Immune Check-Point Inhibitors in Breast Cancer: Current Evidence and Future Directions

Review Article

Austin J Surg. 2021; 8(3): 1270.

Immune Check-Point Inhibitors in Breast Cancer: Current Evidence and Future Directions

Mosteiro M1#, Cejuela M1# and Pernas S1,2*

¹Department of Medical Oncology, Catalan Institute of Oncology (ICO), L’Hospitalet de Llobregat, Barcelona, Spain

²Breast Cancer Group, Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), L’Hospitalet de Llobregat, Barcelona, Spain

#Contributed Equally to this Work

*Corresponding author: Sonia Pernas, Department of Medical Oncology, Catalan Institute of Oncology, Gran Via de l’Hospitalet 199-203, Spain

Received: April 27, 2021; Accepted: June 02, 2021; Published: June 09, 2021

Abstract

Check-point inhibitors have erupted as a treatment option for numerous kinds of neoplasms. Although there have been some achievements, the evidence supporting their use in breast cancer is scarce. Combinations with chemotherapy seem to provide better outcomes, and triple negative is the subtype most likely to benefit from them. New combination strategies are undergoing research to improve these results. Other approaches to determining biomarkers that identify which populations clearly benefit from these therapies are needed. Here, we review the clinical data of the role of immune check-point inhibitors in early and advanced breast cancer and present emerging strategies.

Keywords: Breast cancer; Immunotherapy; Immune checkpoints; PD-L1; Tumor-infiltrating lymphocytes

Abbreviations

BC: Breast Cancer; ICIs: Immune Checkpoint Inhibitors; PD-1: Programmed cell Death protein 1; CTLA-4: Cytotoxic T-Lymphocyte- Associated Protein 4; PD-L1: Programmed Death-Ligand 1; PD-L2: Programmed Death-Ligand 2; TILs: Tumor Infiltrating Lymphocytes; TNBC: Triple Negative Breast Cancer; HR: Hormone Receptor; HER2: Human Epidermal Growth Factor Receptor 2; OS: Overall Survival; ORR: Overall Response Rate; PFS: Progression Free Survival; CPS: Combined Positive Score; ITT: Intention To Treat; pCR: Pathological Complete Response; EFS: Event-Free Survival; AEs: Adverse Events; TMB: Tumor Mutational Burden; PR: Partial Response; SD: Stable Disease; T-DM1: Trastuzumab Emtansine; PARPi: Poly-ADP-Ribose-Polymerase inhibitors; DCR: Disease Control Rate; TPS: Tumor Proportion Score

Introduction

Breast Cancer (BC) is the most common malignancy and the second leading cause of cancer death in women worldwide. There were over 2.1 million newly diagnosed cases in 2018, accounting for one out of four cancer cases in women, and a total of 630,000 deaths [1]. Prognosis in western countries has improved in recent years, due to advances in treatment and earlier detection [2,3]. Nevertheless, metastatic disease is still a deadly illness, and finding new therapeutic strategies is of the utmost importance.

The host immune system has an important role in tumor initiation and progression. Exploiting intrinsic mechanisms of the host immune system to eradicate cancer cells has achieved impressive success. James Allison and Tasuku Honjo developed Immune Checkpoint Inhibitors (ICIs) which have dramatically changed the prognosis of multiple types of neoplasms such as lung cancer and melanoma, among others. Under normal conditions, the immune system uses an inhibitory checkpoint pathway to stop the immune response against pathogens and prevent autoimmune activity. This mechanism is carried out by the Programmed cell Death protein 1 (PD-1) and the Cytotoxic T-Lymphocyte-Associated protein 4 (CTLA-4) which down-regulate and inhibit T-cells by binding to their ligands: Programmed Death- Ligand 1 (PD-L1), Programmed Death-Ligand 2 (PD-L2) and CD80/ CD86 [4]. Tumor cells take advantage of this mechanism to create an immunosuppressive microenvironment in which they can hide from the immune system [5]. The anti-PD-1, anti-PD-L1 and anti-CTLA-4 monoclonal antibodies circumvent this immune down-regulation and boost the immune response to tumor cells [6-12].

BC is a heterogeneous disease with different molecular and clinical features. It has not traditionally been considered a highly immunogenic disease since it is characterized by a relatively low mutation burden in comparison to other neoplasms [13]. Nevertheless, BC immunogenicity is also heterogeneous, with different rates of immune infiltration depending on tumor subtype. The capacity to induce an immune response is also determined by other factors such as tumor neoantigens [14] or PD-L1 expression in the tumor and its microenvironment [15]. Additionally, some genetic mutations such as BRCA1 and BRCA2 result in homologous repair deficiency, which cause more genomic instability and high mutational loads [16]. Triple-Negative Breast Cancers (TNBC) are generally considered more immunogenic than Hormone Receptor (HR)-positive/Human Epidermal Growth Factor Receptor 2 (HER2)- negative BC, and differences in immunogenicity exist also among intrinsic molecular subtypes [17].

Tumor microenvironment includes a wide range of immune cells from both the innate and adaptive response. The quantification and morphological evaluation of these immune infiltrates have acquired great transcendence as a prognostic and predictive factor for response. Currently, PD-L1 has been established as the main biomarker for response to ICIs. In BC, it is up-regulated in approximately 20% - 34% of cases and has been linked to younger patients, high-grade and more aggressive tumors [18]. Tumors infiltrating lymphocytes (TILs) and some of its subpopulations have also been related to ICIs effectiveness. Increased TILs infiltration usually correlates with high PD-L1 expression, especially in TNBC. Among BC subtypes, high PD-L1 expression and TILs are more frequent in HER2-positive and TNBC [19,20].

The aim of this review is to summarize the current evidence of ICIs in both early and advanced BC, as well as review future directions and perspectives.

Triple Negative Breast Cancer

TNBC represents around 15% of BC cases. It is characterized by a lack of estrogen and progesterone receptors and HER2 [21]. It is often related to an earlier age at diagnosis, a more aggressive course, and a worse prognosis with more frequent visceral involvement. Although early-stage TNBC is often associated with high rates of response to chemotherapy, relapse is common and tends to appear in the first 3 years after the treatment [22-24]. Once metastasis occurs, TNBC is incurable, with a median Overall Survival (OS) of only 10-13 months [25-27]. At present, there are no specific treatments other than chemotherapy, but efforts are being made to find new therapeutic approaches for these patients.

Some features make TNBC more likely to respond to immunotherapy than other BC. For that reason, ICIs have mainly been tested in this subtype. It has the highest PD-L1 expression and TILs. In fact, an increased TILs infiltration and high PD-L1 expression have been both associated with better prognosis in early TNBC [28,29]. Moreover, TNBC holds a greater mutational load, which is related to higher tumor-specific neoantigens [30]. This may activate more neoantigen-specific T cells to trigger an anti-tumor response that can be strengthened by ICIs.

Several trials have evaluated therapies with the anti-PD-1 antibody pembrolizumab and the anti-PD-L1 antibodies avelumab, durvalumab and atezolizumab in TNBC.

ICI as single-agent in metastatic TNBC

The first trial reporting the clinical benefit of ICIs in TNBC was the KEYNOTE-012 (NCT01848834) which studied pembrolizumab in patients with metastatic TNBC with at least 1% of PD-L1 expression in either immune or tumor cells. Although most patients had previously been treated (84.4%), and over 46% of them had received =3 previous lines, the trial showed promising results with an Overall Response Rate (ORR) of 18.5% [31].

Atezolizumab was also tested in metastatic TNBC in the phase I trial PCD4989g (NCT01375842). Of the 116 total patients included, 58% had received at least one prior line of treatment. Those who received atezolizumab as first-line therapy with PD-L1 positive tumors presented better ORR, (Table 1) whereas none of the PD-L1 negative patients responded [32].