We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine
Meta-AnalysisOpen Accesscc iconby iconnc iconnd icon

A meta-analysis of application of PD-1/PD-L1 inhibitor-based immunotherapy in unresectable locally advanced triple-negative breast cancer

    Wei Zhang‡

    Department of Breast Surgery, Ningbo No. 2 Hospital, Ningbo, Zhejiang, 315010, China

    ‡Authors contributed equally

    Search for more papers by this author

    ,
    Yujing He‡

    The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China

    ‡Authors contributed equally

    Search for more papers by this author

    ,
    Yuning Tang

    The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China

    ,
    Wei Dai

    The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China

    ,
    Yuexiu Si

    School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China

    ,
    Feiyan Mao

    Department of General Surgery, Ningbo No. 2 Hospital, Ningbo, Zhejiang, 315010, China

    ,
    Jiaxuan Xu

    The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China

    ,
    Chiyuan Yu

    The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China

    &
    Xing Sun

    *Author for correspondence: Tel.: +18 757 495 672;

    E-mail Address: wake2022@163.com

    Department of General Surgery, Ningbo No. 2 Hospital, Ningbo, Zhejiang, 315010, China

    Published Online:https://doi.org/10.2217/imt-2023-0023

    Abstract

    Aims: The purpose of this study was to explore the efficacy of immunotherapy for patients with triple-negative breast cancer (TNBC). Materials & methods: Randomized clinical trials comparing immunotherapy with chemotherapy for advanced TNBC patients were included. Results: A total of six articles (3183 patients) were eligible for this meta-analysis. PD-1/PD-L1 inhibitor-based immunotherapy combined with chemotherapy can significantly increase the progression-free survival (hazard ratio [HR] = 0.82; 95% CI = 0.76–1.14; p < 0.001) of unresectable locally advanced or metastatic TNBC patients without effect on overall survival, compared with chemotherapy. Conclusion: PD-1/PD-L1 inhibitors-based immunotherapy can safely improve progression-free survival in patients with unresectable locally advanced or metastatic TNBC, but has no effect on overall survival.

    Plain language summary

    Breast cancer is a malignant tumor. It is most common in females. Triple-negative breast cancer is one type of malignant tumor that is not sensitive to treatment and is prone to recurrence. It can easily lead to death. Treatment mainly relies on chemotherapy. Immunotherapy is a new treatment method ad includes PD-1/L1 inhibitors. This research was conducted to assess its effects. Immunotherapy has good effects and can alleviate symptoms. It can improve prognosis and extend life. It has some side effects, mainly in the lungs and thyroid, but these side effects are controllable.

    Breast cancer (BC) is a life-threatening disease [1,2] and has the highest incidence in females worldwide [3,4]. Although the overall prognosis of BC is relatively acceptable, the prognosis of triple-negative BC (TNBC) is considerably poor [5–8]. TNBC, accounting for 15–20% of breast cancers, is more aggressive and more likely to metastasize and recur [9–11]. Due to its special molecular phenotype [4,12], TNBC is not sensitive to endocrine therapy or molecular-targeted therapy [8,13]. Standardized TNBC treatment regimens remain up for discussion [14,15]. Cytotoxic chemotherapy is currently the main treatment for TNBC [16–18]. However, studies in the past two decades have shown the objective response rate of systemic chemotherapy in adjuvant and metastatic treatment to be only 30–40% [19,20]. Moreover, even if TNBC patients are treated with systemic chemotherapy, 5-year survival can only be expected in <30% of metastatic patients after diagnosis [21–23]. Since residual metastatic disease can easily result in tumor recurrence, almost all patients with metastasis will die due to organ failure, reflecting unsatisfactory treatment overall [24,25].

    Therefore, the development of new treatment options and targets is urgently needed for TNBC patients. Over the past few years, immunotherapy has been a novel treatment option for TNBC [26,27]. TNBC has a higher number of tumor-infiltrating lymphocytes (TILs) and higher PD-L1 protein and/or mRNA expression compared with other BC subtypes [28,29]. The ligand, PD-L1, and its receptor, PD-1, are involved in the regulation of immune tolerance [30–32]. Therefore, researchers have developed a series of PD-1/PD-L1 inhibitors, such as atezolizumab [33], pembrolizumab [34], nivolumab [35], durvalumab [36], avelumab [37] and so on, and conducted experiments to identify biomarkers for PD-1/PD-L1 inhibitor therapy. At present, the most important biomarker is considered to be the expression of PD-L1, and related evaluation methods include tumor cell proportion score, combined positive score and immune cell proportion score. However, there is a discrepancy between the expression of PD-L1 and the response to PD-1/PD-L1 inhibitor immunotherapy, and there remain doubts about PD-L1 expression as a key point for PD-1/PD-L1 inhibitor therapy. Many studies are still exploring reliable biomarkers beyond PD-L1, such as microsatellite instability, mismatch repair, tumor mutation burden and so on. Therefore, many studies have conducted immunotherapy with PD-1/PD-L1 inhibitors for TNBC without limiting the expression of PD-L1.

    Given the diversity of survival results among different immunotherapy regimens, the details of newly proposed immunotherapy regimens and immune agents have been hotly discussed issues among healthcare professionals and researchers in related fields. The KEYNOTE-119 trial [38] found that the median overall survival (OS) was 9.9 months in the pembrolizumab group and 10.8 months in the chemotherapy group without a statistically significant difference in OS (hazard ratio [HR] = 0.97; 95% CI = 0.82–1.15), while the SAFIR02-BREAST IMMUNO trial [39] indicated that durvalumab did improve OS in patients with TNBC (HR = 0.54; p = 0.037). Therefore, this meta-analysis was designed to evaluate the efficacy and safety of immunotherapy, mainly focusing on PD-1/PD-L1 inhibitors, in locally advanced or metastatic TNBC that is unresectable, and provide a primary exploration of immunotherapy for TNBC.

    Methods

    Literature search

    A search of relevant studies investigating the efficacy and safety of immunotherapy in TNBC patients published before October 2021 was conducted in PubMed, EBSCO, Web of Science and Cochrane Library databases. The keywords used were: “triple-negative breast cancer,” “programmed cell death ligand 1 inhibitor” and “immunotherapy.” The complete retrieval formula that was used to identify the related studies included: “triple negative breast cancer” OR “TNBC” OR “triple-negative breast carcinoma” OR “triple-negative breast neoplasms” AND “pembrolizumab” OR “nivolumab” OR “atezolizumab” OR “durvalumab” OR “avelumab” OR “cemiplimab” OR “programmed cell death 1” OR “programmed cell death ligand 1” OR “PD-L1” OR “PD-1” OR “immunotherapy” OR “immune checkpoint.” Only the most recent article or the one with the highest quality was included when duplicates were found. Literature in the reference lists of retrieved studies and recent reviews were also thoroughly reviewed to avoid missing possible studies that may need to be included. The population, intervention/exposure, comparison, outcome and setting criteria were used to describe the research aim. This meta-analysis was conducted following the guidelines of the Preferred Reporting Items for Systematic Review and Meta-Analysis 2009 Checklist [40]. The Prospero registration number for this meta-analysis is CRD42021289859.

    Selection criteria

    The specific eligibility criteria for this study were as follows: all included studies were randomized controlled trials (RCTs) and were limited to clinical studies. The included studies assessed only patients who were diagnosed with unresectable locally advanced or metastatic TNBC or had been categorized as a subgroup with accessible data attached. Patients in the experimental groups in the RCTs were treated with PD-1/PD-L1 inhibitor-based immunotherapy while those in the control groups were treated with other treatments. All studies had full-text articles available. Exclusion criteria: patients with early TNBC or postoperative adjuvant therapy; studies not reporting relevant outcomes or detailed data; preclinical studies or phase I clinical trials; and studies not published in English.

    Two authors independently applied the search strategy to the selected studies and independently reviewed the titles and abstracts of these articles to judge whether they met the inclusion criteria. Full texts were retrieved in the case of doubts. When necessary, authors were contacted for additional information. In case of differences of opinion, a discussion was held with a third author. When consensus failed to be reached, the study was excluded.

    Data extraction & quality assessment

    A jointly agreed upon data collection form was used to extract all data. The extracted information included the author's name, year of publication, trial duration, NCT number, country, age of patients, therapeutic regimen, trial phase, follow-up time, number of patients, primary outcomes and secondary outcomes. To ensure the objectivity and accuracy of the input data, two researchers independently extracted data from each study. Disagreements were resolved by consensus or consultation with a third author. The quality of each RCT was recorded according to the following indicators from the Cochrane Risk-of-Bias assessment tool score: sequence generation, allocation concealment, blinding, incomplete outcome data, selective outcome reporting and free of other bias.

    Objectives & end points

    The primary objective of this meta-analysis was to assess the OS and progression-free survival (PFS) of patients treated with PD-1/PD-L1 inhibitor-based immunotherapy for unresectable locally advanced or metastatic TNBC compared with others. OS was defined as the proportion of tumor patients who survived after various comprehensive treatments from random allocation at the beginning of the trial to the latest follow-up time. The PFS was defined as the proportion of patients without disease progression or death for any reason from random allocation at the beginning of the trial to the latest follow-up time. The secondary objectives included safety and efficacy based on subgroup analysis of PD-L1 status.

    Statistical analysis

    RevMan 5.3.5 software for Windows® and Stata software version 12 (StataCrop, TX, USA) were used for analysis. Heterogeneity across included studies was tested by Q statistics and the I2 statistic. In accordance with the Cochrane manual and experimental characteristics, I2 values between 0 and 30% indicate mild or insignificant heterogeneity, 30∼70% indicate moderate heterogeneity and 70∼100% indicate high or significant heterogeneity [41]. The CIs of HRs and risk ratios (RRs) were set at 95% to determine the efficacy of immunotherapy. A random-effects model was applied to incorporate data from different therapeutic regimens to increase the credibility of the results. When more than ten studies [42,43] were included, sensitivity analysis and publication bias tests were performed to evaluate the stability and reliability of the results. Begg's rank correlation test was applied to test publication bias; p-values < 0.05 were considered statistically significant.

    Results

    Study selection

    Based on the previously described search formula, a total of 2346 relevant articles were identified through preliminary searches in PubMed, EBSCO, Web of Science and Cochrane Library databases. No other records were identified from other sources. A total of 393 duplicate articles were recognized and deleted; 1501 articles were excluded based on the content in the title or abstract. The remaining 452 articles were reviewed through full-text reading. Among them, 446 articles were eliminated because they were not RCTs (n = 354), had no TNBC (n = 60) or immunotherapy (n = 27) involved and were not in English (n = 5). Finally, 6 articles [39,44–48] comprising 3183 patients were eligible for this meta-analysis. The detailed search and study selection process is shown in Figure 1.

    Figure 1. Flowchart for selection of articles included in this meta-analysis comparing efficacy and safety of PD-1/PD-L1 inhibitor-based immunotherapy combined with chemotherapy and other treatments on unresectable locally advanced or metastatic triple-negative breast cancer.

    Study characteristics

    Of 3183 patients (1839 in the experimental groups and 1344 in the control groups), 55.2% were diagnosed with PD-L1-positive (PD-L1+) TNBC and 44.2% were diagnosed with PD-L1-negative (PD-L1-) TNBC. The remaining 0.6% of patients were unknown in terms of PD-L1 status. All participants were female. Four trials were in phase III and two were in phase II. Patients in the experimental groups were all treated with PD-1/PD-L1 inhibitor-based immunotherapy such as pembrolizumab, nivolumab, atezolizumab, durvalumab, avelumab and cemiplimab, while those in the control groups were treated with other treatments such as paclitaxel, gemcitabine, vinorelbine, capecitabine, eribulin or carboplatin. All six studies were published between 2020 and 2021, with median follow-up times ranging from 7.7 to 31.5 years. Two trials are still ongoing. The random allocation ratio was 2:1 in three of the included studies and 1:1 in the other three. Three studies were open-label trials, two were double-blind trials (participant, investigator) and one was a quadruple-blind trial (participant, care provider, investigator, outcomes assessor). All studies had accessible data for PFS and five studies had accessible data for OS. All studies defined PD-L1+ as the covering rate of PD-L1-stained tumor-infiltrating immune cells over the total tumor area is equal to more than 1% in the metastatic lesion sample. Most of the PD-L1 protein expressions were assessed by immunohistochemistry using the SP142 antibody. Details of the included clinical trials are listed in Table 1 & Supplementary Table 1.

    Table 1. Characteristics of included randomized controlled trials in meta-analysis comparing efficacy and safety of PD-1/PD-L1 inhibitor-based immunotherapy combined with chemotherapy and other treatments in unresectable locally advanced or metastatic triple-negative breast cancer.
    Author, yearCountryCharacteristicsTrial phasePrimary outcome measuresSecondary outcome measuresTherapeutic regimenNumber of ITT participants
    TreatmentControlTreatmentControl
    Bachelot, 2021FranceSAFIR02-BREAST IMMUNOIIPFSOS, ORR, AEsDurvalumabMaintenance chemotherapy4735
    Brufsky, 2021USCOLETIIPFS, OR, PR, CROS, DOR, EOT, AEsAtezolizumab + cobimetinib + paclitaxelCobimetinib + paclitaxel3247
    Cortes, 2020SpainKEYNOTE-355IIIPFS, OS, AEsORR, DOR, DCRPembrolizumab + chemotherapy (nab-paclitaxel, paclitaxel or gemcitabine + carboplatin)Placebo + chemotherapy (nab-paclitaxel, paclitaxel or gemcitabine + carboplatin)566281
    Miles, 2021UKIMpassion131IIIPFSOS, OR, CR, PR, DOR, TTD, AEsAtezolizumab + paclitaxelPlacebo + paclitaxel431220
    Schmid, 2020UKIMpassion130IIIPFS, OSOR, CR, PR, DOR, TTD, AEsAtezolizumab + nab-paclitaxelPlacebo + nab-paclitaxel451451
    Winer, 2021USKEYNOTE-119IIIOSPFS, ORR, DOR, DCR, AEsPembrolizumabChemotherapy (capecitabine, eribulin, gemcitabine or vinorelbine)312310

    AEs: Adverse events; CR: Complete response; DCR: Disease control rate; DOR: Duration of response; EOT: End of treatment; ITT: Intent-to-treat; OR: Overall response; ORR: Objective response rate; OS: Overall survival; PFS: Progression-free survival; PR: Partial response; TNBC: Triple-negative breast cancer; TTD: Time to deterioration.

    Bias risk assessment

    The Cochrane Collaboration tool was applied to objectively evaluate the quality of RCTs included in this meta-study. The tool employs six targets overall and risk of bias falls into either the ‘low risk’, ‘high risk' or 'unclear risk' categories. All included studies were of higher quality, according to the quality evaluation results (Supplementary Figure 1).

    OS of intention-to-treat & PD-L1+/- populations

    In five studies (1273 experimental patients and 1063 control patients) OS was selected to evaluate the efficacy of PD-1/PD-L1 inhibitor-based immunotherapy with other chemotherapy combined for the treatment of unresectable locally advanced or metastatic TNBC. Among them, two studies (283 experimental patients and 279 control patients) had available PD-L1- population data and four studies (597 experimental patients and 501 control patients) had available PD-L1+ population data. The analysis of the results showed that immunotherapy did not improve the OS of the intent-to-treat (ITT) population (HR = 0.90; 95% CI = 0.78–1.04; p = 0.144) with mild heterogeneity (I2 = 24.0%). Immunotherapy based on PD-1/PD-L1 inhibitors could not prolong the OS of unresectable locally advanced or metastatic TNBC compared with nonimmunotherapy treatment (Figure 2). In addition, a subgroup analysis based on PD-L1 status showed that there was no statistically significant difference in OS between the immunotherapy group and the nonimmunotherapy group no matter the TNBC subtype, whether PD-L1- TNBC (HR = 0.83; p = 0.545) or PD-L1+ TNBC (HR = 0.85; p = 0.22; Figure 3). Analysis of the relevant binary classification data yielded similar results. PD-1/PD-L1 inhibitor-based immunotherapy did not prolong the OS of the ITT population (RR = 1.00; 95% CI = 0.88–1.14; p = 0.98; I2 = 75.0%) or the PD-L1+ TNBC population (RR = 0.96; 95% CI = 0.82–1.12; p = 0.59; I2 = 62.0%; Table 2). Hence, PD-1/PD-L1 inhibitor-based immunotherapy has no effect on the OS of unresectable locally advanced or metastatic TNBC and has no relationship to PD-L1 status.

    Figure 2. Forest plot of overall survival of patients with unresectable locally advanced or metastatic triple-negative breast cancer on PD-1/PD-L1 inhibitor-based immunotherapy.

    p = 0.144.

    Figure 3. Forest plot of overall survival of patients with unresectable locally advanced or metastatic triple-negative breast cancer on PD-1/PD-L1 inhibitor-based immunotherapy.

    (A) PD-L1- (p = 0.545) and (B) PD-L1+ (p = 0.220).

    Table 2. Analysis of overall survival and progression-free survival of unresectable locally advanced or metastatic triple-negative breast cancer with PD-1/PD-L1 inhibitor-based immunotherapy combined with chemotherapy versus other treatments.
    GroupNo. of studiesNo. of experimental groupsNo. of control groupsRR95% CIp-valueHeterogeneity (I2) (%)
    OS (ITT)4122610281.000.88–1.140.9875
    OS (PD-L1+)35794870.960.82–1.120.5962
    PFS (ITT)5179213091.160.92–1.450.2196
    PFS (PD-L1+)38135970.830.80–0.970.0288

    ITT: Intent-to-treat; OS: Overall survival; PFS: Progression-free survival; RR: Risk ratio.

    PFS of ITT & PD-L1+/- populations

    For six studies (1839 experimental patients and 1344 control patients), PFS was determined to evaluate the efficacy of immunotherapy. Among them, three studies (424 experimental patients and 419 control patients) had PD-L1- population data and five studies (1022 experimental patients and 712 control patients) had PD-L1+ population data. The results demonstrated that the PFS of the ITT population showed statistically significant differences compared with others after various kinds of chemotherapy (HR = 0.82; 95% CI = 0.76–1.14; p < 0.001; I2 = 0%; Figure 4). That is, PD-1/PD-L1 inhibitor-based immunotherapy combined with chemotherapy can prolong the PFS of patients with unresectable locally advanced or metastatic TNBC and benefit the ITT population. In addition, subgroup analysis further illustrated that immunotherapy has no effect on the PD-L1- TNBC population (HR = 1.17; 95% CI = 0.82–1.66; p = 0.389; I2 = 71.8%; Figure 5A), but can prolong the PFS of the PD-L1+ population specifically, with a significant difference and no heterogeneity (HR = 0.68; 95% CI = 0.61–0.76, p < 0.001; Figure 5B). Analysis of relevant binary classification data suggested that PD-1/PD-L1 inhibitor-based immunotherapy has no effect on the PFS of the ITT population (RR = 1.16; p = 0.21; I2 = 96.0%; Table 2), but can prolong the PFS of the PD-L1+ TNBC population compared with any other discussed chemotherapies with a statistically significant difference (RR = 0.83; p = 0.02; I2 = 88.0%; Table 2) That is, PD-1/PD-L1 inhibitor-based immunotherapy has a beneficial effect on the PFS of unresectable locally advanced or metastatic TNBC and has a greater effect on the PD-L1+ population among all PD-L1 situations in the targeted population.

    Figure 4. Forest plot of progression-free survival of patients with unresectable locally advanced or metastatic triple-negative breast cancer on PD-1/PD-L1 inhibitor-based immunotherapy.

    p < 0.001.

    Figure 5. Forest plot of progression-free survival of patients with unresectable locally advanced or metastatic triple-negative breast cancer on PD-1/PD-L1 inhibitor-based immunotherapy.

    (A) PD-L1- (p = 0.389) and (B) PD-L1+ (p < 0.001).

    Safety analysis

    The incidence of adverse events (AEs) was used to evaluate the safety of PD-1/PD-L1 inhibitor-based immunotherapy combined with chemotherapy. The results showed that there was no difference between immunotherapy combined with chemotherapy and other chemotherapies in any-grade all-cause AEs (RR = 1.01; p = 0.07), any grade (RR = 1.01; p = 0.07) or higher than grade 3 (RR = 0.92; p = 0.60) treatment-related AEs and AEs that potentially lead to treatment discontinuation (RR = 1.49; p = 0.21). However, statistically significant differences were calculated in higher than grade 3 all-cause AEs (RR = 1.08; p = 0.02), any grade (RR = 2.31; p < 0.001) and higher than grade 3 (RR = 2.48; p < 0.001) immune-mediated AEs. This indicates that, compared with other chemotherapies, PD-1/PD-L1 inhibitor-based immunotherapy is more likely to cause higher than grade 3 all-cause AEs, any grade and higher than grade 3 immune-mediated AEs in involved patients (Supplementary Table 2). PD-1/PD-L1 inhibitor-based immunotherapy can lead to an increase in the risk of cough (RR = 1.39; 95% CI = 1.14–1.69; p = 0.001) and abnormal thyroid function (hyperthyroidism; RR = 15.17; 95% CI = 5.77–39.83; p < 0.001) and hypothyroidism (RR = 3.85; 95% CI = 2.78–5.32; p < 0.001)) and pneumonia (RR = 2.95; 95% CI = 1.67–5.22; p < 0.001), but no change in the frequency and incidence of other AEs (including alopecia, anemia, diarrhea, dyspnea, fatigue, nausea, neutropenia and pyrexia; p > 0.05; Supplementary Table 3). When it comes to grade 3 or higher AEs, PD-1/PD-L1 inhibitor-based immunotherapy mainly increased the risk of pneumonia (RR = 3.74; 95% CI = 1.45–9.65; p = 0.006), with no statistical difference in other AEs (i.e., alopecia, ALT increased, anemia, decreased appetite, diarrhea, dyspnea, fatigue, nausea, neutropenia and pyrexia; p > 0.05; Supplementary Table 4).

    Discussion

    Based on the data, PD-1/PD-L1 inhibitor-based immunotherapy combined with chemotherapy had significantly improved PFS in the ITT population with unresectable locally advanced or metastatic TNBC but had no contribution to OS compared with chemotherapy. Subgroup analysis, which was stratified according to PD-L1 receptor status, illustrated that PD-1/PD-L1 inhibitor-based immunotherapy combined with chemotherapy had significantly prolonged PFS in the PD-L1+ TNBC population and not in the PD-L1- TNBC population. As for OS, there was no statistically significant difference between PD-1/PD-L1 inhibitor-based immunotherapy combined with chemotherapy and other types of chemotherapies for either the PD-L1- population or the PD-L1+ population. Similar conclusions were also reached by the related dichotomous data analysis. Safety analysis suggests that, compared with chemotherapy, PD-1/PD-L1 inhibitor-based immunotherapy combined with chemotherapy will increase higher than grade 3 all-cause AEs, any grade and higher than grade 3 immune-mediated AEs. More specifically, AEs of any grade caused by immunotherapy combined with chemotherapy were mainly cough, abnormal thyroid function (including hyperthyroidism and hypothyroidism) and pneumonia, while AEs above grade 3 mainly included pneumonia.

    TNBC mainly occurs in young women under the age of 40 before menopause [7,8]. Compared with other BC subtypes (luminal A, luminal B or HER2+ BC), TNBC is more aggressive and more likely to metastasize and recur, resulting in poor prognosis [10,49]. Chemotherapy and targeted therapy have shown unsatisfactory curative effects on TNBC in terms of poor improvement in prognosis [21–23]. Therefore, in recent years, immunotherapy has provided novel treatment options for TNBC due to its remarkable activity and great therapeutic potential in a wide range of tumors [27]. It has been widely recognized that the immune system plays an important role in the development and progression of BC in terms of occurrence, progression and treatment [50,51]. In the early stages of BC, acute inflammation activates innate immunity and sequentially triggers tumor-specific T-cell responses [52]. At this stage, the body can show immune-mediated rejection to initial tumors, or produce tumor cell variants that can evade immune response [53,54]. A transition from acute inflammation to chronic inflammation will eventually be accomplished, creating a complex tumor microenvironment that greatly allows immune escape and tumor progression [55,56].

    Under normal physiological conditions, T cells can maintain the equilibrium of self-tolerance and tumor immunity by switching activation and inhibition signals [57–60]. Tumors can express immune inhibitory signals, such as CTLA-4 [61,62] and PD-1 [63], leading to a weakened immune response against pathological antigens [64,65]. The CTLA-4 signal pathway is involved in limiting the initiation of T-cell responses in lymph nodes. PD-1 limits the activity of T cells in the tumor microenvironment after initiation and is responsible for the immune escape of cancer [58,66].

    The higher genomic instability [67] and mutational burden [68] of TNBC lead to a greater possibility of generating new antigens [28]. Meanwhile, compared with other BC subtypes, TNBC has a higher number of TILs [29] and can produce more PD-L1 protein [69,70] or express more related mRNA [30,31]. It is believed that the expression of PD-L1 is significantly related to the presence of TILs [71]. There is a linear relationship between matrix TIL content and clinical results [71]. Solid tumors that carry TILs and produce PD-L1 are more likely to respond to PD-1/PD-L1 blockade, suggesting the possible mechanism of immunotherapy being active in the management of TNBC [72–74]. The regulatory mechanism of PD-L1 expression in TNBC patients may participate in the regulatory feedback of immunity [32,75,76].

    In TNBC cells, high PD-L1 expression can induce the generation of a tumor microenvironment that can inhibit antitumor immune response [69,77]. PD-1/PD-L1 inhibitors inhibit the interaction between PD-1 and PD-L1 on the membrane of tumor cells and immune cells, thereby antagonizing PD-1 signal transduction in inhibiting T-cell activation in the effective stage of the immune response [78–80]. At the same time, PD-1/PD-L1 inhibitors can induce the cytotoxic function of T cells by upregulating the gene expression of proinflammatory cytokines (IL-2, IFN-γ and TNF-α) [81–84], downregulating the gene expression of anti-inflammatory cytokines (IL-10 and TGF-β) [85,86], prevent T-cell lysis and inhibit antigen tolerance. By reactivating the specific immune response against tumors and effectively inhibiting the metastasis of tumor cells [72–74], immunotherapy based on PD-1/PD-L1 inhibitors can significantly prolong the PFS of the ITT population and, to be more specific, effective in the PD-L1+ population but not in PD-L1- population.

    Immunotherapy based on PD-1/PD-L1 inhibitors did not improve the OS of the ITT population and had no effect on either the PD-L1+ subtype or the PD-L1- subtype. The following three points may help explain this finding. First, the activity of immune response in the human body (mainly) depends on the duration of interaction between antigen-presenting cells and target cells [87,88]. When T cells are activated by the PD-1/PD-L1 inhibitor, the PD-1/PD-L1 axis is blocked, leading to disturbance of tumor cells evading from the immune response, thus indicating pharmacological effect [89,90]. However, the anticancer immune response usually fails when antigen-specific T cells and cytokines are strongly regulated, called “immune exhaustion” by researchers [91]. Therefore, PD-1/PD-L1 inhibitors can significantly improve the PFS of unresectable locally advanced or metastatic TNBC in the early follow-up time, but effectiveness, in the long run, is subsequently dependent on the immune regulatory response over time. As the occurrence of immune failure does not significantly improve OS, researchers can try to compare intermittent dosing regimens and continuous dosing regimens to explore the best dosing regimen of inhibitor-based immunotherapy. This may also be related to drug holidays, where a conscious decision is made to discontinue a certain medication for a period of time after long-term use. Second, the activity of immune responses in the human body also depends on the exercise capacity of T cells [88]. PD-L1 can reduce the motility of CD8+ T cells to prevent CD8+ T cells from performing their functions, to keep them in a state of negative immunoregulation for a long time [92–94]. The resulting decline in the body's immunity and the binding obstacles of antigen-presenting cells and target cells (e.g., the lymphocytes' challenge to completely move to the tumor cell area for infiltration) are factors that limit the effect of PD-1/PD-L1 inhibitors [95,96]. Therefore, the obstacle of T-cell motor function may also explain why immunotherapy failed to significantly improve OS in this study. Last, the number of trials included in this study was limited and potentially accidental factors may cause false negative results in the analysis. The improvement with immune therapy in OS in human trials will be further explored with the development of more clinical trials and the release of experimental data.

    Immunotherapy based on PD-1/PD-L1 inhibitors combined with other chemotherapies can increase the risk of cough, abnormal thyroid function (including hyperthyroidism and hypothyroidism) and pneumonia in TNBC patients. It is believed that the most likely mechanism of developing those AEs is that the immune system activated by PD-1/PD-L1 inhibitors not only targets tumor cells, but is also inductive to the death of normal cells, tissues and organs [94]. The thyroid gland is more vulnerable to autoimmune attacks than any other organ [97]. PD-1/PD-L1 blockade can induce autoantibodies' production and secretion in T-cell-dependent B cells [98,99]. The presence of thyroid autoantibodies and early elevation of serum thyroglobulin levels may increase the risk of thyroid dysfunction [100,101]. In addition, Krieg et al. [102] found that IFN-γ and IL-2 are increasingly expressed by CD4+ Th1 after the anti-PD-1 treatment. Higher levels of IL-2 can not only induce the binding between human major histocompatibility antigen II and thyroid cell self-antigens but also encourage the killing effect of CD8+ cytotoxic T lymphocytes on the cells in the thyroid gland [103,104]. This may explain why the anti-PD-1 treatment group has a higher incidence of developing thyroid dysfunction than other treatment groups. Similarly, pneumonia is also suspected to be caused by attack from the immune system, which is mainly manifested as interstitial pneumonia-like changes [105]. Studies have shown that patients with chronic pneumonia, patients who have received lung radiotherapy and patients with a history of chronic obstructive pulmonary disease are more prone to develop interstitial pneumonia after immunotherapy, and cough maybe is one of the symptoms of interstitial pneumonia [106]. Generally speaking, since most of the trials included in this meta-analysis adopted a grouping scheme of immunotherapy combined with chemotherapy versus chemotherapy alone, an increase in the incidence of certain adverse reactions, mainly focusing on thyroid and lung, in the experimental group is inevitable but safe and manageable.

    Reviewing similar studies, only Ali et al. [107] and Lazarus et al. [108] conducted a meta-analysis on the efficacy of immunotherapy in patients with advanced TNBC, but only 2 of the 12 trials included in the former were RCTs (KEYNOTE-355, KEYNOTE-119), only one of 7 trials included in the latter was an RCT (IMpassion130) and the rest were single-arm studies. The smaller sample size and the smaller number of RCTs limit the credibility and comparability of their research. Therefore, this meta-analysis is expected to be the first to fully and systematically explore the efficacy and safety of PD-1/PD-L1 inhibitor-based immunotherapy for unresectable locally advanced or metastatic TNBC, as the included studies were all high-quality RCT studies, and all trials were phase II and above, making this research more convincing and meriting this research over others in terms of convincingness and credibility. Also, data was grouped according to PD-L1 receptor status to explore the efficacy of PD-1/PD-L1 inhibitor-based immunotherapy on different detailed TNBC subtypes to increase the applicability of the research conclusions.

    The limitations of this study should also be discussed. First, the number of studies included in this meta-analysis was limited, and long-term survival outcomes for some trials were not published or have not been updated. Only two studies involved available OS data for the PD-L1- population, and four studies had OS data of the PD-L1+ population available, resulting in relatively poor effective data that is not constructive enough to identify the advantages of immunotherapy. Second, owing to obstacles to obtaining more information on the clinical trials, this meta-analysis only included one subgroup analysis of PD-L1 receptors. Subgroup analysis based on CD8+ or CD4+ T-cell levels is expected in the future to reflect the efficacy of immunotherapy. Lastly, this meta-analysis contained a variety of treatment options, including different dosing regimens, resulting in difficulty in clarifying the best treatment option for TNBC. Therefore, a large-scale RCT is still needed to further improve novel treatment options and verify the relevant conclusions. In short, this meta-analysis had drawn some meaningful conclusions, which may provide a new reference for immunotherapy, especially for patients with unresectable locally advanced or metastatic TNBC.

    Conclusion

    This meta-analysis found that PD-1/PD-L1 inhibitor-based immunotherapy combined with chemotherapy can significantly increase the PFS of unresectable locally advanced or metastatic TNBC population, but had no effect on OS, compared with chemotherapy alone. Immunotherapy had a significantly prolonged effect on the PFS in the PD-L1+ population but had no effect on OS. No effect on the PD-L1- population (PFS and OS) was found. AEs of any grade caused by immunotherapy mainly focused on cough, abnormal thyroid function (including hyperthyroidism and hypothyroidism) and pneumonia, and AEs above grade 3 were mainly pneumonia, which was all generally safe and manageable.

    Summary points
    • Triple-negative breast cancer (TNBC), accounting for 15–20% of breast cancers, is more aggressive and more likely to metastasize and recur.

    • Limited treatment strategies are available for patients with unresectable locally advanced or metastatic TNBC.

    • The development of PD-1/PD-L1 inhibitors has initiated a paradigm shift in therapeutic strategies for TNBC patients.

    • A meta-analysis was performed to explore the efficacy and safety of combined therapy with immunotherapy based on PD-1/PD-L1 inhibitors and chemotherapy for unresectable locally advanced or metastatic TNBC.

    • The selected articles were retrieved from PubMed, EBSCO, Web of Science and Cochrane Library databases before October 2021.

    • The primary end points were overall survival (OS) and progression-free survival (PFS) in TNBC patients. The secondary objective was safety.

    • A total of six articles comprising 3183 patients were eligible for this meta-analysis.

    • Compared with chemotherapy, PD-1/PD-L1 inhibitor-based immunotherapy combined with chemotherapy can significantly increase the PFS of unresectable locally advanced or metastatic TNBC (hazard ratio [HR] = 0.82; 95% CI = 0.76–1.14; p < 0.001) without effect on OS.

    • Immunotherapy had a significant prolongation effect on the PFS of the PD-L1+ population (HR = 0.68; 95% CI = 0.61–0.76; p < 0.001) but had no effect on OS.

    • No effect was discovered in the PD-L1- population (PFS and OS).

    • Adverse events of any grade caused by immunotherapy were mainly cough, abnormal thyroid function (including hyperthyroidism and hypothyroidism) and pneumonia and adverse events above grade 3 were mainly pneumonia.

    • PD-1/PD-L1 inhibitors-based immunotherapy can safely improve PFS in patients with unresectable locally advanced or metastatic TNBC but had no effect on OS.

    Supplementary data

    To view the supplementary data that accompany this paper please visit the journal website at: www.futuremedicine.com/doi/suppl/10.2217/imt-2023-0023

    Author contributions

    Each author contributed significantly to the conception and development of the present paper. W Zhang and Y He designed the research process. Y Tang and W Dai searched the databases for corresponding articles and extracted useful information from the articles. Y Si and F Mao used statistical software for analysis. J Xu, C Yu and X Sun drafted the meta-analysis. All authors read and approved the final manuscript.

    Financial & competing interests disclosure

    This study was funded by Ningbo Medical Key Discipline (grant no. 2022-B09), China. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties.

    No writing assistance was utilized in the production of this manuscript.

    Open access

    This work is licensed under the Attribution-NonCommercial-NoDerivatives 4.0 Unported License. To view a copy of this license, visit http://creativecommons.org/licenses/by-nc-nd/4.0/

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    Reference

    • 1. Ge L, Tang Y, Zhang QN et al. A network meta-analysis on the efficacy of targeted agents in combination with chemotherapy for treatment of advanced/metastatic triple-negative breast cancer. Oncotarget 8(35), 59539–59551 (2017).
    • 2. Harbeck N, Gnant M. Breast cancer. Lancet 389(10074), 1134–1150 (2017). • A comprehensive and systematic review of breast cancer with clear logic.
    • 3. Akram M, Iqbal M, Daniyal M et al. Awareness and current knowledge of breast cancer. Biol. Res. 50(1), 33 (2017).
    • 4. Garcia-Guasch M, Navarro L, Rivero V et al. A high extra-virgin olive oil diet induces changes in metabolic pathways of experimental mammary tumors. J. Nutr. Biochem. 99, 108833 (2022).
    • 5. Cherbal F, Gaceb H, Mehemmai C et al. Distribution of molecular breast cancer subtypes among Algerian women and correlation with clinical and tumor characteristics: a population-based study. Breast Dis. 35(2), 95–102 (2015).
    • 6. Effi AB, Aman NA, Koui BS et al. Breast cancer molecular subtypes defined by ER/PR and HER2 status: association with clinicopathologic parameters in Ivorian patients. Asian Pac. J. Cancer Prev. 17(4), 1973–1978 (2016).
    • 7. Kumar P, Aggarwal R. An overview of triple-negative breast cancer. Arch. Gynecol. Obstet. 293(2), 247–269 (2016).
    • 8. Waks AG, Winer EP. Breast cancer treatment: a review. JAMA 321(3), 288–300 (2019).
    • 9. Keenan TE, Tolaney SM. Role of immunotherapy in triple-negative breast cancer. J. Natl Compr. Cancer Netw. 18(4), 479–489 (2020).
    • 10. Lehmann BD, Pietenpol JA, Tan AR. Triple-negative breast cancer: molecular subtypes and new targets for therapy. Am. Soc. Clin. Oncol. Educ. Book. e31–e39 (2015).
    • 11. Sharma P. Biology and management of patients with triple-negative breast cancer. Oncologist 21(9), 1050–1062 (2016).
    • 12. Li X, Yang J, Peng L et al. Triple-negative breast cancer has worse overall survival and cause-specific survival than non-triple-negative breast cancer. Breast Cancer Res. Treat. 161(2), 279–287 (2017).
    • 13. Feng W, He Y, Xu J et al. A meta-analysis of the effect and safety of platinum-based neoadjuvant chemotherapy in treatment of resectable triple-negative breast cancer. Anticancer Drugs 33(1), e52–e60 (2022).
    • 14. Lehmann BD, Bauer JA, Chen X et al. Identification of human triple-negative breast cancer subtypes and preclinical models for selection of targeted therapies. J. Clin. Invest. 121(7), 2750–2767 (2011).
    • 15. Oualla K, Kassem L, Nouiakh L et al. Immunotherapeutic approaches in triple-negative breast cancer: state of the art and future perspectives. Int. J. Breast Cancer 2020, 8209173 (2020).
    • 16. Bayraktar S, Gluck S. Molecularly targeted therapies for metastatic triple-negative breast cancer. Breast Cancer Res. Treat. 138(1), 21–35 (2013).
    • 17. da Silva JL, Cardoso Nunes NC, Izetti P et al. Triple negative breast cancer: a thorough review of biomarkers. Crit. Rev. Oncol. Hematol. 145, 102855 (2020).
    • 18. Navratil J, Fabian P, Palacova M et al. Triple negative breast cancer. Klin. Onkol. 28(6), 405–415 (2015).
    • 19. Li ZY, Zhang Z, Cao XZ et al. Platinum-based neoadjuvant chemotherapy for triple-negative breast cancer: a systematic review and meta-analysis. J. Int. Med. Res. 48(10), 300060520964340 (2020).
    • 20. Poggio F, Bruzzone M, Ceppi M et al. Platinum-based neoadjuvant chemotherapy in triple-negative breast cancer: a systematic review and meta-analysis. Ann. Oncol. 29(7), 1497–1508 (2018).
    • 21. Bonotto M, Gerratana L, Poletto E et al. Measures of outcome in metastatic breast cancer: insights from a real-world scenario. Oncologist 19(6), 608–615 (2014). • Monitoring methods for metastatic breast cancer are clearly described.
    • 22. Carey LA, Dees EC, Sawyer L et al. The triple negative paradox: primary tumor chemosensitivity of breast cancer subtypes. Clin. Cancer Res. 13(8), 2329–2334 (2007).
    • 23. von Minckwitz G, Untch M, Blohmer JU et al. Definition and impact of pathologic complete response on prognosis after neoadjuvant chemotherapy in various intrinsic breast cancer subtypes. J. Clin. Oncol. 30(15), 1796–1804 (2012).
    • 24. Bergin ART, Loi S. Triple-negative breast cancer: recent treatment advances. F1000Res 8, 1–11(2019).
    • 25. Gerson R, Alban F, Villalobos A et al. Recurrence and survival rates among early breast cancer cases with triple negative immunophenotype. Gac. Med. Mex. 144(1), 27–34 (2008).
    • 26. Gu-Trantien C, Loi S, Garaud S et al. Cd4(+) follicular helper T cell infiltration predicts breast cancer survival. J. Clin. Invest. 123(7), 2873–2892 (2013).
    • 27. Schmidt M, Bohm D, von Torne C et al. The humoral immune system has a key prognostic impact in node-negative breast cancer. Cancer Res. 68(13), 5405–5413 (2008).
    • 28. Brown SD, Warren RL, Gibb EA et al. Neo-antigens predicted by tumor genome meta-analysis correlate with increased patient survival. Genome Res. 24(5), 743–750 (2014).
    • 29. Loi S, Sirtaine N, Piette F 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. 31(7), 860–867 (2013).
    • 30. Mittendorf EA, Philips AV, Meric-Bernstam F et al. PD-L1 expression in triple-negative breast cancer. Cancer Immunol. Res. 2(4), 361–370 (2014).
    • 31. Sabatier R, Finetti P, Mamessier E et al. Prognostic and predictive value of PDL1 expression in breast cancer. Oncotarget 6(7), 5449–5464 (2015). •• An interesting discussion of the expression of PD-L1 in breast cancer.
    • 32. Spranger S, Spaapen RM, Zha Y et al. Up-regulation of PD-L1, IDO, and T(regs) in the melanoma tumor microenvironment is driven by cd8(+) T cells. Sci. Transl. Med. 5(200), 200ra116 (2013).
    • 33. Schmid P, Adams S, Rugo HS et al. Atezolizumab and nab-paclitaxel in advanced triple-negative breast cancer. N. Engl. J. Med. 379(22), 2108–2121 (2018).
    • 34. Schmid P, Cortes J, Pusztai L et al. Pembrolizumab for early triple-negative breast cancer. N. Engl. J. Med. 382(9), 810–821 (2020).
    • 35. Smith KM, Desai J. Nivolumab for the treatment of colorectal cancer. Expert Rev. Anticancer Ther. 18(7), 611–618 (2018).
    • 36. Paz-Ares L, Dvorkin M, Chen Y et al. Durvalumab plus platinum-etoposide versus platinum-etoposide in first-line treatment of extensive-stage small-cell lung cancer (CASPIAN): a randomised, controlled, open-label, phase 3 trial. Lancet 394(10212), 1929–1939 (2019).
    • 37. Dirix LY, Takacs I, Jerusalem G et al. Avelumab, an anti-PD-L1 antibody, in patients with locally advanced or metastatic breast cancer: a phase 1b JAVELIN solid tumor study. Breast Cancer Res. Treat. 167(3), 671–686 (2018).
    • 38. Winer EP, Lipatov O, Im SA et al. Pembrolizumab versus investigator-choice chemotherapy for metastatic triple-negative breast cancer (KEYNOTE-119): a randomised, open-label, phase 3 trial. Lancet Oncol. 22(4), 499–511 (2021).
    • 39. Bachelot T, Filleron T, Bieche I et al. Durvalumab compared to maintenance chemotherapy in metastatic breast cancer: the randomized phase II SAFIR02-BREAST IMMUNO trial. Nat. Med. 27(2), 250–255 (2021).
    • 40. Moher D, Liberati A, Tetzlaff J et al. Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statement. BMJ 339, b2535 (2009).
    • 41. Higgins JP, Thompson SG, Deeks JJ et al. Measuring inconsistency in meta-analyses. BMJ 327(7414), 557–560 (2003).
    • 42. Gu L, Huang X, Li S et al. A meta-analysis of the medium- and long-term effects of laparoscopic sleeve gastrectomy and laparoscopic roux-en-y gastric bypass. BMC Surg. 20(1), 30 (2020).
    • 43. Li S, Gu L, Shen Z et al. A meta-analysis of comparison of proximal gastrectomy with double-tract reconstruction and total gastrectomy for proximal early gastric cancer. BMC Surg. 19(1), 117 (2019).
    • 44. Brufsky A, Kim SB, Zvirbule Z et al. A phase II randomized trial of cobimetinib plus chemotherapy, with or without atezolizumab, as first-line treatment for patients with locally advanced or metastatic triple-negative breast cancer (COLET): primary analysis. Ann. Oncol. 32(5), 652–660 (2021).
    • 45. Cortes J, Cescon DW, Rugo HS et al. Pembrolizumab plus chemotherapy versus placebo plus chemotherapy for previously untreated locally recurrent inoperable or metastatic triple-negative breast cancer (KEYNOTE-355): a randomised, placebo-controlled, double-blind, phase 3 clinical trial. Lancet 396(10265), 1817–1828 (2020).
    • 46. Miles D, Gligorov J, Andre F et al. Primary results from IMPASSION131, a double-blind, placebo-controlled, randomised phase iii trial of first-line paclitaxel with or without atezolizumab for unresectable locally advanced/metastatic triple-negative breast cancer. Ann. Oncol. 32(8), 994–1004 (2021).
    • 47. Schmid P, Rugo HS, Adams S et al. 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. 21(1), 44–59 (2020).
    • 48. Winer EP, Lipatov O, Im S-A et al. Pembrolizumab versus investigator-choice chemotherapy for metastatic triple-negative breast cancer (KEYNOTE-119): a randomised, open-label, phase 3 trial. Lancet Oncol. 22(4), 499–511 (2021).
    • 49. Yin L, Duan JJ, Bian XW et al. Triple-negative breast cancer molecular subtyping and treatment progress. Breast Cancer Res. 22(1), 61 (2020).
    • 50. Emens LA. Breast cancer immunobiology driving immunotherapy: vaccines and immune checkpoint blockade. Expert Rev. Anticancer Ther. 12(12), 1597–1611 (2012).
    • 51. Smyth MJ, Dunn GP, Schreiber RD. Cancer immunosurveillance and immunoediting: the roles of immunity in suppressing tumor development and shaping tumor immunogenicity. Adv. Immunol. 90, 1–50 (2006).
    • 52. DeNardo DG, Barreto JB, Andreu P et al. Cd4(+) T cells regulate pulmonary metastasis of mammary carcinomas by enhancing protumor properties of macrophages. Cancer Cell 16(2), 91–102 (2009).
    • 53. Ribas A. Adaptive immune resistance: how cancer protects from immune attack. Cancer Discov. 5(9), 915–919 (2015).
    • 54. Schutz F, Stefanovic S, Mayer L et al. PD-1/PD-L1 pathway in breast cancer. Oncol. Res. Treat. 40(5), 294–297 (2017).
    • 55. Coussens LM, Pollard JW. Leukocytes in mammary development and cancer. Cold Spring Har.b Perspect. Biol. 3(3), 1–24 (2011).
    • 56. Ho PC, Kaech SM. Reenergizing T cell anti-tumor immunity by harnessing immunometabolic checkpoints and machineries. Curr. Opin. Immunol. 46, 38–44 (2017).
    • 57. Ceeraz S, Nowak EC, Noelle RJ. B7 family checkpoint regulators in immune regulation and disease. Trends Immunol. 34(11), 556–563 (2013).
    • 58. Han Y, Liu D, Li L. PD-1/PD-L1 pathway: current researches in cancer. Am. J. Cancer Res. 10(3), 727–742 (2020).
    • 59. Jung K, Choi I. Emerging co-signaling networks in T cell immune regulation. Immune Netw. 13(5), 184–193 (2013).
    • 60. Topalian SL, Drake CG, Pardoll DM. Immune checkpoint blockade: a common denominator approach to cancer therapy. Cancer Cell 27(4), 450–461 (2015).
    • 61. Rotte A. Combination of CTLA-4 and PD-1 blockers for treatment of cancer. J. Exp. Clin. Cancer Res. 38(1), 255 (2019).
    • 62. Van Coillie S, Wiernicki B, Xu J. Molecular and cellular functions of CTLA-4. Adv. Exp. Med. Biol. 1248, 7–32 (2020).
    • 63. Xu T, Wang Z, Dong M et al. Chloride intracellular channel protein 2: prognostic marker and correlation with PD-1/PD-L1 in breast cancer. Aging (Albany) 12(17), 17305–17327 (2020).
    • 64. Dong P, Xiong Y, Yue J et al. Tumor-intrinsic PD-L1 signaling in cancer initiation, development and treatment: beyond immune evasion. Front. Oncol. 8, 386 (2018).
    • 65. Pentcheva-Hoang T, Corse E, Allison JP. Negative regulators of T-cell activation: potential targets for therapeutic intervention in cancer, autoimmune disease, and persistent infections. Immunol. Rev. 229(1), 67–87 (2009).
    • 66. Fife BT, Bluestone JA. Control of peripheral T-cell tolerance and autoimmunity via the CTLA-4 and PD-1 pathways. Immunol. Rev. 224, 166–182 (2008).
    • 67. Liu L, Shen Y, Zhu X et al. Eralpha is a negative regulator of PD-L1 gene transcription in breast cancer. Biochem. Biophys. Res. Commun. 505(1), 157–161 (2018).
    • 68. Liu S, Chen S, Yuan W et al. PD-1/PD-L1 interaction up-regulates mdr1/p-gp expression in breast cancer cells via pi3k/akt and mapk/erk pathways. Oncotarget 8(59), 99901–99912 (2017).
    • 69. Ali HR, Glont SE, Blows FM et al. PD-L1 protein expression in breast cancer is rare, enriched in basal-like tumours and associated with infiltrating lymphocytes. Ann. Oncol. 26(7), 1488–1493 (2015).
    • 70. Wimberly H, Brown JR, Schalper K et al. PD-L1 expression correlates with tumor-infiltrating lymphocytes and response to neoadjuvant chemotherapy in breast cancer. Cancer Immunol. Res. 3(4), 326–332 (2015).
    • 71. Emens LA. Breast cancer immunotherapy: facts and hopes. Clin. Cancer Res. 24(3), 511–520 (2018).
    • 72. Ajona D, Ortiz-Espinosa S, Moreno H et al. A combined pd-1/c5a blockade synergistically protects against lung cancer growth and metastasis. Cancer Discov. 7(7), 694–703 (2017).
    • 73. Nunes-Xavier CE, Angulo JC, Pulido R et al. A critical insight into the clinical translation of PD-1/PD-L1 blockade therapy in clear cell renal cell carcinoma. Curr. Urol. Rep. 20(1), 1 (2019).
    • 74. You Y, Ren Y, Liu J et al. Promising epigenetic biomarkers associated with cancer-associated-fibroblasts for progression of kidney renal clear cell carcinoma. Front. Genet. 12, 736156 (2021).
    • 75. Glorieux C, Xia X, Huang P. The role of oncogenes and redox signaling in the regulation of PD-L1 in cancer. Cancers (Basel) 13(17), 1–25(2021).
    • 76. Hosseini A, Gharibi T, Marofi F et al. CTLA-4: from mechanism to autoimmune therapy. Int. Immunopharmacol. 80, 106221 (2020).
    • 77. Yao S, Chen L. Reviving exhausted t lymphocytes during chronic virus infection by b7-h1 blockade. Trends Mol. Med. 12(6), 244–246 (2006).
    • 78. Kim JM, Chen DS. Immune escape to PD-L1/PD-1 blockade: seven steps to success (or failure). Ann. Oncol. 27(8), 1492–1504 (2016).
    • 79. Lenouvel D, Gonzalez-Moles MA, Talbaoui A et al. An update of knowledge on PD-L1 in head and neck cancers: physiologic, prognostic and therapeutic perspectives. Oral Dis. 26(3), 511–526 (2020).
    • 80. Salmaninejad A, Valilou SF, Shabgah AG et al. PD-1/PD-L1 pathway: basic biology and role in cancer immunotherapy. J. Cell. Physiol. 234(10), 16824–16837 (2019).
    • 81. Briesemeister D, Sommermeyer D, Loddenkemper C et al. Tumor rejection by local interferon gamma induction in established tumors is associated with blood vessel destruction and necrosis. Int. J. Cancer 128(2), 371–378 (2011).
    • 82. Michel C, Montravers P, Viron B et al. Renovascular hypertension with renal failure: value of renal revascularization. Arch. Mal. Coeur Vaiss. 79(6), 851–856 (1986).
    • 83. Nie H, Zheng Y, Li R et al. Phosphorylation of foxp3 controls regulatory T cell function and is inhibited by TNF-alpha in rheumatoid arthritis. Nat. Med. 19(3), 322–328 (2013).
    • 84. Patsoukis N, Sari D, Boussiotis VA. PD-1 inhibits T cell proliferation by upregulating p27 and p15 and suppressing cdc25a. Cell Cycle 11(23), 4305–4309 (2012).
    • 85. Abdoli Shadbad M, Hajiasgharzadeh K, Baradaran B. Cross-talk between myeloid-derived suppressor cells and mucin1 in breast cancer vaccination: on the verge of a breakthrough. Life Sci. 258, 118128 (2020).
    • 86. Kwasniak K, Czarnik-Kwasniak J, Maziarz A et al. Scientific reports concerning the impact of interleukin 4, interleukin 10 and transforming growth factor beta on cancer cells. Cent. Eur. J. Immunol. 44(2), 190–200 (2019).
    • 87. Emens LA, Kok M, Ojalvo LS. Targeting the programmed cell death-1 pathway in breast and ovarian cancer. Curr. Opin. Obstet. Gynecol. 28(2), 142–147 (2016).
    • 88. Honda T, Egen JG, Lammermann T et al. Tuning of antigen sensitivity by T cell receptor-dependent negative feedback controls T cell effector function in inflamed tissues. Immunity 40(2), 235–247 (2014).
    • 89. Emens LA, Ascierto PA, Darcy PK et al. Cancer immunotherapy: opportunities and challenges in the rapidly evolving clinical landscape. Eur. J. Cancer 81, 116–129 (2017).
    • 90. Vibhakar R, Juan G, Traganos F et al. Activation-induced expression of human programmed death-1 gene in T-lymphocytes. Exp. Cell Res. 232(1), 25–28 (1997).
    • 91. Wherry EJ. T cell exhaustion. Nat. Immunol. 12(6), 492–499 (2011).
    • 92. Barber DL, Wherry EJ, Masopust D et al. Restoring function in exhausted cd8 T cells during chronic viral infection. Nature 439(7077), 682–687 (2006).
    • 93. Curran MA, Montalvo W, Yagita H et al. 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. USA 107(9), 4275–4280 (2010).
    • 94. Delivanis DA, Gustafson MP, Bornschlegl S et al. Pembrolizumab-induced thyroiditis: comprehensive clinical review and insights into underlying involved mechanisms. J. Clin. Endocrinol. Metab. 102(8), 2770–2780 (2017).
    • 95. Iwai Y, Ishida M, Tanaka Y et al. Involvement of PD-L1 on tumor cells in the escape from host immune system and tumor immunotherapy by PD-L1 blockade. Proc Natl Acad Sci USA 99(19), 12293–12297 (2002).
    • 96. Velu V, Titanji K, Zhu B et al. Enhancing SIV-specific immunity in vivo by PD-1 blockade. Nature 458(7235), 206–210 (2009).
    • 97. Luty J, Ruckemann-Dziurdzinska K, Witkowski JM et al. Immunological aspects of autoimmune thyroid disease–complex interplay between cells and cytokines. Cytokine 116, 128–133 (2019).
    • 98. Chen DS, Mellman I. Elements of cancer immunity and the cancer-immune set point. Nature 541(7637), 321–330 (2017).
    • 99. Elia G, Ferrari SM, Galdiero MR et al. New insight in endocrine-related adverse events associated to immune checkpoint blockade. Best Pract. Res. Clin. Endocrinol. Metab. 34(1), 101370 (2020).
    • 100. Ohara N, Kobayashi M, Ohashi K et al. Isolated adrenocorticotropic hormone deficiency and thyroiditis associated with nivolumab therapy in a patient with advanced lung adenocarcinoma: a case report and review of the literature. J. Med. Case Rep. 13(1), 88 (2019).
    • 101. Zhan L, Feng HF, Liu HQ et al. Immune checkpoint inhibitors-related thyroid dysfunction: epidemiology, clinical presentation, possible pathogenesis, and management. Front. Endocrinol. (Lausanne) 12, 649863 (2021).
    • 102. Krieg C, Nowicka M, Guglietta S et al. High-dimensional single-cell analysis predicts response to anti-PD-1 immunotherapy. Nat. Med. 24(2), 144–153 (2018).
    • 103. Kethireddy N, Thomas S, Bindal P et al. Multiple autoimmune side effects of immune checkpoint inhibitors in a patient with metastatic melanoma receiving pembrolizumab. J. Oncol. Pharm. Pract. 27(1), 207–211 (2021).
    • 104. Kurimoto C, Inaba H, Ariyasu H et al. Predictive and sensitive biomarkers for thyroid dysfunctions during treatment with immune-checkpoint inhibitors. Cancer Sci. 111(5), 1468–1477 (2020).
    • 105. Disayabutr S, Calfee CS, Collard HR et al. Interstitial lung diseases in the hospitalized patient. BMC Med. 13, 245 (2015).
    • 106. Naidoo J, Wang X, Woo KM et al. Pneumonitis in patients treated with anti-programmed death-1/programmed death ligand 1 therapy. J. Clin. Oncol. 35(7), 709–717 (2017).
    • 107. Ali MA, Aiman W, Shah SS et al. Efficacy and safety of pembrolizumab based therapies in triple-negative breast cancer: a systematic review of clinical trials. Crit. Rev. Oncol. Hematol. 157, 103197 (2021).
    • 108. Lazarus G, Audrey J, Iskandar AWB. Efficacy and safety profiles of programmed cell death-1/programmed cell death ligand-1 inhibitors in the treatment of triple-negative breast cancer: a comprehensive systematic review. Oncol. Rev. 13(2), 425 (2019).