Elsevier

The Lancet

Volume 373, Issue 9664, 21–27 February 2009, Pages 673-683
The Lancet

Review
Use of tumour-responsive T cells as cancer treatment

https://doi.org/10.1016/S0140-6736(09)60404-9Get rights and content

Summary

The stimulation of a tumour-specific T-cell response has several theoretical advantages over other forms of cancer treatment. First, T cells can home in to antigen-expressing tumour deposits no matter where they are located in the body—even in deep tissue beds. Additionally, T cells can continue to proliferate in response to immunogenic proteins expressed in cancer until all the tumour cells are eradicated. Finally, immunological memory can be generated, allowing for eradication of antigen-bearing tumours if they reoccur. We will highlight two direct methods of stimulating tumour-specific T-cell immunity: active immunisation with cancer vaccines and infusion of competent T cells via adoptive T-cell treatment. Preclinical and clinical studies have shown that modulation of the tumour microenvironment to support the immune response is as important as stimulation of the most appropriate effector T cells. The future of T-cell immunity stimulation to treat cancer will need combination approaches focused on both the tumour and the T cell.

Introduction

T lymphocytes can be found infiltrating human tumours and have been associated with both an improved or poor prognosis. Investigations suggest that this contradiction can be explained by close analysis of the phenotype of the tumour-infiltrating T lymphocyte. T cells can be broadly classified as cytotoxic CD8+ T cells, which can directly kill an antigen-expressing cell or cytokine-secreting CD4+ T cells. The CD4+ T-cell response can elicit both immune stimulatory or immune inhibitory effects. Specific CD4+ T-helper (Th) cell phenotypes are crucial for the expansion and persistence of tissue-destructive CD8+ T cells. Th1 CD4+ T cells secrete type I cytokines such as interferon (IFN) γ, resulting in the activation of antigen-presenting cells, which stimulate a CD8+ T-cell response.1 Th2 CD4+ T-cells secrete type II cytokines, such as interleukin 4 (IL4), in response to antigen. Th2 CD4+ T cells can limit the activation of antigen-presenting cells and enhance humoral immunity as well as the influx of innate immune cells such as eosinophils and granulocytes.2 The newly identified Th17 CD4+ T cell secretes IL17, eliciting tissue inflammation implicated in autoimmunity.3 Finally, CD4+FOXP3+ T regulatory (Treg) cells will inhibit the development of an adaptive T-cell response directed against self-molecules, especially self-tumour antigens, via secretion of immunosuppressive cytokines such as IL10 or direct inhibition of antigen-presenting cells.4

The interplay of specific T-cell phenotypes is highlighted in an analysis5 of more than 400 colon cancers for tumour-infiltrating lymphocytes (TIL). With tissue microarrays, investigators showed that a high density of CD8+ effector memory T cells in the tumour parenchyma was associated with increased survival. In a more detailed study of gene-expression patterns in a subset of tumours from 75 of the patients with colon cancer,6 data suggested that an upregulation of genes related to Th1 adaptive immune response was associated with a decreased risk of relapse.6

Conversely, the presence of Treg cells seems to correlate with a poor prognosis in several tumour types in large population-based studies. After assessing tumours in more than 300 patients with hepatocellular carcinoma, investigators showed both improved disease-free and overall survival in patients whose tumours had low numbers of Treg cells and high numbers of activated CD8+ T cells, measured by granzyme B staining, compared with patients whose tumours had high numbers of Treg cells and low numbers of activated CD8+ T cells.7 Intratumoural Treg cells were assessed by immunohistochemical staining in more than 200 patients with invasive breast cancer.8 Individuals with high numbers of Treg cells in their tumours had a shorter relapse-free and overall survival than did those with low Treg-cell infiltrate. These population-based analyses, using large numbers of well-defined cases, provide new insight into the role of T cells in cancer progression.

Direct evidence that tumour-specific T cells can induce an antitumour response is shown by the infusion of T cells used to treat patients with cancer. Donor lymphocyte infusions, used once patients with haematological malignant diseases have relapsed after allogeneic transplantation, have become a standard of care resulting in durable complete remissions in many individuals.9, 10 The clinical response is presumably due to a graft-versus-tumour effect.11 Infusion of tumour-specific T cells in solid tumours has also met with some success. Transfer of autologous T cells derived from TIL resulted in a 51% response in 35 treated patients with refractory metastatic melanoma.12 Although responses were not durable, evidence of substantial immunological remodelling of the tumour was seen, resulting in immune escape.

Unfortunately, tumours possess many mechanisms to escape immune recognition. Most defined cancer-associated immunogenic proteins, or tumour antigens, are non-mutated self-proteins; thus, systemic tolerance is a major mechanism of tumour immune escape. As described, Treg cells limit inflammation and prevent autoimmunity and are important in inhibiting self-antitumour immune responses. Other forms of peripheral tolerance include deletion, ignorance, and anergy. Deletion in the periphery is similar to central deletion, in which a cell expressing a T-cell receptor with high affinity for a self-antigen is deleted. When self-reactive T cells encounter self-antigen, the T cells can upregulate the death receptor Fas, resulting in T-cell apoptosis.13 Ignorance is thought to occur when self-reactive T cells persist but are not activated by the antigen. T cells can also remain ignorant of an antigen if the antigen is not easily accessible to the peripheral blood or lymphatic system.14 Anergy is a state of unresponsiveness, and is traditionally thought to occur when T-cell-receptor ligation happens in the absence of costimulation.15 Immune receptor and costimulatory molecules have often been downregulated or lost in tumour cells.16

Tumours directly produce a local suppressive milieu that affects the activity of the infiltrating immune cells. For instance, tumours can downregulate various factors in antigen processing (eg, MHC molecules).17 Furthermore, tumours can secrete inhibitory cytokines, such as IL10 and transforming growth factor (TGF) β, which can aid recruitment of Treg cells and inhibition of dendritic-cell maturation.18 Many tumours also express ligands that can interact with infiltrating T cells to provide negative stimulation, which inhibits or reduces the effector functions of specific cytotoxic T lymphocytes.19, 20 The tumour stroma can also limit the therapeutic efficacy of activated T cells by acting as a physical barrier as well as by elaborating cytokines and other soluble factors that promote cell growth and remodelling.21 Immune-based treatments will probably be most effective at low or non-detectable levels of tumour burden.

Section snippets

Human tumour antigens

Tumour antigens have been identified in nearly every human cancer, by virtue of these proteins being immunogenic in patients and not in volunteer controls. The development of highly quantitative assays to measure antigen-specific T cells allows a precise assessment of the endogenous tumour-associated T-cell response in patients. Patients with melanoma have been reported to have endogenous tumour-specific T-cell precursor frequencies as robust as 1:1000 CD8+ T cells.22 Moreover, the numbers of

Tumour-cell-based vaccines

A benefit of whole-cell tumour vaccines is that many tumour-associated antigens (known and unknown) are simultaneously delivered, eliminating the need to predetermine which antigens are the most immunogenic and mediate tumour rejection.63 In the classic antigen-processing pathway, extracellular proteins are presented on MHC class II molecules to CD4+ T cells, whereas intracellular antigens are presented on MHC class I molecules to CD8+ T cells.64, 65 However, evidence has shown that exogenous

Adoptive T-cell treatment

The main purpose of adoptive T-cell treatment is to augment a tumour-specific T-cell response above what is possible by vaccination alone. Infusion of tumour-competent T cells could allow levels of immunity to be capable of eradicating established disease. Anergic T cells can be rescued if removed from the tolerising milieu and stimulated ex vivo.86, 87 The rationale for adoptive T-cell transfer is based on attempts to circumvent or break tolerance using ex-vivo stimulated autologous T

Future directions

Many compounds can modulate the environment and be useful in the development of combination immunotherapy to elicit tumour-specific immunity. For example, the in-vivo administration of an antibody can be used to block CTLA4 (cytotoxic T-lymphocyte-associated antigen 4), an inhibitory receptor that dampens the ability of T cells to respond to antigen. The use of CTLA4 monoclonal antibodies alone can result in a 14% clinical response rate in advanced-stage melanoma.120 However, removal of natural

Search strategy and selection criteria

We searched Medline (1990–2008) using search terms such as “T cell” and “cellular immunity” in combination with terms such as “cancer”, “tumor”, and “immune therapy”. We largely selected publications from the past 5 years, but did not exclude highly regarded older publications. We also searched the reference lists of articles identified by this search strategy and selected those we judged relevant. Review articles are cited to provide readers with more details and references if needed.

References (129)

  • MJ Smyth et al.

    Cancer immunosurveillance and immunoediting: the roles of immunity in suppressing tumor development and shaping tumor immunogenicity

    Adv Immunol

    (2006)
  • PJ Lehner et al.

    Processing and delivery of peptides presented by MHC class I molecules

    Curr Opin Immunol

    (1996)
  • D Jocham et al.

    Adjuvant autologous renal tumour cell vaccine and risk of tumour progression in patients with renal-cell carcinoma after radical nephrectomy: phase III, randomised controlled trial

    Lancet

    (2004)
  • RA Kroczek et al.

    Emerging paradigms of T-cell co-stimulation

    Curr Opin Immunol

    (2004)
  • C Palena et al.

    Potential approach to immunotherapy of chronic lymphocytic leukemia (CLL): enhanced immunogenicity of CLL cells via infection with vectors encoding for multiple costimulatory molecules

    Blood

    (2005)
  • SR Riddell et al.

    The use of anti-CD3 and anti-CD28 monoclonal antibodies to clone and expand human antigen-specific T cells

    J Immunol Methods

    (1990)
  • F Ossendorp et al.

    Specific T helper cell requirement for optimal induction of cytotoxic T lymphocytes against major histocompatibility complex class II negative tumors

    J Exp Med

    (1998)
  • JI Ellyard et al.

    Th2-mediated anti-tumour immunity: friend or foe?

    Tissue Antigens

    (2007)
  • C Dong

    Diversification of T-helper-cell lineages: finding the family root of IL-17-producing cells

    Nat Rev Immunol

    (2006)
  • W Zou

    Regulatory T cells, tumour immunity and immunotherapy

    Nat Rev Immunol

    (2006)
  • F Pages et al.

    Effector memory T cells, early metastasis, and survival in colorectal cancer

    N Engl J Med

    (2005)
  • J Galon et al.

    Type, density, and location of immune cells within human colorectal tumors predict clinical outcome

    Science

    (2006)
  • Q Gao et al.

    Intratumoral balance of regulatory and cytotoxic T cells is associated with prognosis of hepatocellular carcinoma after resection

    J Clin Oncol

    (2007)
  • GJ Bates et al.

    Quantification of regulatory T cells enables the identification of high-risk breast cancer patients and those at risk of late relapse

    J Clin Oncol

    (2006)
  • HM Lokhorst et al.

    Donor lymphocyte infusions for relapsed multiple myeloma after allogeneic stem-cell transplantation: predictive factors for response and long-term outcome

    J Clin Oncol

    (2000)
  • B Sprangers et al.

    Experimental and clinical approaches for optimization of the graft-versus-leukemia effect

    Nat Clin Pract Oncol

    (2007)
  • ME 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)
  • J Alferink et al.

    Control of neonatal tolerance to tissue antigens by peripheral T cell trafficking

    Science

    (1998)
  • RH Schwartz

    T cell anergy

    Sci Am

    (1993)
  • FM Marincola et al.

    Escape of human solid tumors from T-cell recognition: molecular mechanisms and functional significance

    Adv Immunol

    (2000)
  • HL Kaufman et al.

    Immune system versus tumor: shifting the balance in favor of DCs and effective immunity

    J Clin Invest

    (2004)
  • H Dong et al.

    B7-H1 pathway and its role in the evasion of tumor immunity

    J Mol Med

    (2003)
  • C Germeau et al.

    High frequency of antitumor T cells in the blood of melanoma patients before and after vaccination with tumor antigens

    J Exp Med

    (2005)
  • C Lurquin et al.

    Contrasting frequencies of antitumor and anti-vaccine T cells in metastases of a melanoma patient vaccinated with a MAGE tumor antigen

    J Exp Med

    (2005)
  • GV Yamshchikov et al.

    Sequential immune escape and shifting of T cell responses in a long-term survivor of melanoma

    J Immunol

    (2005)
  • M Inokuma et al.

    Functional T cell responses to tumor antigens in breast cancer patients have a distinct phenotype and cytokine signature

    J Immunol

    (2007)
  • A Banerjea et al.

    Immunogenic hsp-70 is overexpressed in colorectal cancers with high-degree microsatellite instability

    Dis Colon Rectum

    (2005)
  • V Goodell et al.

    Level of HER-2/neu protein expression in breast cancer may affect the development of endogenous HER-2/neu-specific immunity

    Mol Cancer Ther

    (2008)
  • DJ Loftus et al.

    Identification of epitope mimics recognized by CTL reactive to the melanoma/melanocyte-derived peptide MART-1(27–35)

    J Exp Med

    (1996)
  • U Behn

    Idiotypic networks: toward a renaissance?

    Immunol Rev

    (2007)
  • K Falk et al.

    Allele-specific motifs revealed by sequencing of self-peptides eluted from MHC molecules

    Nature

    (1991)
  • R Cibotti et al.

    Tolerance to a self-protein involves its immunodominant but does not involve its subdominant determinants

    Proc Natl Acad Sci USA

    (1992)
  • EE Sercarz et al.

    Dominance and crypticity of T cell antigenic determinants

    Annu Rev Immunol

    (1993)
  • MR Parkhurst et al.

    Improved induction of melanoma-reactive CTL with peptides from the melanoma antigen gp100 modified at HLA-A*0201-binding residues

    J Immunol

    (1996)
  • JW Smith et al.

    Adjuvant immunization of HLA-A2-positive melanoma patients with a modified gp100 peptide induces peptide-specific CD8+ T-cell responses

    J Clin Oncol

    (2003)
  • DE Speiser et al.

    Rapid and strong human CD8+ T cell responses to vaccination with peptide, IFA, and CpG oligodeoxynucleotide 7909

    J Clin Invest

    (2005)
  • KL Knutson et al.

    Immunization of cancer patients with a HER-2/neu, HLA-A2 peptide, p369–377, results in short-lived peptide-specific immunity

    Clin Cancer Res

    (2002)
  • PA Cohen et al.

    CD4+ T cells in adoptive immunotherapy and the indirect mechanism of tumor rejection

    Crit Rev Immunol

    (2000)
  • P van der Bruggen et al.

    Processing and presentation of tumor antigens and vaccination strategies

    Curr Opin Immunol

    (2006)
  • KL Knutson et al.

    Immunization with a HER-2/neu helper peptide vaccine generates HER-2/neu CD8 T-cell immunity in cancer patients

    J Clin Invest

    (2001)
  • Cited by (146)

    • Angiogenic biomolecules specific nanobodies application in cancer imaging and therapy; review and updates

      2022, International Immunopharmacology
      Citation Excerpt :

      According to the latest achievements in cancer immunotherapy, the augmentation of host’s antitumor immune response is highly recommended for the treatment of different types of carcinomas [215]. As well, the administration of those agents capable of provoking cytotoxic T lymphocytes proliferation (for instance, low doses of anti-CD3 antibody) is known as one of main approaches used to achieve augmentation in anticancer immune response [216]. Besides, angiogenesis may also be blocked during tumor overgrowth via the administration of immunomodulatory agents, which is a procedure referred to as ‘anti-angiogenic surveillance’ [217].

    View all citing articles on Scopus
    View full text