Abstract

Lessons Learned

  • The combination of enobosarm and pembrolizumab was well tolerated and showed a modest clinical benefit rate of 25% at 16 weeks.

  • Future trials investigating androgen receptor‐targeted therapy in combination with immune checkpoint inhibitors are warranted.

Background

Luminal androgen receptor is a distinct molecular subtype of triple‐negative breast cancer (TNBC) defined by overexpression of androgen receptor (AR). AR‐targeted therapy has shown modest activity in AR‐positive (AR+) TNBC. Enobosarm (GTx‐024) is a nonsteroidal selective androgen receptor modulator (SARM) that demonstrates preclinical and clinical activity in AR+ breast cancer. The current study was designed to explore the safety and efficacy of the combination of enobosarm and pembrolizumab in patients with AR+ metastatic TNBC (mTNBC).

Methods

This study was an open‐label phase II study for AR+ (≥10%, 1+ by immunohistochemistry [IHC]) mTNBC. Eligible patients received pembrolizumab 200 mg intravenous (IV) every 3 weeks and enobosarm 18 mg oral daily. The primary objective was to evaluate the safety of enobosarm plus pembrolizumab and determine the response rate. Peripheral blood, tumor biopsies, and stool samples were collected for correlative analysis.

Results

The trial was stopped early because of the withdrawal of GTx‐024 drug supply. Eighteen patients were enrolled, and 16 were evaluable for responses. Median age was 64 (range 36–81) years. The combination was well tolerated, with only a few grade 3 adverse events: one dry skin, one diarrhea, and one musculoskeletal ache. The responses were 1 of 16 (6%) complete response (CR), 1 of 16 (6%) partial response (PR), 2 of 16 (13%) stable disease (SD), and 12 of 16 (75%) progressive disease (PD). Response rate (RR) was 2 of 16 (13%). Clinical benefit rate (CBR) at 16 weeks was 4 of 16 (25%). Median follow‐up was 24.9 months (95% confidence interval [CI], 17.5–30.9). Progression‐free survival (PFS) was 2.6 months (95% CI, 1.9–3.1) and overall survival (OS) was 25.5 months (95% CI, 10.4–not reached [NR]).

Conclusion

The combination of enobosarm and pembrolizumab was well tolerated, with a modest clinical benefit rate of 25% at 16 weeks in heavily pretreated AR+ TNBC without preselected programmed death ligand‐1 (PD‐L1). Future clinical trials combining AR‐targeted therapy with immune checkpoint inhibitor (ICI) for AR+ TNBC warrant investigation.

Discussion

There has been strong interest in targeting AR in breast cancer; however, single‐agent androgen receptor antagonists have shown modest benefit to date. Single‐agent bicalutamide showed a CBR of 19% at 6 months and no PR. Single‐agent enzalutamide trial reported a CBR of 35% at 4 months with a median PFS of 14 weeks and a RR of 5.9% [1]. Early literature supports synergy of AR‐targeted therapy with ICI, and concurrent expression of PD‐L1 and AR in TNBC has the potential to be a molecular biomarker.

Enobosarm (GTx‐024) is an oral nonsteroidal, tissue‐SARM that was initially developed for anabolic activity with minimal androgenic activity [2,3,4]. Preclinical study demonstrated that enobosarm inhibited proliferation and tumor growth of AR+ TNBC [5]. In this clinical trial of combined enobosarm and pembrolizumab in heavily pretreated patients with AR+ TNBC without preselected PD‐L1, we observed an RR of 12% and CBR of 25% at 16 weeks, with one patient showing complete response (Fig. 1). The modest response rate is comparable to the single‐agent pembrolizumab RR of 12% observed in KEYNOTE‐12 but was higher than the RR of 5% observed in KEYNOTE‐86 [6,7]. There were no grade ≥ 4 adverse events (AEs) observed on the study and minimal grade 3 AEs: one (6%) musculoskeletal ache, one (6%) dry skin, and one (6%) diarrhea.

Response to therapy and PFS. Black triangle, response started.
FIGURE 1

Response to therapy and PFS. Black triangle, response started.

Abbreviations: CR, complete response; NA, not applicable; PD, progression of disease; PR, partial response; SD, stable disease; X, off treatment for progression; Y, treatment completed.

Median follow‐up was 24.9 months (95% CI, 17.5–30.9). PFS was 2.6 months (95% CI, 1.9–3.1), and overall survival was 25.5 months (95% CI, 10.4–NR). The trial was stopped early because of withdrawal of the enobosarm drug supply. Our results may reflect modest efficacy of enobosarm as an AR‐targeted agent. Future clinical trials investigating AR‐targeted therapy in combination with ICI for AR+ TNBC are warranted.

Trial Information

DiseaseBreast cancer
Stage of Disease/TreatmentMetastatic/advanced
Prior TherapyMore than 2 prior regimens
Type of StudyPhase II, single arm
Primary EndpointsSafety, overall response rate
Secondary EndpointsProgression‐free survival, overall survival, correlative endpoint, toxicity
Additional Details of Endpoints or Study Design
This study was a phase II trial to evaluate efficacy and safety for pembrolizumab with enobosarm in AR‐positive mTNBC conducted from April 2017 to April 2019 at City of Hope (NCT 02971761). All patients provided written informed consent before enrollment. Patients received pembrolizumab 200 mg IV on day 1 and enobosarm 18 mg oral daily in a 21‐day cycle. The primary objectives were to evaluate the tolerability and determine the response rate of the combination regimen. The AEs were graded by CTCAE version 4.0, and responses were assessed by RECIST 1.1. The secondary objective was to evaluate clinical outcomes, including CBR at 16 weeks, PFS, and OS.
The initial patients were followed in a safety lead‐in with only three patients at risk for first‐cycle toxicities to observe if a dose adjustment was appropriate. If in the first six patients we observed two or more patients with dose limiting toxicities (DLTs) in the first cycle, this would mandate a reduction in the starting dose for all subsequent patients. If dose level 1 had two patients with DLTs in the first six, the study would hold accrual.
Initially, based only on KEYNOTE‐12 (PD‐L1+ patients), we designed the study to discriminate between a 19% vs. 39% response rate. As the larger KEYNOTE‐86 results became available and demonstrated a 5% response rate with pembrolizumab (not restricted to PD‐L1+ patients), we amended the design at interim analysis to consider 2 of 15 (13.3%) with response to be sufficient justification for additional patients (previously requiring at least 3 of 15), and changed the primary focus to obtaining an estimate of the response of the combination, whereas with 29 patients, the standard error of the response rate would be 9% or less. Nevertheless, the trial was stopped early because of the withdrawal of GTx‐024 drug supply when 18 patients were enrolled (16 evaluable for response). Survival endpoints were estimated by the Kaplan‐Meier method, and median follow‐up was estimated by reverse Kaplan‐Meier. All eligible patients who started treatment were considered in the calculation of the response rate.
Although both drugs were well tolerated with nonoverlapping toxicities, the initial patients would be followed in a safety lead‐in with only three patients at risk for first‐cycle toxicities to observe if a dose adjustment was appropriate [8]. If in the first six patients we observed two or more patients with dose limiting toxicities in the first cycle, this would mandate a reduction in the starting dose for all subsequent patients.
The correlative studies were planned to refine patient selection for future studies and understand the role of AR pathway activities and immune changes on the activity of the combination of pembrolizumab plus enobosarm. These correlative studies were considered exploratory in the context of this limited phase II study. Tumor biomarkers and peripheral immune characteristics were evaluated using unpaired Mann‐Whitney tests. In addition, the absolute abundance of 18 immune cell types using CIBERSORT analysis of immune subsets and AR signaling pathway analysis of responders versus nonresponders were compared by unpaired Mann‐Whitney tests. Gut microbiome analysis was performed using hierarchical clustering of species classified by MetaPhIAn2 by patient. Linear discriminant analysis effect size (LEfSe) was used to determine the species most likely to explain differences between classes by coupling standard tests for statistical significance.
Eligible patients were 18 years or older with mTNBC who were willing to provide a sample from a recently obtained (within 42 days prior to initiation of day 1) biopsy of a tumor lesion. All patients had histologically proven diagnosis of TNBC: added ER negative (≤10% positive tumor nuclei), Progression Receptor (PR) negative (≤10% positive tumor nuclei), and HER2 expression 0 or 1+ by IHC staining or negative amplification by fluorescence in situ hybridization. AR+ was defined as ≥10% nuclear AR staining by IHC using SP107 antibody in the metastatic lesion. Other inclusion criteria were (a) Eastern Cooperative Oncology Group performance status of 0–1, (b) measurable disease per RECIST 1.1, (c) adequate hepatic, renal, and bone marrow functions defined as: liver enzymes <2.5 × upper limit of normal (ULN) or ≤ 5.0 × ULN if liver metastases present; serum total bilirubin ≤1.5 × ULN or direct bilirubin ≤ULN if total bilirubin levels >1.5 ULN; serum creatinine ≤1.5 × ULN or creatinine clearance ≥60 mL/min if creatinine levels >1.5 x ULN; absolute neutrophil count 1,500/mL; platelets 100,000/mL; and hemoglobin ≥9 g/dL. Patients who received prior treatment with immune checkpoint inhibitors ICIs or any antibody targeting other immunoregulatory receptors or mechanisms were excluded from the study. Other exclusion criteria were (a) prior exposure to AR‐targeted agents (including enobosarm, enzalutamide, or other AR‐targeted therapies); (b) testosterone or testosterone‐like agents, or other androgenic compounds or antiandrogens within 30 days prior to day 1 of protocol therapy; or (c) chronic systemic steroid therapy with prednisone dose ≥10 mg daily or equivalent, or on any other form of immunosuppressive medication. Patients with unstable or untreated brain or leptomeningeal metastasis were also removed from the study.
Investigator's Analysis
The trial was stopped early because of the withdrawal of GTx‐024 drug supply. Full assessment of activity of this combination could not be completed as planned.
DiseaseBreast cancer
Stage of Disease/TreatmentMetastatic/advanced
Prior TherapyMore than 2 prior regimens
Type of StudyPhase II, single arm
Primary EndpointsSafety, overall response rate
Secondary EndpointsProgression‐free survival, overall survival, correlative endpoint, toxicity
Additional Details of Endpoints or Study Design
This study was a phase II trial to evaluate efficacy and safety for pembrolizumab with enobosarm in AR‐positive mTNBC conducted from April 2017 to April 2019 at City of Hope (NCT 02971761). All patients provided written informed consent before enrollment. Patients received pembrolizumab 200 mg IV on day 1 and enobosarm 18 mg oral daily in a 21‐day cycle. The primary objectives were to evaluate the tolerability and determine the response rate of the combination regimen. The AEs were graded by CTCAE version 4.0, and responses were assessed by RECIST 1.1. The secondary objective was to evaluate clinical outcomes, including CBR at 16 weeks, PFS, and OS.
The initial patients were followed in a safety lead‐in with only three patients at risk for first‐cycle toxicities to observe if a dose adjustment was appropriate. If in the first six patients we observed two or more patients with dose limiting toxicities (DLTs) in the first cycle, this would mandate a reduction in the starting dose for all subsequent patients. If dose level 1 had two patients with DLTs in the first six, the study would hold accrual.
Initially, based only on KEYNOTE‐12 (PD‐L1+ patients), we designed the study to discriminate between a 19% vs. 39% response rate. As the larger KEYNOTE‐86 results became available and demonstrated a 5% response rate with pembrolizumab (not restricted to PD‐L1+ patients), we amended the design at interim analysis to consider 2 of 15 (13.3%) with response to be sufficient justification for additional patients (previously requiring at least 3 of 15), and changed the primary focus to obtaining an estimate of the response of the combination, whereas with 29 patients, the standard error of the response rate would be 9% or less. Nevertheless, the trial was stopped early because of the withdrawal of GTx‐024 drug supply when 18 patients were enrolled (16 evaluable for response). Survival endpoints were estimated by the Kaplan‐Meier method, and median follow‐up was estimated by reverse Kaplan‐Meier. All eligible patients who started treatment were considered in the calculation of the response rate.
Although both drugs were well tolerated with nonoverlapping toxicities, the initial patients would be followed in a safety lead‐in with only three patients at risk for first‐cycle toxicities to observe if a dose adjustment was appropriate [8]. If in the first six patients we observed two or more patients with dose limiting toxicities in the first cycle, this would mandate a reduction in the starting dose for all subsequent patients.
The correlative studies were planned to refine patient selection for future studies and understand the role of AR pathway activities and immune changes on the activity of the combination of pembrolizumab plus enobosarm. These correlative studies were considered exploratory in the context of this limited phase II study. Tumor biomarkers and peripheral immune characteristics were evaluated using unpaired Mann‐Whitney tests. In addition, the absolute abundance of 18 immune cell types using CIBERSORT analysis of immune subsets and AR signaling pathway analysis of responders versus nonresponders were compared by unpaired Mann‐Whitney tests. Gut microbiome analysis was performed using hierarchical clustering of species classified by MetaPhIAn2 by patient. Linear discriminant analysis effect size (LEfSe) was used to determine the species most likely to explain differences between classes by coupling standard tests for statistical significance.
Eligible patients were 18 years or older with mTNBC who were willing to provide a sample from a recently obtained (within 42 days prior to initiation of day 1) biopsy of a tumor lesion. All patients had histologically proven diagnosis of TNBC: added ER negative (≤10% positive tumor nuclei), Progression Receptor (PR) negative (≤10% positive tumor nuclei), and HER2 expression 0 or 1+ by IHC staining or negative amplification by fluorescence in situ hybridization. AR+ was defined as ≥10% nuclear AR staining by IHC using SP107 antibody in the metastatic lesion. Other inclusion criteria were (a) Eastern Cooperative Oncology Group performance status of 0–1, (b) measurable disease per RECIST 1.1, (c) adequate hepatic, renal, and bone marrow functions defined as: liver enzymes <2.5 × upper limit of normal (ULN) or ≤ 5.0 × ULN if liver metastases present; serum total bilirubin ≤1.5 × ULN or direct bilirubin ≤ULN if total bilirubin levels >1.5 ULN; serum creatinine ≤1.5 × ULN or creatinine clearance ≥60 mL/min if creatinine levels >1.5 x ULN; absolute neutrophil count 1,500/mL; platelets 100,000/mL; and hemoglobin ≥9 g/dL. Patients who received prior treatment with immune checkpoint inhibitors ICIs or any antibody targeting other immunoregulatory receptors or mechanisms were excluded from the study. Other exclusion criteria were (a) prior exposure to AR‐targeted agents (including enobosarm, enzalutamide, or other AR‐targeted therapies); (b) testosterone or testosterone‐like agents, or other androgenic compounds or antiandrogens within 30 days prior to day 1 of protocol therapy; or (c) chronic systemic steroid therapy with prednisone dose ≥10 mg daily or equivalent, or on any other form of immunosuppressive medication. Patients with unstable or untreated brain or leptomeningeal metastasis were also removed from the study.
Investigator's Analysis
The trial was stopped early because of the withdrawal of GTx‐024 drug supply. Full assessment of activity of this combination could not be completed as planned.

Drug Information

Drug 1
Generic/Working NamePembrolizumab
Trade NameKeytruda
Company NameMerck
Drug TypeAntibody
Drug ClassImmune therapy
Dose200 mg per flat dose
RouteIV
Schedule of AdministrationEvery 3 weeks (21 day cycle)
Drug 2
Generic/Working NameEnobosarm
Trade NameEnobosarm
Company NameGTx, Inc.
Drug TypeBiological
Drug ClassAndrogen receptor
Dose18 mg per flat dose
Routeoral (po)
Schedule of AdministrationDaily on 21‐day cycle
Drug 1
Generic/Working NamePembrolizumab
Trade NameKeytruda
Company NameMerck
Drug TypeAntibody
Drug ClassImmune therapy
Dose200 mg per flat dose
RouteIV
Schedule of AdministrationEvery 3 weeks (21 day cycle)
Drug 2
Generic/Working NameEnobosarm
Trade NameEnobosarm
Company NameGTx, Inc.
Drug TypeBiological
Drug ClassAndrogen receptor
Dose18 mg per flat dose
Routeoral (po)
Schedule of AdministrationDaily on 21‐day cycle

Patient Characteristics

Number of Patients, Male0
Number of Patients, Female16
StageIV
AgeMedian (range): 64 (36–81) years
Performance Status: ECOG

0 — 7

 

1 — 9

 

2 —

 

3 —

 

Unknown —

Cancer Types or Histologic SubtypesTriple‐negative breast cancer, 16
Number of Patients, Male0
Number of Patients, Female16
StageIV
AgeMedian (range): 64 (36–81) years
Performance Status: ECOG

0 — 7

 

1 — 9

 

2 —

 

3 —

 

Unknown —

Cancer Types or Histologic SubtypesTriple‐negative breast cancer, 16

Primary Assessment Method

TitleResponse assessment
Number of Patients Screened22
Number of Patients Enrolled18
Number of Patients Evaluable for Toxicity18
Number of Patients Evaluated for Efficacy16
Evaluation MethodRECIST 1.1
Response Assessment CRn = 1 (6%)
Response Assessment PRn = 1 (6%)
Response Assessment SDn = 2 (13%)
Response Assessment PDn = 12 (75%)
(Median) Duration Assessments PFS2.6 months, CI: 1.9–3.1
(Median) Duration Assessments OS25.5 months, CI: 10.4‐
(Median) Duration Assessments Duration of Treatment12 (6–105) weeks
Outcome Notes
CBR at 16 weeks was 4 of 16 (25%). Durable CR was found in one patient who continues to be progression‐free for over 32 months. The median follow‐up was 24.9 months (95% CI, 17.5–30.9). PFS was 2.6 months (95% CI, 1.9–3.1) and OS was 25.5 months (95% CI, 10.4–NR).
TitleResponse assessment
Number of Patients Screened22
Number of Patients Enrolled18
Number of Patients Evaluable for Toxicity18
Number of Patients Evaluated for Efficacy16
Evaluation MethodRECIST 1.1
Response Assessment CRn = 1 (6%)
Response Assessment PRn = 1 (6%)
Response Assessment SDn = 2 (13%)
Response Assessment PDn = 12 (75%)
(Median) Duration Assessments PFS2.6 months, CI: 1.9–3.1
(Median) Duration Assessments OS25.5 months, CI: 10.4‐
(Median) Duration Assessments Duration of Treatment12 (6–105) weeks
Outcome Notes
CBR at 16 weeks was 4 of 16 (25%). Durable CR was found in one patient who continues to be progression‐free for over 32 months. The median follow‐up was 24.9 months (95% CI, 17.5–30.9). PFS was 2.6 months (95% CI, 1.9–3.1) and OS was 25.5 months (95% CI, 10.4–NR).

Adverse Events

All Cycles
NameNC/NA12345All grades
Fatigue66%28%6%0%0%0%34%
Edema limbs88%6%6%0%0%0%12%
Pain88%0%6%6%0%0%12%
Headache94%0%6%0%0%0%6%
Arthralgia88%6%6%0%0%0%12%
Hot flashes94%0%6%0%0%0%6%
Hyperhidrosis83%11%6%0%0%0%17%
Dry skin94%0%0%6%0%0%6%
Diarrhea66%22%6%6%0%0%34%
Cough94%0%6%0%0%0%6%
Dyspnea88%6%6%0%0%0%12%
Palpitations94%0%6%0%0%0%6%
Hyperthyroidism94%0%6%0%0%0%6%
Aspartate aminotransferase increased77%17%6%0%0%0%23%
Adrenal insufficiency94%0%6%0%0%0%6%
Anemia94%0%6%0%0%0%6%
Cholesterol high94%0%6%0%0%0%6%
Hypophosphatemia94%0%6%0%0%0%6%
Hypoglycemia94%0%6%0%0%0%6%
Weight gain88%6%6%0%0%0%12%
Alanine aminotransferase increased83%6%11%0%0%0%17%
Edema face94%0%6%0%0%0%6%
Bone pain94%0%6%0%0%0%6%
Myalgia88%6%6%0%0%0%12%
All Cycles
NameNC/NA12345All grades
Fatigue66%28%6%0%0%0%34%
Edema limbs88%6%6%0%0%0%12%
Pain88%0%6%6%0%0%12%
Headache94%0%6%0%0%0%6%
Arthralgia88%6%6%0%0%0%12%
Hot flashes94%0%6%0%0%0%6%
Hyperhidrosis83%11%6%0%0%0%17%
Dry skin94%0%0%6%0%0%6%
Diarrhea66%22%6%6%0%0%34%
Cough94%0%6%0%0%0%6%
Dyspnea88%6%6%0%0%0%12%
Palpitations94%0%6%0%0%0%6%
Hyperthyroidism94%0%6%0%0%0%6%
Aspartate aminotransferase increased77%17%6%0%0%0%23%
Adrenal insufficiency94%0%6%0%0%0%6%
Anemia94%0%6%0%0%0%6%
Cholesterol high94%0%6%0%0%0%6%
Hypophosphatemia94%0%6%0%0%0%6%
Hypoglycemia94%0%6%0%0%0%6%
Weight gain88%6%6%0%0%0%12%
Alanine aminotransferase increased83%6%11%0%0%0%17%
Edema face94%0%6%0%0%0%6%
Bone pain94%0%6%0%0%0%6%
Myalgia88%6%6%0%0%0%12%

See also Table 2.

Abbreviation: NC/NA, no change from baseline/no adverse event.

Assessment, Analysis, and Discussion

CompletionStudy terminated before completion
Terminated ReasonCompany stopped development
Investigator's AssessmentThe trial was stopped early because of the withdrawal of GTx‐024 drug supply. Full assessment of activity of this combination could not be completed as planned.
CompletionStudy terminated before completion
Terminated ReasonCompany stopped development
Investigator's AssessmentThe trial was stopped early because of the withdrawal of GTx‐024 drug supply. Full assessment of activity of this combination could not be completed as planned.

Triple‐negative breast cancer (TNBC) is defined by lack of hormone receptor (HR) expression and human epidermal growth factor receptor 2 (HER2) overexpression and accounts for 15% of all breast cancer. This is an aggressive type of breast cancer with poor prognosis in the metastatic setting. TNBC is molecularly heterogeneous, with at least four distinct TNBC subtypes identified through genomic profiling, including luminal androgen receptor (LAR), mesenchymal, basal‐like immunosuppressed, and basal‐like immune‐activated [9, 10]. Recent U.S. Food and Drug Administration (FDA)–approved targeted therapies make precision medicine feasible in the treatment of TNBC. These include PARP inhibitors for BRCA mutated tumors [11, 12], immune checkpoint inhibitors for programmed death ligand‐1 (PD‐L1)–positive TNBC [13], and antibody drug conjugate targeting Trop‐2 receptor [14]. Despite these advances, treatment choice for metastatic androgen receptor (AR) + TNBC remain limited. The LAR subtype accounts for approximately 10%–15% of TNBC, and LAR cell lines have shown sensitivity to AR antagonist [10, 15]. There is strong interest in the use of AR as a potential therapeutic target.

Expression of AR has been observed in 70%–80% of all breast cancer [1618], and HR‐positive (+) breast cancer exhibits significantly higher AR expression than HR‐negative (−) breast cancer [17, 18]. Moreover, AR expression seems to be associated with favorable clinical outcome in estrogen receptor (ER) + breast cancer [19, 20]. In TNBC, 10%–40% of tumors express AR across several studies, with immunohistochemistry (IHC) cutoff values ranging between 1% and 10% [18, 21, 22]. The prognostic value and precise mechanism of AR in TNBC remain controversial [2123]. In preclinical studies, androgen induces migration and invasiveness in AR+ TNBC cell lines, whereas the AR antagonists bicalutamide and enzalutamide inhibit growth of TNBC cell lines in vitro [24, 25]. Gucalp et al. reported the efficacy of bicalutamide in patients with AR+ metastatic TNBC (mTNBC) [1]. The clinical benefit rate (CBR) of 19% observed with bicalutamide shows proof of concept for the efficacy of androgen blockade in AR+ (≥10% IHC) TNBC. The efficacy of enzalutamide has been studied in patients with AR+ mTNBC with a CBR 25% or 33% in AR with ≥1% or ≥ 10% by IHC, respectively, at 16 weeks [26]. Similarly, the 17α‐Hydroxylase/C17,20‐lyase (CYP17) inhibitor abiraterone showed modest clinical activity in AR+ (≥10% IHC) mTNBC with a response rate (RR) of 6.7%, CBR of 20%, and progression‐free survival (PFS) of 2.8 months [27]. The regimen was well tolerated, with grade 1–2 fatigue, hypertension, hypokalemia, and nausea being the most common adverse events (AE). Despite these encouraging results, efficacy of these single‐agent AR‐targeted therapies appears to be modest, and more effective combination therapies are needed.

Enobosarm (GTx‐024) is an oral nonsteroidal, tissue‐selective, AR modulator that was initially developed for anabolic activity with minimal androgenic activity [24]. Preclinical study demonstrated that enobosarm inhibited proliferation and tumor growth of AR+ TNBC [5]. Enobosarm provides a novel targeted approach to exploit the therapeutic benefits of AR modulation without virilization or estrogenic effects. In a proof‐of‐concept phase II study evaluating the safety and efficacy of 9 mg daily dosing of enobosarm in patients with AR+ HR+ metastatic breast cancer, the drug was well tolerated, and 8 of 22 (36%) patients had stable disease (SD). Pembrolizumab is a highly selective humanized monoclonal antibody of programmed death receptor (PD‐1) and has shown promising antitumor effects in solid tumors. Single‐agent pembrolizumab showed an RR of 18.5% and a median response of 17.9 weeks in KEYNOTE‐12, which enrolled 27 patients with PD‐L1+ mTNBC [6]. In PD‐L1 nonselected heavily pretreated patients with mTNBC, the RR was only 5.3% in the KEYNOTE‐86 cohort A (n = 170) [7]. Preclinical evidence has suggested a potential therapeutic synergy by combining AR modulation and PD‐1 blockade. Androgens are immunosuppressive and target both innate and adaptive immune systems to dampen the immune response [2834]. Inhibition of AR sensitizes cancer cells to immune‐mediated killing. AR suppresses PD‐L1 expression by binding to the PD‐L1 promotor and directly attenuating PD‐L1 gene transcription [29, 30]. In addition, AR signaling inhibits thymocyte production [31], whereas AR blockade has been shown to increase naive T cell generation resulting in a larger diversity of T cell repertoire that could potentially respond to PD‐1 blockade [32, 33]. Moreover, AR blockade yielded increased survival benefits to vaccination in a murine prostate cancer model [34].

The complementary modes of action and low potential of overlapping toxicities of these two modalities make the combination therapy of AR‐targeted agent and immune checkpoint inhibitor an attractive option for AR+ TNBC. A proof‐of‐concept KEYNOTE‐199 trial showed that pembrolizumab plus enzalutamide demonstrated a RR of 12% and disease control rate of 51% for patients with metastatic castration‐resistant prostate cancer. We hypothesized that enobosarm could enhance pembrolizumab‐induced immune response while targeting AR. The current study was designed to assess the safety and efficacy of enobosarm plus pembrolizumab in patients with AR+ mTNBC.

A total of 18 patients were enrolled from April 2017 to April 2019. In April 2019, GTx Inc. notified the study team of withdrawal of enobosarm drug supply, and further enrollment was halted. Two patients were ineligible because of undiagnosed brain metastasis. Patient characteristics and treatment variables are described in Table 1 (n = 16). Median age was 64 (range, 36–81), with 50% non‐Hispanic white, 31% Hispanic, 13% Asian, and 6% African American. A total of 56% received 0–1 previous lines of therapy, and 44% received ≥ 2 prior lines of therapy.

The combination of pembrolizumab and enobosarm was well tolerated. Median dose delay and dose modification were both 0 (range, 0–2). Five of 16 (31%) had more than one dose delay, and 3 of 16 (19%) had more than one dose reduction. Causes of dose delay and/or dose reduction were grade 2 elevated liver function test (LFT); hypoglycemia; fever, nausea, or diarrhea; elective tissue expander removal; and patient's choice.

There were no grade ≥ 4 AEs observed in the study. Grade 1–3 AEs are shown in Table 2. Grade 3 AEs were one musculoskeletal ache (pain), one dry skin, and one diarrhea. AEs associated with potential pembrolizumab‐induced immune side effects were consistent with known side effects per FDA package insert.

Among 16 patients evaluable for response, 1 of 16 (6%) achieved complete response (CR), 1 of 16 (6%) achieved partial response (PR), 2 of 16 (13%) had SD, and 12 of 16 (75%) had progressive disease. Response rate (RR) was 2 of 16 (13%) (Fig. 1). CBR at 16 weeks was 4 of 16 (25%). Durable CR was found in one patient who continues to be progression‐free for over 32 months (Fig. 2). The median follow‐up was 24.9 months (95% confidence interval [CI], 17.5–30.9). PFS was 2.6 months (95% CI, 1.9–3.1), and overall survival (OS) was 25.5 months (95% CI, 10.4–NR; Fig. 3).

Stromal tumor infiltrating lymphocytes (TILs), tumor mutation burden (TMB), and PD‐L1 expression were evaluated in pretreatment tumor biopsies as potential biomarkers of response. Stromal tumor infiltration levels ranged from 2% to 12.5% and showed no correlation with PFS or response (Fig. 4A). TMB was low in all patients, ranging from 0–4.6 mutations per megabase (m/MB) (Fig. 4B).

Gene expression patterns in tumor tissues were assessed by whole transcriptome analysis (n = 16) and showed clustering of the top 20 variants for responders versus nonresponders (Fig. 5A). Although principle component analysis identified two distinct subgroups of patients, no noticeable differences in response to therapy or time point in therapy were found (Fig. 5B).

We next quantified potential differences in the activity of the androgen‐related pathway. The gene expression and signature score of the androgen‐related pathway were calculated and compared with a clinically relevant AR signature [39]. No significant differences in the AR signature score between responders and nonresponders were identified (Fig. 6).

CIBERSORTx with merged LM22 signatures were used to deconvolute RNA expression profiles to overall immune infiltration levels and the relative abundances of 18 immune cell subtypes. No significant difference in overall immune infiltration levels (absolute immune score) were found (p = .10) (Fig. 7A). Of note, the absolute abundance of naive B cells (p = .028) were observed to be significantly higher in responding patient tumors compared with nonresponding patient tumors (Fig. 7B).

Peripheral blood mononuclear cells were collected from patients at pretreatment, immediately prior to cycle 2, and immediately prior to cycle 4 for evaluation of immune phenotypes by flow cytometry. At pretreatment, there were no differences between responding and nonresponding patients in memory T cell composition in either CD4+ T cells or CD8+ T cells (Fig. 8). Similarly, we found no differences at pretreatment between responding and nonresponding patients in CD4+ T cell or CD8+ T cell expression of PD‐1, TIGIT, TIM‐3, KLRG1, or CD137 or frequencies of regulatory T cells or follicular helper T cells. We also found no differences in CD8+ T cell expression of PD‐1, TIGIT, TIM‐3, KLRG1, and CD137. To assess the potential impact of treatment on T cell phenotypes, we calculated fold change in expression percentages relative to pretreatment for cycle 2 and cycle 4 collected samples. No significant differences between responder and nonresponder patients were found over the course of therapy in CD4+ T cell or CD8+ T cell subsets. We do note that the patient with CR was observed to have a relatively high expression of TIGIT on CD4+ T cells at pretreatment that increased over the course of therapy. Similarly, the patient with CR was observed to have relatively high fold change in increased CD137 expression on both CD4+ and CD8+ T cells. Together, these data suggest that both pretreatment and on‐therapy peripheral T cell activation markers may be useful in monitoring patient responses to immune checkpoint inhibitors (ICIs).

Genomic alterations in responders versus nonresponders were analyzed, and the gene alterations were reported (Fig. 9). A total of 9 of 13 (69%) patients had a TP53 mutation, and 2 of 13 (15%) had EGFR, NF1, PIK3R1, and/or PTEN alterations. Results from this analysis were consistent with published breast cancer profiles [9, 10].

Our final exploratory analysis evaluated the microbiome. Patient samples classified by MetaPlAn2 were examined [35] by Bray‐Curtis hierarchical clustering (Fig. 10A). One cluster (green) consists of samples from a patient 11 and was notable for a marked lack of diversity. This patient's microbiome is dominated by Escherichia coli, and patient had PFS of 1 month. Two clusters (gold and black) contained stable disease and responders (patient 4 had CR and patient 8 had PR) with high levels of some Bacteroides spp. and Alistipes spp., indicating a potentially healthier gut microbiome. Alpha diversity by patient is relatively stable for most (Fig. 10B). Patient 9 had high diversity at baseline that decreased at cycle 5 (patient had PFS of 2 months). In contrast, patient 11 had increased diversity at cycle 5. Significant organisms determined by PFS (< 6month vs. >6 month) is shown in Figure 10C. There was no clear correlation between diversity and response or PFS.

The trial was stopped early because of GTx's withdraw of drug supply, with a total of 16 evaluable patients included in the analysis.

In the current study, we observed an RR of 12% and CBR of 25% with pembrolizumab plus enobosarm in heavily pretreated patients without PD‐L1 preselection. One patient showed complete response and remains in remission at the time of this manuscript submission. This study was originally designed to test the hypothesis that enobosarm plus pembrolizumab would lead to an RR of 29% with 29 patients. With the early termination of the accrual, we did not meet this aim. Although from a strict statistical point of view, these data do not allow a rejection of the null hypothesis, from a clinical perspective, our results remain meaningful. The modest response rate is comparable with the single‐agent pembrolizumab RR of 12% observed in KEYNOTE‐12 [6] yet was numerically higher than RR of 5% observed in KEYNOTE‐86 [7]. Within the bicalutamide study, a CBR of 19% at 6 months and no PR was reported. In the enzalutamide trial, CBR at 4 months was 35% and median PFS was 14 weeks. A total of 2 of 118 (1.7%) CR and 5 of 118 (4.2%) PR were observed, which translates to 5.9% RR. Early literature supported synergy of AR‐targeted therapy with ICI. Concurrent expression of PD‐L1 and AR in TNBC showed potential to apply combination treatment of AR inhibitor and ICIs [36]. In addition, preclinical studies demonstrated that blockade of AR stimulates thymic volume and maturation of naive T cell clones, which can facilitate antitumor immune responses and induce synergic effect with ICI [32, 37]. Currently, a phase II trial studying dual immune checkpoint blockade (nivolumab plus ipilimumab) plus bicalutamide is ongoing in patients with metastatic AR+ HER2− breast cancer (BC) and TNBC [33]. Our modest result could also reflect modest efficacy of enobosarm as an AR‐targeted agent.

AR is a highly expressed steroid receptor in breast cancer, with 75–95% of ER+ and 50% of ER− BCs expressing AR [38]. AR is an intracellular steroid hormonal receptor of the nuclear receptor family along with ER, PR, glucocorticoid, and mineralocorticoid receptors. It links a transcription factor that regulates specific genes involved in cellular processes with a diverse range of actions such as stimulating both cell proliferation or apoptosis, depending on the concurrent activated signaling pathways [39, 40]. The role of AR in breast cancer is complicated and varies in different subtypes. It was observed that ligand‐bound AR binds to an estrogen‐related element in the nucleus leading to cell apoptosis in ER+ AR+ cell lines. In contrast, AR binds to an androgen‐related element in the nucleus, leading to cell proliferation in ER negative AR+ cell lines [41, 42]. In TNBC, the AR pathway shows cross‐talk with several other key signaling pathways, including the PI3K/AKT/mTOR pathway [4345]. AR status, by itself, may not be a reliable prognostic marker and it varies across different race and ethnicity [46].

Despite the enthusiasm for targeting AR for LAR TNBC, single‐agent AR‐targeted therapies have been associated with modest response [26, 27]. Combination of AR‐targeted therapy with other cancer driver pathway inhibitors have been investigated. LAR tumors have focal gains twice as frequently on 11q13.3 (CCND1, fibroblast growth factor family) and 14q21.3 (MDGA2) [9]. Lehmann et al. reported that PIK3CA mutations were highly clonal and more frequent in AR + versus AR− TNBC (40% vs. 4%) and often associated with concurrent amplification of the PIK3CA locus. The Pan‐PI3K inhibitor GDC‐0941 or the dual PI3K/mTOR inhibitor GDC‐0980 had an additive growth inhibitory effect when combined with genetic or pharmacological AR targeting with bicalutamide in AR + TNBCs in vitro. A phase II trial combining the α‐specific PIK3CA inhibitor alpelisib with enzalutamide in patients with AR+/PTEN loss metastatic breast cancer is ongoing (NCT03207529). In addition, phase II trials combining CDK 4/6 inhibitors ribociclib (NCT03090165) or palbociclib (NCT02605486) with bicalutamide are actively being conducted [47, 48] in AR+ metastatic breast cancer, and results are eagerly awaited.

Immune biomarkers to predict responses have been extensively studied for personalized treatment. We analyzed TILs [49], TMB [50], PD‐L1 expression(IHC 22C3 pharmDx (Dako, Inc.) at QualTek Molecular Laboratory), and subsets of T cells in the study. Whereas breast cancer has low TMB compared with other immunogenic cancers, such as melanoma and lung cancer that have shown good responses to PD‐1 inhibitors [51], TNBC has relatively higher TMB than other types of breast cancer [52]. In our study, TMB was generally low in all patients, ranging from 0–4.6 m/MB, whereas the patient with CR had the highest TMB. TILs were not associated with treatment responses, and PD‐L1 positivity was observed in two patients, including one who achieved CR. There were no significant differences in CD4+ T cell nor CD8+ T cell subsets between responder and nonresponder patients.

In our exploratory CIBERSORTx analysis [53, 54, 55], M2 macrophages and mast cells were more enriched in the tumors of nonresponders, which is consistent with the immune suppressive roles of M2 macrophages and mast cells in cancer. M2 macrophages inhibit inflammation and promote tissue remodeling and angiogenesis [56]. In preclinical studies, mast cell‐secreted angiogenic cytokines directly facilitate tumor vascularization and thereby stimulate other inflammatory cells of the tumor microenvironment to release other angiogenic mediators. Hence, increased numbers of mast cells have been associated with angiogenesis in breast cancer [57].

The relation between the human microbiomes and cancer has been explored recently. Although many questions still remain unanswered, emerging evidence from various studies show that the presence of specific microbiomes play a role in breast cancer tumorigenesis [58]. In TNBC, one study that analyzed stool microbiomes using 16S rRNA amplicon sequencing identified that microbial signatures (Bacteroides and Ruminococcaceae) were associated with treatment response to chemotherapy [59]. Several other studies showed that microbiome may affect the response to ICI in solid tumors [60, 61]. In metastatic melanoma, baseline gut microbiomes were associated with clinical responses to CTLA‐4 inhibitor or PD‐1 inhibitor [62, 63]. We analyzed gut microbiomes of patients using whole metagenome sequencing and classification by MetaPhlAn2. One patient with a lack of diversity on the microbiomes had a very short PFS, but there was no clear correlation between diversity and response when compared responders with nonresponders. Alpha diversity by patient was relatively stable for most patients. Several organisms of potential interest associated with patients with increased PFS were identified by LEfSe. Further study is warranted.

Acknowledgments

Merck Sharp & Dohme Corp., a subsidiary of Merck & Co., Inc., Kenilworth, NJ, USA provided drug, and financial support for the study. We thank GTx, Inc., for providing the study drug enobosarm. We also thank the STOP Cancer Foundation (principal investigator [PI] Yuan Yuan), phase I Foundation (PI Yuan Yuan), NCI K‐12 Career Development Award (K12CA001727, PI Joanne Mortimer), and the National Institutes of Health (P30CA033572). We thank the patients who participated this study. The content of this article is solely the responsibility of the authors and does not necessarily represent the official views of the National Cancer Institute.

Disclosures

Yuan Yuan: Puma, Pfizer, Immunomedics (C/A), Merck, Eisai, Novartis, Puma, Genentech, Celgene and Pfizer (RF), Eisai, Novartis, Genentech, AstraZeneca, Daiichi Sankyo, Immunomedics (SAB); Mina Sedrak: Eli Lilly, Seattle Genetics, Novartis, Pfizer (RF‐institutional). The other authors indicated no financial relationships.

(C/A) Consulting/advisory relationship; (RF) Research funding; (E) Employment; (ET) Expert testimony; (H) Honoraria received; (OI) Ownership interests; (IP) Intellectual property rights/inventor/patent holder; (SAB) Scientific advisory board.

Figures and Tables

CT imaging of exceptional response. (A): Baseline CT on July 24, 2017, showed subcarinal and right hilar conglomerate lymph adenopathy (the red arrows pointing the target lesions). (B): The bulky subcarinal adenopathy and right hilar is no longer seen on August 24, 2018. Patient continues to have no evidence of disease as of July 2020.
FIGURE 2

CT imaging of exceptional response. (A): Baseline CT on July 24, 2017, showed subcarinal and right hilar conglomerate lymph adenopathy (the red arrows pointing the target lesions). (B): The bulky subcarinal adenopathy and right hilar is no longer seen on August 24, 2018. Patient continues to have no evidence of disease as of July 2020.

Abbreviation: CT, computed tomography.
Kaplan‐Meier survival analysis. (A): Median PFS was 2.6 months (95% CI, 1.9.–3.1). (B): Median OS was 25.5 months (95% CI, 10.4–NR).
FIGURE 3

Kaplan‐Meier survival analysis. (A): Median PFS was 2.6 months (95% CI, 1.9.–3.1). (B): Median OS was 25.5 months (95% CI, 10.4–NR).

Abbreviations: CI, confidence interval; OS, overall survival; NR, not reached; PFS, progression‐free survival.
Tumor biomarkers. Established biomarkers of response to immune checkpoint blockade were examined in pretreatment tumors. (A): Stromal TILs were evaluated by pathologists at City of Hope (n = 13). (B): Tumor mutation burden, depicted as m/MB, was evaluated by the TEMPUS xT genome alteration panel (n = 14). Statistics were assessed by two‐tailed Mann‐Whitney test. For both graphs, the median is represented by a line. Nonresponders (progressive disease + stable disease) in red, responders (complete response + partial response) in blue.
FIGURE 4

Tumor biomarkers. Established biomarkers of response to immune checkpoint blockade were examined in pretreatment tumors. (A): Stromal TILs were evaluated by pathologists at City of Hope (n = 13). (B): Tumor mutation burden, depicted as m/MB, was evaluated by the TEMPUS xT genome alteration panel (n = 14). Statistics were assessed by two‐tailed Mann‐Whitney test. For both graphs, the median is represented by a line. Nonresponders (progressive disease + stable disease) in red, responders (complete response + partial response) in blue.

Abbreviations: m/MB, mutations per megabase; NR, nonresponders; R, responders; TIL, tumor infiltrating lymphocyte.
Principal component analysis (PCA) of whole transcriptome gene expression in tumor tissues. (A): Heat map showing scaled expressions of top 10 genes with significantly differential expressions (false discovery rate ≤ 0.10) for responders versus nonresponders . (B): PCA diagram with type of specimen and response to treatment.
FIGURE 5

Principal component analysis (PCA) of whole transcriptome gene expression in tumor tissues. (A): Heat map showing scaled expressions of top 10 genes with significantly differential expressions (false discovery rate ≤ 0.10) for responders versus nonresponders . (B): PCA diagram with type of specimen and response to treatment.

Abbreviations: CR, complete response; LN, lymph node; NR, nonresponders; PC1, principle component 1; PC2, principle component 2; PD, progressive disease; PR, partial response; Pre‐Tx, pretreatment; R, responders; SD, stable disease.
Androgen receptor signaling pathway. Baseline tumor biopsies were assessed for gene differential gene expression of a defined AR signature. (A): Heatmap of gene expression of AR signature. Row orders are based on hierarchical clustering. Column orders are based on the specific response groups. (B): AR signature scores between responder and nonresponder groups. Statistics assessed by two‐tailed Mann‐Whitney test. Nonresponders (PD + SD) in red, responders (CR + PR) in blue.
FIGURE 6

Androgen receptor signaling pathway. Baseline tumor biopsies were assessed for gene differential gene expression of a defined AR signature. (A): Heatmap of gene expression of AR signature. Row orders are based on hierarchical clustering. Column orders are based on the specific response groups. (B): AR signature scores between responder and nonresponder groups. Statistics assessed by two‐tailed Mann‐Whitney test. Nonresponders (PD + SD) in red, responders (CR + PR) in blue.

Abbreviations: AR, androgen receptor; CR, complete response; LN, lymph node; NR, nonresponders; PD, progressive disease; PR, partial response; R, responders; SD, stable disease.
CIBERSORT analysis of immune subsets. (A): Overall immune infiltration levels between responders and nonresponders. (B): Relative abundance of 18 immune cell types using CIBERSORTx with LM22 signature. The p values from two‐tailed Mann‐Whitney Nonresponders (progressive disease + stable disease) in red, responders (complete response + partial response) in blue.
FIGURE 7

CIBERSORT analysis of immune subsets. (A): Overall immune infiltration levels between responders and nonresponders. (B): Relative abundance of 18 immune cell types using CIBERSORTx with LM22 signature. The p values from two‐tailed Mann‐Whitney Nonresponders (progressive disease + stable disease) in red, responders (complete response + partial response) in blue.

Abbreviations: NR, nonresponders; R, responders.
Peripheral immune characteristics. Peripheral blood mononuclear cells were evaluated by flow cytometry for immune phenotype distributions. (A): CD4+ T cells and (B) CD8+ T cells were evaluated for distribution of naive (CD45RA+ CCR7+), CM (CD45RA− CCR7+), EM (CD45RA− CCR7−), and EMRA (CD45RA+ CCR7−) subsets in pretreatment samples (n = 15). Within (C) non‐naive CD4+ T cells and (D) non‐naive CD8+ T cells, expression of molecules PD‐1, TIGIT, TIM‐3, KLRG1, and CD137 were evaluated in addition to fractions of Tregs (CD25+ CD127−) and Tfh (PD‐1+, CXCR5+) at pretreatment (n = 15). Fold change in expression of depicted molecules and cell subsets relative to pretreatment is shown for (E) CD4+ T cells, cycle 2; (F) CD8+ T cells, cycle 2; (G) CD4+ T cells, cycle 4; (H) CD8+ T cells cycle 4 (cycle 2, n = 15; cycle 4, n = 11). For all graphs, the median is represented by a line. Nonresponders (progressive disease + stable disease) in red, responders (complete response + partial response) in blue.
FIGURE 8

Peripheral immune characteristics. Peripheral blood mononuclear cells were evaluated by flow cytometry for immune phenotype distributions. (A): CD4+ T cells and (B) CD8+ T cells were evaluated for distribution of naive (CD45RA+ CCR7+), CM (CD45RA− CCR7+), EM (CD45RA− CCR7−), and EMRA (CD45RA+ CCR7−) subsets in pretreatment samples (n = 15). Within (C) non‐naive CD4+ T cells and (D) non‐naive CD8+ T cells, expression of molecules PD‐1, TIGIT, TIM‐3, KLRG1, and CD137 were evaluated in addition to fractions of Tregs (CD25+ CD127−) and Tfh (PD‐1+, CXCR5+) at pretreatment (n = 15). Fold change in expression of depicted molecules and cell subsets relative to pretreatment is shown for (E) CD4+ T cells, cycle 2; (F) CD8+ T cells, cycle 2; (G) CD4+ T cells, cycle 4; (H) CD8+ T cells cycle 4 (cycle 2, n = 15; cycle 4, n = 11). For all graphs, the median is represented by a line. Nonresponders (progressive disease + stable disease) in red, responders (complete response + partial response) in blue.

Abbreviations: CM, central memory; EM, effector memory; EMRA, effector memory RA+; NR, nonresponder; R, responder; Tfh, follicular helper CD4+ T cell; Treg, regulatory CD4+ T cell.
Tumor genomic mutations (n = 13). Exome sequencing was performed for pretreatment specimens using Tempus (n = 12) and Foundation Medicine (n = 1).
FIGURE 9

Tumor genomic mutations (n = 13). Exome sequencing was performed for pretreatment specimens using Tempus (n = 12) and Foundation Medicine (n = 1).

Abbreviations: CR, complete response; LN, lymph node; PD, progressive disease; PFS, progressive‐free survival; PR, partial response; Pre‐Tx, pretreatment; SD, stable disease.
Gut Microbiome Analysis. (A): Hierarchical clustering of species classified by MetaPhlAn2 by patient. (B): Shannon diversity by patient and day of collection. (C): Significant organisms determined by linear discriminant analysis effect size per survival outcomes (blue, progression‐free survival [PFS] >6 months; red, PFS <6 months).
FIGURE 10

Gut Microbiome Analysis. (A): Hierarchical clustering of species classified by MetaPhlAn2 by patient. (B): Shannon diversity by patient and day of collection. (C): Significant organisms determined by linear discriminant analysis effect size per survival outcomes (blue, progression‐free survival [PFS] >6 months; red, PFS <6 months).

Abbreviations: CR, complete response; LDA, linear discriminant analysis; PD, progressive disease; PR, partial response; SD, stable disease.
Table 1

Patient characteristics

CharacteristicPatients (n = 16)
Median age (range), yr64 (36‐81)
Race and ethnicity, n (%)
Non‐Hispanic White8 (50)
Hispanic5 (31)
Asian2 (13)
African American1 (6)
Performance status (ECOG), n (%)
07 (44)
19 (56)
Histological grade, n (%)
12 (13)
25 (31)
39 (56)
Initial tumor stage, n (%)
I6 (38)
II8 (50)
III2 (13)
Median prior lines of therapy for MBC, n (%)1 (0‐9)
03 (18)
16 (38)
≥27 (44)
Prior surgery, n (%)
Lumpectomy7 (44)
Mastectomy9 (56)
Prior radiation, n (%)
Yes15 (94)
No1 (6)
CharacteristicPatients (n = 16)
Median age (range), yr64 (36‐81)
Race and ethnicity, n (%)
Non‐Hispanic White8 (50)
Hispanic5 (31)
Asian2 (13)
African American1 (6)
Performance status (ECOG), n (%)
07 (44)
19 (56)
Histological grade, n (%)
12 (13)
25 (31)
39 (56)
Initial tumor stage, n (%)
I6 (38)
II8 (50)
III2 (13)
Median prior lines of therapy for MBC, n (%)1 (0‐9)
03 (18)
16 (38)
≥27 (44)
Prior surgery, n (%)
Lumpectomy7 (44)
Mastectomy9 (56)
Prior radiation, n (%)
Yes15 (94)
No1 (6)

Abbreviations: ECOG, Eastern Cooperative Oncology Group; MBC, metastatic breast cancer.

Table 1

Patient characteristics

CharacteristicPatients (n = 16)
Median age (range), yr64 (36‐81)
Race and ethnicity, n (%)
Non‐Hispanic White8 (50)
Hispanic5 (31)
Asian2 (13)
African American1 (6)
Performance status (ECOG), n (%)
07 (44)
19 (56)
Histological grade, n (%)
12 (13)
25 (31)
39 (56)
Initial tumor stage, n (%)
I6 (38)
II8 (50)
III2 (13)
Median prior lines of therapy for MBC, n (%)1 (0‐9)
03 (18)
16 (38)
≥27 (44)
Prior surgery, n (%)
Lumpectomy7 (44)
Mastectomy9 (56)
Prior radiation, n (%)
Yes15 (94)
No1 (6)
CharacteristicPatients (n = 16)
Median age (range), yr64 (36‐81)
Race and ethnicity, n (%)
Non‐Hispanic White8 (50)
Hispanic5 (31)
Asian2 (13)
African American1 (6)
Performance status (ECOG), n (%)
07 (44)
19 (56)
Histological grade, n (%)
12 (13)
25 (31)
39 (56)
Initial tumor stage, n (%)
I6 (38)
II8 (50)
III2 (13)
Median prior lines of therapy for MBC, n (%)1 (0‐9)
03 (18)
16 (38)
≥27 (44)
Prior surgery, n (%)
Lumpectomy7 (44)
Mastectomy9 (56)
Prior radiation, n (%)
Yes15 (94)
No1 (6)

Abbreviations: ECOG, Eastern Cooperative Oncology Group; MBC, metastatic breast cancer.

Table 2

Treatment‐related grade 1–3 adverse events per CTCAE 4.0 (n = 18)

Adverse eventGrade 2, n (%)Grade 3, n (%)
Fatigue1 (6)
Edema2 (12)
Pain3 (18)1 (6)
Headache1 (6)
Arthralgia1 (6)
Hot flashes1 (6)
Hyperhidrosis1 (6)
Dry skin1 (6)
Diarrhea1 (6)1 (6)
Cough1 (6)
Dyspnea1 (6)
Palpitations1 (6)
Hyperthyroidism1 (6)
Elevated LFTs3 (17)
Adrenal insufficiency1 (6)
Anemia1 (6)
Hypercholesterolemia1 (6)
Hypophosphatemia1 (6)
Hypoglycemia1 (6)
Adverse eventGrade 2, n (%)Grade 3, n (%)
Fatigue1 (6)
Edema2 (12)
Pain3 (18)1 (6)
Headache1 (6)
Arthralgia1 (6)
Hot flashes1 (6)
Hyperhidrosis1 (6)
Dry skin1 (6)
Diarrhea1 (6)1 (6)
Cough1 (6)
Dyspnea1 (6)
Palpitations1 (6)
Hyperthyroidism1 (6)
Elevated LFTs3 (17)
Adrenal insufficiency1 (6)
Anemia1 (6)
Hypercholesterolemia1 (6)
Hypophosphatemia1 (6)
Hypoglycemia1 (6)

Abbreviation: LFT, liver function test.

Table 2

Treatment‐related grade 1–3 adverse events per CTCAE 4.0 (n = 18)

Adverse eventGrade 2, n (%)Grade 3, n (%)
Fatigue1 (6)
Edema2 (12)
Pain3 (18)1 (6)
Headache1 (6)
Arthralgia1 (6)
Hot flashes1 (6)
Hyperhidrosis1 (6)
Dry skin1 (6)
Diarrhea1 (6)1 (6)
Cough1 (6)
Dyspnea1 (6)
Palpitations1 (6)
Hyperthyroidism1 (6)
Elevated LFTs3 (17)
Adrenal insufficiency1 (6)
Anemia1 (6)
Hypercholesterolemia1 (6)
Hypophosphatemia1 (6)
Hypoglycemia1 (6)
Adverse eventGrade 2, n (%)Grade 3, n (%)
Fatigue1 (6)
Edema2 (12)
Pain3 (18)1 (6)
Headache1 (6)
Arthralgia1 (6)
Hot flashes1 (6)
Hyperhidrosis1 (6)
Dry skin1 (6)
Diarrhea1 (6)1 (6)
Cough1 (6)
Dyspnea1 (6)
Palpitations1 (6)
Hyperthyroidism1 (6)
Elevated LFTs3 (17)
Adrenal insufficiency1 (6)
Anemia1 (6)
Hypercholesterolemia1 (6)
Hypophosphatemia1 (6)
Hypoglycemia1 (6)

Abbreviation: LFT, liver function test.

  • ClinicalTrials.gov  Identifier:  NCT02971761

  • Sponsors: City of Hope Comprehensive Cancer Center, Merck, GTx, Inc.

  • Principal Investigator: Yuan Yuan

  • IRB Approved: Yes

References

1

Gucalp
 
A
,
Tolaney
 
S
,
Isakoff
 
SJ
et al.
Phase II trial of bicalutamide in patients with androgen receptor‐positive, estrogen receptor‐negative metastatic Breast Cancer
.
Clin Cancer Res
 
2013
;
19
:
5505
5512
.

2

Yin
 
D
,
Gao
 
W
,
Kearbey
 
JD
et al.
Pharmacodynamics of selective androgen receptor modulators
.
J Pharmacol Exp Ther
 
2003
;
304
:
1334
1340
.

3

Yin
 
D
,
He
 
Y
,
Perera
 
MA
et al.
Key structural features of nonsteroidal ligands for binding and activation of the androgen receptor
.
Mol Pharmacol
 
2003
;
63
:
211
223
.

4

Srinath
 
R
,
Dobs
 
A
.
Enobosarm (GTx‐024, S‐22): A potential treatment for cachexia
.
Future Oncol
 
2014
;
10
:
187
194
.

5

Narayanan
 
R
,
Ahn
 
S
,
Cheney
 
MD
et al.
Selective androgen receptor modulators (SARMs) negatively regulate triple‐negative breast cancer growth and epithelial:mesenchymal stem cell signaling
.
PLoS One
 
2014
;
9
:
e103202
.

6

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
 
2006
;
34
:
2460
2467
.

7

Adams
 
S
,
Schmid
 
P
,
Rugo
 
HS
et al.
Pembrolizumab monotherapy for previously treated metastatic triple‐negative breast cancer: Cohort A of the phase II KEYNOTE‐086 study
.
Ann Oncol
 
2019
;
30
:
397
404
.

8

Frankel
 
PH
,
Chung
 
V
,
Tuscano
 
J
et al.
Model of a queuing approach for patient accrual in phase 1 oncology studies
.
JAMA Netw Open
 
2020
;
3
:
e204787
.

9

Burstein
 
MD
,
Tsimelzon
 
A
,
Poage
 
GM
et al.
Comprehensive genomic analysis identifies novel subtypes and targets of triple‐negative breast cancer
.
Clin Cancer Res
 
2015
;
21
:
1688
1698
.

10

Lehmann
 
BD
,
Jovanović
 
B
,
Chen
 
X
et al.
Refinement of triple‐negative breast cancer molecular subtypes: Implications for neoadjuvant chemotherapy selection
.
PLoS One
 
2016
;
11
:
e0157368
.

11

Litton
 
JK
,
Rugo
 
HS
,
Ettl
 
J
et al.
Talazoparib in patients with advanced breast cancer and a germline BRCA mutation
.
N Engl J Med
 
2018
;
379
:
753
763
.

12

Robson
 
M
,
Im
 
S‐A
,
Senkus
 
E
et al.
Olaparib for metastatic breast cancer in patients with a germline BRCA mutation
.
N Engl J Med
 
2017
;
377
:
523
533
.

13

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
 
2020
;
21
:
44
59
.

14

Bardia
 
A
,
Mayer
 
IA
,
Vahdat
 
LT
et al.
Sacituzumab govitecan‐hziy in refractory metastatic triple‐negative breast cancer
.
N Engl J Med
 
2019
;
380
:
741
751
.

15

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
 
2011
;
121
:
2750
2767
.

16

Gonzalez
 
LO
,
Corte
 
MD
,
Vazquez
 
J
et al.
Androgen receptor expresion in breast cancer: relationship with clinicopathological characteristics of the tumors, prognosis, and expression of metalloproteases and their inhibitors
.
BMC Cancer
 
2008
;
8
:
149
.

17

Collins
 
LC
,
Cole
 
KS
,
Marotti
 
JD
et al.
Androgen receptor expression in breast cancer in relation to molecular phenotype: Results from the Nurses' Health Study
.
Mod Pathol
 
2011
;
24
:
924
931
.

18

Niemeier
 
LA
,
Dabbs
 
DJ
,
Beriwal
 
S
et al.
Androgen receptor in breast cancer: Expression in estrogen receptor‐positive tumors and in estrogen receptor‐negative tumors with apocrine differentiation
.
Mod Pathol
 
2009
;
23
:
205
212
.

19

Castellano
 
I
,
Allia
 
E
,
Accortanzo
 
V
et al.
Androgen receptor expression is a significant prognostic factor in estrogen receptor positive breast cancers
.
Breast Cancer Res Treat
 
2010
;
124
:
607
617
.

20

Hu
 
R
,
Dawood
 
S
,
Holmes
 
MD
et al.
Androgen receptor expression and breast cancer survival in postmenopausal women
.
Clin Cancer Res
 
2011
;
17
:
1867
1874
.

21

Park
 
S
,
Koo
 
J
,
Park
 
HS
et al.
Expression of androgen receptors in primary breast cancer
.
Ann Oncol
 
2010
;
21
:
488
492
.

22

Astvatsaturyan
 
K
,
Yue
 
Y
,
Walts
 
AE
et al.
Androgen receptor positive triple‐negative breast cancer: Clinicopathologic, prognostic, and predictive features
.
PLoS One
 
2018
;
13
:
e0197827
.

23

Kucukzeybek
 
BB
,
Bayoglu
 
IV
,
Kucukzeybek
 
Y
et al.
Prognostic significance of androgen receptor expression in HER2‐positive and triple‐negative breast cancer
.
Pol J Pathol
 
2018
;
69
:
157
168
.

24

Giovannelli
 
P
,
Di Donato
 
M
,
Auricchio
 
F
et al.
Androgens induce invasiveness of triple‐negative breast cancer cells through AR/Src/PI3‐K complex assembly
.
Sci Rep
 
2019
;
9
:
4490
.

25

Barton
 
VN
,
D'Amato
 
NC
,
Gordon
 
MA
et al.
Multiple molecular subtypes of triple‐negative breast cancer critically rely on androgen receptor and respond to enzalutamide in vivo
.
Mol Cancer Ther
 
2015
;
14
:
769
778
.

26

Traina
 
TA
,
Miller
 
K
,
Yardley
 
DA
et al.
Enzalutamide for the treatment of androgen receptor‐expressing triple‐negative breast cancer
.
J Clin Oncol
 
2018
;
36
:
884
890
.

27

Bonnefoi
 
H
,
Grellety
 
T
,
Tredan
 
O
et al.
A phase II trial of abiraterone acetate plus prednisone in patients with triple‐negative androgen receptor positive locally advanced or metastatic breast cancer (UCBG 12‐1)
.
Ann Oncol
 
2016
;
27
:
812
818
.

28

Trigunaite
 
A
,
Dimo
 
J
,
Jørgensen
 
TN
.
Suppressive effects of androgens on the immune system
.
Cell Immunol
 
2015
;
294
:
87
94
.

29

Jiang
 
G
,
Shi
 
L
,
Zheng
 
X
et al.
Androgen receptor affects the response to immune checkpoint therapy by suppressing PD‐L1 in hepatocellular carcinoma
.
Aging (Albany NY)
 
2020
;
12
:
11466
11484
.

30

Kwilas
 
AR
,
Ardiani
 
A
,
Gameiro
 
SR
et al.
Androgen deprivation therapy sensitizes triple‐negative breast cancer cells to immune‐mediated lysis through androgen receptor independent modulation of osteoprotegerin
.
Oncotarget
 
2016
;
7
:
23498
23511
.

31

Greenstein
 
BD
,
Fitzpatrick
 
FT
,
Adcock
 
IM
et al.
Reappearance of the thymus in old rats after orchidectomy: Inhibition of regeneration by testosterone
.
J Endocrinol
 
1986
;
110
:
417
422
.

32

Velardi
 
E
,
Tsai
 
JJ
,
Holland
 
AM
et al.
Sex steroid blockade enhances thymopoiesis by modulating Notch signaling
.
J Exp Med
 
2014
;
211
:
2341
2349
.

33

Page
 
D
,
Kim
 
I
,
Chun
 
B
et al.
A phase II study of dual immune checkpoint blockade (ICB) plus androgen receptor (AR) blockade to enhance thymic T‐cell production and immunotherapy response in metastatic breast cancer (MBC)
.
J Clin Oncol
 
2019
;
37
:
TPS1106a
.

34

Ardiani
 
A
,
Farsaci
 
B
,
Rogers
 
CJ
et al.
Combination therapy with a second‐generation androgen receptor antagonist and a metastasis vaccine improves survival in a spontaneous prostate cancer model
.
Clin Cancer Res
 
2013
;
19
:
6205
6218
.

35

Liu
 
CM
,
Aziz
 
M
,
Kachur
 
S
et al.
BactQuant: An enhanced broad‐coverage bacterial quantitative real‐time PCR assay
.
BMC Microbiol
 
2012
;
12
:
56
.

36

Tung
 
N
,
Garber
 
JE
,
Hacker
 
MR
et al.
Prevalence and predictors of androgen receptor and programmed death‐ligand 1 in BRCA1‐associated and sporadic triple‐negative breast cancer
.
NPJ Breast Cancer
 
2016
;
2
:
16002
.

37

Sutherland
 
JS
,
Goldberg
 
GL
,
Hammett
 
MV
et al.
Activation of thymic regeneration in mice and humans following androgen blockade
.
J Immunol
 
2005
;
175
:
2741
2753
.

38

Overmoyer
 
B
,
Sanz‐Altamira
 
P
,
Taylor
 
RP
et al.
Enobosarm: A targeted therapy for metastatic, androgen receptor positive, breast cancer
.
J Clin Oncol
 
2014
;
32
:
568a
.

39

Davey
 
RA
,
Grossmann
 
M
.
Androgen receptor structure, function and biology: From bench to bedside
.
Clin Biochem Rev
 
2016
;
37
:
3
15
.

40

Gerratana
 
L
,
Basile
 
D
,
Buono
 
G
et al.
Androgen receptor in triple‐negative breast cancer: A potential target for the targetless subtype
.
Cancer Treat Rev
 
2018
;
68
:
102
110
.

41

Pietri
 
E
,
Conteduca
 
V
,
Andreis
 
D
et al.
Androgen receptor signaling pathways as a target for breast cancer treatment
.
Endocr Relat Cancer
 
2016
;
23
:
R485
R498
.

42

Kono
 
M
,
Fujii
 
T
,
Lim
 
B
et al.
Androgen receptor function and androgen receptor‐targeted therapies in breast cancer: A review
.
JAMA Oncol
 
2017
;
3
:
1266
1273
.

43

Gonzalez‐Angulo
 
AM
,
Stemke‐Hale
 
K
,
Palla
 
SL
et al.
Androgen receptor levels and association with PIK3CA mutations and prognosis in breast cancer
.
Clin Cancer Res
 
2009
;
15
:
2472
2478
.

44

Lehmann
 
BD
,
Bauer
 
JA
,
Schafer
 
JM
et al.
PIK3CA mutations in androgen receptor‐positive triple‐negative breast cancer confer sensitivity to the combination of PI3K and androgen receptor inhibitors
.
Breast Cancer Res
 
2014
;
16
:
406
.

45

Shi
 
Y
,
Yang
 
F
,
Huang
 
D
et al.
Androgen blockade based clinical trials landscape in triple‐negative breast cancer
.
Biochim Biophys Acta Rev Cancer
 
2018
;
1870
:
283
290
.

46

Bhattarai
 
S
,
Klimov
 
S
,
Mittal
 
K
et al.
Prognostic role of androgen receptor in triple‐negative breast cancer: A multi‐institutional study
.
Cancers (Basel)
 
2019
;
11
:
995
.

47

Gucalp
 
A
,
Proverbs‐Singh
 
T
,
Corben
 
A
et al.
Phase I/II trial of palbociclib in combination with bicalutamide for the treatment of androgen receptor (AR)+ metastatic breast cancer (MBC)
.
J Clin Oncol
 
2016
;
34
:
TPS1103a
.

48

Sharifi
 
M
,
Wisinski
 
K
,
Burkard
 
ME
et al.
Abstract OT1‐02‐01: Phase I trial of bicalutamide and ribociclib in androgen receptor‐positive triple‐negative breast cancer
.
Can Res
 
2019
;
79
:
OT1
02a
.

49

Salgado
 
R
,
Denkert
 
C
,
Demaria
 
S
et al.
The evaluation of tumor‐infiltrating lymphocytes (TILs) in breast cancer: Recommendations by an International TILs Working Group 2014
.
Ann Oncol
 
2015
;
26
:
259
271
.

50

Beaubier
 
N
,
Tell
 
R
,
Lau
 
D
et al.
Clinical validation of the tempus xT next‐generation targeted oncology sequencing assay
.
Oncotarget
 
2019
;
10
:
2384
2396
.

51

Yarchoan
 
M
,
Hopkins
 
A
,
Jaffee
 
EM
.
Tumor mutational burden and response rate to PD‐1 inhibition
.
N Engl J Med
 
2017
;
377
:
2500
2501
.

52

Barroso‐Sousa
 
R
,
Jain
 
E
,
Cohen
 
O
et al.
Prevalence and mutational determinants of high tumor mutation burden in breast cancer
.
Ann Oncol
 
2020
;
31
:
387
394
.

53

Chen
 
B
,
Khodadoust
 
MS
,
Liu
 
CL
et al.
Profiling tumor infiltrating immune cells with CIBERSORT
.
Methods Mol Biol
 
2018
;
1711
:
243
259
.

54

Newman
 
AM
,
Steen
 
CB
,
Liu
 
CL
et al.
Determining cell type abundance and expression from bulk tissues with digital cytometry
.
Nat Biotechnol
 
2019
;
37
:
773
782
.

55

Sharma
 
NL
,
Massie
 
CE
,
Ramos‐Montoya
 
A
et al.
The androgen receptor induces a distinct transcriptional program in castration‐resistant prostate cancer in man
.
Cancer Cell
 
2013
;
23
:
35
47
.

56

Lee
 
C
,
Jeong
 
H
,
Bae
 
Y
et al.
Targeting of M2‐like tumor‐associated macrophages with a melittin‐based pro‐apoptotic peptide
.
J Immunother Cancer
 
2019
;
7
:
147
.

57

Cimpean
 
AM
,
Tamma
 
R
,
Ruggieri
 
S
et al.
Mast cells in breast cancer angiogenesis
.
Crit Rev Oncol Hematol
 
2017
;
115
:
23
26
.

58

Banerjee
 
S
,
Tian
 
T
,
Wei
 
Z
et al.
Distinct microbial signatures associated with different breast cancer types
.
Front Microbiol
 
2018
;
9
:
951
.

59

Swoboda
 
A
,
Sharma
 
A
,
Olopade
 
O
et al.
Characterizing the gut microbiome of patients with triple‐negative breast cancer
.
J Clin Oncol
 
2019
;
37
:
e14186a
.

60

Gong
 
J
,
Chehrazi‐Raffle
 
A
,
Placencio‐Hickok
 
V
et al.
The gut microbiome and response to immune checkpoint inhibitors: preclinical and clinical strategies
.
Clin Transl Med
 
2019
;
8
:
9
.

61

Routy
 
B
,
Le Chatelier
 
E
,
Derosa
 
L
et al.
Gut microbiome influences efficacy of PD‐1‐based immunotherapy against epithelial tumors
.
Science
 
2018
;
359
:
91
97
.

62

Chaput
 
N
,
Lepage
 
P
,
Coutzac
 
C
et al.
Baseline gut microbiota predicts clinical response and colitis in metastatic melanoma patients treated with ipilimumab
.
Ann Oncol
 
2017
;
28
:
1368
1379
.

63

Frankel
 
AE
,
Coughlin
 
LA
,
Kim
 
J
et al.
Metagenomic shotgun sequencing and unbiased metabolomic profiling identify specific human gut microbiota and metabolites associated with immune checkpoint therapy efficacy in melanoma patients
.
Neoplasia
 
2017
;
19
:
848
855
.

This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://dbpia.nl.go.kr/journals/pages/open_access/funder_policies/chorus/standard_publication_model)