Clinical Implications of Erb-B2 Receptor Tyrosine Kinase 2 S310 Mutations in Non–Small-Cell Lung Cancer: Two Case Reports
CASE REPORT
Hong Kong J Radiol 2024;27:Epub 3 December 2024
Clinical Implications of Erb-B2 Receptor Tyrosine Kinase 2 S310 Mutations in Non–Small-Cell Lung Cancer: Two Case Reports
Q Du, TY Kam, MW Yeung
Department of Clinical Oncology, Pamela Youde Nethersole Eastern Hospital, Hong Kong SAR, China
Correspondence: Dr Q Du, Department of Clinical Oncology, Pamela Youde Nethersole Eastern Hospital, Hong Kong SAR, China. Email: dq083@ha.org.hk
Submitted: 4 January 2024; Accepted: 10 July 2024. This version may differ from the final version when published in an issue.
Contributors: QD and TYK designed the study. QD acquired and analysed the data and drafted the manuscript. All authors critically revised
the manuscript for important intellectual content. All authors had full access to the data, contributed to the study, approved the final version for
publication, and take responsibility for its accuracy and integrity.
Conflicts of Interest: All authors have disclosed no conflicts of interest.
Funding/Support: This study received no specific grant from any funding agency in the public, commercial, or not-for-profit sectors.
Data Availability: All data generated or analysed during the present study are available from the corresponding author on reasonable request.
Ethics Approval: The patients were treated in accordance with the Declaration of Helsinki. Informed consent for publication of the case reports was obtained from the patient and the patient’s family for Case 1 and Case 2, respectively.
Supplementary Material: The supplementary material was provided by the authors and some information may not have been peer reviewed. Any opinions or recommendations discussed are solely those of the author(s) and are not endorsed by the Hong Kong College of Radiologists. The
Hong Kong College of Radiologists disclaims all liability and responsibility arising from any reliance placed on the content.
INTRODUCTION
Alterations to Erb-B2 receptor tyrosine kinase 2
(ERBB2) have been reported in approximately 1%
to 4% of patients with non–small-cell lung cancer
(NSCLC).[1] [2] Several somatic mutations associated with
ERBB2 have been identified, with the most prevalent
subtype being in-frame insertion in exon 20, located
in the tyrosine kinase domain (TKD) of the human
epidermal growth factor receptor 2 (HER2) protein, and
accounting for 50% to 90% of cases.[3] [4] [5] [6] Other mutations
such as the ERBB2 exon 8 mutation (S310) in the
extracellular domain (ECD) have been infrequently
reported (11% among patients with ERBB2 mutation)
and remain underinvestigated.[3] [4] The latest National
Comprehensive Cancer Network guideline version
2024 recommends antibody-drug conjugates (ADCs) of
trastuzumab deruxtecan (T-DXd) and ado-trastuzumab
emtansine (T-DM1) for ERBB2-mutant NSCLC
patients progressing on platinum-based chemotherapy with or without immunotherapy.[7] The recommendation
for T-DXd is primarily based on the DESTINY-Lung01
and DESTINY-Lung02 studies that demonstrated a
promising overall response rate, duration of response
(DOR), overall survival, and favourable toxicity
profile.[7] [8] It is noteworthy that most patients recruited
in these two studies exhibited ERBB2 mutations located
in the TKD (93%-100%).[8] [9] Only a small proportion
of patients (2.9%, n = 3) in the DESTINY-Lung02
study presented ERBB2 exon 8 substitutions on the
ECD.9 To date, reports on the efficacy of ADCs on
the ERBB2 S310 mutation remain scarce.[3] We present
two patients with ERBB2 S310 mutations in different
clinical scenarios: the first had a de novo ERBB2 S310F
mutation on diagnosis; the second showed concurrent
ERBB2 S310Y and epidermal growth factor receptor
(EGFR) L858R mutations, and MET amplification upon
progression on first-line EGFR tyrosine kinase inhibitor
(TKI) therapy.
CASE PRESENTATIONS
Case 1
A 65-year-old male non-smoker with a medical
history of radically treated early-stage intrahepatic
cholangiocarcinoma and lung adenocarcinoma presented
with back pain in May 2022. Magnetic resonance imaging
(MRI) and positron emission tomography–computed
tomography (PET-CT) of the whole spine in August
2022 revealed new lytic bone metastasis involving the
L1-L2 vertebrae as well as right thoracic and mediastinal
lymph node metastasis (Figure 1a-c). Pathology of the
bone biopsy over the L2 vertebra confirmed metastasis
of primary lung adenocarcinoma to the bone (thyroid
transcription factor–1 positive). Tissue next-generation
sequencing (NGS) using the Oncomine Precision Assay
GX (Thermo Fisher Scientific, Waltham [MA], US)
revealed an ERBB2 S310F mutation, and the PD-L1
tumour proportion score was 0%.
Figure 1. Case 1. Serial positron emission tomography–computed tomography scans at baseline and following first and subsequent
lines of systemic treatment. (a) Right thoracic lymph node metastasis, (b) L2 bone metastasis and (c) manubrium bone metastasis at
baseline (August 2022). (d) Right thoracic lymph node metastasis, (e) L2 bone metastasis and (f) manubrium bone metastasis after cycle 4
pemetrexed/carboplatin/pembrolizumab (January 2023). (g) Right thoracic lymph node metastasis, (h) L2 bone metastasis and (i) manubrium
bone metastasis after cycle 6 pemetrexed/carboplatin/pembrolizumab (March 2023). (j) Right thoracic lymph node metastasis, (k) L2 bone
metastasis and (l) manubrium bone metastasis after cycle 9 maintenance pemetrexed/pembrolizumab (July 2023). (m) Right thoracic lymph
node metastasis, (n) L2 bone metastasis and (o) manubrium bone metastasis after cycle 4 trastuzumab deruxtecan (November 2023).
First-line treatment with pemetrexed (500 mg/m2
administered as an intravenous infusion on Day 1),
carboplatin (AUC 5 administered as an intravenous
infusion on Day 1), and pembrolizumab (200 mg
administered as an intravenous infusion on Day 1)
every 3 weeks was started in October 2022. PET-CT
after cycle 4 in January 2023 revealed increased
hypermetabolism in the right thoracic and mediastinal
lymph nodes as well as several new hypermetabolic
bone metastases involving the manubrium, ribs, pelvis,
and spine (Figure 1d-f). Notably, the carcinoembryonic
antigen (CEA) level decreased significantly from 198
ng/mL to 35.4 ng/mL within 3 months. Given the
improved clinical symptoms, the overall impression
was of immune unconfirmed progressive disease per
iRECIST (Immune-based Response Evaluation Criteria
in Solid Tumors) criteria,[10] and two additional cycles
of pemetrexed, carboplatin, and pembrolizumab were
administered (online supplementary Table).
Subsequent PET-CT in March 2023 showed a partial
response of the involved lymph nodes and largely
unchanged bone metastasis (Figure 1g-i). Therefore,
systemic treatment was switched to maintenance
pemetrexed and pembrolizumab from cycle 7 in April
2023. Worsening back pain was reported and the CEA
level had increased to 59.6 ng/mL in June 2023. Progress
PET-CT in July 2023 confirmed disease progression of
the right thoracic and mediastinal lymph nodes and bone
metastases (Figure 1j-l). Second-line systemic treatment
with T-DXd (5.4 mg/kg administered as an intravenous
infusion on Day 1 every 3 weeks) was initiated. The patient tolerated treatment well without significant side-effects.
Serial chest X-rays did not show evidence of
interstitial pneumonitis, even though he had previously
undergone stereotactic body radiation therapy to the lung
and immunotherapy. The CEA level rose to 214 ng/mL
after four cycles of T-DXd. PET-CT in November 2023
showed disease progression with new hypermetabolic
bone metastasis and mixed response of the previously
seen multiple mixed lytic and sclerotic bone metastases,
despite a partial response in the right thoracic and
mediastinal lymph nodes (Figure 1m-o). The current
plan is for the patient to receive third-line docetaxel
(online supplementary Table).
Case 2
A 79-year-old male non-smoker presented with left
hip pain in November 2022. PET-CT and MRI of the
brain revealed a left lower lobe lung tumour with
pleural, lymph node, liver, and bone metastases (Figure 2a-c). Pathology of lung biopsy indicated primary
adenosquamous carcinoma with lung (both thyroid
transcription factor–1 and p40 positive). Conventional
tissue polymerase chain reaction via the Idylla platform
(Biocartis, Mechelen, Belgium) revealed an EGFR
L858R mutation. The patient commenced EGFR TKI
erlotinib (150 mg daily) in December 2022 and CEA
level had decreased from baseline 111.4 ng/mL to 33.6
ng/mL by March 2023 (online supplementary Table).
The patient tolerated treatment well except for a grade 1
skin reaction and diarrhoea with reference to the CTCAE
(Common Terminology Criteria for Adverse Events)
version 5.0.[11]
Figure 2. Case 2. Serial positron emission tomography–computed tomography scans at baseline and following first and subsequent lines of
systemic treatment. (a) Primary lung tumour in the left lower lobe, (b) left lower rib metastasis and (c) liver metastasis at baseline (November
2022). (d) Primary lung tumour in the left lower lobe, (e) left lower rib metastasis and (f) liver metastasis after 4 months of erlotinib (March
2023). (g) Primary lung tumour in the left lower lobe, (h) left lower rib metastasis and (i) liver metastasis after cycle 1 paclitaxel/carboplatin
(May 2023). (j) Primary lung tumour in the left lower lobe, (k) left lower rib metastasis and (l) liver metastasis after 3 months of osimertinib/tepotinib (August 2023). (m) Primary lung tumour in the left lower lobe, (n) left lower rib metastasis and (o) liver metastasis after cycle 2
trastuzumab deruxtecan (October 2023).
Worsening bone pain and deranged liver function were
reported in March 2023. The EGFR TKI was switched
to osimertinib due to the impaired liver function. CEA
level rapidly increased from 33.6 ng/mL to 563 ng/mL
from March to April 2023. PET-CT showed progression
of the primary lung tumour in the left lower lobe and
mixed response in the liver and bone with new emerging
metastases (Figure 2d-f). Liquid NGS through the
LiquidHALLMARK test (Lucence, Alto [CA], US) in
March 2023 revealed EGFR L858R and TP53 S215G
mutations, while EGFR T790M was negative. Pathology
of the liver biopsy at the progressive site in April 2023
revealed non–small-cell carcinoma, and subsequent
conventional polymerase chain reaction found only
EGFR L858R mutation, without T790M mutation. Due
to the rapid progression on EGFR TKI and the absence
of T790M mutation in both liquid and tissue biopsies,
the patient was prescribed one cycle of paclitaxel (175 mg/m2 × 70% administered as an intravenous
infusion on Day 1) and carboplatin (AUC 5 × 70%
administered as an intravenous infusion on Day 1) in
early May 2023 (Figure 2g-i). Nonetheless the treatment
course was complicated by non-neutropenic fever,
malaise, and gastrointestinal side-effects despite a dose
reduction to 70% of the full dose. Therefore, tissue NGS
from the liver biopsy specimen using ACT Precis Thorax
(ACT Genomics, Taiwan) was conducted, revealing an
EGFR L858R mutation, MET amplification, and ERBB2
S310Y mutation. Osimertinib (80 mg daily) and tepotinib
(450 mg daily) were started in May 2023 at the patient’s
own expense (online supplementary Table).
The patient experienced grade 1 vomiting, transaminitis,
lower limb oedema and grade 2 diarrhoea with reference
to CTCAE version 5.0[11] during treatment, leading to a
dose reduction of tepotinib to 225 mg daily in June 2023.
CEA level initially decreased from 969 ng/mL to 607 ng/mL from May to July 2023 but rebounded to 1170 ng/mL in August 2023. PET-CT confirmed further disease
progression of lung, pleural, liver, and bone metastases
(Figure 2j-l). Given the previous suboptimal tolerance
to chemotherapy and detection of the ERBB2 S310Y
mutation by tissue NGS upon progression, T-DXd (5.4
mg/kg administered as an intravenous infusion every 3
weeks) was started in August 2023 as a self-financed
medication. CEA level decreased from 1170 ng/mL to
550 ng/mL within the initial 2 months. Nonetheless the
patient experienced recurrent non-neutropenic fever and
required repeated courses of antibiotics and naproxen
for possible sepsis and tumour fever. Serial chest X-rays
did not suggest definite pneumonitis changes. PET-CT
after two cycles of T-DXd showed disease progression
in the liver with new emerging metastasis and
superimposed infection (Figure 2m-o). In view of the
suboptimal performance status and rapidly progressing
disease, symptomatic care was initiated. Unfortunately,
the patient passed away in November 2023 (online supplementary Table).
DISCUSSION
Data indicated that patients with ERBB2 mutations
exhibit a favourable response to first-line immunotherapy
in combination with chemotherapy, with an overall
response rate up to 52% and a median progression-free
survival of 6 months.[12] Nonetheless most of these studies
focused on patients with ERBB2 exon 20 insertion (66%),
while patients with ERBB2 S310 mutations accounted
for only 13% of the studied population.[12] In Case 1 with
primary adenocarcinoma of lung harbouring de novo ERBB2 S310F mutation, the efficacy of pemetrexed,
carboplatin and pembrolizumab was maintained with
the DOR up to 7 months, consistent with the currently
available data.[12] The National Comprehensive Cancer
Network guideline recommends ADCs for patients
with ERBB2-mutant NSCLC after failure of first-line
platinum-based chemotherapy with or without
immunotherapy.[7] To date, the clinical treatment response
to ADCs specific to the S310 mutations have been rarely
reported. A phase II basket trial reported two patients
with the S310F mutation, with or without concurrent
ERBB2 amplification, who achieved a partial response
or stable disease, respectively, to T-DM1.[13] In our cases,
Case 1 failed to exhibit a clinically meaningful response
after four cycles of T-DXd. This may have been due to
genuine treatment failure although another explanation
could be tumour heterogeneity since the progress PET-CT
indicated a partial response of lymph node metastasis
but frank progression of bone metastasis (Figure 1).
It is hypothesised that the ERBB2 S310 mutation triggers
hydrophobic interactions and non-covalent dimerisation,
activating the downstream signalling pathway,[14] thereby
playing a pivotal role in oncogenesis as a driver mutation.
The oncogenic de novo ERBB2 mutations in NSCLC
are believed to be mutually exclusive with other driver
genes.[15] Nonetheless other studies and case series have
shown a minority of NSCLCs harbouring both EGFR
and ERBB2 alterations (0.46%),[5] with a significantly
higher frequency in the non-TKD S310 locus.[15] [16] It is
speculated that patients treated with EGFR TKIs have
compensated by developing ERBB2 ECD mutation
bypassing EGFR to continue on the mitogen-activated
protein kinase signalling pathway,[5] driving resistance
to EGFR TKIs with associated poor survival. In Case
2 with EGFR-mutant adenosquamous lung carcinoma
demonstrating ERBB2 S310Y and MET amplification
upon progression on first-line EGFR TKI, there were
short progression-free intervals of 3 to 4 months on
first-line EGFR TKI and the subsequent combination of
EGFR/MET inhibitors, evidence that ERBB2 S310 may
be a poor predictive and prognostic factor. Despite two
cycles of T-DXd upon progression on TKIs, the patient
continued to experience rapid disease progression with
no clinically meaningful response (online supplementary Table). This may have been due to tumour heterogeneity
given the mixed histology of adenosquamous carcinoma.
Another plausible explanation is that the ERBB2 S310
mutation may mediate resistance to EGFR TKIs, rather
than playing an oncogenic role, thereby rendering
HER2-targeting therapy alone ineffective. Moreover, the concurrent mutation of EGFR in the ERBB2-altered
NSCLC may also impair the efficacy of anti-HER2
agents.[15] Therefore, concomitant treatment of EGFR
TKIs and an HER2-targeted agent may be a promising
therapeutic strategy. Jia et al[17] reported a patient with
Li-Fraumeni syndrome and chemotherapy-refractory
metastatic lung adenocarcinoma harbouring EGFR
L858R and ERBB2 S310F mutations treated with a
dual EGFR/ERBB2 inhibitor, afatinib. A complete
response was achieved and maintained after 12 months.[17]
Nevertheless further investigations are needed to provide
the rationale in these clinical settings.[15]
CONCLUSION
These two cases demonstrate that de novo ERBB2
S310 mutations and concurrent EGFR and ERBB2
S310 mutations did not yield a clinically meaningful
response to T-DXd. The co-occurrence of ERBB2 S310
mutations with EGFR±MET amplification resulted in
short DOR following the EGFR±MET inhibitors. This
supports the association of ERBB2 S310 alterations with
unfavourable clinical outcomes in NSCLC. Given the
scarcity of studies focused on ERBB2 S310 mutations,
it is essential to continue gathering valuable clinical data
on these disease entities to enable real-world outcomes
analysis.[5]
REFERENCES
1. Mazieres J, Peters S, Lepage B, Cortot AB, Barlesi F, Beau-Faller M, et al. Lung cancer that harbors an HER2 mutation:
epidemiologic characteristics and therapeutic perspectives. J Clin
Oncol. 2013;31:1997-2003. Crossref
2. Zheng Y, Shen G, Zhang C, Huo X, Xin Y, Fang Q, et al. Efficacy
of anti-HER2 drugs in the treatment of patients with HER2-mutated
cancers: a systematic review and meta-analysis. Clin Exp Med.
2023;23:3205-16. Crossref
3. Wang J, Wen Y, Ding G, Ding P, Zhang L, Liu J, et al. Efficacy
generated by afatinib in a lung adenocarcinoma patient harboring
HER2 S310Y mutation. Cancer Biol Ther. 2018;19:450-3. Crossref
4. Jebbink M, de Langen AJ, Boelens MC, Monkhorst K, Smit EF.
The force of HER2—a druggable target in NSCLC? Cancer Treat Rev. 2020;86:101996. Crossref
5. Nagasaka M, Singh V, Baca Y, Sukari A, Kim C, Mamdani H, et al. The effects of HER2 alterations in EGFR mutant non–small cell lung cancer. Clin Lung Cancer. 2022;23:52-9. Crossref
6. Loeffler E, Ancel J, Dalstein V, Deslée G, Polette M, Nawrocki-Raby B. HER2 alterations in non–small cell lung cancer: biologico-clinical consequences and interest in therapeutic strategies. Life
(Basel). 2023;14:64. Crossref
7. National Comprehensive Cancer Network. NCCN Guidelines. Guidelines for patients: non–small cell lung cancer. Available from: https://www.nccn.org/guidelines/guidelines-detail?category=1&id=1450. Accessed 22 Nov 2024.
8. Li BT, Smit EF, Goto Y, Nakagawa K, Udagawa H, Mazières J, et al. Trastuzumab deruxtecan in HER2-mutant non–small-cell lung cancer. N Engl J Med. 2022;386:241-51. Crossref
9. Goto K, Goto Y, Kubo T, Ninomiya K, Kim SW, Planchard D, et al.
Trastuzumab deruxtecan in patients with HER2-mutant metastatic
non–small-cell lung cancer: primary results from the randomized,
phase II DESTINY-Lung02 trial. J Clin Oncol. 2023;41:4852-63. Crossref
10. Seymour L, Bogaerts J, Perrone A, Ford R, Schwartz LH, Mandrekar S, et al. iRECIST: guidelines for response criteria for use in trials testing immunotherapeutics. Lancet Oncol. 2017;18:e143-52. Crossref
11. United States Department of Health and Human Services. Common
Terminology Criteria for Adverse Events (CTCAE) Version 5.0.
2017 November 27. Available from: https://ctep.cancer.gov/protocoldevelopment/electronic_applications/docs/CTCAE_v5_Quick_Reference_5x7.pdf. Accessed 29 Nov 2024.
12. Saalfeld FC, Wenzel C, Christopoulos P, Merkelbach-Bruse S, Reissig TM, Laßmann S, et al. Efficacy of immune checkpoint
inhibitors alone or in combination with chemotherapy in NSCLC
harboring ERBB2 mutations. J Thorac Oncol. 2021;16:1952-8. Crossref
13. Li BT, Shen R, Buonocore D, Olah ZT, Ni A, Ginsberg MS, et al.
Ado-trastuzumab emtansine for patients with HER2-mutant
lung cancers: results from a phase II basket trial. J Clin Oncol.
2018;36:2532-7. Crossref
14. Greulich H, Kaplan B, Mertins P, Chen TH, Tanaka KE, Yun CH,
et al. Functional analysis of receptor tyrosine kinase mutations in
lung cancer identifies oncogenic extracellular domain mutations
of ERBB2. Proc Natl Acad Sci U S A. 2012;109:14476-81. Crossref
15. Yu X, Ji X, Su C. HER2-altered non–small cell lung cancer:
biology, clinicopathologic features, and emerging therapies. Front
Oncol. 2022;12:860313. Crossref
16. Wei XW, Gao X, Zhang XC, Yang JJ, Chen ZH, Wu YL, et al.
Mutational landscape and characteristics of ERBB2 in non–small
cell lung cancer. Thorac Cancer. 2020;11:1512-21. Crossref
17. Jia Y, Ali SM, Saad S, Chan CA, Miller VA, Halmos B. Successful
treatment of a patient with Li-Fraumeni syndrome and metastatic
lung adenocarcinoma harboring synchronous EGFR L858R and
ERBB2 extracellular domain S310F mutations with the pan-HER
inhibitor afatinib. Cancer Biol Ther. 2014;15:970-4. Crossref