Download Neoadjuvant Therapy for Early-Stage Breast

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts
no text concepts found
Transcript
Neoadjuvant Therapy for Early-Stage Breast Cancer: Current Pr
Published on Cancer Network (http://www.cancernetwork.com)
Neoadjuvant Therapy for Early-Stage Breast Cancer: Current
Practice, Controversies, and Future Directions
Review Article [1] | November 15, 2015 | Oncology Journal [2], Breast Cancer [3]
By Cesar Augusto Santa-Maria, MD [4], Melissa Camp, MD, MPH [5], Ashley Cimino-Mathews, MD [6],
Susan Harvey, MD [7], Jean Wright, MD [8], and Vered Stearns, MD [9]
In this review, we will discuss multidisciplinary considerations in treating patients with neoadjuvant
therapy and highlight areas of controversy and ongoing research.
Introduction
Surgical resection of breast cancer remains the cornerstone of therapy for patients diagnosed with
early-stage breast cancer. Systemic therapy can reduce the risk of relapse in patients with early
breast cancer by killing cancer cells that have escaped the breast or local lymph nodes. Generally,
systemic therapy administered either before or after surgery provides equal benefit in terms of
disease-free survival (DFS) and overall survival (OS) for patients with early-stage breast cancer.[1]
Administration of neoadjuvant therapy has benefits and limitations; therefore, patient selection is
crucial for optimizing outcomes. In this review, we will discuss multidisciplinary considerations in
treating patients with neoadjuvant therapy and highlight areas of controversy and ongoing research.
Neoadjuvant Therapy: Benefits, Indications, and Limitations
Benefits and indications
Historically, the primary indication for neoadjuvant therapy has been to facilitate breast surgery (see
Table).[2] Neoadjuvant chemotherapy has the potential to convert unresectable tumors to resectable
ones, and can reduce the extent of surgery needed to achieve adequate resection. For patients with
inflammatory breast cancer, neoadjuvant therapy is considered a standard of care (unless a
contraindication exists) and may confer a survival benefit in this population.[3] If patients are not
candidates for surgery at the time of diagnosis, neoadjuvant therapy can be used as a bridge to
operability in selected cases. Similarly, patients who want to undergo breast-conserving surgery
(BCS) but are not candidates for this approach at diagnosis may also be considered for neoadjuvant
therapy; among such patients, BCS rates as high as 72.3% (odds ratio [OR], 1.7 [95% confidence
interval (CI), 1.6–1.8]) have been reported.[4,5]
There are many other instances when neoadjuvant therapy may be considered (see Table). For
example, patients with chemoresponsive breast cancers may benefit from this treatment. Of all
clinical breast cancer subtypes, human epidermal growth factor receptor (HER) 2–positive breast
cancer and triple-negative breast cancer (TNBC) are the most chemosensitive, and therefore are
most amenable to neoadjuvant chemotherapy; indeed, patients with HER2-positive disease and
TNBC have the highest pathologic complete response (pCR) rates.[6] Patients with aggressive
estrogen receptor (ER)-positive breast cancer subtypes, or luminal B–like phenotype breast cancers,
tend to have higher pCR rates than those with more indolent ER-positive subtypes (pCR, 15% vs
7.5%), and so may be more suitable for neoadjuvant therapy.[7] Patients with classic invasive
lobular breast cancer tend to have inferior responses to neoadjuvant chemotherapy compared with
those who have invasive ductal carcinomas (pCR, 11% vs 25%; P = .01), and are therefore less likely
to benefit from neoadjuvant chemotherapy.[8] In patients with node-positive disease, for which
chemotherapy is the standard of care regardless of the sequencing of drug administration, response
to neoadjuvant treatment has a potential impact on locoregional management, including the extent
of axillary surgery and the radiation targets selected. In clinical research trials, the neoadjuvant
setting is useful for evaluation of the biologic and clinical effects of novel agents, and is thus a
powerful research tool. Importantly, because tissue is readily available both before and after therapy
in this setting, investigation of tissue-based biomarkers is logistically more feasible.
Page 1 of 16
Neoadjuvant Therapy for Early-Stage Breast Cancer: Current Pr
Published on Cancer Network (http://www.cancernetwork.com)
Limitations
Neoadjuvant chemotherapy is not recommended routinely for patients with stage I breast
cancer[9]—particularly if it is unclear whether to administer chemotherapy, or when third-generation
vs first- or second-generation chemotherapy regimens are being considered. In these cases, the final
surgical pathology report is often essential to making decisions about the need for chemotherapy
and the type of regimen. The extent of tumor as defined by focality may affect BCS rates after
neoadjuvant chemotherapy, when multifocal or multicentric tumors have lower chances of achieving
eligibility for BCS (unifocal, 71.6%; multifocal, 58.5%; multicentric, 30%).[10] Rates of pCR may also
differ slightly depending on tumor focality (unifocal, 19.4%; multifocal, 16.5%; multicentric, 14.4%).
Patients with multicentric rather than unifocal tumors may have inferior DFS (P < .001) and OS (P <
.009) after neoadjuvant chemotherapy; however, this disadvantage is abrogated if pCR is
achieved.[10]
There are insufficient data to support use of BCS after neoadjuvant chemotherapy in multicentric or
multifocal disease; therefore, cases should be considered on an individual basis.[2] For patients with
multicentric disease (ie, multiple tumors in different quadrants of the breast) at presentation,
mastectomy would be the standard of care regardless of response to neoadjuvant therapy. If
multifocal disease (ie, multiple distinct tumors in the same quadrant of the breast) is present, BCS
may become possible. It is important to bear in mind, however, that although the individual lesions
may decrease in size in response to neoadjuvant therapy, the distance between lesions remains the
same.
Progression during neoadjuvant chemotherapy is uncommon. While most patients are still able to
undergo surgery, studies have reported that up to 12% of patients who started neoadjuvant
chemotherapy were not able to undergo definitive surgery.[11] Even more uncommon, distant
metastasis has been described in 4% of patients who experience disease progression while receiving
neoadjuvant chemotherapy.[11] Factors that predict progression of disease during neoadjuvant
therapy include African-American race, advanced tumor stage, high nuclear grade, high Ki-67 levels,
and hormone receptor negativity.[12] Patients with breast cancer subtypes that are relatively less
chemosensitive, such as luminal A or classic invasive lobular breast cancer, may also be less likely to
benefit from neoadjuvant therapy.[7,8] These data underscore the importance of careful patient
selection and monitoring during treatment.
In the neoadjuvant setting, therapeutic decisions are largely based on clinical staging; this includes
physical examination and results obtained with imaging modalities, which may be less accurate than
surgical staging. Nodal staging, in particular, may affect radiation therapy options.[13]
Initial Assessment of the Patient
Breast evaluation
Locoregional assessment should be performed with careful physical examination and the use of
mammography and ultrasound as recommended by the National Comprehensive Cancer Network
(NCCN) guidelines.[9,14] Magnetic resonance imaging (MRI) of the breast is more sensitive in
determining the extent of the tumor but it may also overestimate the tumor size.[15] MRI may be
performed if there are other areas of concern on initial imaging that may warrant additional
evaluation.[16] A baseline MRI has been shown to decrease re-excision rates, and this procedure
also allows for assessment of the contralateral breast, where there is a 3% to 10% likelihood of
synchronous disease.[17,18] The decision to pursue an MRI should be tailored to each patient’s
specific clinical situation and needs, particularly if BCS is desired.
According to NCCN guidelines, image-guided core needle biopsy with placement of an
image-detectable marker of the breast abnormality is necessary to confirm the diagnosis; provide
tissue to assess pathologic markers, including ER, progesterone receptor (PR), and HER2 status; and
demarcate the tumor bed for post-neoadjuvant management.[9] Tumor grade and Ki-67 levels may
also be assessed, as these may estimate a more aggressive phenotype and may predict response to
chemotherapy. Other testing on tissue obtained by core needle biopsy, such as Oncotype DX or
other commercial genotyping, is not a standard of care in the preoperative setting at this time and
should not be obtained routinely outside the context of a clinical trial. Many biomarkers are currently
under development, including tumor-infiltrating lymphocytes (TILs), which have been associated with
higher likelihood of pCR in HER2-positive disease and TNBC, and the homologous recombination
deficiency score, which has been found to be an excellent predictor of pCR in patients with TNBC
receiving platinum-based neoadjuvant chemotherapy.[19-21]
Page 2 of 16
Neoadjuvant Therapy for Early-Stage Breast Cancer: Current Pr
Published on Cancer Network (http://www.cancernetwork.com)
Axillary evaluation
Patients being considered for neoadjuvant chemotherapy should undergo careful physical
examination of the axilla and review of mammography and ultrasonography.[16] If the clinical or
breast imaging results are notable for lymphadenopathy, a dedicated axillary ultrasound is
recommended for further evaluation. Fine needle aspiration (FNA) or core needle biopsy should be
performed on any suspicious-appearing lymph nodes (Figure). Core needle biopsy is more accurate,
particularly in invasive lobular carcinomas, and when feasible, it may be preferred to FNA; either
procedure is acceptable, however, according to the NCCN guidelines.[9,22] The placement of a
biopsy clip should be strongly considered when performing an FNA or core needle biopsy, because
use of the clip can improve the rate of successful surgical resection of biopsy-proven metastatic
axillary lymph nodes. This allows for more accurate assessment of pathologic response in the axilla,
and decreases the false-negative rate when performing a sentinel lymph node biopsy (SLNB) after
neoadjuvant chemotherapy in patients with known axillary nodal involvement prior to systemic
treatment.[23,24]
For patients with clinically node-negative breast cancer at diagnosis, the role of performing an SLNB
prior to or following neoadjuvant chemotherapy is controversial. In the SENTINA study, 35% of
clinically node-negative patients were found to have pathologically node-positive disease on SLNB
performed prior to neoadjuvant chemotherapy.[25] Among these patients, repeat SLNB after
neoadjuvant chemotherapy detected additional sentinel lymph nodes (SLNs) in 61% of patients, with
a high false-negative rate of 51.6%. Thus, SLNB performed after excision of a positive node prior to
neoadjuvant therapy has poor reliability.
In another study, compared with the same procedure performed prior to treatment, SLNB following
neoadjuvant chemotherapy resulted not only in lower SLN identification rates (98% vs 95%, before
vs after neoadjuvant therapy, respectively; P = .032), but also in less frequent axillary dissections or
radiation (45% vs 33%; P = .006); therefore, performing an SLNB after neoadjuvant therapy
potentially reduced morbidity.[26] Furthermore, one key limitation of SLNB prior to neoadjuvant
therapy is that removal of affected nodes prior to treatment precludes the ability to assess
pathologic response and decreases the significance of achieving a pCR.[27] Given that compelling
data on routine SLNB before neoadjuvant therapy are lacking, our preference is to perform SLNB
after neoadjuvant therapy in patients with a clinically node-negative axilla at presentation; however,
patient management in this setting must be individualized.
Evaluation of distant disease
Patients with stage I and II disease who are asymptomatic at presentation do not require routine
systemic imaging; however, patients with stage III disease should be considered for systemic
imaging even in the absence of symptoms, since they are at higher risk for harboring systemic
disease.[9] According to NCCN guidelines, however, routine systemic imaging in the absence of
symptoms is not indicated, but any patient experiencing systemic symptoms should undergo
laboratory testing and systemic staging as indicated.[9] Nuclear bone scan and computed
tomography (CT) scans with contrast of the chest, abdomen, and pelvis can adequately assess for
systemic disease, and should be considered as a first step unless a contraindication exists.
Additional imaging, including positron emission tomography (PET) scans and MRI, may be required in
select cases. Areas suspicious for distant metastasis should undergo biopsy to confirm metastatic
disease. Patients with central nervous system symptoms should undergo brain MRI.
Additional considerations
The treatment of breast cancer requires a multidisciplinary approach. Although initial evaluation may
be performed by a surgical or medical oncologist, there are several additional factors that should be
considered during the initial assessment. In complex cases, the use of a multidisciplinary tumor
board, in which pathologists; radiologists; and surgical, medical, and radiation oncologists review the
case, can facilitate the development of a treatment strategy. An early referral to radiation oncology
as part of the initial assessment should be recommended, particularly when clinical trials are
available or there are questions of disease burden, since response to treatment may impact
radiation planning. Early involvement of plastic surgery can aid in surgical planning, particularly as it
may pertain to cosmetic outcomes. Genetic counseling early in the process can also be helpful, since
this may inform final surgical treatment decisions. In cases of patients whose subsequent cancer risk
is affected by hereditary mutations, such as BRCA1 or BRCA2, the decision to perform mastectomy
and/or prophylactic mastectomy may be considered. In addition, the neoadjuvant setting also
Page 3 of 16
Neoadjuvant Therapy for Early-Stage Breast Cancer: Current Pr
Published on Cancer Network (http://www.cancernetwork.com)
provides numerous opportunities for research and clinical trial participation, and these opportunities
should be discussed with all eligible patients.
Systemic Therapy Considerations
Hormone receptor (HR)-positive breast cancer
Unless in the context of a clinical trial, patients with aggressive phenotype HR-positive breast cancer
who are candidates for chemotherapy should be treated with neoadjuvant chemotherapy rather than
endocrine therapy, since compared with indolent HR-positive subtypes they have superior rates of
response.[7] There are data, however, that suggest that neoadjuvant endocrine therapy may be
effective. Studies comparing neoadjuvant endocrine therapy with chemotherapy have shown similar
clinical response rates (48% to 67% vs 63% to 66%, respectively), and pCR rates were comparable
but low (0% to 3% vs 1% to 6%, respectively).[28,29] Of note, the majority of patients included in
these studies had ER and PR positivity and had grade 1/2 histology, suggesting that these may be
less aggressive in nature and more likely to be sensitive to endocrine therapy. Since these studies
demonstrated low pCR rates with chemotherapy in HR-positive breast cancer, primary surgery
should be considered, unless there is an indication for neoadjuvant therapy. Several studies
investigating neoadjuvant aromatase inhibitors (AIs) vs tamoxifen have also been conducted,
demonstrating either equal efficacy or superiority of AIs, with clinical response rates ranging from
38% to 70% for AIs and 36% to 51% for tamoxifen.[30-33] In a phase II study comparing different
AIs, clinical response rates were highest with letrozole (74.8% [95% CI, 66.3–82.1]) and anastrozole
(69.1% [95% CI, 60.1–77.1]), and lowest with exemestane (62.9% [95% CI, 53.8–71.4]); however,
future studies are needed to definitively demonstrate that one AI is superior to another.[34] The
duration of therapy is also an important factor, with several studies suggesting treatment duration in
excess of 3 to 4 months is optimal.[35-37] In patients who are unable to undergo surgery or receive
chemotherapy, neoadjuvant endocrine therapy may be an appropriate alternative. Assays that
reflect tumor biology may be useful in determining who is a good candidate for neoadjuvant
chemotherapy vs hormonal therapy.
HER2-positive breast cancer
The addition of the anti-HER2 monoclonal antibody trastuzumab to chemotherapy has resulted in an
increase in pCR rates, from 20% to 43% (relative risk [RR], 2.07 [95% CI, 1.41–3.03]; P = .0002), and
a decreased relapse rate, from 20% to 12% (RR, 0.67 [95% CI, 0.48–0.94]).[38] Furthermore,
event-free survival (EFS) and OS benefits (hazard ratio [HR], 0.64; P = .016 and HR, 0.66; P = .055,
respectively) have been demonstrated in the NOAH study, which was presented at the 2013
American Society of Clinical Oncology (ASCO) Annual Meeting.[39]
The success of lapatinib, a tyrosine kinase inhibitor (TKI) targeting the intracellular domain of HER2,
in the metastatic setting motivated several studies investigating this drug in the neoadjuvant
setting. Investigators from the European Organisation for Research and Treatment of Cancer
(EORTC) 10054 study, the National Surgical Adjuvant Breast and Bowel Project (NSABP) B-41 study,
the Cancer and Leukemia Group B (CALGB) 40601 study, and the Neoadjuvant Lapatinib and/or
Trastuzumab Treatment Optimization (NeoALTTO) study reported that the addition of lapatinib to
trastuzumab-based neoadjuvant chemotherapy improved pCR rates by 4% to 21%.[40-43]
NeoALTTO randomized patients to paclitaxel and either lapatinib, trastuzumab, or a combination of
lapatinib and trastuzumab. Following completion of chemotherapy, the patients underwent definitive
surgical treatment and then received adjuvant anthracycline-based chemotherapy. While this study
demonstrated a striking improvement in pCR rate with the addition of lapatinib to trastuzumab (pCR,
30% for the trastuzumab arm vs 51% for the trastuzumab/lapatinib arm), combination treatment was
not associated with improved EFS (HR, 0.78 [95% CI, 0.47–1.28]; P = .33) or OS (HR, 0.62 [95% CI,
0.30–1.25]; P = .19). The study was not powered to detect small differences in survival outcomes,
however.
The confirmatory Adjuvant Lapatinib and/or Trastuzumab Treatment Optimization (ALTTO) study,
presented at the 2014 ASCO Annual Meeting, failed to demonstrate that adding lapatinib to
trastuzumab-based chemotherapy improves DFS (HR, 0.84 [95% CI, 0.70–1.02]; P = .048, with P ≤
.025 needed to achieve statistical significance).[44] The discordant results between NeoALTTO and
ALTTO may be explained in part by the timing of the anthracycline-based therapy, since in
NeoALTTO this regimen was given after surgery, and therefore did not have an impact on pCR rates.
The findings inform future study designs, suggesting that all chemotherapy should be given upfront
Page 4 of 16
Neoadjuvant Therapy for Early-Stage Breast Cancer: Current Pr
Published on Cancer Network (http://www.cancernetwork.com)
before surgery. Given that there is no survival benefit to adding lapatinib to neoadjuvant
trastuzumab-based chemotherapy, at this time it is not considered a standard of care for patients
with early HER2-positive breast cancer, and should not be routinely prescribed except in the context
of a clinical trial.
Pertuzumab is a monoclonal antibody against HER2 and HER3. It has demonstrated a survival benefit
for patients with metastatic HER2-positive breast cancer, and has also been studied in the
neoadjuvant setting.[45] The NeoSphere trial found that the addition of pertuzumab to trastuzumab
and docetaxel improved pCR rates from 29% to 45.8%.[46] A recent update reported at the 2015
ASCO Annual Meeting suggested a trend for improved DFS, but this was not statistically significant
(HR, 0.60 [95% CI, 0.28–1.27]).[47] The TRYPHAENA study compared a combination regimen of
pertuzumab, trastuzumab, docetaxel, and carboplatin vs two other pertuzumab-based regimens
(both including epirubicin), and showed an unprecedented pCR rate of 66.2% in the
non–anthracycline-containing arm.[48] Based on the results of NeoSphere and TRYPHAENA, the US
Food and Drug Administration (FDA) granted approval for the use of pertuzumab in the neoadjuvant
setting.
The results of the APHINITY study will confirm whether the addition of pertuzumab improves survival
outcomes in early HER2-positive breast cancer. Until then, the strict approval for use of pertuzumab
in early breast cancer remains in the neoadjuvant setting, but the NCCN guidelines have made
provisional statements to also consider its use in the adjuvant setting.[9] The success of several
novel therapeutics in the metastatic setting has led to studies in the neoadjuvant setting. The effects
of neoadjuvant trastuzumab emtansine (T-DM1) with or without hormone therapy in HR-positive,
HER2-positive early breast cancer were reported at the 2015 ASCO Annual Meeting. Compared with
trastuzumab administered with endocrine therapy, pCR rates were substantially higher in the T-DM1
arms (6.7% vs 40.5% and 45.8%, respectively; P < .001).[49] The novel TKI neratinib, which
irreversibly inhibits HER2, was studied in the I-SPY 2 trial, in which it demonstrated overall pCR rates
of 32% (95% CI, 28–36); a larger registration trial is planned.[50]
TO PUT THAT INTO CONTEXT
Debu Tripathy, MD
The University of Texas MD Anderson Cancer Center
Houston, Texas
What Do We Already Know About Neoadjuvant Therapy for Breast Cancer?
Santa-Maria and colleagues review the evolving field of neoadjuvant therapy for breast cancer,
nicely addressing the very practical questions of why and how this approach should be considered.
They appropriately point out that neoadjuvant therapy should include adjuvant regimens proven to
significantly lengthen disease-free or overall survival. The additional advantage of neoadjuvant
therapy lies in its ability to downstage tumors and thereby enable selected patients to undergo
breast-conserving surgery. The neoadjuvant approach may even enable patients who present with
clinical N1 disease to avoid full axillary dissections, although this possibility is still being formally
studied. The most responsive breast cancer subtypes are triple-negative and human epidermal
growth factor receptor 2 (HER2)-positive (especially hormone receptor–negative) tumors, and in the
case of inflammatory breast cancer, neoadjuvant chemotherapy has become the standard approach
in the absence of controlled trials, because of the dismal historical results obtained with upfront
surgery.
What Holds Promise for the Future—And Why
One of the most interesting aspects of the neoadjuvant approach is the widening range of research
Page 5 of 16
Neoadjuvant Therapy for Early-Stage Breast Cancer: Current Pr
Published on Cancer Network (http://www.cancernetwork.com)
opportunities. Several investigative groups are using the neoadjuvant platform to accelerate the
development of relevant drugs and biomarkers/ signatures, given the strong link between pathologic
complete response and long-term survival. However, very careful stratification by tumor subtype is
necessary to confirm whether or not the neoadjuvant model can identify therapies with a high
chance of success at improving survival in a large-scale trial. While the US Food and Drug
Administration will consider this model as a pathway to accelerated drug approval, it must be
coupled with definitive trials showing a disease-free survival benefit. The authors provide some
examples of neoadjuvant research designs for triple-negative breast cancer, including the use of
carboplatin, that have generated much interest and even premature changes in practice patterns.
Accelerated approval of pertuzumab for neoadjuvant use in HER2-positive breast cancer, based on
the results of NeoSphere, was also heavily supported by the large body of evidence of a survival
benefit in the metastatic setting, as well as a favorable safety record. For platinum agents, the road
will be different, as two large cooperative groups have launched definitive trials of post-neoadjuvant
and adjuvant therapy for patients with early-stage breast cancer.
Triple-negative breast cancer
Although there are no approved drugs specifically indicated for TNBC, this subtype is associated with
relatively high pCR rates following chemotherapy, with many novel agents under investigation. The
addition of the anti–vascular endothelial growth factor receptor (VEGFR) monoclonal antibody
bevacizumab to chemotherapy has been studied extensively in the setting of neoadjuvant treatment
for breast cancer. The German Breast Group (GBG) 44 study and the Avastin Randomized Trial With
Neoadjuvant Chemotherapy for Patients With Early Breast Cancer (ARTemis) found that in TNBC
cohorts, the addition of bevacizumab improved the rates of ypT0N0 pCR by 11.4% (P = .003) and
14% (P = .03), respectively.[51,52] The recently presented Austrian Breast and Colorectal Cancer
Study Group (ABCSG) 32 study and the Southwestern Oncology Group (SWOG) S0800 study have
also reported improvements in pCR rates, especially in patients with TNBC.[53,54] The NSABP B-40
and CALGB 40603 studies found statistically significant improvements in ypT0Nx pCR in the breast
with the addition of bevacizumab, but differences in ypT0N0 rates were not statistically significant (P
= .08 and P = .057, respectively).[55,56] While these neoadjuvant studies demonstrated
improvements in pCR, three large randomized studies in multiple breast cancer subtypes in the
adjuvant setting have failed to demonstrate a survival advantage.[57-59] These data demonstrate
no role for bevacizumab at this time in unselected populations with early-stage breast cancer.
Platinum agents such as carboplatin have been investigated in the neoadjuvant
setting—administered at different dosing schedules and in a variety of combinations with other
agents—and have consistently been shown to improve pCR rates. The CALGB 40603 study found
rates of ypT0N0 pCR improved from 41% to 54% (P = .0029); in GeparSixto, pCR rates in the TNBC
cohort improved from 36.9% to 53.2% (P = .005); and in the I-SPY 2 study reported at the 2013 San
Antonio Breast Cancer Symposium, the estimated pCR rate with carboplatin/veliparib was
52%.[55,59,60] Notably, these studies used various doses and schedules of carboplatin, and in
combinations with non–standard-of-care agents such as bevacizumab and veliparib. The addition of
carboplatin is associated with significant myelosuppression and nausea. Grade 3/4 hematologic
adverse events ranged from 59% to 82% in the GeparSixto study.[59] In the CALGB 40603 study,
patients who received carboplatin were more likely to miss more than two doses of paclitaxel (36%
vs 16%). Additionally, 20% of patients did not receive all planned doses of anthracyclines and
taxanes, and many required dose reductions.[55] This is an important factor to consider, since
long-term survival data for anthracyclines and taxanes are robust.[61] In summary, given the
significant toxicity of carboplatin, its incorporation into the neoadjuvant management of breast
cancer requires additional studies, which should use consistent doses and schedules of carboplatin,
integrate it into standard treatment regimens, and provide long-term survival data. Furthermore,
adjuvant studies have been initiated to study the benefit of adding platinum drugs to treatment
regimens for early-stage TNBC.
Residual disease
Patients treated with neoadjuvant chemotherapy who have residual disease at the end of treatment
are at increased risk of recurrence. This relationship is particularly true of patients with TNBC (DFS
HR, 6.02 [95% CI, 3.92–9.25]; P < .001), HER2-positive disease (DFS HR, 8.74 [95% CI, 3.17–24.12];
P < .001), and luminal B breast cancer (DFS HR, 5.95 [95% CI, 1.46–24.25]; P = .013).[62] Because
of the complexities of assessing pathologic characteristics of residual disease, the Breast
Page 6 of 16
Neoadjuvant Therapy for Early-Stage Breast Cancer: Current Pr
Published on Cancer Network (http://www.cancernetwork.com)
International Group and the North American Breast Cancer Group have set forth guidelines that
focus on a multidisciplinary approach, rigorous pathologic sampling, use of biopsy clips, and the
consideration of receptor retesting when it may affect clinical management (ie, patients with initial
receptor-negative status).[27]
Receptor status has been demonstrated to change with neoadjuvant chemotherapy, and may be
associated with prognosis when there is loss of the ER.[63] In one series examining receptor status
after neoadjuvant therapy, 10.3% of patients with tumors that were ER-positive and HER2-negative
on core needle biopsy became ER-negative on excision, whereas 34.5% of patients with TNBC on
core needle biopsy became ER-positive at the time of surgical resection. This study is limited
because it did not report the change in percentage of ER labeling in tumors with discordant ER status
results, and the differences may reflect tumor heterogeneity, tissue sampling, or technical issues
with the assays. Nevertheless, changes did correspond with outcomes, as patients whose tumors
were ER-negative on excision had inferior recurrence-free survival (RFS; HR, 3.54 [95% CI,
1.60–7.85]), while patients whose tumors became ER-positive had a trend toward improved RFS (HR,
1.32 [95% CI, 0.64–2.74]).[63]
In addition to standard American Joint Committee on Cancer/Union for International Cancer Control
TNM staging, the Residual Cancer Burden (RCB) system can be used for patients who have residual
disease. The RCB system incorporates not only residual disease, but also the primary tumor size and
nodal burden, and it can be used to determine the risk of recurrence.[64] Biomarkers are under
development to further characterize patients with residual disease after neoadjuvant therapy. For
instance, the presence of TILs has been found to be a favorable prognostic factor for metastasis-free
survival and OS in patients with TNBC who have residual disease after neoadjuvant
chemotherapy.[65] Furthermore, biomarkers such as multigene assays may be useful in the
development of targeted therapies for patients with an increased risk of recurrence. Unfortunately,
there is no standard additional therapy at this time for patients who have residual disease after
neoadjuvant chemotherapy.
The KATHERINE study is investigating the role of T-DM1 in HER2-positive breast cancer with residual
disease (ClinicalTrials.gov identifier: NCT01772472). For TNBC patients with residual disease, the
role of cisplatin is being evaluated by investigators from the Eastern Cooperative Oncology Group
(ECOG)/American College of Radiology Imaging Network (ACRIN) (ClinicalTrials.gov identifier:
NCT02445391). A deeper molecular understanding of tumor biology is needed to help identify and
overcome mechanisms of resistance to standard therapy.
Interpreting Pathologic Complete Response
For patients who achieve a pCR, numerous data demonstrate that they are more likely to experience
a survival benefit when pCR is defined as ypT0/is ypN0.[66] Naturally, pCR achieved in a single
patient may not correlate to the pCR rate in a population of patients as a function of a therapy. To
date, only our experience with trastuzumab has demonstrated that improvements in pCR are linked
to improved survival; investigations of lapatinib and bevacizumab have failed to show this, and we
await results from the APHINITY trial to see if pertuzumab will demonstrate a correlation between
pCR and survival endpoints.[39,44,58,59] Similarly, studies are ongoing in the neoadjuvant and
adjuvant settings to investigate the role of carboplatin in early TNBC. Certainly the anticancer
potency of the drug under investigation matters, and perhaps the degree of change in pCR rate may
also matter. When considering neoadjuvant therapy, a clinician must weigh the potential benefits of
agents with no proven survival benefit against their potential toxicity. The impact of toxicity on the
ability of patients to complete standard-of-care therapy should also be considered. Furthermore,
clinicians should be cautious in extrapolating neoadjuvant data to the adjuvant setting in the
absence of survival data.
Surgical Considerations
Additional imaging following neoadjuvant therapy prior to surgery
In addition to clinical examination, imaging at the conclusion of neoadjuvant therapy can aid in the
assessment of treatment response. Particularly when the patient desires BCS, critical to surgical
decision making is the determination of whether neoadjuvant therapy has downstaged the tumor to
the point where lumpectomy is feasible. MRI of the breast has been shown to be more accurate than
mammography for evaluation of treatment response. Although the sensitivity of MRI is high (0.92),
the specificity tends to be lower (0.6).[67] Correlation of MRI performed after neoadjuvant
Page 7 of 16
Neoadjuvant Therapy for Early-Stage Breast Cancer: Current Pr
Published on Cancer Network (http://www.cancernetwork.com)
chemotherapy with pathology obtained from surgery has shown that MRI can both underestimate
and overestimate pathologic size, although the discrepancy may be particularly related to
HR-positive disease.[67-69] MRI alone, however, may not be adequate to assess for residual disease
after neoadjuvant therapy. In one study, MRI alone correctly predicted suitability for BCS in 88% of
patients, but MRI plus mammography improved this to 92%. Mammography after neoadjuvant
therapy is important to assess the patient for extensive residual calcifications that may preclude
BCS.[70] Close collaboration between surgery and radiology departments is crucial for assessing
response to neoadjuvant therapy and surgical planning.
Partial vs complete mastectomy
Surgery after neoadjuvant therapy can be performed safely despite the potential toxicities
associated with the therapy.[71] A primary objective of neoadjuvant therapy is to enhance surgical
options for patients. One key measure of this is a decrease in mastectomy rate, and studies have
found that mastectomies can be decreased by 16.6% to 27% after neoadjuvant therapy.[72,73]
Interestingly, as pCR rates have increased with the use of modern neoadjuvant regimens, rates of
BCS have not.[74,75] Reasons for this discrepancy may be related to tumor focality and centricity,
patient preference, and residual ductal carcinoma in situ.[2] While, in general, BCS can be performed
safely after neoadjuvant therapy, advanced nodal involvement, residual tumor > 2 cm, multifocal
residual disease, and lymphovascular invasion predict higher rates of locoregional and ipsilateral
breast tumor recurrence.[76]
Surgical axillary approach
The approach to initial evaluation of the axilla is discussed in an earlier section of this article. In
women with clinically negative axillary lymph nodes, SLNB after neoadjuvant therapy yields similar
identification rates compared with those of women who undergo primary surgery (97.4% vs 98.7%,
respectively); recurrence rates are statistically similar as well (1.2% vs 0.9%, respectively).[77]
Therefore, SLNB after neoadjuvant chemotherapy in patients with clinically negative lymph nodes at
diagnosis is acceptable and considered to be the standard of care.[78] The role of SLNB vs axillary
lymph node dissection in patients with clinically node-positive disease prior to neoadjuvant
chemotherapy is more controversial. The American College of Surgeons Oncology Group (ACOSOG)
Z1071 investigators found that in women with clinically node-positive breast cancer (cN1) who
received neoadjuvant chemotherapy and then underwent SLNB, the false-negative rate was 12.6%
(90% Bayesian credible interval, 9.85–16.05) when two or more SLNs were examined, and 9.1%
(95% CI, 5.6–13.7) when three or more SLNs were examined; notably, in this study, false-negative
rates > 10% were considered to be unacceptable.[79]
Additional analysis revealed that radiolabeled colloid alone or with blue dye, compared with blue dye
alone, led to improved SLN identification rates.[80] The SENTINA study found that in women with
clinically node-positive disease at diagnosis who converted to clinically node-negative status after
neoadjuvant chemotherapy, SLNB following neoadjuvant chemotherapy resulted in a false-negative
rate of 14.2% (95% CI, 9.9–19.4). If three or more lymph nodes were removed, the false-negative
rate dropped to 7.3%, and the addition of blue dye to radiocolloid decreased the false-negative rate
from 16% to 8.6%.[25] The Sentinel Node Biopsy Following Neoadjuvant Chemotherapy in Biopsy
Proven Node Positive Breast Cancer (SN FNAC) study also found that increasing the number of SLNs
removed decreased false-negative rates (with removal of one lymph node associated with a
false-negative rate of 18.2% vs a rate of 4.9% for removal of two lymph nodes), as did mandatory
use of immunohistochemistry and a broad definition of lymph node positivity with metastasis of any
size, including isolated tumor cells.[81] While the NCCN guidelines state that axillary lymph node
dissection should be performed in patients with clinically positive lymph nodes, the previously
discussed data suggest that SLNB after neoadjuvant therapy is a feasible option for patients with
initial node-positive disease when the procedure yields three or more lymph nodes, and dual
radiotracer and dye techniques are used.[9] Alternative approaches are under investigation—for
example, “targeted axillary dissection,” which includes SLNB in combination with targeted removal
of biopsy-proven positive lymph nodes that were clipped at the time of initial core needle biopsy or
FNA.[82] Furthermore, additional studies are needed to identify breast cancer subtypes for which
patients with biopsy-proven nodal disease at presentation may derive the most benefit from SLNB
after neoadjuvant therapy.
Radiation Considerations
Page 8 of 16
Neoadjuvant Therapy for Early-Stage Breast Cancer: Current Pr
Published on Cancer Network (http://www.cancernetwork.com)
For patients who undergo BCS after neoadjuvant therapy, radiation therapy to the breast is
considered a standard of care whether or not pCR is achieved, as it is for patients who undergo
upfront surgery.[9,83] For patients who undergo mastectomy, the roles of postmastectomy radiation
therapy (PMRT) and regional nodal irradiation (RNI) are more controversial, particularly among those
who achieve pCR. NCCN guidelines simply state that radiation therapy should be based on the worst
stage either prior to or after neoadjuvant therapy.[9] Patients who have negative lymph nodes prior
to and following neoadjuvant therapy generally do not require PMRT or RNI, and conversely those
with residual node-positive disease are at higher risk of locoregional recurrence (LRR) and require
PMRT/RNI.[84] However, studies evaluating the risk of LRR after neoadjuvant therapy have
consistently demonstrated a lower risk of LRR in patients who achieve pCR compared with those who
do not.[85-88] Subtype of breast cancer may additionally influence LRR rates in those achieving pCR,
with TNBC generally associated with a higher LRR rate.[89] A pooled analysis of the NSABP B-18 and
B-27 studies demonstrated low rates of LRR in patients with pCR who had mastectomy without
PMRT; in these studies, the 10-year risk of LRR was < 10% in patients with pCR, regardless of initial
nodal status or tumor size, suggesting that PMRT may not be needed in these patients.[87]
Conversely, a recent meta-analysis of patients treated in the German Gepar trials, presented at the
2015 ASCO Annual Meeting, found that among patients who achieved pCR, the use of PMRT was an
independent prognostic factor for locoregional relapse–free survival (HR, 0.54 [95% CI, 0.035–0.82];
P = .004) and DFS (HR, 0.69 [95% CI, 0.51–0.93]; P = .016).[90] To address this conundrum, the
Radiation Therapy Oncology Group (RTOG) and the NSABP have designed the randomized phase III
NSABP B-51/RTOG 1304 trial (ClinicalTrials.gov identifier: NCT01872975) for patients who have
positive axillary nodes before neoadjuvant chemotherapy but convert to pathologically negative
axillary nodes. Patients will be randomized to PMRT with RNI vs no radiation in those who had
mastectomy, and breast irradiation with or without RNI in those who have undergone BCS. This study
will help to determine which patients can safely have PMRT and/or RNI omitted in the setting of nodal
pCR.[91] In summary, at this time there are no prospective data to guide clinicians regarding use of
radiation after neoadjuvant chemotherapy in the setting of pCR. Outside the setting of a clinical trial,
the current NCCN guidelines recommend strong consideration of PMRT or RNI in patients who have
positive lymph nodes either before or after neoadjuvant chemotherapy; treatment decisions should
be made on a case-by-case basis (Figure).[84]
Conclusions
In carefully selected patients, neoadjuvant therapy can provide improved surgical outcomes,
prognostic information, and access to clinical trials investigating novel agents and locoregional
therapies. Management involves a multidisciplinary team that should include surgical, medical, and
radiation oncologists; pathologists; radiologists; and, in certain cases, plastic surgeons, genetic
counselors, and research staff. (A suggested algorithm, based on data presented in this review
article, is provided in the Figure.) Since optimization of systemic and local therapies continues to be
an evolving field, participation in a clinical trial should be considered for eligible patients.
Financial Disclosure: Dr. Stearns receives research funding from AbbVie, Celgene, MedImmune,
Merck, Novartis, Pfizer, and Puma. The other authors have no significant financial relationship with
the manufacturer of any product or provider of any service mentioned in this article.
Oncology (Williston Park). 29(11):828-838.
Table. Indications, Considerations, and Relative
Contraindications for...
Page 9 of 16
Neoadjuvant Therapy for Early-Stage Breast Cancer: Current Pr
Published on Cancer Network (http://www.cancernetwork.com)
Figure: Algorithm Describing Approach to
Neoadjuvant Therapy
References:
1. Mauri D, Pavlidis N, Ioannidis JP. Neoadjuvant versus adjuvant systemic treatment in breast
cancer: a meta-analysis. J Natl Cancer Inst. 2005;97:188-94.
2. King TA, Morrow M. Surgical issues in patients with breast cancer receiving neoadjuvant
chemotherapy. Nat Rev Clin Oncol. 2015;12:335-43.
3. Perez CA, Fields JN, Fracasso PM, et al. Management of locally advanced carcinoma of the breast.
II. Inflammatory carcinoma. Cancer. 1994;74:466-76.
4. Barranger E, Antomarchi J, Chamorey E, et al. Effect of neoadjuvant chemotherapy on the surgical
treatment of patients with locally advanced breast cancer requiring initial mastectomy. Clin Breast
Cancer. 2015;15:e231-e235.
5. Killelea BK, Yang VQ, Mougalian S, et al. Neoadjuvant chemotherapy for breast cancer increases
the rate of breast conservation: results from the National Cancer Database. J Am Coll Surg.
2015;220:1063-9.
6. Connolly RM, Stearns V. Current approaches for neoadjuvant chemotherapy in breast cancer. Eur J
Pharmacol. 2013;717:58-66.
7. Bonnefoi H, Litiere S, Piccart M, et al. Pathological complete response after neoadjuvant
chemotherapy is an independent predictive factor irrespective of simplified breast cancer intrinsic
subtypes: a landmark and two-step approach analyses from the EORTC 10994/BIG 1-00 phase III
trial. Ann Oncol. 2014;25:1128-36.
8. Lips EH, Mukhtar RA, Yau C, et al. Lobular histology and response to neoadjuvant chemotherapy in
invasive breast cancer. Breast Cancer Res Treat. 2012;136:35-43.
9. Sikov WM, Berry DA, Perou CM, et al. Impact of the addition of carboplatin and/or bevacizumab to
neoadjuvant once-per-week paclitaxel followed by dose-dense doxorubicin and cyclophosphamide on
pathologic complete response rates in stage II to III triple-negative breast cancer: CALGB 40603
(Alliance). J Clin Oncol. 2015;33:13-21.
10. Ataseven B, Lederer B, Blohmer JU, et al. Impact of multifocal or multicentric disease on surgery
and locoregional, distant and overall survival of 6,134 breast cancer patients treated with
neoadjuvant chemotherapy. Ann Surg Oncol. 2015;22:1118-27.
11. Caudle AS, Gonzalez-Angulo AM, Hunt KK, et al. Impact of progression during neoadjuvant
chemotherapy on surgical management of breast cancer. Ann Surg Oncol. 2011;18:932-8.
12. Caudle AS, Gonzalez-Angulo AM, Hunt KK, et al. Predictors of tumor progression during
neoadjuvant chemotherapy in breast cancer. J Clin Oncol. 2010;28:1821-8.
13. Feng Y, Huang R, He Y, et al. Efficacy of physical examination, ultrasound, and ultrasound
combined with fine-needle aspiration for axilla staging of primary breast cancer. Breast Cancer Res
Treat. 2015;149:761-5.
14. Cocconi G, Di Blasio B, Alberti G, et al. Problems in evaluating response of primary breast cancer
to systemic therapy. Breast Cancer Res Treat. 1984;4:309-13.
15. Berg WA, Gutierrez L, NessAiver MS, et al. Diagnostic accuracy of mammography, clinical
Page 10 of 16
Neoadjuvant Therapy for Early-Stage Breast Cancer: Current Pr
Published on Cancer Network (http://www.cancernetwork.com)
examination, US, and MR imaging in preoperative assessment of breast cancer. Radiology.
2004;233:830-49.
16. Dialani V, Chadashvili T, Slanetz PJ. Role of imaging in neoadjuvant therapy for breast cancer.
Ann Surg Oncol. 2015;22:1416-24.
17. Lehman CD, Gatsonis C, Kuhl CK, et al. MRI evaluation of the contralateral breast in women with
recently diagnosed breast cancer. N Engl J Med. 2007;356:1295-303.
18. Sung JS, Li J, Da Costa G, et al. Preoperative breast MRI for early-stage breast cancer: effect on
surgical and long-term outcomes. AJR Am J Roentgenol. 2014;202:1376-82.
19. Mayer EL, Vaz-Luis I, Richardson AL, et al. TBCRC030: A randomized phase II study of
preoperative cisplatin versus paclitaxel in patients (pts) with BRCA1/2-proficient triple-negative
breast cancer (TNBC)—evaluating the homologous recombination deficiency (HRD) biomarker. J Clin
Oncol. 2014;32(suppl 5s):abstr TPS1145.
20. Denkert C, Loibl S, Noske A, et al. Tumor-associated lymphocytes as an independent predictor of
response to neoadjuvant chemotherapy in breast cancer. J Clin Oncol. 2010;28:105-13.
21. Mao Y, Qu Q, Zhang Y, et al. The value of tumor infiltrating lymphocytes (TILs) for predicting
response to neoadjuvant chemotherapy in breast cancer: a systematic review and meta-analysis.
PLoS One. 2014;9:e115103.
22. Ohashi R, Matsubara M, Watarai Y, et al. Diagnostic value of fine needle aspiration and core
needle biopsy in special types of breast cancer. Breast Cancer. 2015 Jul 2. [Epub ahead of print]
23. Plecha D, Bai S, Patterson H, et al. Improving the accuracy of axillary lymph node surgery in
breast cancer with ultrasound-guided wire localization of biopsy proven metastatic lymph nodes. Ann
Surg Oncol. 2015 Mar 27. [Epub ahead of print]
24. Braeuning MP, Burke ET, Pisano ED. Embolization coils as tumor markers for mammography in
patients undergoing neoadjuvant chemotherapy for carcinoma of the breast. AJR Am J Roentgenol.
2000;174:251-2.
25. Kuehn T, Bauerfeind I, Fehm T, et al. Sentinel-lymph-node biopsy in patients with breast cancer
before and after neoadjuvant chemotherapy (SENTINA): a prospective, multicentre cohort study.
Lancet Oncol. 2013;14:609-18.
26. van der Heiden-van der Loo M, de Munck L, Sonke GS, et al. Population based study on sentinel
node biopsy before or after neoadjuvant chemotherapy in clinically node negative breast cancer
patients: identification rate and influence on axillary treatment. Eur J Cancer. 2015;51:915-21.
27. Bossuyt V, Provenzano E, Symmans WF, et al. Recommendations for standardized pathological
characterization of residual disease for neoadjuvant clinical trials of breast cancer by the BIG-NABCG
collaboration. Ann Oncol. 2015;26:1280-91.
28. Semiglazov VF, Semiglazov VV, Dashyan GA, et al. Phase 2 randomized trial of primary
endocrine therapy versus chemotherapy in postmenopausal patients with estrogen receptor-positive
breast cancer. Cancer. 2007;110:244-54.
29. Alba E, Calvo L, Albanell J, et al. Chemotherapy (CT) and hormonotherapy (HT) as neoadjuvant
treatment in luminal breast cancer patients: results from the GEICAM/2006-03, a multicenter,
randomized, phase-II study. Ann Oncol. 2012;23:3069-74.
30. Cataliotti L, Buzdar AU, Noguchi S, et al. Comparison of anastrozole versus tamoxifen as
preoperative therapy in postmenopausal women with hormone receptor-positive breast cancer: the
Pre-Operative “Arimidex” Compared to Tamoxifen (PROACT) trial. Cancer. 2006;106:2095-103.
Page 11 of 16
Neoadjuvant Therapy for Early-Stage Breast Cancer: Current Pr
Published on Cancer Network (http://www.cancernetwork.com)
31. Smith IE, Dowsett M, Ebbs SR, et al. Neoadjuvant treatment of postmenopausal breast cancer
with anastrozole, tamoxifen, or both in combination: the Immediate Preoperative Anastrozole,
Tamoxifen, or Combined with Tamoxifen (IMPACT) multicenter double-blind randomized trial. J Clin
Oncol. 2005;23:5108-16.
32. Eiermann W, Paepke S, Appfelstaedt J, et al. Preoperative treatment of postmenopausal breast
cancer patients with letrozole: a randomized double-blind multicenter study. Ann Oncol.
2001;12:1527-32.
33. Masuda N, Sagara Y, Kinoshita T, et al. Neoadjuvant anastrozole versus tamoxifen in patients
receiving goserelin for premenopausal breast cancer (STAGE): a double-blind, randomised phase 3
trial. Lancet Oncol. 2012;13:345-52.
34. 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. 2011;29:2342-9.
35. Llombart-Cussac A, Guerrero A, Galan A, et al. Phase II trial with letrozole to maximum response
as primary systemic therapy in postmenopausal patients with ER/PgR[+] operable breast cancer. Clin
Transl Oncol. 2012;14:125-31.
36. Hille U, Soergel P, Länger F, et al. Aromatase inhibitors as solely treatment in postmenopausal
breast cancer patients. Breast J. 2012;18:145-50.
37. Dixon JM, Renshaw L, Macaskill EJ, et al. Increase in response rate by prolonged treatment with
neoadjuvant letrozole. Breast Cancer Res Treat. 2009;113:145-51.
38. Petrelli F, Borgonovo K, Cabiddu M, et al. Neoadjuvant chemotherapy and concomitant
trastuzumab in breast cancer: a pooled analysis of two randomized trials. Anticancer Drugs.
2011;22:128-35.
39. Gianni L, Eiermann W, Semiglazov V, et al. Follow-up results of NOAH, a randomized phase III
trial evaluating neoadjuvant chemotherapy with trastuzumab (CT+H) followed by adjuvant H versus
CT alone, in patients with HER2-positive locally advanced breast cancer. J Clin Oncol.
2013;31(suppl):abstr 503.
40. Bonnefoi H, Jacot W, Saghatchian M, et al. Neoadjuvant treatment with docetaxel plus lapatinib,
trastuzumab, or both followed by an anthracycline-based chemotherapy in HER2-positive breast
cancer: results of the randomised phase II EORTC 10054 study. Ann Oncol. 2015;26:325-32.
41. Robidoux A, Tang G, Rastogi P, et al. Lapatinib as a component of neoadjuvant therapy for
HER2-positive operable breast cancer (NSABP protocol B-41): an open-label, randomised phase 3
trial. Lancet Oncol. 2013;14:1183-92.
42. Carey LA, Berry DA, Ollila D, et al. Clinical and translational results of CALGB 40601: a
neoadjuvant phase III trial of weekly paclitaxel and trastuzumab with or without lapatinib for
HER2-positive breast cancer. J Clin Oncol. 2013;31(suppl):abstr 500.
43. 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.
2014;15:1137-46.
44. 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 (T+L) in the adjuvant treatment of
HER2-positive early breast cancer (EBC). J Clin Oncol. 2014;32(suppl 5s):abstr LBA4.
Page 12 of 16
Neoadjuvant Therapy for Early-Stage Breast Cancer: Current Pr
Published on Cancer Network (http://www.cancernetwork.com)
45. Swain SM, Baselga J, Kim SB, et al. Pertuzumab, trastuzumab, and docetaxel in HER2-positive
metastatic breast cancer. N Engl J Med. 2015;372:724-34.
46. 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. 2012;13:25-32.
47. Gianni L, Pienkowski T, Im YH, et al. Five-year analysis of the phase II NeoSphere trial evaluating
four cycles of neoadjuvant docetaxel (D) and/or trastuzumab (T) and/or pertuzumab (P). J Clin Oncol.
2015;33(suppl):abstr 505.
48. 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. 2013;24:2278-84.
49. Harbeck N, Gluz O, Christgen M, et al. Efficacy of 12-weeks of neoadjuvant TDM1 with or without
endocrine therapy in HER2-positive hormone-receptor-positive early breast cancer: WSG-ADAPT
HER2+/HR+ phase II trial. J Clin Oncol. 2015;33(suppl):abstr 506.
50. Park JW, Liu MC, Yee D, et al. Neratinib plus standard neoadjuvant therapy for high-risk breast
cancer: efficacy results for the I-SPY 2 trial. Cancer Res. 2014;74:abstr CT227.
51. Gerber B, Loibl S, Eidtmann H, et al. Neoadjuvant bevacizumab and anthracycline-taxane-based
chemotherapy in 678 triple-negative primary breast cancers; results from the GeparQuinto study
(GBG 44). Ann Oncol. 2013;24:2978-84.
52. 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. 2015;16:656-66.
53. Steger GG, Greil R, Hubalek M, et al. Bevacizumab in combination with docetaxel+trastuzumab
+/– non-pegylated liposomal doxorubicin: final results of ABCSG-32, a prospective, randomized
phase II-study. Cancer Res. 2015;75:abstr P3-11-06.
54. Nahleh ZA, Barlow WE, Hayes DF, et al. S0800: Nab-paclitaxel, doxorubicin, cyclophosphamide,
and pegfilgrastim with or without bevacizumab in treating women with inflammatory or locally
advanced breast cancer. Cancer Res. 2015;75:abstr P3-11-16.
55. Sikov WM, Berry DA, Perou CM, et al. Impact of the addition of carboplatin and/or bevacizumab
to neoadjuvant once-per-week paclitaxel followed by dose-dense doxorubicin and cyclophosphamide
on pathologic complete response rates in stage II to III triple-negative breast cancer: CALGB 40603
(Alliance). J Clin Oncol. 2015;33:13-21.
56. Bear HD, Tang G, Rastogi P, et al. Bevacizumab added to neoadjuvant chemotherapy for breast
cancer. N Engl J Med. 2012;366:310-20.
57. 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.
2013;14:933-42.
58. Miller K, O’Neill AM, Dang CT, et al. Bevacizumab (Bv) in the adjuvant treatment of
HER2-negative breast cancer: final results from Eastern Cooperative Oncology Group E5103. J Clin
Oncol. 2014;32(suppl 5s):abstr 500.
59. 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. 2014;15:747-56.
Page 13 of 16
Neoadjuvant Therapy for Early-Stage Breast Cancer: Current Pr
Published on Cancer Network (http://www.cancernetwork.com)
60. 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. Cancer Res. 2013;73:abstr
S5-02.
61. Early Breast Cancer Trialists’ Collaborative Group (EBCTCG); Peto R, Davies C, Godwin J, et al.
Comparisons between different polychemotherapy regimens for early breast cancer: meta-analyses
of long-term outcome among 100,000 women in 123 randomised trials. Lancet. 2012;379:432-44.
62. von Minckwitz G, Untch M, Blohmer JU, et al. Definition and impact of pathologic complete
response on prognosis after neoadjuvant chemotherapy in various intrinsic breast cancer subtypes. J
Clin Oncol. 2012;30:1796-804.
63. Lim SK, Lee MH, Park IH, et al. Impact of molecular subtype conversion of breast cancers after
neoadjuvant chemotherapy on clinical outcome. Cancer Res Treat. 2015 Apr 7. [Epub ahead of print]
64. Symmans WF, Peintinger F, Hatzis C, et al. Measurement of residual breast cancer burden to
predict survival after neoadjuvant chemotherapy. J Clin Oncol. 2007;25:4414-22.
65. Dieci MV, Criscitiello C, Goubar A, et al. Prognostic value of tumor-infiltrating lymphocytes on
residual disease after primary chemotherapy for triple-negative breast cancer: a retrospective
multicenter study. Ann Oncol. 2015;26:1518.
66. Cortazar P, Zhang L, Untch M, et al. Pathological complete response and long-term clinical
benefit in breast cancer: the CTNeoBC pooled analysis. Lancet. 2014;384:164-72.
67. Marinovich ML, Houssami N, Macaskill P, et al. Meta-analysis of magnetic resonance imaging in
detecting residual breast cancer after neoadjuvant therapy. J Natl Cancer Inst. 2013;105:321-33.
68. Straver ME, Loo CE, Rutgers EJ, et al. MRI-model to guide the surgical treatment in breast cancer
patients after neoadjuvant chemotherapy. Ann Surg. 2010;251:701-7.
69. McGuire KP, Toro-Burguete J, Dang H, et al. MRI staging after neoadjuvant chemotherapy for
breast cancer: does tumor biology affect accuracy? Ann Surg Oncol. 2011;18:3149-54.
70. Jochelson MS, Lampen-Sachar K, Gibbons G, et al. Do MRI and mammography reliably identify
candidates for breast conservation after neoadjuvant chemotherapy? Ann Surg Oncol.
2015;22:1490-5.
71. Abt NB, Flores JM, Baltodano PA, et al. Neoadjuvant chemotherapy and short-term morbidity in
patients undergoing mastectomy with and without breast reconstruction. JAMA Surg.
2014;149:1068-76.
72. Mieog JS, van der Hage JA, van de Velde CJ. Neoadjuvant chemotherapy for operable breast
cancer. Br J Surg. 2007;94:1189-200.
73. Fisher B, Brown A, Mamounas E, et al. Effect of preoperative chemotherapy on local-regional
disease in women with operable breast cancer: findings from National Surgical Adjuvant Breast and
Bowel Project B-18. J Clin Oncol. 1997;15:2483-93.
74. Bear HD, Anderson S, Brown A, et al. The effect on tumor response of adding sequential
preoperative docetaxel to preoperative doxorubicin and cyclophosphamide: preliminary results from
National Surgical Adjuvant Breast and Bowel Project Protocol B-27. J Clin Oncol. 2003;21:4165-74.
75. Baselga J, Bradbury I, Eidtmann H, et al. Lapatinib with trastuzumab for HER2-positive early
breast cancer (NeoALTTO): a randomised, open-label, multicentre, phase 3 trial. Lancet.
2012;379:633-40.
76. Chen AM, Meric-Bernstam F, Hunt KK, et al. Breast conservation after neoadjuvant
chemotherapy: the MD Anderson cancer center experience. J Clin Oncol. 2004;22:2303-12.
Page 14 of 16
Neoadjuvant Therapy for Early-Stage Breast Cancer: Current Pr
Published on Cancer Network (http://www.cancernetwork.com)
77. Hunt KK, Yi M, Mittendorf EA, et al. Sentinel lymph node surgery after neoadjuvant
chemotherapy is accurate and reduces the need for axillary dissection in breast cancer patients. Ann
Surg. 2009;250:558-66.
78. Lyman GH, Giuliano AE, Somerfield MR, et al. American Society of Clinical Oncology guideline
recommendations for sentinel lymph node biopsy in early-stage breast cancer. J Clin Oncol.
2005;23:7703-20.
79. Boughey JC, Suman VJ, Mittendorf EA, et al. Sentinel lymph node surgery after neoadjuvant
chemotherapy in patients with node-positive breast cancer: the ACOSOG Z1071 (Alliance) clinical
trial. JAMA. 2013;310:1455-61.
80. Boughey JC, Suman VJ, Mittendorf EA, et al. Factors affecting sentinel lymph node identification
rate after neoadjuvant chemotherapy for breast cancer patients enrolled in ACOSOG Z1071
(Alliance). Ann Surg. 2015;261:547-52.
81. Boileau JF, Poirier B, Basik M, et al. Sentinel node biopsy after neoadjuvant chemotherapy in
biopsy-proven node-positive breast cancer: the SN FNAC study. J Clin Oncol. 2015;33:258-64.
82. Caudle AS, Yang WT, Mittendorf EA, et al. Selective surgical localization of axillary lymph nodes
containing metastases in patients with breast cancer: a prospective feasibility trial. JAMA Surg.
2015;150:137-43.
83. Early Breast Cancer Trialists’ Collaborative Group (EBCTCG); Darby S, McGale P, Correa C, et al.
Effect of radiotherapy after breast-conserving surgery on 10-year recurrence and 15-year breast
cancer death: meta-analysis of individual patient data for 10,801 women in 17 randomised trials.
Lancet. 2011;378:1707-16.
84. Mak KS, Harris JR. Radiotherapy issues after neoadjuvant chemotherapy. J Natl Cancer Inst
Monogr. 2015;2015:87-9.
85. Buchholz TA, Tucker SL, Masullo L, et al. Predictors of local-regional recurrence after
neoadjuvant chemotherapy and mastectomy without radiation. J Clin Oncol. 2002;20:17-23.
86. Huang EH, Tucker SL, Strom EA, et al. Postmastectomy radiation improves local-regional control
and survival for selected patients with locally advanced breast cancer treated with neoadjuvant
chemotherapy and mastectomy. J Clin Oncol. 2004;22:4691-9.
87. Mamounas EP, Anderson SJ, Dignam JJ, et al. Predictors of locoregional recurrence after
neoadjuvant chemotherapy: results from combined analysis of National Surgical Adjuvant Breast and
Bowel Project B-18 and B-27. J Clin Oncol. 2012;30:3960-6.
88. McGuire SE, Gonzalez-Angulo AM, Huang EH, et al. Postmastectomy radiation improves the
outcome of patients with locally advanced breast cancer who achieve a pathologic complete
response to neoadjuvant chemotherapy. Int J Radiat Oncol Biol Phys. 2007;68:1004-9.
89. Yang TJ, Morrow M, Modi S, et al. The effect of molecular subtype and residual disease on
locoregional recurrence in breast cancer patients treated with neoadjuvant chemotherapy and
postmastectomy radiation. Ann Surg Oncol. 2015 Jul 1. [Epub ahead of print]
90. Krug D, Lederer B, Debus J, et al. Relationship of omission of adjuvant radiotherapy to outcomes
of locoregional control and disease-free survival in patients with or without pCR after neoadjuvant
chemotherapy for breast cancer: a meta-analysis on 3481 patients from the Gepar trials. J Clin Oncol.
2015;33(suppl):abstr 1008.
91. Mamounas EP, White JR, Bandos H, et al. NSABP B-51/RTOG 1304: randomized phase III clinical
trial evaluating the role of postmastectomy chest wall and regional nodal XRT (CWRNRT) and
post-lumpectomy RNRT in patients (pts) with documented positive axillary (Ax) nodes before
Page 15 of 16
Neoadjuvant Therapy for Early-Stage Breast Cancer: Current Pr
Published on Cancer Network (http://www.cancernetwork.com)
neoadjuvant chemotherapy (NC) who convert to pathologically negative Ax nodes after NC. J Clin
Oncol. 2014;32(suppl 5s):abstr TPS1141.
Source URL:
http://www.cancernetwork.com/oncology-journal/neoadjuvant-therapy-early-stage-breast-cancer-curr
ent-practice-controversies-and-future-directions
Links:
[1] http://www.cancernetwork.com/review-article
[2] http://www.cancernetwork.com/oncology-journal
[3] http://www.cancernetwork.com/breast-cancer
[4] http://www.cancernetwork.com/authors/cesar-augusto-santa-maria-md
[5] http://www.cancernetwork.com/authors/melissa-camp-md-mph
[6] http://www.cancernetwork.com/authors/ashley-cimino-mathews-md
[7] http://www.cancernetwork.com/authors/susan-harvey-md
[8] http://www.cancernetwork.com/authors/jean-wright-md
[9] http://www.cancernetwork.com/authors/vered-stearns-md
Page 16 of 16