Elsevier

Biomaterials

Volume 265, January 2021, 120421
Biomaterials

Photo-activated chemo-immunotherapy for metastatic cancer using a synergistic graphene nanosystem

https://doi.org/10.1016/j.biomaterials.2020.120421Get rights and content

Abstract

We developed a novel treatment strategy for metastatic cancer by synergizing photothermal therapy (PTT), chemotherapy, and immunotherapy through a nanosystem to trigger host antitumor immunity. The nanosystem was constructed by loading mitoxantrone (MTX), a chemotherapeutic agent, and SB-431542 (SB), a transforming growth factor beta (TGF-β) inhibitor, onto reduced graphene oxide (rGO). Intratumoral administration of rGO/MTX/SB, followed by non-invasive irradiation of a near-infrared laser, destroyed local primary tumors and inhibited distant metastases in 4T1 mouse mammary tumor model, which is poorly immunogenic and highly metastatic. After treatment, 70% of the tumor-bearing mice became long-term survivors and developed a tumor type-specific immunity to resist rechallenged tumor cells. We found that rGO-based PTT provided an immunogenic antigen source, forming in situ vaccination with rGO as an immune-adjuvant. The use of SB changed the tumor microenvironment and improved the therapeutic effect of MTX-generated chemotherapy and rGO-based PTT. The immunological functions of MTX, SB, and rGO acted synergistically to induce an effective tumor vaccination, as evidenced by the increased infiltration of tumor-specific cytotoxic CD8+ T lymphocytes and decreased infiltration of regulatory T cells (Tregs) in distal tumors. Collectively, we demonstrated that rGO/MTX/SB combined with laser irradiation provided a synergistic chemo-immuno-photothermal effect against tumors by in situ vaccination and inhibition of immunosuppressive microenvironment. This unique combination embodies a promising approach to treat metastatic cancers by inducing a systemic antitumor response through a local intervention.

Introduction

Triple-negative breast cancer (TNBC) accounts for 10–20% of diagnosed breast cancers, with limited options and poor prognosis [[1], [2], [3]]. More importantly, compared to hormone-positive breast cancer, TNBC is common in women younger than 40 years [4]. The cure rate of cancer has gradually increased in the past decades with the development of new cancer therapies, including target molecules, immune checkpoint inhibitors, vaccines, and CAR-T. However, the cure for TNBC is still elusive [[5], [6], [7], [8]]. For the unsatisfactory outcomes in treating TNBC, the major reason is the poor tumor immunogenicity and the strong immunosuppressive tumor microenvironment [9,10].

Chemotherapy has been one of the primary clinical modalities. However, the immunosuppressive effects of chemotherapeutic agents have been well known. As a few exceptions, doxorubicin (Dox) and mitoxantrone (MTX) induce topoisomerase II associated DNA damage in the cell nucleus, causing immunogenic cell death (ICD) that could trigger a tumor-specific immunity by releasing damage-associated molecular patterns (DAMPs) and tumor antigens [[11], [12], [13], [14]]. However, drug resistance is still the big challenge in chemotherapy, resulting in recurrence and drug-resistant metastases in the treated TNBC patients [15].

TGF-β is a pleiotropic cytokine that maintains an immunosuppressive environment to help established tumor escape from the host immune system [16]. Many tumors overexpression of TGF-β correlate with metastasis and poor prognosis. Therefore, TGF-β was considered as an ideal target and TGF-β inhibitors were developed for the treatment of cancer [17,18]. One of the TGF-β inhibitors, SB-431542, was found to decrease lung metastasis, however, there was no effect on the growth of the primary tumor in 4T1 xenograft tumor model [17]. LY2157299 prevented reestablishment of tumors after paclitaxel treatment of 4T1 xenograft [18]. Either chemotherapy agents or TGF-β inhibitors face severe limitations in treating TNBC due to delivery challenges and toxicity profiles.

Nano-graphene oxide (nGO) has been widely used as a drug carrier and photo-agent for cancer theranostics, due to its principal characteristics of 2D film structure and near-infrared absorption spectrum. Especially, PEG-nGO was found to activate macrophages with a potential to enhance the antitumor immunity [19]. We hypothesized that a combination of an in situ tumor vaccine with improved tumor microenvironment, should lead to an effective antitumor immunity. In addition, local application of chemotherapeutic and immunotherapeutic agents could be effective against tumors while reducing the systemic side effect.

Herein, we developed a graphene-based nano-system by conjugating rGO with MTX and SB for photo-activated, synergistic chemo-immunotherapy to treat metastatic TNBC using 4T1 mouse breast tumor model Fig. 1. The therapeutic efficacy of photo-activated chemo-immunotherapy was investigated. The host immunity, including induced ICD, antitumor vaccination, tumor-specific T cells, and the abscopal effect, were also investigated.

Section snippets

Synthesis and characterization of rGO/MTX/SB

The base of the nanoplatform, rGO, was conjugated with mPEG2000-NH2 following a well-established EDC/NHS protocol and purified by a 100 kDa filter through centrifugation. MTX and SB were then loaded onto rGO-PEG via π-π stacking interaction. The detailed protocol and analysis were shown in Supplemental Methods.

Cell culture and cellular treatment

4T1 murine breast cancer cell line and CT26 colon carcinoma cell line were both purchased from ATCC (Manassas, VA, USA), bone marrow macrophages and DCs were harvested from BALB/c mice,

Characterization of rGO/MTX/SB

The biocompatibility of rGO was improved by conjugating with mPEG2000-NH2, followed by MTX and SB loading via π-π stacking (Figs. S1-5). A UV–vis absorption spectrum of rGO/MTX/SB showed the characteristic absorption peaks of MTX and SB, indicating successful loading of MTX and SB, with a final size of 100–200 nm (Fig. 2A and B). The temperature increase of rGO/MTX/SB solutions detected under laser irradiation with an 805-nm laser indicated that rGO/MTX/SB kept the photothermal transfer effects

Discussion

The potent therapeutic strategies for metastatic cancer require efficient antitumor responses by eradicating primary tumors and controlling distal metastases. To this end, chemo-immunotherapy was developed as a combination therapeutic strategy by synergizing the tumor killing effect and antitumor immunomodulatory effect to control primary tumors and metastases with reduced drug doses. However, drug resistance and toxicity are still the big challenges due to the systemic delivery of

Credit author statement

Feifan Zhou, Designed and directed the project, Performed experiments, Analyzed data, Wrote the paper. Meng Wang, Designed and performed experiments, Analyzed data. Teng Luo, Performed experiments. Junle Qu, Designed and directed the project, Provided scientific input, Analyzed data, Wrote the paper. Wei R. Chen, Designed and directed the project, Provided scientific input, Analyzed data, Wrote the paper.

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

This study was supported in part by grants from the US National Institutes of Health R01 CA205348 (W.R.C.), the Oklahoma Center for Advancement of Science and Technology HR16-085 (W.R.C.), the National Natural Science Foundation of China (61722508/61525503/61620106016/61835009/81727804) (J.Q.), Guangdong Province Key Area R&D Program 2019B110233004 and Hainan University R&D Program (KYQD(ZR)20074) (F.Z.), Shenzhen Fundamental Research Program JCYJ20190808114609361 (M.W.).

References (34)

  • P. Sharma et al.

    The future of immune checkpoint therapy

    Science

    (2015)
  • R. Zhou et al.

    CAR T cells targeting the tumor MUC1 glycoprotein reduce triple-negative breast cancer growth

    Front. Immunol.

    (2019)
  • J.P. Bates et al.

    Mechanisms of immune evasion in breast cancer

    BMC Canc.

    (2018)
  • N. Casares et al.

    Caspase-dependent immunogenicity of doxorubicin-induced tumor cell death

    J. Exp. Med.

    (2005)
  • M. Michaud et al.

    Autophagy-dependent anticancer immune responses induced by chemotherapeutic agents in mice

    Science

    (2011)
  • T. Blankenstein et al.

    The determinants of tumour immunogenicity

    Nat. Rev. Canc.

    (2012)
  • G. Kroemer et al.

    Immunogenic cell death in cancer therapy

    Annu. Rev. Immunol.

    (2013)
  • Cited by (73)

    • Z-scheme MoS<inf>2</inf>/Co<inf>3</inf>S<inf>4</inf>@PEG nanoflowers: Intracellular NIR-II photocatalytic O<inf>2</inf> production facilitating hypoxic tumor therapy

      2023, Biomaterials Advances
      Citation Excerpt :

      In summary, with excellent therapy efficiency and good biological safety, MSC-2@PEG+NIR-II can be served as a potential nanoagent for anticancer. In view of the previous reports, immunogenic cell death (ICD) could facilitate the maturation of dendritic cells (DCs) to arouse antitumor immune response [37,38]. After treatment for 3 days, the inguinal lymph of mouse was collected to make cell suspensions, and lymphoid CD80 and CD86 (typical DC-maturated indicators) levels were evaluated by the flow cytometry method.

    • Chitosan-functionalized graphene oxide as adjuvant in HEV P239 vaccine

      2022, Vaccine
      Citation Excerpt :

      Compared with those conventional adjuvants, functionalized GO was capable of enhancing both humoral and cellular immunity [22]. At present, targeted tumor photodynamic therapy utilizing functional GO loaded with photosensitizer can not only deliver drugs to the target, but also trigger an immune response and enhance the efficacy of photodynamic therapy [23]; lentinan (LNT) modified nano-GO can improve the degree of innate and adaptive immune response as well as sustain the body's long-term immune response [24]. Thus far, numerous studies have shown that GO enhances the secretion of macrophages by activating the NF-kB signaling pathway, and activates the immune system by promoting the proliferation and differentiation of lymphocytes (especially CD8+T cells) and the secretion of inflammatory mediators [25].

    View all citing articles on Scopus
    1

    Both authors contributed equally to this manuscript.

    View full text