Skip to main content
Oncology Reviews logoLink to Oncology Reviews
. 2019 Oct 10;13(2):425. doi: 10.4081/oncol.2019.425

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

Gilbert Lazarus 1,, Jessica Audrey 1, Anthony William Brian Iskandar 1
PMCID: PMC6886008  PMID: 31857857

Abstract

Triple-negative breast cancer (TNBC) is associated with worse prognosis, with limited treatment regiments available and higher mortality rate. Immune checkpoint inhibitors targeting programmed cell death-1 (PD-1) or programmed cell death-ligand 1 (PD-L1) showed great potentials in treating malignancies and may serve as potential therapies for TNBC. This systematic review aims to evaluate the efficacy and safety profiles of PD-1/PD-L1 inhibitors in the treatment of TNBC. Literature search was performed via PubMed, EBSCOhost, Scopus, and CENTRAL databases, selecting studies which evaluated the use of anti-PD-1/PDL1 for TNBC from inception until February 2019. Risk of bias was assessed by the Newcastle-Ottawa Scale (NOS). Overall, 7 studies evaluating outcomes of 1395 patients with TNBC were included in this systematic review. Anti-PD-1/PD-L1 showed significant antitumor effect, proven by their promising response (objective response rate (ORR), 18.5-39.4%) and survival rates (median overall survival (OS), 9.2-21.3 months). Moreover, anti- PD-1/PD-L1 yielded better outcomes when given as first-line therapy, and overexpression of PD-L1 in tumors showed better therapeutic effects. On the other hands, safety profiles were similar across agents and generally acceptable, with grade ≥3 treatment- related adverse effects (AEs) ranging from 9.5% to 15.6% and no new AEs were experienced by TNBC patients. Most grade ≥3 AEs are immune-mediated, which are manifested as neutropenia, fatigue, peripheral neuropathy, and anemia. PD-1/PD-L1 inhibitors showed promising efficacy and tolerable AEs, and thus may benefit TNBC patients. Further studies of randomized controlled trials with larger populations are needed to better confirm the potential of these agents.

Key words: Checkpoint inhibitor, programmed cell death-1, programmed cell death-ligand 1, triple-negative breast cancer

Introduction

Breast cancer (BC) remains a global health concern, especially for females.1,2 The Global Cancer Observatory1 stated that breast cancer constitutes 6.6% of all deaths from cancer in 2018 and is the leading cause of cancer deaths in females.2 Furthermore, the 5-year survival rate diminished greatly across each stage from stage 0 and I (100%), stage II and III (93% and 72%, respectively), to stage IV (22%).3

Among all breast cancer subtypes, triple-negative BC (TNBC) is associated with the highest mortality rate.4-7 TNBC is mostly, but not always, included in basal-like BC subtypes4 because it expresses epithelial cytokeratin 5/6/14/17, laminin, and/or epidermal growth factor receptor.8 It is defined by the lack of expression of prognostic factors of BC such as estrogen receptor (ER), progesterone receptor (PgR), and human epidermal growth factor receptor-2 (HER2),9-11 which makes it often associated with worse prognosis since there are no commonly utilized targeting agents for this case.10 There are several pathways known in the pathogenesis of TNBC, some of which include under-expression of receptors by BRCA1-related pathway and de novo mutations.11 Although it only accounts for 10% to 20% of all BC cases, it is responsible for around 30% BC associated deaths.5 Moreover, some TNBC has deregulated integrin expression which may contribute to its highly metastatic behavior.8

Due to the inexpressiveness of hormone receptors, patients with TNBC cannot benefit from trastuzumab and/or hormonalbased therapy.12 That being said, surgery11,12 and chemotherapy11,12 are the only treatment available for TNBC, unlike other types of BC that can fully benefit from common adjuvant/neoadjuvant therapies.13 There are currently several trials conducted to develop chemotherapies for TNBC (e.g. anthracycline and taxane14, cytotoxic chemotherapy15). However, optimal chemotherapy regiments have yet to be established.16 Furthermore, usage of chemotherapies have showed significant adverse events, ranging from pain,17 nausea,17 neurocognitive dysfunction,18 cardiovascular diseases,18 and often deterioration of quality life,17 to ultimately death.18

Recently, researchers have extensively conducted trials about the utilization of immunotherapy for the management of TNBC. The identification of tumor-infiltrating lymphocytes (TILs) as a prognostic biomarker for TNBC has led to the study of immunecheckpoint inhibitors (ICIs). Current researches have focused on the role of programmed cell death-1 (PD-1) and its ligand (PD-L1), and also cytotoxic T-lymphocyte antigen-4 (CTLA-4), in maintaining immunosuppression in the tumor microenvironment.19 PD-1 is an inhibitory immune checkpoint that limits immune effector cells’ function within tissues, which is expressed on B cells, T cells, dendritic cells, NK cells, and TILs.20 It interacts with PD-L1 and PDL2 and mediate immune tolerance by attenuating T-cell function, expansion, and survival.19 On the other hand, CTLA-4 diminishes signaling via CD28 receptor indirectly and thus also mediates immunosuppression. Inhibitors of those immune checkpoints yield promising activity by increasing patients’ survival.21 Several PD- 1/PD-L1 and CTLA-4 inhibitors have been tested for TNBC, including atezolizumab, avelumab, durvalumab, ipilimumab, and tremelimumab.22 This systematic review aims to integrate and assess the efficacy and safety profiles of PD-1/PD-L1 inhibitor in treating TNBC.

Materials and methods

Search strategy

This systematic review was conducted according to the Cochrane Handbook for Systematic Reviews of Interventions23 and Preferred Reporting Items for Systematic Reviews and Meta- Analysis (PRISMA) statement.24 We retrieved relevant studies from PubMed, EBSCOhost, Scopus, and Cochrane Controlled Register of Trials (CENTRAL) databases from inception to 20 February 2019, with search terms as following: (“PD-1 antibody”[ All Fields] OR “PD-L1 antibody”[All Fields] OR “avelumab”[ All Fields] OR “atezolizumab”[All Fields] OR “pembrolizumab”[ All Fields] OR “nivolumab”[All Fields] OR “checkpoint inhibitor”[All Fields] OR “immune checkpoint”[All Fields] OR “checkpoint blockade”[All Fields] OR “checkpoint blocker”[All Fields] OR “durvalumab”[All Fields] OR “immune blockade inhibitor”[All Fields]) AND (“triple-negative breast cancer”[All Fields] OR “triple negative breast cancer”[All Fields] OR “TNBC”[All Fields]). The search was limited to human participants and no language restrictions were applied. However, studies included in the review was restricted to English and Bahasa Indonesia, which were the only languages readable by the reviewers. Details of the literature search strategy are shown on Figure 1.

Figure 1.

Figure 1.

Diagram flow of literature search strategy for this systematic review.

Inclusion and exclusion criteria

The following inclusion criteria were applied: i) study design, clinical trials (either randomized or non-randomized); ii) study population, patients over 18 years old with confirmed TNBC; iii) language, English and Bahasa Indonesia; iv) comparison and intervention, PD-1 and/or PD-L1 inhibitor either as neoadjuvant or adjuvant therapy; and v) outcome parameter, overall response rate (ORR), disease control rate (DCR), duration of response (DOR), progression-free survival (PFS), overall survival (OS), and adverse effects (AEs) grade ≥3. Furthermore, exclusion criteria comprising of i) no extractable data (i.e. ORR, DCR, DOR, PFS, OS, or AEs grade ≥3) and ii) irretrievable full-text articles were also set.

Data extraction and quality assessment

Literature screening and data extraction were performed by two independent reviewers (GL and JA), and discrepancies were resolved by the consensus of all authors. The following information was extracted from each trial: first author; publication year; study design; trial phase; subject characteristics, consisting of median age and sample size; intervention, consisting of treatment groups, target molecule, and line of therapy; duration of follow-up; ORR, defined as proportion of patients with complete (CR) or partial response (PR); DCR, defined as the percentage of patients with CR, PR, or stable disease for 24 weeks, PFS, defined as time from patient enrollment to disease progression or death; OS, defined as time from patient enrollment to death from any cause; and significant AEs (grade 3).

Two reviewers (GL and JA) independently assessed the quality of included trials, disagreements between authors were adjudicated by a third investigator (AW). Risk of bias and methodological quality assessment was performed using the Newcastle-Ottawa Scale (NOS).25 Appendix 1 provides details of quality assessment of included studies.

Results

Study selection

The selection process for included studies in the systematic review is shown in Figure 1. The initial search yielded 949 relevant studies from PubMed, EBSCOhost, CENTRAL, and Scopus. Among them, 648 studies were deduplicated, and 253 studies were excluded after title and/or abstract screening. Furthermore, 42 studies were excluded because 2126-46 did not provide outcomes of our interest, 2047-66 were irretrievable, and 167 was not in English or Bahasa Indonesia. Finally, 7 clinical trials were identified for the qualitative analysis, consisting of 6 non-randomized trials and 1 randomized trial.

Study characteristics

The main patient characteristics of included studies are shown in Table 1. A pooled total of 1395 patients were included in our systematic review. Patient characteristics were matched for locally advanced and metastatic (stage III/IV) TNBC (ER-negative, PgRnegative, and HER2-negative). The trials were published between 2016 and 2018, and mostly were international, multicentered, and/or multi-cohort studies. All trials aimed to investigate the efficacy and safety of immune checkpoint inhibitors. In addition, two trials68,69 also evaluated the clinical activity of checkpoint inhibitors.

Table 1.

Study characteristics.

Year; Author Study characteristics Subject characteristics Intervention NOS Score
Study design Phase Aims/ Purpose Median age (years) Sample size Disease stage Target molecule Intervention Treatment groups Line of therapy Follow-up (months)
2018; Adams S68 Single-arm trial, multi-cohort, multicenter Ib To examine the safety, tolerability, and preliminary clinical activity of atezolizumab + nab-paclitaxel in mTNBC 55 33 IV PD-L1 Atezolizumab + nab-paclitaxel ≥1L 24.4 (22.1-28.8) 8
2018; Adams S Cohort A70 Single-arm trial, open-label II To examine the efficacy and safety of pembrolizumab monotherapy in large cohort of patients with previously treated mTNBC regardless of PD-L1 expression 53.5 170 IV PD-1 Pembrolizumab ≥1L 9.6 (0.1-25.7) 7
2018; Adams S Cohort B71 Single-arm trial, open-label II To examine the safety and efficacy of single-agent pembrolizumab in patients with previously untreated PD-L1+ mTNBC 52.5 (26-91) 84 IV PD-1 Pembrolizumab 1L 12.3 (0.9-23.5) 7
2018; Schmid P72 Randomized, multi-cohort III To investigate the efficacy and safety of first line atezolizumab + nab-paclitxel, as compared with placebo + nabnab- paclitaxel, in patients with locally advanced or mTNBC Atezolizumab– nab-paclitaxel: 55 Placebo–nab- paclitaxel: 56 902 III/IV PD-L1 Atezolizumab + nab-paclitaxel vs placebo vs nab-paclitaxel 1L Atezolizumab–nab- paclitaxel: 13.0 Placebo–nab- paclitaxel: 12.5 9
2018; Emens LA69 Single-arm trial, multi-cohort, open-label I To evaluate the safety, clinical activity, and biomarkers associated with the use of single agent atezolizumab in patients with mTNBC 53 116 IV PD-L1 Atezolizumab ≥1L 25.3 (0.4-45.6) 6
2017; Dirix LY73 Single-arm trial, open-label, international I To assess the activity of avelumab, a PD-L1 inhibitor, in patients with mBC 52.5 (31-80) 58 III/IV PD-L1 Avelumab ≥1L 10.0 (6.0-15.2) 7
2016; Nanda R74 Non-randomized, multicenter Ib To assess the safety and antitumor activity of PD-1 inhibitor pembrolizumab in patients with advanced TNBC 50.5 32 IV PD-1 Pembrolizumab ≥1L 10.0 (0.4-19.5) 7

NOS, Newcastle-Ottawa Scale; mTNBC, metastatic triple-negative breast cancer; mBC, metastatic breast cancer; TNBC, triple-negative breast cancer; PD-1, programmed

Efficacy

Four studies evaluated the effects of PD-L1 inhibitors, while the remaining three analyzed the effects of PD-1 inhibitors, all on TNBC type (Table 2). Outcomes extracted to assess clinical activity include ORR, DCR, DOR, PFS, and OS.

Table 2.

Reported outcomes related to efficacy and adverse effects.

Progression-free survival (PFS)Overall survival (PFS) Adverse effects (Grade ≥3)
Year; Author ORR (%) [95% CI] DCR (%) [95% CI] DOR (months) [95% CI] Median PFS (months) [95% CI] PFS6m (%) PFS1y (%) HR [95% CI] Median OS (months) [95% CI] OS6m (%) OS1y (%) OS2y (%) HR [95% CI] Treatment related n (%)
2018; Adams S68 39.4 [22.9-57.9] 51.5 [33.5-69.2] 9.1 [2.9-20.9] 5.5 [5.1-7.7] 45.5 - - 14.7 [10.1-NE] - 57.4 40.6 - All AEs 24(73)
Neutropenia 15(46)
Thrombocytopenia 3(9)
Diarrhea 2(6)
Anemia 2(6)
Pneumonia 2(6)
Leukopenia 2(6)
Peripheral neuropathy 1(3)
Myalgia 1(3)
Bone pain 1(3)
Colitis 1(3)
Syncope 1(3)
Elevated AST 1(3)
Febrile neutropenia 1(3)
Type 1 diabetes mellitus 1(3)
Pneumonia mycoplasma 1(3)
Paronychia 1(3)
2018; Adams S Cohort A70 2018; Adams S Cohort A70 5.3 [2.7-9.9] 7.6 [4.4-12.7] 2.0 [1.9-2.0] 14.9 8.1 - 9.0 [7.6-11.2] 69.7 39.8 - - Any AEs 22(13)
Diarrhea 3 (1.8)
Fatigue 1 (0.6)
Nausea 1 (0.6)
2018; Adams S Cohort B71 21.4 [13.9-31.4] 23.8 [15.9-34.0] 10.4 [4.9-19.2] 2.1 [2.0-2.2] 27.0 - - 18.0 [12.9-23.0] 81.0 61.7 - - Any AEs 8 (9.5)
Diarrhea 1 (1.2)
Fatigue 1 (1.2)
Anemia 1 (1.2)
2018; Schmid P72 56.0 [51.3-60.6] - 7.4 [6.9-9.0] 7.2 [5.6-7.5] 50.1 23.7 0.62* [0.49-0.78] 21.3 [17.3-23.4] - 60.09 42.1 0.62* [0.45-0.86] Neutropenia 37 (8.2)
Peripheral neuropathy 25 (5.5)
Nausea 5 (1.1)
Alopecia 3 (0.7)
Pyrexia 3 (0.7)
2018; Emens LA69 24.0 [8.0-47.0] 29.0 21.0 [10.0-38.0] 1.4 [1.3-1.6] - - - 17.6 [10.2-NE] - 41.0 19.0 2.21° 1.17-4.16] Any AEs 46(40)
Hyperglycemia 1 (0.9)
Pneumonitis 1 (0.9)
Pulmonary hypertension 1 (0.9)
2017; irix LY73 5.2 [1.1-14.4] 31.0 - 5.9 [5.7-6.9] 12.4 - - 9.2 [4.3-NE] - 37.9 - - Any AEs 23 (13.7)
Fatigue 3 (1.8)
Anemia 3 (1.8)
Autoimmune hepatitis 3 (1.8)
Elevated GGT 3 (1.8)
Arthralgia 1 (0.6)
Acute hepatic failure 1 (0.6)
Dyspnea exertional 1 (0.6)
Elevated AST 1 (0.6)
Elevated ALT 1 (0.6)
Hypoxia 1 (0.6)
Hypokalemia 1 (0.6)
Pneumonitis 1 (0.6)
Axillary pain 1 (0.6)
Pleuritic pain 1 (0.6)
Non-cardiac chest pain 1 (0.6)
Thrombocytopenia 1 (0.6)
Cardiac arrest 1 (0.6)
Hypertriglyceridemia 1 (0.6)
Neutropenia 1 (0.6)
Proteinuria 1 (0.6)
Respiratory failure 1 (0.6)
Respiratory distress 1 (0.6)
Pulmonary arterial hypertension1 (0.6)
2016; Nanda R74 18.5 [6.3-38.1] 25.9 - 1.9 1.7-5.5] 24.4 - - 11.2 [5.3-NE] 66.7 43.0 - - Anemia 1 (3.1)
Aseptic meningitis 1 (3.1)
Disseminated intravascular (3.1)
coagulation
Headache 1 (3.1)
Lymphopenia 1 (3.1)
Pyrexia 1 (3.1)

ORR, overall response rate; DCR, disease control rate; DOR, duration of response; PFS, progression-free survival; PFS6m, 6-months PFS rate; PFS1y, 1-year PFS rate; OS, overall survival; OS6m, 6-months OS rate; OS1y, 1-year OS rate; OS2y, 2-year OS rate; HR, hazard ratio; NE, not estimable; AEs, adverse effects; AST, aspartate aminotransferase; ALT, alanine aminotransferase; GGT, -glutamyl transferase

*HR calculated between intervention arm and placebo arm

°HR calculated between 1st and 2nd line of therapy.

Overall, ORR ranges from 5.2–56.0 months and DCR ranges from 7.6 –51.5 months. DOR ranges from 9.1-21+ months, as median DOR was not reached in several studies. Median PFS ranges from 1.4-7.2 months, whereas median OS ranges from 9.2– 21.3 months.68-74 In one randomized trial by Schmid et al.,72 effects of PD-L1 inhibitor combined with nab-paclitaxel were compared to effects of nab-paclitaxel alone with placebo. Results showed a higher PFS (7.2 vs 5.5 months) and OS (21.3 vs 17.6 months) in intervention group when compared to placebo, with HR of 0.80 and 0.84 respectively.

Safety profiles

AEs in each study were assessed using National Cancer Institute Common Terminology Criteria for Adverse Events (CTCAE) version 4.0. In the included studies, AEs were described as generally manageable, acceptable, or well tolerable. Their results were consistent with known safety profiles of each agent and no new adverse effects were reported. AEs of grade ≥3 is summarized in Table 2. The rate of treatment-related AEs (TRAEs) of any grade ranges from 56.3% to 100%.68-74 Grade 3 or higher AEs were experienced by 9.5%-15.6% (n=35) patients administered pembrolizumab,70,71,74 13.7% (n=23) administered avelumab,73 and 12.9% (n=15) administered atezolizumab as monotherapy.69 In patients treated with combination of atezolizumab plus nab-paclitaxel, grade 3 or higher AEs were found in 48,7% (n=220) patients compared to 42.2% in placebo-nab-paclitaxel group,72 while in another study the rate is 73% (n=24) in atezolizumab-nab-paclitaxel group.68 Treatment-related deaths were found in 1, 2, 2 and 3 patients receiving pembrolizumab,74 avelumab,73 atezolizumab,69 and atezolizumab plus nab-paclitaxel,72 respectively.

Discussion

Plenty of studies have shown the potential of checkpoint inhibitor as a treatment candidate against cancer, and this type of immunotherapy has been approved in several cancer types, such as melanoma75 and lung cancer.76 Based on our systematic review, we found that checkpoint inhibitor also shows positive effects on breast cancer, especially TNBC, proven by increase in patients’ response rates and survival.

The mechanism by which immune checkpoint inhibitors function involves a unique way of modulating immune response against tumor cells. Unlike previous targeted therapy which target specific receptors on tumor cells, checkpoint inhibitors work by removing certain inhibitions which under normal conditions help to keep the immune system in check. This is based on the findings that cancer cells have certain immunosuppressive abilities which make use of normal immune regulatory mechanism.77 Under normal conditions, presentation of antigens to T cells, together with co-stimulatory signals from the binding of CD28 to its ligand B7-1/2, would result in T cell activation. T cells also express protein receptor molecules, such as CTLA-4 and PD-1, which function as checkpoint molecules. Binding of CTLA-4 to B7-1/2 or PD-1 to PD-L1 would inhibit T cell activation, thus preventing T cells from turning over-reactive. However, when this regulatory mechanism is hijacked by cancer cells, it helps to shield tumor cells from the host immune response.78,79

Preclinical studies with mice aimed to examine in vivo activities of anti-PD-1 against TNBC have previously yielded encouraging results. Anti-PD-1 was shown to succeed in inhibiting tumor growth in humanized mice implanted with tumor xenografts.36,80,81 Mice treated with anti-PD-1 had an increased level of antitumor Tcell responses and decreased activities of regulatory T cells as well as myeloid populations which may play an important role in suppressing immune response against tumor cells in host.80 These preclinical models give valuable insights regarding the mechanism of PD-1 blockade therapy against TNBC, and provide the basis to facilitate further investigation and clinical studies.

Clinical therapy of TNBC with PD-1 inhibitor shows a promising response and survival rate. Studies by Adams71 and Nanda74 shows response rates of 21.4% and 18.5% respectively, and median overall survival of 18 and 11.2 months respectively. Meanwhile, treatment with PD-L1 inhibitor also exhibit favorable outcomes, with an ORR as high as 39.4% and median OS of 14.7 months in a study by Adams.68 Another study by Schmid72 also compared the effects of PD-L1 inhibitor combined with chemotherapy nab-paclitaxel to the effects of nab-paclitaxel with placebo, and results show a significant increase in PFS and OS of the patients. Aside from its response rate, checkpoint inhibitors also show a durable response, as expressed by median DOR which ranges from 9.1 to 21+ months, compare to low DOR in chemotherapy of TNBC patients, ranging from 4 to 12 months.82 These outcomes demonstrate positive clinical antitumor activities when used as single or combination therapy, highlighting the potential of checkpoint inhibitors for the treatment of TNBC.

Effects of PD-1/PD-L1 inhibitors also differ when given as first-line therapy compared to second-line or greater treatment. Pembrolizumab, a PD-1 inhibitor, shows higher response rate and survival (ORR 21.4% and OS 18 months) when given as first-line therapy compared to when given after other systemic therapy (ORR 5.3% and OS 9 months).70,71 A study by Emens69 with PDL1 inhibitor also shows similar results, with ORR of 24% and OS 17.6 months when atezolizumab was given as first line therapy, compared to ORR 6% and OS 7.3 months when administered as second-line or greater treatment.69

The positive results discussed above were comparable to the effects of PD-1/PD-L1 blockade therapy in cancer types where it had been officially approved for treatment. A meta-analysis comparing the effects of such checkpoint inhibitor over docetaxel in patients with non-small-cell lung carcinoma (NSCLC), exhibited favorable outcomes in immunotherapy arms, shown by the significant increase in OS (HR=0.71) and PFS (HR=0.86).83 Similar results were shown with studies involving advanced melanoma, where significant difference were found in PFS (HR=0.53), OS (HR=0.60), and ORR (RR=2.87), all in favor of PD-1 inhibitors.84 These results demonstrated the possibility of PD-1/PD-L1 inhibitors to emerge as one of main choices of therapy for TNBC in the future, as how it had been for other cancer types.

Although clinical trials on PD-1/PD-L1 blockade show encouraging antitumor results, responses were seen better in certain subgroups of patients. This emphasize the need of certain markers to predict response to PD-1/PD-L1 blockade therapy, as well as to select proper candidates which would benefit most from the therapy. Patients with increased tumor PD-L1 expression are found to respond better to checkpoint inhibitor therapy. Pembrolizumab, when given to PD-L1 positive population, gives an ORR of 5.7% and DCR of 9.5%, higher than PD-L1 negative population with ORR 4.7% and DCR 4.7%. Another study by Nanda74 shows similar results, with increasing ORR as PD-L1 expression increases (P=0.028).74 Overexpression of PD-L1 in tumors help tumor cells to inhibit cytotoxic T cells, thereby allowing the cancer to evade the immune system, correlating with poorer prognosis. However, with PD-1/PD-L1 blockade therapy, the expression of these molecules could predict a better therapeutic effect, as these pre-existing binding of PD-1/PD-L1 provide a target for the drug’s blocking action.85

These results with TNBC is consistent with a meta-analysis conducted by Khunger86 which studied the predictive value PD-L1 expression as biomarker in other cancer types, including NSCLC, melanoma, renal, and bladder carcinoma. Except for bladder carcinoma, PD-L1 expression was found to be predictive of favorable responses in tumors treated with PD-1/PD-L1 inhibitors in the other cancer types (OR=2.77), where its effect was largest in NSCLC (OR=3.33).86 Nevertheless, studies found that the expression of PD-L1 is a dynamic process, thus sampling of tissues at one time may not accurately depict a patient’s response accurately. Moreover, up until now, there is still no exact cut-off values for PD-L1 overexpression.87 Thus, further studies regarding its use as a prognostic biomarker are still needed.

Checkpoint inhibitor therapy also demonstrated a lower response rate in patients with poor clinical prognostic factors. These include an elevated lactate dehydrogenase level which is associated with faster disease progression, presence of liver metastases, more metastatic sites, and other visceral diseases.69,70,74 Alternative therapy strategies as well as combination therapy should be considered in these settings to help improve response rate to therapy.70

Safety profiles of PD-1/PD-L1 inhibitors are generally acceptable. Grade 3 or higher AEs were found in 9.5%-15.6%, 13.7%, and 12.9% patients receiving pembrolizumab, avelumab, and atezolizumab, respectively. These numbers indicated that each agent have a similar safety profile. Furthermore, our results are comparable with recent studies on other types of metastatic cancer.88-91

Some common TRAEs of pembrolizumab therapy were fatigue (18,8%-26.2%; n=63),70,71,74 nausea (11.2%-13.1%; n=35),69,70,74 diarrhea (11.9%; n=10),70 arthralgia (18.8%; n=6),74 and myalgia (18,8%; n=6).74A study on metastatic esophageal cancer reported less incidence of fatigue (10.7%; n=13) and diarrhea (4.9%), despite similar proportion of pruritus (6.6% vs 6.3%- 7.1%70,71,74), rash (7.4% vs 6.0%70), and total AEs of grade 3-5 (12.4%).88 Fatigue was also the most common AE in patients with advanced recurrent ovarian cancer (33.8%; n=127), followed by nausea (15.4%).91 Additionally, the incidence of diarrhea (10.1%), pruritus (8.2%), and rash (7.2%) were similar;91 however, less patients experienced arthralgia (5.3%)91 compared to our included studies. Grade 3 or higher AEs including aseptic meningitis, headache, anemia, lymphopenia and pyrexia were in line with previous reports.92-94

The most common AEs of any grade in anti PD-L1 monotherapy were fatigue (13%-19.0%; n=47), pyrexia (16%; n=19),69 infusion- related reaction (14.3%; n=24),73 and nausea (11%-13.1%; n=35).69,73 A study in which patients with metastatic melanoma had received avelumab reported less grade 3 AEs (7.8%; n=4)89 when compared with avelumab monotherapy in patients with TNBC (13.7%; n=23).73 Infusion-related reaction had higher prevalence (29.4%) and was the most AE experienced by melanoma patients, followed by fatigue (17.6%), chills (11.8%), and diarrhea (9.8%).89 Atezolizumab monotherapy in patients with TNBC was assessed by Emens et al.69 In this study, grade 3-5 AEs were present in 12.9% (n=15) patients, with frequent AEs being pyrexia (16%), fatigue (13%), nausea (11%), diarrhea (10%), asthenia (10%), and pruritus (11%). In comparison, patients with metastatic urothelial cancer who were given atezolizumab monotherapy experienced less grade 3-5 AEs (9%; n=9). Both studies had similar incidences of fatigue (18%), asthenia (14%), pruritus (13%), nausea (12%) and diarrhea (7%).90

Grade 3 or higher AEs were mostly immune-mediated (Table 2), which might manifest as organ-specific autoimmune reaction.95 Dirix et al.73 reported two treatment-related deaths, one of which was due to respiratory distress in a patient with liver, lung, and soft tissue metastases. Grade 3 or higher AEs related to respiratory system also constituted significantly in this study, including dyspnea (5.4%; n=9) and pleural effusion (5.4%; n=9). Other treatmentrelated adverse events were similar to known safety profiles of anti PD-L1 agents.96

Most patients receiving atezolizumab plus nab-paclitaxel combination therapy experience neutropenia (70%; n=23), fatigue (67%; n=22), alopecia (42%; n=14), diarrhea (39%; n=13), peripheral sensory neuropathy (36%; n=12), and nausea (30%; n=10).68 When compared to placebo, higher frequency of nausea, pyrexia, hypothyroidism, neutropenia, and cough were found in the atezolizumab- nab-paclitaxel group, thus suggesting AEs caused by atezolizumab. The most common grade 3 or 4 AEs in both groups were neutropenia, decreased neutrophil count, peripheral neuropathy, fatigue, and anemia. Percentage of grade 3 or 4 peripheral neuropathy was higher than placebo (5.5%; n=25 vs 2.7%; n=12), which is consistent with a similar study on metastatic non-squamous NSCLC.97

Considering the pharmacodynamics of PD-1/PD-L1 inhibitors, most AEs are due to immune system activation.95 Therefore, immune-related AEs (irAEs) are of special interest and considered by all studies included in our review, regardless of attribution to treatment. According to a meta-analysis on other types of cancer, organ specific irAEs include hypothyroidism, pneumonitis, colitis, hepatitis and hypophysitis, with pneumonitis being the most common serious AE. Additionally, general adverse events such as fatigue, diarrhea, rash, are also the known AEs related to immune activation.95 Similarly, we found hypothyroidism,70,71,73 hyperthyroidism, 70,71,73 type 1 diabetes mellitus,70 pneumonitis,68-70,72,73 colitis,68,74 and hepatitis72-74 as irAEs, while fatigue, rash, and diarrhea are mostly grade 1 or 2.68-74 These results were consistent with the previous studies on checkpoint inhibitors.95,98

Though considerable amount of studies has shown the potential of checkpoint inhibitors in treating TNBC patients, more knowledge about their synergistic effect with other therapeutic agents is needed. In fact, combinations of these agents are currently evaluated. Common examples include the combination of chemotherapeutic drugs with checkpoint inhibitors or other agents of immunotherapy. NeoPACT99 is studying the effect of pembrolizumab when combined with carboplatin and docetaxel in stage I-III TNBC, while multiple combinations of chemotherapy with atezolizumab are included in the Morpheus-TNBC trial-a currently ongoing phase Ib/II trial.100 Another study is evaluating the combination of olaparib, a poly (ADP-ribose) polymerase inhibitor, and durvalumab (anti-PD-L1 monoclonal antibody) in metastatic TNBC.101 Chemotherapies mainly prevent cancer to grow and spread, while checkpoint inhibitors help the immune system to combat tumor cells; thus, combining these agents might work better against TNBC. Treatment of TNBC using combination of PVX-410 vaccine and pembrolizumab is currently on phase 1b.102 Since both agents ameliorate immune system against cancer, such combinations might offer better efficacy; however, increased rate and severity of AEs should be anticipated.

Study limitation

As in other studies, our systematic review also has several limitations. A major limitation is the fact that most studies included in our review are non-randomized and open-labelled. Moreover, checkpoint inhibitor is only recently tried in TNBC, thus most studies are still in phase I of II of clinical trials. A larger, randomized controlled trial with more samples is therefore needed to further confirm these results. Despite these drawbacks, we believe that the result of this review would provide useful clinical implications, serving as a basis to encourage further studies in this research area.

Conclusions

Based on the above systematic review, we found that checkpoint inhibitor exhibits an excellent potential as a novel therapy against TNBC. Clinical trials found that checkpoint inhibitors showed good antitumor activity, demonstrated by their promising response and survival rates. Responses are seen better with increased PD-L1 expression. Adverse effects of PD-1/PD-L1 blockade therapy are generally immune-related; however, these effects are mostly tolerable, with hardly any grade 3 or above adverse effects in each study. We hope that this result could serve as a basis for further studies regarding checkpoint inhibitor immunotherapy to uncover its potential as an effective and tolerable therapy for breast cancer, especially TNBC, thus helping to reduce its mortality rate worldwide.

Acknowledgments

We would like to show our gratitude to Vivian Soetikno, MD, for the guidance in manuscript writing and data interpretation. The corresponding author had the responsibility for the decision for publication.

References

  • 1.Global Cancer Observatory. Breast cancer. Geneva: World Health Organization; 2019. Available from: https://gco.iarc.fr/today/data/factsheets/cancers/20-Breast-fact-sheet.pdf Accessed: Feb 2019. [Google Scholar]
  • 2.Bray F, Ferlay J, Soerjomataram I, et al. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2018;68:394-424. [DOI] [PubMed] [Google Scholar]
  • 3.Cummings MC, Simpson PT, Reid LE, et al. Metastatic progression of breast cancer: Insights from 50 years of autopsies. J Pathol 2014;232:23-31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Jitariu A-A, Cîmpean AM, Ribatti D, Raica M. Triple negative breast cancer: the kiss of death. Oncotarget 2017;8:46652-62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Saraiva DP, Cabral MG, Jacinto A, Braga S. How many diseases is triple negative breast cancer: the protagonism of the immune microenvironment. ESMO Open 2017;2:e000208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Chen L, Linden HM, Anderson BO, Li CI. Trends in 5-year survival rates among breast cancer patients by hormone receptor status and stage. Breast Cancer Res Treat 2014;147:609-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Brewster AM, Chavez-MacGregor M, Brown P. Epidemiology, biology, and treatment of triple-negative breast cancer in women of African ancestry. Lancet Oncol 2015;15:e625-34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Yersal O, Barutca S. Biological subtypes of breast cancer: prognostic and therapeutic implications. World J Clin Oncol 2014;5:412-24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Gierach GL, Burke A, Anderson WF. Epidemiology of triple negative breast cancers. Breast Dis 2010;32:5-24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Plasilova ML, Hayse B, Killelea BK, et al. Features of triple-negative breast cancer. Medicine (Baltimore) 2016;95:e4614. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Al-Mahmood S, Sapiezynski J, Garbuzenko OB, Minko T. Metastatic and triple-negative breast cancer: challenges and treatment options. Drug Deliv Transl Res 2018;8:1483-507. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Reddy KB. Triple-negative breast cancers: an updated review on treatment options. Curr Oncol 2011;18:173-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Miller E, Lee HJ, Lulla A, et al. Current treatment of early breast cancer: adjuvant and neoadjuvant therapy. F1000Res 2014;3:198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Collignon J, Lousberg L, Schroeder H, Jerusalem G. Triple-negative breast cancer: treatment challenges and solutions. Breast Cancer Targets Ther 2016;8:93-107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Isakoff SJ. Triple negative breast cancer: role of specific chemotherapy agents. Cancer J 2010;16:53-61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Lebert JM, Lester R, Powell E, et al. Advances in the systemic treatment of triple-negative breast cancer. Curr Oncol 2018;25:S142-50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Gaston-Johansson F, Watkins CC, Kanu IK, et al. The effects of symptoms on quality of life during chemotherapy in African- American women with breast cancer. J Natl Black Nurse Assoc 2015;26:7-16. [PMC free article] [PubMed] [Google Scholar]
  • 18.Tao JJ, Visvanathan K, Wolff AC. Long term side effects of adjuvant chemotherapy in patients with early breast cancer. Breast 2015;24:S149-53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Kwa MJ, Adams S. Checkpoint inhibitors in triple-negative breast cancer (TNBC): where to go from here. Cancer 2018;124:2086-103. [DOI] [PubMed] [Google Scholar]
  • 20.Lu W, Li Z, Qiu Y, et al. Immunotherapeutic interventions of triple negative breast cancer. J Transl Med 2018;16:147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Seidel JA, Otsuka A, Kabashima K. Anti-PD-1 and anti-CTLA-4 therapies in cancer: mechanisms of action, efficacy, and limitations. Front Oncol 2018;8:86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Polk A, Svane IM, Andersson M, Nielsen D. Checkpoint inhibitors in breast cancer - current status. Cancer Treat Rev 2018;63:122-34. [DOI] [PubMed] [Google Scholar]
  • 23.Higgins J, Green S. Cochrane handbook for systematic reviews of interventions version 5.1.0. The Cochrane Collaboration; 2011. Available from: https://handbook-5-1.cochrane.org/ Accessed: Feb 2019. [Google Scholar]
  • 24.Liberati A, Altman DG, Tetzlaff J, et al. The PRISMA statement for reporting systematic reviews and meta-analyses of studies that evaluate health care interventions: explanation and elaboration. J Clin Epidemiol 2009;62:e1-34. [DOI] [PubMed] [Google Scholar]
  • 25.Wells G, Shea B, O’Connell D, et al. The Newcastle-Ottawa scale (NOS) for assessing the quality of nonrandomised studies in metaanalyses. Ottawa: Ottawa Hospital Research Institute; 2011. Available from: http://www.ohri.ca/programs/clinical_epidemiology/oxford.asp Accessed: Feb 2019. [Google Scholar]
  • 26.Gibson J. Anti-PD-L1 for metastatic triple-negative breast cancer. Lancet Oncol 2015;16:e264. [DOI] [PubMed] [Google Scholar]
  • 27.Nanda R, Specht J, Dees E, et al. 243 - Pembrolizumab for metastatic triple-negative breast cancer (mTNBC): long-lasting responses in the phase Ib KEYNOTE-012 study ECCO: 2017 European Cancer Congress 27 January 2017-30 January 2017. Eur J Cancer 2017;Suppl 1:S38. [Google Scholar]
  • 28.Kok M, Winer E, Loi S. Passion for immune checkpoint blockade in triple negative breast cancer: comment on the IMpassion130 study. Ann Oncol 2019;30:13-6. [DOI] [PubMed] [Google Scholar]
  • 29.Qin Y, Vasilatos S, Chen L, et al. Inhibition of histone lysine-specific demethylase 1 elicits breast tumor immunity and enhances antitumor efficacy of immune checkpoint inhibitor. Oncogene 2019;38:390-405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Reguera-Nuñez E, Xu P, Chow A, et al. Therapeutic impact of nintedanib with paclitaxel and/or a PD-L1 antibody in preclinical models of orthotopic primary or metastatic triple negative breast cancer. J Exp Clin Cancer Res 2019;38:16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Sidaway P. Setting dictates efficacy of pembrolizumab in TNBC. Nat Rev Clin Oncol 2019;16:66. [DOI] [PubMed] [Google Scholar]
  • 32.Gray MJ, Gong J, Hatch MMS, et al. Phosphatidylserine-targeting antibodies augment the anti-tumorigenic activity of anti-PD-1 therapy by enhancing immune activation and downregulating prooncogenic factors induced by T-cell checkpoint inhibition in murine triple-negative breast cancers. Breast Cancer Res 2016;18:1-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.He Q, Davis M, Bae S, et al. AR negative triple negative or “quadruple negative” breast cancers in African American women have an enriched basal and immune signature. PLoS One 2018;13:e0196909. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Mordoh J, Levy EM, Juliá EP, et al. Avelumab, an IgG1 anti-PDL1 immune checkpoint inhibitor, triggers NK cell-mediated cytotoxicity and cytokine production against triple negative breast cancer cells. Front Immunol 2018;9:1-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Walter CB, Grischke E-M, Brucker SY, et al. PD-1 and PD-L1 immune checkpoint blockade to treat breast cancer. Breast Care 2016;11:385-90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Wang M, Yao L-C, Cheng M, et al. Humanized mice in studying efficacy and mechanisms of PD-1-targeted cancer immunotherapy. FASEB J 2018;32:1537-49. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Bernard-Tessier A, Baldini C, Martin P, et al. Outcomes of longterm responders to anti-programmed death 1 and anti-programmed death ligand 1 when being rechallenged with the same anti-programmed death 1 and anti-programmed death ligand 1 at progression. Eur J Cancer 2018;101:160-4. [DOI] [PubMed] [Google Scholar]
  • 38.Eastman P. Atezolizumab improves OS in triple negative breast cancer. Oncol Times 2017;39:43-43. [Google Scholar]
  • 39.Jung K, LoRusso P, Burris H, et al. Phase I study of the indoleamine 2,3-dioxygenase (IDO1) inhibitor navoximod (GDC-0919) administered with PD-L1 inhibitor (atezolizumab) in advanced solid tumors. Clin Cancer Res 2019. [Epub ahead of print]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Tomioka N, Azuma M, Ikarashi M, et al. The therapeutic candidate for immune checkpoint inhibitors elucidated by the status of tumor-infiltrating lymphocytes (TILs) and programmed death ligand 1 (PD-L1) expression in triple negative breast cancer (TNBC). Breast Cancer 2018;25:34-42. [DOI] [PubMed] [Google Scholar]
  • 41.Simoneaux R. Eribulin mesylate plus pembrolizumab shows encouraging efficacy in TNBC. Oncol Times 2018;40:15-6. [Google Scholar]
  • 42.Bernier C, Soliman A, Gravel M, et al. DZ-2384 has a superior preclinical profile of taxanes for the treatment of triple-negative breast cancer and is synergistic with anti-CTLA-4 immunotherapy. Anticancer Drugs 2018;29:774-85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Stirrups R. Atezolizumab for metastatic triple-negative breast cancer. Lancet Oncol 2018;19:e519. [DOI] [PubMed] [Google Scholar]
  • 44.Siomski A. Chemoimmunotherapy for triple-negative breast cancer. JAMA 2018;320:2521. [DOI] [PubMed] [Google Scholar]
  • 45.Rocca LE, Dispinzieri M, Lozza L, et al. Radiotherapy with the anti-programmed cell death ligand-1 immune checkpoint blocker avelumab: acute toxicities in triple-negative breast cancer. Med Oncol 2019;36:1-1. [DOI] [PubMed] [Google Scholar]
  • 46.Heimes A, Schmidt M. Atezolizumab for the treatment of triplenegative breast cancer. Expert Opin Investig Drugs 2019;28:1-5. [DOI] [PubMed] [Google Scholar]
  • 47.Castan JC, Guo Z, Karantza V, Aktan G. KEYNOTE-335: randomized, double-blind, phase III study of pembrolizumab (pembro) 1 chemotherapy (chemo) vs placebo (PBO) 1 chemo for previously untreated, locally recurrent, inoperable or metastatic triplenegative breast cancer (mTNBC). Ann Oncol 2017;28:v72-. [Google Scholar]
  • 48.Emens L, Adams S, Loi S, et al. IMpassion130: a phase III trial comparing 1L atezolizumab with nab-paclitaxel in treatmentnaive patients with mTNBC. Ann Oncol 2016;27:ix40-. [Google Scholar]
  • 49.Mittendorf E, Barrios C, Harbeck N, et al. IMpassion031: a phase III study comparing neoadjuvant atezolizumab vs placebo in combination with nab-paclitaxel-based chemotherapy in early triplenegative breast cancer (TNBC). Cancer Res 2018;78:[1 p.]. [Google Scholar]
  • 50.Page D, Yu W, Conlin A, et al. A pilot/phase II study evaluating pembrolizumab plus standard-of-care chemotherapy in metastatic triple-negative breast cancer, with companion comprehensive immunological monitoring. J Immunother Cancer 2016;4:[1 p.]. [Google Scholar]
  • 51.Obeid E, Miller K, Sparano J, et al. A phase II randomized trial of pembrolizumab with carboplatin and gemcitabine for treatment of patients with metastatic triple-negative breast cancer (mTNBC). Cancer Res 2017;77:[1 p.]. [Google Scholar]
  • 52.Pohlmann P, Diamond J, Hamilton E, et al. Atezolizumab (atezo) + nab-paclitaxel (nab-pac) in metastatic triple-negative breast cancer (mTNBC): 2-year update from a ph Ib trial. Cancer Res 2018;78:[1 p.]. [Google Scholar]
  • 53.Pusztai L, Silber A, Hofstatter E, et al. Safety of MEDI4736 (anti- PD-L1 antibody) administered concomitant with weekly nabpaclitaxel and dose dense doxorubicin/cyclophosphamide (ddAC) as neoadjuvant chemotherapy for stage I-III triple negative breast cancer (TNBC): a phase I/II trial. J Clin Oncol 2017;35:[1 p.]. [Google Scholar]
  • 54.Schmid P, Park YH, Munoz-Couselo E, et al. Pembrolizumab (pembro) + chemotherapy (chemo) as neoadjuvant treatment for triple negative breast cancer (TNBC): preliminary results from KEYNOTE-173. J Clin Oncol 2017;35:[1 p.]. [Google Scholar]
  • 55.Schmid P, Castan JC, Bergh J, et al. KEYNOTE-522: phase III study of pembrolizumab (pembro) 1 chemotherapy (chemo) vs placebo 1 chemo as neoadjuvant followed by pembro vs placebo as adjuvant therapy for triple-negative breast cancer (TNBC). Ann Oncol 2017;28:v72-. [Google Scholar]
  • 56.Spira A, Hamid O, Bauer T, et al. Efficacy/safety of epacadostat plus pembrolizumab in triple-negative breast cancer and ovarian cancer: phase I/II ECHO-202 study. J Clin Oncol 2017;35:[1 p.]. [Google Scholar]
  • 57.Tolaney S, Savulsky C, Aktan G, et al. Phase 1b/2 study to evaluate eribulin mesylate in combination with pembrolizumab in patients with metastatic triple-negative breast cancer. Cancer Res 2017;77:[1 p.]. [Google Scholar]
  • 58.Wilsanen LS. Long-term clinical outcomes and biomarker analyses of atezolizumab therapy in metastatic triple-negative breast cancer. Clin Breast Cancer 2018;Suppl:6-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Forero A, Stroyakovskiy D, Cha E, et al. ENCORE 602: a randomized, placebo-controlled, double-blind, multicenter phase 2 study (with a phase 1b lead-in) of atezolizumab with or without entinostat in patients with advanced triple negative breast cancer (aTNBC). Cancer Res. 2017;77:[1 p.]. [Google Scholar]
  • 60.Iglesias J, Prathikanti R, Ma B, et al. A multicenter, open-label, phase II study of PGG beta-glucan and pembrolizumab in patients (pts) with advanced melanoma (MEL) following progression on treatment with checkpoint inhibitors (CPI) or triple negative breast cancer (TNBC) failing front-line ch. J Clin Oncol 2017;35:[1 p.]. [Google Scholar]
  • 61.Isakoff S, Tolaney SM, Tung N, et al. A phase 1b study of safety and immune response to PVX-410 vaccine alone and in combination with durvalumab (MEDI4736) in HLA-A2+ patients following adjuvant therapy for stage 2/3 triple negative breast cancer. J Clin Oncol 2017;35:[1 p.]. [Google Scholar]
  • 62.Kok M, Horlings H, van de Vijver K, et al. Adaptive phase II randomized non-comparative trial of nivolumab after induction treatment in triple negative breast cancer: tONIC-trial. Ann Oncol 2017;28:v608-. [Google Scholar]
  • 63.Litton J, Moulder S, Helgason T, et al. Triple-negative first-line study: neoadjuvant trial of nab-paclitaxel and atezolizumab, a PDL1 inhibitor, in patients with triple negative breast cancer (TNBC) (NCT02530489). Cancer Res 2017;77:[1 p.]. [Google Scholar]
  • 64.Loibl S, Untch M, Burchardi N, et al. A randomized phase II neoadjuvant study (GeparNuevo) to investigate the addition of durvalumab, a PD-L1 antibody, to a taxane-anthracycline containing chemotherapy in triple negative breast cancer (TNBC). J Clin Oncol 2017;35:[1 p.]. [Google Scholar]
  • 65.Miles D, Andre F, Gligorov J, et al. IMpassion131: phase III study comparing 1L atezolizumab with paclitaxel vs placebo with paclitaxel in treatment-naive patients with inoperable locally advanced or metastatic triple negative breast cancer (mTNBC). Ann Oncol 2018;28:v105-. [Google Scholar]
  • 66.Miles D, Kim S-B, McNally V, et al. COLET: a multistage, phase 2 study evaluating the safety and efficacy of a doublet regimen of cobimetinib (C) in combination with paclitaxel (P) or triplet regimens of C in combintion with atezolizumab (atezo) plus either P or nabpaclitaxel (nab-P) in metastatic teiple-negative breast cancer (TNBC). Cancer Res 2017;77:[1 p.]. [Google Scholar]
  • 67.Yu X-R, Wen Q-S, Xiao Y, et al. [Blocking programmed deathligand 1 attenuates maturation inhibition of dendritic cells by cocultured breast cancer cells]. Nan Fang Yi Ke Da Xue Xue Bao 2018;38:561-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Adams S, Diamond JR, Hamilton E, et al. Atezolizumab plus nabpaclitaxel in the treatment of metastatic triple-negative breast cancer with 2-year survival follow-up: a phase 1b clinical trial. JAMA Oncol 2018;E1-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Emens LA, Cruz C, Eder JP, et al. Long-term clinical outcomes and biomarker analyses of atezolizumab therapy for patients with metastatic triple-negative breast cancer. JAMA Oncol 2018;5:74-82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Adams S, Schmid P, Rugo H, et al. Pembrolizumab monotherapy for previously treated metastatic triple-negative breast cancer: cohort A of the phase 2 KEYNOTE-086 study. Ann Oncol 2018:1-31. [DOI] [PubMed] [Google Scholar]
  • 71.Adams S, Loi S, Toppmeyer D, et al. Pembrolizumab monotherapy for previously untreated, PD-L1-positive, metastatic triplenegative breast cancer: cohort B of the phase 2 KEYNOTE-086 study. Ann Oncol 2018:1-28. [DOI] [PubMed] [Google Scholar]
  • 72.Schmid P, Adams S, Rugo H, et al. Atezolizumab and nab-paclitaxel in advanced triple-negative breast cancer. N Engl J Med 2018;379:2108-21. [DOI] [PubMed] [Google Scholar]
  • 73.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 2017;167:671-86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Nanda R, Chow LQ, Dees EC, et al. Pembrolizumab in patients with advanced triple-negative breast cancer: phase Ib KEYNOTE-012 study. J Clin Oncol 2016;34:2460-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Lugowska I, Teterycz P, Rutkowski P. Immunotherapy of melanoma. Contemp Oncol (Pozn) 2018;22:61-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Jain P, Jain C, Velcheti V. Role of immune-checkpoint inhibitors in lung cancer. Ther Adv Respir Dis 2018;12:1753465817750075. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.National Cancer Institute. What is cancer?. United States: National Cancer Institute; 2015. Available from: https://www.cancer.gov/about-cancer/understanding/what-is-cancer Accessed: Jan 2019. [Google Scholar]
  • 78.Webb ES, Liu P, Baleeiro R, et al. Immune checkpoint inhibitors in cancer therapy. J Biomed Res 2018;32:317-26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Sharpe AH. Introduction to checkpoint inhibitors and cancer immunotherapy. Immunol Rev. 2017;276:5-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Capasso A, Lang J, Pitts TM, et al. Characterization of immune responses to anti-PD-1 mono and combination immunotherapy in hematopoietic humanized mice implanted with tumor xenografts. J Immunother Cancer 2019;7:37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Rosato RR, Dávila-González D, Choi DS, et al. Evaluation of anti-PD-1-based therapy against triple-negative breast cancer patient-derived xenograft tumors engrafted in humanized mouse models. Breast Cancer Res 2018;20:108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Senkus E, Cardoso F, Pagani P. Time for more optimism in metastatic breast cancer?. Cancer Treat Rev 2014;40:220-8. [DOI] [PubMed] [Google Scholar]
  • 83.Liu J, Zhong Y, Peng S, et al. Efficacy and safety of PD1/PDL1 blockades versus docetaxel in patients with pretreated advanced non-small-cell lung cancer: a meta-analysis. Onco Targets Ther 2018;11:8623-32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Li J, Gu J. Efficacy and safety of PD-1 inhibitors for treating advanced melanoma: a systematic review and meta-analysis. Immunotherapy 2018;10:1293-302. [DOI] [PubMed] [Google Scholar]
  • 85.Geng Z, Xiao Y, Zhu XJ, et al. Anti-PD-1 therapy for clinical treatment of lymphoma: a single-arm meta-analysis. Oncotarget 2018;9:35343-55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Khunger M, Rakshit S, Schalper KA, et al. Meta-analysis of tumor PD-L1 expression as a predictive biomarker of benefit from PD-1/PD-L1 axis inhibitors in solid tumors. J Clin Oncol 2016;34:11603. [DOI] [PubMed] [Google Scholar]
  • 87.Meng X, Huang Z, Teng F, et al. Predictive biomarkers in PD- 1/PD-L1 checkpoint blockade immunotherapy. Cancer Treat Rev 2015;41:868-76. [DOI] [PubMed] [Google Scholar]
  • 88.Shah MA, Kojima T, Hochhauser D, et al. Efficacy and safety of pembrolizumab for heavily pretreated patients with advanced, metastatic adenocarcinoma or squamous cell carcinoma of the esophagus: the phase 2 KEYNOTE-180 study. JAMA Oncol 2018;10021:1-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Keilholz U, Mehnert JM, Bauer S, et al. Avelumab in patients with previously treated metastatic melanoma: phase 1b results from the JAVELIN solid tumor trial. J Immunother Cancer 2019;7:12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Petrylak DP, Powles T, Bellmunt J, et al. Atezolizumab (MPDL3280A) monotherapy for patients with metastatic urotherial cancer: long term outcomes from a phase 1 study. JAMA Oncol 2018;4:537-44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Matulonis UA, Shapira-Frommer R, Santin AD, et al. Antitumor activity and safety of pembrolizumab in patients with advanced recurrent ovarian cancer: results from the phase 2 KEYNOTE- 100 study. Ann Oncol 2019;Pii:mdz135. [Epub ahead of print] [DOI] [PubMed] [Google Scholar]
  • 92.Atwal D, Joshi KP, Ravilla R, Mahmoud F. Pembrolizumabinduced pancytopenia: a case report. Perm J 2017;21:17-004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Linardou H, Gogas H. Toxicity management of immunotherapy for patients with metastatic melanoma. Ann Transl Med 2016;4:272. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Blackmon JT, Viator TM, Conry RM. Central nervous system toxicities of anti-cancer immune checkpoint blockade. J Neurol Neuromed 2016;1:39-45. [Google Scholar]
  • 95.Baxi S, Yang A, Gennarelli RL, et al. Immune-related adverse events for anti-PD-1 and anti-PD-L1 drugs: systematic review and meta-analysis. BMJ 2018;360:k793. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Davies M, Duffield EA. Safety of checkpoint inhibitors for cancer treatment: strategies for patient monitoring and management of immune-mediated adverse events. Immunotargets Ther 2017;6:51-71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Socinski MA, Jotte RM, Cappuzzo F, et al. Atezolizumab for firstline treatment of metastatic nonsquamous NSCLC. N Engl J Med 2018;378:2288-301. [DOI] [PubMed] [Google Scholar]
  • 98.Ferrari SM, Fallahi P, Galetta F, et al. Thyroid disorders induced by checkpoint inhibitors. Rev Endocr Metab Disord 2018;19:325-33. [DOI] [PubMed] [Google Scholar]
  • 99.ClinicalTrials.gov. Neoadjuvant phase II study of pembrolizumab and carboplatin plus docetaxel in triple negative breast cancer (NeoPACT). Bethesda: National Library of Medicine (US); 2018. Available from: https://clinicaltrials.gov/ct2/show/NCT03639948 Accessed Aug 2019. [Google Scholar]
  • 100.ClinicalTrials.gov. A study evaluating the efficacy and safety of multiple immunotherapy-based treatment combinations in patients with metastatic or inoperable locally advanced triple-negative breast cancer (Morpheus-TNBC) (Morpheus-TNBC). Bethesda: National Library of Medicine (US); 2018. Available from: https://clinicaltrials.gov/ct2/show/NCT03424005 Accessed: Aug 2019. [Google Scholar]
  • 101.ClinicalTrials.gov. Olaparib and durvalumab in treating participants with metastatic triple negative breast cancer. Bethesda: National Library of Medicine (US); 2019. https://clinicaltrials.gov/ct2/show/NCT03544125 Accessed: Aug 2019. [Google Scholar]
  • 102.ClinicalTrials.gov. PVX-410 vaccine plus pembrolizumab in HLA-A2+ metastatic triple negative breast cancer. Bethesda: National Library of Medicine (US); 2017. Available from: https://clinicaltrials.gov/ct2/show/NCT03362060 Accessed: Aug 2019. [Google Scholar]

Articles from Oncology Reviews are provided here courtesy of Frontiers Media SA

RESOURCES