Figure 1
Figure 1. Vaccination with tumor cells secreting the heat shock protein fusion gp96-Ig led to the activation, expansion, and functional competence of tumor-reactive CD8+ T lymphocytes in syngeneic HSCT recipients. Conditioned (9.5 Gy) B6 mice were transplanted with 5.0 × 106 B6-CD45.1+ TCD-BM cells and adoptively transferred with 1.0 × 106 CD8+ T cells specific for OVA257-264 (OT-I) 5 days later. After a 2-day resting period, recipients were vaccinated intraperitoneally with irradiated (120 Gy) EL-4 lymphoma cells expressing OVA (E.G7) engineered to secrete gp96-Ig (E.G7-gp96-Ig). (A) Macrophages (F4/80hi CD11bhi Gr-1lo) and inflammatory monocytes (Gr-1hi CD11bmid F4/80lo) infiltrated the peritoneal cavity 2 days following vaccination with gp96-Ig-secreting tumor cells; n = 9 from a pool of 3 experiments. (B) Tumor-reactive CD8+ T lymphocytes (OT-I: CD8+ CD45.1− Vα2+ Vβ5+) expanded in the peritoneal cavity 5 days following vaccination with tumor cells secreting gp96-Ig; n = 8 from a pool of 3 experiments. (C) Tumor-reactive CD8+ T cells accumulated in secondary lymphoid organs 5 days following a second vaccination (13 days after HSCT) with gp96-Ig-secreting tumor cells; n = 3. (D) Vaccination with gp96-Ig-secreting tumor cells up to 2 weeks after HSCT resulted in expansion of cotransplanted tumor-reactive CD8+ T cells 5 days later; n = 3 to 8 from 3 independent experiments. (E) Tumor-reactive CD8+ T lymphocytes transitioned from central-memory (TCM, CD62L+ CD44+) to effector cells (Teff, CD62L− CD44+) following vaccination with tumor cells secreting gp96-Ig: (left) representative dot plots; (right) n = 5 to 6 from a pool of 2 experiments. (F) Vaccination with gp96-Ig-secreting tumor cells induced highly IFN-γ+ tumor-reactive CD8+ T cells: (left) representative dot plots; (right) n = 4 from a representative of 3 experiments.

Vaccination with tumor cells secreting the heat shock protein fusion gp96-Ig led to the activation, expansion, and functional competence of tumor-reactive CD8+ T lymphocytes in syngeneic HSCT recipients. Conditioned (9.5 Gy) B6 mice were transplanted with 5.0 × 106 B6-CD45.1+ TCD-BM cells and adoptively transferred with 1.0 × 106 CD8+ T cells specific for OVA257-264 (OT-I) 5 days later. After a 2-day resting period, recipients were vaccinated intraperitoneally with irradiated (120 Gy) EL-4 lymphoma cells expressing OVA (E.G7) engineered to secrete gp96-Ig (E.G7-gp96-Ig). (A) Macrophages (F4/80hi CD11bhi Gr-1lo) and inflammatory monocytes (Gr-1hi CD11bmid F4/80lo) infiltrated the peritoneal cavity 2 days following vaccination with gp96-Ig-secreting tumor cells; n = 9 from a pool of 3 experiments. (B) Tumor-reactive CD8+ T lymphocytes (OT-I: CD8+ CD45.1 Vα2+ Vβ5+) expanded in the peritoneal cavity 5 days following vaccination with tumor cells secreting gp96-Ig; n = 8 from a pool of 3 experiments. (C) Tumor-reactive CD8+ T cells accumulated in secondary lymphoid organs 5 days following a second vaccination (13 days after HSCT) with gp96-Ig-secreting tumor cells; n = 3. (D) Vaccination with gp96-Ig-secreting tumor cells up to 2 weeks after HSCT resulted in expansion of cotransplanted tumor-reactive CD8+ T cells 5 days later; n = 3 to 8 from 3 independent experiments. (E) Tumor-reactive CD8+ T lymphocytes transitioned from central-memory (TCM, CD62L+ CD44+) to effector cells (Teff, CD62L CD44+) following vaccination with tumor cells secreting gp96-Ig: (left) representative dot plots; (right) n = 5 to 6 from a pool of 2 experiments. (F) Vaccination with gp96-Ig-secreting tumor cells induced highly IFN-γ+ tumor-reactive CD8+ T cells: (left) representative dot plots; (right) n = 4 from a representative of 3 experiments.

Close Modal

or Create an Account

Close Modal
Close Modal