Share this post on:

Vity of EGF-SubA in U251 cells using this platform. Continuous exposure of U251 cells to 1.0 pM of EGF-SubA, which represents a concentration that led to significant cytotoxicity in the clonogenic assay (Fig. 3A), demonstrated a similarly potent anti-tumor activity on the xCELLigence platform (Fig. S3A). In addition, as this assay was performed in real-time, we were able to identify that EGF-SubA induced cytotoxicity began approximately 8 h following exposure, which corresponds to the observed temporal dynamics of GRP78 cleavage presented in Fig. 2B, further supporting its underlying mechanism of action. Interestingly, as opposed to U251 controls, in which surviving cell populations quickly resumed proliferation, U251 cells grown in acidic conditions (pH 6.7) maintained an attenuated repopulation, supporting our previous findings of increased cellular sensitivity to EGF-SubA in acidic conditions. We then SPI 1005 biological activity extended this assay to the GNS cell line G179 and normal human astrocytes. Similar to U251, G179 cells also demonstrated potent cytotoxicity of EGF-SubA (1.0 pM) when 15481974 compared to SubA toxin alone and attenuated repopulation in cells grown in acidic conditions (Fig. S2B). To support the therapeutic potential of this approach, we did similar studies using normal human astrocytes. As shown in Fig. S2C, EGF-SubA (1.0 pM) demonstrated no activity in human astrocytes, which corresponds to our previous findings suggesting higher concentrations of EGF-SubA would be required to induce GRP78 cleavage (Fig. 2A). Lastly, we extended our in vitro findings in vivo using a mouse xenograft model. U251 cells were implanted s.c. into the hind leg of nude mice and randomized to control (PBS) or EGF-SubA (125 ug/kg) delivered s.c. every other day for 3 days. As demonstrated in Fig. 6A, although this approach did not result in any notable tumor regression, a significant growth delay was observed with EGF-SubA (p = 0.0009). In addition, this regimen was well tolerated, demonstrating no significant weight loss in EGF-SubA ML 264 biological activity treated mice (Fig. 6B; p = 0.47). Next, to confirm in vivo target engagement of EGF-SubA and to evaluate for potential normal tissue toxicity of this compound, we performed western blot on tissue lysates 24 h following EGF-SubA treatment. As demonstrated in Fig. 6C, GRP78 was expressed in U251 tumors and in mouse liver. Consistent with in vitro data, EGF-SubA cleaved GRP78 in U251 tumors grown subcutaneously. Normal liver cells express EGFR; therefore as expected, there was modest GRP78 cleavage observed in the mouse liver, although it was not associated with any significant weight loss or activity. This finding is consistent with the previous report that up to 50 decrease in GRP78 expression does not affect physiologically normal organs and tissues, however significantly impedes tumor growth and angiogenesis [23]. Nevertheless this may represent a potential dose-limiting toxicity of this compound. In summary, the UPR is emerging as an important adaptive pathway contributing to malignant glioma survival. Targeting its primary mediator, the chaperone protein GRP78, through specific, proteolytic cleavage with the immunotoxin EGF-SubA represents a novel and promising multi-targeted approach to cancer therapy. Our work confirms the potential of GRP78 to serve as a molecular target in malignant glioma and demonstratesTargeting the UPR in Glioblastoma with EGF-SubApotent tumor specific cytotoxicity of EGF-SubA in a panel of glioblastoma models in.Vity of EGF-SubA in U251 cells using this platform. Continuous exposure of U251 cells to 1.0 pM of EGF-SubA, which represents a concentration that led to significant cytotoxicity in the clonogenic assay (Fig. 3A), demonstrated a similarly potent anti-tumor activity on the xCELLigence platform (Fig. S3A). In addition, as this assay was performed in real-time, we were able to identify that EGF-SubA induced cytotoxicity began approximately 8 h following exposure, which corresponds to the observed temporal dynamics of GRP78 cleavage presented in Fig. 2B, further supporting its underlying mechanism of action. Interestingly, as opposed to U251 controls, in which surviving cell populations quickly resumed proliferation, U251 cells grown in acidic conditions (pH 6.7) maintained an attenuated repopulation, supporting our previous findings of increased cellular sensitivity to EGF-SubA in acidic conditions. We then extended this assay to the GNS cell line G179 and normal human astrocytes. Similar to U251, G179 cells also demonstrated potent cytotoxicity of EGF-SubA (1.0 pM) when 15481974 compared to SubA toxin alone and attenuated repopulation in cells grown in acidic conditions (Fig. S2B). To support the therapeutic potential of this approach, we did similar studies using normal human astrocytes. As shown in Fig. S2C, EGF-SubA (1.0 pM) demonstrated no activity in human astrocytes, which corresponds to our previous findings suggesting higher concentrations of EGF-SubA would be required to induce GRP78 cleavage (Fig. 2A). Lastly, we extended our in vitro findings in vivo using a mouse xenograft model. U251 cells were implanted s.c. into the hind leg of nude mice and randomized to control (PBS) or EGF-SubA (125 ug/kg) delivered s.c. every other day for 3 days. As demonstrated in Fig. 6A, although this approach did not result in any notable tumor regression, a significant growth delay was observed with EGF-SubA (p = 0.0009). In addition, this regimen was well tolerated, demonstrating no significant weight loss in EGF-SubA treated mice (Fig. 6B; p = 0.47). Next, to confirm in vivo target engagement of EGF-SubA and to evaluate for potential normal tissue toxicity of this compound, we performed western blot on tissue lysates 24 h following EGF-SubA treatment. As demonstrated in Fig. 6C, GRP78 was expressed in U251 tumors and in mouse liver. Consistent with in vitro data, EGF-SubA cleaved GRP78 in U251 tumors grown subcutaneously. Normal liver cells express EGFR; therefore as expected, there was modest GRP78 cleavage observed in the mouse liver, although it was not associated with any significant weight loss or activity. This finding is consistent with the previous report that up to 50 decrease in GRP78 expression does not affect physiologically normal organs and tissues, however significantly impedes tumor growth and angiogenesis [23]. Nevertheless this may represent a potential dose-limiting toxicity of this compound. In summary, the UPR is emerging as an important adaptive pathway contributing to malignant glioma survival. Targeting its primary mediator, the chaperone protein GRP78, through specific, proteolytic cleavage with the immunotoxin EGF-SubA represents a novel and promising multi-targeted approach to cancer therapy. Our work confirms the potential of GRP78 to serve as a molecular target in malignant glioma and demonstratesTargeting the UPR in Glioblastoma with EGF-SubApotent tumor specific cytotoxicity of EGF-SubA in a panel of glioblastoma models in.

Share this post on: