Cancer Letters

Cancer Letters

Volume 503, 10 April 2021, Pages 151-162
Cancer Letters

Immunization with alloantibodies-covered melanoma cells induces regional antitumor effects that become systemic when combined with 5-FU treatment

https://doi.org/10.1016/j.canlet.2021.01.027Get rights and content

Highlights

  • High titer allo-IgG against melanoma promote tumor immunogenicity of allo-IgG-B16.

  • cDCs in lymph nodes capture and present tumor antigens released by allo-IgG-B16.

  • Allo-IgG-B16, together with 5-FU, induce a systemic antitumor response.

Abstract

Alloantibodies, in particular immunoglobulin G (allo-IgG), confer a rejection advantage to tumors sharing the same major histocompatibility complex (MHC) in mice. However, when administrated intratumorally, this effect can only be achieved in combination with dendritic cells (DCs) activation. Here, we developed high titer allo-IgG by multiple rounds of immunization with allogenic B16 melanoma cells, which allows for the strong binding with B16 cells. We demonstrate that B16 cells incubated with these allo-IgG (referred to as allo-IgG-B16) become highly immunogenic, which release tumor antigens that are efficiently presented by classic DCs in lymph nodes (LNs). Injection of allo-IgG-B16 turns the tumor into an immune hot one and even elicits a systemic antitumor response when used together with 5-fluorouracil (5-FU). This systemic response is tumor-specific and relies on the critical site – LNs. Our findings provide a rationale for the use of allo-IgG in cancer treatment.

Introduction

The host immune response to major histocompatibility complex (MHC)-mismatched antigens, i.e., alloantigen, present on donor cells or organs (called donor graft), is called alloimmunization [1]. This process leads to the generation of alloantibodies produced by B cells that are capable of binding specifically to alloantigen [2]. When released into the bloodstream, alloantibodies may allow the immune system to destroy the donor organs, thereby strongly correlating with allograft rejection [3,4]. Conversely, the removal and prevention of alloantibodies production appears to correlate with allograft tolerance [5]. In a similar manner, allogenic tumor cells injected subcutaneously can be rejected by the host immune system through the production of alloantibodies, in particular IgG class (allo-IgG) [6]. When injected intravenously, e.g., B16 melanoma cells (H-2Kblo) Balb/c hosts (H-2Kd), induction of allo-IgG may contribute significantly to the rapid and complete rejection of the secondary subcutaneous B16 [7].

As such, adoptive transfer of allo-IgG can lead to the rejection of syngeneic tumors [6]. However, the use of intratumoral injection of allo-IgG as a therapeutic intervention against established tumors fails to delay tumor growth [6]. This limitation can be bypassed by the combination of allo-IgG with dendritic cells (DCs) stimuli that induce tumor associated DCs activation and accumulation [6]. Presumably, the addition of DCs stimuli increased the IgG binding, resulting in tumor eradication [6]. This notion is supported by the uptake of ovalbumin-immune complexes (IC) by DCs, which were subsequently presented to ovalbumin-specific T cells to elicit strong antitumor immune responses [8,9]. However, the antitumor potential of the administration of tumor cells coated with allo-IgG to acquire the immunogenicity of tumor cells per se and its subsequent immune effects in vivo (i.e., the uptake and presentation of tumor-released antigens), was never tested experimentally.

Current limitations in the use of allo-IgG, i.e., natural antibodies, which are present in the circulation of healthy donors and are apparently produced without overt exposure to the corresponding alloantigens [6], make it particularly challenging to ascertain the effectiveness of allo-IgG to tumor-specific immunotherapy. Here, we developed high titer allo-IgG by multiple rounds of alloimmunization with weakly immunogenic B16 melanoma cells. When incubated with B16 cells, these allo-IgG formed an IC with B16 cells (i.e., allo-IgG-B16), conferred high immunogenicity to B16 cells, which released tumor antigens that can be taken up and presented efficiently by classical DCs. We demonstrate that injection of allo-IgG-B16 results in effective local tumor control and when used together with 5-fluorouracil (5-FU), also in systemic antitumor response that is tumor-specific (against contralateral B16 tumor, but not MC38 tumor).

Section snippets

Tumor cells and mice

The melanoma cell line B16, T cell lymphoma cell line EL-4, and lymphocytic leukemia cell line L1210 were obtained from the ATCC (Rockville, MD, USA). The colon carcinoma cell line MC38 was kindly provided by Prof. Max Mazzone (VIB-KU Leuven Center for Cancer Biology, Leuven, Belgium). Tumor cells were cultured in DMEM (Gibco) supplemented with 10% heat-inactivated FCS, 2 mM l-glutamine, 100 U/mL penicillin, and 100 g/mL streptomycin (Gibco) under standard conditions and were not grown for more

High titer allo-IgG against B16 melanoma

To efficiently produce alloantibodies against tumor cells, we developed an alloimmunization profile using a MHC-mismatched mouse tumor model, in which Balb/c mice (H-2Kd) were challenged with multiple rounds of subcutaneous injection of B16 melanoma cells (H-2Kb). We analyzed alloantibodies as previously defined by their IgG production [7] and responsiveness to B16 cells and found elevated IgG levels in serum (i.e., high titers) in melanoma recipients compared with naïve Balb/c mice (Fig. 1A).

Discussion

The use of nature alloantibodies, especially IgG, elicits strong antitumor responses that can be exploited as a powerful therapeutic approach against cancer [6]. However, this in situ vaccination effect is achieved only when combined with either DCs stimuli or tumor associated DC-intrinsic checkpoint inhibitors [6,18]. To overcome this limitation, we developed allo-IgG against less immunogenic tumors (e.g., B16 melanoma) to study the immunogenic changes of melanoma cells per se when loaded with

Author contributions

Conception and design: N. Dang, M. Waer, B. Sprangers

Acquisition of data: N. Dang, Y. Lin, M. Waer

Analysis and interpretation of data: N. Dang, Y. Lin, M. Waer, B. Sprangers

Writing, review, and/or revision of the manuscript: N. Dang, Y. Lin, M. Waer, B. Sprangers

Study supervision: N. Dang, M. Waer, B. Sprangers.

Declaration of competing interest

No potential conflicts of interest were disclosed.

Acknowledgments

This work was supported by Olivia Hendrickx Research Fund (http://www.olivia.be); Belgium; Grant ID: HHRF01-0210 to Nana Dang. We thank past and present members of the Waer lab for their assistance and feedback. B. Sprangers is a senior clinical investigator for The Research Foundation Flanders (to Fonds Wetenschappelijk Onderzoek – Vlaanderen; 1842919 N).

References (49)

  • Y. Carmi et al.

    Allogeneic IgG combined with dendritic cell stimuli induce antitumour T-cell immunity

    Nature

    (2015)
  • N. Dang et al.

    Improved anti-tumour adaptive immunity can overcome the melanoma immunosuppressive tumour microenvironment

    Cancers

    (2019)
  • K. Rafiq et al.

    Immune complex–mediated antigen presentation induces tumor immunity

    J. Clin. Invest.

    (2002)
  • D.H. Schuurhuis et al.

    Immune complex-loaded dendritic cells are superior to soluble immune complexes as antitumor vaccine

    J. Immunol.

    (2006)
  • N. Dang et al.

    Solid tumor–induced immune regulation alters the GvHD/GvT paradigm after allogenic bone marrow transplantation

    Canc. Res.

    (2019)
  • N. Dang et al.

    Donor lymphocyte-derived nature killer cells control MHC class I-negative melanoma

    Cancer Immunol Res.

    (2020)
  • L.I. Brasoveanu et al.

    Expression of protectin (CD59) in human melanoma and its functional role in cell- and complement-mediated cytotoxicity

    Int. J. Canc.

    (1995)
  • R. Zhang et al.

    Pancreatic cancer-educated macrophages protect cancer cells from complement-dependent cytotoxicity by up-regulation of CD59

    Cell Death Dis.

    (2019)
  • N.T. Joncker et al.

    The site of tumor development determines immunogenicity via temporal mobilization of antigen-laden dendritic cells in draining lymph nodes,

    Eur. J. Immunol.

    (2015)
  • A. Malesci et al.

    Tumor-associated macrophages and response to 5-fluorouracil adjuvant therapy in stage III colorectal cancer

    OncoImmunology

    (2017)
  • S.L. Topalian et al.

    Neoadjuvant checkpoint blockade for cancer immunotherapy

    Science

    (2020)
  • Y. Carmi et al.

    Akt and SHP-1 are DC-intrinsic checkpoints for tumor immunity

    JCI Insight

    (2016)
  • M.J. Frank et al.

    Situ vaccination with a TLR9 agonist and local low-dose radiation induces systemic responses in untreated indolent lymphoma

    Canc. Discov.

    (2018)
  • A.M. Salazar et al.

    Therapeutic in situ autovaccination against solid cancers with intratumoral poly-ICLC: case report, hypothesis, and clinical trial

    Cancer Immunol Res.

    (2014)
  • Cited by (1)

    • Establishment of operational tolerance to sustain antitumor immunotherapy

      2022, Journal of Heart and Lung Transplantation
      Citation Excerpt :

      Cardiac allografts upon rejection at the end of the experiments were removed by surgical procedures and fixed in 4% formaldehyde solution for 24 hours at room temperature and embedded in paraffin. For each organ, 5-mm sections were stained with hematoxylin and eosin (H&E) at VIB-KU LEUVEN Center for Cancer Biology (Belgium).12,19-22 Slides were examined by microscope (Zeiss Axio Scan, Zeiss, Koln, Germany).

    View full text