Abstract

Cancer immunotherapy including adoptive T cell therapy (ACT) is widely used in the clinic and is highly beneficial for patients with hematological malignancies; however, it remains a challenge to develop effective immunotherapy strategies for the treatment of solid cancers, due to the inefficiency of the immune response and the immunosuppressive tumor microenvironment (TME). Immunogenic cell death (ICD) converts dying cancer cells into a therapeutic vaccine and stimulate a systemic antigen-specific antitumor immune response, which can effectively subvert the immunosuppressive TME and enhance the efficiency of immune responses, relative to conventional immunotherapeutic regimens. However, the application of traditional inducers of ICD in anti-cancer immunotherapy has been limited because of low levels of ICD induction and a lack of tumor-targeting accumulation. Mitochondria are important for tumor-targeting strategies and have emerged as organelles with key roles in the immune system. We hypothesized that the alteration of mitochondria in cancer cells could be an important target for the development of an efficient ICD inducer for use in cancer immunotherapy. Here, we report the evaluation of a mitochondria-targeted small molecule, IR-780, that acts as an ICD inducer and exhibits exceptional antineoplastic activity. IR-780 specifically accumulated in tumor cells to elicit ICD in vitro and in vivo, effectively suppressed tumor growth and lung metastasis, and enhanced adoptive T-cell therapy effects against solid tumors in mouse models. These anticancer effects were linked to dendritic cell maturation and synergistic effector T cell priming and infiltration into tumors. The underlying mechanism involves the direct targeting of the mitochondria by IR-780, to destroy cancer cells, including drug-resistant cancer cells, leading to the full exposure of tumor-associated antigens (TAAs), thereby enhancing antigen-specific antitumor immune responses. These features of IR-780 suggest that it has the advantage of leading to complete TAA exposure and the stimulation of efficient antitumor immune responses in the TME. IR-780 has potential for use as a preparative ICD inducer, in combination with conventional immunostimulatory regimens for cancer immunotherapy, particularly in the context of solid tumor treatment.

Highlights

  • In the last decade, immunotherapy has emerged as the most promising cancer treatment strategy, alongside chemotherapy, radiation therapy, and surgery [1]

  • We describe the assessment of a mitochondria-targeting small molecule, IR780, for use as an Immunogenic cell death (ICD) inducer that can effectively suppress tumor growth and lung metastasis of colorectal cancer (CRC) in a mouse model

  • All these results indicate that IR-780 may act FIGURE 3 | was measured by detecting the percentage of (E) CD4+CD69+ and (F) CD8+ CD69+ T cells populations. (G) The activation of CD80+CD86+ DCs was analyzed by flow cytometry. (H) Schedule representation of different treatment-mediated antitumor vaccination effects in CT26 tumor models. (I) After subcutaneous injection of different treated CT26 cells (10 μM IR-780, 1 μM MTX, and 20 μM CIS, respectively) in the left flank of mice, the tumor volumes in the right flank of each group mice were detected and compared. (J) The frequency of tumor-free mice in each group after vaccinated with different pretreated CT26 cells (n = 10)

Read more

Summary

Introduction

Immunotherapy has emerged as the most promising cancer treatment strategy, alongside chemotherapy, radiation therapy, and surgery [1]. CAR T therapy in particular has been widely used in the clinic and is highly beneficial for patients with B cell malignancies [6]; it remains a challenge to develop effective ACT strategies for the treatment of solid cancers, due to the inefficiency of effector T cell activation and the immunosuppressive tumor microenvironment (TME), resulting from chronic, but suboptimal exposure to tumor-associated antigens (TAAs) [7]. There has been considerable interest in the development of complementary approaches, including therapeutic chemotherapy, which can fully expose TAAs and stimulate an antitumor microenvironment [8], to induce increased and prolonged adoptive T cell responses at tumor sites, exerting strong antitumor immunity against solidtumors [9]

Methods
Results
Conclusion
Full Text
Paper version not known

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.