Your browser doesn't support javascript.
loading
Cancer vaccine formulation dictates synergy with CTLA-4 and PD-L1 checkpoint blockade therapy.
Hailemichael, Yared; Woods, Amber; Fu, Tihui; He, Qiuming; Nielsen, Michael C; Hasan, Farah; Roszik, Jason; Xiao, Zhilan; Vianden, Christina; Khong, Hiep; Singh, Manisha; Sharma, Meenu; Faak, Faisal; Moore, Derek; Dai, Zhimin; Anthony, Scott M; Schluns, Kimberly S; Sharma, Padmanee; Engelhard, Victor H; Overwijk, Willem W.
Afiliación
  • Hailemichael Y; Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
  • Woods A; Department of Microbiology, Immunology, and Cancer Biology, Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA.
  • Fu T; Department of Genitourinary Medical Oncology/Immunology and.
  • He Q; Department of Genitourinary Medical Oncology/Immunology and.
  • Nielsen MC; Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
  • Hasan F; Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
  • Roszik J; Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
  • Xiao Z; Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
  • Vianden C; Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
  • Khong H; Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
  • Singh M; Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
  • Sharma M; Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
  • Faak F; Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
  • Moore D; Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
  • Dai Z; Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
  • Anthony SM; Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
  • Schluns KS; Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
  • Sharma P; The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas, USA.
  • Engelhard VH; Department of Genitourinary Medical Oncology/Immunology and.
  • Overwijk WW; Department of Microbiology, Immunology, and Cancer Biology, Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA.
J Clin Invest ; 128(4): 1338-1354, 2018 04 02.
Article en En | MEDLINE | ID: mdl-29480817
ABSTRACT
Anticancer vaccination is a promising approach to increase the efficacy of cytotoxic T lymphocyte-associated protein 4 (CTLA-4) and programmed death ligand 1 (PD-L1) checkpoint blockade therapies. However, the landmark FDA registration trial for anti-CTLA-4 therapy (ipilimumab) revealed a complete lack of benefit of adding vaccination with gp100 peptide formulated in incomplete Freund's adjuvant (IFA). Here, using a mouse model of melanoma, we found that gp100 vaccination induced gp100-specific effector T cells (Teffs), which dominantly forced trafficking of anti-CTLA-4-induced, non-gp100-specific Teffs away from the tumor, reducing tumor control. The inflamed vaccination site subsequently also sequestered and destroyed anti-CTLA-4-induced Teffs with specificities for tumor antigens other than gp100, reducing the antitumor efficacy of anti-CTLA-4 therapy. Mechanistically, Teffs at the vaccination site recruited inflammatory monocytes, which in turn attracted additional Teffs in a vicious cycle mediated by IFN-γ, CXCR3, ICAM-1, and CCL2, dependent on IFA formulation. In contrast, nonpersistent vaccine formulations based on dendritic cells, viral vectors, or water-soluble peptides potently synergized with checkpoint blockade of both CTLA-4 and PD-L1 and induced complete tumor regression, including in settings of primary resistance to dual checkpoint blockade. We conclude that cancer vaccine formulation can dominantly determine synergy, or lack thereof, with CTLA-4 and PD-L1 checkpoint blockade therapy for cancer.
Asunto(s)
Palabras clave

Texto completo: 1 Colección: 01-internacional Banco de datos: MEDLINE Asunto principal: Péptidos / Vacunas contra el Cáncer / Antígeno gp100 del Melanoma / Antígeno B7-H1 / Antígeno CTLA-4 / Puntos de Control del Ciclo Celular / Melanoma / Neoplasias Experimentales Tipo de estudio: Prognostic_studies Límite: Animals Idioma: En Revista: J Clin Invest Año: 2018 Tipo del documento: Article País de afiliación: Estados Unidos

Texto completo: 1 Colección: 01-internacional Banco de datos: MEDLINE Asunto principal: Péptidos / Vacunas contra el Cáncer / Antígeno gp100 del Melanoma / Antígeno B7-H1 / Antígeno CTLA-4 / Puntos de Control del Ciclo Celular / Melanoma / Neoplasias Experimentales Tipo de estudio: Prognostic_studies Límite: Animals Idioma: En Revista: J Clin Invest Año: 2018 Tipo del documento: Article País de afiliación: Estados Unidos