Molecular aberrations that can activate the PI3K/Akt/mTOR axis are common in mesenchymal/metaplastic breast cancer- and temsirolimus-based regimens offer promise for treatment of these rare cancers. Response rates (CR + PR) were 32% including 2 CRs in patients treated with liposomal doxorubicin and temsirolimus (±bevacizumab).
Keywords: metaplastic breast cancer, liposomal doxorubicin, bevacizumab, temsirolimus
Abstract
Background
Mesenchymal/metaplastic breast cancers (MpBCs) are often triple-negative (TNBC), and chemo-refractory, and can harbor phosphoinositide 3-kinase (PI3kinase) alterations; thus, therapy with mTor inhibitors may demonstrate activity.
Patients and methods
Patients with mesenchymal/MpBC treated with temsirolimus-based regimens were evaluated. Mutational analyses [polymerase chain reaction (PCR)-based DNA sequencing method, mass spectrometric detection (Sequenom MassARRAY), or next-generation sequencing] as well as loss of phosphatase and tensin homolog (PTEN) (immunohistochemistry) were performed (archived tissue when available).
Results
Twenty-three patients (one of whom was on two separate trials) were treated using temsirolimus-containing regimens: temsirolimus alone (n = 1 patient) or combined with the following: liposomal doxorubicin and bevacizumab (DAT, n = 18); liposomal doxorubicin (DT, n = 1); paclitaxel and bevacizumab (TAT, n = 2); paclitaxel (TT, n = 1); carboplatin and bevacizumab (CAT, n = 1). Response rate [complete response (CR) + partial response (PR)] was 25% across all regimens; 32% in the anthracycline-based regimens [DAT and DT (CR = 2, PR = 4; N = 19)]. An additional two patients achieved stable disease (SD) ≥6 months [total SD ≥6 months/CR/PR = 8 (33%)]. Molecular aberrations in the PI3K pathway were common: PIK3CA mutation = 6/15 (40%), PTEN mutation = 3/11 (27%), and PTEN loss = 2/11 (18%). A point mutation in the NF2 gene (K159fs*16; NF2 alterations can activate mTor) was found in one patient who attained CR (3+ years). Of the eight patients who achieved SD ≥6 months/CR/PR, all 4 patients with available tissue had a molecular aberration that activate the PIK3CA/Akt/mTOR axis: PIK3CA mutation = 2; PTEN loss = 1; NF2 aberration = 1.
Conclusions
DAT has activity in MpBCs including complete CRs. Molecular aberrations that can activate the PI3 K/Akt/mTOR axis are common in MpBC.
introduction
MpBCs are defined as invasive carcinomas that have either squamous or mesenchymal differentiation and are commonly TNBC [1]. Despite variation in prognosis depending on grade and histologic variant, high-grade MpBCs are relatively chemo-refractory and carry a worse prognosis than nonmetaplastic, triple-negative breast cancers (TNBCs) [2–7].
Advances in molecular testing have further characterized TNBCs suggesting that subsets such as ‘claudin-low’, ‘mesenchymal’, or ‘mesenchymal-stem cell like’ are enriched in epithelial-to-mesenchymal transition (EMT), maintain stem cell-like characteristics, have PI3K pathway activation, and respond to PI3K inhibitors in preclinical models [8, 9]. Further characterization of claudin-low tumors revealed that 20%–30% are MpBCs [10]. Like claudin-low and mesenchymal/mesenchymal-stem cell-like tumors, MpBCs are also enriched in EMT, display PI3K pathway activation, and commonly carry PIK3CA mutations [11]. MpBCs also show strong correlation with a circulating stem cell-derived genomic profile heavily weighted for PI3K activity [11, 12]. Thus, treatment regimens that induce response in MpBCs may be used to treat the subsets of TNBC that lack metaplastic morphology, but share similar molecular characteristics. Given these molecular features, we report outcomes of patients with MpBC who were treated within the M.D. Anderson Cancer Center (MDACC) using regimens containing the mTOR inhibitor temsirolimus. Data from the first five patients treated have been previously published but were included to capture the entire experience for this rare cancer subtype [13].
patients and methods
patient selection and data collection
Patients who carried a diagnosis of MpBC (invasive myoepithelial carcinoma for one patient) confirmed by pathology review at MDACC were treated in accordance with MDACC internal review board (IRB) guidelines. Response assessments occurred every 6–12 weeks using Response Evaluation Criteria in Solid Tumors (RECIST) [14].
molecular correlative studies
When archived tissue was available, DNA was analyzed by polymerase chain reaction (PCR)-based DNA sequencing method for select genes of interest, particularly PIK3CA mutations [15, 16]. After January 2011, the assay was changed to mass spectrometric detection (Sequenom MassARRAY) [17]. In some patients, loss of PTEN was measured using immunohistochemistry (DAKO, Carpinteria, CA). Genes analyzed by PCR-based sequencing were not re-analyzed by Sequenom. Two patients' tumors underwent next-generation sequencing through Foundation Medicine (182 and 236 gene panel, Cambridge, MA).
results
patients and treatment
From May 2009 until September 2012, 23 patients were treated using six different temsirolimus-based therapy regimens (1 patient treated on two separate trials), yielding 24 analyzable outcomes (Table 1). All but one patient had metastatic cancer and this patient had T4, locally advanced cancer that was unresponsive to neoadjuvant anthracycline-based chemotherapy. All patient tumors were negative for ER/PR or HER2 with the majority (91%) having visceral disease. The most common histology was sarcomatoid carcinomas [including keratin-positive, pure sarcomatoid morphology (n = 7) or both sarcomatoid and epithelial components (n = 7)], followed by matrix-producing carcinomas (n = 7) Table 1. All of the matrix-producing carcinomas had chondroid or chondromyxoid stroma. Median number of prior chemotherapy regimens was 2 (range 0–7).
Table 1.
Patient characteristics (n = 23 patients)
Characteristics | No. | % |
---|---|---|
Age | ||
Median | 59 | |
Range | 21–86 | |
ECOG | ||
0 | 3 | 13 |
1 | 18 | 78 |
2 | 2 | 9 |
Metastatic disease | ||
Yes | 22 | 96 |
No | 1 | 4 |
Visceral organ disease | ||
Yes | 21 | 91 |
No | 2 | 12 |
Hormone receptor status | ||
Positive | 0 | 0 |
Negative | 23 | 100 |
HER2 status | ||
Positive | 0 | 0 |
Negative | 21 | 91 |
Unknown | 2 | 9 |
Prior therapies for MBC | ||
0 | 5 | 22 |
1 | 5 | 22 |
>1 | 13 | 56 |
Prior anthracycline | ||
Yes | 15 | 65 |
No | 8 | 35 |
Prior bevacizumab | ||
Yes | 6 | 26 |
No | 17 | 74 |
Predominant histology | ||
Sarcomatoid, pure | 7 | 30 |
Sarcomatoid, mixed | 7 | 30 |
Matrix-producing | 7 | 30 |
Other | 2 | 9 |
All patients received therapy with the temsirolimus, most in combination with chemotherapy ± bevacizumab (Table 2). Liposomal doxorubicin was the usual chemotherapy backbone (n = 19), followed by paclitaxel (n = 3; including one patient previously treated with DAT) and carboplatin (n = 1). One patient received temsirolimus alone. Several patients (n = 6) who began therapy with liposomal doxorubicin lost access during a nationwide drug shortage and received targeted agents alone for ∼6 months. Two patients developed PD and discontinued therapy; one subsequently received therapy with PAT.
Table 2.
Response by treatment regimen
Treatment regimen | Dose | No. of treated | Median no. of cycles | CBR | RR |
---|---|---|---|---|---|
DAT | 20 mg/m2, 15 mg/kg, 25 mg | 5 | 6 | 2 | 2 |
30 mg/m2, 15 mg/kg, 25 mg | 13 | 5 | 4 | 3 | |
PATa | 60 mg/m2, 15 mg/kg, 25 mg | 2a | 7a | 1a | 0 |
CAT | 4 AUC, 10 mg/kg, 25 mg | 1 | 2 | ||
DT | 30 mg/m2, 25 mg | 1 | 12 | 1 | 1 |
PT | 60 mg/m2, 25 mg | 1 | 3 | ||
T | 25 mg | 1 | 2 |
aOne patient previously treated with DAT and one patient treated neoadjuvantly.
D, liposomal doxorubicin; A, bevacizumab; T, temsirolimus; P, paclitaxel; C, carboplatin.
efficacy
The response rate [complete response (CR)/partial response (PR)] to PI3K/Akt/mTor pathway-directed therapy in treated patients (n = 24 outcomes) with MpBC was 25% (n = 6). All responding patients received liposomal doxorubicin for a RR of 32% to the DAT/DT regimens. One patient attained a CR by RECIST with DAT after eight cycles of therapy (two without bevacizumab due to toxicity) followed by temsirolimus alone for a year before transitioning to oral everolimus (10 mg daily), which she continues to date (4 years on) without evidence of disease (Figure 1). Notably, this patient was treated with DAT as first-line therapy for her metastatic disease and was anthracycline naïve as she had only received prior docetaxel and cyclophosphamide (five cycles) in the adjuvant setting. A second patient who achieved CR was also treated with DAT [progression free survival (PFS) = 22 months] (Table 4). This patient had received four cycles of doxorubicin and cyclophosphamide in the adjuvant setting and was treated with carboplatin and paclitaxel followed by radiation for a chest wall recurrence prior to beginning DAT for metastatic disease in the lung. Four patients achieved PRs; three were treated with DAT; and one with DT. Two additional patients receiving PI3K pathway-directed therapy in combination with chemotherapy achieved SD ≥6 months (DAT = 1, PAT = 1) for an overall clinical benefit rate (CBR = SD ≥6 months/CR/PR) of 35% (n = 8 of 23 patient outcomes) and 37% in those receiving DAT/DT.
Figure 1.
Scans of the chest showining response in patients treated with liposomal doxorubicin, bevacizumab and temsirolimus (DAT).
Table 4.
Characteristics of responders
Patient # | Response | Histology | Molecular | Treatment regimen | # of prior cytotoxic regimens (metastatic disease) | Prior anthracycline | # of cycles |
---|---|---|---|---|---|---|---|
01 | CR | Sarcomatoid, pure | NF2 truncation; TP53 mutation | D 20 mg/m2; A 15 mg/kg; T 25 mg | 1 (0) | None | 8 |
09 | CRa | Sarcomatoid, pure |
PIK3CA mutation H1047R |
D 30 mg/m2; A 15 mg/kg; T 25 mg | 2 (0) | Adjuvant | 17 |
02 | PR (−40%) | Sarcomatoid, pure | Unknown | D 20 mg/m2; A 15 mg/kg; T 25 mg | 1 (0) | None | 4 |
12b | PR (−61%) | Matrix-producing | No loss of PTEN; mutation status unknown | D 30 mg/m2; A 15 mg/kg; T 25 mg | 2 (0) | Neoadjuvant | 5 |
17 | PR (−32%) | Sarcomatoid, mixed | PTEN loss | D 20 mg/m2; A 15 mg/kg; T 25 mg | 1 (0) | Neoadjuvant | 15 |
18 | PR (−40%) | Myoepithelial | Unknown | D 30 mg/m2; A 15 mg/kg; T 25 mg | 1 (1) | None | 22 |
06 | SD ≥6 months (+8%) | Sarcomatoid, pure |
PIK3CA mutation H1047R |
D 20 mg/m2; A 15 mg/kg; T 25 mg | 1 (0) | Adjuvant | 7 |
aPatient had PET avid disease prior to therapy with a minimal residual linear abnormality on CT chest that demonstrated no residual FDG uptake after 17 cycles of therapy; considered a CR. She transitioned to oral everolimus (10 mg daily) and later received consolidative stereotactic radiation to her isolated, treated pulmonary metastasis. PFS was 22 months (developed chest wall recurrence).
bAlso experienced SD ≥6 months on PAT.
toxicity
Toxicities associated with DAT have been previously published and thus not detailed here [18]. However, it is important to note that one patient with a history of postobstructive pneumonia died of pneumonia during cycle 2 of therapy with DAT. A second patient treated with DAT had grade 3 congestive heart failure simultaneously with disease progression within the brain and chose to enter hospice, so it is unknown if her heart function improved after discontinuing therapy.
molecular correlative studies
Archived tissue was available for molecular analysis in 16 of 23 patients (one patient had breast recurrence and lung metastasis analyzed) (Tables 3–5). Half of the samples tested (n = 8) were from primary tumors and half (n = 8) were from recurrent disease (4 from local recurrences and 4 from distant metastatic disease). Nine samples were evaluated prior to 2011 using PCR-based sequencing and 7 were evaluated using mass spectrometric detection (Table 5). No genes analyzed by PCR-based sequencing were reanalyzed by mass spectrometric techniques; however, tumor samples from patient 15 underwent analysis of different genes, initially by PCR (PIK3CA, PTEN) and then by mass spectrometric techniques (BRAF, KRAS, AKT1). A molecular aberration in the PI3K pathway was detected in 69% of patients tested (Table 5). The majority harbored mutations in PIK3CA (6 of 15, 40%) or PTEN aberrations (5 of 15, 33%). Two of 11 (18%) had loss of PTEN by IHC and 3 of 11 (27%) harbored aberrations in PTEN (Table 5). Both patients who achieved CRs harbored molecular aberrations: one a PIK3CA mutation, the other an NF2 mutation (Table 5).
Table 3.
Response according to aberration status
Mutation (# tested) | # positive | CR | PR | SD ≥6 months | PD/SD ≤6 months |
---|---|---|---|---|---|
PI3 Kinase (n = 15) | 6 | 1 | 0 | 1 | 4 |
PTEN mutation (n = 11) | 3 | 0 | 0 | 0 | 3 |
PTEN loss by IHC (n = 11) | 2 | 0 | 1 | 0 | 1 |
NF2 mutation (n = 2) | 1 | 1 | 0 | 0 | 0 |
Unknowna or no aberration present (n = 7) | NA | 0 | 2 | 1 | 4 |
aInadequate tissue available.
Table 5.
Genomic aberrations and associated clinical features in patients with tissue available for analysis
Patient # | Histology | Regimen | Aberration | Genes or protein tested | Testing | Best response |
---|---|---|---|---|---|---|
1 | Sarcomatoid, pure | DAT | None PTEN detected NF2 K159fs*16 TP53 F109fs*39 |
PIK3CA*, EGFR, BRAF, KRAS, PTEN PTEN expression 182 gene panel |
PCR-based IHC Foundation Medicine |
CR |
3 | Sarcomatoid, pure | DAT | None PTEN detected |
PIK3CA*, EGFR, BRAF,
KRAS, C-KIT PTEN expression |
PCR-based IHC |
SD (2 cycles) |
5 | Matrix producinga | DAT | PTEN (D92G) | PTEN, PIK3CA*, EGFR, BRAF, KRAS, C-KIT | PCR-based | SD (6 cycles) |
6 | Sarcomatoid, pure | DAT | PI3K (H1047R) | PIK3CA*, EGFR, BRAF, KRAS, C-KIT | PCR-based | SD ≥6 months (8 cycles) |
7 | Sarcomatoid, pure | DAT | PTEN (14 base pair deletion in exon 1) | PIK3CA**, PTEN | Sequenom | SD (4 cycles) |
8 | Sarcomatoid, mixed | DAT |
PI3K (E545A and H1047Y) PTEN detected |
PIK3CA*
PTEN expression |
PCR-based IHC |
SD (4 cycles) |
9 | Sarcomatoid, pure | DAT |
PI3K (H1047R) PTEN detected |
PIK3CA*, BRAF, KRAS,
C-KIT, TP53 PTEN expression |
PCR-based IHC |
CRb |
10 | Sarcomatoid, mixed | DAT |
PI3K (H1047R) PTEN detected |
PIK3CA*, BRAF, KRAS,
PTEN PTEN expression |
PCR-based IHC |
NEc |
12 | Matrix producing | DAT and PAT | None PTEN detected |
EGFR, KRAS, NRAS,
C-KIT, AKT PTEN expression |
Sequenom IHC |
PR (DAT) then SD ≥6 months (PAT) |
14 | Sarcomatoid, mixed | DAT |
PI3K (E545K) PTEN detected |
PIK3CA*, EGFR, BRAF,
KRAS, PTEN PTEN expression |
PCR-based IHC |
PD (2 cycles) |
15 | Sarcomatoid, mixed | T | None None PTEN detected |
PIK3CA*, PTEN BRAF, KRAS, AKT1 PTEN expression |
PCR-based Sequenom IHC |
SD (2 cycles) |
16 | Matrix producing | DAT | None PTEN detected |
PIK3CA**, EGFR, BRAF,
KRAS, C-KIT, PTEN,
MET, GNAQ PTEN expression |
Sequenom IHC |
PD (2 cycles) |
17 | Sarcomatoid, mixed | DT | No mutation PTEN loss |
PIK3CA**, PTEN, MET PTEN expression |
Sequenom IHC |
PR (12 cycles) |
20 | Matrix producing | CAT | None | PIK3CA**, EGFR, BRAF, KRAS, C-KIT, PTEN, MET, GNAQ, GNAS, IDH1, IDH2, NRAS, RET | Sequenom | PD (2 cycles) |
21 | Sarcomatoid, pure | DAT | None | PIK3CA**, EGFR, BRAF, KRAS, C-KIT, PTEN, MET, GNAQ, GNAS, IDH1, IDH2, NRAS, RET | Sequenom | PD (2 cycles) |
22 | Matrix producing | DAT |
PTEN loss PIK3R1 (N453_T454insNN) PTEN (K327fs*16) MCL1 amplification TP53 (N131del) KDM6A (Q692fs*37) RB1 (splice site 1421+1G > C) |
PTEN expression 236 gene panel |
IHC Foundation Medicine |
PD (2 cycles) |
Genes ‘evaluated by PCR’: EGFR (exons 18 to 21), BRAF (codons 595 to 600 in exon 15), KRAS (codons 12, 13 or 61), C-KIT (exons 9, 11, 13, 17) PTEN, exons 1–9, PIK3CA* (codons 532 to 554 in exon 9 or codons 1011 to 1062 in exon 20), PIK3CA**(codons 60, 88, 110, 111, 345, 405, 418, 420, 453, 539, 542, 545, 546, 909, 1021, 1025, 1043, 1046, 1047, or 1049), TP53 (exons 4 to 9), AKT1 (codons 17, 173, or 179), MET (codons 375, 848, 988, 1010, 1112, 1124, 1248, 1253, or 1268), GNAQ (codon 209), GNAS (codon 201), IDH1 (codon 132), IDH2 (codon 172), NRAS (codon 12, 13, or 61), RET (codon 918)
aBy outside pathology review, slides not reviewed at M.D. Anderson.
bPatient had PET avid disease prior to therapy with a minimal residual linear abnormality on CT chest that demonstrated no residual FDG uptake after 17 cycles of therapy; considered a CR. She transitioned to oral everolimus (10 mg daily) and later received consolidative stereotactic radiation to her isolated, treated pulmonary metastasis. PFS was 22 months (developed chest wall recurrence).
cDeath during cycle 2 of therapy due to pneumonia, not evaluated for response (NE).
Of the four patients who attained PR, one had PTEN loss and the other three did not have adequate tissue available for complete testing (Table 3); of the two patients who achieved CR, both had aberrations that could lead to PI3K pathway activation (Tables 4 and 5). Overall, of the eight patients with clinical benefit, all four with correlative tissue had an aberration that could lead to activation of the PI3K/Akt/mTOR axis (Table 3). An additional patient with a detectable mutation in PIK3CA (H1047R) died of pneumonia during cycle 2 of therapy with DAT and was, thus, not assessed for response using RECIST; however, a reduction in size of her lung metastases was noted on chest X-ray prior to starting her second cycle of DAT. Not all patients with mutations responded. A patient with detected PIK3CA mutation E545K developed rapid disease progression after two cycles of DAT and the patient with detected PIK3CA mutations E545A and H1047Y developed disease progression after four cycles of DAT.
discussion
We describe the largest prospective series documenting treatment of MpBC, a rare but historically aggressive subtype of TNBC. MpBC includes a spectrum of histologic types, usually high grade and considered chemo-refractory. In retrospective series short-lived, partial responses have been noted with doxorubicin in patients with metastatic disease [5, 19]. Our prospectively collected data demonstrates that a variety of aberrations in the PI3K/Akt/mTor axis are a hallmark of these patients. Indeed, 10 of 16 patients with available tissue had an aberration in one or more of the following genes that affect this pathway (Table 5): PIK3CA, PTEN, NF2, or PIK3R1. Since next-generation sequencing was not available for many patients, it is plausible that other patients who had less comprehensive molecular testing might have also harbored an aberration in this pathway.
Additionally, we identified a CR/PR rate of 26% using chemotherapy with temsirolimus (±bevacizumab) and a 32% CR/PR rate with DAT/DT. Responses to DAT/DT included 2 CRs, one ongoing (with maintenance everolimus) 3+ years after discontinuing protocol therapy; time to progression from initiation of therapy in the second patient was 22 months.
Additionally, two other patients attained SD for ≥6 months for a CBR of 33% overall, 35% among those who received chemotherapy with temsirolimus (±bevacizumab) and 37% in those treated with DAT/DT. These response rates are consistent with rates published for PI3K-directed therapy in nonbreast cancer patients whose tumors harbor PIK3CA mutations [15, 16, 20, 21]. Most patients also received bevacizumab as a component of their therapeutic regimen. MpBCs commonly express VEGF and HIF-1, suggesting bevacizumab may be key to the success of the DAT regimen [22, 23].
The regimens were generally well tolerated with expected myelosuppression, mucositis, hand–foot syndrome, dyslipidemia and rarely pneumonitis [18]. One death occurred from postobstructive pneumonia and one patient had grade 3 congestive heart failure during DAT.
Though considered rare, MpBCs share similar molecular features to claudin-low and mesenchymal/mesenchymal-stem cell-like tumors, subsets comprising up to 30% of TNBCs [8, 9, 11]. In fact, MpBCs make-up a portion of tumors molecularly characterized as claudin-low [10]. Unlike other mesenchymal tumors, MpBCs can be identified using microscopy, thus have undergone more extensive molecular characterization. We have confirmed previously published rates of PIK3CA/PTEN mutations rates in MpBC and one CR tumor harbored a PIK3CA mutation. Additionally, a patient with durable CR (3+ years) to therapy exhibited NF2 mutation. NF2 encodes merlin, a negative regulator of the mammalian target of rapamycin complex 1 (mTORC1). Loss of function mutations in NF2 is associated with sensitivity to rapamycin in preclinical models of mesothelioma [24]. Interestingly, a durable complete response was seen with single-agent everolimus in bladder cancer with an NF2 nonsense mutation; further corroborating the potential for mTOR dependence in tumors with aberrations in NF2 [25]. Additionally, bevacizumab ± mTOR inhibition has reduced schwannomas, stabilized disease, and/or improved in symptoms in neurofibromatosis type 2 [26–28]. Trials are ongoing to determine the role of rapamycin analogs in neurofibromatosis [29, 30].
While the association of molecular aberrations with treatment response requires confirmation, three of six patients with CR or PR potentially activated the PI3K/Akt/mTOR axis (PIK3CA mutation, n = 1; PTEN loss, n = 1; NF2 aberration, n = 1; tissue unavailable in the other three). Both the patient with CR and another patient with SD ≥6 months had PIK3CA/H1047R mutation that is associated with response to mTOR inhibitors in early phase trials.
This study has important limitations. Notably, a range of regimens were employed, but each included temsirolimus, and all CRs and PRs occurred in regimens where liposomal doxorubicin (±bevacizumab) were included. Correlative data were also limited by tissue availability, although several molecular aberrations linked to activation of the PI3K/Akt/mTOR were identified. All patients with clinical benefit (CR/PR/SD ≥6months) and available tissue for molecular testing demonstrated these anomalies. Finally, comparative data consist of small, retrospective analyses involving diverse treatments.
In conclusion, patients with advanced MpBC harbor diverse molecular abnormalities potentially actionable by temsirolimus-based therapies which may achieve durable CR. These promising results justify exploration of the DAT regimen in a randomized clinical trial, including molecular correlates and nationwide recruitment of patients with this rare tumor subtype.
disclosure
RK receives consultant funds from Sequenom and is a founder of RScueRx, Inc. All remaining authors have declared no conflicts of interest.
references
- 1. Tse GM, Tan PH, Putti TC et al. Metaplastic carcinoma of the breast: a clinicopathological review. J Clin Pathol 2006; 59: 1079–1083. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Hennessy BT, Giordano S, Broglio K et al. Biphasic metaplastic sarcomatoid carcinoma of the breast. Ann Oncol 2006; 17: 605–613. [DOI] [PubMed] [Google Scholar]
- 3. Jung SY, Kim HY, Nam BH et al. Worse prognosis of metaplastic breast cancer patients than other patients with triple-negative breast cancer. Breast Cancer Res Treat 2010; 120: 627–637. [DOI] [PubMed] [Google Scholar]
- 4. Luini A, Aguilar M, Gatti G et al. Metaplastic carcinoma of the breast, an unusual disease with worse prognosis: the experience of the European Institute of Oncology and review of the literature. Breast Cancer Res Treat 2007; 101: 349–353. [DOI] [PubMed] [Google Scholar]
- 5. Rayson D, Adjei AA, Suman VJ et al. Metaplastic breast cancer: prognosis and response to systemic therapy. Ann Oncol 1999; 10: 413–419. [DOI] [PubMed] [Google Scholar]
- 6. Barnes PJ, Boutilier R, Chiasson D, Rayson D. Metaplastic breast carcinoma: clinical-pathologic characteristics and HER2/neu expression. Breast Cancer Res Treat 2005; 91: 173–178. [DOI] [PubMed] [Google Scholar]
- 7. Lester TR, Hunt KK, Nayeemuddin KM et al. Metaplastic sarcomatoid carcinoma of the breast appears more aggressive than other triple receptor-negative breast cancers. Breast Cancer Res Treat 2012; 131: 41–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Herschkowitz JI, Simin K, Weigman VJ et al. Identification of conserved gene expression features between murine mammary carcinoma models and human breast tumors. Genome Biol 2007; 8: R76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Prat A, Parker JS, Karginova O et al. Phenotypic and molecular characterization of the claudin-low intrinsic subtype of breast cancer. Breast Cancer Res 2010; 12: R68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Hennessy BT, Gonzalez-Angulo AM, Stemke-Hale K et al. Characterization of a naturally occurring breast cancer subset enriched in epithelial-to-mesenchymal transition and stem cell characteristics. Cancer Res 2009; 69: 4116–4124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Creighton CJ, Li X, Landis M et al. Residual breast cancers after conventional therapy display mesenchymal as well as tumor-initiating features. Proc Natl Acad Sci USA 2009; 106: 13820–13825. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Moulder S, Moroney J, Helgason T et al. Responses to liposomal Doxorubicin, bevacizumab, and temsirolimus in metaplastic carcinoma of the breast: biologic rationale and implications for stem-cell research in breast cancer. J Clin Oncol 2011; 29: e572–e575. [DOI] [PubMed] [Google Scholar]
- 14. Therasse P, Arbuck SG, Eisenhauer EA et al. New guidelines to evaluate the response to treatment in solid tumors. European Organization for Research and Treatment of Cancer, National Cancer Institute of the United States, National Cancer Institute of Canada. J Natl Cancer Inst 2000; 92: 205–216. [DOI] [PubMed] [Google Scholar]
- 15. Janku F, Tsimberidou AM, Garrido-Laguna I et al. PIK3CA mutations in patients with advanced cancers treated with PI3K/AKT/mTOR axis inhibitors. Mol Cancer Ther 2011; 10: 558–565. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Janku F, Wheler JJ, Naing A et al. PIK3CA mutations in advanced cancers: characteristics and outcomes. Oncotarget 2012; 3: 1566–1575. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Janku F, Wheler JJ, Naing A et al. PIK3CA mutation H1047R is associated with response to PI3K/AKT/mTOR signaling pathway inhibitors in early-phase clinical trials. Cancer Res 2013; 73: 276–284. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Moroney JW, Schlumbrecht MP, Helgason T et al. A phase I trial of liposomal doxorubicin, bevacizumab, and temsirolimus in patients with advanced gynecologic and breast malignancies. Clin Cancer Res 2011; 17: 6840–6846. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Al Sayed AD, El Weshi AN, Tulbah AM et al. Metaplastic carcinoma of the breast clinical presentation, treatment results and prognostic factors. Acta Oncol 2006; 45: 188–195. [DOI] [PubMed] [Google Scholar]
- 20. Holsinger FC, Piha-Paul SA, Janku F et al. Biomarker-directed therapy of squamous carcinomas of the head and neck: targeting PI3K/PTEN/mTOR pathway. J Clin Oncol 2013; 31: e137–e140. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Janku F, Wheler JJ, Westin SN et al. PI3K/AKT/mTOR inhibitors in patients with breast and gynecologic malignancies harboring PIK3CA mutations. J Clin Oncol 2012; 30: 777–782. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Bellino R, Arisio R, D'Addato F et al. Metaplastic breast carcinoma: pathology and clinical outcome. Anticancer Res 2003; 23: 669–673. [PubMed] [Google Scholar]
- 23. Kochhar R, Howard EM, Umbreit JN, Lau SK. Metaplastic breast carcinoma with squamous differentiation: molecular and clinical analysis of six cases. Breast J 2005; 11: 367–369. [DOI] [PubMed] [Google Scholar]
- 24. Lopez-Lago MA, Okada T, Murillo MM et al. Loss of the tumor suppressor gene NF2, encoding merlin, constitutively activates integrin-dependent mTORC1 signaling. Mol Cell Biol 2009; 29: 4235–4249. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Iyer G, Hanrahan AJ, Milowsky MI et al. Genome sequencing identifies a basis for everolimus sensitivity. Science 2012; 338: 221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Subbiah V, Slopis J, Hong DS et al. Treatment of patients with advanced neurofibromatosis type 2 with novel molecularly targeted therapies: from bench to bedside. J Clin Oncol 2012; 30: e64–e68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Plotkin SR, Merker VL, Halpin C et al. Bevacizumab for progressive vestibular schwannoma in neurofibromatosis type 2: a retrospective review of 31 patients. Otol Neurotol 2012; 33: 1046–1052. [DOI] [PubMed] [Google Scholar]
- 28. Plotkin SR, Stemmer-Rachamimov AO, Barker FG II et al. Hearing improvement after bevacizumab in patients with neurofibromatosis type 2. N Engl J Med 2009; 361: 358–367. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. James MF, Han S, Polizzano C et al. NF2/merlin is a novel negative regulator of mTOR complex 1, and activation of mTORC1 is associated with meningioma and schwannoma growth. Mol Cell Biol 2009; 29: 4250–4261. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. James MF, Stivison E, Beauchamp R et al. Regulation of mTOR complex 2 signaling in neurofibromatosis 2-deficient target cell types. Mol Cancer Res 2012; 10: 649–659. [DOI] [PubMed] [Google Scholar]