targeting of CD19 and CD22 in adult patients with recurrent or refractory B cell malignancies: a phase 1 trial. Nat Med. 2021; 27: 1419-1431.
- Liu S, Deng B, Yin Z, Lin Y, An L, Liu D, et al. Combination of CD19 and CD22 CAR T cell therapy in relapsed B-cell acute lymphoblastic leukemia after allogeneic transplantation. Am J Hematol. 2021; 96: 671-679.
- Wei G, Zhang Y, Zhao H, Wang Y, Liu Y, Liang B, et al. CD19/CD22 Dual- Targeted CAR T-cell Therapy for Relapsed/Refractory Aggressive B-cell Lymphoma: A Safety and Efficacy Study. Cancer Immunol Res. 2021.
- Cao Y, Yi X, Wang N, Wang G, Huang L, Hong Z, et al. CD19/CD22 CAR T cell Cocktail Therapy Following Autologous Transplantation in Patients with Relapsed/Refractory Aggressive B Cell Lymphomas: Combined CAR T Cell Therapy and Transplantation. Transplant Cell Ther. 2021: S2666- 6367(21)01148-9.
- Fry TJ, et al. CD22-targeted CAR T cells induce remission in B-ALL that is naive or resistant to CD19-targeted CAR immunotherapy. Nat. Med. 2018; 24: 20-28.
- Cohen AD, et al. B cell maturation antigen-specific CAR T cells are clinically active in multiple myeloma. J. Clin. Invest. 2019; 129: 2210-2221.
- Majzner RG, et al. CAR T cells targeting B7-H3, a pan-cancer antigen, demonstrate potent preclinical activity against pediatric solid tumors and brain tumors. Clin. Cancer Res. 2019; 25: 2560-2574.
- Watanabe K, et al. Target antigen density governs the efficacy of anti-CD20- CD28-CD3 ζ chimeric antigen receptor-modified effector CD8+ T cells. J. Immunol. 2015; 194: 911-920.
- Hombach AA, et al. Superior therapeutic index in lymphoma therapy: CD30+ CD34+ hematopoietic stem cells resist a chimeric antigen receptor T-cell attack. Mol. Ther. 2016; 24: 1423-1434.
- Walker AJ, et al. Tumor antigen and receptor densities regulate efficacy of a chimeric antigen receptor targeting anaplastic lymphoma kinase. Mol. Ther. 2017; 25: 2189-2201.
- Majzner RG, et al. Tuning the antigen density requirement for CAR T-cell activity. Cancer Discov. 2020; 10: 702-723.