characterization of a mc38 mouse syngeneic tumor model ... · characterization of a mc38 mouse...

1
Characterization of a MC38 Mouse Syngeneic Tumor Model Expressing Human PD-L1 in the Transgenic C57BL/6J Mouse System Expressing Human PD-1 and PD-L1 Michael Koratich, Ted Green, Jay Liu, Charlotte Hammond, LaJuana Durbin, Anna Chen*, Jerry Zhou*, and Murray Stackhouse Southern Research Institute, Birmingham, AL; *Nanjing Galaxy Biopharma, Nanjing, China The human checkpoint targets PD-1 and PDL-1 continue to demonstrate great promise in the clinic. With much of the current focus turning toward combination regimens, an animal model using the human clinical antibodies will be beneficial for evaluating new combination strategies. Here we continue characterization of a C57BL/6 transgenic mouse model expressing both the human PD-1 and PD-L1 combined with a modified murine MC38 colon tumor cell line expressing human PD-L1. PD-1 expression of the transgenic mice were verified in ex vivo stimulated splenocytes by flow cytometry analysis. Expression of PD-L1 on the genetically modified MC38 cells was also demonstrated by flow cytometry. For in vivo efficacy evaluation, tumor-implanted mice were treated with the clinical agents nivolumab and pembrolizumab at 100 μg and atezolizumab at 1 mg on Days 3, 7, 10, and 14 post implant. Treatment with nivolumab and pembrolizumab initiated tumor regression by Day 17. Complete tumor regressions were seen at Day 28 in nivolumab, 62.5% complete regression, and pembrolizumab, 71.4% complete regression. Growth inhibition was 83.9%, 69.7%, and 95.0% on Day 28 for nivolumab, atezolizumab, and pembrolizumab, respectively, compared to the control animals. There was no significant body weight loss and no signs of toxicity in any of the treated animals. To compare the human anti-PD-1 and PD-L1 clinical agent specificity, the non-transgenic parent C57BL/6 mouse strain was implanted with the unmodified MC38 colon tumor cells. Treatment with pembrolizumab and atezolizumab was conducted as with the transgenic animals. Through 28 days no growth inhibition, tumor size 105.9% of control, was seen in the pembrolizumab treated group, demonstrating lack of cross reactivity of the human therapeutic in the standard mouse model. Atezolizumab did demonstrate a 56.9% growth inhibition compared to controls and is consistent with the known cross reactivity of atezolizumab between human and mouse. We have shown a genetically modified MC38 colon tumor expressing human PD-L1 in transgenic mice expressing both human PD-1 and PD-L1 to be a suitable model for checkpoint inhibitor evaluation of the human form of the particular checkpoint therapeutic. Further research will involve combining each of the checkpoint antibodies with various chemotherapeutic agents. Figure 1 Summary 2000 Ninth Avenue South ● Birmingham, AL 35205 ● www.SouthernResearch.org ● 1-800-967-6774 (USA) ● 1-205-581-2000 We have shown that MC38.huPD-L1 cells in transgenic C57BL/6J huPD- 1/PD-L1 mice respond to treatment with the human clinical agents atezolizumab, pembrolizumab, and nivolumab. Importantly, the genetically modified MC38 tumor and transgenic mouse model expressing human checkpoint genes allows direct evaluation of human checkpoint inhibitor therapies without testing the murine analogs. The wild-type C57BL/6 with wild-type MC-38 tumor may be a suitable model for anti-PD-L1 therapeutics, but not for PD-1. Abstract #1504 Human PD-L1 Expression in MC-38.WT versus MC-38.huPD-L1 transfected cells Genetically modified MC-38 cells stably express human PD-L1. Homology of mouse and human PD-L1 allows for activity of atezolizumab in the wild-type syngeneic MC-38 model. Lack of mouse/human homology for PD-1 requires use of transgenic model system to evaluate human therapeutics. Nivolumab, atezolizumab, and pembrolizumab demonstrated growth inhibition of MC-38.huPD-L1 tumor in the huPD-1/L1 transgenic C57BL/6J mouse model. Nivolumab and pembrolizumab demonstrated complete tumor regression of MC-38.huPD-L1 tumor in the huPD-1/L1 transgenic C57BL/6J mouse model. MC-38.WT MC-38.huPD-L1 Figure 2 Figure 3 Atezolizumab and Pembrolizumab Efficacy in Wild-Type C57BL/6:MC-38 Model System Atezolizumab, Pembrolizumab, & Nivolumab Efficacy in huPD-1/L1 Transgenic C57BL6J Mouse model with MC-38 Tumor Expressing huPD-L1 Control Pembrolizumab Atezolizumab Nivolumab Conclusions Homology of mouse & human PD-L1 allows for atezolizumab activity Transgenic huPD-1/L1 C57BL/6J mice implanted with MC-38.huPD-L1 cells and dosed with clinical agents atezolizumab (1 mg), pembrolizumab (100 μg), or nivolumab (100 μg), on Days 3, 7, 10, & 14. Tumor measurements collected twice weekly. Individual animal data presented. Wild-type MC-38 mouse colon tumor cells and MC-38.huPD-L1 cells were stained with anti-human APC anti-human CD274 (B7-H1, PD-L1), Biolegend (Cat 124311) and analyzed for expression of human PD-L1. Wild-type C57BL/6 mice were implanted with wild-type MC-38 mouse colon tumor cells and dosed with clinical agents atezolizumab (1 mg) or pembrolizumab (100 μg) on Days 3, 7, 10, & 14. Tumor measurements collected twice weekly. 62.5% Complete Regression 83.9% Growth Inhibition 71.4% Complete Regression 95.0% Growth Inhibition 69.7% Growth Inhibition

Upload: others

Post on 16-Oct-2020

14 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Characterization of a MC38 Mouse Syngeneic Tumor Model ... · Characterization of a MC38 Mouse Syngeneic Tumor Model Expressing Human PD-L1 in the ... PD-L1), Biolegend (Cat 124311)

Characterization of a MC38 Mouse Syngeneic Tumor Model Expressing Human PD-L1 in the Transgenic C57BL/6J Mouse System Expressing Human PD-1 and PD-L1

Michael Koratich, Ted Green, Jay Liu, Charlotte Hammond, LaJuana Durbin, Anna Chen*, Jerry Zhou*, and Murray StackhouseSouthern Research Institute, Birmingham, AL; *Nanjing Galaxy Biopharma, Nanjing, China

The human checkpoint targets PD-1 and PDL-1 continue to demonstrate great promise in the clinic. With much of the current focusturning toward combination regimens, an animal model using the human clinical antibodies will be beneficial for evaluating newcombination strategies. Here we continue characterization of a C57BL/6 transgenic mouse model expressing both the human PD-1 andPD-L1 combined with a modified murine MC38 colon tumor cell line expressing human PD-L1. PD-1 expression of the transgenic micewere verified in ex vivo stimulated splenocytes by flow cytometry analysis. Expression of PD-L1 on the genetically modified MC38 cellswas also demonstrated by flow cytometry. For in vivo efficacy evaluation, tumor-implanted mice were treated with the clinical agentsnivolumab and pembrolizumab at 100 µg and atezolizumab at 1 mg on Days 3, 7, 10, and 14 post implant. Treatment with nivolumab andpembrolizumab initiated tumor regression by Day 17. Complete tumor regressions were seen at Day 28 in nivolumab, 62.5% completeregression, and pembrolizumab, 71.4% complete regression. Growth inhibition was 83.9%, 69.7%, and 95.0% on Day 28 for nivolumab,atezolizumab, and pembrolizumab, respectively, compared to the control animals. There was no significant body weight loss and no signsof toxicity in any of the treated animals. To compare the human anti-PD-1 and PD-L1 clinical agent specificity, the non-transgenic parentC57BL/6 mouse strain was implanted with the unmodified MC38 colon tumor cells. Treatment with pembrolizumab and atezolizumabwas conducted as with the transgenic animals. Through 28 days no growth inhibition, tumor size 105.9% of control, was seen in thepembrolizumab treated group, demonstrating lack of cross reactivity of the human therapeutic in the standard mouse model.Atezolizumab did demonstrate a 56.9% growth inhibition compared to controls and is consistent with the known cross reactivity ofatezolizumab between human and mouse. We have shown a genetically modified MC38 colon tumor expressing human PD-L1 intransgenic mice expressing both human PD-1 and PD-L1 to be a suitable model for checkpoint inhibitor evaluation of the human form ofthe particular checkpoint therapeutic. Further research will involve combining each of the checkpoint antibodies with variouschemotherapeutic agents.

Figure 1

Summary

2000 Ninth Avenue South ● Birmingham, AL 35205 ● www.SouthernResearch.org ● 1-800-967-6774 (USA) ● 1-205-581-2000

• We have shown that MC38.huPD-L1cells in transgenic C57BL/6J huPD-1/PD-L1 mice respond to treatmentwith the human clinical agentsatezolizumab, pembrolizumab, andnivolumab.

• Importantly, the genetically modifiedMC38 tumor and transgenic mousemodel expressing human checkpointgenes allows direct evaluation ofhuman checkpoint inhibitor therapieswithout testing the murine analogs.

• The wild-type C57BL/6 with wild-typeMC-38 tumor may be a suitable modelfor anti-PD-L1 therapeutics, but not forPD-1.

Abstract #1504

Human PD-L1 Expression in MC-38.WT versus

MC-38.huPD-L1 transfected cells

• Genetically modified MC-38 cells stably expresshuman PD-L1.

• Homology of mouse and human PD-L1 allows foractivity of atezolizumab in the wild-type syngeneicMC-38 model.

• Lack of mouse/human homology for PD-1 requiresuse of transgenic model system to evaluate humantherapeutics.

• Nivolumab, atezolizumab, and pembrolizumabdemonstrated growth inhibition of MC-38.huPD-L1tumor in the huPD-1/L1 transgenic C57BL/6J mousemodel.

• Nivolumab and pembrolizumab demonstratedcomplete tumor regression of MC-38.huPD-L1tumor in the huPD-1/L1 transgenic C57BL/6J mousemodel.

MC-38.WT

MC-38.huPD-L1

Figure 2

Figure 3

Atezolizumab and Pembrolizumab Efficacy in

Wild-Type C57BL/6:MC-38 Model System

Atezolizumab, Pembrolizumab, & Nivolumab Efficacy in

huPD-1/L1 Transgenic C57BL6J Mouse model with MC-38 Tumor Expressing huPD-L1

Control Pembrolizumab

Atezolizumab Nivolumab

Conclusions

Homology of mouse & human PD-L1 allows for atezolizumab activity

Transgenic huPD-1/L1 C57BL/6J mice implanted with MC-38.huPD-L1 cells and dosed with clinical agents atezolizumab (1 mg), pembrolizumab(100 µg), or nivolumab (100 µg), on Days 3, 7, 10, & 14. Tumor measurements collected twice weekly. Individual animal data presented.

Wild-type MC-38 mouse colon tumor cells and MC-38.huPD-L1cells were stained with anti-human APC anti-human CD274(B7-H1, PD-L1), Biolegend (Cat 124311) and analyzed forexpression of human PD-L1.

Wild-type C57BL/6 mice were implanted with wild-type MC-38mouse colon tumor cells and dosed with clinical agentsatezolizumab (1 mg) or pembrolizumab (100 µg) on Days 3, 7,10, & 14. Tumor measurements collected twice weekly.

62.5% Complete Regression83.9% Growth Inhibition

71.4% Complete Regression95.0% Growth Inhibition

69.7% Growth Inhibition