Elsevier

Vaccine

Volume 34, Issue 1, 2 January 2016, Pages 134-141
Vaccine

The efficacy of a novel vaccine approach using tumor cells that ectopically express a codon-optimized murine GM-CSF in a murine tumor model

https://doi.org/10.1016/j.vaccine.2015.10.106Get rights and content

Abstract

Granulocyte macrophage-colony stimulating factor (GM-CSF) is a potent immunomodulatory cytokine that is known to facilitate vaccine efficacy by promoting the development and prolongation of both humoral and cellular immunity. Here, we investigated a novel vaccine approach using a human papillomavirus (HPV)-16 E6/E7-transformed cell line, TC-1, that ectopically expresses a codon-optimized 26-11-2015 murine GM-CSF (cGM-CSF). Ectopically expressing cGM-CSF in TC-1 (TC-1/cGM) cells significantly increased expression of a GM-CSF that was functionally identical to wt GM-CSF by 9-fold compared with ectopically expressed wild type GM-CSF in TC-1 cells (TC-1/wt). Mice vaccinated with irradiated TC-1/cGM cells exhibited enhanced survival compared with mice vaccinated with TC-1/wt cells when both groups were subsequently injected with live TC-1. Consistently, mice vaccinated with irradiated TC-1/cGM cells exhibited stronger IFN-γ production in HPV E7-specific CD8+ T cells. More dendritic cells were recruited to the draining lymph nodes (dLNs) of mice vaccinated with TC-1/cGM cells than C-1/wt cells. Regarding dLN cell recall responses, both proliferation and IFN-γ production in the HPV E7-specific CD8+ T cells were enhanced in mice that were vaccinated with TC-1/cGM cells. Our results demonstrate that a novel practical molecular strategy utilizing a codon-optimized GM-CSF gene overcomes the limitation and improves the efficacy of tumor cell-based vaccines.

Introduction

Cervical cancer is the second most frequent gynecological malignancy worldwide and accounts for approximately 12% of all cancers in women [1]. Greater than 99% of cervical cancer patients are carriers of human papillomaviruses (HPV), which makes HPV a major risk factor for cervical cancer [2]. HPVs are double-strand DNA viruses with a genome size of approximately 8 kb that encode 8 or 9 open reading frames consisting of eight early (E1–E9) and two late (L1 and L2) gene products [3]. HPV integration into the genome and high-level E6 and E7 expression are often associated with the manifestation of high-grade cervical neoplasia [2], [4], [5]. Although virus-like particle (VLP)-based vaccines have been developed with prophylactic activities to prevent most HPV infections [6], [7], the therapeutic effect of VLP vaccines has yet to be demonstrated for those who were already infected [8]. Additionally, although prophylactic vaccines induce a humoral immune response against the viral capsid protein L1, a cellular immune response against virus early proteins, E6 and E7, is required to eliminate previously infected cells [9], [10]. The viral transforming proteins, E6 and E7, are consistently expressed in cervical cancer cell lines and HPV-associated neoplasms. Thus, E6 and E7 represent true tumor-specific antigens and serve as a target for the development of immunotherapeutic strategies to combat HPV-associated cancers.

Granulocyte macrophage-colony stimulating factor (GM-CSF) was initially characterized as a factor that can support the in vitro colony formation of granulocyte-macrophage progenitors [11]. GM-CSF also serves as a growth factor for erythroid, megakaryocyte, and eosinophil progenitors. GM-CSF is produced by various cell types, including T cells, B cells, macrophages, mast cells, endothelial cells, fibroblasts, and adipocytes, in response to cytokine or inflammatory stimuli. In mature hematopoietic cells, GM-CSF is a survival factor and activates the effector functions of granulocytes, monocytes/macrophages, and eosinophils. In recent years, both murine tumor models and human clinical trials revealed that GM-CSF-secreting tumors cells can serve as an option for immunotherapy in several tumor types, including non-small cell lung carcinoma [12], pancreatic [13], prostate [14], melanoma [15], leukemia [16], gliomas [17], cervical [18], and renal cell carcinoma [19]. However, GM-CSF gene expression is highly regulated at multiple levels [20], [21], [22]. Native GM-CSF protein is poorly expressed in a tissue-specific and activation-dependent manner. A clinical trial demonstrated that a threshold level of GM-CSF is required for the induction of effective immunity [23]. We previously described that the co-administration of plasmids encoding the codon-optimized murine GM-CSF (cGM) sequence with a DNA vaccine resulted in a strong and protective antibody and cytotoxic T lymphocyte (CTL) immune response against recombinant vaccinia virus challenge [24].

In the present study, we applied a lentiviral-delivered codon-optimized murine GM-CSF into a HPV-16 E6/E7-transformed cell line, TC-1, as a vaccine adjuvant approach. The goal of the present study was to assess whether codon-optimized murine GM-CSF that is transduced into cancer cells as a vaccine would increase (dendritic cell) DC recruitment and result in enhanced antigen (Ag)-specific immune responses [28]. The lentiviral-modified TC-1/cGM cells showed significantly increased GM-CSF protein expression levels as demonstrated by Western blot and (enzyme-linked immunosorbent assay) ELISA assays. The GM-CSF secreted from the lentiviral-modified TC-1/cGM cell culture medium exhibited identical biological functions to the WT GM-CSF in its ability to support NFS-60 cell growth. The infected cells were continuously propagated for more than 30 passages, and stable lentiviral transgene expression was confirmed. We subcutaneously (s.c.) immunized C57BL/6 mice with a lethally irradiated TC-1/cGM cell-based vaccine and subsequently quantified CD11c+ DC and CD8+ by flow cytometry analysis. Vaccination with TC-1/cGM induced strong systemic CD8+ T cell immune responses against lethal TC-1 challenge and specific MHC class I HPV E7 epitopes, as demonstrated by an enzyme-linked immunospot (ELISPOT) assay. Peak DC recruitment was detected at 72 h post-inoculation in dLNs and was significantly increased (p < 0.05) in mice that were vaccinated with TC-1/cGM cells compared with control mice or mice inoculated with TC-1/wt cells. These results support the use of this novel codon-optimized GM-CSF for immunotherapeutic cancer vaccine strategies.

Section snippets

Mice

Female C57BL/6 (B6) mice were purchased from National Laboratory Animal Center (Taipei, Taiwan) and housed under specific pathogen-free (SPF) conditions at the animal facility of Chang-Gung University. All of the experiments were approved by the Animal Ethics Committee, Chang Gung University. All of the mice that were used were 8–12 weeks in age.

Cell lines

TC-1 cells were generated by co-transformation of primary B6 mouse lung epithelial cells with HPV-16 E6/E7 in combination with an activated H-ras

TC-1 cells transfected with LV-cGM (TC-1/cGM) expressed increased levels of functional GM-CSF compared with TC-1 cells transfected with LV-wtGM (TC-1/wtGM)

We previously demonstrated that cGM-CSF could be used as an adjuvant to enhance the immunity of a DNA vaccine against HIV-1 infection [24]. To explore its use as a prophylactic vaccine adjuvant in cancer immunotherapy, we generated lentiviral vectors that expressed eGFP, wt GM-CSF (wtGM) and cGM-CSF (cGM) to infect TC-1 cells. Stable clones were then screened and cultured for 24 h to determine the levels of secreted GM-CSF. As shown in Fig. 1A, all TC-1 clones stably infected with LV-cGM

Discussion

The efficacy of therapeutic vaccines generally correlates with the adjuvants that may generate strong antigen-specific T and B cell responses, and enhancement of such responses may increase the overall potency of immunotherapies. GM-CSF is one of the most potent adjuvants and is used in many tumor immunization schemes, including DNA-, peptide-, tumor cell-, or dendritic cell-based vaccines. Despite numerous demonstrations of its strong immune-stimulatory effects in animal models and clinical

Acknowledgments

We are grateful to Long-Ji Chang and Becky Chen for their helpful discussions. This study was supported by Chang Gung Memorial Hospital (CMRPG3C0681 and CMRPG3C0682) and Ministry of Science and Technology (NSC 102-2314-B-182-041-MY2) of Taiwan, respectively. The authors wish to thank Chang Gung Medical Foundation Kaohsiung and Taoyuan Chang Gung Memorial Hospital Tissue Bank for their excellent technical support.

References (38)

  • D.M. Parkin et al.

    Global cancer statistics, 2002

    CA: Cancer J Clin

    (2005)
  • J. Doorbar

    Molecular biology of human papillomavirus infection and cervical cancer

    Clin Sci (Lond)

    (2006)
  • M. von Knebel Doeberitz et al.

    Inhibition of tumorigenicity of cervical cancer cells in nude mice by HPV E6-E7 anti-sense RNA

    Int J Cancer

    (1992)
  • H. zur Hausen

    Papillomaviruses causing cancer: evasion from host-cell control in early events in carcinogenesis

    J Natl Cancer Inst

    (2000)
  • L.L. Villa et al.

    Quadrivalent vaccine against human papillomavirus to prevent high-grade cervical lesions

    N Engl J Med

    (2007)
  • S.M. Garland et al.

    Quadrivalent vaccine against human papillomavirus to prevent anogenital diseases

    N Engl J Med

    (2007)
  • N.B. Miller et al.

    Human papillomavirus vaccine

    N Engl J Med

    (2007)
  • M.F. Duggan-Keen et al.

    Papillomavirus vaccines

    Front Biosci

    (1998)
  • T.L. Cornelison

    Human papillomavirus genotype 16 vaccines for cervical cancer prophylaxis and treatment

    Curr Opin Oncol

    (2000)
  • Cited by (13)

    • Enhancing immunogenicity of HPV16 E<sup>7</sup> DNA vaccine by conjugating codon-optimized GM-CSF to HPV16 E<sup>7</sup> DNA

      2021, Taiwanese Journal of Obstetrics and Gynecology
      Citation Excerpt :

      After 72 h, the cell supernatants were collected for ELISA of IL-4 and IFN-γ. The intracellular IFN-γ levels of CD8+ or CD4+ T cells were characterized using a previously described flow cytometry protocol with modifications [22]. For the restimulation assay, a BD GolgiStop protein transport inhibitor containing monensin (BD Biosciences, USA) was added to the cells to block IFN-γ release 8 h before surface marker staining, to provide time for the protein to react.

    • Development and application of a bioluminescent imaging mouse model for Chikungunya virus based on pseudovirus system

      2017, Vaccine
      Citation Excerpt :

      In addition, an important area of research aims at improving the immunogenicity of DNA vaccines using molecular adjuvants such as cytokines. Among these cytokines, GM-CSF is one of the better-studied molecules and has many roles in immune regulation [28]. Our results showed that by combination of DNA vaccine and GM-CSF or followed by electroporation could enhance humoral immune responses in mice.

    • Current Immunotherapy Techniques for Cancer Treatment: A Scoping Review

      2023, Undergraduate Research in Natural and Clinical Sciences and Technology Journal
    View all citing articles on Scopus
    1

    These authors contributed equally to this work.

    View full text