Tumor microenvironment regulation - enhanced radio - immunotherapy

https://doi.org/10.1016/j.bioadv.2022.212867Get rights and content

Highlights

  • SM NPs regulated tumor microenvironment through oxygenation, ROS generation, GSH depletion, and immune activation.

  • SM NPs enhanced the efficacy of radio-immunotherapy through inhibiting both primary tumors and distant untreated tumors.

  • The combination of SM NPs and the anti-CTLA-4 antibody enhanced the efficacy of RT at a low dose of radiation.

Abstract

Radiotherapy (RT) is frequently utilized for cancer treatment in clinical practice and has been proved to have immune stimulation potency in recent years. However, its inhibitory effect on tumor growth, especially on tumor metastasis, is still limited by many factors, including the complex tumor microenvironment (TME). Therefore, the TME - regulating SiO2@MnO2 nanoparticles (SM NPs) were prepared and applied to the combination of RT and immunotherapy. In a bilateral animal model, SM NPs not only enhanced the inhibitory effect of RT on primary tumor growth, but also strengthened the abscopal effect to inhibit the growth of distant untreated tumors. As for the distant untreated tumor, 40% of mice showed complete inhibition of tumor growth and 40% showed a suppressed tumor growth. Moreover, SM NPs showed modulation functions for TME through inducing the increase in intracellular levels of oxygen and reactive oxygen species after their reaction with hydrogen peroxide and the main antioxidative agent glutathione in TME. Lastly, SM NPs also effectively induced the increase in the amounts of cytokines secreted by macrophage - like cells, indicating modulation functions for immune responses. This work highlighted a potential strategy of simultaneously inhibiting tumor growth and metastasis through the regulation of TME and immune responses by SM NPs - enhanced radio - immunotherapy.

Introduction

RT is one of the vital cancer treatment strategies in clinical practice, and one of the major radio-toxic effects of RT is DNA damage [1], [2]. In recent years, the abscopal effect of RT has been proved to stimulate immune responses, but it is seen in only 10% of cases in clinical practice [3]. Therefore, the inhibitory effect of RT on tumor metastasis is still far from satisfactory [4], [5]. Immunotherapy has emerged in recent years with promising clinical applications owing to its modulation functions for immune responses [6]. Interestingly, compared with RT alone, the abscopal effect of RT can become more common when combined with immunotherapy both in preclinical and clinical models of programmed cell death 1 (PD - 1) and cytotoxic T lymphocyte antigen 4 (CTLA - 4) inhibitors [7], [8]. However, owing to the host mechanism to remain immunologically silent after cell death caused by DNA damage, the DNA damage generated will be inactivated through many mechanisms, including cancer cell autophagy [9], apoptotic caspase activation during apoptosis [10], and digestion by host deoxyribonuclease (DNase) [10], [11]. Moreover, the efficacy of RT and immunotherapy is limited by the complex TME, which is usually characterized by hypoxia, acidic pH value [12], elevated hydrogen peroxide (H2O2) levels [13], and powerful antioxidative systems [14]. Hypoxia of TME contributes to tumor recurrence and poor prognosis in RT [15] and limits the efficacy of RT through the support of the development of cancer stem cells and inhibition of the radical generation [16], [17]. Specifically, hypoxic cells require a threefold higher dose of radiation than normoxic cells [18]. Moreover, a hypoxic TME can suppress anticancer immune responses through the promotion of the infiltration and accumulation of suppressor T - cells and the inhibition of the adaptive immune system, resulting in tumor angiogenesis and cancer metastasis [19], [20]. Moreover, glutathione (GSH), one of the main reducing agents of antioxidative systems in cancer cells, can scavenge reactive oxygen species (ROS), which decreases the efficacy of ROS - based therapies and promotes the expression of glutathione peroxidase 4 (GPX - 4), which further enhances the antioxidation barrier in TME, thus strongly suppressing the efficacy of RT [14]. These findings emphasize the fact that regulating TME can be a promising strategy to enhance the synergistic efficacy of radio-immunotherapy.

In recent years, TME - responsive anticancer therapies have been widely studied, which modulate TME through the unique physical and chemical properties of nanomaterials [21], [22]. They have been proved effective in improving treatment outcomes of various anticancer therapies [23], [24]. For example, H2O2, an ideal prodrug for Fenton reactions or Fenton-like reactions in chemodynamic therapy (CDT), can react with various nanomaterials to produce highly toxic hydroxyl radicals (·OH), including manganese (Mn) [25], [26], [27]. Recently, Mn-based nanomaterials have attracted tremendous interest in the field of anticancer therapies. Mn plays an important role in many in vivo processes, such as neuronal function, immune regulation, antioxidant defenses, and the biosynthesis of some metalloenzymes [28], [29]. Studies have shown that Mn2+ can be excreted rapidly by the kidneys leading to fewer side effects on the body [30]. Moreover, its unique properties in regulating TME make Mn - based nanomaterials potentially applicable to anticancer therapies. Typically, Mn - based nanomaterials can induce increase in oxygen (O2) and ROS levels through reactions with H2O2 and GSH in TME, thus enhancing the efficacy of many anticancer therapies [31], [32], [33]. Notably, Mn2+ has been proved by recent studies to have immune stimulation potential [34], [35], [36], [37], [38]. It has been proved that Mn2+ is critical in the innate immune sensing of tumors and enhancing adaptive immune responses, which can promote the sensitivity of the DNA sensor of the cyclic GMP - AMP synthase (cGAS) and the stimulator of interferon genes (STING) [34], [35]. Interestingly, RT - induced DNA damage has also been proved to activate the cGAS / STING pathway and enhance immune responses [39], [40]. Therefore, Mn - based nanomaterials may be promising in enhancing the immune responses and the abscopal effect of RT. Except for Mn-based nanomaterials, various inorganic nanomaterials have been proved to stimulate immune responses [41], especially silica. In the previous studies, silica-based nanomaterials have shown significant immunogenicity, which can promote antigen presentation, cytokine secretion, CD4+ and CD8+ T cell proliferation, and effector memory T cell population [42], [43], [44], [45], [46], [47], [48], [49]. Therefore, it is considered that combining different nanomaterials with immune stimulation capacity may maximize the immune responses in immunotherapy. However, the applications of Si - Mn - based NPs to radiotherapy to synergistically inhibit both tumor growth and tumor recurrence and metastasis through the regulation of TME and immune responses are still rare. Whereas, tumor recurrence and metastasis are still challenging for tumor treatment by RT, indicating that simply aiming at enhancing primary tumors treatment is far from satisfaction, and the problems of tumor recurrence and metastasis are urgent to be solved. The Si-Mn-based NPs have been synthesized to regulate the TME to a beneficial status before exposure to radiation in radio-immunotherapy from different aspects: oxygenation, GSH depletion, ROS generation, and immune activation. Notably, due to the ROS-involved redox metabolism equilibrium, only generating ROS may trigger more GSH production and result in more ROS consumption by GSH. Here, Mn-based nanomaterials not only induce ROS generation, but also deplete GSH, which breaks the ROS-involved redox metabolism equilibrium to regulate TME. In conclusion, the Si-Mn-based NPs have not only acted as the radiosensitizer to enhance the treatment of primary tumors by RT at a low dose of radiation, but also played a role in activating immune response to inhibit the tumor recurrence and metastasis.

Herein, we prepared Si - Mn - based NPs by coating MnO2 onto SiO2 NPs (SM NPs) by a hydrothermal method to solve the limitation of TME - induced resistance and inhibit tumor growth and tumor metastasis in radio-immunotherapy (Scheme 1). The obtained SM NPs induced the generation of O2, which could relieve hypoxia to reduce radiation tolerance and immune suppression, as well as depleted GSH and induced radical dotOH generation to increase the levels of ROS and lipid peroxidation (LPO) and enhance DNA damage in Lewis lung carcinoma (LLC) cells. Thus, the cancer cell killing effect of RT was improved at a low dose, at which dose radiation alone could'nt kill cancer cells. Furthermore, SM NPs stimulated antitumor immune responses through improving the secretion of interleukin 1β (IL - 1β) and tumor necrosis factor - α (TNF - α) by macrophage - like cells in vitro. Interestingly, combining low - dose radiation with SM NPs and the anti - CTLA - 4 antibody induced a significant inhibitory effect on the growth of primary tumors and distant untreated tumors. Therefore, it was demonstrated that SM NPs may provide a new strategy for radio - immunotherapy to inhibit primary tumor growth at a low dose of radiation and suppress metastatic tumor growth even at distant sites not exposed to radiation.

Section snippets

Materials

Hexadecyltrimethylammonium bromide (CTAB), tetraethyl orthosilicate (TEOS), potassium permanganate (KMnO4), and N - acetyl - l - cysteine (NAC) were from Wako Chemical, Ltd. MB and hydrogen peroxide (H2O2, 30%) were from FUJIFILM Wako Pure Chemical Co. 5 mol/L - sodium hydroxide solution (NaOH) was from Nacalai Tesque, Inc. Glutathione reduced form (GSH) was from Tokyo Chemical Industry Co., Ltd. Liperfluo was from Dojindo Molecular Technologies, Inc. The anti - CTLA- 4 antibody (9D9; Cat.

Preparation and characterization

As shown in Fig. 1a–b, uniform SM NPs were obtained by coating MnO2 onto SiO2 NPs by the hydrothermal method. Results of element distribution analysis are shown in Fig. 1c, which confirmed that Mn was successfully and uniformly coated onto the surface of SiO2 NPs. The mass ratio of Mn in SM NPs was 14.0% ± 2.6% as shown by ICP analysis. Moreover, the phase of Mn in SM NPs was demonstrated by XRD analysis. As shown in the XRD patterns of SM NPs (Fig. 1d), three new diffraction peaks appeared for

Conclusions

In this work, core - shell structured SM NPs were synthesized for use in synergistic radio-immunotherapy. SM NPs regulated TME to a status beneficial for radio - immunotherapy through O2 production in situ to relieve hypoxia, radical dotOH production, and GSH depletion to increase ROS levels, as well as through promotion of cytokine secretion to stimulate anticancer immune responses. Notably, SM NPs not only inhibited the growth of primary tumors at a low radiation dose at which radiation alone has no

Funding

This work was supported in part by AIST, Japan Society for the Promotion of Science (JSPS, KAKENHI Grant Number 22H03964, 17K01399).

CRediT authorship contribution statement

X.Y. performed the experiments, analyzed the results and wrote the manuscript. X.W. conceived the experiments, analyzed the results and revised the manuscript. L.S., A.Y. and X.L. supported parts of the experiments, discussed the results and revised the manuscript. X.W. oversaw the whole project.

Declaration of competing interest

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

Acknowledgments

We thank Dr. Atsuo Ito for his fruitful discussion, thank Dr. Yu Sogo and Ms. Hisako Sugino for their discussion and technical assistance. X.P. YU thanks the support of Japanese Government Scholarship. This work was supported in part by NIMS Molecule & Material Synthesis Platform as a program of “Nanotechnology Platform” of the Ministry of Education, Culture, Sports, Science and Technology (MEXT), Japan. FE - SEM and XPS measurements were performed at the Materials Characterization Central

References (87)

  • L. Deng et al.

    STING-dependent cytosolic DNA sensing promotes radiation-induced type I interferon-dependent antitumor immunity in immunogenic tumors

    Immunity

    (2014)
  • S.-R. Woo et al.

    STING-dependent cytosolic DNA sensing mediates innate immune recognition of immunogenic tumors

    Immunity

    (2014)
  • X. Wang et al.

    Particle-size-dependent toxicity and immunogenic activity of mesoporous silica-based adjuvants for tumor immunotherapy

    Acta Biomater.

    (2013)
  • X. Wang et al.

    Si-doping increases the adjuvant activity of hydroxyapatite nanorods

    Colloids Surf. B: Biointerfaces

    (2019)
  • Z. Zou et al.

    Programmed packaging of mesoporous silica nanocarriers for matrix metalloprotease 2-triggered tumor targeting and release

    Biomaterials

    (2015)
  • J.-G. Wang et al.

    A high-performance asymmetric supercapacitor based on carbon and carbon–MnO2 nanofiber electrodes

    Carbon

    (2013)
  • J. Huang et al.

    Tumor microenvironment responsive hollow mesoporous Co9S8@MnO2-ICG/DOX intelligent nanoplatform for synergistically enhanced tumor multimodal therapy

    Biomaterials

    (2020)
  • H. Bi et al.

    Glutathione and H2O2 consumption promoted photodynamic and chemotherapy based on biodegradable MnO2–Pt@Au25 nanosheets

    Chem. Eng. J.

    (2019)
  • Y.-J. Cai et al.

    Salvicine triggers DNA double-strand breaks and apoptosis by GSH-depletion-driven H2O2 generation and topoisomerase II inhibition

    Free Radic. Biol. Med.

    (2008)
  • A. Morales et al.

    Oxidative damage of mitochondrial and nuclear DNA induced by ionizing radiation in human hepatoblastoma cells

    Int. J. Radiat. Oncol.Biol.Phys.

    (1998)
  • B.R. Stockwell et al.

    The chemistry and biology of ferroptosis, cell

    Chem. Biol.

    (2020)
  • R.M. Locksley et al.

    The TNF and TNF receptor superfamilies

    Cell

    (2001)
  • L. Deng et al.

    STING-dependent cytosolic DNA sensing promotes radiation-induced type I interferon-dependent antitumor immunity in immunogenic tumors

    Immunity

    (2014)
  • S.-R. Woo et al.

    STING-dependent cytosolic DNA sensing mediates innate immune recognition of immunogenic tumors

    Immunity

    (2014)
  • L. Ding et al.

    PARP inhibition elicits STING-dependent antitumor immunity in Brca1-deficient ovarian cancer

    Cell Rep.

    (2018)
  • G.M. Ross

    Induction of cell death by radiotherapy

    Endocr. Relat. Cancer

    (1999)
  • W. Ngwa et al.

    Using immunotherapy to boost the abscopal effect

    Nat. Rev. Cancer

    (2018)
  • G. Song et al.

    Emerging nanotechnology and advanced materials for cancer radiation therapy

    Adv. Mater.

    (2017)
  • C. Wang et al.

    Coordination polymer-coated CaCO3 reinforces radiotherapy by reprogramming the immunosuppressive metabolic microenvironment

    Adv. Mater.

    (2021)
  • D.N. Khalil et al.

    The future of cancer treatment: immunomodulation, CARs and combination immunotherapy, nature reviews

    Clin. Oncol.

    (2016)
  • M.Z. Dewan et al.

    Fractionated but not single-dose radiotherapy induces an immune-mediated abscopal effect when combined with anti-CTLA-4 antibody

    Clin. Cancer Res.

    (2009)
  • S.J. Dovedi et al.

    Fractionated radiation therapy stimulates antitumor immunity mediated by both resident and infiltrating polyclonal T-cell populations when combined with PD-1 blockade

    Clin. Cancer Res.

    (2017)
  • T. Yamazaki et al.

    Mitochondrial DNA drives abscopal responses to radiation that are inhibited by autophagy

    Nat. Immunol.

    (2020)
  • M.A. Swartz et al.

    Tumor microenvironment complexity: emerging roles in cancer therapy

    Cancer Res.

    (2012)
  • K. Liang et al.

    Breaking the redox homeostasis: an albumin-based multifunctional nanoagent for GSH depletion-assisted chemo-/Chemodynamic combination therapy

    Adv. Funct. Mater.

    (2021)
  • T. van den Beucken et al.

    Hypoxia promotes stem cell phenotypes and poor prognosis through epigenetic regulation of DICER

    Nat. Commun.

    (2014)
  • L.H. Gray et al.

    The concentration of oxygen dissolved in tissues at the time of irradiation as a factor in radiotherapy

    Br. J. Radiol.

    (1953)
  • D.K.-C. Chiu et al.

    Hypoxia inducible factor HIF-1 promotes myeloid-derived suppressor cells accumulation through ENTPD2/CD39L1 in hepatocellular carcinoma

    Nat. Commun.

    (2017)
  • Y. Li et al.

    Hypoxia-driven immunosuppressive metabolites in the tumor microenvironment: new approaches for combinational immunotherapy

    Front. Immunol.

    (2018)
  • F. Gong et al.

    Ultrasmall oxygen-deficient bimetallic oxide MnWOX nanoparticles for depletion of endogenous GSH and enhanced sonodynamic cancer therapy

    Adv. Mater.

    (2019)
  • H. Wang et al.

    Nanoscale covalent organic polymers as a biodegradable nanomedicine for chemotherapy-enhanced photodynamic therapy of cancer

    Nano Res.

    (2018)
  • C. Zhang et al.

    Synthesis of iron nanometallic glasses and their application in cancer therapy by a localized Fenton reaction

    Angew. Chem. Int. Ed.

    (2016)
  • B. Yang et al.

    A metal-organic framework (MOF) Fenton nanoagent-enabled nanocatalytic cancer therapy in synergy with autophagy inhibition

    Adv. Mater.

    (2020)
  • Cited by (5)

    • Manganese Dioxide-Based Nanomaterials for Medical Applications

      2023, ACS Biomaterials Science and Engineering
    1

    X Li's current address: Smart Polymers Group, National Institute for Materials Science (NIMS), 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan.

    View full text