Elsevier

International Immunopharmacology

Volume 62, September 2018, Pages 29-39
International Immunopharmacology

Review
Immune checkpoint blockade therapy for cancer: An overview of FDA-approved immune checkpoint inhibitors

https://doi.org/10.1016/j.intimp.2018.06.001Get rights and content

Highlights

  • CTLA-4, PD-1, and PD-L1 are key negative regulators of anti-tumor T cell reactivity.

  • Inhibitors of CTLA-4, PD-1, and PD-L1 can enhance anti-tumor T cell reactivity.

  • Immune checkpoint blockade yields significant clinical benefit in cancer patients.

Abstract

Although T lymphocytes have long been appreciated for their role in the immunosurveillance of cancer, it has been the realization that cancer cells may ultimately escape a response from tumor-reactive T cells that has ignited efforts to enhance the efficacy of anti-tumor immune responses. Recent advances in our understanding of T cell immunobiology have been particularly instrumental in informing therapeutic strategies to overcome mechanisms of tumor immune escape, and immune checkpoint blockade has emerged as one of the most promising therapeutic options for patients in the history of cancer treatment. Designed to interfere with inhibitory pathways that naturally constrain T cell reactivity, immune checkpoint blockade releases inherent limits on the activation and maintenance of T cell effector function. In the context of cancer, where negative T cell regulatory pathways are often overactive, immune checkpoint blockade has proven to be an effective strategy for enhancing the effector activity and clinical impact of anti-tumor T cells. Checkpoint inhibitors targeting CTLA-4, PD-1, and PD-L1 have yielded unprecedented and durable responses in a significant percentage of cancer patients in recent years, leading to U.S. FDA approval of six checkpoint inhibitors for numerous cancer indications since 2011. In this review, we highlight the clinical success of these FDA-approved immune checkpoint inhibitors and discuss current challenges and future strategies that must be considered going forward to maximize the efficacy of immune checkpoint blockade therapy for cancer.

Introduction

Immunotherapy has recently emerged as a viable and attractive treatment option for many cancer patients. In particular, monoclonal antibody-based immune checkpoint blockade (ICB) therapies that enhance the function of anti-tumor T lymphocytes have been especially promising, yielding unprecedented results in the clinic. Based on these successes, there is now great optimism that ongoing work in immunooncology will further enhance the efficacy of these revolutionary cancer therapies, which have already led to durable remissions in many cancer patients.

The transition of cancer immunotherapy from hopeful vision to standard-of- care treatment for many tumor types has been driven by years of work in the fields of T cell biology and tumor immunology. Some of the first experimental evidence supporting a role for T cells in tumor immune surveillance was provided by seminal studies in murine models showing that either T lymphocytes or effector molecules released by these cells are critical to the control of spontaneous, inducible, or transplanted tumors [[1], [2], [3], [4], [5], [6], [7]]. Additionally, a role for T lymphocytes in tumor immune surveillance in humans was supported by clinical observations correlating spontaneous tumor regression with either autoimmune reactivity (i.e., melanoma regression in the context of vitiligo) [8] or the detection of tumor-associated CD4+ and CD8+ T cell responses in cancer patients [9, 10]. The significance of such T cell responses to the immunologic control of many tumor types was further highlighted by subsequent studies documenting the recovery of dysfunctional T cells from tumors and tumor-draining lymph nodes of patients with progressive disease [[11], [12], [13]]. During this time, the realization that T cell receptor engagement by itself is insufficient to promote T cell activation [14] propelled significant efforts to understand the nature of signals that regulate T cell responses to a source of antigen. In addition to the discovery that T cell activation requires co-stimulation and cytokine support as 2nd and 3rd signals over and above the “first signal” provided by antigenic stimulation through the T cell receptor [15], the identification of co-inhibitory signaling pathways that negatively regulate T cell activation provided key insights into the complex network of interactions that ultimately control the balance between T cell activation and tolerance [16]. While these co-inhibitory immune checkpoints act as fail-safes to prevent aberrant or chronic activation of the immune system and are thus critical to the maintenance of immune homeostasis, they may be co-opted by cancer cells or tumor-associated antigen-presenting cells (APC) as a means of dampening anti-tumor T cell responses and promoting tumor immune escape. Importantly, checkpoint blockade therapies that target these co-inhibitory pathways in the context of cancer effectively “release the brakes” on the immune system at both the induction and effector phases of anti-tumor T cell responses, thereby overcoming tumor immune subversion (Fig. 1). In this review, we highlight the current landscape of FDA-approved immune checkpoint inhibitors for cancer (Table 1), with emphasis on: 1) the ways in which ICB regimens have already revolutionized cancer immunotherapy and 2) the factors that must be considered going forward in order to maximize the potential of immune checkpoint inhibitors as anti-cancer therapeutic agents.

Section snippets

The CTLA-4 immune checkpoint

The first negative regulator of T cell activation to be identified was CTLA-4, a co-inhibitory receptor that is upregulated early during the course of T cell activation [17]. Bound by the same ligands (CD80/CD86) that provide co-stimulatory signals through CD28 on T lymphocytes, CTLA-4 conversely impedes acquisition of T cell effector function by 1) mediating trans-endocytosis and degradation of the ligands needed for co-stimulation [18] and 2) delivering inhibitory signals that block T cell

The PD-1 immune checkpoint

Though first discovered prior to CTLA-4 in 1992 as a molecule associated with programmed T cell death, PD-1 was not appreciated as a co-inhibitory receptor that negatively regulates T cell effector function until several years later, when autoimmune disease was observed in transgenic mice harboring PD-1−/− T cells [30]. At the same time, PD-L1 and PD-L2 were discovered as dual ligands for PD-1, and both were shown to inhibit T cell effector activity following PD-1 engagement [31, 32]. Although

Combinatorial blockade of the CTLA-4 and PD-1 immune checkpoint pathways

The clinical benefits of checkpoint blockade therapies targeting either the CTLA-4 or PD-1 pathways in cancer patients, along with the realization that these checkpoint pathways each regulate T cell function by distinct mechanisms that act on particular T cell subsets and at different stages of T cell differentiation, have prompted the investigation of combinatorial ICB regimens in the treatment of cancer. Combinatorial therapy with ipilimumab and nivolumab has led to unprecedented responses in

Toxicities associated with immune checkpoint blockade therapy for cancer

In addition to the clinical benefits that have been achieved thus far with ICB therapy, another striking observation that has emerged from the early trials with immune checkpoint inhibitors is the undesirable (and previously unforeseen) side effects that can result from their use. Indeed, the fact that immune checkpoint inhibitors are not directed solely to tumor-specific T cells means that the ability of these drugs to unleash desirable anti-tumor immune reactivity may be accompanied by the

Current challenges to immune checkpoint blockade therapy for cancer

In order to maximize both the efficacy and reach of ICB therapy for cancer patients, many challenges in this field must be addressed. In particular, it is necessary to gain insight into factors that influence response outcomes to ICB therapy and better understand why some patients exhibit complete and durable responses that prolong survival while others experience disease relapse within the first few months of treatment or fail to respond to ICB therapy at all. While efficacy varies by cancer

The future of immune checkpoint blockade therapy for cancer

Despite the challenges still being faced in the field of ICB therapy, the success of this approach in the treatment of a broad spectrum of cancers over the last decade has highlighted just how significant a role the immune system plays in the control of tumors, and historic outcomes in several clinical trials evaluating the efficacy of ICB regimens have reinvigorated the field of cancer immunotherapy. Marked by FDA approval of 6 immune checkpoint inhibitors in the last 7 years, the current

Funding

Research in the Hargadon laboratory is funded by a Jeffress Trust Awards Program in Interdisciplinary Research Grant from the Thomas F. and Kate Miller Jeffress Memorial Trust, Bank of America, N.A., Trustee, a Hampden-Sydney College Research Grant from the Arthur Vining Davis endowment, and generous donations from Mr. Michael Hargadon and Mrs. Patricia Hargadon to support the involvement of undergraduate students in research at Hampden-Sydney College. These funding sources were not involved in

Declarations of interest

None.

References (100)

  • A.B. El-Khoueiry et al.

    Nivolumab in patients with advanced hepatocellular carcinoma (CheckMate 040): an open-label, non-comparative, phase 1/2 dose escalation and expansion trial

    Lancet

    (2017)
  • M.J. Overman et al.

    Nivolumab in patients with metastatic DNA mismatch repair-deficient or microsatellite instability-high colorectal cancer (CheckMate 142): an open-label, multicentre, phase 2 study

    Lancet Oncol.

    (2017)
  • A. Younes et al.

    Nivolumab for classical Hodgkin's lymphoma after failure of both autologous stem-cell transplantation and brentuximab vedotin: a multicentre, multicohort, single-arm phase 2 trial

    Lancet Oncol.

    (2016)
  • A. Ribas et al.

    Pembrolizumab versus investigator-choice chemotherapy for ipilimumab-refractory melanoma (KEYNOTE-002): a randomised, controlled, phase 2 trial

    Lancet Oncol.

    (2015)
  • J. Schachter et al.

    Pembrolizumab versus ipilimumab for advanced melanoma: final overall survival results of a multicentre, randomised, open-label phase 3 study (KEYNOTE-006)

    Lancet

    (2017)
  • R.S. Herbst et al.

    Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced non-small-cell lung cancer (KEYNOTE-010): a randomised controlled trial

    Lancet

    (2016)
  • T. Powles et al.

    Atezolizumab versus chemotherapy in patients with platinum-treated locally advanced or metastatic urothelial carcinoma (IMvigor211): a multicentre, open-label, phase 3 randomised controlled trial

    Lancet

    (2018)
  • A. Rittmeyer et al.

    Atezolizumab versus docetaxel in patients with previously treated non-small-cell lung cancer (OAK): a phase 3, open-label, multicentre randomised controlled trial

    Lancet

    (2017)
  • M.R. Patel et al.

    Avelumab in metastatic urothelial carcinoma after platinum failure (JAVELIN Solid Tumor): pooled results from two expansion cohorts of an open-label, phase 1 trial

    Lancet Oncol.

    (2018)
  • H.L. Kaufman et al.

    Avelumab in patients with chemotherapy-refractory metastatic Merkel cell carcinoma: a multicentre, single-group, open-label, phase 2 trial

    Lancet Oncol.

    (2016)
  • M.D. Hellmann et al.

    Nivolumab plus ipilimumab as first-line treatment for advanced non-small-cell lung cancer (CheckMate 012): results of an open-label, phase 1, multicohort study

    Lancet Oncol.

    (2017)
  • S.J. Antonia et al.

    Nivolumab alone and nivolumab plus ipilimumab in recurrent small-cell lung cancer (CheckMate 032): a multicentre, open-label, phase 1/2 trial

    Lancet Oncol.

    (2016)
  • G.V. Long et al.

    Standard-dose pembrolizumab in combination with reduced-dose ipilimumab for patients with advanced melanoma (KEYNOTE-029): an open-label, phase 1b trial

    Lancet Oncol.

    (2017)
  • S. Antonia et al.

    Safety and antitumour activity of durvalumab plus tremelimumab in non-small cell lung cancer: a multicentre, phase 1b study

    Lancet Oncol.

    (2016)
  • B. El Osta et al.

    Not all immune-checkpoint inhibitors are created equal: meta-analysis and systematic review of immune-related adverse events in cancer trials

    Crit. Rev. Oncol. Hematol.

    (2017)
  • J.M. Pitt et al.

    Resistance mechanisms to immune-checkpoint blockade in cancer: tumor-intrinsic and -extrinsic factors

    Immunity

    (2016)
  • G.T. Gibney et al.

    Predictive biomarkers for checkpoint inhibitor-based immunotherapy

    Lancet Oncol.

    (2016)
  • N. Chaput et al.

    Baseline gut microbiota predicts clinical response and colitis in metastatic melanoma patients treated with ipilimumab

    Ann. Oncol.

    (2017)
  • J. Gao et al.

    Loss of IFN-γ pathway genes in tumor cells as a mechanism of resistance to anti-CTLA-4 therapy

    Cell

    (2016)
  • D.H. Munn et al.

    Immune suppressive mechanisms in the tumor microenvironment

    Curr. Opin. Immunol.

    (2016)
  • H.I. Levitsky et al.

    In vivo priming of two distinct antitumor effector populations: the role of MHC class I expression

    J. Exp. Med.

    (1994)
  • M.E. van den Broek et al.

    Decreased tumor surveillance in perforin-deficient mice

    J. Exp. Med.

    (1996)
  • D.H. Kaplan et al.

    Demonstration of an interferon gamma-dependent tumor surveillance system in immunocompetent mice

    Proc. Natl. Acad. Sci. U. S. A.

    (1998)
  • K. Hung et al.

    The central role of CD4(+) T cells in the antitumor immune response

    J. Exp. Med.

    (1998)
  • M.J. Smyth et al.

    Perforin-mediated cytotoxicity is critical for surveillance of spontaneous lymphoma

    J. Exp. Med.

    (2000)
  • V. Shankaran et al.

    IFNgamma and lymphocytes prevent primary tumour development and shape tumour immunogenicity

    Nature

    (2001)
  • J.L. Smith et al.

    Spontaneous regression of primary malignant melanomas with regional metastases

    Cancer

    (1965)
  • G.M. Halliday et al.

    Spontaneous regression of human melanoma/nonmelanoma skin cancer: association with infiltrating CD4+ T cells

    World J. Surg.

    (1995)
  • F.H. Saleh et al.

    Primary melanoma tumour regression associated with an immune response to the tumour-associated antigen melan-A/MART-1

    Int. J. Cancer

    (2001)
  • R. Mortarini et al.

    Lack of terminally differentiated tumor-specific CD8+ T cells at tumor site in spite of antitumor immunity to self-antigens in human metastatic melanoma

    Cancer Res.

    (2003)
  • L. Deng et al.

    Accumulation of foxp3+ T regulatory cells in draining lymph nodes correlates with disease progression and immune suppression in colorectal cancer patients

    Clin. Cancer Res.

    (2010)
  • J.J. Severson et al.

    PD-1+Tim-3+ CD8+ T lymphocytes display varied degrees of functional exhaustion in patients with regionally metastatic differentiated thyroid cancer

    Cancer Immunol. Res.

    (2015)
  • A. Shahinian et al.

    Differential T cell costimulatory requirements in CD28-deficient mice

    Science

    (1993)
  • O. Joffre et al.

    Inflammatory signals in dendritic cell activation and the induction of adaptive immunity

    Immunol. Rev.

    (2009)
  • L. Chen et al.

    Molecular mechanisms of T cell co-stimulation and co-inhibition

    Nat. Rev. Immunol.

    (2013)
  • O.S. Qureshi et al.

    Trans-endocytosis of CD80 and CD86: a molecular basis for the cell-extrinsic function of CTLA-4

    Science

    (2011)
  • M.F. Krummel et al.

    CD28 and CTLA-4 have opposing effects on the response of T cells to stimulation

    J. Exp. Med.

    (1995)
  • M.F. Krummel et al.

    CTLA-4 engagement inhibits IL-2 accumulation and cell cycle progression upon activation of resting T cells

    J. Exp. Med.

    (1996)
  • C.A. Chambers et al.

    Secondary but not primary T cell responses are enhanced in CTLA-4-deficient CD8+ T cells

    Eur. J. Immunol.

    (1998)
  • K. Wing et al.

    CTLA-4 control over Foxp3+ regulatory T cell function

    Science

    (2008)
  • Cited by (819)

    • The BTLA-HVEM complex – The future of cancer immunotherapy

      2024, European Journal of Medicinal Chemistry
    • Peptide nanovaccine in melanoma immunotherapy

      2024, International Immunopharmacology
    View all citing articles on Scopus
    View full text