Advances in Medical Management of Early Stage and Advanced Breast Cancer: 2015 Sabrina Witherby, MD,*,† Tina Rizack, MD,* Bachir J. Sakr, MD,* Robert D. Legare, MD,* and William M. Sikov, MD, FACP* Standard management of early stage and advanced breast cancer has been improved over the past few years by knowledge gained about the biology of the disease, results from a number of eagerly anticipated clinical trials and the development of novel agents that offer our patients options for improved outcomes or reduced toxicity or both. This review highlights recent major developments affecting the systemic therapy of breast cancer, broken down by clinically relevant patient subgroups and disease stage, and briefly discusses some of the ongoing controversies in the treatment of breast cancer and promising therapies on the horizon. Semin Radiat Oncol 26:59-70 C 2016 Elsevier Inc. All rights reserved.
Introduction
A
lthough the number of breast cancer deaths in the United States has fallen over the past 15 years, it remains the most common serious cancer and second leading cause of cancer deaths among U.S. women. The past few years have seen a number of new drug approvals and the presentation of results from large, randomized studies that have changed treatment algorithms and offer hope of substantial improvements in outcomes in both early stage and advanced breast cancer. In this review, we discuss those advances and how they have affected the standard of care for many breast cancer patients, focusing on the 3 major subtypes—hormone receptor (HR)– positive/human epidermal growth factor receptor-2 (HER2)– negative, HER2-positive, and triple-negative.
*Program in Women's Oncology, Breast Health Center, Women and Infants Hospital of Rhode Island and Alpert Medical School of Brown University, Providence, RI. †Department of Medicine, Memorial Hospital of Rhode Island, Pawtucket, RI. Robert D. Legare, Sabrina Witherby, Tina Rizack, and Bachir J. Sakr have no conflict of interest. William M. Sikov is on the advisory board (uncompensated) of AbbVie and also received an honoraria (o$5000) from Celgene. Address reprint requests to William M. Sikov, MD, FACP, Breast Health Center, Women and Infants Hospital of Rhode Island, 101 Dudley St., Providence, RI 02906. E-mail:
[email protected]
http://dx.doi.org/10.1016/j.semradonc.2015.09.005 1053-4296//& 2016 Elsevier Inc. All rights reserved.
Hormone Receptor–Positive/ HER2-Negative Breast Cancer Stage I-III HRþ/HER2 Disease Of patients diagnosed with breast cancer in the United States, 60%-70% have HR-positive and HER2-negative (HRþ/ HER2) cancers, the vast majority with stage I-II disease at presentation, especially among those whose cancers are detected by screening mammography (Fig. 1A). A major challenge in the management of these patients is to differentiate those who are at low risk for local or distant recurrence, in whom treatment should be limited and, ideally, have minimal effect on the patient's quality of life, from those at high risk for disease recurrence and death, in whom more intensive and potentially toxic treatments are warranted. Results from recent studies have addressed the duration of adjuvant endocrine therapy, assessed the role of ovarian function suppression (OFS) in premenopausal women and expanded the potential indications for neoadjuvant endocrine therapy. There has also been widespread adoption of the use of genomic assays to select patients in whom chemotherapy may, or may not, offer significant benefit over endocrine therapy alone. Although we have long been aware that more than half of the patients with these typically less-aggressive cancers who eventually recur will do so more than 5 years after initiation of adjuvant endocrine therapy, until fairly recently the benefit of extending that treatment beyond 5 years was uncertain. Studies such as National Surgical Adjuvant Breast and Bowel Project (NSABP) B-14 failed to demonstrate improved outcomes from extending tamoxifen from 5-10 years, though this 59
S. Witherby et al.
60 B Stage IV
A Stage I-III
Stage I-IIA
Surgery
If path stage T1c or T2 N0 send Oncotype; ?role in T1b N0 and T1N1
Stage IIA-IIB
Stage IIB-III
Surgery, unless response could improve surgical options
NACT with AC-T
Neoadjuvant endocrine therapy with AI, unless grade 3 or high-risk Oncotype, then NACT with AC-T or TC
Surgery
If low risk (by clinical stage +/- Oncotype), adjuvant endocrine therapy with tamoxifen if premenopausal and AI > tamoxifen if postmenopausal If high-risk, (neo)adjuvant chemotherapy (AC-T or TC 4-6) followed by adjuvant endocrine therapy, including OFS in premenopausal
Premenopausal: Consider trial of tamoxifen; otherwise OFS and then follow postmenopausal algorithm
Postmenopausal: Letrozole + palbociclib, fulvestrant, or AI PD
Depending on 1st-line choice: fulvestrant (if AI +/- palbociclib) + palbociclib (if FDA approved and did not receive as 1st-line), any AI (if fulvestrant), exemestane (if a non-steroidal AI) +/- everolimus PD Depending on 1st- and 2nd- line treatments: fulvestrant +/-palbociclib, exemestane +/- everolimus, megestrol acetate or medroxyprogesterone, estradiol, or fluoxymesterone
Abbreviations: NACT = neoadjuvant chemotherapy; AI = aromatase inhibitor; AC-T = doxorubicin & cyclophosphamide followed or preceded by docetaxel or paclitaxel (weekly or dose-dense); TC= docetaxel & cyclophosphamide; OFS = ovarian function suppression; PD = progressive disease
Figure 1 Treatment algorithm for hormone receptor–positive, HER2-negative breast cancer (HRþ/HER2 BC). AC-T, doxorubicin and cyclophosphamide followed or preceded by docetaxel or paclitaxel (weekly or dose dense); NACT, neoadjuvant chemotherapy; PD, progressive disease; TC, docetaxel and cyclophosphamide. (Color version of figure is available online.)
was a relatively small study (n ¼ 1172).1 However, the National Cancer Institute of Canada (NCIC) MA.17 trial, which compared 5 years of the aromatase inhibitor (AI) letrozole with placebo in postmenopausal women who were free of disease after 5 years of tamoxifen, demonstrated a 48% improvement in disease-free survival (DFS) and a 39% improvement in overall survival (OS), taking into account crossover from placebo to letrozole when the initial results of the study were announced, illustrating the potential benefit of extended adjuvant endocrine therapy.2,3 Further analysis revealed that younger patients, those who were premenopausal when they started tamoxifen but postmenopausal 5 years later when they transitioned to the AI, received the largest benefit from this treatment.4 The dilemma was how to treat women who were either still premenopausal after 5 years of tamoxifen or unable to tolerate an AI owing to side effects, most often joint stiffness and pain, vaginal dryness and atrophy, and mood swings. The Adjuvant Tamoxifen: Longer Against Shorter (ATLAS) and adjuvant Tamoxifen—To offer more (aTTOM) trials, which were initiated in 1996 and included more than 22,000 women, assessed the benefits and risks of extended adjuvant therapy with tamoxifen.5,6 Results reported in 2012-2013 demonstrated that although patients with HRþ cancers (n ¼ 9600)
assigned to continue tamoxifen had minimal reductions in recurrences and breast cancer–attributed deaths during their additional 5 years of treatment, their risks of recurrence and death from breast cancer were significantly (25%-30%) lower in years 10-15 and beyond, reflecting the indolent and insidious nature of these cancers. The only major side effect of extended treatment with tamoxifen was a doubling of the incidence of endometrial cancer, from 1.2%-2.4%. The benefit of extending adjuvant endocrine therapy beyond 5 years in patients treated with an AI—either alone or after 2-3 years of tamoxifen—are less clear; reports from 2 studies that address this matter—NSABP B-42 and MA.17 R —are eagerly awaited. Pending those findings, oncologists may recommend extending treatment with an AI to 5 years in patients who started it after 2-3 years of tamoxifen or, in higher risk patients who are tolerating it well, simply continuing the AI until these study results become available. Patients whose oncologist recommends stopping the AI (for now) may be comforted by the knowledge that, in both MA.17 and the recently reported LATER trial, patients who started an AI after a break of up to several years still benefited from this treatment.7,8 Despite its potential advantages, the decision to recommend extending adjuvant endocrine therapy should take into
Advances in medical management of breast cancer: 2015 account the patient's likelihood of benefiting as well as the side effects and other risks of that treatment. For example, a patient with a small, node-negative cancer would be expected to have no more than a 1%-2% additional reduction in risk of recurrence with continued treatment with either tamoxifen or an AI, whereas patients with larger or node-positive cancers have a much higher residual risk of recurrence after 5 years. Although there is no way of knowing which patients are going to develop a late recurrence, genomic tests such as the Breast Cancer Index, performed on the original surgical specimens, may help to classify patients as either low risk, in whom endocrine therapy could be discontinued, or high risk, who are more likely to benefit from continuation of treatment.9 The superiority of an AI over tamoxifen as adjuvant therapy in postmenopausal patients was confirmed by a recently published meta-analysis,10 with significant reductions in recurrences (26% at 5 years and 20% at 10 years, resulting in a 3.6% absolute improvement) and breast cancer mortality (15% at 10 years, resulting in a 2.1% absolute improvement). With an AI, endometrial cancer rates were lower (0.4% vs 1.2% at 10 years), fracture rates were higher (8.2% vs 5.5% at 5 years) and non–breast cancer mortality was similar. Patients who received 2-3 years of tamoxifen before switching to an AI had higher recurrence rates for the first few years than patients treated initially with an AI, though the difference in breast cancer mortality at 10 years was not significant, and results with this group were still superior to those who received only tamoxifen. The value of OFS, either surgical (bilateral oophorectomy) or medical (with a long-acting luteinizing hormone–releasing hormone analog such as goserelin or leuprolide), in premenopausal women with HRþ cancers has been investigated for many years, but prior studies were confounded by either the absence of an appropriate control arm or the uncertainty of ovarian recovery after chemotherapy, which varies by patient age and regimen administered. As tamoxifen competes with estrogen for binding to the estrogen receptor (ER), lowering the estrogen level might improve its efficacy; moreover, with OFS patients have the option of being treated with an AI in place of tamoxifen. In the Tamoxifen and Exemestane Trial (TEXT), premenopausal patients were stratified by whether or not they had received chemotherapy, but all underwent OFS and were randomized to tamoxifen or exemestane (an AI). In the Suppression of Ovarian Function Trial (SOFT), a control arm—tamoxifen without OFS—was added, and patients who had received chemotherapy were required to have evidence of recovery of ovarian function—either resumption of menses or a premenopausal estrogen level—within 8 months of completing that treatment. A combined analysis of the 2 common arms of the trials confirmed the superiority of the AI over tamoxifen in the setting of OFS.11 The more eagerly awaited results from SOFT revealed a dichotomy—although low-risk patients, who had not received chemotherapy, did very well with tamoxifen alone and did not achieve significantly better results with OFS with either tamoxifen or exemestane, highrisk patients, who had received chemotherapy, did much better with OFS and with exemestane over tamoxifen.12
61 Patients under the age of 35 had the largest benefit from OFS and exemestane. OFS was associated with increased hot flashes, vaginal dryness, and reduced libido, and would be expected to increase bone mineral loss, thus in recommending this treatment oncologists have to balance its effect on a patient's quality of life against her likelihood of benefit. Whether OFS should be extended beyond 5 years in very young women with HRþ cancers has not been studied. As risk categorization for patients enrolled on SOFT did not include a genomic assay such as Oncotype Dx (see later), its availability raises challenging questions, such as whether patients who would have received chemotherapy in the past but do not as a result of a low-risk genomic assay and patients with early stage cancers who might not have received chemotherapy in the past but get this treatment owing to a high-risk genomic assay would benefit from OFS and administration of an AI over tamoxifen alone. In the past, neoadjuvant endocrine therapy, initially with tamoxifen, more recently with an AI, was reserved for women with larger or unresectable cancers believed to be too old or frail for neoadjuvant chemotherapy. With the realization that many HRþ/HER2 cancers achieve little response to chemotherapy but may be very sensitive to estrogen deprivation, the population of patient in whom this approach is contemplated has expanded considerably. ACOSOG Z1031 randomized postmenopausal patients with HRþ/HER2 cancers to one of the 3 US Food and Drug Administration (FDA) approved AIs—anastrozole, letrozole or exemestane—for 14 weeks.13 Although no substantial differences were seen in clinical response rates between the 3 agents, and few patients achieved a pathologic complete response (pCR), many patients who would have required a mastectomy at study entry became candidates for breastconserving surgery (BCS). The Preoperative Endocrine Prognostic Index (PEPI) score, which takes into account tumor and nodal stage, endocrine sensitivity (ER expression), and proliferative rate (Ki67) after neoadjuvant treatment, has been developed to distinguish patients with a good prognosis who would likely not benefit from subsequent adjuvant chemotherapy (PEPI ¼ 0) from those who might (PEPI Z 1). Measurement of Ki67 on a biopsy taken after 2-4 weeks of neoadjuvant endocrine therapy has been proposed to identify patients more (low Ki67) or less (high Ki67) likely to achieve a PEPI ¼ 0. Recognizing the slower response to endocrine therapies than with neoadjuvant chemotherapy, the standard duration of neoadjuvant endocrine therapy (in responding patients) has been expanded from 3-4 months to 6 months. The recognition of significant biologic diversity within HRþ/HER2 tumors, which may determine both risk of recurrence and benefit from both endocrine therapy and chemotherapy, encouraged the development of genomic assays, including Oncotype Dx and Mammoprint. At some institutions, submission of tissue for one of these assays—most often Oncotype Dx—has become “reflex” for node-negative HRþ/HER2 cancers of a defined size (such as 41 cm) in certain patient populations (such as younger than the age of 60 or 70 years). That way, results are available for the patient's postoperative consultation with medical oncology to discuss the role of adjuvant chemotherapy in an early stage cancer if
62 the test suggests a biologically more aggressive cancer than previously suspected. Studies suggest that this test alters treatment recommendations in 20%-30% of patients, more often leading the oncologist to recommend omitting chemotherapy than suggesting chemotherapy when endocrine therapy alone would have been the recommendation. The use of these assays in node-positive patients is less well established, but may be useful in patients with limited nodal involvement. Results from trials like Trial Assigning IndividuaLized Options for Treatment (Rx) (TAILORx) (also known as Program for the Assessment of Clinical Cancer Tests [PACCT-1]), which randomized node-negative patients with an intermediate risk Oncotype Dx Recurrence Score to chemotherapy or not, are eagerly anticipated. A similar trial, Rx for Positive Node, Endocrine Responsive Breast Cancer (RxPONDER), also known as SouthWest Oncology Group (SWOG) 1007, is ongoing for patients with limited node-positive cancers (1-3 nodes with macrometastases) with low- or intermediate-risk Recurrence Scores. A controversial issue in the management of stage I-III HRþ/ HER2 breast cancers is the role of bisphosphonates in the adjuvant setting. Bone is the most common site of distant disease recurrence in patients with this subtype of breast cancer. Preclinical studies suggest that bisphosphonates may possess inherent antitumor activity, and that bisphosphonateinduced osteoclast inhibition may make the bone microenvironment less supportive of the establishment of bone metastases. Reports from studies performed primarily to assess the effect of bisphosphonates on treatment-related osteoporosis and bone fractures in patients with induced menopause suggested a recurrence-free survival benefit, leading to a number of studies that examined this question directly. Although results from the individual studies were mixed, a recently published meta-analysis that included more than 18,000 women from 26 clinical trials demonstrated an 18% reduction in distant recurrences at 10 years in postmenopausal patients, owing solely to a 28% reduction in the incidence of bone metastases, which translated into a 3.3% absolute reduction in breast cancer mortality, with no improvement seen in younger women, distant recurrences outside of bone or contralateral breast cancers.14 This has led some to hypothesize an interaction between osteoclast activation in the setting of a low estrogen level and a more hospitable environment for circulating breast cancer cells that is altered by these agents. In a recently reported study (that did not include a no-treatment arm) of more than 6000 women with stage I-III cancers treated for 3 years there was no difference in DFS and minimal differences in toxicities between the intravenous bisphosphonate zoledronic acid and the oral agents clodronate and ibandronate.15 Although most patients expressed a preference for an oral agent if equally effective, as neither clodronate nor ibandronate (at the dose used in this study) is approved in the United States, the currently available treatment option is zoledronic acid 4 mg administered intravenous every 6 months for 3 years. Its usage varies widely, with some oncologists opting to wait for further data and reserving its use for patients with documented osteoporosis or a
S. Witherby et al. significant decline in bone mineral density following initiation of adjuvant endocrine therapy, others selectively administering it to high-risk patients, and others recommending it more broadly to their postmenopausal patients (and also to higherrisk premenopausal patients undergoing OFS).
HRþ/HER2 Metastatic Breast Cancer Except in patients who present with extensive visceral (usually liver or lung) or very symptomatic metastatic disease, who often receive chemotherapy first, HRþ/HER2 metastatic breast cancer (MBC) is typically treated with a sequence of endocrine therapies unless or until they exhibit resistance to such treatments, at which point they are switched to chemotherapy (Fig. 1B). Recent clinical trial results have led medical oncologists to rethink the sequence of endocrine therapies, especially in postmenopausal women, and to consider the addition of targeted therapies to delay the development of resistance. In 2015, first-line endocrine therapy options for postmenopausal patients include an AI, fulvestrant (an ER antagonist), the combination of fulvestrant and an AI, and the combination of an AI with the cyclin-dependent kinase (CDK) 4/6 inhibitor palbociclib. First-line endocrine therapy for premenopausal patients consists of tamoxifen alone or, following OFS, tamoxifen or any of the options available to postmenopausal patients. Fulvestrant binds to the ER with much greater avidity than tamoxifen, and not only blocks activation of estrogenresponsive genes but promotes degradation of the ER, which is why it is sometimes referred to as a selective ER downregulator (SERD). As a second- or third-line endocrine agent, its use is associated with a median time to progression of only 3-4 months, comparable to that seen with switching to the steroidal AI exemestane after progression on a nonsteroidal AI.16,17 A more intensive regimen (500 mg intramuscularly (IM) on days 1 and 15 then every 28 days) has been shown to be more effective than the initially approved dose and schedule (250 mg IM every 28 days).18 In the phase II FIRST trial, first-line fulvestrant improved median time to progression (mTTP) by 10 months (23 vs 13 months) over anastrozole despite similar overall response rates (ORR) and was associated with fewer side effects.19 In updated results presented at the 2014 San Antonio Breast Cancer Symposium (SABCS), patients assigned to fulvestrant also had an increase in median OS (mOS) from 48-54 months.20 A much larger phase III trial comparing fulvestrant with anastrozole in the first-line setting (FALCON) has completed accrual. The phase III SWOG 0226 trial compared singleagent anastrozole with the combination of anastrozole and fulvestrant as first- or second-line endocrine therapy, using a fulvestrant dose of 250 mg IM for days 1 and 15, then every 28 days.21 Although treatment with the combination yielded only a modest improvement in median progression-free survival (mPFS), there was a more significant increase in mOS (48 vs 41 months), though this benefit appeared to be limited to first-line patients. Pending future results, some oncologists are recommending first-line treatment with fulvestrant, whereas others suggest combination therapy with fulvestrant and an AI,
Advances in medical management of breast cancer: 2015 reserving treatment with an AI alone for patients who decline the IM injections. Although a significant fraction of HRþ/HER2 patients with MBC initially respond to first-line endocrine therapy, some progress rapidly and a significant fraction progress within 6-12 months of starting this treatment. One of the hallmarks of endocrine resistance is increased cellular proliferation and overexpression of cyclin D1, which in turn activates CDK 4/6, which enable progression through the cell cycle and cell division. Palbociclib is a highly selective oral CDK 4/6 inhibitor that demonstrated preferential inhibition of HRþ/HER2 breast cancer cell lines in vitro.22 The phase II PALOMA-1 trial compared letrozole with the combination of letrozole and palbociclib as first-line therapy for postmenopausal women with HRþ/HER2 MBC. Adding palbociclib significantly improved mPFS (from 10-20 months) and ORR (from 39%-55%).23 Although there was no significant improvement in mOS (37.5 vs 33 months, P ¼ 0.81), the FDA granted palbociclib accelerated approval in February 2015, pending confirmation from the PALOMA-2 phase III study. The recently reported phase III PALOMA-3 trial demonstrated that adding palbociclib to fulvestrant in patients with HRþ/HER2 MBC who had progressed on prior endocrine therapy increased mPFS from 3.8-9.2 months compared with fulvestrant alone.24 Given these results, it seems likely that the FDA will approve the use of palbociclib with fulvestrant in the near future, though whether that will include patients receiving fulvestrant as first-line endocrine therapy is unclear. Owing to its antiproliferative effects, treatment with palbociclib is associated with a 450% incidence of grade 3-4 neutropenia, though this is typically asymptomatic, and with modest increases in fatigue, nausea, anemia, diarrhea, and low-grade infections. To what extent these toxicities and the cost of this agent ($10,000-$11,000 per month) may limit its use are not yet known. Endocrine resistance has also been attributed to aberrant activation of intracellular signaling pathways that bypass the block caused by estrogen deprivation. One of the most commonly mutated genes in HRþ/HER2 cancers is PIK3CA, which can lead to constitutive activation of the PI3K/Akt/mTOR signaling pathway. In the phase III BOLERO-2 trial, patients with HRþ/HER2 MBC who had progressed on a nonsteroidal AI assigned to treatment with the oral mTOR inhibitor everolimus in combination with exemestane had superior mPFS (7 vs 3 months) compared with exemestane alone.25 Although this resulted in FDA approval of this combination in the second-line setting, uptake by oncologists and their patients has been limited by toxicities (stomatitis, anemia, fatigue, dyspnea, hyperglycemia, and noninfectious pneumonitis), drug costs, and the lack of a significant survival benefit. In premenopausal women with HRþ/HER2 MBC, although some oncologists will try single-agent tamoxifen in patients with asymptomatic or minimally symptomatic disease, in a meta-analysis of 4 randomized trials the addition of OFS suppression was shown to improve mPFS and mOS.26 Although randomized trial data on the benefits of substituting either an AI or fulvestrant for tamoxifen following OFS in this setting are lacking, given their superiority in postmenopausal
63 women many oncologists have adopted this approach. Finally, as the standard of care is to not permit a patient's ovarian function, once suppressed, to recover, and as recovery of ovarian function on luteinizing hormone–releasing hormone analogs has been known to occur, patients who are initially treated with one of these agents are often encouraged to undergo bilateral oophorectomies. An unanswered question in the management of HRþ/ HER2 MBC is whether patients without symptomatic or extensive visceral disease would benefit from upfront chemotherapy, rather than deferring this treatment until they progress on one or more endocrine therapies. There are no good trials addressing this question, and perhaps the advent of effective targeted therapies will make it moot. Similarly, although endocrine therapy is often administered after first- or subsequent-line chemotherapy in responding patients when response plateaus or excessive toxicity develops, the benefits of this approach in improving mPFS or mOS have not been tested in a randomized trial. Progestational agents like megestrol acetate, pharmacologic doses of estrogen (such as estradiol 2 mg 3 times a day), and androgens like fluoxymesterone have all demonstrated some activity in HRþ/HER2 MBC after progression on the agents discussed above. As with all subtypes of MBC, early incorporation of palliative care in the setting of incurable disease is appropriate.
HER2-Positive Breast Cancer Identification of HER2 and the gene that encodes for it on chromosome 17, recognition that amplification, and consequent overexpression, of that gene in approximately 20% of human breast cancers results in a biologically aggressive and relatively endocrine therapy- and chemotherapy-resistant malignancy, and demonstration that administration of agents, initially the monoclonal antibody trastuzumab, which interfere with the ability of that receptor to activate signaling pathways that promote tumor cell survival, proliferation, and metastasis results in sizable improvements in mPFS and mOS in women with HER2þ breast cancer have been among the most important advances in medical oncology in the past 15 years (Fig. 2). Studies completed, presented and published over the past few years have addressed efforts to enhance the efficacy or reduce the toxicity of standard chemotherapy plus trastuzumab regimens, in both early stage and advanced disease, and to deal with patients whose cancers have become refractory to those treatments. HER2 is a transmembrane receptor. Trastuzumab binds to the extracellular portion of the receptor adjacent to the cell membrane, blocking the conformational change triggered by formation of HER2 homodimers, which is necessary to activate the tyrosine kinase moiety on the intracellular portion of the receptor, thus, preventing initiation of the signal transduction cascade responsible for its biologic activities. One proposed mechanism for resistance to trastuzumab is loss of this extracellular domain; by directly inhibiting the receptor's tyrosine
S. Witherby et al.
64
B Stage IV
A Stage I-III
Stage I
Stage IIA-IIB
Surgery
Surgery, unless response could improve surgical options
If path stage unchanged adjuvant wP + H for T1b/cN0; ? benefit in T1aN0
Stage IIB-III
NACT with TCHP or AC-TH
T + HP or wP + HP (in poor PS patients could consider endocrine therapy + H if HR+ or T-DM1 if covered) PD T-DM1 PD
NACT with TCH (+P if T>3cm or N+) or ACTH+/- P Adjuvant TCH, TCyH or AC-TH, then
Surgery
Non-cross-resistant chemo (liposomal doxorubicin, vinorelbine, gemcitabine, capecitabine, eribulin, ixabepilone, etc.) + H, capecitabine + lapatinib, lapatinib + H (may favor lapatinib-containing regimen if patient has brain mets)
Adjuvant T to complete a year and adjuvant endocrine therapy if HR+ Abbreviations: wP = weekly paclitaxel; H= trastuzumab; NACT = neoadjuvant chemotherapy; TCH = docetaxel, carboplatin & trastuzumab; P=pertuzumab; AC-TH = AC followed or preceded by docetaxel or paclitaxel + H; TCyH = docetaxel, cyclophosphamide & H; HR+ = hormone receptorpositive; T-DM1= trastuzumab emtansine; PD = progressive disease
Figure 2 Treatment algorithm for HER2-positive breast cancer (HER2þ BC). AC-TH, AC followed or preceded by docetaxel or paclitaxel þ H; H, trastuzumab; HRþ, hormone receptor positive; NACT, neoadjuvant chemotherapy; P, pertuzumab; PD, progressive disease; TCH, docetaxel, carboplatin, and trastuzumab; TCyH, docetaxel, cyclophosphamide, and H; T-DM1, trastuzumab emtansine; wP, weekly paclitaxel. (Color version of figure is available online.)
kinase, lapatinib, which, in combination with the oral chemotherapy agent capecitabine, may be effective in HER2þ cancers that progressed on trastuzumab, retains activity against these mutated cells. However, lapatinib also inhibits the tyrosine kinase of the epidermal growth factor receptor (also known as HER1), leading to doselimiting diarrhea and rash similar to that seen with other epidermal growth factor receptor–targeted agents. Another proposed mechanism for trastuzumab resistance is the formation of HER2:HER3 heterodimers, the biologic activity of which is not blocked by trastuzumab. Pertuzumab binds to HER2 at a different site than trastuzumab, and blocks the formation of these dimers. In patients who had progressed on a trastuzumab-containing regimen, the combination of pertuzumab with trastuzumab yielded an ORR of 25%, and half of these patients either responded or had stable disease lasting at least 6 months.27 The CLEOPATRA trial assessed the addition of pertuzumab to the combination of docetaxel and trastuzumab as first-line therapy for HER2þ MBC, and demonstrated statistically significant improvements in PFS (18.5 vs 12.4 months), ORR from (80% vs 69%), and, as reported earlier this year, mOS (56.5 vs 40.8 months).28,29 This led to FDA approval of pertuzumab for HER2þ MBC,
and general acceptance of the docetaxel, trastuzumab, and pertuzumab (THP) regimen as standard first-line therapy, though it should be noted that patients assigned to the control arm of the study did not cross over to pertuzumab as salvage therapy. The addition of pertuzumab was associated with higher rates of diarrhea, mucositis, and rash, raising interesting questions as to role of HER2 heterodimers in normal tissues, and febrile neutropenia, but no increase in the incidence of cardiac toxicity. A recently published phase II study demonstrated similar results with weekly paclitaxel, trastuzumab, and pertuzumab with fewer acute chemotherapy-related toxicities,30 and other chemotherapeutic agents such as vinorelbine have also been given with the antibody combination. If a patient develops persistent chemotherapy-related toxicities, or her response to treatment plateaus, the chemotherapeutic agent can be discontinued and the dual antibody therapy continued until disease progression, as was allowed on CLEOPATRA, sometimes with the addition of an endocrine agent in the 50%-60% of patients who are also HRþ. Although studies support continuation of trastuzumab with second- and subsequent-line therapy in HER2þ patients who progress, no such data exists for pertuzumab and thus this cannot be recommended.
Advances in medical management of breast cancer: 2015 Another promising agent for patients with HER2þ MBC is the antibody-drug conjugate (ADC) trastuzumab emtansine, also known as T-DM1, which consists of a potent antimicrotubule agent (emtansine) linked to trastuzumab which is activated only after the trastuzumab moiety, bound to HER2, is endocytosed and the linker digested, designed to minimize exposure of normal tissues. After demonstrating an ORR of 26% in patients who had progressed while on or shortly after discontinuing a trastuzumab-containing regimen,31 T-DM1 was compared in 2 phase III studies with either the combination of capecitabine and lapatinib (EMILIA) and to treatment of physician's choice (TPC) (TH3RESA) in patients previously treated with a trastuzumab-containing regimen.32,33 In EMILIA, T-DM1 was not only significantly more effective (mPFS ¼ 9.6 vs 6.4 months and mOS ¼ 30.9 vs 25.1 months) but also better tolerated, with thrombocytopenia and elevated transaminases the only toxicities seen more commonly with the ADC. Similar results were reported from the Th3RESA trial, with mPFS of 6.2 months for T-DM1 vs 3.3 for TPC and a mOS trend favoring the ADC. On the basis of the EMILIA results, trastuzumab emtansine was approved as second-line therapy for HER2þ MBC, and is currently considered the standard of care in this setting. Treatment with this agent is usually continued until disease progression or toxicity (neuropathy, thrombocytopenia), with a significant proportion of patients continuing treatment for 12 months and longer with minimal toxicity. The MARIANNE study was designed to determine if singleagent T-DM1 or the combination of T-DM1 and pertuzumab could replace a taxane-trastuzumab regimen as standard firstline therapy for HER2þ MBC. As reported at this year's American Society of Clinical Oncology (ASCO) meeting, whereas both T-DM1-containing regimens were equivalent in efficacy to docetaxel or paclitaxel (physician's choice) and trastuzumab (mPFS ¼ 13.7 vs 14.1 and 15.2 months for single-agent T-DM1 and T-DM1 plus pertuzumab, respectively), neither was superior.34 Responses on the T-DM1 arms were more durable, likely owing to chemotherapy-related toxicities limiting the duration of treatment on the control arm, and side effects, ranging from febrile neutropenia to alopecia, were much less common. The investigators could not explain the lack of added benefit with the addition of pertuzumab to TDM1. Given the results from the CLEOPATRA trial, THP remains the standard first-line regimen in HER2þ MBC, but in patients hoping to avoid hair loss and other toxicities, singleagent T-DM1 may be an appealing alternative, assuming coverage for both first-line T-DM1 and salvage therapy with a pertuzumab-containing regimen, neither of which are consistent with their current FDA indications. In patients who progress on first-line THP and second-line T-DM1, third-line options range from combinations of nontaxane chemotherapeutic agents (vinorelbine, liposomal doxorubicin, gemcitabine, capecitabine, eribulin, ixabepilone, etc.) with trastuzumab, the combination of capecitabine and lapatinib, which may be especially appealing in patients with brain metastases, as lapatinib can penetrate the CNS, whereas trastuzumab does not, trastuzumab and lapatinib together, or a clinical trial of any of a number of novel HER2-targeted agents.
65 Combinations of endocrine therapy and HER2-targeted agents (such as anastrozole or tamoxifen with trastuzumab or letrozole with lapatinib) have demonstrated superior efficacy to endocrine therapy alone in patients with HRþ/ HER2þ cancers, but appear to be less effective than chemotherapy-based combinations, though a randomized study to test this has not been performed, and thus are generally reserved for older or frail patients, except when given to patients who responded to a chemotherapy-based combination as mentioned above. The addition of trastuzumab to adjuvant chemotherapy for HER2þ early stage breast cancer improves DFS by 40%-50% over chemotherapy alone. The first wave of HER adjuvant trails used anthracycline-containing regimens—AC followed by a taxane, with trastuzumab given concurrent with or sequentially after the taxane (AC-TH). BCIRG 006 demonstrated that either AC-TH or a non–anthracycline-containing regimen, docetaxel, carboplatin, and trastuzumab (TCH) was superior to AC-T alone.35 Although DFS and OS curves for the AC-TH arm tracked above those for the TCH arm, the study was not powered to compare these 2 regimens, and any modest advantages favoring AC-TH may be offset by a higher incidence of congestive heart failure rate (2% vs 0.4%) and a doubling of patients with asymptomatic declines in cardiac function (18.6% vs 9.4%). No other study has directly compared AC-TH with TCH in the adjuvant setting, but on studies where treating physicians were allowed to choose between the two, U.S. and European medical oncologists have “voted with their feet” in favor of TCH by a margin of 9:1. The BETH trial failed to demonstrate a benefit from the addition of the antiangiogenic antibody bevacizumab to adjuvant chemotherapy and trastuzumab, but confirmed excellent outcomes associated with the TCH regimen, with invasive DFS ¼ 92% (88% in node positive, 96% in node negative) at 38 months.36 Results from the APHINITY trial, which tests the addition of pertuzumab to either TCH or AC-TH in a largely node-positive population, are eagerly awaited to see if this already low recurrence rate can be reduced further. Although effective, both TCH and AC-TH may be too toxic to justify their use in lower risk HER2þ patients, especially those with T1a/bN0 cancers, even though their risk of recurrence without adjuvant treatment may be as high as 15%-20%. After concluding that a randomized study with a no-treatment control arm could not accrue, a single arm study was conducted in node-negative patients with tumors up to 3 cm given weekly paclitaxel and trastuzumab for 12 weeks followed by trastuzumab every 3 weeks to complete a year of treatment. At 3 years, DFS is 98.7%, leading this less toxic regimen to be widely adopted to treat these lower risk patients.37 Given the challenges of conducting studies in the adjuvant setting—large patient numbers and relatively long follow-up required, especially as results with standard therapy improve —researchers have turned to the neoadjuvant setting and the use of pCR as a surrogate end point for longer-term outcomes to assess the benefit of novel therapies and combinations. The validity of this end point is supported by results from the NOAH trial and the FDA-sponsored meta-analysis, both of
66 which demonstrated a correlation between pCR and superior event-free survival (EFS) in HER2þ patients.38,39 The NeoALTTO trial compared the combination of trastuzumab and lapatinib with either single agents administered for 6 weeks by themselves then for 12 weeks in combination with weekly paclitaxel. Dual HER2-targeted therapy was associated with a significant increase in the pCR rate (51.3% vs 29.5% for trastuzumab and 24.7% for lapatinib), and patients who achieved a pCR fared much better than those who did not (3-year EFS of 86% vs 72%, OS ¼ 94% vs 87%).40 However, there was no significant difference in long-term outcomes between patients who received dual HER-targeted therapy compared with singleagent trastuzumab (EFS ¼ 84% vs 76%), and when the much larger ALTTO adjuvant trial also failed to show a significant benefit for the addition of lapatinib to chemotherapy and trastuzumab,41 and that it was associated with increases in diarrhea, rash, and early treatment discontinuation, the question as to the role of lapatinib in early stage HER2þ cancers appears to have been settled. Fortunately, at around the same time, data on the benefit of adding pertuzumab to neoadjuvant chemotherapy and trastuzumab began to emerge. The randomized phase II NeoSphere trial demonstrated that the addition of pertuzumab to 4 cycles of docetaxel and trastuzumab increased the pCR rate from 29% (TH) to 46% (THP), whereas substitution of pertuzumab for trastuzumab reduced the pCR rate to 24%, and treatment with the 2 HER2-targeted antibodies alone yielded a pCR rate of 17%, demonstrating the power of HER2-blockade, especially in HR/HER2þ patients, in whom there was a pCR rate of 29%.42 All patients eventually received anthracycline-based adjuvant chemotherapy and completed a year of trastuzumab. The study was not powered to demonstrate a benefit in longterm outcomes, but patients who received THP had 5-year PFS of 86% compared with 81% for TH.43 In another randomized phase II study, TRYPHAENA, 3 different pertuzumabcontaining neoadjuvant regimens—FEC with or without concurrent trastuzumab and pertuzumab followed by THP or TCH with pertuzumab (TCHP)—demonstrated pCR rates exceeding 50%.44 On the basis of these 2 phase II studies, the FDA granted accelerated approval for the use of pertuzumab in the neoadjuvant setting in any of the regimens used in those trials. This approach has been widely adopted by the medical oncology community in patients with HER2þ cancers clinical stage IIA and higher, with TCHP chosen as the control neoadjuvant regimen for a number of randomized trials, and has increased the percentage of HER2þ patients being treated in the neoadjuvant setting, given that pertuzumab is not approved in the adjuvant setting, pending results from the APHINITY trial described above. A recently reported, moderated-sized (n¼130) randomized phase II neoadjuvant study in HRþ/HER2þ patients demonstrated a pCR rate of 40.5% with 4 cycles of T-DM1 alone and 45.8% with the combination of T-DM1 and endocrine therapy, rates that are higher for this patient subset than in NeoSphere, compared with only 6.7% with trastuzumab and endocrine therapy.45 Additional studies with this agent in the neoadjuvant setting are ongoing.
S. Witherby et al.
Triple-Negative Breast Cancer Triple-negative breast cancer (TNBC) accounts for 15%-20% of breast cancers in the United States, but is more common in younger women, African-Americans and Hispanics, and in carriers of deleterious mutations in the BRCA1 gene (Fig. 3). Although some degree of variability within this entity, defined by what it does not have, has long been appreciated—pure medullary and the rare adenoid cystic carcinomas of the breast do not express hormone receptors or HER2, but behave very differently from “classic” TNBC—over the past few years the extent of this heterogeneity has become more apparent. By one type of gene expression analysis, 70%-80% of TNBC express a “basal-like” pattern, whereas the other 20%-30% are a mix of luminal, HER2-enriched (despite the absence of expression of hormone receptors or HER2), “normal-like” and “claudin-low” subtypes.46 In another analysis, 6 different subtypes were identified—2 basal-like and others labeled immunomodulatory, mesenchymal, mesenchymal stem-like, and luminal androgen—all of which may have different biologic behaviors and different susceptibilities to treatment,47 but have not been validated for routine use in clinical practice. Patients with metastatic TNBC continue to have a poor prognosis, with mOS of less than a year. Although patients often initially respond to chemotherapy, duration of response is typically brief, and briefer still with subsequent lines of therapy. Combination chemotherapy may yield a higher ORR, and thus is useful in symptomatic patients, but does not change the overall prognosis. Based on promising results from a randomized phase II study48 there was hope that the addition of a PARP inhibitor, iniparib (BSI-201), to the combination of carboplatin and gemcitabine, chosen to maximize DNA damage, would significantly prolong survival in metastatic TNBC, but the larger phase III study was negative,49 and it was subsequently discovered that iniparib was actually not an effective inhibitor of PARP, and there is little reason to favor the carboplatin-gemcitabine combination over other options except perhaps in patients who relapse within months of completing anthracycline- and taxane-containing adjuvant chemotherapy, a group with a particularly poor prognosis given demonstrated chemotherapy-resistant disease. Given the similarities in gene expression patterns between BRCA1-mutated cancers and sporadic TNBC, and the activity reported for single-agent cisplatin in the neoadjuvant setting for BRCA1-associated breast cancers,50 there has been a great deal of interest in determining whether platinum analogs should replace other agents as first-line therapy for metastatic TNBC. A study reported at the 2014 SABCS compared single-agent carboplatin with docetaxel in metastatic TNBC and BRCAassociated cancers. Response rates, median PFS and mOS were similar, except in the small (n ¼ 43) cohort of BRCA-associated cancers, in which carboplatin had a decided advantage (ORR ¼ 68% vs 33%, mPFS ¼ 6.8 vs 4.8 months).51 In patients with stage I-III TNBC, except perhaps those with very small (T1a/b) node-negative cancers, in whom risk of recurrence and thus the role of adjuvant chemotherapy remain controversial, results of a series of clinical trials performed over the last 15-20 years have determined optimal adjuvant therapy
Advances in medical management of breast cancer: 2015
67
A Stage I-III
Stage I T1a/bN0
Surgery
If path stage unchanged OBS vs. adjuvant chemo TC or AC x4
B Stage IV
Stage I-IIA (T<3 cm)
Surgery, unless response could improve surgical options
Stage IIA (>3cm), IIB-III
NACT with ddAC + wP or ddP
Chemotherapy Single agent (anthracycline, taxane or other, depending on prior treatment; consider carboplatin if BRCA carrier) unless extensive or symptomatic disease (‘visceral crisis’) in which case combination (such as AC, A+Taxane, carboplatin/paclitaxel, carboplatin/gemcitabine, etc.) should be considered
NACT with ddAC + wP or ddP Adjuvant chemo with ddAC + wP or ddP
PD
Surgery
Salvage chemotherapy with noncross resistant agent, could add eribulin, ixabepilone, vinorelbine, capecitabine, gemcitabine, etc., to list
Abbreviations: OBS = observation; TC = docetaxel & cyclophosphamide; AC = doxorubicin & cyclophosphamide; NACT = neoadjuvant chemotherapy; ddAC = dose-dense AC; wP = weekly paclitaxel; ddP = dose-dense paclitaxel; PD = progressive disease
Figure 3 Treatment algorithm for triple-negative breast cancer (TNBC). AC, doxorubicin and cyclophosphamide; ddAC, dose-dense AC; ddP, dose-dense paclitaxel; NACT, neoadjuvant chemotherapy; OBS, observation; PD, progressive disease; TC, docetaxel and cyclophosphamide; wP, weekly paclitaxel. (Color version of figure is available online.)
to be a sequence of anthracycline-based chemotherapy such as dose-dense AC (ddAC) and paclitaxel administered either weekly or every 2 weeks. In recently updated results (median follow-up 12.1 years) from the Eastern Cooperative Oncology Group 1199 study, patients assigned to weekly paclitaxel had 31% improvements in both DFS and OS compared with every 3 week paclitaxel,52 whereas in SWOG 0221, which compared weekly vs every 2 weeks adjuvant chemotherapy, DFS and OS trends favored every 2 weeks paclitaxel 6 over weekly paclitaxel 12 in patients who received ddAC.53 Thus far, no additions to this regimen, including chemotherapeutic agents like gemcitabine or capecitabine or biologic agents such as bevacizumab, have been shown to improve outcomes. As in other breast cancer subtypes, investigators have turned to the neoadjuvant setting in the hope of more rapidly and efficiently identifying promising agents and regimens. The sequential regimens used in the adjuvant setting—weekly paclitaxel and ddAC—typically yield pCR rates of 30%-40% when administered to patients with stage II-III TNBC as neoadjuvant therapy, and there are compelling data from the FDA-sponsored metaanalysis as to the superior outcomes associated with achievement of a pCR in this subtype.38 In a number of neoadjuvant studies the addition of bevacizumab to a standard chemotherapy regimen increased the pCR rate,54-56 but thus far this has failed to demonstrate improvements in long-term outcomes.57,58 Coupled with its failure to improve DFS or OS in adjuvant trials,59,60 and the incidence of serious and sometimes life-threatening
toxicities associated with bevacizumab, this has dampened interest in further study of its role in early stage TNBC. In contrast, there is ongoing interest in the potential value of adding carboplatin. Significant increases in the pCR rates with the addition of carboplatin, with higher rates of hematologic toxicities, and dose modifications were demonstrated in 2 fairly large studies (315 and 443 patients, respectively).55,61 One of these studies also demonstrated higher rates of conversion from mastectomy-requiring to BCS-eligible and from node positive to pathologically node negative in patients assigned to carboplatin.55 However, until there is evidence that these higher pCR rates translate into improvements in long-term outcomes, adding carboplatin is unlikely to be accepted as the standard of care, though some oncologists in the United States and elsewhere are incorporating it in the neoadjuvant regimen, especially for patients with larger or locally advanced TNBC. Whether a PARP inhibitor will enhance the efficacy of the platinum analog in this setting is also being studied.62
Controversies and Promising Directions Immunotherapy As our understanding of the immune system and how cancers avoid its surveillance and control increases, we are just
S. Witherby et al.
68 beginning to harness its capacities to assist in our battle against malignancies. The uses of immune checkpoint inhibitors to circumvent ways in which cancers block immune recognition and attack have resulted in newly approved agents in metastatic melanoma and non–small cell lung cancer, and are being studied in a wide range of cancers including breast cancer. Studies presented at the 2014 SABCS and the 2015 AACR meetings demonstrated antitumor activity in patients with heavily pretreated metastatic TNBC for both the PD-1 inhibitor pembrolizumab and the PD-L1 inhibitor MPDL3280A.63,64 These and similar agents are being tested, alone and in combination with chemotherapy, in less heavily pretreated TNBC patients, including as adjuvant therapy in high-risk patients, such as those with significant residual disease after neoadjuvant chemotherapy, and will certainly be tested in other breast cancer subtypes as well. Within a few years, we should know if this approach can have a substantial effect on breast cancer recurrence, progression, and mortality.
Role of Bevacizumab and Other Antiangiogenic Agents As mentioned above, although the importance of neoangiogenesis in supporting the survival and spread of breast cancer is well supported, and there have been tantalizing hints of antitumor activity for bevacizumab when given in combination with chemotherapy in both the metastatic and neoadjuvant settings, this has not translated into clinically relevant improvements in long-term outcomes (recurrence-free survival or OS). But the questions remains—is there an identifiable subset of tumors in which antiangiogenic therapy could demonstrate more substantial and durable efficacy; and—is bevacizumab simply not the best choice for breadth and depth of antiangiogenic activity. Novel agents able to block multiple angiogenic pathways and thus prevent resistance to the relatively selective action of bevacizumab are being tested.
Genomic Assays—Ready for Prime Time? HER2-Targeted Therapy in HER2 “Negative” Patients As we do not have an accurate marker of HER2-dependence, it is likely that some of the patients currently received HER2targeted therapies do not benefit from these treatments and that some patients labeled HER2-negative would benefit from their inclusion. One hint of this came from the NSABP B-31 adjuvant trial, which included a small number of patients whose HER2-positive status, defined by a local lab, was not be confirmed on central testing. Despite being “HER2-negative” these patients derived as much benefit from the addition of trastuzumab to their adjuvant chemotherapy as patients who were HER2þ by standard criteria.65 To determine whether this was a chance observation or a biologically relevant phenomenon, NSABP conducted the B-47 trial, on which patients with HER2-1þ and HER2-2þ cancers without HER2 gene amplification by fluorescence in situ hybridization (FISH) were randomized to standard chemotherapy with or without trastuzumab. This study has completed accrual, and although we will likely not see results for several years, it could revolutionize our thinking about HER2-positivity and which patients may benefit from anti-HER2 therapy.
Endocrine Therapy in HRþ/HER2þ Patients In the neoadjuvant setting, pCR rates with neoadjuvant chemotherapy and HER2-targeted therapy are typically significantly lower in patients whose tumors are HRþ compared with those which are HR. Is this caused by biologic differences between the 2 subtypes—FISH ratios are typically higher in HR/HER2þ tumors, and higher FISH ratios are predictive of a greater likelihood of achieving pCR—or is “cross-talk” between the ER and the pathways activated by HER2 responsible for relative resistance to therapy? NSABP B-52 was designed to assess whether the addition of estrogen deprivation will significantly raise the pCR rate achieved with the TCHP neoadjuvant regimen.
The increased speed and decreased cost of performing whole exome sequencing has resulted in an explosion of interest in testing patients’ tumors to identify hitherto unsuspected driver mutations, which could potentially be targeted by agents already approved in other settings or by investigational agents. Although this technology is intriguing, and the testing often identifies a number of abnormalities in the cancer cells, the percentage of patients in whom a targetable mutation is detected is low, and the overall benefit of this still costly testing is unclear. Instead of paying for a commercial test off study, patients should be encouraged to have this type of testing done in the setting of a clinical trial that would not only cover the cost of the test but also help them gain access to investigational agents should the test identify a potential target. Such studies are being initiated by both the National Cancer Institute and American Society of Clinical Oncology.
Conclusions The last few years have witnessed major advances in our understanding of the biology of breast cancer that have led to promising new treatments for all the major subtypes of the disease, and to some success in tailoring or “personalizing” therapy to try to reduce toxicity without sacrificing efficacy. It is reasonable to expect that patient outcomes will continue to improve. However, we need to use the increasing sophistication of the tools at our disposal to analyze tumors, and to comb through the volume of data produced by those analyses, to identify biomarkers predictive of benefit, or lack thereof, from our novel therapies, given the toxicities, and sometimes staggering costs associated with them, especially when we seek to test agents associated with (often modest) improvements in outcomes in the metastatic setting in early stage disease.
References 1. Fisher B, Dignam J, Bryant J, et al: Five versus more than five years of tamoxifen for lymph node-negative breast cancer: Updated findings from
Advances in medical management of breast cancer: 2015
2.
3.
4.
5.
6.
7.
8.
9.
10.
11.
12.
13.
14.
15.
16.
the National Surgical Adjuvant Breast and Bowel Project B-14 randomized trial. J Natl Cancer Inst 93:684-690, 2001 Goss PE, Ingle JN, Martino S, et al: A randomized trial of letrozole in postmenopausal women after five years of tamoxifen therapy for earlystage breast cancer. N Engl J Med 349:1793-1802, 2003 Jin H, Tu D, Zhao N, et al: Longer-term outcomes of letrozole versus placebo after 5 years of tamoxifen in the NCIC CTG MA.17 trial: Analyses adjusting for treatment crossover. J Clin Oncol 30:718-721, 2012 Goss PE, Ingle JN, Martino S, et al: Impact of premenopausal status at breast cancer diagnosis in women entered on the placebo-controlled NCIC CTG MA17 trial of extended adjuvant letrozole. Ann Oncol 24:355-361, 2013 Davies C, Pan H, Godwin J, et al: Long-term effects of continuing adjuvant tamoxifen to 10 years versus stopping at 5 years after diagnosis of oestrogen receptor-positive breast cancer: ATLAS, a randomised trial. Lancet 381:805-816, 2013 Gray RG, Rea D, Handley K, et al.: aTTom: Long-term effects of continuing adjuvant tamoxifen to 10 years versus stopping at 5 years in 6,953 women with early breast cancer. Presented at the 49th Annual Meeting of the American Society of Clinical Oncology, Chicago, IL, May 31-June 4, 2013 Goss PE, Ingle JN, Pater JL, et al: Late extended adjuvant treatment with letrozole improves outcome in women with early-stage breast cancer who complete 5 years of tamoxifen. J Clin Oncol 26:1948-1955, 2008 Zdenkowski N, Green M, Boyle FM, et al: Final analysis of a randomized comparison of letrozole vs observation as late reintroduction of adjuvant endocrine therapy (AET) for postmenopausal women with hormone receptor positive breast cancer completion of prior AET: ANZBCTG 0501 (LATER). Proc Am Soc Clin Oncol 33:514, 2015. [abstract] Malamud SC, Schroeder B, Operana TN, et al: Predictive testing for selection of patients for extended endocrine therapy: Clinical utilization of Breast Cancer Index (BCI) in early-stage, ERþ, LN breast cancer. Proc Am Soc Clin Oncol 33:545, 2015. [abstract] Early Breast Cancer Trialists’ Collaborative Group (EBCTCG): Aromatase inhibitors versus tamoxifen in early breast cancer: Patient-level metaanalysis of the randomised trials. Lancet, 2015. http://dx.doi.org/10.1016/ S0140-6736(15)61074-1 Pagani O, Regan MR, Walley B, et al.: Randomized comparison of adjuvant aromatase inhibitor exemestane plus ovarian function suppression (OFS) vs tamoxifen plus OFS in premenopausal women with hormone receptor-positive early breast cancer: Joint analysis of IBCSG TEXT and SOFT trials. Presented at the 50th Annual Meeting of the American Society of Clinical Oncology, Chicago, IL, May 30-June 3, 2014 Francis PA, Regan MM, Fleming GF, et al.: Randomized comparison of adjuvant tamoxifen (T) plus ovarian function suppression (OFS) versus tamoxifen in premenopausal women with hormone receptor-positive (HRþ) early breast cancer (BC): Analysis of the SOFT trial. Presented at the 37th San Antonio Breast Cancer Symposium, San Antonio, TX, December 9-13, 2014 Ellis MJ, Suman VJ, Hoog J, et al: Randomized phase II neoadjuvant comparison between letrozole, anastrozole, and exemestane for postmenopausal women with estrogen receptor-rich stage 2 to 3 breast cancer: Clinical and biomarker outcomes and predictive value of the baseline PAM50-based intrinsic subtype–ACOSOG Z1031. J Clin Oncol 29:2342-2349, 2011 Early Breast Cancer Trialists’ Collaborative Group (EBCTCG): Adjuvant bisphosphonate treatment in early breast cancer: Meta-analyses of individual patient data from randomised trials. Lancet, 2015. http://dx. doi.org/10.1016/S0140-6736(15)60908-4 Gralow J, Barlow WE, Paterson AHG, et al.: Phase III trial of bisphosphonates as adjuvant therapy in primary breast cancer: SWOG/Alliance/ ECOG-ACRIN/NCIC Clinical Trials Group/NRG Oncology study S0307. Presented at the 51st Annual Meeting of the American Society of Clinical Oncology, Chicago, IL, May 29-June 2, 2015 Johnston SR, Kilburn LS, Ellis P, et al: Fulvestrant plus anastrozole or placebo versus exemestane alone after progression on non-steroidal aromatase inhibitors in postmenopausal patients with hormonereceptor-positive locally advanced or metastatic breast cancer (SoFEA):
69
17.
18.
19.
20.
21. 22.
23.
24.
25.
26.
27.
28. 29.
30.
31.
32. 33.
34.
A composite, multicentre, phase 3 randomised trial. Lancet Oncol 14:989-998, 2013 Chia S, Gradishar W, Mauriac L, et al: Double-blind, randomized placebo controlled trial of fulvestrant compared with exemestane after prior nonsteroidal aromatase inhibitor therapy in postmenopausal women with hormone receptor-positive, advanced breast cancer: Results from EFECT. J Clin Oncol 26:1664-1670, 2008 Di Leo A, Jerusalem G, Petruzelka L, et al: Final overall survival: Fulvestrant 500 mg vs 250 mg in the randomized CONFIRM trial. J Natl Cancer Inst 106:djt337, 2014. http://dx.doi.org/10.1093/jnci/djt337 Robertson JFR, Llombart-Cussac A, Rolski J, et al: Activity of fulvestrant 500 mg versus anastrozole 1 mg as first-line treatment for advanced breast cancer: Results from the FIRST study. J Clin Oncol 27:4530-4535, 2009 Robertson JF, Llombart-Cussac A, Feltl D, et al.: Fulvestrant 500 mg versus anastrozole as first-line treatment for advanced breast cancer: Overall survival from the phase II “FIRST” study. Presented at the 37th San Antonio Breast Cancer Symposium, San Antonio, TX, December 9-13, 2014 Mehta RS, Barlow WE, Albain KS, et al: Combination anastrozole and fulvestrant in metastatic breast cancer. N Engl J Med 367:435-444, 2012 Finn RS, Dering J, Conklin D, et al: PD 0332991, a selective cycle D kinase 4/6 inhibitor, preferentially inhibits proliferation of luminal estrogen receptor-positive human breast cancer cell lines in vitro. Breast Cancer Res 11:R77, 2009 Finn RS, Crown JP, Lang I, et al: The cyclin-dependent kinase 4/6 inhibitor palbociclib in combination with letrozole versus letrozole alone as first-line treatment of oestrogen receptor-positive, HER2-negative, advanced breast cancer (PALOMA-1/TRIO-18): A randomised phase 2 study. Lancet Oncol 16:25-35, 2015 Turner N, Ro J, Andre F, et al.: PALOMA3: A double-blind, phase III trial of fulvestrant with or without palbociclib in pre- and post-menopausal women with hormone receptor-positive, HER2-negative metastatic breast cancer that progressed on prior endocrine therapy. Presented at the 51st Annual Meeting of the American Society of Clinical Oncology, Chicago, IL, May 29-June 2, 2015 Piccart M, Hortobagyi GN, Campone M, et al: Everolimus plus exemestane for hormone-receptor-positive, human epidermal growth factor receptor-2-negative advanced breast cancer: Overall survival results from BOLERO-2. Ann Oncol 12:2357-2362, 2014 Klijn JG, Blamey RW, Boccardo F, et al: Combined tamoxifen and luteinizing hormone-releasing hormone (LHRH) agonist versus LHRH agonist alone in premenopausal advanced breast cancer: A meta-analysis of four randomized trials. J Clin Oncol 19:343-353, 2001 Baselga J, Gelmon KA, Verma S, et al: Phase II trial of pertuzumab and trastuzumab in patients with human epidermal growth factor receptor 2positive metastatic breast cancer that progressed during prior trastuzumab therapy. J Clin Oncol 28:1138-1144, 2010 Baselga J, Cortés J, Kim SB, et al: Pertuzumab plus trastuzumab plus docetaxel for metastatic breast cancer. N Engl J Med 366:109-119, 2012 Swain SM, Baselga J, Kim S-B, et al: Pertuzumab, trastuzumab, and docetaxel in HER2-positive metastatic breast cancer. N Engl J Med 372:724-734, 2015 Dang C, Iyengar N, Datko F, et al: Phase II study of paclitaxel given once per week along with trastuzumab and pertuzumab in patients with human epidermal growth factor receptor 2–positive metastatic breast cancer. J Clin Oncol 33:442-447, 2015 Krop IE, LoRusso P, Miller KD, et al: A phase II study of trastuzumab emtansine in patients with human epidermal growth factor receptor 2positive metastatic breast cancer who were previously treated with trastuzumab, lapatinib, an anthracycline, a taxane, and capecitabine. J Clin Oncol 30:3234-3241, 2012 Verma S, Miles D, Gianni L, et al: Trastuzumab emtansine for HER2positive advanced breast cancer. N Engl J Med 367:1783-1791, 2012 Krop IE, Kim S-B, Gonzalez-Martin A, et al: Trastuzumab emtansine versus treatment of physician's choice for pretreated HER2-positive advanced breast cancer (TH3RESA): A randomised, open-label, phase 3 trial. Lancet Oncol 15(33):689-699, 2014 Ellis PA, Barrios CH, Eiermann W, et al.: Phase III, randomized study of trastuzumab emtansine (T-DM1) ⫾ pertuzumab (P) vs trastuzumab þ taxane (HT) for first-line treatment of HER2-positive MBC: Primary results
S. Witherby et al.
70
35. 36.
37.
38.
39.
40.
41.
42.
43.
44.
45.
46. 47.
48.
49.
from the MARIANNE study. Presented at the 51st Annual Meeting of the American Society of Clinical Oncology, Chicago, IL, May 29-June 2, 2015 Slamon D, Eiermann W, Robert N, et al: Adjuvant trastuzumab in HER2positive breast cancer. N Engl J Med 365:1273-1283, 2011 Slamon DJ, Swain SM, Buyse M, et al.: Primary results from BETH, a phase 3 controlled study of adjuvant chemotherapy and trastuzumab ⫾ bevacizumab in patients with HER2-positive, node-positive or high risk node-negative breast cancer. Presented at the 36th San Antonio Breast Cancer Symposium, San Antonio, TX, December 10-14, 2013 Tolaney SM, Barry WT, Dang CT, et al: Adjuvant paclitaxel and trastuzumab for node-negative, HER2-positive breast cancer. N Engl J Med 372:134-141, 2015 Gianni L, Eiermann W, Semiglazov V, et al: Neoadjuvant and adjuvant trastuzumab in patients with HER2-positive locally advanced breast cancer (NOAH): Follow-up of a randomised controlled superiority trial with a parallel HER2-negative cohort. Lancet Oncol. 15:640-647, 2014 Cortazar P, Zhang L, Untch M, et al: Pathologic complete response and long-term clinical benefit in breast cancer: The CTNeoBC pooled analysis. Lancet 384:164-172, 2014 de Azambuja E, Holmes AP, Piccart-Gebhart M, et al: Lapatinib with trastuzumab for HER2-positive early breast cancer (NeoALTTO): survival outcomes of a randomised, open-label, multicentre, phase 3 trial and their association with pathological complete response. Lancet Oncol 15:1137-1146, 2014 Piccart-Gebhart MJ, Holmes AP, Baselga J, et al.: First results from the phase III ALTTO trial (BIG 2-06; NCCTG [Alliance] N063D) comparing one year of anti-HER2 therapy with lapatinib alone (L), trastuzumab alone (T), their sequence (T-L), or their combination (TþL) in the adjuvant treatment of HER2-positive early breast cancer (EBC). Presented at the 50th Annual Meeting of the American Society of Clinical Oncology, Chicago, IL, May 30-June 3, 2014 Gianni L, Pienkowski T, Im YH, et al: Efficacy and safety of neoadjuvant pertuzumab and trastuzumab in women with locally advanced, inflammatory, or early HER2-positive breast cancer (NeoSphere): A randomised multicentre, open-label, phase 2 trial. Lancet Oncol 15:640-647, 2014 Gianni L, Pienkowski T, Im YH, et al.: Five-year analysis of the phase II NeoSphere trial evaluating four cycles of neoadjuvant docetaxel and/or trastuzumab and/or pertuzumab. Presented at the 51st Annual Meeting of the American Society of Clinical Oncology, Chicago, IL, May 29-June 2, 2015 Schneeweiss A, Chia S, Hickish T, et al: Pertuzumab plus trastuzumab in combination with standard neoadjuvant anthracycline-containing and anthracycline-free chemotherapy regimens in patients with HER2-positive early breast cancer: A randomized phase II cardiac safety study (TRYPHAENA). Ann Oncol 24:2278-2284, 2013 Harbeck N, Gluz O, Christgen M, et al.: Efficacy of 12-weeks of neoadjuvant TDM1 with or without endocrine therapy in HER2positive hormone-receptor-positive early breast cancer: WSG-ADAPT HER2þ/HRþ phase II trial. Presented at the 51st Annual Meeting of the American Society of Clinical Oncology, Chicago, IL, May 29-June 2, 2015 Perou CM: Molecular stratification of triple-negative breast cancers. Oncologist 15:39-48, 2010 (suppl 5) Lehmann BD, Bauer JA, Chen X, et al: Identification of human triplenegative breast cancer subtypes and preclinical models for selection of targeted therapies. J Clin Invest 121:2750-2767, 2011 O'Shaughnessy J, Osborne C, Pippen JE, et al: Iniparib plus chemotherapy in metastatic triple-negative breast cancer. N Engl J Med 364:205-214, 2011 O'Shaughnessy J, Schwartzberg L, Danso MA, et al: Phase III study of iniparib plus gemcitabine and carboplatin versus gemcitabine and carboplatin in patients with metastatic triple-negative breast cancer. J Clin Oncol 32:3840-3847, 2014
50. Byrski T, Huzarski T, Dent R, et al: Pathologic complete response to neoadjuvant cisplatin in BRCA1-positive breast cancer patients. Breast Cancer Res Treat 147:401-405, 2014 51. Tutt A, Ellis P, Kilburn L, et al.: TNT: A randomized phase III trial of carboplatin compared with docetaxel for patients with metastatic or recurrent locally advanced triple negative or BRCA1/2 breast cancer (CRUK/07/012). Presented at the 37th Annual San Antonio Breast Cancer Symposium, San Antonio, TX, December 9-13, 2014 52. Sparano JA, Zhao F, Martino S, et al: Long-term follow-up of the E1199 phase III trial evaluating the role of taxane and schedule in operable breast cancer. J Clin Oncol 33:2353-2360, 2015 53. Budd GT, Barlow WE, Moor HCF, et al: SWOG S0221: A phase III trial comparing chemotherapy schedules in high-risk early-stage breast cancer. J Clin Oncol 33:58-64, 2015 54. von Minckwitz G, Eidtmann H, Rezai M, et al: Neoadjuvant chemotherapy and bevacizumab for HER2-negative breast cancer. N Engl J Med 366:299-309, 2012 55. Sikov WM, Berry DA, Perou CM, et al: Impact of the addition of carboplatin and/or bevacizumab to neoadjuvant weekly paclitaxel followed by dose-dense doxorubicin and cyclophosphamide on pathologic complete response rates in Stage II-III triple-negative breast cancer: CALGB 40603 (Alliance). J Clin Oncol 33:13-21, 2015 56. Earl HM, Hiller L, Dunn JA, et al: Efficacy of neoadjuvant bevacizumab added to docetaxel followed by fluorouracil, epirubicin, and cyclophosphamide, for women with HER2-negative early breast cancer (ARTemis): An open-label, randomised, phase 3 trial. Lancet Oncol 16:656-666, 2015 57. von Minckwitz G, Loibl S, Untch M, et al: Survival after neoadjuvant chemotherapy with or without bevacizumab or everolimus for HER2negative primary breast cancer (GBG 44-GeparQuinto). Ann Oncol 25:2363-2372, 2014 58. Bear HD, Tang G, Rastogi P, et al.: The effect on overall and disease-free survival (OS & DFS) by adding bevacizumab and/or antimetabolites to standard neoadjuvant chemotherapy: NSABP Protocol B-40. Presented at the 37th Annual San Antonio Breast Cancer Symposium, San Antonio, TX, December 9-13, 2014 59. Cameron D, Brown J, Dent R, et al: Adjuvant bevacizumab-containing therapy in triple-negative breast cancer (BEATRICE): Primary results of a randomised, phase 3 trial. Lancet Oncol 14:933-942, 2013 60. Miller K, O'Neill AM, Dang CT, et al.: Bevacizumab in the adjuvant treatment of HER2-negative breast cancer: Final results from Eastern Cooperative Oncology Group E5103. Presented at the 50th Annual Meeting of the American Society of Clinical Oncology, Chicago, IL, May 30-June 3, 2014 61. von Minckwitz G, Schneeweiss A, Loibl S, et al: Neoadjuvant carboplatin in patients with triple-negative and HER2-positive early breast cancer (GeparSixto; GBG 66): A randomised phase 2 trial. Lancet Oncol 15:747-756, 2014 62. Rugo HS, Olopade O, DeMichele A, et al.:Veliparib/carboplatin plus standard neoadjuvant therapy for high-risk breast cancer: First efficacy results from the I-SPY 2 TRIAL. Presented at the 36th Annual San Antonio Breast Cancer Symposium, San Antonio, TX, December 10-14, 2013 63. Nanda R, Chow LQ, Dees EC,et al.: A phase Ib study of pembrolizumab (MK-3475) in patients with advanced triple-negative breast cancer. Presented at the 37th Annual San Antonio Breast Cancer Symposium, San Antonio, TX, December 9-13, 2014 64. Emens LA, Braiteh FS, Cassier P, et al.: Inhibition of PD-L1 by MPDL3280A leads to clinical activity in patients with metastatic triplenegative breast cancer (TNBC). Presented at the American Association for Cancer Research Annual Meeting 2015, Philadelphia, PA, April 18-22, 2015 65. Pogue-Geile KL, Kim C, Jeong JH, et al: Predicting degree of benefit from adjuvant trastuzumab in NSABP trial B-31. J Natl Cancer Inst 105:1782-1788, 2013