دورية أكاديمية

Adaptive antitumor immune response stimulated by bio-nanoparticle based vaccine and checkpoint blockade

التفاصيل البيبلوغرافية
العنوان: Adaptive antitumor immune response stimulated by bio-nanoparticle based vaccine and checkpoint blockade
المؤلفون: Xuewei Bai, Yanmei Zhou, Yuki Yokota, Yoshihiro Matsumoto, Bo Zhai, Nader Maarouf, Hikaru Hayashi, Rolf Carlson, Songhua Zhang, Aryanna Sousa, Bei Sun, Hossein Ghanbari, Xiaoqun Dong, Jack R. Wands
المصدر: Journal of Experimental & Clinical Cancer Research, Vol 41, Iss 1, Pp 1-18 (2022)
بيانات النشر: BMC, 2022.
سنة النشر: 2022
المجموعة: LCC:Neoplasms. Tumors. Oncology. Including cancer and carcinogens
مصطلحات موضوعية: Aspartate β-hydroxylase (ASPH), Immune checkpoint inhibitor, Lambda phage vaccine, Triple negative breast cancer, Hepatocellular carcinoma, Metastasis, Neoplasms. Tumors. Oncology. Including cancer and carcinogens, RC254-282
الوصف: 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 β-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.
نوع الوثيقة: article
وصف الملف: electronic resource
اللغة: English
تدمد: 1756-9966
العلاقة: https://doaj.org/toc/1756-9966Test
DOI: 10.1186/s13046-022-02307-3
الوصول الحر: https://doaj.org/article/5901491e7a3b4696abc0ff8e9e8b7df4Test
رقم الانضمام: edsdoj.5901491e7a3b4696abc0ff8e9e8b7df4
قاعدة البيانات: Directory of Open Access Journals
الوصف
تدمد:17569966
DOI:10.1186/s13046-022-02307-3