Elsevier

Cancer Treatment Reviews

Volume 41, Issue 10, December 2015, Pages 914-924
Cancer Treatment Reviews

New Drugs
Targeting immune checkpoints: New opportunity for mesothelioma treatment?

https://doi.org/10.1016/j.ctrv.2015.09.006Get rights and content

Highlights

  • Working mechanism and expression pattern of immune checkpoints.

  • Clinical data of immune checkpoint blocking antibodies are very promising.

  • Immune checkpoint blockade deserves further investigation in mesothelioma.

  • Future opportunities for research on immune inhibitory molecules.

Abstract

Malignant pleural mesothelioma is an aggressive cancer linked to asbestos exposure in most patients. Due to the long latency between exposure and presentation, incidence is expected to further increase in the next decade, despite the ban on asbestos import which occurred at the end of last century in industrialized countries. Platinum-based palliative chemotherapy is the only treatment with proven benefit on outcome, resulting in selected patients in a median overall survival of about 1 year. Therefore, there is room for therapeutic improvement using a new strategy to prolong survival. Dealing with cancer cell induced immunosuppression is a promising approach. Reactivating immune responses that are silenced by immune checkpoints recently gained a lot of interest. Checkpoint blockade has already shown promising preclinical and clinical results in several cancer types and is currently also being investigated in mesothelioma. Here, we discuss the expression patterns and mechanisms of action of CTLA-4 and PD-1 as the two most studied and of TIM-3 and LAG-3 as two interesting upcoming immune checkpoints. Furthermore, we review the clinical results of molecules blocking these immune checkpoints and point out their future opportunities with a special focus on mesothelioma.

Introduction

Malignant pleural mesothelioma (MPM) is an aggressive and nearly always fatal cancer, causally linked to previous, mostly professional, exposure to asbestos [1]. The highest incidence rates, around 30 cases per million inhabitants, are reported for Australia, Belgium and the UK [2], [3]. The incidence of MPM is still expected to increase over the next decades due to the long latency between exposure to asbestos and diagnosis and because asbestos is still being used in developing countries [4]. The prognosis of MPM patients remains poor with a median overall survival time in untreated patients of about 10 months and a 5 year survival rate of less than 5% [4], [5]. Palliative platinum–antifolate chemotherapy is the only treatment with proven improvement of outcome in MPM, resulting in a median survival of about 1 year. There is therefore a need for new therapeutic strategies. The discovery of immune checkpoint receptors such as cytotoxic T lymphocyte antigen-4 (CTLA-4) and more recently programmed death-1 (PD-1) introduced a new, exciting era in cancer immunotherapy [6]. Immune checkpoints are responsible for controlling and inactivating the immune system in order to avoid autoimmunity and prevent collateral tissue damage [7]. The new paradigm consists of reactivating silenced immune responses by neutralizing molecules that induce T-cell exhaustion and immune tolerance. Immune checkpoint blocking antibodies have already shown promising results in several cancer types [8], [9], [10], [11], [12], [13]. Recently antibodies blocking immune checkpoints are being investigated in mesothelioma patients. In this review, we discuss the expression pattern and mechanisms of action of CTLA-4 and PD-1 as the two most studied checkpoint receptors and of T-cell immunoglobulin mucin-3 (TIM-3) and lymphocyte activation gene-3 (LAG-3) as two interesting upcoming immune checkpoints. Furthermore, we review the clinical results of therapeutic molecules blocking these immune checkpoints with primary focus on CTLA-4 and PD-1 since FDA approved antibodies are available for both of them. Future opportunities of immune inhibitory molecules will be pointed out, with a special focus on MPM.

Section snippets

CTLA-4: The first clinically targeted immune checkpoint receptor

CTLA-4 is an immune inhibitory receptor that is mainly found on T-cells and to a lower extent on activated B-cells, monocytes, dendritic cells and granulocytes [14], [15], [16], [17]. Its primary role is to regulate T-cell activation upon antigenic stimulation of the T-cell receptor (TCR). T-cell activation can be explained by the two-signal model. The first signal is provided when an antigen, presented by an antigen presenting cell (APC) in combination with a major histocompatibility complex

Other interesting immune checkpoints: LAG-3 and TIM-3

Current research is mainly focused on CTLA-4, PD-1 and their ligands. However, also other molecules on the surface of T lymphocytes can exert inhibitory functions, such as LAG-3 and TIM-3. These two ‘neglected’ immune inhibitory molecules are now gaining more interest since they have been described to be related to T-cell tolerance and exhausted T-cells that are infiltrating the tumor microenvironment [61], [62], [63]. The hypothesis that also other molecules are involved in T-cell exhaustion

Clinical results of blocking antibodies

Different PD-1 blocking compounds are available, such as nivolumab (Opdivo®, BMS-936558, Bristol-Meyers Squibb), pembrolizumab (Keytruda®, MK-3475, Merck) and pidilizumab (Cure-Tech). The latter is the first PD-1 blocking agent that entered clinical trials [81]. It is a humanized (i.e., a mixed human and murine antibody) IgG1 monoclonal antibody, initially investigated during a phase I trial in patients with advanced hematological malignancies. Durable responses of more than 60 weeks were noted,

Immune checkpoints as biomarker

The value of PD-L1 as a prognostic biomarker has been addressed in many cancer types. Data on correlation between expression and overall survival remain controversial. Hino et al. [87] reported PD-L1 expression as a poor prognostic factor in malignant melanoma, while the opposite seems to be suggested by data from Gadiot et al. [88]. Similar discrepancy was also found for NSCLC, ovarian cancer and renal cell carcinoma [42], [89], [90], [91] and might be explained by the use of different sample

Immune checkpoints in mesothelioma: A new treatment opportunity?

Clinical evidence suggests that the immune system plays a critical role in protection against MPM [5], [94], [95]. TILs play an important role in anti tumor immune responses by recognizing tumor-specific antigens and T-cell infiltration has already been associated with a good prognosis in many cancers, such as ovarian and colon cancer. In MPM, high number of CD8+ TILs has shown to be beneficial for prognosis [96], [97], [98], while others reported no association between TILs and survival [99].

Discussion

Inhibiting immune checkpoints with blocking antibodies has already shown promising results in several cancer types [111] and encouraging clinical data with blocking CTLA-4, as well as expression data of PD-1 and PD-L1 in MPM support further investigation of immune checkpoint targeted therapy for MPM treatment. Thorough investigation of the expression of immune checkpoints, also the ‘neglected’ ones, in MPM and the effect of their blockade would help to identify new targets for immunotherapy and

Conclusion

PD-1 and PD-L1 were awarded as cancer breakthrough targets of the year 2013 evolving in a booming attention for immune checkpoints in oncology research. While MPM has interesting immunological features, only few studies until now have focused on this cancer. Research on the expression of immune checkpoints in mesothelioma might help to identify new biomarkers thereby improving patients selection and avoiding toxic side effects in non-responders. Better understanding of the mechanisms of action

Conflict of interest

The authors declare that there are no conflict of interests.

Acknowledgments

This work was performed with the support of the Belgian Foundation Against Cancer (Grant Number: FA/2014/263) and Research Foundation Flanders (Grant Number: 1510215N). E. Marcq is a research fellow of the Agency for Innovation through Science and Technology (fellowship number: 141433).

References (125)

  • M.J. Butte et al.

    Programmed death-1 ligand 1 interacts specifically with the B7-1 costimulatory molecule to inhibit T cell responses

    Immunity

    (2007 Jul)
  • P. Youngnak et al.

    Differential binding properties of B7-H1 and B7-DC to programmed death-1

    Biochem Biophys Res Commun

    (2003 Aug 1)
  • B. Merelli et al.

    Targeting the PD1/PD-L1 axis in melanoma: biological rationale, clinical challenges and opportunities

    Crit Rev Oncol Hematol

    (2014 Jan)
  • S. Mumprecht et al.

    Programmed death 1 signaling on chronic myeloid leukemia-specific T cells results in T-cell exhaustion and disease progression

    Blood

    (2009 Aug 20)
  • L. Zhang et al.

    PD-1/PD-L1 interactions inhibit antitumor immune responses in a murine acute myeloid leukemia model

    Blood

    (2009 Aug 20)
  • K.C. Ohaegbulam et al.

    Human cancer immunotherapy with antibodies to the PD-1 and PD-L1 pathway

    Trends Mol Med

    (2015 Jan)
  • A.J. Gehring et al.

    Profile of tumor antigen-specific CD8 T cells in patients with hepatitis B virus-related hepatocellular carcinoma

    Gastroenterology

    (2009 Aug)
  • Q. Zhou et al.

    Coexpression of Tim-3 and PD-1 identifies a CD8+ T-cell exhaustion phenotype in mice with disseminated acute myelogenous leukemia

    Blood

    (2011 Apr 28)
  • V. Velcheti et al.

    Programmed death ligand-1 expression in non-small cell lung cancer

    Lab Invest

    (2014 Jan)
  • V. Izzi et al.

    Immunity and malignant mesothelioma: from mesothelial cell damage to tumor development and immune response-based therapies

    Cancer Lett

    (2012 Sep 1)
  • M. Anraku et al.

    Impact of tumor-infiltrating T cells on survival in patients with malignant pleural mesothelioma

    J Thorac Cardiovasc Surg

    (2008 Apr)
  • H.S. Mudhar et al.

    No relationship between tumour infiltrating lymphocytes and overall survival is seen in malignant mesothelioma of the pleura

    Eur J Surg Oncol

    (2002 Aug)
  • C. Bianchi et al.

    Malignant mesothelioma: global incidence and relationship with asbestos

    Ind Health

    (2007 Jun)
  • C. Bianchi et al.

    Malignant mesothelioma in central and Eastern Europe

    Acta Med Croat

    (2000)
  • B.W. Robinson et al.

    Advances in malignant mesothelioma

    N Engl J Med

    (2005 Oct 13)
  • M. Bagia et al.

    Novel targeted therapies and vaccination strategies for mesothelioma

    Curr Treat Options Oncol

    (2011 Jun)
  • R.M. Wong et al.

    Immunotherapy for malignant pleural mesothelioma. Current status and future prospects

    Am J Respir Cell Mol Biol

    (2014 May)
  • O. Hamid et al.

    Safety and tumor responses with lambrolizumab (anti-PD-1) in melanoma

    N Engl J Med

    (2013 Jul 11)
  • E.J. Lipson et al.

    Durable cancer regression off-treatment and effective reinduction therapy with an anti-PD-1 antibody

    Clin Cancer Res

    (2013 Jan 15)
  • S.L. Topalian et al.

    Safety, activity, and immune correlates of anti-PD-1 antibody in cancer

    N Engl J Med

    (2012 Jun 28)
  • J.R. Brahmer et al.

    Safety and activity of anti-PD-L1 antibody in patients with advanced cancer

    N Engl J Med

    (2012 Jun 28)
  • E.A. van et al.

    Combination immunotherapy of B16 melanoma using anti-cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) and granulocyte/macrophage colony-stimulating factor (GM-CSF)-producing vaccines induces rejection of subcutaneous and metastatic tumors accompanied by autoimmune depigmentation

    J Exp Med

    (1999 Aug 2)
  • C.Y. Pico de et al.

    Myeloid-derived suppressor cells and their role in CTLA-4 blockade therapy

    Cancer Immunol Immunother

    (2014 Sep)
  • C. Pioli et al.

    Inhibition of IgG1 and IgE production by stimulation of the B cell CTLA-4 receptor

    J Immunol

    (2000 Nov 15)
  • X.B. Wang et al.

    Expression of CTLA-4 by human monocytes

    Scand J Immunol

    (2002 Jan)
  • W. Zou et al.

    Inhibitory B7-family molecules in the tumour microenvironment

    Nat Rev Immunol

    (2008 Jun)
  • X.B. Wang et al.

    Regulation of surface and intracellular expression of CTLA-4 on human peripheral T cells

    Scand J Immunol

    (2001 Nov)
  • B.C. Creelan

    Update on immune checkpoint inhibitors in lung cancer

    Cancer Control

    (2014 Jan)
  • F. Vanky et al.

    Human ex vivo carcinoma cells produce transforming growth factor beta and thereby can inhibit lymphocyte functions in vitro

    Cancer Immunol Immunother

    (1997 Jan)
  • E. Christeli et al.

    TGF-beta 1 overexpression in breast cancer

    Oncol Rep

    (1996 Nov)
  • L. Zitvogel et al.

    Cancer despite immunosurveillance: immunoselection and immunosubversion

    Nat Rev Immunol

    (2006 Oct)
  • P.A. van der Merwe et al.

    CD80 (B7-1) binds both CD28 and CTLA-4 with a low affinity and very fast kinetics

    J Exp Med

    (1997 Feb 3)
  • D.A. Mandelbrot et al.

    B7-dependent T-cell costimulation in mice lacking CD28 and CTLA4

    J Clin Invest

    (2001 Apr)
  • C.E. Rudd et al.

    CD28 and CTLA-4 coreceptor expression and signal transduction

    Immunol Rev

    (2009 May)
  • H.T. Kissick et al.

    Tumour eradication and induction of memory against murine mesothelioma by combined immunotherapy

    Immunol Cell Biol

    (2012 Sep)
  • S. Laurent et al.

    The engagement of CTLA-4 on primary melanoma cell lines induces antibody-dependent cellular cytotoxicity and TNF-alpha production

    J Transl Med

    (2013)
  • M.J. Selby et al.

    Anti-CTLA-4 antibodies of IgG2a isotype enhance antitumor activity through reduction of intratumoral regulatory T cells

    Cancer Immunol Res

    (2013 Jul)
  • N.J. Vogelzang et al.

    Phase III study of pemetrexed in combination with cisplatin versus cisplatin alone in patients with malignant pleural mesothelioma

    J Clin Oncol

    (2003 Jul 15)
  • Birgitta I. Hiddinga et al.

    Mesothelioma treatment: are we on target?

    J Adv Res

    (2014)
  • D.M. Pardoll

    The blockade of immune checkpoints in cancer immunotherapy

    Nat Rev Cancer

    (2012 Apr)
  • Cited by (0)

    1

    Shared senior authors.

    View full text