ArticlePDF Available

NSABP FB-10: a phase Ib/II trial evaluating ado-trastuzumab emtansine (T-DM1) with neratinib in women with metastatic HER2-positive breast cancer

Authors:

Abstract and Figures

Background We previously reported our phase Ib trial, testing the safety, tolerability, and efficacy of T-DM1 + neratinib in HER2-positive metastatic breast cancer patients. Patients with ERBB2 amplification in ctDNA had deeper and more durable responses. This study extends these observations with in-depth analysis of molecular markers and mechanisms of resistance in additional patients. Methods Forty-nine HER2-positive patients (determined locally) who progressed on-treatment with trastuzumab + pertuzumab were enrolled in this phase Ib/II study. Mutations and HER2 amplifications were assessed in ctDNA before (C1D1) and on-treatment (C2D1) with the Guardant360 assay. Archived tissue (TP0) and study entry biopsies (TP1) were assayed for whole transcriptome, HER2 copy number, and mutations, with Ampli-Seq, and centrally for HER2 with CLIA assays. Patient responses were assessed with RECIST v1.1, and Molecular Response with the Guardant360 Response algorithm. Results The ORR in phase II was 7/22 (32%), which included all patients who had at least one dose of study therapy. In phase I, the ORR was 12/19 (63%), which included only patients who were considered evaluable, having received their first scan at 6 weeks. Central confirmation of HER2-positivity was found in 83% (30/36) of the TP0 samples. HER2-amplified ctDNA was found at C1D1 in 48% (20/42) of samples. Patients with ctHER2-amp versus non-amplified HER2 ctDNA determined in C1D1 ctDNA had a longer median progression-free survival (PFS): 480 days versus 60 days (P = 0.015). Molecular Response scores were significantly associated with both PFS (HR 0.28, 95% CI 0.09–0.90, P = 0.033) and best response (P = 0.037). All five of the patients with ctHER2-amp at C1D1 who had undetectable ctDNA after study therapy had an objective response. Patients whose ctHER2-amp decreased on-treatment had better outcomes than patients whose ctHER2-amp remained unchanged. HER2 RNA levels show a correlation to HER2 CLIA IHC status and were significantly higher in patients with clinically documented responses compared to patients with progressive disease (P = 0.03). Conclusions The following biomarkers were associated with better outcomes for patients treated with T-DM1 + neratinib: (1) ctHER2-amp (C1D1) or in TP1; (2) Molecular Response scores; (3) loss of detectable ctDNA; (4) RNA levels of HER2; and (5) on-treatment loss of detectable ctHER2-amp. HER2 transcriptional and IHC/FISH status identify HER2-low cases (IHC 1+ or IHC 2+ and FISH negative) in these heavily anti-HER2 treated patients. Due to the small number of patients and samples in this study, the associations we have shown are for hypothesis generation only and remain to be validated in future studies. Clinical Trials registration NCT02236000
This content is subject to copyright. Terms and conditions apply.
Jacobsetal. Breast Cancer Research (2024) 26:69
https://doi.org/10.1186/s13058-024-01823-8
RESEARCH Open Access
© The Author(s) 2024, corrected publication 2024. Open Access This article is licensed under a Creative Commons Attribution 4.0
International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you
give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes
were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated
otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not
permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To
view a copy of this licence, visit http:// creat iveco mmons. org/ licen ses/ by/4. 0/. The Creative Commons Public Domain Dedication waiver
(http:// creat iveco mmons. org/ publi cdoma in/ zero/1. 0/) applies to the data made available in this article, unless otherwise stated in a
credit line to the data.
Breast Cancer Research
NSABP FB-10: aphase Ib/II trial evaluating
ado-trastuzumab emtansine (T-DM1)
withneratinib inwomen withmetastatic
HER2-positive breast cancer
Samuel A. Jacobs1*, Ying Wang1, Jame Abraham1,2, Huichen Feng1, Alberto J. Montero1,2,3, Corey Lipchik1,
Melanie Finnigan1, Rachel C. Jankowitz1,4,5, Mohamad A. Salkeni1,6,7, Sai K. Maley1, Shannon L. Puhalla1,8,9,
Fanny Piette10, Katie Quinn11, Kyle Chang11, Rebecca J. Nagy11, Carmen J. Allegra1,12, Kelly Vehec1,
Norman Wolmark1,8, Peter C. Lucas1,8,9,13, Ashok Srinivasan1,14 and Katherine L. Pogue‑Geile1
Abstract
Background We previously reported our phase Ib trial, testing the safety, tolerability, and efficacy of T‑DM1 + ner
atinib in HER2‑positive metastatic breast cancer patients. Patients with ERBB2 amplification in ctDNA had deeper
and more durable responses. This study extends these observations with in‑depth analysis of molecular markers
and mechanisms of resistance in additional patients.
Methods Forty‑nine HER2‑positive patients (determined locally) who progressed on‑treatment with trastu‑
zumab + pertuzumab were enrolled in this phase Ib/II study. Mutations and HER2 amplifications were assessed
in ctDNA before (C1D1) and on‑treatment (C2D1) with the Guardant360 assay. Archived tissue (TP0) and study
entry biopsies (TP1) were assayed for whole transcriptome, HER2 copy number, and mutations, with Ampli‑Seq,
and centrally for HER2 with CLIA assays. Patient responses were assessed with RECIST v1.1, and Molecular Response
with the Guardant360 Response algorithm.
Results The ORR in phase II was 7/22 (32%), which included all patients who had at least one dose of study therapy.
In phase I, the ORR was 12/19 (63%), which included only patients who were considered evaluable, having received
their first scan at 6 weeks. Central confirmation of HER2‑positivity was found in 83% (30/36) of the TP0 samples.
HER2‑amplified ctDNA was found at C1D1 in 48% (20/42) of samples. Patients with ctHER2‑amp versus non‑amplified
HER2 ctDNA determined in C1D1 ctDNA had a longer median progression‑free survival (PFS): 480 days versus 60 days
(P = 0.015). Molecular Response scores were significantly associated with both PFS (HR 0.28, 95% CI 0.09–0.90,
P = 0.033) and best response (P = 0.037). All five of the patients with ctHER2‑amp at C1D1 who had undetectable
ctDNA after study therapy had an objective response. Patients whose ctHER2‑amp decreased on‑treatment had bet‑
ter outcomes than patients whose ctHER2‑amp remained unchanged. HER2 RNA levels show a correlation to HER2
CLIA IHC status and were significantly higher in patients with clinically documented responses compared to patients
with progressive disease (P = 0.03).
*Correspondence:
Samuel A. Jacobs
jacobssa1945@gmail.com
Full list of author information is available at the end of the article
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 2 of 13
Jacobsetal. Breast Cancer Research (2024) 26:69
Conclusions The following biomarkers were associated with better outcomes for patients treated with T‑DM1 + ner‑
atinib: (1) ctHER2‑amp (C1D1) or in TP1; (2) Molecular Response scores; (3) loss of detectable ctDNA; (4) RNA levels
of HER2; and (5) on‑treatment loss of detectable ctHER2‑amp. HER2 transcriptional and IHC/FISH status identify HER2‑
low cases (IHC 1+ or IHC 2+ and FISH negative) in these heavily anti‑HER2 treated patients. Due to the small number
of patients and samples in this study, the associations we have shown are for hypothesis generation only and remain
to be validated in future studies.
Clinical Trials registration NCT02236000
Keywords Metastatic breast cancer, ctDNA HER2 amplification, Clinical trial, Neratinib + t‑DM1
Introduction
In 2013, T-DM1 was the first HER2-targeted antibody–
drug conjugate (ADC) granted FDA-approval for late-
stage metastatic breast cancer after prior trastuzumab. In
2019, T-DM1 was approved as post-neoadjuvant therapy
in patients with residual disease based on the KATHER-
INE trial, demonstrating that post-neoadjuvant T-DM1
was statistically more beneficial than trastuzumab,
preventing recurrence of invasive disease or deaths in
patients with residual disease in breast or lymph nodes
after treatment with trastuzumab ± pertuzumab (haz-
ard ratio for invasive disease or death, 0.05: 95% CI
0.039–0.64; P < 0.001) [1]. KATHERINE required archi-
val HER2-positivity but did not mandate HER2 status
at study entry. Because multiple studies have confirmed
that HER2 status is plastic with conversion of HER2-pos-
itive disease to HER2-low (IHC = 0–1+ or IHC 2+ /FISH-
negative) under pressure of therapy [24], this raised the
question of ADC efficacy in HER2-low patients—either
de novo (HR + /HER2-negative) or acquired from con-
version of HER2-amplified to HER2-low. ere are now
several breast cancer-targeting ADCs in the pipeline in
addition to the newly approved trastuzumab deruxtecan
(T-DXd). Initial approval of T-DXd was for metastatic
HER2-positive breast cancer after prior progression on
multiple lines of anti-HER2 therapy (DESTINY-Breast01)
[5]. In DESTINY-Breast03, T-DXd improved PFS and
OS compared to T-DM1 [6] in patients with metastatic
disease with progression on trastuzumab. In heavily pre-
treated HER2-low breast cancer patients, T-DXd was
evaluated in a single arm phase II study. e objective
response rate (ORR) to T-DXd by central review was
37%, with median duration of response of 10.4months
[7]. DESTINY-Breast04, a randomized, multicenter trial
in patients with unresectable or metastatic HER2-low
breast cancer, reported highly significant improvements
in PFS and OS in patients receiving T-DXd compared to
physician choice of treatment [8]—a particularly striking
observation, because neither trastuzumab nor T-DM1
has shown consistent activity in HER2-low popula-
tions [9]. HER2 status (expression, mutation, amplifica-
tion) is thus emerging as a predictor of clinical efficacy
for anti-HER2-therapy. In our NSABP phase Ib trial of
HER2-targeted therapies using T-DM1 + neratinib in
HER2-positive patients, we showed a discordance in
HER2 status between archival tissue and a liquid biopsy
obtained at study entry. Loss of ctHER2-amp occurred
in 63% (17 of 27) patients. Deeper and more durable
responses were observed with T-DM1 + neratinib in
patients with ctHER2-amplification [10]. We now report
on an expanded cohort. Our aims were to: (1) confirm
activity of T-DM1 + neratinib in patients progressing on
a taxane with trastuzumab + pertuzumab (HP), (2) evalu-
ate discordance in HER2 amplification between archived
tissue, contemporaneous tissue, and blood, and (3) com-
pare mutation and gene-expression profiles at different
time points. Finally, in a subset of patients, we assessed
response by RECIST 1.1 with the Guardant Molecular
Response score [11, 12].
Methods
Trial design
FB-10 was a single-arm, nonrandomized, unblinded
clinical trial approved by participating institutions’ insti-
tutional review boards. Written informed consent was
required. FB-10 was conducted according to Good Clini-
cal Practices and the Declaration of Helsinki.
e phase Ib trial was a dose escalation study evaluat-
ing T-DM1 + neratinib in women with metastatic HER2-
postive breast cancer based on local determination of
HER2. Patients received 3.6mg/kg T-DM1 intravenously
on a 3-week cycle and oral neratinib was taken daily in
one of four dose cohorts (120, 160, 200 and 240 mg).
Twenty-seven patients enrolled, with 5 experiencing a
dose-limited toxicity. ree withdrew early for other
reasons (Fig. 1). Nineteen patients were evaluable for
response, which required follow-up imaging after the
second cycle of treatment (6weeks). e recommended
phase II dose of neratinib was determined to be 160mg/d
[10]; however, we did not detect a dose response, i.e., ner-
atinib at 120mg/d was as effective as higher doses and
less toxic. [10]
e phase II expansion included all patients (N = 22)
who received at least one dose of study therapy in
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 3 of 13
Jacobsetal. Breast Cancer Research (2024) 26:69
the analysis of safety and efficacy. Eligibility criteria
were identical in phase Ib and phase II [10]. All eligi-
ble patients had prior HP and a taxane as neoadjuvant
therapy or for de novo metastatic disease, had measur-
able disease, were ECOG PS 2, with adequate hema-
tologic, renal, and liver function. Patients with known
stable brain metastases were eligible. Brain imaging at
entry was not required. Treatment in phase II included
T-DM1 at 3.6 mg/kg iv q 3 weeks and neratinib at
160 mg/day. Primary diarrhea prophylaxis was man-
dated as described in phase I [10].
Safety assessment
Safety assessment was similar in phase Ib and phase II
including physical examination, interim history, and
laboratory assessments. Patients remained on treat-
ment until progressive disease or discontinuation
because of withdrawal, physician discretion or toxicity.
For phase Ib patients, adverse event (AE) assessment
occurred on days 1, 8, and 15 of cycle 1; on day 1 of
each cycle and for 30days after therapy discontinuation
or when alternate therapy began. Phase II AE assess-
ments were made on day 1 of each cycle.
Response evaluation
In phase Ib, patients were assessed for best response
beginning with their first follow-up scan after 2 cycles
(6 weeks). Response by RECIST v1.1 was complete
response (CR), partial response (PR), stable disease
(SD), or progression (PD). In phase II, imaging stud-
ies were performed after every 3 cycles (9weeks). e
clinical benefit rate included all CR, PR, and SD patients
with duration 180 days. Patients with stable disease
of < 180 days were included with progressive disease
patients. A confirmatory scan at least one month after
the best response was not required in this study, perhaps
accounting for partial responses of short duration in sev-
eral patients.
Blood andtissue collection
Blood samples were required before treatment at cycle 1,
day 1 (C1D1), and after treatment at cycle 2day 1 (C2D1)
for all patients in phase Ib and II (Fig.1A). Archived tis-
sue (TP0) of diagnostic blocks or slides were required on
all phase Ib and II patients which included 27 patients
from phase Ib and 22 patients in phase II. (Fig.1B). Con-
temporaneous biopsy specimens or slides at study entry
(TP1) were optional in phase Ib but in phase II, after
Fig. 1 Remark Diagram of Blood and Tissue Samples: NSABP FB‑10. A Blood samples collected from patients enrolled into FB‑10 phase Ib
and phase II, and successful assays for ctDNA analysis of HER2 amplification with Guardant360 assays. B Tissue samples collected from patients
enrolled into FB‑10 and their samples that were profiled for mutations and whole transcriptomic analysis and for ERBB2 amplification status
with CLIA and AmpliSeq assays. C The timing and type of sample collections (tissue: TP0 or TP1, or blood: C1D1 or C2D1) are shown
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 4 of 13
Jacobsetal. Breast Cancer Research (2024) 26:69
enrollment of the first 6 patients, the study was amended
to require a study entry biopsy. e timing and type of
collections of samples are shown in Fig.1C.
ctDNA assessment
Samples were analyzed by the Guardant360 assay
(Fig.1A), which detects single-nucleotide variants, indels,
fusions, and copy number alterations in 74 genes. For
HER2 amplification, a cutoff of 2.14 was used. Where
amplification could not be determined because of failed
assays or no blood, samples were categorized as indeter-
minant. Guardant Health (Guardant360 assay) is Clinical
Laboratory Improvement Amendments (CLIA)-certified,
College of American Pathologists-accredited, New York
State Department of Health-approved laboratory.
ctDNA molecular response
Guardant360 Molecular Response is a next generation
sequencing (NGS)based liquid biopsy that assesses
changes in tumorderived cellfree DNA (ctDNA)
between baseline and an early ontreatment timepoint
in patients with solid tumor malignancies. It employs
an algorithm to identify informative somatic single
nucleotide variants (SNVs), insertions/deletions and
gene fusions and calculates the percent ctDNA change
between the two timepoints based on the mean vari-
ant allele frequency (VAF) between two samples (mean
VAF2/mean VAF1) -1 × 100%. Using the Molecular
Response panel and the Guardant bioinformatics pipe-
line, the change in ctDNA levels between baseline and
the initial follow-up scan (6weeks in phase I and 9weeks
in phase II) was calculated and the change in VAF deter-
mined. Molecular Response is calculated as the ratio of
mean VAF on-treatment to baseline with a cutoff of 50%.
Decreases in ctDNA of 50%100% during this timeframe
are associated with clinical benefit in patients on anti
cancer therapies [11, 13, 14]. Kaplan–Meier curves for
PFS are generated for patients above and below a Molec-
ular Response cut off. [11]
Isolation ofnucleic acid
Tumor regions, defined by a certified pathologist, were
macrodissected. DNA and RNA were isolated using the
Qiagen AllPrep DNA/RNA kit, following the manufac-
turer’s recommendations but eliminating the xylene wash
in the first step. Separate tissue sections were used for
RNA and DNA isolation.
Whole transcriptomic proling
10–30ng of RNA from the phase II samples was reverse
transcribed. cDNA libraries were constructed using
whole transcriptomic Ampli-Seq kits, following the
manufacturer’s instructions without a fragmentation step
due to the small size of the RNAs. is same method did
not work well for the phase Ib RNAs. To overcome this
problem, phase Ib RNAs were made library-ready via the
HTG EdgeSeq system and the HTP panel, which includes
probes to interrogate 19,398 genes representing most of
the human transcriptome (details in Additional file1).
Breast cancer molecular subtypes were determined
by applying the AIMs signature [15]. e 8-gene trastu-
zumab-benefit groups were determined using a validated
signature. [16, 17]
HER2 amplication status andanalysis ofvariants intissues
DNA sequencing was performed using a custom Ampli-
Seq panel referred to as the NAR panel, amplifying 3,847
amplicons with 94.25% coverage of exons from 117 genes
in HER2-activated pathways (Additional file1: TableS1).
e panel was designed using the ermo Fisher Ion
AmpliSeq Designer tool (https:// www. ampli seq. com).
Libraries were constructed using 10ng of DNA using the
Ion AmpliSeq kit for Chef DL8. e Ion Chef instru-
ment was used to template and load samples on Ion 550
chips. Up to 32 samples were barcoded, pooled, and
sequenced on the S5 sequencer (ermoFisher) following
manufacturer’s instructions.
We have used two different criteria to identify vari-
ants in FB-10 tissue. For a conservative approach to vari-
ant selection, we selected variants with VAF 10% and
for a less restrictive option we selected variants with
VAF 5%. Additional details are included in Additional
file1 and the rationale for these approaches is discussed.
Ion Torrent data and the Ion Reporter software were
used to determine HER2 copy number.
HER2 IHC FISH
HER2 status was also assessed in tissue samples with IHC
and reflexively for FISH at the discretion of the Director
at the CLIA laboratory (Magee Women’s Hospital, Uni-
versity of Pittsburgh Medical Center). Nine samples were
equivocal IHC 0 or 1+ due to poor tissue quality prompt-
ing examination with FISH. Based on FISH, four were
included as HER2-positive.
Statistical analysis
Phase Ib safety, tolerability, efficacy, and recommended
phase 2 dose (RP2D) of neratinib in combination with
T-DM1 was previously reported [10]. In the phase II
expansion cohort, in which neratinib was administered
at the RP2D of 160mg/d, the intention was to confirm
clinical efficacy and tolerability of the combination and
to extend the correlative findings. Given the small sample
size, the endpoint analyses remain descriptive.
e aim of the single-arm phase II expansion was to
rule out the null hypothesis that the ORR was 25% with
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 5 of 13
Jacobsetal. Breast Cancer Research (2024) 26:69
the alternative hypothesis of an ORR of 45%. With these
assumptions, the sample size required was 22 and the
decision rules are to declare success if > 8 responses; to
declare failure if < 7 responses; and to consider the trial
inconclusive if 7 or 8 responses (7/22 = 32%, 8/22 = 36%).
At the outset of the study, we did not anticipate the large
number of patients with loss of HER2-amplification in
blood as determined by the Guardant assay. us, the
subset analyses based upon HER-amplification detected
in blood were performed post-hoc.
Results
Patient characteristics
In the phase Ib portion of this study, 27 patients were
enrolled between February 2015 and July 2017. Nineteen
patients were evaluable having had at least one follow-up
imaging study; three patients withdrew from the study
in cycle 1 and 5 patients with a dose-limited toxicity in
cycle 1 did not have an imaging assessment. All phase
II patients who received at least one dose of study drugs
were included in the analysis. Twenty-two patients were
evaluable for toxicity and 20 were evaluated for efficacy
with at least one scan performed after their third cycle.
Two non-evaluable patients who withdrew from the
study did not have their first scan but are included as PD.
Median age was 55.5years (range 32–70). Hormone sta-
tus (ER and/or PR) was positive in 13 patients and nega-
tive in 9. All patients were HER2-positive at baseline by
local determination (Additional file1: TableS3).
Safety assessment
Similar to phase Ib patients, diarrhea was the most fre-
quent toxicity in phase II: grade 2, 6 patients (27%);
grade 3, 8 (36%). Other grade 3/4 toxicities included:
thrombocytopenia, 2 patients (10%); transaminase eleva-
tion, 3 patients (15%); and pneumonitis, 1 patient (5%).
ere were no unanticipated toxicities in the phase II
expansion.
Ecacy
Among 19 evaluable patients in phase Ib, there were 3
CRs and 9 PRs for an ORR of 63% (12/19) [10]. In phase
II, including all patients who received at least one dose
of therapy, there were 2 CRs, 5 PRs for an ORR of 32%
(7/22), and 3 SDs of 180days or longer making the clini-
cal benefit rate (CBR) 45% (10/22). In phase Ib and II,
nine patients had sustained objective responses lasting
approximately 1year or longer (range 343–1453 + days,
Additional file1: TableS4; Additional file2: Table S5).
Treatment was discontinued at or before the first scan in
15 patients for a variety of reasons, including 5 DLTs (all
in phase I) and 10 with clinical progression in phase I and
II.
ctDNA clearance andtreatment response
Because clearance of ctDNA has been associated with
treatment response, we compared the outcomes of
patients who were positive or negative for ctDNA after
study treatment. e response rate among the ctDNA-
positive patients who were still ctDNA-positive after
study therapy was 9/19 (47%), but the ctHER2 DNA-pos-
itive patients who became ctDNA-undetectable at C2D1,
the response rate was 6/6 (100%), demonstrating that the
loss of ctDNA was associated with a very good response.
HER2 amplication intissues andblood
We assessed the HER2 amplification status of TP0 and
TP1 with a CLIA HER2 IHC/FISH assay, and with an
Ampli-Seq NGS assay. ese tissue samples were also
compared to the HER2 amplification status in blood sam-
ples collected at C1D1 and C2D1 (Fig. 2B). ere was
good concordance between the CLIA HER2/FISH and
Ampli-Seq assays (100% in TP1 tissues and 85% in all tis-
sues), which demonstrated the technical accuracy of the
Ampli-Seq. Concordance between TP0 tissue with IHC/
FISH and C1D1 ctDNA was 71% (20/28). Concordance
between C1D1 and C2D1 was 54% (14/26). Anti-HER2
treatment is potentially the causal reason for the discord-
ance between C1D1 and C2D1, which showed a total loss
of ctDNA in some samples and a loss of HER2 amplifica-
tion in others.
Using the Guardant360 assay cut point of 2.14 for
amplification among 43 C1D1 samples (22 in phase
Ib, 21 in phase II), 6 patient samples were indetermi-
nate (including 4 for which somatic mutations were not
detected) (Fig.2B, dark green), 1 was not evaluable (NE),
and 1 other failed quality control. Among the remaining
37 samples, there were 21/37 (57%) patients with ampli-
fication and 17/37 (46%) without. e objective response
(CR, PR) rate was 55% (11/20) in amplified patients
and 41% (7/17) in non-amplified patients. e CBR in
patients with ctHER2-amplification was 12/21 (57%)
and in non-amplified patients it was 8/17 (47%). ere
was one patient with SD who was ctHER2-amp indeter-
minate. Mean duration of response (CBR) was substan-
tially longer in amplified patients, 457days compared to
131days in non-amplified patients (P = 0.008) (Fig. 2A;
Additional file1: TableS4).
We compared progression-free survival (PFS) of
patients whose ctDNA or tumor tissues had HER2 ampli-
fication to patients with no HER2 amplification. Patients
with ctHER2-amp at C1D1 or in their TP1 tumor tis-
sue had a significantly longer PFS than patients with no
HER2 amplification (Fig.3A–E).
In phase I and II there were 26 C1D1 and C2D1 pairs,
15 with and 11 without ctHER2-amp at C1D1. Among
the 15 with ctHER2-amp at C1D1, 14 showed HER2 loss
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 6 of 13
Jacobsetal. Breast Cancer Research (2024) 26:69
Fig. 2 Amplification Status of Tissues and Blood: NSABP FB‑10. A Response rates (CR/PR, CBR, and SD) for FB‑10 HER2‑amplified and non‑amplified
patients based on ctDNA results. B HER2‑amplification status of FB‑10 tumor tissues based on CLIA tests (IHC/FISH) and Ampli‑Seq (Tissue)
in baseline (TP0) and study entry ( TP1) samples are shown. HER2‑amplification status was determined in ctDNA at C1D1 and at C2D1
with the Guardant360 assays. Responses, amplification status, and changes in copy number in ctDNA between C1D1 and C2D1 are indicated
as shown in the legend
Fig. 3 Association of HER2‑amplification Status with Patient Outcomes: NSABP FB‑10. A Kaplan‑Meier plots of patients with or without HER2
amplification in ctDNA or in TP0 tissue (B & D), or in TP1 tissue (C & E) based on IHC/FISH (B & C) and on Ampli‑Seq (D & E)
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 7 of 13
Jacobsetal. Breast Cancer Research (2024) 26:69
at C2D1 as defined by a loss 28% of HER2 copy number
or no ctDNA detected. e ORR among these 14 patients
was 71% (10/14). Of the 10 responders, 5 cleared ctDNA
completely by C2D1, 3 had detectable ctDNA but no
ctHER2-amp, and 2 were HER2-positive but the amplifi-
cation level in C2D1had decreased dramatically (Fig.2B;
Additional file2: TableS5). e 2 remaining patients with
detectable ctHER2-amp with no loss of HER2 amplifica-
tion had PD, suggesting that a loss of ctDNA and/or a loss
of HER2 ctDNA amplification was a marker for a good
response to study therapy. However, in 11 patients with
no HER2 ctDNA amplification at C1D1, the ORR was
45% (5/11), indicating that some non-amplified tumors
were responsive to study treatment.
Molecular response byctDNA
We assessed the association between molecular response
and objective radiologic response (Fig.4A). A total of 21
patients (9 phase Ib and 12 phase II) had paired samples
that met criteria for assessment of molecular response.
Criteria included 1 alteration present in one of the
paired samples plus a mutant molecule count of 15 in
either sample. Molecular responders demonstrated a
longer PFS compared to non-responders (median PFS 7.4
vs. 2.8, HR 0.28, 95%CI 0.09-0.90, P=0.033 using Wilcox
test). We also examined the association between molecu-
lar response and best RECIST response. Patients with
CR/PR/SD had significantly lower Molecular Response
values compared to patients with PD (P = 0.037; Fig.4B).
Mutations/variants intissues andctDNA
Because ERBB2 is the target of the study therapies, we
have examined both tissue and ctDNA for mutations in
the ERBB2 gene. No ERBB2 variants in tissue at a VAF
of 10% were observed, however, in ctDNA 3 nonsynon-
ymous, ERBB2 variants (I767M, V777L, and S310Y) were
detected in the C1D1 samples from 3 patients. ese
variants have been associated with sensitivity to neratinib
in breast cancer patients [18]. In this study, the patients
whose tumors had a V777L or a S310Y mutation had a
PR, but the one patient with a I767M mutation had PD
with brain metastasis. e tumor with the I767M muta-
tion also had a P1233L mutation [19]. Interestingly, in
an exhaustive meta-analysis of 37,218 patients, includ-
ing 11,906 primary tumor samples, 5,541 extracerebral
metastasis samples, and with 1485 brain metastasis sam-
ples found that a nearby ERBB2 mutation (P1227S) was
the only mutation restricted to brain metastasis. It is
unknown whether any of these mutations played a role in
the patient responses or the course of disease, but it is of
interest to note them [20].
We examined DNA variants in all available TP0 and
TP1 tissues using our NAR Ampli-Seq panel, which
included ESR1, HER2, and 115 other genes in HER2-
activated pathways. Based on our stringent criteria for
variant detection, i.e., VAF 10%, plus other criteria as
described in Additional file1, we identified 27 variants
among 28 samples, representing 21 patients (Fig.5A).
e frequency of PIK3CA mutations among all
sequenced patients was 34% (12/35), similar to that seen
Fig. 4 Molecular Response and Patient Outcomes: NSABP FB‑10. A Kaplan–Meier curves showing association of MR with PFS using a molecular
response cutoff of 50%. B Association between molecular response and best RECIST response
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 8 of 13
Jacobsetal. Breast Cancer Research (2024) 26:69
in other studies of unselected metastatic and early-stage
breast cancer patients (cBioPortal). All of the mutations
were in exons 9 and 20 at amino acid 545 and 1,047,
respectively. ese PIK3CA variants also have the highest
VAFs (ranging from 10 to 72% across samples), however,
PIK3CA mutations do not appear to influence patient
outcomes, because response rates between PIK3CA
mutant and WT tumors were similar: 42% (4/12) ver-
sus 45% (14/31), respectively. In one case, a PIK3CA
mutation was detected only in TP1 but this patient had
a PR, again indicating that PIK3CA is not a resistance
marker for study therapy. Variants detected only in PD
or CR patients represent potential resistance or sensi-
tivity markers, respectively, to study therapy. Mutations
detected only in TP1 samples among the 12 paired TP0/
TP1 cases, included ADAM17_S770L, ERBB4_E1010K,
ERBB4_R1040T, and IL6ST_S834* in one sample and
an ESR1_EY537S mutation in another (Fig. 5A). Both
patients had PD, perhaps indicating that these mutations
may have emerged in response to prior therapies. Details
of variants are presented in Additional file1.
Whole transcriptomic proling
We examined the PAM50 subtypes and the 8-gene tras-
tuzumab benefit signature in all available tissues [16].
Among the 29 patients with response and gene expres-
sion data for TP0 tissue, we found that 19/34 (56%)
were HER2E, 8 (24%) were luminal B, 4 (12%) were
basal, 2 (5.9%) were normal, and 1 (2.9%) was luminal
A. Patients with luminal subtype tumors had a lower
CR/PR response rate (1/8 [12.5%]) than patients with
a non-luminal subtype (12/23 [52%]) (Additional file2:
TableS5). Among the TP1 samples with gene expression
data, the frequency of CR/PR was 1/4 in luminal patients
and 5/9 in the non-luminal patients. Intrinsic subtypes
differed between TP0 and TP1 tissues in some cases
(Additional file1: TableS4). Although numbers are small,
the frequency of CR/PR rates were consistently lower
among the luminal patients than non-luminal patients.
e 8-gene trastuzumab signature is a validated sig-
nature for identifying patients with large-, moderate- or
no-benefit from trastuzumab when added to chemother-
apy in the adjuvant setting [16, 17] and has been shown
to associate with pCR rates in the neoadjuvant setting
[21, 22]. We questioned whether this signature may also
show an association with response in the metastatic set-
ting. Among the large-, moderate- and no- benefit groups
the percent of CR/PR patients was 67% (4/6), 50% (9/18),
and 29% (2/7), respectively (data in Additional file 2:
TableS5).
As expected, the level of HER2 RNA increased as the
IHC status increased (i.e., 0, 1 + , 2 + , to 3 +) (Addi-
tional file1: Fig. S2). In TP1 samples they were concord-
ant with patient responses suggesting that HER2 RNA
expression in study entry is associated with response to
T-DM1 + neratinib (Fig.6).
Significant differences were detected in RNA levels
between IHC 1 + and 3 + and between 2 + and 3 + but not
between 0 and 1 + nor between 1 + and 2 + (Additional
file1: Fig. S3). Although numbers are limited, these data
Fig. 5 Variant Alleles in Patients and their Responses: NSABP FB‑10. A Variants detected with a VAF of ≥ 10% in patients with PD, SD, PR or CR.
*indicates a stop codon. B Variant alleles with a VAF of ≥ 5% in patients with PD, SD, PR or CR
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 9 of 13
Jacobsetal. Breast Cancer Research (2024) 26:69
show that the RNA levels are not different between 0
and 1 + . ese patients may benefit from treatment with
other ADCs. e DAISY and DESTINY-Breast04 trials
signal that T-DXd may have significant activity in HER2-
low patients [8, 23].
Discussion
Approximately 35% of HER2-positive patients may have a
loss of HER2 amplification after undergoing chemother-
apy + anti-HER2 therapy [2, 3]. In a retrospective analysis
of 525 patients who received neoadjuvant chemotherapy
(NAC) + HP, 141 patients with residual disease had HER2
status determined pre-and post-NAC-HP. HER2 was
concordant (positive/positive) in 84/141 (60%). HER2
protein expression was lost (IHC 0) in 13/57 (23%) and
designated as HER2-low in 44/57 (77%) including IHC
1 + in 31 and IHC 2 + /FISH non-amplified in 13 [4].
HER2 intratumoral heterogeneity is likely one cause of
discordant HER2 status between primary and post-treat-
ment residual or metastatic disease [24]. Other possibili-
ties include decreased HER2 expression, which could be
a transient change or a result of the selection of HER2-
low subclones [4].
We have assessed HER2 status before and after pre-
and post-study therapy in not only solid tissue but also
blood. We have determined the HER2 status in tissues
with CLIA IHC/FISH assays, which is the gold standard
for HER2 assessment, plus with Ampli-Seq, because it
provided a quantitative analysis of HER2 copy number
with a greater dynamic range. Ampli-Seq was able to
detect a decrease in HER2 copy number in samples that
had not lost HER2 amplification based on IHC/FISH.
We have also monitored HER2 status in liquid biopsies,
which has several advantages over genomic analysis
of tissues. Blood has exposure to all potential meta-
static sites allowing for the detection of different vari-
ants from different metastatic sites. us, blood may
be more representative of the metastatic tumor than
examination of a single biopsied lesion, and may reflect
tumor evolution and intratumoral heterogeneity [25].
Blood samples are more easily collected, making multi-
ple serial collections possible. Collecting multiple serial
tissue samples is impractical, costly, and represents a
much greater risk to patients than does serial collec-
tion of blood. e assessment of ctDNA is a powerful
tool, showing very promising results to monitor tumor
recurrence and response to therapy, but it does not yet
replace the current gold standard, IHC/FISH, for the
assessment of HER2 status in solid tumors. However,
the monitoring of the HER2 status in ctDNA does pro-
vide an indicator of tumor response to therapy.
In our phase Ib/II study, HER2 tissue was amplified
in the baseline samples (TP0) (pre- anti-HER2 therapy)
in all patients by local determination, however, in liq-
uid biopsies at C1D1 after chemotherapy + HP, HE R2-
amplification was detected in only 20/42 (48%) of
patients. Patients with ctHER2-amp versus non-ampli-
fied HER2 ctDNA determined in C1D1 ctDNA had a
longer median PFS, 480days versus 60days (P = 0.015).
It is expected that patients with HER2 amplification
would respond to study therapy (chemotherapy + HP).
Fig. 6 RNA Expression Levels and Response to Therapy. RNA expression levels in TP0 tissues (A) and in TP1 tissues (B) from patients with PD, SD
or CR/PR. The units for RNA expression were log 2 expression values
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 10 of 13
Jacobsetal. Breast Cancer Research (2024) 26:69
Loss of HER2 amplification observed after one cycle
of study therapy may indicate that responders are either
clearing ctDNA completely or that the amplification falls
below the limit of detection. In the 5 cases who were
ctHER2 DNA amplified, and completely cleared ctDNA,
the response rate was 100%.
We applied a Molecular Response VAF ratio calculation
to measure the change in ctDNA from baseline to C2D1,
with the hypothesis that an early decrease in ctDNA lev-
els would predict response to T-DM1 + neratinib ther-
apy, as measured by PFS and RECIST response. Indeed,
Molecular Response was associated with both PFS and
best response to therapy. is should be confirmed in
larger dataset, however, our findings are in line with
other studies demonstrating the ability of ctDNA to pre-
dict short- and long-term efficacy. Early data from the
PADA-1 trial suggests that changing therapy based on
alterations detected in ctDNA, prior to evidence of pro-
gression via imaging, may provide clinical benefit. In
that trial, patients with ER + /HER2-neg ative metastatic
breast cancer being treated in the first line setting with an
aromatase inhibitor (AI) + palbociclib were monitored for
hotspot ESR1 alterations via ctDNA using digital droplet
PCR (ddPCR). Patients with rising ESR1 VAF on therapy,
but no synchronous evidence of disease progression via
RECIST 1.1, were randomized to either continue receiv-
ing an AI + palbociclib or switched to fulvestrant + pal-
bociclib. PADA-1 met its primary efficacy objective, with
patients randomized to receive fulvestrant + palbociclib
having a significantly longer PFS compared to those who
stayed on an AI + palbociclib (median PFS 11.9 months
[95% CI 9.1–13.6 months] versus 5.7 months [95% CI
3.9–7.5months]; stratified HR 0.61 [95% CI 0.43–0.86],
two-sided P = 0.004) [26]. More data on mutational evo-
lution is needed to determine whether similar strate-
gies employing ctDNA to inform change in therapy will
be broadly applicable across breast cancer subtypes and
therapy classes in order to further prolong OS.
Although a cross comparison of studies can be prob-
lematic, phase II and III studies suggest that as patients
are more heavily treated with anti-HER2 regimens, the
PFS and ORR decrease with each subsequent anti-HER2
therapy [2730]. However, in a phase III randomized trial
of trastuzumab deruxtecan (T-DXd) versus trastuzumab
emtansine (T-DM1) in patients (N = 524) whose disease
progressed on anti-HER2 therapy, the reported ORR for
patients treated with T-DXd or T-DM1 were 79.7% ver-
sus 34.2%, respectively. e landmark analysis of PFS at
12months was 75.8% with T-DXd as compared to 34.1%
with T-DM1 (HR 0.28, 95% CI 0.22–0.37; P < 0.001)
[6]. Although both T-DXd and T-DM1 have a trastu-
zumab backbone, there are substantial differences in
the linker-payload chemistry, which favors an increased
intracellular payload and a bystander effect with T-DXd
[31, 32].
We have shown in our study that the benefit from
T-DM1 + neratinib is limited in HER2-non-amplified
tumors. is finding is consistent with a study reporting
a PFS with T-DM1 of 1.5 months [33] in patients discord-
ant for HER2 in primary and metastatic tissue (HER2-
positive/negative). Although loss of HER2-amplification
appears to be one mechanism of resistance to T-DM1,
half of the patients with HER2-amplified tumors did not
respond to T-DM1 + neratinib, indicating that resistance
to T-DM1 is not limited to loss of HER2-amplification.
Many other mechanisms of resistance to T-DM1 have
been proposed, such as altered cellular uptake, intracel-
lular transport, and metabolism of the payload. [32]
Based on the low level of activity of T-DM1 mono-
therapy in patients failing HP, we speculate that the
combination of T-DM1 and neratinib is more effective
than T-DM1 monotherapy. We are unaware of any tri-
als in HER2-positive breast cancer in which patients
progressing on HP have been randomized to compare
the response to T-DM1 as monotherapy with a com-
bination of T-DM1 and an irreversible tyrosine kinase
inhibitor (TKI). However, in preclinical lung models with
ERBB2 mutation and/or amplification, the combination
of T-DM1 + neratinib did show increased efficacy over
monotherapy. Anecdotally, enhanced efficacy was dem-
onstrated in a breast cancer patient progressing on mon-
otherapy with T-DM1 who then responded with addition
of neratinib. e mechanism of action of the antibody–
drug conjugates (ADC) such as T-DM1 and T-DXd
involves the recognition and binding of the trastuzumab
backbone to the extracellular HER2 surface receptor. e
ADC-protein complex is internalized with cleavage of the
cytotoxic payload. Irreversible TKIs such as neratinib and
afatinib (unlike reversible TKIs such as lapatinib) have
been shown to enhance HER2 internalization and lysoso-
mal sorting, which has the potential to increase uptake of
bound ADC and release of their cytotoxic payload [34].
e KATHERINE [6] study established T-DM1 as a
standard of care in early HER2-positive breast cancer
patients with residual disease after neoadjuvant therapy
[1]. DESTINY-Breast03 has clearly shown superior-
ity of T-DXd over T-DM1 in HER2-positive metastatic
disease. e currently recruiting DESTINY-Breast05
study will compare T-DXd with T-DM1 in high-risk
HER2-positive patients with residual disease following
NAC-HP (NCT04622319). Another study, DESTINY-
Breast06 (NCT04494425), will address the question
of HER2-low (IHC 1 + or IHC 2 + /FISH-negative or
HER2 IHC > 0, < 1 +) in patients with metastatic hor-
mone-positive disease with progression on at least
two lines of endocrine therapy comparing T-DXd with
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 11 of 13
Jacobsetal. Breast Cancer Research (2024) 26:69
investigator’s choice of chemotherapy [31]. e 30-40%
of HER2-positive patients with residual or metastatic
disease after neoadjuvant therapy who have lost HER2
amplification, while not directly being addressed with
these ADCs studies, warrant further investigation with
newer generation ADCs.
Our study has several limitations including the non-
randomized design, the logistic difficulties in obtain-
ing samples of blood and tissue on all patients and
the small sample size, which limited its power and the
ability to perform multivariant analysis. However, the
strengths of our study include the multiple temporal
sample collections, multiple assessments of HER2 sta-
tus, and molecular assessment of DNA in both tissue
and blood.
Despite the limitations of our study, the findings have
generated several hypotheses that should be further
investigated. First, retrospective confirmation in a large
phase III study that loss of HER2-expression under the
pressure of therapy can be detected with liquid biopsy;
second, the overall response, depth, and duration of
response to anti-HER2 therapy is greater in patients
with HER2-amplified than in non-amplified patients;
third, the activity of T-DM1 or other ADCs with a tras-
tuzumab-backbone may be enhanced with addition of
an irreversible TKI such as neratinib. is hypothesis,
testing the interaction of an ADC with a TKI, reversible
and irreversible, could be evaluated in patient-derived
xenografts or other model systems and should be vali-
dated prior to a randomized trial. Finally, a small frac-
tion of HER2-nonamplified patients did benefit from
T-DM1 + neratinib. Possible explanations, which require
further investigation, include a false negative assay,
enhanced internalization of T-DM1 in presence of ner-
atinib, or EGFR becoming the driver in patients with loss
of HER2-amplification, and inhibition by neratinib [32
34]. We also realize that a low ctDNA fraction could have
prevented the detection of ctHER2 amplification.
Gene expression analysis revealed several important
observations. First, the level of HER2 RNA expression
in TP1 tissues was closely correlated with the response
rate to study therapy. Second, non-luminal subtypes had
a better response rate than luminal tumors, although
this difference was not statistically significant. ird,
there was a non-significant association of the 8-gene
trastuzumab benefit groups with the rate of responses
to study therapy. Fourth, changes in intrinsic subtypes
were seen between TP0 and TP1 tissue samples, indicat-
ing that these changes may be a result of tumors evolv-
ing to become resistant to HP. ese results highlight
the importance of collecting and monitoring molecular
changes in tissue samples as patients move through their
treatments.
PIK3CA mutations are a known oncogenic driver in
breast cancer and drive therapeutic resistance in mul-
tiple HER2-targeted therapies [35]. In the EMILIA
trial, patients with PIK3CA mutations, treated with
capecitabine + lapatinib were associated with a shorter
PFS than were patients with wild-type tumors; how-
ever, in patients treated with T-DM1 this was not the
case [36]. We likewise see that in patients treated with
T-DM1 + neratinib, PIK3CA mutations were not asso-
ciated with outcomes. However, we cannot rule out
the possibility that a subset of patients, refractory to
T-DM1 + neratinib with PIK3CA mutations, may be
responsive to PIK3CA inhibitors.
Conclusions
We demonstrate the usefulness of serial assessment of
HER2 status in blood and tissue in patients with an ini-
tial diagnosis of HER2-positive disease. Loss of HER2
amplification in ctDNA, or the complete loss of ctDNA
on treatment with T-DM1 + neratinib, was associated
with clinical benefit. Further, we show that many of the
patients with short-lived PR or PD were HER2-low in
tissue. ese patients may be better treated with the
recently approved ADC, trastuzumab deruxtecan. We
observed that the ADC, T-DM1, plus neratinib, was
well tolerated. e combination with an irreversible
tyrosine kinase inhibitor with other ADCs warrants
investigation.
Supplementary Information
The online version contains supplementary material available at https:// doi.
org/ 10. 1186/ s13058‑ 024‑ 01823‑8.
Additional le1. Additional Patient information and Methodological
Details.
Additional le2. All Molecular and Response Data Information for NSABP
FB‑10 Patients.
Acknowledgments
We would also like to thank Wendy L. Rea, BA, for editing the manuscript.
Author contributions
Conception &/or Design: SAJ, YW, JA, HF, CL, KPG. Acquisition (of pts/materials)
&/or Analysis: All authors: SAJ, YW, JA, HF, AJM, CL, MF, RCJ, AMS, SKM, SLP, FP,
KQ, KC, RJN, CJA, KV, NW, PCL, AS, KP‑G. Interpretation of the data: SAJ, YW, HF,
FP, KQ, AS, KP‑G. Has drafted the work or substantively revised it: SAJ, YW, AS,
KP‑G.
Funding
We would like to thank our funders BCRF (CONS‑20‑009), Guardant Health Inc.,
Puma Biotechnology, Inc., and the NSABP Foundation. The NSABP Foundation
received funding from Puma Biotechnology to conduct the clinical trial and
for the collection of tissues and blood samples associated with this clinical
trial. No authors received any direct funding for this research, but indirectly
received salary support for efforts to conduct this research. The funder played
no role in the design of the study, or collection, analysis, or interpretation of
the data, or in the writing of the manuscript or submission thereof.
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 12 of 13
Jacobsetal. Breast Cancer Research (2024) 26:69
Availability of data and materials
Anonymized individual participant data that underlie the results reported
in this article will be available in dbGAP or other publicly available site after
publication.
Declarations
Ethics approval and consent to participate
Central IRB approval provided by Adverra IRB, Columbia, MD.
Competing interests
AJ Montero: Honoraria: Celgene, AstraZeneca, OncoSec; Consulting/Advisory
Role: New Century Health, Welwaze, Paragon healthcare; Research Funding:
F. Hoffmann‑La Roche Ltd, Basel, Switzerland; Uncompensated: Roche; Open
Payments: https:// openp aymen tsdata. cms. gov/ physi cian/ 618396. K Quinn:
Guardant Health Shareholder. K Chang: Guardant Health Shareholder. RJ Nagy :
Guardant Health Shareholder. PC Lucas: Equity interest in AMGEN outside
the submitted work. KL Pogue‑Geile: Consulting for Bluestar BioAdvisors and
Provisional Patents filed both outside the submitted work. All other authors
declare no other potential conflicts of interest.
Author details
1 NSABP Foundation, Pittsburgh, PA, USA. 2 Cleveland Clinic, Weston/Taussig
Cancer Institute, Cleveland, OH, USA. 3 University Hospitals/Seidman Cancer
Center, Case Western Reserve University, Cleveland, OH, USA. 4 University
of Pittsburgh, Pittsburgh, PA, USA. 5 Present Address: University of Pennsylva‑
nia Perelman School of Medicine, State College, PA, USA. 6 National Institutes
of Health, Washington, DC, USA. 7 Present Address: Virginia Cancer Specialists,
Fairfax, VA, USA. 8 UPMC Hillman Cancer Center, Pittsburgh, PA, USA. 9 University
of Pittsburgh School of Medicine, Pittsburgh, PA, USA. 10 International Drug
Development Institute, Louvain‑la‑Neuve, Belgium. 11 Guardant Health,
Redwood City, CA, USA. 12 University of Florida Health, Gainesville, FL, USA.
13 Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester,
MN, USA. 14 Autism Impact Fund, Pittsburgh, PA, USA.
Received: 5 October 2023 Accepted: 11 April 2024
Published: 22 April 2024
References
1. von Minckwitz G, Huang CS, Mano MS, et al. Trastuzumab emtan‑
sine for residual invasive HER2‑positive breast cancer. N Engl J Med.
2019;380:617–28.
2. Niikura N, Liu J, Hayashi N, et al. Loss of human epidermal growth factor
receptor 2 (HER2) expression in metastatic sites of HER2‑overexpressing
primary breast tumors. J Clin Oncol. 2012;30:593–9.
3. Mittendorf EA, Wu Y, Scaltriti M, et al. Loss of HER2 amplification fol‑
lowing trastuzumab‑based neoadjuvant systemic therapy and survival
outcomes. Clin Cancer Res Off J Am Assoc Cancer Res. 2009;15:7381–8.
4. Ferraro E, Safonov A, Wen HY, et al. Abstract P2‑13‑06: clinical implica‑
tion of HER2 status change after neoadjuvant chemotherapy with
Trastuzumab and Pertuzumab (HP) in patients with HER2‑positive breast
cancer. Cancer Res. 2022;82:2–13.
5. Modi S, Saura C, Yamashita T, et al. Trastuzumab deruxtecan in previously
treated HER2‑positive breast cancer. N Engl J Med. 2020;382:610–21.
6. Cortes J, Kim SB, Chung WP, et al. Trastuzumab deruxtecan versus trastu‑
zumab emtansine for breast cancer. N Engl J Med. 2022;386:1143–54.
7. Modi S, Park H, Murthy RK, et al. Antitumor activity and safety of
trastuzumab deruxtecan in patients with HER2‑low‑expressing
advanced breast cancer: results from a phase Ib study. J Clin Oncol.
2020;38:1887–96.
8. Modi S, Jacot W, Yamashita T, et al. Trastuzumab deruxtecan in previously
treated HER2‑low advanced breast cancer. N Engl J Med. 2022;387:9–20.
9. Fehrenbacher L, Cecchini RS, Geyer CE Jr, et al. NSABP B‑47/NRG oncol‑
ogy phase III randomized trial comparing adjuvant chemotherapy with
or without trastuzumab in high‑risk invasive breast cancer negative for
HER2 by FISH and with IHC 1+ or 2. J Clin Oncol. 2020;38:444–53.
10. Abraham J, Montero AJ, Jankowitz RC, et al. Safety and efficacy of T‑DM1
plus neratinib in patients with metastatic HER2‑positive breast cancer:
NSABP foundation trial FB‑10. J Clin Oncol. 2019;37:2601–9.
11. Thompson JC, Carpenter EL, Silva BA, et al. Serial monitoring of circulat‑
ing tumor DNA by next‑generation gene sequencing as a biomarker
of response and survival in patients with advanced NSCLC receiving
pembrolizumab‑based therapy. JCO Precis Oncol. 2021;5:510–24.
12. Zhang J‑T, Liu S‑Y, Gao W, et al. Longitudinal undetectable molecular
residual disease defines potentially cured population in localized non‑
small cell lung cancer. Cancer Discov. 2022;12:1690–701.
13. Paik PK, Felip E, Veillon R, et al. Tepotinib in non–small‑cell lung cancer
with MET exon 14 skipping mutations. N Engl J Med. 2020;383:931–43.
14. Martínez‑Sáez O, Pascual T, Brasó‑Maristany F, et al. 5P Circulating
tumour DNA (ctDNA) dynamics using a standardized multi‑gene panel
in advanced breast cancer patients (pts) treated with CDK4/6 inhibitors
(CDK4/6i). Ann Oncol. 2020;31:S17.
15. Paquet ER, Hallett MT. Absolute assignment of breast cancer intrinsic
molecular subtype. J Natl Cancer Inst. 2015;107:357.
16. Pogue‑Geile KL, Kim C, Jeong JH, et al. Predicting degree of benefit
from adjuvant trastuzumab in NSABP trial B‑31. J Natl Cancer Inst.
2013;105:1782–8.
17. Pogue‑Geile KL, Song N, Serie DJ, et al. Validation of the NSABP/NRG
oncology 8‑gene trastuzumab‑benefit signature in alliance/NCCTG
N9831. JNCI Cancer Spectr. 2020. https:// doi. org/ 10. 1093/ jncics/ pkaa0 58.
18. Gaibar M, Beltrán L, Romero‑Lorca A, et al. Somatic mutations in HER2
and implications for current treatment paradigms in HER2‑positive breast
cancer. J Oncol. 2020;2020:6375956.
19. Ng CK, Martelotto LG, Gauthier A, et al. Intra‑tumor genetic heterogeneity
and alternative driver genetic alterations in breast cancers with heteroge‑
neous HER2 gene amplification. Genome Biol. 2015;16:107.
20. Nguyen TT, Hamdan D, Angeli E, et al. Genomics of breast cancer brain
metastases: a meta‑analysis and therapeutic implications. Cancers (Basel).
2023;15:1728.
21. Jacobs SA, Robidoux A, Abraham J, et al. NSABP FB‑7: a phase II rand‑
omized neoadjuvant trial with paclitaxel + trastuzumab and/or neratinib
followed by chemotherapy and postoperative trastuzumab in HER2+
breast cancer. Breast Cancer Res. 2019;21:133.
22. Pogue‑Geile K, Wang Y, Feng H, et al. Association of molecular signatures,
mutations, and sTILs, with pCR in breast cancer patients in NRG Oncol‑
ogy/NSABP B‑52. Cancer Res. 2019;79:4064.
23. Dieras V, Deluche E, Lusque A. Trastuzumab deruxtecan for advanced
breast cancer patients, regardless of HER2 status: a phase II study with
biomarkers analysis (DAISY). SABCS:PD8‑02, 2021
24. Lee HJ, Kim JY, Park SY, et al. Clinicopathologic significance of the intratu‑
moral heterogeneity of HER2 gene amplification in HER2‑positive breast
cancer patients treated with adjuvant trastuzumab. Am J Clin Pathol.
2015;144:570–8.
25. Nakamura Y, Taniguchi H, Ikeda M, et al. Clinical utility of circulating tumor
DNA sequencing in advanced gastrointestinal cancer: SCRUM‑Japan GI‑
SCREEN and GOZILA studies. Nat Med. 2020;26:1859–64.
26. Bidard F‑C, Hardy‑Bessard A‑C, Dalenc F, et al. Switch to fulvestrant and
palbociclib versus no switch in advanced breast cancer with rising
<em>ESR1</em> mutation during aromatase inhibitor and palbociclib
therapy (PADA‑1): a randomised, open‑label, multicentre, phase 3 trial.
Lancet Oncol. 2022;23:1367–77.
27. Verma S, Miles D, Gianni L, et al. Trastuzumab emtansine for HER2‑positive
advanced breast cancer. N Engl J Med. 2012;367:1783–91.
28. Krop IE, Kim SB, Gonzalez‑Martin A, et al. Trastuzumab emtansine versus
treatment of physician’s choice for pretreated HER2‑positive advanced
breast cancer (TH3RESA): a randomised, open‑label, phase 3 trial. Lancet
Oncol. 2014;15:689–99.
29. Conte B, Fabi A, Poggio F, et al. T‑DM1 efficacy in patients with HER2‑posi‑
tive metastatic breast cancer progressing after a taxane plus pertuzumab
and trastuzumab: an Italian multicenter observational study. Clin Breast
Cancer. 2020;20:e181–7.
30. Yokoe T, Kurozumi S, Nozawa K, et al. Clinical benefit of treatment after
trastuzumab emtansine for HER2‑positive metastatic breast cancer: a
real‑world multi‑centre cohort study in Japan (WJOG12519B). Breast
Cancer. 2021;28:581–91.
31. Bardia A, Barrios C, Dent R et al: Abstract OT‑03‑09: Trastuzumab der‑
uxtecan (T‑DXd; DS‑8201) vs investigator’s choice of chemotherapy in
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 13 of 13
Jacobsetal. Breast Cancer Research (2024) 26:69
patients with hormone receptor‑positive (HR+), HER2 low metastatic
breast cancer whose disease has progressed on endocrine therapy in the
metastatic setting: a randomized, global phase 3 trial (DESTINY‑Breast06).
Cancer Res. 2021; 81:OT‑03‑09‑OT‑03‑09
32. Hunter FW, Barker HR, Lipert B, et al. Mechanisms of resistance to trastu‑
zumab emtansine (T‑DM1) in HER2‑positive breast cancer. Br J Cancer.
2020;122:603–12.
33. Van Raemdonck E, Floris G, Berteloot P, et al. Efficacy of anti‑HER2
therapy in metastatic breast cancer by discordance of HER2 expression
between primary and metastatic breast cancer. Breast Cancer Res Treat.
2021;185:183–94.
34. Li BT, Michelini F, Misale S, et al. HER2‑mediated internalization of cyto‑
toxic agents in ERBB2 amplified or mutant lung cancers. Cancer Discov.
2020;10:674–87.
35. Rasti AR, Guimaraes‑Young A, Datko F, et al. PIK3CA mutations drive thera‑
peutic resistance in human epidermal growth factor receptor 2–positive
breast cancer. JCO Precis Oncol. 2022;6:e2100370.
36. Baselga J, Lewis Phillips GD, Verma S, et al. Relationship between tumor
biomarkers and efficacy in EMILIA, a phase III study of trastuzumab
emtansine in HER2‑positive metastatic breast cancer. Clin Cancer Res.
2016;22:3755–63.
Publisher’s Note
Springer Nature remains neutral with regard to jurisdictional claims in pub‑
lished maps and institutional affiliations.
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
1.
2.
3.
4.
5.
6.
Terms and Conditions
Springer Nature journal content, brought to you courtesy of Springer Nature Customer Service Center GmbH (“Springer Nature”).
Springer Nature supports a reasonable amount of sharing of research papers by authors, subscribers and authorised users (“Users”), for small-
scale personal, non-commercial use provided that all copyright, trade and service marks and other proprietary notices are maintained. By
accessing, sharing, receiving or otherwise using the Springer Nature journal content you agree to these terms of use (“Terms”). For these
purposes, Springer Nature considers academic use (by researchers and students) to be non-commercial.
These Terms are supplementary and will apply in addition to any applicable website terms and conditions, a relevant site licence or a personal
subscription. These Terms will prevail over any conflict or ambiguity with regards to the relevant terms, a site licence or a personal subscription
(to the extent of the conflict or ambiguity only). For Creative Commons-licensed articles, the terms of the Creative Commons license used will
apply.
We collect and use personal data to provide access to the Springer Nature journal content. We may also use these personal data internally within
ResearchGate and Springer Nature and as agreed share it, in an anonymised way, for purposes of tracking, analysis and reporting. We will not
otherwise disclose your personal data outside the ResearchGate or the Springer Nature group of companies unless we have your permission as
detailed in the Privacy Policy.
While Users may use the Springer Nature journal content for small scale, personal non-commercial use, it is important to note that Users may
not:
use such content for the purpose of providing other users with access on a regular or large scale basis or as a means to circumvent access
control;
use such content where to do so would be considered a criminal or statutory offence in any jurisdiction, or gives rise to civil liability, or is
otherwise unlawful;
falsely or misleadingly imply or suggest endorsement, approval , sponsorship, or association unless explicitly agreed to by Springer Nature in
writing;
use bots or other automated methods to access the content or redirect messages
override any security feature or exclusionary protocol; or
share the content in order to create substitute for Springer Nature products or services or a systematic database of Springer Nature journal
content.
In line with the restriction against commercial use, Springer Nature does not permit the creation of a product or service that creates revenue,
royalties, rent or income from our content or its inclusion as part of a paid for service or for other commercial gain. Springer Nature journal
content cannot be used for inter-library loans and librarians may not upload Springer Nature journal content on a large scale into their, or any
other, institutional repository.
These terms of use are reviewed regularly and may be amended at any time. Springer Nature is not obligated to publish any information or
content on this website and may remove it or features or functionality at our sole discretion, at any time with or without notice. Springer Nature
may revoke this licence to you at any time and remove access to any copies of the Springer Nature journal content which have been saved.
To the fullest extent permitted by law, Springer Nature makes no warranties, representations or guarantees to Users, either express or implied
with respect to the Springer nature journal content and all parties disclaim and waive any implied warranties or warranties imposed by law,
including merchantability or fitness for any particular purpose.
Please note that these rights do not automatically extend to content, data or other material published by Springer Nature that may be licensed
from third parties.
If you would like to use or distribute our Springer Nature journal content to a wider audience or on a regular basis or in any other manner not
expressly permitted by these Terms, please contact Springer Nature at
onlineservice@springernature.com
ResearchGate has not been able to resolve any citations for this publication.
Article
Full-text available
Breast cancer brain metastases are a challenging daily practice, and the biological link between gene mutations and metastatic spread to the brain remains to be determined. Here, we performed a meta-analysis on genomic data obtained from primary tumors, extracerebral metastases and brain metastases, to identify gene alterations associated with metastatic processes in the brain. Articles with relevant findings were selected using Medline via PubMed, from January 1999 up to February 2022. A critical review was conducted according to the Preferred Reporting Items for Systematic Review and Meta-analysis statement (PRISMA). Fifty-seven publications were selected for this meta-analysis, including 37,218 patients in all, 11,906 primary tumor samples, 5541 extracerebral metastasis samples, and 1485 brain metastasis samples. We report the overall and sub-group prevalence of gene mutations, including comparisons between primary tumors, extracerebral metastases and brain metastases. In particular, we identified six genes with a higher mutation prevalence in brain metastases than in extracerebral metastases, with a potential role in metastatic processes in the brain: ESR1, ERBB2, EGFR, PTEN, BRCA2 and NOTCH1. We discuss here the therapeutic implications. Our results underline the added value of obtaining biopsies from brain metastases to fully explore their biology, in order to develop personalized treatments.
Article
Full-text available
Among breast cancers without human epidermal growth factor receptor 2 (HER2) amplification, overexpression, or both, a large proportion express low levels of HER2 that may be targetable. Currently available HER2-directed therapies have been ineffective in patients with these “HER2-low” cancers. METHODS We conducted a phase 3 trial involving patients with HER2-low metastatic breast cancer who had received one or two previous lines of chemotherapy. (Low expression of HER2 was defined as a score of 1+ on immunohistochemical [IHC] analysis or as an IHC score of 2+ and negative results on in situ hybridization.) Patients were randomly assigned in a 2:1 ratio to receive trastuzumab deruxtecan or the physician’s choice of chemotherapy. The primary end point was progression-free survival in the hormone receptor–positive cohort. The key secondary end points were progression-free survival among all patients and overall survival in the hormone receptor–positive cohort and among all patients. RESULTS Of 557 patients who underwent randomization, 494 (88.7%) had hormone receptor–positive disease and 63 (11.3%) had hormone receptor–negative disease. In the hormone receptor–positive cohort, the median progression-free survival was 10.1 months in the trastuzumab deruxtecan group and 5.4 months in the physician’s choice group (hazard ratio for disease progression or death, 0.51; P<0.001), and overall survival was 23.9 months and 17.5 months, respectively (hazard ratio for death, 0.64; P=0.003). Among all patients, the median progression-free survival was 9.9 months in the trastuzumab deruxtecan group and 5.1 months in the physician’s choice group (hazard ratio for disease progression or death, 0.50; P<0.001), and overall survival was 23.4 months and 16.8 months, respectively (hazard ratio for death, 0.64; P=0.001). Adverse events of grade 3 or higher occurred in 52.6% of the patients who received trastuzumab deruxtecan and 67.4% of those who received the physician’s choice of chemotherapy. Adjudicated, drug-related interstitial lung disease or pneumonitis occurred in 12.1% of the patients who received trastuzumab deruxtecan; 0.8% had grade 5 events. CONCLUSIONS In this trial involving patients with HER2-low metastatic breast cancer, trastuzumab deruxtecan resulted in significantly longer progression-free and overall survival than the physician’s choice of chemotherapy. (Funded by Daiichi Sankyo and AstraZeneca; DESTINY-Breast04 ClinicalTrials.gov number, NCT03734029. opens in new tab.)
Article
Full-text available
The efficacy and potential limitations of molecular residual disease (MRD) detection urgently need to be fully elucidated in a larger population of non–small cell lung cancer (NSCLC). We enrolled 261 patients with stages I to III NSCLC who underwent definitive surgery, and 913 peripheral blood samples were successfully detected by MRD assay. Within the population, only six patients (3.2%) with longitudinal undetectable MRD recurred, resulting in a negative predictive value of 96.8%. Longitudinal undetectable MRD may define the patients who were cured. The peak risk of developing detectable MRD was approximately 18 months after landmark detection. Correspondingly, the positive predictive value of longitudinal detectable MRD was 89.1%, with a median lead time of 3.4 months. However, brain-only recurrence was less commonly detected by MRD (n = 1/5, 20%). Further subgroup analyses revealed that patients with undetectable MRD might not benefit from adjuvant therapy. Together, these results expound the value of MRD in NSCLC. Significance This study confirms the prognostic value of MRD detection in patients with NSCLC after definitive surgery, especially in those with longitudinal undetectable MRD, which might represent the potentially cured population regardless of stage and adjuvant therapy. Moreover, the risk of developing detectable MRD decreased stepwise after 18 months since landmark detection.
Article
Full-text available
The phosphatidylinositol 3-kinase (PI3K) pathway is an intracellular pathway activated in response to progrowth signaling, such as human epidermal growth factor receptor 2 (HER2) and other kinases. Abnormal activation of PI3K has long been recognized as one of the main oncogenic drivers in breast cancer, including HER2-positive (HER2+) subtype. Somatic activating mutations in the gene encoding PI3K alpha catalytic subunit ( PIK3CA) are present in approximately 30% of early-stage HER2+ tumors and drive therapeutic resistance to multiple HER2-targeted agents. Here, we review currently available agents targeting PI3K, discuss their potential role in HER2+ breast cancer, and provide an overview of ongoing trials of PI3K inhibitors in HER2+ disease. Additionally, we review the landscape of PIK3CA mutational testing and highlight the gaps in knowledge that could present potential barriers in the effective application of PI3K inhibitors for treatment of HER2+ breast cancer.
Article
Full-text available
Background: Trastuzumab emtansine is the current standard treatment for patients with human epidermal growth factor receptor 2 (HER2)-positive metastatic breast cancer whose disease progresses after treatment with a combination of anti-HER2 antibodies and a taxane. Methods: We conducted a phase 3, multicenter, open-label, randomized trial to compare the efficacy and safety of trastuzumab deruxtecan (a HER2 antibody-drug conjugate) with those of trastuzumab emtansine in patients with HER2-positive metastatic breast cancer previously treated with trastuzumab and a taxane. The primary end point was progression-free survival (as determined by blinded independent central review); secondary end points included overall survival, objective response, and safety. Results: Among 524 randomly assigned patients, the percentage of those who were alive without disease progression at 12 months was 75.8% (95% confidence interval [CI], 69.8 to 80.7) with trastuzumab deruxtecan and 34.1% (95% CI, 27.7 to 40.5) with trastuzumab emtansine (hazard ratio for progression or death from any cause, 0.28; 95% CI, 0.22 to 0.37; P<0.001). The percentage of patients who were alive at 12 months was 94.1% (95% CI, 90.3 to 96.4) with trastuzumab deruxtecan and 85.9% (95% CI, 80.9 to 89.7) with trastuzumab emtansine (hazard ratio for death, 0.55; 95% CI, 0.36 to 0.86; prespecified significance boundary not reached). An overall response (a complete or partial response) occurred in 79.7% (95% CI, 74.3 to 84.4) of the patients who received trastuzumab deruxtecan and in 34.2% (95% CI, 28.5 to 40.3) of those who received trastuzumab emtansine. The incidence of drug-related adverse events of any grade was 98.1% with trastuzumab deruxtecan and 86.6% with trastuzumab emtansine, and the incidence of drug-related adverse events of grade 3 or 4 was 45.1% and 39.8%, respectively. Adjudicated drug-related interstitial lung disease or pneumonitis occurred in 10.5% of the patients in the trastuzumab deruxtecan group and in 1.9% of those in the trastuzumab emtansine group; none of these events were of grade 4 or 5. Conclusions: Among patients with HER2-positive metastatic breast cancer previously treated with trastuzumab and a taxane, the risk of disease progression or death was lower among those who received trastuzumab deruxtecan than among those who received trastuzumab emtansine. Treatment with trastuzumab deruxtecan was associated with interstitial lung disease and pneumonitis. (Funded by Daiichi Sankyo and AstraZeneca; DESTINY-Breast03 ClinicalTrials.gov number, NCT03529110.).
Article
Background: The HER2-targeted antibody-drug conjugate (ADC) trastuzumab deruxtecan (T-DXd) demonstrated efficacy in heavily pretreated HER2-over- and HER2-low expressing ABC (1, 2). We aimed to assess the activity of T-DXd in HER2-over-, HER2-low and HER2-nul expressing ABC, to describe the drug mechanisms of action in the 3 cohorts and to identify biomarkers associated to drug response or resistance. Study Description: DAISY is a multicenter, open-label phase II trial designed to assess the efficacy of single agent T-DXd at 5.4 mg/kg dose in ABC with extensive biomarkers analysis. Three cohorts of patients were included: Cohort 1 (HER2 over-expressing: HER2 3+ on immunohistochemistry (IHC) or HER2 IHC2+/in situ hybridization [ISH]+), Cohort 2 (HER2 low-expressing: IHC1+ or IHC2+/ISH-) and cohort 3 (HER2-nul: IHC0+). Biopsy of metastatic sites was performed: at baseline, on treatment (mandatory for cohort 1, optional for cohort 2/3) and at tumor progression; blood samples for ctDNA were collected at baseline. The primary endpoint was the Best Overall Response (BOR) in each cohort, according to the investigator assessment. Secondary endpoints were BOR by central assessment, clinical benefit rate, duration of response (DOR), progression-free (PFS), overall survival (OS) and safety. Results:185 women and 1 man were enrolled between November 2019 and March 2021. Among the patients enrolled in the safety population (see Table 1), median (range) age was 55 (24-82) years, all received at least one prior line of therapy and 12 patients were TN. Table 2 shows investigator-reported T-Dxd activity in the 3 cohorts at a median follow-up of 10.1 months [95%CI: 9.2-11.1]. A total of 170 patients (95%) had at least one treatment-related toxicity. Key grade ≥3 treatment-related toxicities included neutropenia (10.6% of patients), fatigue (5.6%), leucopenia (4.5%), vomiting (4.5%) and anemia (3.4%). A total of 4 patients had drug-related interstitial lung disease or pneumonitis (grade 1 in 3 patients and grade 2 in 1 patient), 11 patients discontinued treatment due to treatment-related adverse events. No drug-related deaths occurred. Conclusions: T-DXd showed clinically meaningful activity in patients with HER2-overexpressing ABC and interestingly also in those with HER2low and HER2-nul ABC. Safety profile was consistent with previous reports. 1.Modi S et al N Engl J Med 2020 2.Mosi S et al J Clin Oncol 2020 Table 1.Analysis populationsTotalCohort 1 (HER2 over-expressing)Cohort 2 (HER2 low-expressing)Cohort 3 (HER2 non-detected)Enrolled population186727440Safety population*179687338 (including 12 TN)Full analysis Set**176687236TN: Triple Negative. *: safety population = enrolled population except 7 patients who did not receive at least one dose of study drug. **: Full Analysis Set = safety population except 3 patients (2 who did not have a valid first post-baseline assessment of disease status or who did not have progressive disease and 1 who did not have at least one radiologically measurable lesion according to RECIST v1.1) Table 2.T-DXd activity in the three cohorts according to investigator assessmentTotalCohort 1Cohort 2Cohort 3BOR confirmedn/N82/176 (46.6%)47/68 (69.1%)24/72 (33.3%)11/36 (30.6%)[95%CI][39.1; 54.2][56.7; 79.8][22.7; 45.4]16.3; 48.1]Median DORmonths7.69.97.66.8[95%CI][6.2; 9.7][5.4; NR][4.4; 8.7][2.8; 8.3]Median PFSmonths6.911.16.74.2[95%CI][6.7; 8.7][8.4; NR][4.6; 8.5][2.1; 6.9]NR: Not Reached Citation Format: Véronique Diéras, Elise Deluche, Amélie Lusque, Barbara Pistilli, Thomas Bachelot, Jean-Yves Pierga, Frédéric Viret, Christelle Levy, Laura Salabert, Fanny Le Du, Florence Dalenc, Christelle Jouannaud, Laurence Venat-Bouvet, Jean-Philippe Jacquin, Xavier Durando, Thierry Petit, Céline Mahier - Aït Oukhatar, Thomas Filleron, Maria Fernanda Mosele, Magali Lacroix-Triki, Agnès Ducoulombier, Fabrice André. Trastuzumab deruxtecan (T-DXd) for advanced breast cancer patients (ABC), regardless HER2 status: A phase II study with biomarkers analysis (DAISY) [abstract]. In: Proceedings of the 2021 San Antonio Breast Cancer Symposium; 2021 Dec 7-10; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2022;82(4 Suppl):Abstract nr PD8-02.
Article
Background In advanced oestrogen receptor-positive, HER2-negative breast cancer, acquired resistance to aromatase inhibitors frequently stems from ESR1-mutated subclones, which might be sensitive to fulvestrant. The PADA-1 trial aimed to show the efficacy of an early change in therapy on the basis of a rising ESR1 mutation in blood (bESR1mut), while assessing the global safety of combination fulvestrant and palbociclib. Methods We did a randomised, open-label, phase 3 trial in 83 hospitals in France. Women aged at least 18 years with oestrogen receptor-positive, HER2-negative advanced breast cancer and an Eastern Cooperative Oncology Group performance status of 0–2 were recruited and monitored for rising bESR1mut during first-line aromatase inhibitor (2·5 mg letrozole, 1 mg anastrozole, or 25 mg exemestane, orally once per day, taken continuously) and palbociclib (125 mg orally once per day on days 1–21 of a 28-day cycle) therapy. Patients with newly present or increased bESR1mut in circulating tumour DNA and no synchronous disease progression were randomly assigned (1:1) to continue with the same therapy or to switch to fulvestrant (500 mg intramuscularly on day 1 of each 28-day cycle and on day 15 of cycle 1) and palbociclib (dosing unchanged). The randomisation sequence was generated within an interactive web response system using a minimisation method (with an 80% random factor); patients were stratified according to visceral involvement (present or absent) and the time from inclusion to bESR1mut detection (<12 months or ≥12 months). The co-primary endpoints were investigator-assessed progression-free survival from random assignment, analysed in the intention-to-treat population (ie, all randomly assigned patients), and grade 3 or worse haematological adverse events in all patients. The trial is registered with Clinicaltrials.gov (NCT03079011), and is now complete. Findings From March 22, 2017, to Jan 31, 2019, 1017 patients were included, of whom 279 (27%) developed a rising bESR1mut and 172 (17%) were randomly assigned to treatment: 88 to switching to fulvestrant and palbociclib and 84 patients to continuing aromatase inhibitor and palbociclib. At database lock on July 31, 2021, randomly assigned patients had a median follow-up of 35·3 months (IQR 29·2–41·4) from inclusion and 26·0 months (13·8–34·3) from random assignment. Median progression-free survival from random assignment was 11·9 months (95% CI 9·1–13·6) in the fulvestrant and palbociclib group versus 5·7 months (3·9–7·5) in the aromatase inhibitor and palbociclib group (stratified HR 0·61, 0·43–0·86; p=0·0040). The most frequent grade 3 or worse haematological adverse events were neutropenia (715 [70·3%] of 1017 patients), lymphopenia (66 [6·5%]), and thrombocytopenia (20 [2·0%]). The most common grade 3 or worse adverse events in step 2 were neutropenia (35 [41·7%] of 84 patients in the aromatase inhibitor and palbociclib group vs 39 [44·3%] of 88 patients in the fulvestrant and palbociclib group) and lymphopenia (three [3·6%] vs four [4·5%]). 31 (3·1%) patients had grade 3 or worse serious adverse events related to treatment in the overall population. Three (1·7%) of 172 patients randomly assigned had one serious adverse event in step 2: one (1·2%) grade 4 neutropenia and one (1·2%) grade 3 fatigue among 84 patients in the aromatase inhibitor and palbociclib group, and one (1·1%) grade 4 neutropenia among 88 patients in the fulvestrant and palbociclib group. One death by pulmonary embolism in step 1 was declared as being treatment related. Interpretation PADA-1 is the first prospective randomised trial showing that the early therapeutic targeting of bESR1mut results in significant clinical benefit. Additionally, the original design explored in PADA-1 might help with tackling acquired resistance with new drugs in future trials. Funding Pfizer.
Article
Background: Patients with early HER2-positive breast cancer (BC) and residual disease after HER2-targeted neoadjuvant chemotherapy (NAC) are at high risk of recurrence. It is estimated that 10-30% of HER2-positive breast cancers change HER2 status after trastuzumab alone, but the effects of adding pertuzumab on this phenomenon and clinical outcomes remain unclear. We previously reported a high rate (~50%) of HER2 status change after HP in a small subset of patients. Herein, we present an updated analysis incorporating pathological review of additional cases.Methods: We identified patients with HER2-positive BC who received NAC with pertuzumab and trastuzumab (NAC-HP) followed by surgery at our institution between September 1, 2013 to November 1, 2019. Patients with HER2 status performed either at MSKCC or outside institutions were included. Change in HER2 status on residual disease from baseline was evaluated. We defined HER2 positivity as immunohistochemistry (IHC) IHC3+ or IHC0-2+ FISH amplified (ratio ≥ 2 or ratio < 2 and HER2 copy number ≥ 6 signals/cell). HER2-low was defined as IHC 1+ or 2+, FISH non-amplified. Disease free survival (DFS) and overall survival (OS) were analyzed using the Kaplan-Meier method. Differences between patients with concordant and discordance HER2 status were assessed using the log-rank test.Results: Of 525 patients receiving NAC with HP, 229 (44%) patients had residual disease post NAC-HP. Among these 229 patients, 141 had both pre and post NAC-HP HER2 status available and were included in this analysis. HER2 status on biopsy specimens was determined at MSKCC in 35/141 (25%) and at external institution in 106/141 (75%). The majority of patients (84%) received dose-dense AC-THP; the remainder received TCHP or other HP-based regimens. Most (96%) of patients continued HP after surgery, and 2 patients received T-DM1. Of the 141 patients, 84/141(60%) were found to be HER2 concordant, while 57 (40%) were found to be HER2 discordant. In 13/57 (23%) patients, HER2 expression was lost (IHC 0), while in 44/57 (77%) patients, HER2-low profile was detected (IHC 1+ in 31, and IHC 2+, FISH non-amplified in 13). Further details are reported in the table. Patients with HER2 discordance after NAC-HP had similar survival outcome compared with patients who remained HER2 concordant (5-years DFS: 92.3% versus 88.7%, p=0.49 and 5-yr OS 93.6% versus 88.4%, p=0.70).Conclusions: In a single center cohort, discordant HER2 status after NAC-HP appeared frequently without statistically significant impact on survival outcome, although this finding may be due to the small size and hence low statistical power. Of these, HER2-low profile is the most frequent post treatment HER2 status change. This raises the possibility that patients with change in HER2 status may have heterogenous expression of HER2 at baseline, and HER2-loss or low sub-clones survive as residual disease due to the selection pressure of HP. Alternatively, anti-HER2 therapy may suppress HER2 expression in surviving cells. These findings could inform studies of tailored approaches in the post-neoadjuvant setting based on the biological profile of residual disease. Pre NAC-HP HER2 statusNPost NAC-HP HER2 statusNDiscordantN=57IHC 3+: 19IHC0: 4IHC1+: 9IHC2+ FISH not ampl: 6IHC 0-2+ FISH ampl: 38IHC0: 9IHC1+: 22IHC2+ FISH not ampl: 7Concordant N= 84IHC 3+: 59IHC 3+: 47IHC 0- 2+ FISH ampl: 12IHC 0-2+ FISH ampl: 25IHC 3+: 4IHC 0- 2+ FISH ampl: 21 Citation Format: Emanuela Ferraro, Anton Safonov, Hanna Y Wen, Edi Brogi, Mithat Gonan, Andrea V. Barrio, Pedram Razavi, Sarat Chandarlapaty, Shanu Modi, Andrew D. Seidman, Larry Norton, Mark E. Robson, Chau T. Dang. Clinical implication of HER2 status change after neoadjuvant chemotherapy with Trastuzumab and Pertuzumab (HP) in patients with HER2-positive breast cancer [abstract]. In: Proceedings of the 2021 San Antonio Breast Cancer Symposium; 2021 Dec 7-10; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2022;82(4 Suppl):Abstract nr P2-13-06.
Article
PURPOSE Although the majority of patients with metastatic non–small-cell lung cancer (mNSCLC) lacking a detectable targetable mutation will receive pembrolizumab-based therapy in the frontline setting, predicting which patients will experience a durable clinical benefit (DCB) remains challenging. MATERIALS AND METHODS Patients with mNSCLC receiving pembrolizumab monotherapy or in combination with chemotherapy underwent a 74-gene next-generation sequencing panel on blood samples obtained at baseline and at 9 weeks. The change in circulating tumor DNA levels on-therapy (molecular response) was quantified using a ratio calculation with response defined by a > 50% decrease in mean variant allele fraction. Patient response was assessed using RECIST 1.1; DCB was defined as complete or partial response or stable disease that lasted > 6 months. Progression-free survival and overall survival were recorded. RESULTS Among 67 patients, 51 (76.1%) had > 1 variant detected at a variant allele fraction > 0.3% and thus were eligible for calculation of molecular response from paired baseline and 9-week samples. Molecular response values were significantly lower in patients with an objective radiologic response (log mean 1.25% v 27.7%, P < .001). Patients achieving a DCB had significantly lower molecular response values compared to patients with no durable benefit (log mean 3.5% v 49.4%, P < .001). Molecular responders had significantly longer progression-free survival (hazard ratio, 0.25; 95% CI, 0.13 to 0.50) and overall survival (hazard ratio, 0.27; 95% CI, 0.12 to 0.64) compared with molecular nonresponders. CONCLUSION Molecular response assessment using circulating tumor DNA may serve as a noninvasive, on-therapy predictor of response to pembrolizumab-based therapy in addition to standard of care imaging in mNSCLC. This strategy requires validation in independent prospective studies.
Conference Paper
Background: HER2 directed therapies have substantially improved clinical outcomes in patients with HER2 positive (immunohistochemistry [IHC] 3+ or in situ hybridization [ISH] positive) metastatic breast cancer. However, no HER2 directed therapies are currently available for breast cancer with lower HER2 expression (IHC 2+/ISH− or IHC 1+). Patients with HER2 low and HR+ breast cancer typically receive initial treatment with endocrine therapy ± targeted therapies (CDK4/6, PI3K, or mTOR inhibitors). After disease progression, patients are treated with chemotherapy, which has shown limited clinical benefit. There is an unmet need for treatments that provide a superior risk-benefit profile compared with standard chemotherapy. T-DXd is an antibody-drug conjugate consisting of an anti-HER2 antibody, a cleavable tetrapeptide-based linker, and a membrane permeable topoisomerase I inhibitor payload. Results from a phase 1 study demonstrated promising antitumor activity with a confirmed objective response rate (ORR) of 37.0% (20 of 54) per independent central review (ICR) and a median progression-free survival (PFS) of 11.1 months in patients with heavily pretreated (median 7.5 prior regimens) HER2 low metastatic breast cancer. Among patients with HR+ disease, the ORR was 40.4% (19 of 47) per ICR (Modi S, et al. . 2020;38:1887-1896). Here, we describe a phase 3 trial evaluating the efficacy and safety of T-DXd vs chemotherapy in patients with HR+, HER2 low metastatic breast cancer that has progressed on prior endocrine therapy. In addition to the primary population of patients with HER2 low disease being studied, this trial will also study the efficacy and safety of T-DXd in an IHC > 0 < 1+ (detectable HER2 staining < 1+) population. Study Description: DESTINY-Breast06 is a global, randomized, multicenter, open-label, phase 3 trial designed to demonstrate superiority of T-DXd vs investigator’s choice of chemotherapy in patients with HR+, HER2 low metastatic breast cancer who had prior progression on endocrine therapy. Approximately 850 patients (HER2 low, n = 700; IHC > 0 < 1+, n = 150) from ≈ 300 centers globally will be randomized 1:1 to receive T-DXd 5.4 mg/kg every 3 weeks or investigator’s choice of chemotherapy (paclitaxel, nab-paclitaxel, or capecitabine) until disease progression, discontinuation due to intolerable toxicity, or death. Patients must have progression on ≥ 2 prior lines of endocrine therapy and cannot have received prior chemotherapy or any anti-HER2 therapy for metastatic disease. Randomization will be stratified by prior CDK4/6 inhibitor use (yes vs no), HER2 IHC expression (IHC2 +/ISH− vs IHC 1+ vs IHC > 0 < 1+), and prior taxane use in the non-metastatic setting (yes vs no). The primary endpoint is PFS per blinded ICR (BICR) in the HER2 low population. Key secondary endpoints are overall survival in the HER2 low and intent-to-treat (ITT; HER2 low and HER2 IHC > 0 < 1+) populations and PFS by BICR in the ITT population. Primary and key secondary endpoints will be tested in a hierarchical order. Other secondary endpoints are ORR by BICR and investigator assessment (according to RECIST 1.1), duration of response by BICR and investigator, time to second progression or death per investigator, time to first subsequent treatment or death, and time to second subsequent treatment or death (all in the HER2 low and ITT populations); PFS per investigator assessment in the HER2 low population; and safety, pharmacokinetics, patient-reported outcomes, and immunogenicity. Citation Format: Aditya Bardia, Carlos Barrios, Rebecca Dent, Xichun Hu, Joyce O’Shaughnessy, Kan Yonemori, Annie Darilay, Sarice Boston, Yufan Liu, Gargi Patel, Giuseppe Curigliano. Trastuzumab deruxtecan (T-DXd; DS-8201) vs investigator’s choice of chemotherapy in patients with hormone receptor-positive (HR+), HER2 low metastatic breast cancer whose disease has progressed on endocrine therapy in the metastatic setting: A randomized, global phase 3 trial (DESTINY-Breast06) [abstract]. In: Proceedings of the 2020 San Antonio Breast Cancer Virtual Symposium; 2020 Dec 8-11; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2021;81(4 Suppl):Abstract nr OT-03-09.