Your browser doesn't support javascript.
loading
Adaptive antitumor immune response stimulated by bio-nanoparticle based vaccine and checkpoint blockade.
Bai, Xuewei; Zhou, Yanmei; Yokota, Yuki; Matsumoto, Yoshihiro; Zhai, Bo; Maarouf, Nader; Hayashi, Hikaru; Carlson, Rolf; Zhang, Songhua; Sousa, Aryanna; Sun, Bei; Ghanbari, Hossein; Dong, Xiaoqun; Wands, Jack R.
Afiliación
  • Bai X; Liver Research Center, Rhode Island Hospital, Department of Medicine, The Warren Alpert Medical School of Brown University, RI, 02903, Providence, USA.
  • Zhou Y; Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 150081, Heilongjiang Province, People's Republic of China.
  • Yokota Y; Liver Research Center, Rhode Island Hospital, Department of Medicine, The Warren Alpert Medical School of Brown University, RI, 02903, Providence, USA.
  • Matsumoto Y; Department of Anesthesiology, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, 150081, Heilongjiang Province, People's Republic of China.
  • Zhai B; Liver Research Center, Rhode Island Hospital, Department of Medicine, The Warren Alpert Medical School of Brown University, RI, 02903, Providence, USA.
  • Maarouf N; Liver Research Center, Rhode Island Hospital, Department of Medicine, The Warren Alpert Medical School of Brown University, RI, 02903, Providence, USA.
  • Hayashi H; Liver Research Center, Rhode Island Hospital, Department of Medicine, The Warren Alpert Medical School of Brown University, RI, 02903, Providence, USA.
  • Carlson R; Department of Surgical Oncology and Hepatobiliary Surgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China.
  • Zhang S; Liver Research Center, Rhode Island Hospital, Department of Medicine, The Warren Alpert Medical School of Brown University, RI, 02903, Providence, USA.
  • Sousa A; Liver Research Center, Rhode Island Hospital, Department of Medicine, The Warren Alpert Medical School of Brown University, RI, 02903, Providence, USA.
  • Sun B; Liver Research Center, Rhode Island Hospital, Department of Medicine, The Warren Alpert Medical School of Brown University, RI, 02903, Providence, USA.
  • Ghanbari H; Liver Research Center, Rhode Island Hospital, Department of Medicine, The Warren Alpert Medical School of Brown University, RI, 02903, Providence, USA.
  • Dong X; Liver Research Center, Rhode Island Hospital, Department of Medicine, The Warren Alpert Medical School of Brown University, RI, 02903, Providence, USA.
  • Wands JR; Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 150081, Heilongjiang Province, People's Republic of China.
J Exp Clin Cancer Res ; 41(1): 132, 2022 Apr 08.
Article en En | MEDLINE | ID: mdl-35392977
ABSTRACT

BACKGROUND:

Interactions between tumor and microenvironment determine individual response to immunotherapy. Triple negative breast cancer (TNBC) and hepatocellular carcinoma (HCC) have exhibited suboptimal responses to immune checkpoint inhibitors (ICIs). Aspartate ß-hydroxylase (ASPH), an oncofetal protein and tumor associated antigen (TAA), is a potential target for immunotherapy.

METHODS:

Subcutaneous HCC and orthotopic TNBC murine models were established in immunocompetent BALB/c mice with injection of BNL-T3 and 4 T1 cells, respectively. Immunohistochemistry, immunofluorescence, H&E, flow cytometry, ELISA and in vitro cytotoxicity assays were performed.

RESULTS:

The ASPH-MYC signaling cascade upregulates PD-L1 expression on breast and liver tumor cells. A bio-nanoparticle based λ phage vaccine targeting ASPH was administrated to mice harboring syngeneic HCC or TNBC tumors, either alone or in combination with PD-1 blockade. In control, autocrine chemokine ligand 13 (CXCL13)-C-X-C chemokine receptor type 5 (CXCR5) axis promoted tumor development and progression in HCC and TNBC. Interactions between PD-L1+ cancer cells and PD-1+ T cells resulted in T cell exhaustion and apoptosis, causing immune evasion of cancer cells. In contrast, combination therapy (Vaccine+PD-1 inhibitor) significantly suppressed primary hepatic or mammary tumor growth (with distant pulmonary metastases in TNBC). Adaptive immune responses were attributed to expansion of activated CD4+ T helper type 1 (Th1)/CD8+ cytotoxic T cells (CTLs) that displayed enhanced effector functions, and maturation of plasma cells that secreted high titers of ASPH-specific antibody. Combination therapy significantly reduced tumor infiltration of immunosuppressive CD4+/CD25+/FOXP3+ Tregs. When the PD-1/PD-L1 signal was inhibited, CXCL13 produced by ASPH+ cancer cells recruited CXCR5+/CD8+ T lymphocytes to tertiary lymphoid structures (TLSs), comprising effector and memory CTLs, T follicular helper cells, B cell germinal center, and follicular dendritic cells. TLSs facilitate activation and maturation of DCs and actively recruit immune subsets to tumor microenvironment. These CTLs secreted CXCL13 to recruit more CXCR5+ immune cells and to lyse CXCR5+ cancer cells. Upon combination treatment, formation of TLSs predicts sensitivity to ICI blockade. Combination therapy substantially prolonged overall survival of mice with HCC or TNBC.

CONCLUSIONS:

Synergistic antitumor efficacy attributable to a λ phage vaccine specifically targeting ASPH, an ideal TAA, combined with ICIs, inhibits tumor growth and progression of TNBC and HCC.
Asunto(s)
Palabras clave

Texto completo: 1 Colección: 01-internacional Base de datos: MEDLINE Asunto principal: Carcinoma Hepatocelular / Vacunas contra el Cáncer / Neoplasias de la Mama Triple Negativas / Inmunoterapia / Neoplasias Hepáticas Tipo de estudio: Prognostic_studies Límite: Animals / Humans Idioma: En Revista: J Exp Clin Cancer Res Año: 2022 Tipo del documento: Article País de afiliación: Estados Unidos

Texto completo: 1 Colección: 01-internacional Base de datos: MEDLINE Asunto principal: Carcinoma Hepatocelular / Vacunas contra el Cáncer / Neoplasias de la Mama Triple Negativas / Inmunoterapia / Neoplasias Hepáticas Tipo de estudio: Prognostic_studies Límite: Animals / Humans Idioma: En Revista: J Exp Clin Cancer Res Año: 2022 Tipo del documento: Article País de afiliación: Estados Unidos