Original Contribution
Dendritic Cells Loaded with Ultrasound-Ablated Tumour Induce in vivo Specific Antitumour Immune Responses

https://doi.org/10.1016/j.ultrasmedbio.2009.12.004Get rights and content

Abstract

Previous studies have shown that high-intensity focused ultrasound (HIFU) ablation can induce a local inflammation with marked infiltration of dendritic cells (DCs). The purpose of this study was to investigate whether DCs could capture and present activating signals delivered by necrotic tumour cells that remain in situ after HIFU, thus initiating specific antitumour immunity. Tumour debris was derived from a mouse H22 tumour model after HIFU ablation. Bone marrow-derived DCs were loaded with HIFU-treated tumour, tumour lysate and mouse serum. Syngeneic naïve C57BL/6J mice were immunised with three loaded DCs followed by a subsequent H22 tumour challenge. Tumour size and survival were then recorded in each vaccinated mouse. The results showed that both HIFU-ablated tumour and tumour lysate could significantly increase the number of mature DCs and the secretion of IL-12 and IFN-γ (p < 0.001). The proliferation of splenic lymphocytes co-incubated with the loaded-DCs was significantly higher in both HIFU-ablated tumour and tumour lysate groups (p < 0.01). Cytotoxocity and TNF-α and IFN-γ secretion of cytotoxic T lymphocytes against H22 cells were significantly higher in HIFU-ablated tumour group than that in tumour lysate group (p < 0.01). After the H22 tumour challenge, a significant decrease of tumour volume was observed in HIFU-ablated tumour group (p < 0.01). However, there was no statistical difference of long-term survival rates among three groups (p > 0.05). It is concluded that DCs can be activated by HIFU-ablated tumour debris and, thus, initiate host specific antitumour immune response after HIFU therapy. (E-mail: [email protected])

Introduction

With development of medical techniques, minimally invasive therapies, such as radio-frequency, microwave, laser, high-intensity focused ultrasound (HIFU) and cryosurgery, have recently become an alternative approach to surgical intervention in the clinical management of solid malignancies. The main advantage of these alternatives is less invasive than surgical procedure, resulting in an associated reduction in mortality, morbidity, hospital stay and cost for cancer patients (Hong et al., 2006, Liapi and Geschwind, 2007, Timmerman et al., 2009).

Of all of these minimally invasive therapies, HIFU is the only noninvasive approach proposed to date. This technique employs extracorporeal ultrasound energy to ablate a targeted tumour at depth, with no damage to the skin and overlying tissues. It has been used for the treatment of patients with solid tumours, including those of prostate, liver, pancreas, breast, kidney, uterus, bone and soft tissue (Chaussy et al., 2005, Kennedy, 2005, Wu, 2006). Much of the clinical application is recent, where clinical results are very promising for local destruction of targeted tumours. In addition, previous studies have shown that HIFU therapy can enhance host antitumour immune response (Hu et al. 2007; Hundt et al. 2007; Lu et al., 2009, Rosberger et al., 1994, Wang and Sun, 2002, Wu et al., 2004). This may be of benefit in local recurrence and metastasis control in cancer patients who have previous dysfunction of antitumour immunity.

Dendritic cells (DCs) are the potent antigen presenting cells, which play a major role in induction of adaptive immunity against cancer (Melief 2008). They capture and present tumour antigens to naïve T lymphocytes in a major histocompatibility complex (MHC) restricted fashion, thus initiating specific antitumour immune responses. Activating signals, delivered directly or indirectly by tumour cells, can induce the progression of DCs from an immature to a mature stage (Lutz and Schuler 2002). Recent studies have shown that bone marrow-derived DCs are able to promote prophylactic antitumour immunity when loaded with relevant apoptotic or necrotic tumour cells (Goldszmid et al., 2003, Neidhardt-Berard et al., 2004; Schnurr et al. 2005).

It has been noted that large amounts of tumour debris remain in situ after HIFU therapy, which may contain tumour antigens and heat shock proteins (HSP), such as HSP60 (Hu et al. 2005), HSP27 (Madersbacher et al. 1998), HSP72 and HSP73 (Kramer et al. 2004), and HSP70 (Wu et al. 2007). Our previous findings have revealed that HIFU therapy can significantly induce local infiltration of activated DCs in the debris of HIFU-treated breast cancer (Xu et al. 2009). However, it is still unknown whether DCs could capture and present activating signals delivered by necrotic tumour cells and, thus, initiate host specific antitumour immunity. In this study, we used tumour debris created by in situ HIFU therapy to load with bone marrow-derived DCs and then inoculated naïve animals against a subsequent tumour challenge. The purpose of this study was to investigate whether DCs could be activated by HIFU-ablated tumour debris and, thus, initiate host specific antitumour immune response in a murine tumour model.

Section snippets

Antibodies and reagents

PE-anti-CD80 (RM80 clone), PE-anti-CD86 (PO3 clone) and FITC-anti-DEC205 (NLDC-145 clone) were purchased from Serotec (Oxford, UK). PE-anti-CD11c (N418 clone) and PE-anti-MHCII (M5/114.15.2) were obtained from eBioscience (San Diego, CA, USA). They were rat antimouse monoclonal antibodies. Mouse recombinant granulocyte macrophage colony-stimulating factor (mrGM-CSF) and mouse recombinant interleukin-4 (mrIL-4) were purchased from PeproTech (London, UK). Murine enzyme-linked immunosorbent assay

HIFU-ablated tumour induces maturation of DCs

Immature DCs isolated from 7-day C57BL/6 J bone marrow cultures were used to incubate with HIFU-ablated tumour, tumour lysate and mouse serum (a control). After 5-day incubation, expression of MHC class II, CD80 and CD86 molecules on DCs was determined by flow cytometry and IL-12 and IFN-γ levels were then measured by ELISA method in the culture supernatants of each group. As shown in Figure 1, incubation with either HIFU-ablated tumour or tumour lysate significantly increased the number of

Discussion

HIFU therapy is a noninvasive thermal ablation for local destruction of diseased tissues. It induces coagulation necrosis of a targeted tumour and, thus, results in large amounts of tumour debris remained in situ, which contain tumour antigens and HSPs (Hu et al., 2005, Kramer et al., 2004, Madersbacher et al., 1998, Wu et al., 2007). In addition, an acute inflammatory response has been observed at the margins of the treated tissue after HIFU. This leads to the rapid infiltration of a large

Acknowledgements

This work was supported by the Foundation of Ministry of Education of China (grant No. IRT0454).

References (39)

  • D.W. O'Neill et al.

    Manipulating dendritic cell biology for the active immunotherapy of cancer

    Blood

    (2004)
  • A.G. Pockley

    Heat shock proteins as regulators of the immune response

    Lancet

    (2003)
  • M. Schnurr et al.

    Tumor antigen processing and presentation depend critically on dendritic cell type and the mode of antigen delivery

    Blood

    (2005)
  • U. Wagner et al.

    Evaluation of canine lymphocyte proliferation: Comparison of three different colorimetric methods with the 3H-thymidine incorporation assay

    Vet Immunol Immunopathol

    (1999)
  • F. Wu et al.

    Pathological changes in human malignant carcinoma treated with high-intensity focused ultrasound

    Ultrasound Med Bio

    (2001)
  • F. Wu et al.

    Activated anti-tumor immunity in cancer patients after high-intensity focused ultrasound ablation

    Ultrasound Med Biol

    (2004)
  • Z.L. Xu et al.

    Activation of tumor-infiltrating antigen presenting cells by high-intensity focused ultrasound ablation of human breast cancer

    Ultrasound Med Biol

    (2009)
  • L.J. Appleman et al.

    T cell energy and co-stimulation

    Immunol Rev

    (2003)
  • D.I. Bounous et al.

    Comparison of MTT colorimetric assay and tritiated thymidine uptake for lymphocyte proliferation assays using chicken splenocytes

    Avian Dis

    (1992)
  • Cited by (58)

    • Synergies between therapeutic ultrasound, gene therapy and immunotherapy in cancer treatment

      2021, Advanced Drug Delivery Reviews
      Citation Excerpt :

      Therefore, reducing tumor burden through UMGT may impact treatment efficacy even in the absence of an effective transfection. Following specific focused ultrasound protocols, wound healing and inflammation in the TME upregulate danger signals, such as HSPs, damage-associated molecular patterns (DAMPs) [97] and ATP [132,133]. In [134], a non-ablative pulsed ultrasound protocol (1.15 MHz, PNP of 6 MPa, intensity spatial average temporal peak (ISATP) of 2683 W/cm2, 10% duty cycle, 5 Hz PRF, 100 pulses per site), altered the expression of cytokines, chemokines, and cell adhesion molecules, and immune cell phenotypes shifted to an inflamed TME.

    • Ultrasound in tumor immunotherapy: Current status and future developments

      2020, Journal of Controlled Release
      Citation Excerpt :

      The US-ablated tumor debris could also facilitate vaccine delivery conferring specific protective immunity. Bone-marrow-derived immature DCs primed with US-ablated tumor debris produced an obvious increase in the number of mature DCs and the secretion of IL-12 and IFN-γ by CTLs [34]. Nonetheless, the up-regulated expressions of MHC-II, CD80, and CD86 were observed, suggesting that US-ablation-generated vaccines could improve tumor immunogenicity [35].

    View all citing articles on Scopus
    View full text