Tumor associated antigen specific T-cell populations identified in ex vivo expanded TIL cultures

https://doi.org/10.1016/j.cellimm.2011.12.004Get rights and content

Abstract

Ex vivo expanded tumor infiltrating lymphocytes (TILs) from malignant melanoma (MM) and head & neck squamous cell carcinoma (HNSCC) share a similar oligoclonal composition of T effector memory cells, with HLA class I restricted lysis of tumor cell lines. In this study we show that ex vivo expanded TILs from MM and HNSCC demonstrate a heterogeneous composition in frequency and magnitude of tumor associated antigen specific populations by Elispot IFNγ quantitation. TILs from MM and HNSCC shared reactivity towards NY ESO-1, cyclin B1 and Bcl-x derived peptides. Additionally we show that dominating T-cell clones and functionality persists through out expansion among an oligoclonal composition of T-cells. Our findings mirror prior results on the oligoclonal composition of TIL cultures, further indicating a potential for a broader repertoire of specific effector cells recognizing the heterogeneous tumors upon adoptive transfer; increasing the probability of tumor control by minimizing immune evasion by tumor cell escape variants.

Highlights

TILs from MM and SCC recognize a broad repertoire of TAAs. ► TILs share specificity in MM and SCC patients. ► TILs have the potential of engaging the heterogeneous tumor structure.

Introduction

Head & neck squamous cell carcinoma (HNSCC) is the most frequent cancer derived from the aero-digestive tract. Patients with end stage (stage III–IV) and recurrent disease have a very poor prognosis when radiotherapy and/or surgery are no longer an option. Malignant melanoma (MM) is also a poorly controllable disease once it has reached an advanced stage. Unlike HNSCC, MM is often asymptomatic which allows for a disease spread prior to diagnosis. In spite of massive experimental clinical trials in both patient groups, survival has not improved significantly over the past two decades. Meanwhile, a recent phase III clinical trial has shown promising results in MM patients treated with Ipilimumab (CTLA-4 antibody) by significantly increased survival in the Ipilimumab treated patient group [1]. Although the molecular and cellular background for efficacy is largely unknown these data suggest that manipulation with cellular suppressive mechanisms may tip the balance from immune tolerance of cancer towards immunological attack.

Tumors are frequently infiltrated by cells of the immune system, which controls tumor growth at an early stage. Due to genetic instability the tumor cells undergo mutations leading to escape variants and progression [2]. In addition the tumor acquires immune suppressive mechanisms at a cellular (Treg [3], MDSC [4]), soluble (TH2 cytokines) and metabolic (ex. IDO [5]) level. Thereby the tumor environment is skewed towards a tolerogenic state.

In an attempt to tip the balance towards a tumor immunogenic state, different approaches of cancer immunotherapy have been tested including administration of cytokines and therapeutic vaccination. To the latter, although measurable immune responses were induced, limited clinical responses were obtained. However, recent studies in adoptive T-cell therapy have shown very promising results where 50% of MM patients with advanced disease obtained long lasting clinical objective responses after preconditioning lymphodepletion, infusion of ex vivo expanded tumor infiltrating lymphocytes (TILs) and high dose IL-2 [6]. TILs comprised T-cell populations with specificity towards autologous tumor, serving as a source of unknown tumor associated antigens (TAAs) [7]. Regarding identification of TAA peptide antigen specific T-cells among TILs, most attention has been directed at single epitopes derived from differentiation antigens such as MART-1 in MM patients. Thus, the antigen specific repertoire of TILs remains unknown and further attention is needed to identify tumor targets of potential clinical importance in adoptive T-cell therapy.

On the basis of the published methods from Dudley et al. we have established a method for successful expansion of TILs from both MM and HNSCC patients [8]. This has let us to initiate clinical pilot trials in MM patients (NCT00937625) and HNSCC patients (NCT00937300).

To identify the TAA specific populations in the TILs, we screened for reactivity against a panel of peptides covering three main categories of over expressed (OE), cancer testis (CT) and differentiation (D) TAAs. Belonging to the first category, the BCL-2 family proteins (Mcl-1, Bcl-2 and Bclx L) and survivin inhibits apoptosis and are in many cases crucial for tumor cell survival [9], [10], [11], [12]. Telomerase is another over expressed protein that ensures the capacity of cancer cells to continued proliferation [13], [14], while cyclin B1 ensures sustained mitosis [15], [16] and RhoC facilitates increased metastatic potential [17], [18]. Cancer testis antigens (CT) (MAGE 1, MAGE 3 and NY ESO-1) are silenced in normal tissue and upregulated in MM [19] and HNSCC [20], [21], rendering them suited targets for immune recognition. Finally, the differentiation antigen MART-1 is recognized as a highly immunogenic target in MM [22]. Specific T-cell populations for all the included peptides have previously been identified in the circulation of cancer patients. Here we show an occurrence of a broad repertoire of TAA specific T-cell populations from multiple individually established TIL cultures from MM and HNSCC patients. Although there is a tendency of an unequal distribution of TAA specific subgroups between TIL cultures and between patients, there is an equal broad representation of responses towards over expressed antigens (e.g. cyclin B1 and Bcl-x) and cancer testis antigens (e.g. NY ESO-1) in both patient categories.

Section snippets

Patients

Patients referred to surgery for primary or recurrent stage II–IV malignant melanoma or oral squamous cell carcinoma were eligible for the study. The study protocol was approved by the Local ethics committee and all patients were included after signing informed consent. Tumor material from the patients was obtained from the surgically removed tumor and processed for culturing within 30 min after surgery.

Tumor infiltrating lymphocytes (TILs)

TILs were generated accordingly to a previously described two-step expansion method

Patients

We examined the occurrence of TAA peptide specific T-cell populations, determined by IFNγ release measurement in Elispot, in ex vivo expanded TIL cultures from six HNSCC and six MM patients. In HNSCC (two HLA-A2+, three HLA-A3+ and one HLA-A2/A3+), all patients showed TAA specific responses in both alleles. In MM (four HLA-A2+ and two HLA-A3+), 5/6 (83%) patients responded in both alleles. Patient characteristics are summarized in Table 2.

TIL cultures

Lymphocytes migrated out from the fragments within two

Discussion

In recent years adoptive T-cell therapy has gained increased attention, due to the capacity of this treatment to mediate significant clinical responses, including partial or complete responses in heavily pre-treated patients by restoring the immunogenicity of tumors in melanoma patients. The most consistent objective response rates are obtained by infusion of ex vivo expanded autologous TIL bulk cultures supported by immune conditioning [29], [30], [31]. However, very little is known about the

Disclosures

No conflicts of interests to declare.

Acknowledgments

We would like to extend our appreciation of the excellent technical assistance from Mette Noe Vitting and Charlotte Vajhøj on TIL expansion, Kirsten Nikolajsen for flow cytometry analysis, and to Tobias Wirenfeldt Klausen for statistical assistance. The study was supported by the Danielsen Foundation, The Danish Cancer Society, Danish Medical Research Council, and Herlev University Hospital.

References (46)

  • H. Nishikawa et al.

    Regulatory T cells in tumor immunity

    Int. J. Cancer

    (2010)
  • D.H. Munn et al.

    Indoleamine 2,3-dioxygenase and tumor-induced tolerance

    J. Clin. Invest

    (2007)
  • M.E. Dudley et al.

    Adoptive cell transfer therapy following non-myeloablative but lymphodepleting chemotherapy for the treatment of patients with refractory metastatic melanoma

    J. Clin. Oncol.

    (2005)
  • D.J. Schwartzentruber et al.

    In vitro predictors of therapeutic response in melanoma patients receiving tumor-infiltrating lymphocytes and interleukin-2

    J. Clin. Oncol.

    (1994)
  • M.H. Andersen et al.

    Spontaneous cytotoxic T-cell responses against survivin-derived MHC class I-restricted T-cell epitopes in situ as well as ex vivo in cancer patients

    Cancer Res.

    (2001)
  • M.H. Andersen et al.

    The antiapoptotic member of the Bcl-2 family Mcl-1 is a CTL target in cancer patients

    Leukemia

    (2005)
  • M.H. Andersen et al.

    Spontaneous immunity against Bcl-xL in cancer patients

    J. Immunol.

    (2005)
  • L. Wenandy et al.

    The immunogenicity of the hTERT540–548 peptide in cancer

    Clin. Cancer Res.

    (2008)
  • H. Kao et al.

    Identification of cyclin B1 as a shared human epithelial tumor-associated antigen recognized by T cells

    J. Exp. Med.

    (2001)
  • R.S. Andersen et al.

    Identification of a cyclin B1-derived CTL epitope eliciting spontaneous responses in both cancer patients and healthy donors

    Cancer Immunol. Immunother.

    (2010)
  • E.A. Clark et al.

    Genomic analysis of metastasis reveals an essential role for RhoC

    Nature

    (2000)
  • L. Wenandy et al.

    RhoC a new target for therapeutic vaccination against metastatic cancer

    Cancer Immunol. Immunother.

    (2008)
  • O.L. Caballero et al.

    Cancer/testis (CT) antigens: potential targets for immunotherapy

    Cancer Sci.

    (2009)
  • Cited by (21)

    • CAR-T cells and combination therapies: What's next in the immunotherapy revolution?

      2018, Pharmacological Research
      Citation Excerpt :

      The presence of naturally occurring tumor-reactive T cells has been thoroughly documented in the peripheral blood, and within the tumors, of cancer patients [1–5].

    • Methods to improve adoptive T-cell therapy for melanoma: IFN-γ enhances anticancer responses of cell products for infusion

      2013, Journal of Investigative Dermatology
      Citation Excerpt :

      Previous studies have shown that IFN-γ differently modulates the sensitivity of melanoma cells to cytolytic T-cell clones recognizing distinct antigens, and does not increase melanoma killing by clones specific for differentiation antigens (Kirkin et al., 1996). However, we have recently shown that clinically relevant TIL products contain a variety of T cells specific not only for differentiation antigens but also for cancer-testis, mutation, and overexpressed tumor antigens (Andersen et al., 2012; Junker et al., 2012). Altogether, these data suggest that T-cell responses of TIL products might be increased with immuno-sensitizing agents.

    • Novel Immunotherapies for Osteosarcoma

      2022, Frontiers in Oncology
    View all citing articles on Scopus
    View full text