Abstract
Among the new cancer cases and resulting deaths among women worldwide, breast cancer is the most significant threat to women’s health. In recent years, immunotherapy was initially used to treat patients with metastatic breast cancer, where it demonstrated its unique value by providing a novel way to improve therapeutic effects and prolong survival time. With the development of clinical trials related to immunotherapy for breast cancer, tumour vaccines, such as DNA vaccines, have been observed to improve the disease-free survival (DFS) and overall survival (OS) of patients. Monoclonal antibodies have also shown good efficacy, and adoptive cell therapies, such as CAR-T, exhibit strong tumour killing ability and good safety, and thus, these therapies may comprise a new strategy for the treatment of breast cancer. These breakthrough successes have promoted the achievement of “individualized” breast cancer treatment. Moreover, a recent study showed that patients with various cancer types with a higher tumour mutational burden (TMB) are more likely to benefit from immunotherapy. As research progresses, TMB may also demonstrate a certain clinical significance in the treatment of breast cancer. This paper reviews the latest research progress on breast cancer immunotherapy and the predictive value and application status of TMB in immunotherapy regimens for breast cancer patients to provide a reference for further in-depth studies of breast cancer immunotherapy.
Keywords: Breast cancer, immunotherapy, tumour vaccine, adoptive cell therapy, immunobiomarker, tumour mutational burden
Introduction
Breast cancer is a global disease and is the most common malignant cancer in females worldwide [1]. Although substantial progress has been made in the diagnosis and treatment of breast cancer, the effects of current conventional treatment are not ideal for patients with advanced breast cancer, especially for those with triple-negative breast cancer (TNBC). The median survival time of patients with advanced TNBC who received conventional chemotherapy was reported to be only 13 months [2]. In recent years, immunotherapy has achieved remarkable results in tumour treatment and has become the fourth bell mode in addition to surgery, chemotherapy, and radiotherapy. Immunotherapy plays an antitumour role by regulating the immune system of patients. Historically, breast cancer has not been regarded as a highly immunogenic tumour because of the low mutation rate of breast cancer genes and a limited ability to form tumour-causing neoantigens [3]. However, a growing body of research has confirmed a strong link between breast cancer and the immune system. Studies have found that immunotherapy also has a good therapeutic effect in some breast cancer patients, among which studies are also called anti-programmed cell death-1/programmed death ligand-1 (PD-1/PD-L1) at most [4]. Moreover, a recent study showed that among various cancer types, patients with higher tumour mutational burden (TMB) are more likely to benefit from immunotherapy. This article reviews the latest research and current immunobiomarkers of breast cancer immunotherapy to provide a reference for further research on immune-precision therapy for breast cancer.
Immunotherapy strategies for breast cancer
Immunotherapy for breast cancer can be divided into three categories according to its mechanism of action: active immunotherapy (including various tumour vaccines), passive immunotherapy (including monoclonal antibodies, adoptive cell therapy, etc.), and nonspecific immunomodulator therapy (mainly anti-PD-1/PD-L1 antibodies, anti-CTLA-4 antibodies, etc.) [5]. Current immunotherapy that targets breast cancer has progressed in regard to tumour vaccines, monoclonal antibodies, and immunomodulators (Figure 1).
Figure 1.
Classification of cancer immunotherapy [42]. Progress of Immunotherapy and Its Application in Triple Negative Breast Cancer. Note: AR-T, chimeric antigen receptor T cell; TNF, tumor necrosis factor; IFN, interferon; IL, interleukin; GM-CSF, granulocyte-macrophagecolony stimulating factor; PD-1, programmed cell death-1; PD-L1, programmed cell death-ligand 1; CTLA-4, cytotoxic T lymphocyte-associated antigen 4.
Tumour vaccines
Tumour vaccines play an antitumour role by stimulating specific immune responses to tumours through immunization. Tumour vaccines can maintain long-term immune memory and exert long-lasting effects.
Tumour cell vaccine
A tumour whole cell vaccine is rich in tumour antigens and can theoretically generate nonspecific costimulatory signals through antigen presenting cells (APCs) to promote an immune response. Whole cell vaccines were administered to patients with seven types of advanced tumours, including breast cancer, to prolong OS [currently in a phase I/II clinical trial (NCT00722228)]. Due to the absence of costimulatory molecules, an adjuvant is often added to enhance the immune response to tumour whole cell vaccines. However, adjuvants can also induce autoimmunity, which limits their usefulness [6].
Cancer polypeptide (protein) vaccine
DNA vaccine
DNA vaccines can induce the synergistic effect of humoral and cellular immunity by taking up the DNA sequence encoding the target tumour-associated antigen (TAA), translating it, and expressing it into protein, which is then treated and presented as an antigen. DNA vaccines can be produced on a large scale and can easily induce antitumour immune responses, but low long-term levels of tumour antigens can cause immune tolerance. Guardino et al. [7] found that the MVA-BN®-HER2 vaccine has biological activity against metastatic breast cancer with HER2 overexpression and can break HER2 tolerance. At present, although it is still a challenge to find suitable vectors for a DNA vaccine, the electroporation method has achieved preliminary results, which may be a breakthrough [8].
DC vaccine
Dendritic cells (DCs) are the only specialized APCs that can activate primary T cells, and thus, they play an important role in the immune response. A DC vaccine is not restricted by HLA and can induce class I and II immune responses. Studies [9] found that a DC vaccine based on HER2/Neu could induce a decrease and even cause loss of HER2/Neu expression. In addition, the Indoximod + AD.p53DC vaccine may be sensitized to chemotherapy, but its safety and efficacy in combination with chemotherapy drugs require further study [10]. Abdellateif et al. [11] induced mature DCs using viable MCF-7 breast cancer cells in vitro and reported that CD83+, CD86+, and MHC-II+ DCs were significantly increased after induction and that CD4+ and CD8+ T cells were also increased. Numbers of CD4+, CD25+, and Foxp3+ Tregs were significantly decreased, and Foxp3 gene expression levels were significantly decreased. In addition, the secretion of IL-12 and IFN-γ was increased, while the release of lactate dehydrogenase was significantly increased, which indicates the enhanced cytotoxicity of CTLs to the tumour. Therefore, a live breast cancer cell-DC vaccine is a novel approach for breast cancer immunotherapy.
HER2/neu
The HER2/Neu protein is immunogenic and can induce antigen-specific immune responses by CD4+ and CD8+ T lymphocytes. High expression of HER2/Neu can therefore be an effective target for immunotherapy in breast cancer. At present, advanced research is being performed on three types of polypeptide vaccines, namely, E75, GP2 and AE37.
(I) The HER2 antigenic peptide E75 is an HLA-A2/A3-restricted polypeptide derived from the extracellular domain of the HER2/Neu protein that plays an antitumour role by effectively stimulating the expansion of E75-specific cytotoxic lymphocytes (CTLs). Preocular phase I/II clinical trials [12] have confirmed that the E75 vaccine can increase disease-free survival (DFS) in patients with early breast adenocarcinoma, and related phase III clinical trials have also been completed [13]. Benavides et al. [14] found that breast cancer patients with HER2/Neu overexpression may have immune tolerance to HER2/Neu, as patients with low expression of HER2/Neu experience a greater benefit.
(II) GP2 is derived from the HER2/Neu transmembrane domain and is also an HLA-A2 restricted polypeptide, which means it is more immunogenic than E75. Early results of a phase II clinical trial of GP2 for the treatment of breast cancer (NCT00524277) [15] showed that DFS was higher in the experimental group than in the control group (94% vs. 85%).
(III) AE37 is a HER2/Neu-derived MHC class II epitope peptide vaccine hybridized with the li-Key peptide, which significantly enhances the activity of CD4+ T cells and has a longer-lasting immune effect. A phase II clinical trial showed that the relative risk of patients with non-HER2 overexpression was reduced after treatment with the AE37 vaccine and that AE37 could benefit patients with non-HER2 overexpression, especially those with TNBC.
Monoclonal antibody therapy
Monoclonal antibody against HER2/Neu
Previous studies [16,17] showed that dual-targeted therapy can improve the pathologic complete response (pCR) of patients, but the difference was not statistically significant. Recently, a phase III randomized controlled trial (CALGB40601) of a dual target (lapatinib + trastuzumab) combined with paclitaxel in HER2-positive breast cancer [18] suggested that the prolongation of invasive disease-free survival (iDFS) was correlated with pCR. Powles et al. [19] found that patients with high levels of the T-cell receptor β-chain variable (TRBV) 11-3 gene or TRBV-MG2 benefited more from dual-targeted therapy. Thus, the TRBV gene pattern can predict the efficacy of dual-targeted therapy. In addition, primary or secondary drug resistance is a key factor that affects the clinical efficacy of trastuzumab. In the Panacea study [20] trastuzumab-resistant HER2-positive metastatic breast cancer was treated with a combination of pembrolizumab and trastuzumab, the results of which are forthcoming. Pertuzumab is a novel HER2 recombinant monoclonal antibody that binds to the extracellular domain II of HER2 and remains effective against breast cancers with low HER2 under-expressed breast expression and poor trastuzumab response. A phase III randomized clinical trial (NCT00567190) found that the median overall survival (OS) and median progression-free survival (PFS) in patients with nonearly HER2-positive breast cancer treated with trastuzumab plus docetaxel were significantly longer after the addition of pertuzumab. In another phase III clinical trial (APHINITY; LBA500) 4,805 patients with early HER2-positive breast cancer were treated with standard chemotherapy, and the results showed that iDFS in the pertuzumab group was 94.1% vs. 93.2% (P = 0.045) in the other group [21]. The benefit in the lymph node-positive group was greater than that in the trastuzumab group (92% vs. 90.2%). Therefore, dual-targeted therapy combined with chemotherapy is recommended for the first time as an adjunctive therapy for patients with lymph node-positive HER2-positive breast cancer [22]. Keith et al. [23] further confirmed that T-DM1 can effectively delay the growth of brain metastasis (BMS) in patients with HER2-positive BMS and results in a survival benefit. In addition, Borges et al. [24] showed that tucatinib + T-DM1 exhibited preliminary antitumour activity and acceptable toxicity in refractory HER2-positive breast cancer patients with or without brain metastasis.
Recently, the results of the open-label phase III trial of margetuximab were presented [25]. SOPHIA trial (NCT02492711) enrolled 538 patients with advanced HER2 positive metastatic BC and randomly assigned in a 1:1 fashion to margetuximab (15 mg/kg intravenously every 3 weeks) plus chemotherapy or trastuzumab (6 mg/kg [8-mg/kg loading dose]) plus chemotherapy given every 3 weeks. All patients had received trastuzumab and pertuzumab, and more than 90% had also received T-DM1. Most patients were treated with taxane, more than 40% were treated with anthracycline, and almost half had received an endocrine drugs. Compared with trastuzumab, magtuximab improved primary PFS with a 24% relative risk reduction (P = 0.03), with a median PFS of 5.8 months versus 4.9 months with trastuzumab. Margetuximab improved primary PFS over trastuzumab with 24% relative risk reduction (p = 0.03) with a median PFS of 5.8 vs. 4.9 months. The ORR was higher in the magtuximab group: 25.2% vs. 13.7%, increasing the clinical benefit rate from 35.6% with trastuzumab to 48.1% with margetuximab (p = 0.0025). The median duration of response was similar in the two arms. In the planned exploratory analysis of the FcRIII genotype, the benefit was enhanced in patients with low-affinity FcγIIIa genotypes containing a 158F allele, in which disease progression was reduced by 32% [25,26]. In the second interim analysis, for 85% of patients with the FcIIIa 158F allele, median OS was extended by 4.3 months (23.7 months) in the Margetuximab arm compared with 19.4 months (P = 0.087) in the trastuzumab arm. Safety profiles and treatment discontinuation rates were comparable in the margetuximab and trastuzumab arms [27].
Bispecific monoclonal antibody (mAb)
Bispecific mAbs can bind two different TAAs, such as HER3/EGFR, VEGF/HER2, and HER2/HER3, and this double blockade can improve the efficacy of immunotherapy. MM 111 is a bispecific single chain antibody that simultaneously targets HER2/HER3 in HER2-overexpressing breast cancer. Preclinical studies [28] found that MM-111 showed antitumour activity when combined with trastuzumab and lapatinib. T cells lack FcyR, and thus, typical antibodies cannot directly recruit T cells. The Fv region of the triple functional antibody can bind to tumour cells and T cells, while the Fc region can bind to FCR-expressing immune cells, such as NK cells, and kill tumours through antibody-dependent cellular cytotoxicity. In vitro studies [29] found that ertumaxomab, a trifunctional antibody against HER2 and CD3, can be used to treat tumours with low HER2 expression that do not respond to trastuzumab. In addition, many other types of bispecific antibodies, such as BiTEs, are also under investigation [30]. To overcome the problems of immunogenicity and short half-life, we focused on a T-cell-dependent bispecific antibody (HER2-TDB) that can target HER2 and induce an antitumour immune response of polyclonal T cells, which reduces the occurrence of tumour immunity. Other studies [31] found that HER2-TDB has a strong effect at low concentrations in HER2-overexpressing cell lines and showed certain efficacy in cell lines resistant to trastuzumab, lapatinib, TDM-1, and other drugs. HER2-TDB was also effective in hormone receptor-positive breast cancer (HPBC). Although PD-L1 expression limits HER2-TDB activity, the inhibitory effect can be reversed with anti-PD-L1 antibodies, which suggests that a combination with immune checkpoint inhibitors can increase immunotherapy efficacy. However, since HER2 is also expressed at low levels in many normal somatic cells, such as cardiomyocytes, the safety of HER2-TDB should be further evaluated.
Non-specific immunomodulator therapy
PD-1/PD-1/PD-L1 inhibitors
A meta-analysis showed that PD-L1 expression was significantly correlated with lymph node metastasis and histological grade, which suggests that PD-L1 expression may be a prognostic biomarker for breast cancer (Figure 2).
Figure 2.
Schematic diagram of aPD-1mAb in treatment of triple negative breast cancer [42]. Progress of Immunotherapy and Its Application in Triple Negative Breast Cancer. A: PD-L1 expressed on tumor cells combing with PD-1 expressed on TILs play an immunosuppressive role by inhibiting the anti-tumor activity of T cells and tumor cells survive through immune escape before anti-PD1 or anti-PD-L1 monoclonal antibody (aPD-1/PD-L1 mAb); B: PD-1/PD-L1 axis becomes immunoreactive after adding aPD-1/PD-L1 mAb owing to the activation of CD8+ T cells. aPD-1 mAb, anti-PD-1 monoclonal antibody; TILs, tumor infiltrating lymphocytes.
Although patients with metastatic TNBC who received immunotherapy benefited more in terms of objective response rate (ORR) compared with second-line and above chemotherapy, no significant difference was observed in overall survival time (OS) between the two groups [32]. Advanced breast cancer patients with rapid tumour progression or visceral crisis often require rapid control of tumour growth in the short term to alleviate symptoms. Most breast cancer patients are resistant to monotherapy with checkpoint inhibitors, and radiation therapy has many immune-stimulating effects, including activation of the immune system, recruitment of immune cells to the tumour environment and immunosuppressive effects that alter the tumour microenvironment. Radiotherapy combined with checkpoint inhibitors can not only synergistically enhance antitumour efficacy but can also induce long-lasting field responses to radiotherapy [33,34]. A phase II study [35] reported that in patients with advanced TNBC who were treated with pembrolizumab combined with radiotherapy, 3 of 9 evaluable patients (33%) responded outside the irradiation zone and survived up to 49 weeks. Several additional clinical trials evaluating the efficacy of radiotherapy combined with new immunotherapy approaches are ongoing [36]. PD-1/PD-L1 inhibitors are also emerging in TNBC neoadjuvant therapy [37]. One study [38] evaluated the efficacy of adding durvalumab to standard neoadjuvant chemotherapy in primary TNBC, and the results showed that the addition of durvulumab to anthracycline/taxane standard neoadjuvant chemotherapy increased pCR rates (53% vs. 44%) compared with durvulumab alone prior to the initiation of chemotherapy. Although immunotherapy is not used as a neoadjuvant or advanced treatment, its combination with chemotherapy significantly increased the benefit in patients compared with conventional chemotherapy or immunotherapy alone, and no unacceptable immune-related adverse reactions were observed. In addition to TNBC, in which efficacy has been established, clinical trials of PD-1/PD-L1 have been conducted in HER2-positive and hormone-receptor-positive (HR+) breast cancer patients. Tumour-infiltrating lymphocytes (TILs) and PD-L1 expression are lower in HR+/HER2- breast cancer than in HER-2+ breast cancer and TNBC, and thus, patients with HR+/HER2- cancer may not respond significantly to checkpoint inhibitor therapy. The Keynote-028 study treated patients with PD-L1-positive HR+/HER2- advanced breast cancer with pembrolizumab monotherapy, and the ORR was approximately 12%, whereas the ORR was only 3% in patients with metastatic HR+/HER2- breast cancer treated with avelumab. In early HR+ breast cancer, the opposite result is likely to be observed; with the addition of pembrolizumab to standard neoadjuvant chemotherapy, the estimated pCR rate increased from 13% to 34%, which suggests that the combination in neoadjuvant therapy may be effective for this subtype [39,40]. The 229 studies performed on trastuzumab and durvalumab reported disappointing results, with no significant clinical activity observed in patients with HER-2-positive PD-L1-negative metastatic breast cancer [41]. These are similar to the results of PD-L1-negative patients in the Keynote-14 study [42,43], where the ORR of PD-L1-positive patients was 15%. In addition, overall survival data from the Pherexa trial [44] suggest that trastuzumab and pertuzumab combined with the clinical activity of double HER-2 inhibition may induce immune activation in patients with advanced HER-2-positive progressive breast cancer who previously received trastuzumab. HER2-positive-resistant advanced breast cancer was resensitized to trastuzumab therapy, although the prolongation in PFS was not significant.
CTLA-4 inhibitors
Similar to PD-1, CTLA-4 is a negative costimulatory molecule expressed on the surface of the T-cell membrane, and its ligand is the B7 molecule present on the surface of APCs and target cells (tumour cells). The combination of the two can inhibit T-cell activation, proliferation, and cytokine secretion and can negatively regulate the body’s immune response to tumour cells [45]. In addition, CTLA-4 can also negatively regulate the immune response by inhibiting the maturation and antigen presenting ability of APCs, promoting the amplification of Tregs, and inducing the production of indoleamine 2,3-dioxygenase in APCs [46]. Thus, blocking the CTLA-4 signalling pathway restores the immune system’s ability to recognize tumour cells. Currently, two humanized CTLA-4 monoclonal antibodies are clinically available: tremelimumab and ipilimumab. Vonderheide RH [47] combined tremelimumab and exemestane for the treatment of patients with ER-positive, HER2-negative advanced breast cancer, and of these, approximately 42% (11 cases) achieved disease stability over 12 weeks, and the main adverse reactions were mild to moderate diarrhoea, itching, constipation, and fatigue. Ipilimumab is mainly used as a single drug or in combination with PD-1/PD-L1 inhibitors for melanoma and other tumours [48]. Preclinical studies have demonstrated significant antitumor efficacy of combination therapy targeting both PD-1 and CTLA-4 immune checkpoints. The combination of blocking and PD-1 and CTLA-4-based vaccines can promote the rejection of B16 melanoma tumour cells in mice and can synergistically increase the proportion of CD4+ TILs and MDSCs on the tumour surface of these mice [49]. The phase III Check Mate 067 study [50] confirmed that nivolumab combined with ipilimumab significantly improved PFS and 3-year survival in patients with melanoma. However, the exact mechanism and efficacy of this immunocombination therapy in breast cancer are still unclear and require further exploration in relevant animal models and clinical trials.
Adoptive cell therapy
Adoptive cell therapy can improve the state of low cellular immune function in the body by activating immune effects in autologous or allogeneic cells in vitro and then transferring them back into the body where they exert antitumour effects. This therapy can also directly play an antitumour role and repair immune damage to a certain extent. However, limited cell sources, low cytotoxicity, and a low number of amplifications limit its application. Cytokine-induced killer (CIK) cells are mainly CD3+ CD56+ T cells, with the non-MHC restriction of natural killer cells and the antitumour activity of T lymphocytes, that can directly kill tumour cells. Activated CIK cells can inhibit and kill tumour cells by expressing high levels of cytokines. Their tumour killing spectrum is broad, their proliferation rate is fast, their killing activity is high, and their associated adverse reactions are infrequent. Therefore, these cells have certain advantages as an immunotherapy treatment for breast cancer.
Pan et al. [51] found that CIKs are an effective treatment for TNBC patients with positive lymph nodes, high TNM stage, and poor pathological grade.
TILs
TILs are cells with MHC restriction and tumour specificity that exert a novel antitumor effect [52]. Zhang et al. [53] purified and expanded TILs from breast cancer and lymph nodes to obtain >1×109 cells after which they were transfused back into patients. The results showed that TILs had the highest killing rate at 78.0%, which suggests that TILs can enhance cellular immune function in breast cancer patients. Recently, Zacharakis et al. [54] reported a case of hormone receptor-positive and HER2-negative metastatic breast cancer that failed to respond to multiple chemotherapy regimens. TILs were cultured in vitro, and TILs that responded to only four somatic mutant proteins were further cultured to a certain number and transfused back into the body. At the same time, the combination of TILs with IL-2 and an immune checkpoint inhibitor (ICI) showed that the disease foci completely disappeared for more than 22 months, which represents a new way for immunotherapy to treat refractory cancer.
Genetically modified T cells
CAR T therapy (CAR-T) is an adoptive cell immunotherapy based on the use of genetically engineered lymphocytes that express chimeric antigen receptors that kill tumour cells in a nonmajor histocompatibility complex-restrictive manner [55]. CAR-T therapy has been successful in haematological malignancies, but challenges remain in solid tumours. Clinical trials of CAR-T cells targeting HER2 in breast cancer are ongoing. A phase I/II clinical trial (NCT01935843) [56] investigated the clinical efficacy of CAR-T cells in the treatment of HER2-positive advanced solid tumours. Another phase II trial (NCT01022138) [57] evaluated the efficacy of activated T cells modified with an anti-CD3X, anti-HER2/Neu bispecific monoclonal antibody in HER2/Neu-negative metastatic breast cancer. Byrd et al. [58] proposed that specific CAR-T cells targeting TEM8/ANTXR1 could be used as an immunotherapy for TNBC. In addition, clinical trials of CAR-T cells targeting other breast cancer antigens, such as cMet and tumour-associated antigen-mesothelin, are also ongoing. In conclusion, in solid tumours such as breast cancer, some key issues related to CAR-T therapy persist, such as scarcity of target antigens and limited CAR-T-cell duration, which require further research (Figure 3).
Figure 3.
Chimeric antigen receptor T cell immune therapy [56]. Advances in research on tumor immunotherapy. CAR-T, Chimeric antigen receptor T cell immune therapy; TILs, Tumor-infiltrating lymphocytes.
Immunobiomarkers
Tumour mutation load
Tumour mutational burden (TMB) refers to the total number of somatic mutations detected per million bases or the number of mutations detected per tumour. Neoantigens generated by somatic mutations in tumours can induce the body’s antitumour immune response. Therefore, patients with higher TMB levels respond better to immunotherapy drugs. Compared with the TMB of lung cancer, the TMB of breast cancer is lower and related to molecular typing. TNBC has the highest TMB, followed by HER2+ breast cancer [59]. Among patients with TNBC and HER2+ refractory metastatic breast cancer, those with high TMB have significantly better OS than patients with low TMB [60,61]. TMB is closely related to the prognosis of breast cancer, and studies on the ability of TMB to predict the efficacy of PD-1/PD-L1 antibody therapy for breast cancer need to be performed.
The predictive role of TMB in triple-negative breast cancer (TNBC) immunotherapy
The IMpassion130 phase III clinical trial demonstrated that treatment with atlizumab plus albumin-paclitaxel significantly extended PFS (7.5 months vs. 5 months) and overall survival (25 months vs. 18 months) in patients with advanced TNBC with PD-L1-positive tumours compared with placebo plus albumin-paclitaxel [62]. Emens et al. [63] performed a retrospective study based on this finding and confirmed that higher TMB was associated with an overall survival benefit in the atlizumab combined with albumin-paclitaxel group; however, a clinical benefit was only observed in patients with PD-L1-positive tumours, and TMB was not related to PD-L1 expression. TMB also plays a predictive role in immune neoadjuvant therapy for breast cancer. In a study of needle neoadjuvant therapy for early TNBC, the predictive value of TMB and TMB combined with gene expression profiling (GEP) for pathologic complete response (pCR) was shown. The results showed that both TMB and immune GEP were independent predictors of pCR, and that when patients were stratified according to the upper third and median GEP of TMB, the pCR rate was 82% in patients with higher TMB and GEP (95% CI: 60-95%), while the pCR rate was only 28% (95% CI: 16-43%) in patients with low TMB and GEP [64,65].
Predictive effect of TMB on immunotherapy efficacy in other molecular types of breast cancer
Yin et al. [66] analysed whole-exon sequencing data of 366 cases of breast cancer in the Cancer Genome Atlas (TCGA) database and next-generation sequencing data of 335 cases of patients with breast adenocarcinoma. We confirmed that human epithelial growth factor receptor 2 (HER2) amplification is associated with higher TMB in breast cancer, which may be helpful in screening breast cancer patients to determine who is more suitable for immunotherapy. TMB can not only predict the efficacy of immunotherapy for HER2-positive breast cancer but can also serve as a prognostic factor for patients treated with HER2-targeted drugs combined with chemotherapy. In a clinical study of patients with metastatic breast cancer, 31 of 46 patients (67.4%) with HER2-positive metastatic breast cancer elected to receive docetaxel plus HER2-targeting trastuzumab, while 7 patients (15.2%) received docetaxel and trastuzumab plus pertuzumab; 16 patients (34.6%) were predefined as part of the high TMB group, and 30 patients (65.4%) were in the low TMB group. The results showed that the difference in median overall survival of those in the low and high TMB groups (44.9 months vs. 85.8 months) was statistically significant [61]. TMB and antitumour immunogenicity were found to be higher in HR-negative than in HR-positive breast cancer patients. HR-negative breast cancer patients may show higher immunogenicity. Patients with HER2-positive breast cancer may exhibit higher immunogenic activity than those with HER2-negative breast cancer [67]. HR-negative or HER2-positive breast cancers exhibit higher TMB and immunogenic activity, which suggests that patients with these tumours may benefit from immunotherapy. Generally, the effect of TMB on the ICI treatment of breast cancer varies according to the molecular type of breast cancer and antitumour immune response. TMB in breast cancer was reported to be the highest in clinically aggressive TNBC, followed by HER2-positive, luminal B, and luminal A cancers [68,69]. It is important to note that the primary source of TMB detection is primary or metastatic tumour tissue, which may lead to systematic bias because metastatic tumours tend to have more monoclonal structures. Schnidrig et al. [70] retrospectively analysed 1,662 patients with advanced cancer who were treated with ICI therapy and whose TMB was measured using the MSK-IMPACT technology platform; they then explored whether primary or metastatic TMB predicted overall survival after ICI treatment. The results showed revealed a positive correlation between primary TMB and metastatic TMB. However, primary TMB and metastatic TMB were equally effective in predicting overall survival during ICI treatment (primary TMB: HR = 0.61, 95% CI: 45-82%; metastatic TMB: HR = 0.59, 95% CI: 45-76%).
TMB combined with other biomarkers to predict the efficacy of immunotherapy for breast cancer
TMB has been demonstrated to be a biomarker that can predict the efficacy of immunotherapy in breast cancer. However, as additional research has been performed, methods to optimize the integration of TMB and other biomarkers have been proposed to improve the accuracy of prediction. A meta-analysis published in JAMA Oncology showed that the combination of multiple biomarkers had the best predictive value for response to ICI therapy compared with individual biomarkers (e.g., PD-L1, GEP, TMB) [71]. Thus, while higher TMB is associated with better treatment outcomes for ICI therapy, the complexity of the immune response means that TMB should be considered in conjunction with other factors to optimize the prediction of ICI outcomes. As for other potential biomarkers in breast cancer immunotherapy, such as new antigen load, mismatch repair defects, BRCA1/2 mutation, immunogenicity of cell death, MHC II expression, tumour-infiltrating lymphocytes, oestrogen receptor expression, and some markers of serum lactate dehydrogenase, their integration as a composite biomarker may lead to better prediction of tumour immunotherapy efficacy.
Tumour-infiltrating lymphocytes
Tumour-infiltrating lymphocytes (TILs) are a heterogeneous population of lymphocytes that are primarily present in tumour nests and in the interstitium [72]. The predictive effect of TILs on immunotherapy for breast cancer is related to the type of TIL and the molecular classification and development stage of breast cancer, but the relationship between the two is still controversial. Studies have shown that TNBC has the highest degree of TIL invasion, followed by HER2+ breast cancer [73]. The higher the level of TILs in TNBC patients, the better the effect of immunotherapy, and the number of TILs is positively correlated with prognosis, such as disease-free survival [74]. The Keynote-086 study found that in metastatic TNBC, a higher abundance of interstitial TILs was closely associated with better therapeutic efficacy of pembrolizumab [75]. However, the predictive effect of TILs on immunotherapy for breast cancer is still unclear, and further studies are needed.
PD-L1
With FDA approval of PD-1/PD-L1 antibodies for the treatment of malignant tumours, PD-L1 expression has also been included in all levels of guidelines and has thus become an important immunobiomarker for screening patients for appropriate antibody therapy. In addition to tumour cells that express PD-L1, immune cells such as lymphocytes and macrophages also express PD-L1. Therefore, the following three methods can be used to evaluate the expression level of PD-L1: (1) the tumour proportion score (TPS), which refers to the proportion of PD-L1+ tumour cells; (2) the combined positive score (CPS), which refers to the ratio of the total number of PD-L1+ tumour cells, lymphocytes, and macrophages to the total number of tumour cells; and (3) the expression of PD-L1 in immune cells only [76]. PD-L1 expression is detected by immunohistochemistry. Currently, FDA-approved antibodies for immunohistochemical detection include 28-8, 22C3, SP142, and SP263 [77]. The Blueprint project compared these four antibodies and showed that 28-8, 22C3, and SP263 predominantly stained tumour cells with similar results, while SP142 showed strong staining of immune cells. All four antibodies stained immune cells and showed both greater heterogeneity compared with tumour cell staining and different judgement criteria due to different critical values [78]. The IMpassion130 study used SP142 to detect PD-L1 expression in tumour and immune cells. In TNBC, PD-L1 expression is primarily found on tumour-infiltrating immune cells, and the PD-L expression level predicts whether patients will benefit from alemtuzumab combined with albumin-bound paclitaxel. Patients in the PD-L1+ group had longer median PFS and OS [79]. CD8+ T cells were abundant in the population of PD-L1+ immune cells. Only when the PD-L1 expression of immune cells was positive could CD8+ T cells predict any benefit. More PD-L1+ cells were also observed in the TIL population. Similarly, patients with high TIL content could benefit only when immune cells were positive for PD-L1 expression. PD-L1 expression on tumour cells and immune cells was evaluated by SP142 and SP263. The CPS score was used for 22C3. Over 97% of patients with positive PD-L1 by SP142 staining were also positive by 22C3 and SP263 staining, and when both SP142 and 22C3 were positive, the PFS and OS benefits of alemtuzumab combined with albumin-bound paclitaxel could be predicted. In conclusion, in different studies, the predictive effect of PD-L1 expression on the efficacy of PD-1/PD-L1 antibodies is highly variable. Due to the heterogeneity of PD-L1 expression, nonunified detection methods and interpretation standards, as well as for other reasons, more studies are needed on the molecular typing of breast cancer, detection methods, and PD-L1 expression to accurately predict the efficacy of PD-1/PD-L1 antibodies.
Conclusion and prospects
Immunotherapy has been shown to prolong the survival time of patients with a variety of solid tumours and is a promising treatment option for metastatic breast cancer. Immune checkpoint inhibitors and secondary cellular immunotherapy, which enable immune-mediated tumour clearance, are two major achievements in cancer immunotherapy that play a leading role in overcoming tumour-induced immunosuppression.
However, some patients will still experience drug resistance to existing immunotherapies, which will lead to immune escape and a poor overall therapeutic effect. In triple-negative breast cancer, the combination of chemotherapy, radiotherapy, and new targeted therapies with existing immunotherapy methods, especially immune checkpoint inhibitors, has greatly improved the efficacy of immunotherapy. This suggests that an effective combination of immune checkpoint inhibitors with chemotherapy or other immune-targeted therapies may be an effective means to improve long-term survival of patients with triple-negative breast cancer who do not have options for specific effective endocrine therapy or targeted therapy.
As breast cancer has entered a new era, immunotherapy has shown an ability to treat metastatic breast cancer. Among patients with breast cancer, those with PD-L1-positive and triple-negative metastatic breast cancer are most likely to benefit from immunotherapy, and multiple retrospective studies have preliminarily confirmed that TMB is an effective biomarker for predicting immunotherapy efficacy in breast cancer. Additional studies have analysed TMB in different molecular types of breast cancer. Due to the complexity of the immune system, the integration of TMB and several other immune-related biomarkers into a composite biomarker can more accurately predict the outcome of ICI treatment. Since the definition of high TMB is not unified with the detection method of TMB, and because clinical trial data support is lacking, TMB is not currently able to better guide clinical treatment.
It is believed that with the increasing momentum of TMB as a biomarker of immunotherapy response and the emergence of standardized methods for clinical application, TMB detection as a way to guide treatment will become a new trend in precision medicine in the foreseeable future.
Acknowledgements
We thank Ruifang An, Department of Obstetrics and Gynecology, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shannxi, P. R. China who took part with great efforts in revising the paper in English. All authors have given their consent for publication.
Disclosure of conflict of interest
None.
Abbreviations
- TNBC
Negative breast cancer
- DFS
Disease-free survival
- OS
Overall survival
- PD-L1
Programmed cell death 1
- TMB
Tumor mutational burden
- AR-T
chimeric antigen receptor T cell
- TNF
tumor necrosis factor
- IFN
interferon
- IL
interleukin
- GM-CSF
granulocyte-macrophagecolony stimulating factor
- CTLA-4
cytotoxic T lymphocyte-associated antigen 4
- CTL
cytotoxic lymphocyte
- MUC1-C
t MUC1 transmembrane C-terminal
- ADCC
antibo-dependent cell-mediated cytotoxicity
- ADCP
antibose-dependent cell-mediated phagosis
- CDC
Complement dependent cytotoxicity
- TRBV
cell β-chain variable
- HPBC
hormone receptor positive breast cancer
- PFS
disease-free progression
- LAK
flymphokine-activated killer cell
- ICI
immune checkpoint inhibitor
- CAR T
Chimeric antigen receptor T cell immune therapy
- TMB
Tumor mutation burden
- HER2
Human epithelial growth factor receptor 2
- TIL
Tumor-infiltrating lymphocyte
- TPS
Tumor positive score
- CPS
Combined positive score
References
- 1.Emens LA. Immunotherapy in triple-negative breast cancer. Cancer J. 2021;27:59–66. doi: 10.1097/PPO.0000000000000497. [DOI] [PubMed] [Google Scholar]
- 2.Keenan TE, Tolaney SM. Role of immunotherapy in triple-negative breast cancer. J Natl Compr Canc Netw. 2020;18:479–489. doi: 10.6004/jnccn.2020.7554. [DOI] [PubMed] [Google Scholar]
- 3.Topalian SL, Drake CG, Pardoll DM. Immune checkpoint blockade: a common denominator approach to cancer therapy. Cancer Cell. 2015;27:450–461. doi: 10.1016/j.ccell.2015.03.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Loibl S, Poortmans P, Morrow M, Denkert C, Curigliano G. Breast cancer. Lancet. 2021;397:1750–1769. doi: 10.1016/S0140-6736(20)32381-3. [DOI] [PubMed] [Google Scholar]
- 5.Emens LA, Adams S, Cimino-Mathews A, Disis ML, Gatti-Mays ME, Ho AY, Kalinsky K, McArthur HL, Mittendorf EA, Nanda R, Page DB, Rugo HS, Rubin KM, Soliman H, Spears PA, Tolaney SM, Litton JK. Society for immunotherapy of cancer (SITC) clinical practice guideline on immunotherapy for the treatment of breast cancer. J Immunother Cancer. 2021;9:e002597. doi: 10.1136/jitc-2021-002597. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Pernot S, Terme M, Voron T, Colussi O, Marcheteau E, Tartour E, Taieb J. Colorectal cancer and immunity: what we know and perspectives. World J Gastroenterol. 2014;20:3738–3750. doi: 10.3748/wjg.v20.i14.3738. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Guardino A, Cassidy M, Pienkowski T, Radulovic S, Legrand F, Nguyen A, Fernandez L, Coutts J, Moore N, Hwang O, Trieger B, Brand L, Reiner L, Delcayre A, Godfrey W. Results of two phase I clinical trials of MVA-BN-HER2 in HER-2 overexpressing metastatic breast cancer patients. Cancer Res. 2009;69:5089. [Google Scholar]
- 8.Trimble CL, Morrow MP, Kraynyak KA, Shen X, Dallas M, Yan J, Edwards L, Parker RL, Denny L, Giffear M, Brown AS, Marcozzi-Pierce K, Shah D, Slager AM, Sylvester AJ, Khan A, Broderick KE, Juba RJ, Herring TA, Boyer J, Lee J, Sardesai NY, Weiner DB, Bagarazzi ML. Safety, efficacy, and immunogenicity of VGX-3100, a therapeutic synthetic DNA vaccine targeting human papillomavirus 16 and 18 E6 and E7 proteins for cervical intraepithelial neoplasia 2/3: a randomised, double-blind, placebo-controlled phase 2b trial. Lancet. 2016;386:2078–2088. doi: 10.1016/S0140-6736(15)00239-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Sharma A, Koldovsky U, Xu S, Mick R, Roses R, Fitzpatrick E, Weinstein S, Nisenbaum H, Levine BL, Fox K, Zhang P, Koski G, Czerniecki BJ. HER-2 pulsed dendritic cell vaccine can eliminate HER-2 expression and impact ductal carcinoma in situ. Cancer. 2012;118:4354–4362. doi: 10.1002/cncr.26734. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Soliman H, Khambati F, Han HS, Ismail-Khan R, Bui MM, Sullivan DM, Antonia S. A phase 1/2 study of Ad. p53 DC vaccine with indoximod immunotherapy in metastatic breast cancer. Oncotarget. 2018;10:10110–10117. doi: 10.18632/oncotarget.24118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Abdellateif MS, Shaarawy SM, Kandeel EZ, El-Habashy AH, Salem ML, El-Houseini ME. A novel potential effective strategy for enhancing the antitumor immune response in breast cancer patients using a viable cancer cell-dendritic cell-based vaccine. Oncol Lett. 2018;16:529–535. doi: 10.3892/ol.2018.8631. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Fernández A, Oliver L, Alvarez R, Fernández LE, Lee KP, Mesa C. Adjuvants and myeloid-derived suppressor cells: enemies or allies in therapeutic cancer vaccination. Hum Vaccin Immunother. 2014;10:3251–3260. doi: 10.4161/hv.29847. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Benavides LC, Gates JD, Carmichael MG, Patil R, Holmes JP, Hueman MT, Mittendorf EA, Craig D, Stojadinovic A, Ponniah S, Peoples GE. The impact of HER2/neu expression level on response to the E75 vaccine: from U.S. military cancer institute clinical trials group study I-01 and I-02. Clin Cancer Res. 2009;15:2895–2904. doi: 10.1158/1078-0432.CCR-08-1126. [DOI] [PubMed] [Google Scholar]
- 14.Clifton GT, Litton JK, Arrington K, Ponniah S, Ibrahim NK, Gall V, Alatrash G, Peoples GE, Mittendorf EA. Results of a phase I b trial of combination immunotherapy with a CD8+ T cell eliciting vaccine and trastuzumab inbreast cancer patients. Ann Oncol. 2017;24:2161–2167. doi: 10.1245/s10434-017-5844-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Anstasopoulou EA, Tzonis P, Fortis SP, Mahaira L, Vaxevanis C, Ardavanis A, Mittendorf EA, Peoples GE, Baxevanis CN, Papamichail M, Perez SA. DRB1* 11 allele expression and HER2 pre-existing immunity may predict benefit in breast cancer patients vaccinated with the HER2 modified AE37 peptide vaccine. J Immuno Ther Cancer. 2015;3:P427. [Google Scholar]
- 16.Spellman A, Tang SC. Immunotherapy for breast cancer: past, present, and future. Cancer Metastasis Rev. 2016;35:525–546. doi: 10.1007/s10555-016-9654-9. [DOI] [PubMed] [Google Scholar]
- 17.Robidoux A, Tang G, Rastogi P, Geyer CE Jr, Azar CA, Atkins JN, Fehrenbacher L, Bear HD, Baez-Diaz L, Sarwar S, Margolese RG, Farrar WB, Brufsky AM, Shibata HR, Bandos H, Paik S, Costantino JP, Swain SM, Mamounas EP, Wolmark N. Lapatinib as a component of neoadjuvant therapy for HER2-positive operable breast cancer (NSABP protocol B-41): an open-label, randomised phase 3 trial. Lancet Oncol. 2013;14:1183–1192. doi: 10.1016/S1470-2045(13)70411-X. [DOI] [PubMed] [Google Scholar]
- 18.Carey LA, Berry DA, Cirrincione CT, Barry WT, Pitcher BN, Harris LN, Ollila DW, Krop IE, Henry NL, Weckstein DJ, Anders CK, Singh B, Hoadley KA, Iglesia M, Cheang MC, Perou CM, Winer EP, Hudis CA. Molecular heterogeneity and response to neoadjuvant human epidermal growth factor receptor 2 targeting in CALGB 40601, a randomized phase III trial of paclitaxel plus trastuzumab with or without lapatinib. J. Clin. Oncol. 2016;34:542–549. doi: 10.1200/JCO.2015.62.1268. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Powles RL, Redmond D, Sotiriou C, Loi S, Fumagalli D, Nuciforo P, Harbeck N, de Azambuja E, Sarp S, Di Cosimo S, Huober J, Baselga J, Piccart-Gebhart M, Elemento O, Pusztai L, Hatzis C. Association of T cell receptor repertoire use with response to combined trastuzumablapatinib treatment of HER2-positive breast cancer: secondary analysis of the neo ALTTO randomized clinical trial. JAMA Oncol. 2018;4:e181564. doi: 10.1001/jamaoncol.2018.1564. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Loi S, Andre F, Maibach R, Hui R, Bartsch R, Jerusalem G, Gombos A, Campone M, Bonnefoi H, Bachelot T. PANACEA (IBCSG 45-13/BIG 4-13): a phase Ib/II trial evaluating the efficacy of pembrolizumab and trastuzumab in patients with trastuzumab-resistant, HER2-positive, metastatic breast cancer. Cancer Res. 2016;76:OT3-01-05. [Google Scholar]
- 21.Bartsch R, Bergen E. ASCO 2017: highlights in breast cancer. Memo. 2017;10:228–232. doi: 10.1007/s12254-017-0368-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Askoxylakis V, Ferraro GB, Kodack DP, Badeaux M, Shankaraiah RC, Seano G, Kloepper J, Vardam T, Martin JD, Naxerova K, Bezwada D, Qi X, Selig MK, Brachtel E, Duda DG, Huang P, Fukumura D, Engelman JA, Jain RK. Preclinical efficacy of ado-trastuzumab emtansine in the brain microenvironment. J Natl Cancer Inst. 2015;108:763. doi: 10.1093/jnci/djv313. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Keith KC, Lee Y, Ewend MG, Zagar TM, Anders CK. Activity of trastuzumabemtansine (TDM1) in HER2-positive breast cancer brain metastases: a case series. Cancer Treat Commun. 2016;7:43–46. doi: 10.1016/j.ctrc.2016.03.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Borges VF, Ferrario C, Aucoin N, Falkson C, Khan Q, Krop I, Welch S, Conlin A, Chaves J, Bedard PL, Chamberlain M, Gray T, Vo A, Hamilton E. Tucatinib combined with Ado-trastuzumab emtansine in advanced ERBB2/HER2- positive metastatic breast cancer: a phase 1b clinical trial. JAMA Oncol. 2018;4:1214–1220. doi: 10.1001/jamaoncol.2018.1812. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Rugo HS, Im SA, Wright GLS, Escriva-de-Romani S, DeLaurentiis M, Cortes J. SOPHIA primary analysis: a phase 3 (P3) study of margetuximab (M) + chemotherapy (C) versus trastuzumab (T) + C in patients (pts) with HER2+ metastatic (met) breast cancer (MBC) after prior anti-HER2 therapies (Tx) J. Clin. Oncol. 2019;37:1000. [Google Scholar]
- 26.Mandó P, Rivero SG, Rizzo MM, Pinkasz M, Levy EM. Targeting ADCC: a different approach to HER2 breast cancer in the immunotherapy era. Breast. 2021;60:15–25. doi: 10.1016/j.breast.2021.08.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Rugo HS, Im SA, Cardoso F, Cortés J, Curigliano G, Musolino A. Efficacy of margetuximab vs trastuzumab in patients with pretreated ERBB2-positive advanced breast cancer: a phase 3 randomized clinical trial. JAMA Oncol. 2021;7:573–584. doi: 10.1001/jamaoncol.2020.7932. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Mcdonagh CF, Huhalov A, Harms BD, Adams S, Paragas V, Oyama S, Zhang B, Luus L, Overland R, Nguyen S, Gu J, KohliN N, Wallace M, Feldhaus MJ, Kudla AJ, Schoeberl B, Nielsen UB. Antitumor activity of a novel bispecific antibody that targets the ErbB2/ErbB3 oncogenic unit and inhibits heregulin-induced activation of ErbB3. Mol Cancer Ther. 2012;11:582–593. doi: 10.1158/1535-7163.MCT-11-0820. [DOI] [PubMed] [Google Scholar]
- 29.Jager M, Schoberth A, Ruf P, Hess J, Lindhofer H. The trifunctional antibody ertumaxomab destroys tumor cells that express low levels of human epidermal growth factor receptor 2. Cancer Res. 2009;69:4270–4276. doi: 10.1158/0008-5472.CAN-08-2861. [DOI] [PubMed] [Google Scholar]
- 30.Topp MS, Gökbuget N, Stein AS, Zugmaier G, O’Brien S, Bargou RC, Dombret H, Fielding AK, Heffner L, Larson RA, Neumann S, Foà R, Litzow M, Ribera JM, Rambaldi A, Schiller G, Brüggemann M, Horst HA, Holland C, Jia C, Maniar T, Huber B, Nagorsen D, Forman SJ, Kantarjian HM. Safety and activity of blinatumomab for adult patients with relapsed or refractory bprecursor acute lymphoblastic leukaemia: a multicentre, single-arm, phase 2 study. Lancet Oncol. 2015;16:57–66. doi: 10.1016/S1470-2045(14)71170-2. [DOI] [PubMed] [Google Scholar]
- 31.Winer EP, Dang T, Karantza V, Su SC. KEYNOTE-119: phase III study of single-agent pembrolizumab versus single-agent chemotherapy for metastatic triple negative breast cancer (mTNBC) J. Clin. Oncol. 2016:TPS1102. [Google Scholar]
- 32.Cortes J, Andre F, Goncalves A, Kümmel S, Martín Mi, Schmid P, Schuetz F, Swain SM, Easton V, Pollex E, Deurloo R, Dent R. IMpassion132 phase III trial: atezolizumab and chemotherapy in early relapsing metastatic triple-negative breast cancer. Future Oncol. 2019;15:1951–1961. doi: 10.2217/fon-2019-0059. [DOI] [PubMed] [Google Scholar]
- 33.Voorwerk L, Slagter M, Horlings HM, Voorwerk L, Slagter M, Horlings HM, Sikorska K, van de Vijver KK, Maaker MD, Kluin RJ, Warren S, Ong SF, Wiersma TG, Russell NS, Lalezari F, Schouten PC, Bakker NA, Ketelaars SL, Peters D, Lange CA, Werkhoven EV, Tinteren HV, Mandjes IA, Kemper I, Onderwater S, Chalabi M, Wilgenhof S, Haanen JB, Salgado R, de Visser KE, Sonke G, Wessels LF, Linn SC, Schumacher TN, Blank CU, Kok M. Immune in duction strategies in metastatic triple-negative breast cancer to enhance the sensitivity to PD-1 blockade: the TONIC trial. Nat Med. 2019;25:920–928. doi: 10.1038/s41591-019-0432-4. [DOI] [PubMed] [Google Scholar]
- 34.Hu ZI, Ho AY, Marthur HL. Combined radiation therapy and immune checkpoint blockade therapy for breast cancer. Int J Radiat Oncol Biol Phys. 2017;99:153–164. doi: 10.1016/j.ijrobp.2017.05.029. [DOI] [PubMed] [Google Scholar]
- 35.Force J, Leal JHS, Mcarthur HL. Checkpoint blockade strategies in the treatment of breast cancer: where we are and where we are heading. Curr Treat Options Oncol. 2019;20:35. doi: 10.1007/s11864-019-0634-5. [DOI] [PubMed] [Google Scholar]
- 36.Kang J, Demaria S, Formenti S. Current clinical trials testing the combination of immunotherapy with radiotherapy. J Immunother Cancer. 2016;4:51. doi: 10.1186/s40425-016-0156-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Heeke AL, Tan AR. Checkpoint inhibitor therapy for metastatic triple-negative breast cancer. Cancer Metastasis Rev. 2021;40:537–547. doi: 10.1007/s10555-021-09972-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Loibl S, Untch M, Burchardi N, Huober J, Sinn BV, Blohmer JU, Grischke EM, Furlanetto J, Tesch H, Hanusch C, Engels K, Rezai M, Jackisch C, Schmitt WD, von Minckwitz G, Thomalla J, Kümmel S, Rautenberg B, Fasching PA, Weber K, Rhiem K, Denkert C, Schneeweiss A. A randomised phase II study investigating durvalumab in addition to an anthracycline taxane based neoadjuvant therapy in early triple-negative breast cancer: clinical results and biomarker analysis of GeparNuevo study. Ann Oncol. 2019;30:1279–1288. doi: 10.1093/annonc/mdz158. [DOI] [PubMed] [Google Scholar]
- 39.Rugo HS, Delord JP, Im SA, Ott PA, Piha-Paul SA, Bedard PL, Sachdev J, Tourneau CL, Brummelen EM, Varga A, Salgado R, Loi S, Saraf S, Pietrangelo D, Karantza V, Tan AR. Safety and antitumor activity of pembrolizumab in patients with estrogen receptor-positive/human epidermal growth factor receptor 2-negative advanced breast cancer. Clin Cancer Res. 2018;24:2804–2811. doi: 10.1158/1078-0432.CCR-17-3452. [DOI] [PubMed] [Google Scholar]
- 40.Dirix LY, Takacs I, Jerusalem G, Nikolinakos P, Arkenau HT, Forero-Torres A, Boccia R, Lippman ME, Somer R, Smakal M, Emens LA, Hrinczenko B, Edenfield W, Gurtler J, von Heydebreck A, Grote HJ, Chin K, Hamilton EP. Avelumab, an antiPD-L1 antibody, in patients with locally advanced or metastatic breast cancer: a phase 1b JAVELIN solid tumor study. Breast Cancer Res Treat. 2018;167:671–686. doi: 10.1007/s10549-017-4537-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Chia S, Bedard PL, Hilton J, Amir E, Gelmon K, Goodwin R, Villa D, Cabanero M, Tu D, Tsao M, Seymour L. A phase Ib trial of durvalumab in combination with trastuzumab in HER2-positive metastatic breast cancer (CCTG IND.229) Oncologist. 2019;24:1439–1445. doi: 10.1634/theoncologist.2019-0321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Zheng HM, Feng YJ, Xu J, Chen HT, Jing LT, Zhong W, Wu X. Progress of immunotherapy and its application in triple negative breast cancer. Cancer Res Prev Treat. 2020;47:889–893. [Google Scholar]
- 43.Loi S, GiobbieHurder A, Gombos A, Bachelot T, Hui R, Curigliano G, Campone M, Biganzoli L, Bonnefoi H, Jerusalem G. Phase Ib/II study evaluating safety and effificacy of pembrolizumab and trastuzumab in patients with trastuzumab-resistant HER2-positive metastatic breast cancer: results from the PANACEA (IBCSG 45-13/KEYNOTE-014) study. Cancer Res. 2017 [Google Scholar]
- 44.Urruticoechea A, Rizwanullah M, Im SA, Ruiz ACS, Láng I, Tomasello G, Douthwaite H, Badovinac Crnjevic T, Heeson S, Eng-Wong J, Muñoz M. Randomized phase III trial of trastuzumab plus capecitabine with or without pertuzumab in patients with human epidermal growth factor receptor 2-positive metastatic breast cancer who experienced disease progression during or after trastuzumab-based therapy. J. Clin. Oncol. 2017;35:3030–3038. doi: 10.1200/JCO.2016.70.6267. [DOI] [PubMed] [Google Scholar]
- 45.Kern R, Panis C. CTLA-4 expression and its clinical significance in breast cancer. Arch Immunol Ther Exp (Warsz) 2021;69:16. doi: 10.1007/s00005-021-00618-5. [DOI] [PubMed] [Google Scholar]
- 46.Emens LA, Ascierto PA, Darcy PK, Demaria S, Eggermont AMM, Redmond WL, Seliger B, Marincola FM. Cancer immunotherapy: opportunities and challenges in the rapidly evolving clinical landscape. Eur J Cancer. 2017;81:116–129. doi: 10.1016/j.ejca.2017.01.035. [DOI] [PubMed] [Google Scholar]
- 47.Vonderheide RH, Lorusso PM, Khalili M, Gartner EM, Khaira D, Soulieres D, Dorazio P, Trosko JA, Rüter J, Mariani GL, Usari T, Domchek SM. Tremelimumab in combination with exemestane in patients with advanced breast cancer and treatment-associated modulation of inducible costimulator expression on patient T cells. Clin Cancer Res. 2010;16:3485–3494. doi: 10.1158/1078-0432.CCR-10-0505. [DOI] [PubMed] [Google Scholar]
- 48.Weber J, Mandala M, Del Vecchio M, Gogas HJ, Arance AM, Cowey CL, Dalle S, Schenker M, Chiarion-Sileni V, Marquez-Rodas I, Grob JJ, Butler MO, Middleton MR, Maio M, Atkinson V, Queirolo P, Gonzalez R, Kudchadkar RR, Smylie M, Meyer N, Mortier L, Atkins MB, Long GV, Bhatia S, Lebbé C, Rutkowski P, Yokota K, Yamazaki N, Kim TM, de Pril V, Sabater J, Qureshi A, Larkin J, Ascierto PA CheckMate 238 Collaborators. Adjuvant nivolumab versus ipilimumab in resected stage III or IV melanoma. N Engl J Med. 2017;377:1824–1835. doi: 10.1056/NEJMoa1709030. [DOI] [PubMed] [Google Scholar]
- 49.Curran MA, Montalvo W, Yagita H, Allison JP. PD-1 and CTLA-4 combination blockade expands infiltrating T cells and reduces regulatory T and myeloid cells within B16 melanoma tumors. Proc Natl Acad Sci U S A. 2010;107:4275–4280. doi: 10.1073/pnas.0915174107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Hodi FS, Chiarion-Sileni V, Gonzalez R, Grob JJ, Rutkowski P, Cowey CL, Lao CD, Schadendorf D, Wagstaff J, Dummer R, Ferrucci PF, Smylie M, Hill A, Hogg D, Marquez-Rodas I, Jiang J, Rizzo J, Larkin J, Wolchok JD. Nivolumab plus ipilimumab or nivolumab alone versus ipilimumab alone in advanced melanoma (CheckMate 067): 4-year outcomes of a multicentre, randomised, phase 3 trial. Lancet Oncol. 2018;19:1480–1492. doi: 10.1016/S1470-2045(18)30700-9. [DOI] [PubMed] [Google Scholar]
- 51.Pan K, Guan XX, Li YQ, Zhao JJ, Li JJ, Qiu HJ, Weng DS, Wang QJ, Liu Q, Huang LX, He J, Chen SP, Ke ML, Zeng YX, Xia JC. Clinical activity of adjuvant cytokine-induced killer cell immunotherapy in patients with postmastectomy triple-negative breast cancer. Clin Cancer Res. 2014;20:3003–3011. doi: 10.1158/1078-0432.CCR-14-0082. [DOI] [PubMed] [Google Scholar]
- 52.Bates JP, Derakhshandeh R, Jones L, Webb TJ. Mechanisms of immune evasion in breast cancer. BMC Cancer. 2018;18:556. doi: 10.1186/s12885-018-4441-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Zhang L, Lu X. Injection of tumor infiltrating lymphocytes for prevention of tumor recurrence in patients with breast cancel. Journal of Practical Oncology. 2004;26:423–424. [Google Scholar]
- 54.Zacharakis N, Chinnasamy H, Black M, Xu H, Lu YC, Zheng Z, Pasetto A, Langhan M, Shelton T, Prickett T, Gartner J, Jia L, Trebska-McGowan K, Somerville RP, Robbins PF, Rosenberg SA, Goff SL, Feldman SA. Immune recognition of somatic mutations leading to complete durable regression in metastatic breast cancer. Nat Med. 2018;24:724–730. doi: 10.1038/s41591-018-0040-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Ahmed N, Brawley VS, Hegde M, Robertson C, Ghazi A, Gerken C, Liu E, Dakhova O, Ashoori A, Corder A, Gray T, Wu MF, Liu H, Hicks J, Rainusso N, Dotti G, Mei Z, Grilley B, Gee A, Rooney CM, Brenner MK, Heslop HE, Wels WS, Wang LL, Anderson P, Gottschalk S. Human epidermal growth factor receptor 2 (HER2)-specific chimeric antigen receptor-modified T cells for the immunotherapy of HER2-positive sarcoma. J. Clin. Oncol. 2015;33:1688–1696. doi: 10.1200/JCO.2014.58.0225. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Li L, Cheng Y, Zheng M, Jiang H. Advances in research on tumor immunotherapy. Chinese Journal of Nature. 2021;43:391–399. [Google Scholar]
- 57.Lum LG, Thakur A, Al-Kadhimi Z, Colvin GA, Cummings FJ, Legare RD, Dizon DS, Kouttab N, Maizel A, Colaiace W, Liu Q, Rathore R. Targeted T-cell therapy in stage IV breast cancer: a phase I clinical trial. Clin Cancer Res. 2015;21:2305–2314. doi: 10.1158/1078-0432.CCR-14-2280. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Byrd TT, Fousek K, Pignata A, Szot C, Samaha H, Seaman S, Dobrolecki L, Salsman VS, Oo HZ, Bielamowicz K, Landi D, Rainusso N, Hicks J, Powell S, Baker ML, Wels WS, Koch J, Sorensen PH, Deneen B, Ellis MJ, Lewis MT, Hegde M, Fletcher BS, St Croix B, Ahmed N. TEM8/ANTXR1-specific CAR T cells as a targeted therapy for triple-negative breast cancer. Cancer Res. 2018;78:489–500. doi: 10.1158/0008-5472.CAN-16-1911. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Wang J, Zhou P. New approaches in CAR-T cell immunotherapy for breast cancer. Adv Exp Med Biol. 2017;1026:371–381. doi: 10.1007/978-981-10-6020-5_17. [DOI] [PubMed] [Google Scholar]
- 60.Yarchoan M, Hopkins A, Jaffee EM. Tumor mutational burden and response rate to PD-1 inhibition. N Engl J Med. 2017;377:2500–2501. doi: 10.1056/NEJMc1713444. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Park SE, Park K, Lee E, Kim JY, Ahn JS, Im YH, Lee C, Jung H, Cho SY, Park WY, Cristescu R, Park YH. Clinical implication of tumor mutational burden in patients with HER2-positive refractory metastatic breast cancer. Oncoimmunology. 2018;7:e1466768. doi: 10.1080/2162402X.2018.1466768. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Schmid P, Adams S, Rugo HS, Schneeweiss A, Barrios CH, Iwata H, Diéras V, Hegg R, Im SA, Shaw Wright G, Henschel V, Molinero L, Chui SY, Funke R, Husain A, Winer EP, Loi S, Emens LA IMpassion130 Trial Investigators. Atezolizumab and nab-paclitaxel in advanced triple-negative breast cancer. N Engl J Med. 2018;379:2108–2121. doi: 10.1056/NEJMoa1809615. [DOI] [PubMed] [Google Scholar]
- 63.Emens LA, Molinero L, Adams S, Rugo HS, Schneeweiss A, Diéras V, Iwata H, Barrios CH, Nechaeva M, Winer EP. Tumour mutational burden and clinical outcomes with first-line atezolizumab and nab-paclitaxel in triple negative breast cancer: exploratory analysis of the phase III IMpassion130 trial. Ann Oncol. 2020;31:360–361. [Google Scholar]
- 64.Zhu Y, Zhu X, Tang C, Guan X, Zhang W. Progress and challenges of immunotherapy in triple-negative breast cancer. Biochim Biophys Acta Rev Cancer. 2021;1876:188593. doi: 10.1016/j.bbcan.2021.188593. [DOI] [PubMed] [Google Scholar]
- 65.Karn T, Denkert C, Weber KE, Holtrich U, Hanusch C, Sinn BV, Higgs BW, Jank P, Sinn HP, Huober J, Becker C, Blohmer JU, Marmé F, Schmitt WD, Wu S, van Mackelenbergh M, Müller V, Schem C, Stickeler E, Fasching PA, Jackisch C, Untch M, Schneeweiss A, Loibl S. Tumor mutational burden and immune infiltration as independent predictors of response to neoadjuvant immune checkpoint inhibition in early TNBC in GeparNuevo. Ann Oncol. 2020;31:1216–1222. doi: 10.1016/j.annonc.2020.05.015. [DOI] [PubMed] [Google Scholar]
- 66.Yin Y, Li W, Huang X. HER-2 amplification is associated with higher tumor mutation burden in breast cancer. Ann Oncol. 2018;29:146. [Google Scholar]
- 67.Xu J, Bao H, Wu X, Wang X, Shao YW, Sun T. Elevated tumor mutation burden and immunogenic activity in patients with hormone receptor-negative or human epidermal growth factor receptor 2-positive breast cancer. Oncol Lett. 2019;18:449–455. doi: 10.3892/ol.2019.10287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Kawaji H, Kubo M, Yamashita N, Yamamoto H, Kai M, Kajihara A, Yamada M, Kurata K, Kaneshiro K, Harada Y, Hayashi S, Shimazaki A, Mori H, Akiyoshi S, Oki E, Oda Y, Baba E, Mori M, Nakamura M. Comprehensive molecular profiling broadens treatment options for breast cancer patients. Cancer Med. 2021;10:529–539. doi: 10.1002/cam4.3619. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Barroso SR, Jain E, Cohen O, Kim D, Buendia JB, Winer E, Lin N, Tolaney SM, Wagle N. Prevalence and mutational determinants of high tumor mutation burden in breast cancer. Ann Oncol. 2020;31:387–394. doi: 10.1016/j.annonc.2019.11.010. [DOI] [PubMed] [Google Scholar]
- 70.Schnidrig D, Turajlic S, Litchfield K. Tumour mutation burden: primary versus metastatic tissue creates systematic bias. Immunooncol Technol. 2019;4:8–14. doi: 10.1016/j.iotech.2019.11.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Lu S, Stein JE, Rimm DL, Wang DW, Bell JM, Johnson DB, Sosman JA, Schalper KA, Anders RA, Wang H, Hoyt C, Pardoll DM, Danilova L, Taube JM. Comparison of biomarker modalities for predicting response to PD-1/PD-L1 checkpoint blockade: a systematic review and meta-analysis. JAMA Oncol. 2019;5:1195–1204. doi: 10.1001/jamaoncol.2019.1549. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Criscitiello C, Esposito A, Trapani D, Curigliano G. Prognostic and predictive value of tumor infiltrating lymphocytes in early breast cancer. Cancer Treat Rev. 2016;50:205–207. doi: 10.1016/j.ctrv.2016.09.019. [DOI] [PubMed] [Google Scholar]
- 73.Stanton SE, Disis ML. Clinical significance of tumor-infiltrating lymphocytes in breast cancer. J Immunother Cancer. 2016;4:59. doi: 10.1186/s40425-016-0165-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Masili-Oku SM, Almeida BGL, Bacchi CE, Filassi JR, Baracat EC, Carvalho FM. Lymphocyte-predominant triple-negative breast carcinomas in premenopausal patients: lower expression of basal immunohistochemical markers. Breast. 2017;31:34–39. doi: 10.1016/j.breast.2016.10.012. [DOI] [PubMed] [Google Scholar]
- 75.Adams S, Loi S, Toppmeyer D, Cescon DW, De Laurentiis M, Nanda R, Winer EP, Mukai H, Tamura K, Armstrong A, Liu MC, Iwata H, Ryvo L, Wimberger P, Rugo HS, Tan AR, Jia L, Ding Y, Karantza V, Schmid P. Pembrolizumab monotherapy for previously untreated, PD-L1-positive, metastatic triple-negative breast cancer: cohort B of thephase II KEYNOTE-086 study. Ann Oncol. 2019;30:405–411. doi: 10.1093/annonc/mdy518. [DOI] [PubMed] [Google Scholar]
- 76.Kulangara K, Zhang N, Corigliano E, Guerrero L, Waldroup S, Jaiswal D, Ms MJ, Shah S, Hanks D, Wang J, Lunceford J, Savage MJ, Juco J, Emancipator K. Clinical utility of the combined positive score for programmed death ligand-1 expression and the approval of pembrolizumab for treatment of gastric cancer. Arch Pathol Lab Med. 2019;143:330–337. doi: 10.5858/arpa.2018-0043-OA. [DOI] [PubMed] [Google Scholar]
- 77.Ancevski HK, Socinski MA, Villaruz LC. PD-L1 testing in guiding patient selection for PD-1/PD-L1 inhibitor therapy in lung cancer. Mol Diagn Ther. 2018;22:1–10. doi: 10.1007/s40291-017-0308-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Hirsch FR, McElhinny A, Stanforth D, Ranger-Moore J, Jansson M, Kulangara K, Richardson W, Towne P, Hanks D, Vennapusa B, Mistry A, Kalamegham R, Averbuch S, Novotny J, Rubin E, Emancipator K, McCaffery I, Williams JA, Walker J, Longshore J, Tsao MS, Kerr KM. PD-L1 immunohistochemistry assays for lung cancer: results from phase 1 of the blueprint PD-L1 IHC assay comparison project. J Thorac Oncol. 2017;12:208–222. doi: 10.1016/j.jtho.2016.11.2228. [DOI] [PubMed] [Google Scholar]
- 79.Schmid P, Rugo HS, Adams S, Schneeweiss A, Barrios CH, Iwata H, Diéras V, Henschel V, Molinero L, Chui SY, Maiya V, Husain A, Winer EP, Loi S, Emens LA IMpassion130 Investigators. Atezolizumab plus nab-paclitaxel as first-line treatment for unresectable, locally advanced or metastatic triple-negative breast cancer (IMpassion130): updated efficacy results from a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 2020;21:44–59. doi: 10.1016/S1470-2045(19)30689-8. [DOI] [PubMed] [Google Scholar]