You are on page 1of 8

Eliminating Breast Surgery for Invasive Cancer with

Exceptional Response to Neoadjuvant Systemic


Therapy: Prospective Multicenter Clinical Trial
Downloaded from http://journals.lww.com/journalacs by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XMi0hCy

Planned Initial Feasibility Endpoint


wCX1AWnYQp/IlQrHD3i3D0OdRyi7TvSFl4Cf3VC1y0abggQZXdtwnfKZBYtws= on 03/22/2024

Helen M Johnson, MD, Vicente Valero, MD, Wei T Yang, MBBS, MHM, Benjamin D Smith, MD,
Savitri Krishnamurthy, MD, Yu Shen, PhD, Heather Lin, PhD, Anthony Lucci, MD, FACS,
Gaiane M Rauch, MD, PhD, Henry M Kuerer, MD, PhD, FACS

BACKGROUND: Response to neoadjuvant systemic therapy (NST) for breast cancer enables tailoring of subse-
quent therapy. Image-guided breast biopsy after NST can accurately predict a pathologic com-
plete response (pCR). The feasibility phase of the clinical trial reported here assesses omission
of breast surgery followed by radiotherapy in terms of local recurrence before trial expansion.
STUDY DESIGN: Women with unicentric, cT1-2 N0-1 M0 triple-negative (TNBC) or human epidermal growth
factor receptor 2-positive breast cancer (HER2+BC) cancer with <2 cm residual disease on post-
NST imaging were eligible to enroll. If no residual invasive or in situ disease was identified by
image-guided, vacuum-assisted core biopsy (VACB), breast surgery was omitted, and radiother-
apy delivered. The primary endpoint for the feasibility phase was ipsilateral breast tumor recur-
rence at 6 months. If any recurrence occurred during the feasibility phase the trial would halt.
RESULTS: Thirteen patients were enrolled from March 2017 to October 2018. The mean age was 60.8
years (range 51 to 75) and most patients were White (69.2%) and non-Hispanic/Latino
(84.6%). All patients had invasive ductal carcinoma (6 TNBC, 7 HER2+BC). Mean tumor
size was 2.4 cm (range 0.9 to 5.0) before NST and 0.7 cm (range 0 to 1.8) after NST. Seven
patients (53.8%) had residual disease identified on VACB; the remaining 6 (46.2%) com-
prised the feasibility cohort. At a median follow-up of 44.3 months (range 41.3 to 51.3) there
was no ipsilateral breast tumor recurrence in this cohort.
CONCLUSIONS: These early data suggest that omission of breast surgery in patients with invasive TNBC and
HER2+BC with no evidence of residual disease on standardized VACB after NST is poten-
tially feasible. Results from the expansion phase of this clinical trial will be reported per pro-
tocol prespecified analyses. (J Am Coll Surg 2023;237:101–108. © 2023 by the American
College of Surgeons. Published by Wolters Kluwer Health, Inc. All rights reserved.)

CME questions for this article available at Shared Resource, as well as a Clinical and Translational Science Award (grant
UL1 RR024148) to MD Anderson Cancer Center.
http://jacscme.facs.org
Presented at the Western Surgical Association 130th Scientific Session, Santa
Disclosure Information: Authors have nothing to disclose. Timothy Barbara, CA, November 2022.
J Eberlein, Editor-in-Chief, has nothing to disclose. Ronald J Weigel,
CME Editor, has nothing to disclose. Received December 19, 2022; Revised January 18, 2023; Accepted January
19, 2023.
Disclosures outside the scope of this work: Dr Yang receives royalty pay-
ments from Elsevier. Dr Smith receives salary support from Varian Medical From the Departments of Breast Surgical Oncology (Johnson, Lucci, Kuerer),
Systems and receives royalty payments and equity interest from Oncora Breast Medical Oncology (Valero), Breast Imaging (Yang, Rauch), Radiation
Medical. Dr Valero is a paid consultant to Hologic, Exact Science, Roche, Oncology (Smith), Pathology (Krishnamurthy), Biostatistics (Shen, Lin),
Novartis, and AstraZeneca. Dr Krishnamurthy is a paid scientific advisory and Abdominal Imaging (Rauch), University of Texas MD Anderson Cancer
board member at AstraZeneca and is a paid consultant to Daichi Sankyo. Center, Houston, TX.
Support: Dr Kuerer is supported by the PH and Fay Etta Robinson Correspondence address: Henry M Kuerer, MD, PhD, FACS, Breast
Distinguished Professorship in Cancer research and funding MD Anderson Programs MD Anderson Cancer Network, University of Texas MD
Clinical Research Funding Award Program. Additional support provided by Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 1434, Houston,
a Cancer Center Support Grant (CA016672) from the National Institutes Texas 77030-4009. email: hkuerer@mdanderson.org
of Health, which supports the Clinical Trials Support Unit and Biostatistics A discussion of this article is available at http://links.lww.com/JACS/A274.

© 2023 by the American College of Surgeons. Published by Wolters Kluwer https://doi.org/10.1097/XCS.0000000000000670


Health, Inc. All rights reserved. 101 ISSN 1879-1190/22
102 Johnson et al    Eliminating Breast Surgery Trial Feasibility J Am Coll Surg

CONTINUING MEDICAL EDUCATION CREDIT INFORMATION


occurred during the first 6 months after radiotherapy in the
Accreditation 6 patients demonstrated to have a breast pCR on image-
The American College of Surgeons is accredited by the Accreditation
Council for Continuing Medical Education (ACCME) to provide continuing
guided VACB, the data safety monitoring committee
medical education for physicians. would halt the protocol. In the absence of recurrence the
accrual would continue to 50 total patients in the expan-
AMA PRA Category 1 Credits™
sion phase of the trial with subsequent protocol-specified
Downloaded from http://journals.lww.com/journalacs by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XMi0hCy

The American College of Surgeons designates this journal-based activity


for a maximum of 1.0 AMA PRA Category 1 Credits™. Physicians should planned outcome analyses. This article reports on the fea-
claim only the credit commensurate with the extent of their participation
sibility phase of this prospective multicenter clinical trial
wCX1AWnYQp/IlQrHD3i3D0OdRyi7TvSFl4Cf3VC1y0abggQZXdtwnfKZBYtws= on 03/22/2024

in the activity.
testing the safety of eliminating breast surgery among
Of the AMA PRA Category 1 Credits™ listed above, a maximum of 1.0 patients with VACB-proven cases of pCR.
credits meet the requirements for Self-Assessment.

METHODS
Cohort selection
This prospective, multicenter, single-arm, phase-2
clinical trial was registered with the National Cancer
Institute (NCT02945579) and approved by the insti-
Abbreviations and Acronyms tutional review boards of all participating centers. All
DCIS = ductal carcinoma in situ
patients provided informed consent. Women 40 years
HER2+BC = human epidermal growth factor receptor 2-posi-
tive breast cancer of age or older with pathologically confirmed invasive,
IBTR = ipsilateral breast tumor recurrence unicentric, cT1-2 N0-1 M0 TNBC or HER2+BC with
NST = neoadjuvant systemic therapy less than 2 cm residual disease (mass, density, suspicious
pCR = pathologic complete response calcifications, or enhancement) on post-NST imaging
TNBC = triple-negative breast cancer were eligible to enroll. HER2+BC was defined as an
VACB = vacuum-assisted core biopsy immunohistochemistry score of 3+ or amplified fluo-
rescence in situ hybridization. TNBC was defined by
estrogen receptor less than 10%, progesterone receptor
Neoadjuvant systemic therapy (NST) is frequently used in less than 10%, and HER2 immunohistochemistry of
the multimodality treatment of early-stage triple-negative 1+ or 2+ or nonamplified fluorescence in-situ hybrid-
breast cancer (TNBC) and human epidermal growth fac- ization. Clinical stage was defined by mammography
tor receptor 2-positive breast cancer (HER2+BC).1 NST and breast ultrasound including axillary nodal ultra-
facilitates less extensive breast and axillary surgery in cases sound and N1 was defined as 1 to 3 abnormal-appear-
of substantive tumor downstaging and enables tailoring of ing nodes on ultrasound and pathologic confirmation
adjuvant therapies in cases of residual disease.2 of metastatic disease on biopsy. MRI was performed
Pathologic complete response (pCR) to NST is asso- at physician discretion. Any NST regimen considered
ciated with improved overall survival, event-free sur- standard for TNBC or HER2+BC was permissible. The
vival, and distant recurrence-free survival for TNBC and primary tumor was localized with image-guided clip
HER2+BC.3,4 With modern systemic therapy for these marker placement before initiation of NST. Exclusion
subtypes, pCR occurs in 46% to 68% of cases.5-8 criteria included previous history of ipsilateral invasive
Image-guided, vacuum-assisted core biopsy (VACB) breast cancer or ductal carcinoma in situ (DCIS), preg-
of the tumor bed after NST can accurately identify resid- nancy, and progression of disease during NST.
ual disease and predict pCR, with a false-negative rate
of approximately 3% when standardized techniques are
applied to highly selected patients.9,10 The ability to pre- Trial design
dict which breast cancers demonstrate pCR to NST in The trial opened to accrual in March 2017. Patients
the preoperative setting calls into question the relative were eligible to enroll before, during, or after NST. At
benefit of local therapy for these patients. This prospec- enrollment, all patients were evaluated with a history
tive multicenter clinical trial aims to evaluate the safety of and physical examination. After completion of standard
omitting breast surgery in women with invasive TNBC or NST, patients underwent mammography and ultrasound
HER2+BC with exceptional response to NST. of the breast and the axilla. The optimal modality for
The feasibility phase of the trial was designed to eval- image-guided biopsy of the breast tumor bed was deter-
uate the safety of this new approach. If a local recurrence mined by the operating radiologist. Image-guided VACB
Vol. 237, No. 1, July 2023 Johnson et al    Eliminating Breast Surgery Trial Feasibility 103

was performed 2 to 6 weeks after completion of NST Patients with HER2+BC completed adjuvant anti-HER2
and required 12 (at minimum) 9-gauge cores. Biopsy of targeted therapy, and hormone receptor-positive patients
the previously placed clip was performed; the clip was also received adjuvant endocrine therapy.
removed along with more than 90% of residual radiologic After completion of radiotherapy, patients were evalu-
abnormality if present in the tumor bed. A new clip was ated with a history and physical examination and mammo-
placed at the biopsy site to facilitate localization in cases gram at 6-month intervals for 5 years. Abnormal radiologic
Downloaded from http://journals.lww.com/journalacs by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XMi0hCy

of residual disease and for ongoing imaging surveillance. findings underwent additional imaging work-up or biopsy
If residual invasive cancer or DCIS was identified on to evaluate for tumor recurrence. Progression of patients
wCX1AWnYQp/IlQrHD3i3D0OdRyi7TvSFl4Cf3VC1y0abggQZXdtwnfKZBYtws= on 03/22/2024

VACB pathology, patients were removed from the study through the trial is illustrated in Figure 1.
and proceeded to standard breast surgical therapy. If no
residual disease was identified, participants were eligible
Feasibility endpoint
to omit breast surgery and proceed to standard whole-
breast radiotherapy. Patients with cN1 disease were eligi- The feasibility phase of the trial was designed to evalu-
ble to participate if they demonstrated a breast pCR on ate 6 patients with no residual disease after NST, with a
VACB. They were required to undergo targeted axillary primary endpoint of ipsilateral breast tumor recurrence
dissection and completion axillary lymph node dissection (IBTR). If any of the 6 patients in the feasibility cohort
in the setting of residual nodal disease. Axillary surgery experienced IBTR within 6 months of post-NST biopsy
was not required for patients with cN0 disease. demonstrating no residual disease, the study would be
External beam radiotherapy was initiated within 12 halted, reviewed by principal investigators and the data
weeks of NST completion. For cN0 patients, whole-breast monitoring group, and either closed or modified. If no
radiotherapy at a dose of 40.05 Gy in 15 fractions was IBTR were observed with this timeframe, patients in the
delivered, followed by a tumor bed boost at a dose of 14 feasibility cohort would be rolled over to the expansion
Gy in 7 fractions. For cN1 patients, the radiation oncol- cohort and continue in the second phase of the trial with
ogist had the option to deliver whole-breast and regional continued prespecified monitoring and outcome analyses
nodal radiation at a dose of 50 Gy in 25 fractions, followed through 5 years of follow-up.
by the same boost. Patients were evaluated on a weekly We chose a feasibility cohort sample size of 6 patients
basis during radiotherapy. In the event of severe radiation for both practical consideration and statistical justification.
toxicity, the treating physician had the option to hold ther- We intended to minimize the number of patients exposed
apy for up to 3 days, with substitution of tumor bed boost to a new treatment strategy with unknown outcome in
for whole-breast therapy during this time, provided that the event that IBTR rates were higher than with stand-
the total final doses met the previously mentioned criteria. ard-of-care treatment. Additionally, we wanted to ensure

Figure 1. Flow diagram depicting progression of patients through the trial. HER2, human epidermal growth factor receptor 2.
104 Johnson et al    Eliminating Breast Surgery Trial Feasibility J Am Coll Surg

Table 1. Patient Characteristics Overall and by Presence or Absence of Residual Disease Identified After Neoadjuvant
Systemic Therapy on Image-Guided Vacuum-Assisted Core Biopsy
Residual disease No residual disease
Overall identified on VACB identified on VACB
Characteristic (n = 13) (n = 7) (n = 6)
Age, y, mean (range) 60.8 (51–75) 59.7 (52–67) 62.2 (51–75)
Downloaded from http://journals.lww.com/journalacs by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XMi0hCy

Race, n (%)
 White 9 (69.2) 4 (57.1) 5 (83.3)
wCX1AWnYQp/IlQrHD3i3D0OdRyi7TvSFl4Cf3VC1y0abggQZXdtwnfKZBYtws= on 03/22/2024

 Black 3 (23.0) 2 (28.6) 1 (16.7)


 Asian 1 (7.7) 1 (14.3) 0
Ethnicity, n (%)
 Hispanic or Latino 2 (15.4) 1 (14.3) 1 (16.7)
 Non-Hispanic or Latino 11 (84.6) 6 (85.7) 5 (83.3)
Breast cancer subtype, n (%)
 Triple-negative 6 (46.2) 3 (42.9) 3 (50)
 HER2-positive 7 (53.8) 4 (57.1) 3 (50)
 Hormone receptor-positive 4 (30.8) 3 (42.9) 1 (16.7)
 Hormone receptor negative 3 (23.0) 1 (14.3) 2 (33.3)
Histology: invasive ductal carcinoma, n (%) 13 (100) 7 (100) 6 (100)
Grade, n (%)
 1 0 0 0
 2 5 (38.5) 2 (28.6) 3 (50)
 3 8 (61.5) 5 (71.4) 3 (50)
Tumor size, initial, cm, mean (range) 2.4 (0.9–5.0) 2.2 (0.9–3.1) 2.5 (1.2–5.0)
Tumor size, after NST, cm, mean (range) 0.7 (0–1.8) 1.1 (0.5–1.5) 0.3 (0–1.8)
Radiologic complete response, n (%) 5 (38.5) 0 5 (83.3)
No. of VACB cores, mean (range) 17.3 (12–30) 18.4 (12–30) 16 (12–24)
HER2, human epidermal growth factor receptor 2; NST, neoadjuvant systemic therapy; VACB, vacuum-assisted core biopsy.

the opportunity to progress to the expansion phase of the trastuzumab, and pertuzumab, except for 1 patient with
study should IBTR rates prove similar to those seen with a 0.9-cm primary tumor who was treated with paclitaxel
standard breast surgical therapy. In a 6-patient cohort, and trastuzumab.
if the true IBTR rate at 6 months is 2%, the probabil- The median initial tumor size was 2.1 cm (range 0.9 to
ity of observing zero IBTR is 89% and the probability of 5.0), which decreased to a median of 0.8 cm (range 0 to
observing at least 1 IBTR is 11%. Alternatively, if the true 1.8) after NST. The median number of VACB cores was
6-month IBTR rate is 15%, these probabilities are 38% 18 (range 12 to 30). Seven patients had residual disease
and 62%, respectively. identified on VACB (4 with invasive cancer; 3 with DCIS
only). The remaining 6 patients comprised the feasibility
cohort.
RESULTS Among the feasibility cohort, the median age was 62
From March 2017 to October 2018, 13 patients were years (range 51 to 75) and most women were White
enrolled, which led to 6 patients with pCR by VACB for (83%) or non-Hispanic/Latino (83%). Half of the cohort
the feasibility cohort. Patient characteristics are shown in had TNBC and were treated with adriamycin and cyclo-
Table 1. Overall, the median age was 62 years (range 51 phosphamide followed by paclitaxel, while the other half
to 75). Most patients were White (69%) and non-His- had HER2+BC and were treated with docetaxel, carbo-
panic/Latino (84%). All patients had either interme- platin, trastuzumab, and pertuzumab. The median initial
diate- (38.5%) or high-grade (61.5%) invasive ductal tumor size was 2.1 cm (range 1.2 to 5.0). Five patients
carcinoma. Six patients had TNBC and 7 had HER2+BC. (83.3%) had radiologically complete responses; the sixth
All patients with TNBC were treated with adriamycin had a residual mass measuring 1.8 cm on post-NST imag-
and cyclophosphamide followed by paclitaxel. All patients ing. The median number of VACB cores was 15 (range 12
with HER2+BC were treated with docetaxel, carboplatin, to 24).
Vol. 237, No. 1, July 2023 Johnson et al    Eliminating Breast Surgery Trial Feasibility 105

Two patients with initial biopsy-proven N1 disease in Of note, 2 patients underwent surveillance ultrasound
the feasibility cohort underwent planned targeted axillary in addition to mammogram in the absence of mam-
dissection per protocol, and no lymph node metastases mographic abnormality per radiologist’s preference. One
were identified. These patients with initial N1 disease were of these patients was found to have 3 indeterminate,
treated with whole-breast and regional nodal radiotherapy sub-centimeter level 2 and 3 axillary lymph nodes at 48
at a dose of 50 Gy in 25 fractions, whereas the other 4 months after radiation and underwent ultrasound-guided
Downloaded from http://journals.lww.com/journalacs by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XMi0hCy

patients who did not present with N1 disease received 40 biopsy which confirmed lymphoid tissue and was negative
Gy in 15 fractions. All radiotherapy included a tumor bed for malignancy.
wCX1AWnYQp/IlQrHD3i3D0OdRyi7TvSFl4Cf3VC1y0abggQZXdtwnfKZBYtws= on 03/22/2024

boost per protocol criteria.


At a median follow-up of 44.3 months (range 41.3 to
51.3), there were no IBTRs in the feasibility cohort. At DISCUSSION
6 months follow-up, 3 patients had mammograms with The results of the feasibility phase of this prospective mul-
abnormal results and were evaluated further with addi- ticenter clinical trial suggest the potential safety of omit-
tional breast imaging. In 1 case, the mammogram noted ting breast surgery in patients with invasive TNBC and
focal asymmetry in proximity to the biopsy clip; further HER2+BC with no evidence of residual disease using
evaluation by ultrasound showed scarring and shadowing standardized, image-guided VACB after NST. The feasi-
consistent with postbiopsy changes. Contralateral breast bility phase was planned because this was a high-risk and
ultrasound was recommended for another patient because high-potential impact trial where standard surgical ther-
of a sub-centimeter oval mass visualized on mammogram apy has shown to be effective. None of the patients in the
that had the ultrasonographic appearance of fibrocystic feasibility cohort experienced an IBTR and therefore, the
changes classified as Breast Imaging Reporting and Data trial was not halted and was allowed to reach accrual of 50
System category 2. A third patient’s mammogram report patients. Additionally, no feasibility cohort patient expe-
noted postradiation changes at the site of the clip and rienced IBTR at median 44.3-month follow-up (at time
recommendation for ultrasound (read as “postradiation of writing). This indicates potential very early oncologic
changes limiting sonographic evaluation of the breast”), as safety results for the de-escalation of local therapy in tum-
well as noting a stable benign mass in a different quadrant. ors showing exceptional response to NST, demonstrated
MRI was performed demonstrating suspicious nonmass by breast pCR on image-guided-VACB. This concept is
enhancement in the region of the benign-appearing mass supported by the fact that definitive radiotherapy with-
(seen on ultrasound) for which biopsy was recommended. out surgical resection—with or without systemic ther-
Ultrasound-guided biopsy was performed and pathology apy—has shown to be effective for selected patients with
demonstrated fibroadenoma with focal atypical lobular esophageal, anal, laryngeal, prostate, cervical, or lung car-
hyperplasia. cinoma.11 However, until recently, it was not possible to
At 12 months, 4 patients had benign mammographic accurately predict which patients with breast cancer were
findings, including the patient who underwent 6-month free of residual disease after NST and could safely forego
biopsy. One patient underwent stereotactic biopsy of a 0.9- surgery.12
cm irregular mass associated with the biopsy clip attributa- NST is increasingly used for TNBC and HER2+BC.
ble to increased interval size and pathology demonstrating In 2017, the St Gallen International Expert Consensus
dense nodular stromal fibrosis. Another patient under- panel strongly endorsed NST for stage II-III TNBC and
went additional breast imaging because of mammographic HER2+BC, especially for cases in which NST may facilitate
findings of focal asymmetry with indistinct margins which de-escalation of local therapy.1 Similarly, current National
preclude accurate size measurement. Ultrasound showed Comprehensive Cancer Network guidelines recommend
postbiopsy changes classified as Breast Imaging Reporting NST for patients with operable TNBC and HER2+BC
and Data System category 4A and MRI demonstrated a in cases of cT2+ disease, cN1+ disease, or other cases for
correlative 1.5-cm irregular nonenhancing mass with spic- which NST would enable breast conservation therapy or
ulated margins classified as Breast Imaging Reporting and less-extensive axillary surgery, as well as consideration for
Data System category 4 with recommendation for biopsy. NST for cT1cN0 disease.13 With advances in NST result-
Stereotactic biopsy was performed and pathology demon- ing in higher rates of pCR, the next logical step in the push
strated stromal fibrosis and therapy-related changes. This for de-escalation of surgical therapy is omission of breast
patient underwent an additional stereotactic biopsy at 24 surgery for tumors with exceptional response to NST.
months for suspicious calcifications located in a different Higher rate of pCR may be expected in the future
quadrant than the tumor bed, with pathology demonstrat- as NST continues to be refined and new targeted ther-
ing fibroadenoma with stromal calcifications. apies, immunotherapies, and other novel agents are
106 Johnson et al    Eliminating Breast Surgery Trial Feasibility J Am Coll Surg

developed. Indeed, since the initiation of this trial, prac- the excellent locoregional recurrence-free survival rate of
tice-changing trials such as Pembrolizumab (MK-3475) patients with TNBC and HER2+BC who undergo NST
Plus Chemotherapy vs Placebo Plus Chemotherapy as with pCR followed by breast-conservation therapy (98.6%
Neoadjuvant Therapy and Pembrolizumab vs Placebo as and 97.4% at 5 years, respectively),17 we anticipated a sim-
Adjuvant Therapy in Participants with Triple Negative ilar rate in our feasibility cohort. Therefore, both a larger
Breast Cancer (KEYNOTE-522) have been published, number of patients and a longer median follow-up are nec-
Downloaded from http://journals.lww.com/journalacs by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XMi0hCy

demonstrating improvements in pCR for TNBC (from essary to observe a single event of locoregional recurrence.
55% to 69%) with the addition of pembrolizumab to a Five-year IBTR results from the expansion cohort will
wCX1AWnYQp/IlQrHD3i3D0OdRyi7TvSFl4Cf3VC1y0abggQZXdtwnfKZBYtws= on 03/22/2024

standard chemotherapy backbone of NST.14 Studies are provide key information. Early results from the expansion
ongoing to determine optimal NST and adjuvant sys- cohort are very promising and demonstrate no detectable
temic therapy regimens for different stages of HER2+BC, IBTR at an approximate 2-year median follow-up for the
including possible de-escalation of standard cytotoxic entire group of patients.18 Second, because of the nonzero
chemotherapy, with attention to pCR rates.2 As the pCR false-negative rate for predicting pCR by image-guided
rate continues to improve, if longer-term IBTR results VACB, it is possible that we failed to identify residual dis-
from the expansion phase of this trial support omission ease in some patients. While whole-breast radiotherapy
of breast surgery in TNBC and HER2+BC with pCR to would be expected to effectively treat a small volume of
NST, this concept will be tested in other subsequent pro- residual disease, it is possible that IBTR events could be
spective trials and can potentially be offered to a greater observed with a longer follow-up or larger cohort. Based
number of patients in the future. on the reported 5-year locoregional recurrence-free sur-
Meticulous multidisciplinary patient selection is essen- vival rate for patients with HER2+BC and TNBC (86.7%
tial for the oncologic safety of breast surgery omission. and 89.9%, respectively) whose tumors did not demon-
Preoperative identification of patients predicted to have strate pCR after NST and breast-conservation therapy,17
a pCR can be achieved with high accuracy by adhering and assuming that a lower rate would be observed if sur-
to standardized biopsy techniques. A false-negative rate of gery were omitted, we anticipate at least 1 IBTR in the
less than 5% can be achieved through stringent patient expansion cohort of future reports (median follow-up 5
selection criteria, thorough sampling of the tumor bed years) if the nonzero false-negative rate of VACB is clini-
(including removal of the previously placed clip), use of cally meaningful.
a larger-gauge VACB device with removal of multiple
cores, and meticulous specimen processing.9,10 The higher
false-negative rates reported in other studies (37-40%) can CONCLUSIONS
be attributed to differences in selection criteria and biopsy In this high-risk and high-potential prospective clinical
techniques.15,16 In this clinical trial, pCR was predicted trial, the feasibility cohort did not demonstrate any local
by examining a minimum of 12 cores with a single biopsy recurrence at 6 months per planned safety data monitor-
procedure that used a 9G VACB device. This design was ing and no patient had recurrence at follow-up (median
informed by results of an earlier trial that demonstrated a 44.3-months). The trial reached accrual of 50 patients and
reduction in the rate of false negatives when using these subsequent outcomes will be reported per protocol-speci-
measures to ensure adequate tumor bed sampling.12 The fied time-points. If these early limited results pan out, sev-
precision of this approach is further illustrated by the abil- eral other prospective trials testing this novel approach will
ity to discern both residual invasive disease and DCIS in be warranted in multiple medical settings.
the 7 patients who were enrolled but did not meet criteria
for inclusion in the feasibility cohort, which is consistent
with previous reports of 95% sensitivity for residual dis- Author Contributions
ease.12 It is interesting to note that 1 patient in the feasi- Data curation: Johnson, Yang, Smith, Krishnamurthy,
bility cohort did not have a radiologic complete response Lucci, Rauch
to NST (ie with residual 1.8 cm on imaging) but did have Visualization: Johnson
a pCR predicted by VACB. This is congruent with the Writing – original draft: Johnson
known variability in sensitivity and specificity of breast Conceptualization: Valero, Yang, Smith, Rauch, Kuerer
imaging modalities for the detection of residual disease Funding acquisition: Valero, Kuerer
after NST, as well as the described risk of radiologic over- Investigation: Valero, Yang, Smith, Krishnamurthy, Lucci,
estimation of residual disease.11 Rauch, Kuerer
This study is limited by several factors. First, the sam- Methodology: Valero, Shen, Lin, Kuerer
ple size of the initial feasibility cohort is very small. Given Project administration: Valero, Kuerer
Vol. 237, No. 1, July 2023 Johnson et al    Eliminating Breast Surgery Trial Feasibility 107

Resources: Valero, Kuerer FACS, Department of Breast Surgical Oncology, University


Supervision: Valero, Kuerer of Texas MD Anderson Cancer Center, Houston, TX.
Writing – review & editing: Valero, Yang, Smith, Jessica Suarez Colen, MD, MPH, Department of Breast
Krishnamurthy, Shen, Lin, Lucci, Rauch, Kuerer Surgical Oncology, University of Texas MD Anderson
Formal analysis: Shen, Lin Cancer Center, Houston, TX Catherine E Loveland-Jones,
MD, MS, FACS, Division of Breast Surgery, MD Anderson
Downloaded from http://journals.lww.com/journalacs by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XMi0hCy

Cancer Center at Cooper University Health Care,


Acknowledgment: Exceptional Responders Study Group
Camden, NJ Beth Ann Lesnikowski, MD, FACS, The
wCX1AWnYQp/IlQrHD3i3D0OdRyi7TvSFl4Cf3VC1y0abggQZXdtwnfKZBYtws= on 03/22/2024

Participating Investigators: Emilia J Diego, MD, FACS,


Breast Institute at JFK Medical Center, Atlantis, FL Laila
Division of Breast Surgery, University of Pittsburgh Medical
Samiian, MD, FACS, Section of Breast Surgical Oncology,
Center Magee-Women’s Hospital, Pittsburgh, PA Judy C
Baptist MD Anderson Cancer Center, Jacksonville, FL
Boughey, MD, FACS, Division of Breast and Melanoma
Clayton D Chong, MD, Medical Oncology, The Queen’s
Surgical Oncology, Department of Surgery, Mayo Clinic,
Health System, Honolulu, HI Simona F Shaitelman, MD,
Rochester, MN Richard L White, MD, FACS, Division
EDM, Department of Radiation Oncology, University
of Surgical Oncology, Department of Surgery, Carolinas
of Texas MD Anderson Cancer Center, Houston, TX
Medical Center, Levine Cancer Institute, Atrium Health,
Melissa P Mitchell, MD, PhD, Department of Radiation
Charlotte, NC Tanya W Moseley, MD, Department of
Breast Imaging, University of Texas MD Anderson Cancer Oncology, University of Texas MD Anderson Cancer
Center, Houston, TX Jessica WT Leung, MD, Department Center, Houston, TX.
of Breast Imaging, University of Texas MD Anderson Cancer
Center, Houston, TX Monica Huang, MD, Department REFERENCES
of Breast Imaging, University of Texas MD Anderson 1. Curigliano G, Burstein HJ, Winer EP, et al; St. Gallen
Cancer Center, Houston, TX Rosalind P Candelaria, International Expert Consensus on the Primary Therapy of
MD, Department of Breast Imaging, University of Texas Early Breast Cancer 2017. De-escalating and escalating treat-
ments for early-stage breast cancer: the St. Gallen International
MD Anderson Cancer Center, Houston, TX Beatriz E
Expert Consensus Conference on the Primary Therapy of Early
Adrada, MD, Department of Breast Imaging, University Breast Cancer 2017. Ann Oncol 2017;28:1700–1712.
of Texas MD Anderson Cancer Center, Houston, TX Elsa 2. Leon-Ferre RA, Hieken TJ, Boughey JC. The Landmark series:
Arribas, MD, Department of Breast Imaging, University neoadjuvant chemotherapy for triple-negative and her2-positive
of Texas MD Anderson Cancer Center, Houston, TX breast cancer. Ann Surg Oncol 2021;28:2111–2119.
Raquel FD van la Parra, MD, PhD, Department of Breast 3. Cortazar P, Zhang L, Untch M, et al. Pathological complete
Surgical Oncology, University of Texas MD Anderson response and long-term clinical benefit in breast cancer: the
Cancer Center, Houston, TX Kelly K Hunt, MD, FACS, CTNeoBC pooled analysis. Lancet 2014;384:164–172.
4. Consortium IST, Yee D, DeMichele AM, Yau C, et al.
Department of Breast Surgical Oncology, University of Association of event-free and distant recurrence-free survival
Texas MD Anderson Cancer Center, Houston, TX Isabelle with individual-level pathologic complete response in neoadju-
Bedrosian, MD, FACS, Department of Breast Surgical vant treatment of stages 2 and 3 breast cancer: three-year fol-
Oncology, University of Texas MD Anderson Cancer low-up analysis for the I-SPY2 Adaptively Randomized Clinical
Center, Houston, TX Mediget Teshome, MD, MPH, Trial. JAMA Oncol 2020;6:1355–1362.
FACS, Department of Breast Surgical Oncology, University 5. Gianni L, Pienkowski T, Im YH, et al. Efficacy and safety of neo-
of Texas MD Anderson Cancer Center, Houston, TX adjuvant pertuzumab and trastuzumab in women with locally
advanced, inflammatory, or early HER2-positive breast cancer
Susie X Sun, MD, MS, FACS, Department of Breast
(NeoSphere): a randomised multicentre, open-label, phase 2 trial.
Surgical Oncology, University of Texas MD Anderson Lancet Oncol 2012;13:25–32.
Cancer Center, Houston, TX Rosa F Hwang, MD, FACS, 6. Bayraktar S, Gonzalez-Angulo AM, Lei X, et al. Efficacy of neo-
Department of Breast Surgical Oncology, University adjuvant therapy with trastuzumab concurrent with anthracy-
of Texas MD Anderson Cancer Center, Houston, TX cline- and nonanthracycline-based regimens for HER2-positive
Makesha V Miggins, MD, FACS, Department of Breast breast cancer. Cancer 2012;118:2385–2393.
Surgical Oncology, University of Texas MD Anderson 7. van Ramshorst MS, van der Voort A, van Werkhoven ED, et al;
Cancer Center, Houston, TX Matthew J Piotrowski, MD, Dutch Breast Cancer Research Group (BOOG). Neoadjuvant
chemotherapy with or without anthracyclines in the presence of
FACS, Department of Breast Surgical Oncology, University
dual HER2 blockade for HER2-positive breast cancer (TRAIN-
of Texas MD Anderson Cancer Center, Houston, TX 2): a multicentre, open-label, randomised, phase 3 trial. Lancet
Anna P Refinetti, MD, FACS, Department of Breast Oncol 2018;19:1630–1640.
Surgical Oncology, University of Texas MD Anderson 8. Nanda R, Liu MC, Yau C, et al. Effect of pembrolizumab plus
Cancer Center, Houston, TX Richard A Ehlers, MD, neoadjuvant chemotherapy on pathologic complete response in
108 Johnson et al    Eliminating Breast Surgery Trial Feasibility J Am Coll Surg

women with early-stage breast cancer: an analysis of the ongo- 14. Schmid P, Cortes J, Pusztai L, et al; KEYNOTE-522
ing phase 2 adaptively randomized I-SPY2 Trial. JAMA Oncol Investigators. Pembrolizumab for early triple-negative breast
2020;6:676–684. cancer. N Engl J Med 2020;382:810–821.
9. Tasoulis MK, Lee HB, Yang W, et al. Accuracy of post-neo- 15. Lee HB, Han W, Kim SY, et al. Prediction of pathologic complete
adjuvant chemotherapy image-guided breast biopsy to predict response using image-guided biopsy after neoadjuvant chemother-
residual cancer. JAMA Surg 2020;155:e204103. apy in breast cancer patients selected based on MRI findings: a pro-
Downloaded from http://journals.lww.com/journalacs by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XMi0hCy

10. Koelbel V, Pfob A, Schaefgen B, et al. Vacuum-assisted breast spective feasibility trial. Breast Cancer Res Treat 2020;182:97–105.
biopsy after neoadjuvant systemic treatment for reliable exclu- 16. van Loevezijn AA, van der Noordaa MEM, van Werkhoven ED,
sion of residual cancer in breast cancer patients. Ann Surg et al. Minimally Invasive Complete Response Assessment of the
wCX1AWnYQp/IlQrHD3i3D0OdRyi7TvSFl4Cf3VC1y0abggQZXdtwnfKZBYtws= on 03/22/2024

Breast After Neoadjuvant Systemic Therapy for Early Breast


Oncol 2022;29:1076–1084.
Cancer (MICRA trial): interim analysis of a multicenter obser-
11. van la Parra RF, Kuerer HM. Selective elimination of breast can-
vational cohort study. Ann Surg Oncol 2021;28:3243–3253.
cer surgery in exceptional responders: historical perspective and 17. Swisher SK, Vila J, Tucker SL, et al. Locoregional control
current trials. Breast Cancer Res 2016;18:28. according to breast cancer subtype and response to neoadjuvant
12. Kuerer HM, Rauch GM, Krishnamurthy S, et al. A clinical chemotherapy in breast cancer patients undergoing breast-con-
feasibility trial for identification of exceptional responders in serving therapy. Ann Surg Oncol 2016;23:749–756.
whom breast cancer surgery can be eliminated following neoad- 18. Kuerer HM, Smith BD, Krishnamurthy S, et al; Exceptional
juvant systemic therapy. Ann Surg 2018;267:946–951. Responders Clinical Trials Group. Eliminating breast surgery
13. Gradishar WJ, Moran MS, Abraham J, et al. Breast cancer, ver- for invasive breast cancer in exceptional responders to neoadju-
sion 3.2022, NCCN Clinical Practice Guidelines in Oncology. vant systemic therapy: a multicentre, single-arm, phase 2 trial.
J Natl Compr Canc Netw 2022;20:691–722. Lancet Oncol 2022;23:1517–1524.

You might also like