Elsevier

Cancer Treatment Reviews

Volume 63, February 2018, Pages 122-134
Cancer Treatment Reviews

Anti-Tumour Treatment
Checkpoint inhibitors in breast cancer – Current status

https://doi.org/10.1016/j.ctrv.2017.12.008Get rights and content

Highlights

  • The overall response rate of PD-1/PD-L1 monotherapy varied from 5 to 30% in heavily pretreated TNBC.

  • Responses were of long duration in a subset of patients.

  • Median PFS and OS were either not reported or short.

  • In the preoperative setting preliminary results suggest that chemotherapy + CPI increase pCR.

  • The identification of predictive biomarkers is crucial.

Abstract

Introduction

An increasing number of compounds directed against immune checkpoints are currently under clinical development. In this review we summarize current research in breast cancer.

Material and methods

A computer-based literature search was carried out using PubMed and EMBASE; data reported at international meetings and clinicaltrials.gov were included as well.

Results

The obtained overall response rate of PD-1/PD-L1 monotherapy varied from 5 to 30% in heavily pretreated triple negative breast cancer (TNBC). The median duration of progression free survival and overall survival were either not reported or short. Still responses were of long duration in a subset of patients. In the neoadjuvant setting, preliminary results including a very limited number of patients suggest high pathological response rates after combined blockade of the PD-1 pathway and chemotherapy. Multiple trials have been initiated to evaluate the combination of other anticancer agents and checkpoint inhibitors, especially in TNBC. In addition, ongoing studies aim to identify biomarkers to guide patient selection.

Conclusion

Immune checkpoint inhibitors have the potential to produce durable tumor remission and induce long standing anti-tumor immunity in a subgroup of breast cancer patients. However, the identification of predictive biomarkers is crucial for further development of this treatment modality.

Introduction

The overall survival rate for patients with breast cancer (BC) has improved in the past three decades, because of advances in cancer detection and locoregional therapy as well as improvements in systemic treatment i.e., chemotherapy, endocrine therapy, and new molecular targeted drugs. Due to these advances, the 5-year, 10-year, and 15-year relative survival rates for BC are 89%, 83%, and 78%, respectively [1].

Recent advances in immunotherapy in other cancer types e.g. melanoma and non-small cell lung cancer highlight the potential for immunotherapy [2], [3]. BC has historically been considered immunologically silent [4]. Nonetheless, a robust body of literature now suggests that BC, particularly the more aggressive subtypes, does elicit host antitumor immune responses, and that the robustness of responses correlate with prognosis [5]. In triple negative breast cancer (TNBC), substantial evidence shows that the presence of tumor infiltrating lymphocytes (TILs) is associated with improved overall survival (OS) [6] and increased metastasis free survival [7], [8]. Meta-analyses have shown that TILs were significantly correlated with a favorable prognosis in TNBC [9]. In addition, the presence of PD-L1 is significantly associated with response to immunotherapy [10] and TNBC frequently overexpresses programmed death-ligand protein (PD-L1) [11].

The most striking evidence of effective immunotherapy in BC has been the development of the monoclonal antibody trastuzumab directed against the human epidermal growth factor receptor 2 (HER2) protein. In addition to targeting the kinase signaling pathway, trastuzumab also functions via recruitment of natural killer (NK) cells and activation of antibody-dependent cytotoxicity (ADCC) [12]. Finally, chemotherapeutics as anthracyclines, platinum-based agents and cyclophosphamide may induce immunogenic cell death [13], [14].

Therefore, there is great interest in exploring the potential role of immunotherapy in patients with BC [15], [16].

The basal-like and HER2-enriched subtypes have been found to harbor higher genomic instability and be more immunogenic than luminal A carcinomas [5]. Emerging evidence indicates that genomic instability and thus a higher mutational load causes production of higher tumor-specific antigen levels eliciting stronger immune responses [17], [18], [19]. In TNBC, multiple studies suggest an important influence of host anti-tumor immunity on response to chemotherapy and prognosis making this subtype most relevant for investigating immunotherapy [7], [19], [20], [21], [22], [23].

In HER2-positive patients receiving neoadjuvant treatment with taxanes and trastuzumab, a positive correlation between number of NK cells and pCR has been demonstrated [24]. More recently, Luen et al. [19] in a secondary analysis of the CLEOPATRA study evaluating docetaxel + trastuzumab + pertuzumab/placebo in HER2-positive advanced BC found a positive correlation between number of TILs and OS. No correlation to progression free survival (PFS) was found [19]. These studies may provide a rationale for integrating immunotherapies into the care of HER2-positive BC [25]. The rational for immunotherapies in ER-positive BC cancer is very limited [15].

Under normal physiologic conditions, a plethora of suppressive pathways in the immune system exist to promote self-tolerance and protect against autoimmunity [26]. Tumors have been shown to exploit these pathways in order to prevent antitumor responses and escape immune detection and elimination [27], [28]. The pathways are highly regulated by immune checkpoints. The two immune-checkpoint receptors that have been most studied in the context of clinical cancer immunotherapy, cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) and programmed cell death protein 1 (PD-1) are both inhibitory receptors. CTLA-4 is expressed on T cells where it primarily regulates the amplitude of the early stages of T cell activation by down-modulating the activity of effector T cells (Teff cells) and enhancing the immunosuppressive T regulatory cells (Tregs) [27]. PD-1 is also expressed on T cells and signals through the PD-1 pathway limit T cell activation and effector T cell responses and, thus, immune responses within the tumor microenvironment [29]. Immune checkpoint inhibitors are antibodies against CTLA-4, PD-1 or its ligand (PD-L1). The inhibitors modulate the interaction between tumor cells and cytotoxic T lymphocytes which are thought to be exhausted in their function [30]. Targeting CTLA-4 or PD-1/PD-L1 reverses the exhaustion of cytotoxic T lymphocytes leading to elimination of tumor cells via re-induction of the “natural” function of the T cell population [31].

The use of immune checkpoint inhibitors (CPIs) is a promising anticancer therapeutic strategy which aims at blocking specific immune regulatory checkpoints to enhance the endogenous antitumor immunity. The present review summarizes data on clinical studies of CPIs in BC.

Two monoclonal antibodies that inhibit CTLA-4 are currently either approved by The United States Food and Drug Administration (FDA) or under investigation. Ipilimumab, a fully humanized monoclonal IgG1 antibody (Supplementary Table 1) has been approved both as monotherapy and in combination with nivolumab for treatment of malignant melanoma [2], [32], [33], [34]. Tremelimumab, a fully human IgG2 monoclonal antibody has been tested in a variety of tumor types [35] (Supplementary Table 1).

PD-1 pathway signaling is a result of binding to the inhibitory surface receptor PD-1 expressed on immune cells such as T, B and NK cells. Its ligands PD-L1/B7-H1 and PD-L2/B7-DC are displayed on cancer cells, and antigen-presenting cells such as activated monocytes and dendritic cells [27]. Two PD-1 inhibitors, nivolumab and pembrolizumab, are investigated in BC (Supplementary Table 1).

PD-L1 is the best characterized of the two known PD-1 ligands, PD-L1 and PD-L2. It can be expressed by tumor cells as well as by T and B cells, macrophages and dendritic cells [27]. Targeting PD-L1 may result in different biological effects than targeting PD-1. In addition to binding PD-1, PD-L1 exerts negative signals on T cells by interaction with B7 [36]. PD-L1 antibodies prevent this interaction. In addition, PD-L1 antibodies do not prevent PD-1 from interaction with PD-L2 - although the significance of this interaction remains unknown [37]. Altogether five PD-L1 inhibitors are evaluated in BC (Supplementary Table 1). Among these, atezolizumab (MPDL3280A), avelumab (MSB0010718C) and durvalumab (MEDI4736) have been approved for other cancer types.

Section snippets

Search strategy

Articles included in this review were retrieved by searching PubMed (1966–2017), EMBASE (1980–2017), American Society of Clinical Oncology Annual Meeting (ASCO) (2013–2017) and San Antonio Breast Cancer Symposium (SABCS) (2013–2017). We (AP and DN) searched for studies using the following search terms: “checkpoint inhibitor” and “breast cancer” (PubMed 383, EMBASE 70). In addition, we searched for the specific drugs (Supplementary Table 1) and “breast cancer”: ipilimumab (PubMed 18; EMBASE

Reported studies with CTLA-4 inhibitors in early breast cancer

Studies including CTLA-4 directed therapy in BC are given in Table 1. Only one pilot study on ipilimumab monotherapy in early stage BC has been reported [38]. Nineteen women with primary BC for whom a mastectomy was planned were treated preoperatively with cryoablation, a single dose of ipilimumab or both modalities. Ipilimumab and especially combination therapy were found to be associated with increased Th1-cytokine production, peripheral T-cell proliferation/activation and intratumoral

Discussion

Few clinical trials on monotherapy demonstrated modest efficacy of PD-L1/PD-1 blockade in primarily TNBC [41], [44], [47], [49], [50]. Anyhow, response rates have been potentially better than one might expect and interestingly, when responses occurred, they were often durable. In the preoperative setting preliminary encouraging results suggest that the combination of chemotherapy and CPI might significantly increase pCR and thus have the potential to increase survival [64], [65], [66]. It

Funding

This research did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors.

Conflict of interest statement

All authors declare no conflicts of interest and this research did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors.

References (95)

  • F. Triebel

    LAG-3: a regulator of T-cell and DC responses and its use in therapeutic vaccination

    Trends Immunol

    (2003)
  • F. Xie et al.

    Costimulatory molecule OX40/OX40L expression in ductal carcinoma in situ and invasive ductal carcinoma of breast: an immunohistochemistry-based pilot study

    Pathol Res Pract

    (2010)
  • V. Velcheti et al.

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

    Lab Invest

    (2014)
  • H.R. Ali et al.

    PD-L1 protein expression in breast cancer is rare, enriched in basal-like tumours and associated with infiltrating lymphocytes

    Ann Oncol

    (2015)
  • A. Swaika et al.

    Current state of anti-PD-L1 and anti-PD-1 agents in cancer therapy

    Mol Immunol

    (2015)
  • L. Apetoh et al.

    Combining immunotherapy and anticancer agents: the right path to achieve cancer cure?

    Ann Oncol

    (2015)
  • A.M. Cook et al.

    Chemotherapy and immunotherapy: mapping the road ahead

    Curr Opin Immunol

    (2016)
  • P. Zhang et al.

    Chemopreventive agents induce programmed death-1-ligand 1 (PD-L1) surface expression in breast cancer cells and promote PD-L1-mediated T cell apoptosis

    Mol Immunol

    (2008)
  • H. Bonnefoi et al.

    Pathological complete response after neoadjuvant chemotherapy is an independent predictive factor irrespective of simplified breast cancer intrinsic subtypes: a landmark and two-step approach analyses from the EORTC 10994/BIG 1-00 phase III trial

    Ann Oncol

    (2014)
  • F. Schettini et al.

    Nab-paclitaxel for the treatment of triple-negative breast cancer: rationale, clinical data and future perspectives

    Cancer Treat Rev

    (2016)
  • P. Sharma et al.

    Primary, adaptive, and acquired resistance to cancer immunotherapy

    Cell

    (2017)
  • F. Teng et al.

    Radiotherapy combined with immune checkpoint blockade immunotherapy: achievements and challenges

    Cancer Lett

    (2015)
  • S. Champiat et al.

    Management of immune checkpoint blockade dysimmune toxicities: a collaborative position paper

    Ann Oncol

    (2016)
  • T.K. Eigentler et al.

    Diagnosis, monitoring and management of immune-related adverse drug reactions of anti-PD-1 antibody therapy

    Cancer Treat Rev

    (2016)
  • K.D. Miller et al.

    Cancer treatment and survivorship statistics, 2016

    CA Cancer J Clin

    (2016)
  • F.S. Hodi et al.

    Improved survival with ipilimumab in patients with metastatic melanoma

    N Engl J Med

    (2010)
  • P.M. Ellis et al.

    Immune checkpoint inhibitors for patients with advanced non-small-cell lung cancer: a systematic review

    Clin Lung Cancer

    (2017)
  • C. Criscitiello et al.

    Immunotherapeutics for breast cancer

    Curr Opin Oncol

    (2013)
  • L.D. Miller et al.

    Immunogenic subtypes of breast cancer delineated by gene classifiers of immune responsiveness

    Cancer Immunol Res

    (2016)
  • P. Garcia-Teijido et al.

    Tumor-infiltrating lymphocytes in triple negative breast cancer: the future of immune targeting

    Clin Med Insights Oncol

    (2016)
  • E.M. Ibrahim et al.

    The prognostic value of tumor-infiltrating lymphocytes in triple-negative breast cancer: a meta-analysis

    Breast Cancer Res Treat

    (2014)
  • Z. Gatalica et al.

    Programmed cell death 1 (PD-1) and its ligand (PD-L1) in common cancers and their correlation with molecular cancer type

    Cancer Epidemiol Biomarkers Prev

    (2014)
  • O. Kepp et al.

    Consensus guidelines for the detection of immunogenic cell death

    Oncoimmunology

    (2014)
  • N. Casares et al.

    Caspase-dependent immunogenicity of doxorubicin-induced tumor cell death

    J Exp Med

    (2005)
  • A. Cimino-Mathews et al.

    Immune targeting in breast cancer

    Oncology (Williston Park)

    (2015)
  • L.A. Emens et al.

    Targeting the programmed cell death-1 pathway in breast and ovarian cancer

    Curr Opin Obstet Gynecol

    (2016)
  • S. Loi et al.

    Prognostic and predictive value of tumor-infiltrating lymphocytes in a phase III randomized adjuvant breast cancer trial in node-positive breast cancer comparing the addition of docetaxel to doxorubicin with doxorubicin-based chemotherapy: BIG 02-98

    J Clin Oncol

    (2013)
  • C. Denkert et al.

    Tumor-associated lymphocytes as an independent predictor of response to neoadjuvant chemotherapy in breast cancer

    J Clin Oncol

    (2010)
  • C. Denkert et al.

    Tumor-infiltrating lymphocytes and response to neoadjuvant chemotherapy with or without carboplatin in human epidermal growth factor receptor 2-positive and triple-negative primary breast cancers

    J Clin Oncol

    (2015)
  • Y. Mao et al.

    The value of tumor infiltrating lymphocytes (TILs) for predicting response to neoadjuvant chemotherapy in breast cancer: a systematic review and meta-analysis

    PLoS One

    (2014)
  • M. Ono et al.

    Tumor-infiltrating lymphocytes are correlated with response to neoadjuvant chemotherapy in triple-negative breast cancer

    Breast Cancer Res Treat

    (2012)
  • E. Muraro et al.

    Improved Natural Killer cell activity and retained anti-tumor CD8(+) T cell responses contribute to the induction of a pathological complete response in HER2-positive breast cancer patients undergoing neoadjuvant chemotherapy

    J Transl Med

    (2015)
  • G.J. Freeman et al.

    A new therapeutic strategy for malaria: targeting T cell exhaustion

    Nat Immunol

    (2012)
  • D.M. Pardoll

    The blockade of immune checkpoints in cancer immunotherapy

    Nat Rev Cancer

    (2012)
  • E.I. Buchbinder et al.

    CTLA-4 and PD-1 pathways: similarities, differences, and implications of their inhibition

    Am J Clin Oncol

    (2016)
  • J. Matsuzaki et al.

    Tumor-infiltrating NY-ESO-1-specific CD8+ T cells are negatively regulated by LAG-3 and PD-1 in human ovarian cancer

    Proc Natl Acad Sci USA

    (2010)
  • J. Larkin et al.

    Combined Nivolumab and Ipilimumab or Monotherapy in Untreated Melanoma

    N Engl J Med

    (2015)
  • Cited by (102)

    • Our evolving understanding of how 27-hydroxycholesterol influences cancer

      2022, Biochemical Pharmacology
      Citation Excerpt :

      Of those, tumor-infiltrating immune cell populations have been shown to have high predictive value in disease prognosis, even compared to traditional indicators in breast cancer, colorectal cancer, melanoma and pancreatic cancer [116–119]. Anti-PD1 (Pembrolizumab) and anti-PDL1 (Atezolizumab) are now approved for the treatment of several solid tumors, including a subset of patients with advanced TNBC [120–122]. However, these two immune checkpoint inhibitions have limited efficacy in the treatment of many solid tumors, including breast and ovarian.

    View all citing articles on Scopus
    View full text