Amplification of HER2 can drive the proliferation of cancer cells, and several inhibitors of HER2 have been successfully developed. Recent advances in next-generation sequencing now reveal that HER2 is subject to mutation, with over 2,000 unique variants observed in human cancers. Several examples of oncogenic HER2 mutations have been described, and these primarily occur at allosteric sites outside the ATP-binding site. To identify the full spectrum of oncogenic HER2 driver mutations aside from a few well-studied mutations, we developed mutation-allostery-pharmacology (MAP), an in silico prediction algorithm based on machine learning. By applying this computational approach to 820 single-nucleotide variants, a list of 222 known and potential driver mutations was produced. Of these 222 mutations, 111 were screened by Ba/F3-retrovirus proliferation assays; 37 HER2 mutations were experimentally determined to be driver mutations, comprising 15 previously characterized and 22 newly identified oncogenic mutations. These oncogenic mutations mostly affected allosteric sites in the extracellular domain (ECD), transmembrane domain, and kinase domain of HER2, with only a single mutation in the HER2 orthosteric ATP site. Covalent homodimerization was established as a common mechanism of activation among HER2 ECD allosteric mutations, including the most prevalent HER2 mutation, S310F. Furthermore, HER2 allosteric mutants with enhanced covalent homodimerization were characterized by altered pharmacology that reduces the activity of existing anti-HER2 agents, including the mAb trastuzumab and the tyrosine kinase inhibitor lapatinib. Overall, the MAP-scoring and functional validation analyses provided new insights into the oncogenic activity and therapeutic targeting of HER2 mutations in cancer.

Significance:

This study identified new oncogenic HER2 allosteric mutations, including ECD mutations that share covalent dimerization as a mechanism of oncogenicity, suggesting the need for novel inhibitors to treat HER2-mutant cancers.

HER2 belongs to the ERBB family of receptor tyrosine kinases (RTK) that undergo dimerization to trigger downstream signaling pathways involved in cell growth, proliferation, differentiation, and survival (1–4). HER2, like the other ERBB family members, contains an extracellular domain (ECD), a transmembrane domain (TMD), and an intracellular region consisting of a juxtamembrane domain (JMD), a kinase domain (KD), and a C-terminal tail domain (CTD). The ECD is comprised of four subdomains (I–IV): two leucine-rich subdomains, L1 (I) and L2 (III), that typically participate in ligand binding (except for HER2), and two cysteine-rich subdomains, CR1 (II) and CR2 (IV), that contain multiple intramolecular disulfide bonds that line the ECD dimer interfaces (1, 5, 6). For ligand-binding ERBB family members, in absence of ligand, the ECD adopts an autoinhibited tethered (closed) conformation, whereas upon ligand binding, the ECD adopts an untethered (open) conformation, leading to homodimerization or heterodimerization (1, 3). Unlike other members of the ERBB family, HER2 ECD constitutively adopts an untethered, active conformation independently of ligand interactions (4). HER2 is the preferred partner to form heterodimer with other ligand-binding ERBB family members, although HER2 homodimers have been reported in HER2-overexpressing cells (1–3). The formation of ERBB dimers is mediated by the CR1 and CR2 of the ECD, as well as the rearrangement of the TMD and JMD to support the asymmetric dimerization of the KD (7).

Overexpression and amplifications of wild-type (WT) HER2 are known to play an oncogenic role in a subset (∼20%) of breast and gastric/gastroesophageal junction cancers (8). These findings led to the approval of anti-HER2 therapies for the treatment of HER2+ breast cancer, including mAbs (trastuzumab, pertuzumab, and margetuximab), antibody–drug conjugate (ADC; ado-trastuzumab emtansine and trastuzumab deruxtecan), and small molecules (lapatinib, neratinib, and tucatinib; refs. 9–16). Trastuzumab and ADC trastuzumab deruxtecan are currently approved for the treatment of HER2+ gastric or gastroesophageal junction adenocarcinoma (12, 15). More recently, oncogenic mutations in the HER2 gene have been identified in patients with breast, lung, colorectal, gastric/gastroesophageal, bladder cancers, and other cancers (8). Mutations in HER2, including single-nucleotide variants (SNV; point mutations), insertions, deletions, and fusions, are rare events and most commonly occur at allosteric sites including the ECD, the TMD, the JMD, and the KD. According to a comprehensive genomic profiling using next-generation sequencing conducted on a collection of 272,548 unique patient solid tumor samples (the Foundation Medicine, Inc. data: Apr 28, 2020—MI20200428 FMI), the most common HER2 mutations include S310F/Y (0.5%, in the ECD), the exon 20 insertion A775-G776insYVMA (0.2%, in the KD), R678Q (0.2%, in the JMD), L755S (0.2%, in the KD), and V842I (0.1%, in the KD).

As HER2 is subject to over 2,000 unique variants, we sought to define the full spectrum of oncogenic HER2 mutations expressed by solid tumors. Here we describe the screening process that led to the determination of a panel of 37 allosteric HER2 driver mutations, comprising 15 previously characterized and 22 newly identified oncogenic mutations, and further analyses aimed at elucidating the mechanism of oncogenicity associated with these mutations, as well as at understanding the pharmacology of available anti-HER2 agents in these mutants.

Cell culture

Ba/F3 cells were purchased from DSMZ (Braunschweig, Germany) and maintained in RPMI1640 medium (Life Technologies) supplemented with 10% FBS, 1% l-glutamine, and 10 ng/mL mouse IL3 (R&D Systems). Lack of endogenous murine erbB family member protein expression in mammalian hematopoietic cells (17) was confirmed in parental Ba/F3 cells (Supplementary Fig. S1). Ba/F3 transformed cells were maintained in the medium with 15-µg/mL blasticidin S HCl (Life Technologies). U87MG cells were purchased from ATCC and maintained in MEM (Life Technologies) supplemented with 10% FBS and 1% l-glutamine. The cell lines were periodically checked for Mycoplasma using MycoAlert Plus Mycoplasma Detection Kit (Lonza).

Retroviral production

Human HER2 cDNA (RC212583; Origene) was subcloned into pMXs-IRES-Blasticidin (Cell Biolabs). HER2 mutants were generated by site-directed mutagenesis. Retroviral expression vector retrovirus was produced by transient transfection of HEK 293T cells with the retroviral ERBB expression vector pMXs-IRES-Blasticidin, pCMV-Gag-Pol vector, and pCMV-VSV-G-Envelope vector (Cell Biolabs). Briefly, HEK 293T cells were plated (2.5 × 105 per plate) in 6-well plates (Corning) and incubated overnight. The next day, retroviral plasmids (1 µg of ERBB, 0.32 µg of pCMV-Gag-Pol, and 0.16 µg pCMV-VSV-G) were mixed in 150 µL of Opti-MEM (Life Technologies). The mixture was incubated at room temperature for 5 minutes and then added to Opti-MEM containing transfection reagent Lipofectamine (Invitrogen) and incubated for 20 minutes. Mixture was then added dropwise to HEK 293T cells. The next day, the medium was replaced with fresh culture medium, and retrovirus was harvested at 24 hours.

Generation of ERBB2-mutant stable cell lines

Ba/F3 cells (4.0 × 104 cells) were infected with 20 µL of viral supernatant supplemented with 8-µg/mL polybrene by centrifuging for 30 minutes at 1,000 rpm. Cells were placed in a 37°C incubator overnight. Then, 100 µL cells and supernatant were added to 900 µL of RPMI1640 medium (Life Technologies) supplemented with 10% FBS, 1% l-glutamine, and 15 µg/mL blasticidin S HCl (Life Technologies). Cells were placed in a 37°C incubator and monitored for outgrowth over a 2-week period.

U87MG cells (1.0 × 106 cells) were infected with 2 mL of viral supernatant supplemented with 8-µg/mL polybrene in T150 flasks. Cells were placed in a 37°C incubator overnight. After 24 hours, cells were given a media change and were placed in a 37°C incubator overnight. Cells were then selected in presence of 25 µg/mL blasticidin S HCl (Life Technologies) and monitored for outgrowth.

Cell proliferation

Ba/F3 cell lines were resuspended at 1.1 × 105 cells/mL in RPMI containing 10% heat inactivated FBS and 1% l-glutamine and dispensed in triplicate (1.5 × 104 cells/well) into 96-well plates. To determine the effect of drug on cell proliferation, cells were incubated for 3 days in the presence of vehicle control or test drug at varying concentrations. Inhibition of cell growth was determined by luminescent quantification of intracellular ATP content using CellTiter-Glo (Promega). Dose–response curves were plotted by comparing cell numbers on day 0 versus 72 hours posttreatment. The number of viable cells was determined and normalized to vehicle-treated controls. Inhibition of proliferation, relative to vehicle-treated controls was expressed as a fraction of 1 and graphed using GraphPad Prism 9 software (GraphPad Software). EC50 values were determined with the same application.

Cellular protein analysis

Cell extracts were prepared by detergent lysis RIPA (Sigma) containing 10-mmol/L iodoacetamide (G-Biosciences), protease inhibitor (Sigma), and phosphatase inhibitors (Sigma) cocktails. The soluble protein concentration was determined by micro-BCA assay (Pierce Biotechnology). Protein immunodetection was performed by electrophoretic transfer of SDS-PAGE separated proteins to nitrocellulose, incubation with antibody, and chemiluminescent second step detection. Nitrocellulose membranes were blocked with 5% nonfat dry milk in TBS and incubated overnight with primary antibody in 5% BSA. The following primary antibody from Cell Signaling Technology were used at 1:1,000 dilution: phospho-HER2 and total HER2. β-Actin antibody, used as a control for protein loading, was purchased from Sigma. Horseradish peroxidase–conjugated secondary antibodies (Cell Signaling Technology) were incubated in nonfat dry milk at 1:5,000 dilution for 1 hour. SuperSignal chemiluminescent reagent (Pierce Biotechnology) was used according to the manufacturer's directions, and blots were imaged using the Alpha Innotech image analyzer and AlphaEaseFC software (Alpha Innotech).

Development of mutation-allostery-pharmacology oncogenicity prediction through a machine learning algorithm

The mutation-allostery-pharmacology (MAP) algorithm utilizes a well-curated set of genomic and proteomic features to assess the potential oncogenicity of SNVs, based on a prediction model trained on an internally generated validated mutation dataset from a group of known oncogenes. This prediction algorithm determines the potential impact of mutations causing deleterious effects on the structure and function of proteins, taking into account both gain-of-function and loss-of-function mutations. The reported MAP score, a binary classifier, ranges between 0 and 1, and is a proxy for probability of being oncogenic. This approach results in a relatively high number of false-positive predictions on driver mutations, as the MAP score oncogenicity threshold of 0.3 was chosen for higher sensitivity, sacrificing accuracy.

Molecular dynamics simulations

The atomistic model of the HER2 ECD was prepared on the basis of the HER2 crystal structure (PDB ID: 6J71) using Prime from the Schrödinger suite (18). The S310F mutant was built based on the WT model with a single substitution of residue Ser 310 to Phe. In an effort to enhance any structural changes associated with the HER2 residue 310, simulations of both WT and S310F models were conducted with and without a disulfide bond between Cys299 and Cys311 (referred to as oxidized and reduced states, respectively). We generated four distinct models with visual molecular dynamics (VMD; ref. 19): HER2 WT-oxidized, HER2 WT-reduced, HER2 S310F-oxidized, and HER2 S310F-reduced molecular dynamics (MD) simulations were performed with the NAMD 2.13 software package (20). The CHARMM36 parameter set was used for the protein (21, 22) and the TIP3P model was used for the water molecules (23). A cut-off distance of 10 Å was used for the van der Waals and short-range electrostatic interactions. The long-range electrostatic interactions were computed with the particle-mesh Ewald summation method (24). The SHAKE algorithm was applied to all hydrogen-containing bonds (25). With all other atoms fixed, the waters were energy minimized for 20,000 steps using the conjugate gradient algorithm and subsequently all systems were minimized for 100,000 steps with the conjugate gradient algorithm. After heating the system gently with the canonical ensemble NVT (constant number, volume, temperature) run for 200 picoseconds (ps) at 300 K, the systems were further minimized (100,000 steps), followed by equilibration first in the NVT and then in the isothermal-isobaric ensemble NPT for 200 ps each. Finally, unbiased MD simulations were carried out, with the atomic coordinates of the systems saved every 10 ps. For each of four simulations (HER2 WT-oxidized, HER2 WT-reduced, HER2 S310F-oxidized, and HER2 S310F-reduced), three independent MD simulations (three replicas) were run. All structures and trajectories analyses were performed with VMD (19).

Perturbation response scanning

Perturbation response scanning (PRS) analysis was utilized to map the dynamic effect of allosteric mutations on the structure of HER2 (26, 27). PRS simulates a perturbation at a residue and computes the propagation of the response across the protein structure. The procedure is repeated for all residues, resulting in a response matrix P. The averages over the rows and columns of the normalized matrix P reveal the network of sensors and effectors, and allows to visualize the potential dynamic effect of an allosteric mutation. PRS was computed using ProDy on the HER2 crystal structure (PDB ID: 6J71; ref. 28).

Data availability

The project GENIE (Genomics Evidence Neoplasia Information Exchange) data analyzed in this study are available from the Synapse platform (http://www.synapse.org/genie). Restrictions apply to the availability of the FMI cancer-associated HER2 mutation data, which are available from the authors upon reasonable request with the permission of the Foundation Medicine, Inc.

Identification of HER2 oncogenic mutations distributed throughout multiple domains

Comprehensive collection of cancer-associated mutations has revealed that HER2 contains 820 and 2616 unique SNVs based on the project GENIE data and the Foundation Medicine, Inc. data, respectively. These SNVs are distributed throughout the entire amino acid sequence of HER2, encompassing the ECD, the TMD, the JMD, the KD, and the CTD (Fig. 1A). The 2,616 SNVs detected in the Foundation Medicine dataset consist of 2,132 missense mutations, 242 nonsense mutations, 128 indel mutations, and 114 splice site mutations. Of these 2,616 SNVs, a small subset corresponded to previously described driver mutations (Supplementary Table S1; ref. 29). Mutation association data analysis revealed that the vast majority of HER2 driver mutations are observed independently of gene amplification. Among 10,874 instances of HER2-mutant tumors within the Foundation Medicine dataset, only 10% (1,094 cases) presented with copy-number amplification. The majority of HER2 SNVs (2,586/2,616) consists of rarely observed mutations (<0.01%), with only 30 SNVs ranging from intermediate (0.01%–0.1%) prevalence to high (>0.1%) prevalence (Fig. 1B). As there is growing recognition that even rarely occurring mutations can promote potent oncogenic activity (30, 31), we sought to identify the full spectrum of rare and recurrent oncogenic HER2 mutations.

Figure 1.

HER2 mutation prevalence and protein domain architecture. A, Prevalence of somatic HER2 mutations observed throughout the protein domains. Foundation Medicine, Inc. data used in this study contain 2,616 unique SNV HER2 mutations. HER2 domains: The protein architecture consists of L domains 1 and 2 (L1, L2), cysteine-rich domains 1 and 2 (CR1, CR2), TMD, JMD, KD, and CTD. The L domains and the CR domains collectively form the ECD. Prediction: Oncogenicity of cancer-associated mutations are predicted as either oncogenic mutations (dark blue) or neutral mutations (gray). Validation: Select HER2 SNVs are biologically validated as driver mutations (magenta) or neutral (green) mutations, based on Ba/F3 cell proliferation assay results. Refined collection of verified HER2 driver mutations with a varying degree of prevalence is shown on the bottom plot. The most prevalent mutation, S310F, is located in the ECD (CR1), whereas an intracellular mutational hotspot, containing L755S, Exon20ins, V777L, and V842L, is located in the KD. Other notable mutations are labeled. B,HER2 SNVs and oncogenic mutations by prevalence range. Quantification analysis of somatic HER2 mutations in three prevalence groups.

Figure 1.

HER2 mutation prevalence and protein domain architecture. A, Prevalence of somatic HER2 mutations observed throughout the protein domains. Foundation Medicine, Inc. data used in this study contain 2,616 unique SNV HER2 mutations. HER2 domains: The protein architecture consists of L domains 1 and 2 (L1, L2), cysteine-rich domains 1 and 2 (CR1, CR2), TMD, JMD, KD, and CTD. The L domains and the CR domains collectively form the ECD. Prediction: Oncogenicity of cancer-associated mutations are predicted as either oncogenic mutations (dark blue) or neutral mutations (gray). Validation: Select HER2 SNVs are biologically validated as driver mutations (magenta) or neutral (green) mutations, based on Ba/F3 cell proliferation assay results. Refined collection of verified HER2 driver mutations with a varying degree of prevalence is shown on the bottom plot. The most prevalent mutation, S310F, is located in the ECD (CR1), whereas an intracellular mutational hotspot, containing L755S, Exon20ins, V777L, and V842L, is located in the KD. Other notable mutations are labeled. B,HER2 SNVs and oncogenic mutations by prevalence range. Quantification analysis of somatic HER2 mutations in three prevalence groups.

Close modal

To facilitate this discovery of oncogenic mutations beyond the previously identified HER2 driver mutations using a scalable approach, we developed a computational prediction algorithm, called MAP, based on numerous genomic and proteomic features deemed critical for oncogenicity prediction. This initial computational approach was applied to the fully annotated set of 820 mutations within the project GENIE dataset. The in silico MAP analysis produced a list of 222 mutations with predicted oncogenicity, consisting of 20 already known driver mutations and 202 potentially new driver mutations (Fig. 1A). Next, we evaluated the oncogenicity of HER2 mutations in the context of cell proliferation, as the expression of HER2 oncogenic mutants, but not the HER2 WT, has been shown to transform mouse hematopoietic Ba/F3 cells to IL3 independence. Lack of endogenous murine erbB family member protein expression in Ba/F3 cells (Supplementary Fig. S1; ref. 17) provided a suitable model for assessing transformation by overexpressing an oncogene, albeit precluding HER2/erbB heterodimerization, and assessing the ability for the cells to proliferate in the absence of IL3. We selected half of the predicted 222 mutations for experimental validation in the Ba/F3-retrovirus proliferation assay, including mutations located in various domains (48 in the ECD/TMD region and 63 in the JMD/KD/CTD region) and mutations ranging from rare to high prevalence, and determined 37 mutations to be mitogenic/oncogenic and 74 mutations to be neutral (Fig. 1A; Supplementary Table S2). Additional tumor sample data, including ethnicity, age, sex, race, cancer type, and a list of genes with at least one mutation, reported along with these HER2 mutations, are provided in Supplementary Table S3. The analysis of variant allelic fraction (VAF), representing the ratio of each HER2 SNV to WT of these experimentally validated mutations shows that VAF values from different tumor samples vary greatly and do not appear to correspond with the oncogenicity of HER2 mutations (Supplementary Fig. S2). This extensive collection of annotated HER2 mutations includes 15 previously described and 22 newly identified driver mutations. Seven oncogenic driver mutations are expressed with high prevalence (≥0.1%), nine driver mutations with intermediate prevalence (0.01%–0.1%), and 21 driver mutations are considered as rare/private mutations (<0.01%; Fig. 1B). These oncogenic/driver mutations are distributed throughout the entire amino acid sequence of HER2, nearly all occurring outside of the ATP-binding site. They are often found aggregated together within several functionally critical regions of HER2, forming allosteric hotspots or mutational clusters defined by their impact on protein structure and dynamics. Numerous mutations found in the N-terminal half of HER2 form a cluster in the ECD (Fig. 1A). This cluster is represented by the most prevalent HER2 mutation S310F that is allosterically located in the extracellular CR1 domain involved in dimer activation mechanisms (1, 7, 32). A cluster of prevalent intracellular KD mutations, L755S, exon 20 insertions (A775-G776insYVMA), D769Y, V777L, V842I, T862A, and L869R, are found proximal or within several key structural motifs, such the α-C helix, DFG motif, and A-loop (33, 34). Another prevalent mutation, R678Q, is located in the JMD and it is involved in the rearrangement of TMD/JMD dimerization upon activation (1). We confirmed the oncogenicity of these nine mutations in the context of Ba/F3 cell proliferation assays (Fig. 1A), and these findings are consistent with previous findings (1, 34–42). In addition, our validation approach determined the oncogenic driver status of a TMD mutation G660D (1) with medium prevalence (Fig. 1A) and the extracellular G292R/C mutations (39) with low prevalence in CR1 domain (Fig. 1A; Table 1). These findings support the previous assessment of these mutations as likely oncogenic (1, 39, 43), and the presence of allosteric hotspots formed by low-prevalence oncogenic mutations clustering together with highly prevalent driver mutations in discrete domains.

Table 1.

List of oncogenic HER2 mutations in the ECD and TMD/JMD with presumed mechanisms of activation, after validation in Ba/F3 cell proliferation assay.

HER2 mutationDomain involvedActivation mechanism (free cysteine)Reference
G58R ECD (L1) NA This study 
R103Q ECD (L1) Covalent ECD dimerization (NA) This study 
P122 L ECD (L1) Covalent ECD dimerization (NA) This study 
V219I ECD (CR1) Covalent ECD dimerization (NA) This study 
G229R ECD (CR1) NA This study 
MDK-HER2 fusion (271–1225)a ECD (L1/CR1) NA (C295) (40
G292C ECD (CR1) Covalent ECD dimerization (G292C) This study 
G292R ECD (CR1) Covalent ECD dimerization (NA) This study 
A293T ECD (CR1) Covalent ECD dimerization (NA) This study 
G309Aa ECD (CR1) NA (35, 36
S310F ECD (CR1) Covalent ECD dimerization (C299, C311) This study (36
S310Y ECD (CR1) NA This study (36
C311Ra ECD (CR1) Covalent ECD dimerization (C299) (36
E321Ga ECD (CR1) NA (36
C334Sa ECD (CR1) NA (36
R434Q ECD (L2) Covalent ECD dimerization (NA) This study 
A516T ECD (CR2) NA This study 
A588V ECD (CR2) Covalent ECD dimerization (NA) This study 
G603C ECD (CR2) Covalent ECD dimerization (NA) This study 
HER2 p95-M611b ECD (L1/CR1/L2/CR2) Covalent ECD dimerization (C623) This study 
HER2 Δ16 (634–649del)b ECD (CR2) Covalent ECD dimerization (C626/C630) This study 
ZNF207-HER2 fusion (666–1225)a ECD/TMD NA (NA) (40
S653Ca TMD Covalent ECD dimerization (S653C) (41, 42
G660D TMD TMD dimerization This study (1
R678Q JMD TMD/JMD dimerization This study (1, 35, 41
HER2 mutationDomain involvedActivation mechanism (free cysteine)Reference
G58R ECD (L1) NA This study 
R103Q ECD (L1) Covalent ECD dimerization (NA) This study 
P122 L ECD (L1) Covalent ECD dimerization (NA) This study 
V219I ECD (CR1) Covalent ECD dimerization (NA) This study 
G229R ECD (CR1) NA This study 
MDK-HER2 fusion (271–1225)a ECD (L1/CR1) NA (C295) (40
G292C ECD (CR1) Covalent ECD dimerization (G292C) This study 
G292R ECD (CR1) Covalent ECD dimerization (NA) This study 
A293T ECD (CR1) Covalent ECD dimerization (NA) This study 
G309Aa ECD (CR1) NA (35, 36
S310F ECD (CR1) Covalent ECD dimerization (C299, C311) This study (36
S310Y ECD (CR1) NA This study (36
C311Ra ECD (CR1) Covalent ECD dimerization (C299) (36
E321Ga ECD (CR1) NA (36
C334Sa ECD (CR1) NA (36
R434Q ECD (L2) Covalent ECD dimerization (NA) This study 
A516T ECD (CR2) NA This study 
A588V ECD (CR2) Covalent ECD dimerization (NA) This study 
G603C ECD (CR2) Covalent ECD dimerization (NA) This study 
HER2 p95-M611b ECD (L1/CR1/L2/CR2) Covalent ECD dimerization (C623) This study 
HER2 Δ16 (634–649del)b ECD (CR2) Covalent ECD dimerization (C626/C630) This study 
ZNF207-HER2 fusion (666–1225)a ECD/TMD NA (NA) (40
S653Ca TMD Covalent ECD dimerization (S653C) (41, 42
G660D TMD TMD dimerization This study (1
R678Q JMD TMD/JMD dimerization This study (1, 35, 41

Abbreviations: CR, cysteine-rich; ECD, extracellular domain; JMD, juxtamembrane domain; NA, not available; TMD, transmembrane domain.

aThese mutations are previously known oncogenic mutations, not part of the 111 mutations tested with the Ba/F3 biological validation method.

bThese mutations are not part of the 111 mutations tested with the Ba/F3 biological validation method but are included here because we validated their mechanistic oncogenicity.

Tumor-specific analysis of validated oncogenic HER2 mutations reveals a unique tumor tropic distribution of HER2 oncogenic mutations among different cancer types. For example, a cluster of KD mutations at residues L755, V777, and V842 constitutes the predominant mutations in breast and colorectal cancers, while observed at much lower prevalence in lung cancers (Fig. 2). Similarly, the exon 20 insertions and R678Q mutations are the prevalent HER2 mutations in lung and colorectal cancers, respectively, while representing minor fractions in other cancer types (Fig. 2). In contrast, S310F/Y mutations are most prevalent in bladder cancer and consistently represent a sizable fraction (∼10%–15%) in other cancer types, reflecting their agnostic oncogenicity (Fig. 2; ref. 44). Considering that rare driver mutations collectively make up a considerable fraction (10%–19%) of these tumors (Fig. 2; Supplementary Fig. S3), the identification of new driver mutations by employing the MAP technology has provided new opportunities to cluster together rare and prevalent oncogenic mutations to further elaborate on mutational hotspots irrespective of prevalence and tumor type.

Figure 2.

Distribution of HER2 mutations in solid tumors. Compositions of representative HER2 mutations, exon 20 insertions, S310F/Y, R678Q, and the KD hotspot mutations (L755/L777/V842) differ among all solid tumors, breast cancer, lung cancer, bladder cancer, and colorectal cancer.

Figure 2.

Distribution of HER2 mutations in solid tumors. Compositions of representative HER2 mutations, exon 20 insertions, S310F/Y, R678Q, and the KD hotspot mutations (L755/L777/V842) differ among all solid tumors, breast cancer, lung cancer, bladder cancer, and colorectal cancer.

Close modal

Molecular attributes of HER2 mutations in different domains

We mapped 37 driver and 74 neutral mutations on a full-length HER2 dimer model to examine the molecular attributes of oncogenicity (Fig. 3A; Table 1). Among the 111 mutations tested, 16 of 48 ECD/TMD mutations and 21 of 63 JMD/KD/CTD mutations were determined to be driver mutations. Comparison of proteomic features of driver and neutral mutations revealed several key attributes associated with oncogenic mutations in the ECD and KD (Fig. 3B). As expected, driver mutations have higher tendency to occur where highly conserved and functionally important amino acid residues reside. Structurally, driver mutations are associated with relatively stable motifs (medium dynamics), yet more likely to be associated with conformational changes than neutral mutations. Furthermore, the ECD driver mutations, such as S310F, are uniquely found near intramolecular disulfide bonds, whereas the KD driver mutations, such as L755S, are in close proximity to the ATP-binding site and the asymmetrical KD dimer interface (Fig. 3B). Several less frequently occurring driver mutations, such as mutations at residues G292, G660, and L869, are spatially proximal to prominent driver mutations to form the ECD cluster around S310 (Fig. 3C), the TMD/JMD cluster including R678 (Fig. 3D) and the KD cluster with L755, V777, and V842 (Fig. 3E).

Figure 3.

Protein structure analysis of HER2 oncogenic mutations. A,HER2 mutations mapped on a full-length dimer structure model as driver mutations (magenta) or neutral (green) mutations. Individual protein domains are indicated by different colors. B, Radar charts of HER2 mutations on key proteomic features. In general, driver mutations are associated with well-conserved residues that are likely involved in conformational changes. The ECD driver mutations are in close proximity to the disulfide bonds, whereas the KD driver mutations are located near the ligand binding and protein interaction sites. CE, Close-up views of prevalent HER2 mutations: S310F in CR1 (C), R678Q in JMD (D), and V842I in KD (E), and other nearby driver mutations. F, Western blots of Ba/F3-HER2 oncogenic mutants inducing covalent homodimerization in the absence and presence of reducing agents (indicated by − and +). Phospho-HER2 blots and total HER2 blots are shown.

Figure 3.

Protein structure analysis of HER2 oncogenic mutations. A,HER2 mutations mapped on a full-length dimer structure model as driver mutations (magenta) or neutral (green) mutations. Individual protein domains are indicated by different colors. B, Radar charts of HER2 mutations on key proteomic features. In general, driver mutations are associated with well-conserved residues that are likely involved in conformational changes. The ECD driver mutations are in close proximity to the disulfide bonds, whereas the KD driver mutations are located near the ligand binding and protein interaction sites. CE, Close-up views of prevalent HER2 mutations: S310F in CR1 (C), R678Q in JMD (D), and V842I in KD (E), and other nearby driver mutations. F, Western blots of Ba/F3-HER2 oncogenic mutants inducing covalent homodimerization in the absence and presence of reducing agents (indicated by − and +). Phospho-HER2 blots and total HER2 blots are shown.

Close modal

Many HER2 oncogenic mutations affect the extracellular region, and are situated near sites involved in dimerization. Covalent dimerization has been one mechanism described to promote oncogenicity among the ERBB family of RTKs, including oncogenic EGFR ECD mutations expressed in glioblastoma (45) and a select group of HER2 ECD mutations (36). We sought to determine whether this might be a general mechanism for oncogenicity for HER2 ECD mutations. Among 14 ECD driver mutations evaluated, mutations at nine positions result in covalent homodimerization, as detected by resolving proteins under nonreducing conditions (Fig. 3F). This includes the S310F mutation, the most commonly occurring HER2 mutation found in human cancers (Fig. 1; ref. 8), and is the first time this mechanism has been ascribed to this mutation. By analyzing monomer-dimer fractions of phosphorylated HER2, we found that while the covalent dimer constitutes a minor fraction of total HER2 receptors, nearly all phosphorylated receptors, indicating the active form of the receptor, were present in the covalent dimer fraction. Previous analyses that focused on monomer-dimer fractions of total HER2, and not phosphorylated HER2, may have hindered the detection of covalent dimer as the active form of the mutant. Indeed, Greulich and colleagues (36) failed to measure the active form of the commonly occurring HER2 S310F mutation. While covalent dimerization of HER2 has been previously proposed to facilitate oncogenic activation of ERBB proteins (36, 45, 46), the frequent occurrence of ECD driver mutations proximal to disulfide bonds suggests covalent homodimerization as a common mechanism of activation among HER2 ECD mutations. Interestingly, earlier studies by Siegel and colleagues conducted in mouse models expressing high levels of HER2 WT described latent development of mouse mammary tumors being not coincident with the expression of high levels of HER2 (47), but with the subsequent acquisition of HER2 mutations, such as short truncations occurring in the ECD CR2 that produced covalent dimers (46, 47).

Select HER2 mutations drive oncogenicity through reinforcement of dimerization

HER2 S310F/Y mutations induce covalent homodimerization of activated proteins through intermolecular disulfide bonds. This mutation site is directly adjacent to a disulfide bond formed between C299 and C311 within CR1 (Fig. 4A), and is part of a critical interface that engages with the dimer arm of adjacent HER2/ERBB molecule (Fig. 3C; ref. 48). To gain insights into the potential structural changes associated with the substitution of Ser310 with Phe or Tyr, we generated a minimized homology model of HER2 S310F (PDB ID: 6J71) mutant in solvent (Supplementary Fig S4). The model suggests that this amino acid position can accommodate a considerably larger amino acid side chain of either Phe or Tyr without any obvious steric effects on nearby residues. Next, we explored the mutational impact of S310F on the stability of local structural motifs, including a β-hairpin motif formed by residues 299–312 and the C299/C311 disulfide bond. We performed MD equilibrium simulations of the HER2 ECD in the presence and absence of S310F mutation (Fig. 4B). Our initial simulations performed for 100 nanoseconds (ns) with an intact C299–C311 disulfide bond show stable CR1 structures for both HER2 WT and HER2 S310F (Fig. 4B). Minimal deviations of simulated oxidized structures from the starting models are consistent with the stable CR1 structure supported by numerous disulfide bonds and the limitation of simulations time. Subsequent simulations with C299 and C311 as free cysteines, revealed similarly stable WT structure, whereas the associated β-hairpin motif of the S310F mutant became markedly unstable (Fig. 4B), indicated by increased root mean square fluctuations and the decreased stability of secondary structures (Supplementary Fig. S5). These observations suggest that the S310F mutation likely destabilizes the local β-hairpin motif and interferes with the disulfide bond formation between C311 and C299, resulting in unpaired Cys residues at the ECD dimer interface.

Figure 4.

HER2 variants drive oncogenicity through reinforcement of dimerization. A, Altered domain structures of HER2 variants, S310F, p95-M611, and Δ16. The presumed free cysteines associated with these mutations are indicated by yellow lollipops. Trastuzumab binding site (HER2 WT residues 579–625) is altered in HER2 p95-M611and HER2 Δ16. B, The S310F missense mutation disrupts the β-hairpin motif (orange) and the adjacent disulfide bond formed by C299 and C311. Structural comparison of HER2 WT (S310) and HER2 S310F mutant in oxidizing and in reducing conditions. Structural disorder induced by S310F likely results in increased distance between C311 and C299 to interfere with the disulfide bond formation. C, U87MG cells expressing HER2 S310F result in the marked formation of activated covalent dimers. Reduced dimer formation is observed with the introduction of Cys-to-Ser substitutions (C299S and C311S). Residual covalent dimer formation suggests the involvement of other cysteines in intermolecular disulfide bonds. D, U87MG cells individually expressing HER2 variants (S310F, Δ16, and p95-M611) form activated homodimers under nonreducing conditions. E, Trastuzumab effectively reduces the active dimer population of HER2 WT expressed in U87MG cells, but does not reduce the amount of covalently dimerized HER2 variants (S310F, Δ16, and p95-M611) expressed in U87MG cells. Cells were treated with control (C; PBS) or decreasing concentrations of trastuzumab (1.0, 0.5, 0.25, 0.125 µg/mL) for 24 hours. F, Proliferation of BT474 cells expressing HER2 WT is suppressed with 1 µg/mL trastuzumab, whereas Ba/F3 cells expressing HER2 variants (p95-M611 or Δ16) required higher trastuzumab concentration for growth reduction.

Figure 4.

HER2 variants drive oncogenicity through reinforcement of dimerization. A, Altered domain structures of HER2 variants, S310F, p95-M611, and Δ16. The presumed free cysteines associated with these mutations are indicated by yellow lollipops. Trastuzumab binding site (HER2 WT residues 579–625) is altered in HER2 p95-M611and HER2 Δ16. B, The S310F missense mutation disrupts the β-hairpin motif (orange) and the adjacent disulfide bond formed by C299 and C311. Structural comparison of HER2 WT (S310) and HER2 S310F mutant in oxidizing and in reducing conditions. Structural disorder induced by S310F likely results in increased distance between C311 and C299 to interfere with the disulfide bond formation. C, U87MG cells expressing HER2 S310F result in the marked formation of activated covalent dimers. Reduced dimer formation is observed with the introduction of Cys-to-Ser substitutions (C299S and C311S). Residual covalent dimer formation suggests the involvement of other cysteines in intermolecular disulfide bonds. D, U87MG cells individually expressing HER2 variants (S310F, Δ16, and p95-M611) form activated homodimers under nonreducing conditions. E, Trastuzumab effectively reduces the active dimer population of HER2 WT expressed in U87MG cells, but does not reduce the amount of covalently dimerized HER2 variants (S310F, Δ16, and p95-M611) expressed in U87MG cells. Cells were treated with control (C; PBS) or decreasing concentrations of trastuzumab (1.0, 0.5, 0.25, 0.125 µg/mL) for 24 hours. F, Proliferation of BT474 cells expressing HER2 WT is suppressed with 1 µg/mL trastuzumab, whereas Ba/F3 cells expressing HER2 variants (p95-M611 or Δ16) required higher trastuzumab concentration for growth reduction.

Close modal

We confirmed the direct contributions of C299 and C311 in covalent homodimerization of HER2 S310F by showing that Ser substitutions of these Cys results in reduced dimer formation (Fig. 4C). The residual dimer formation of the HER2 S310F/C311S/C299S mutant under nonreducing conditions suggests the possibility of S310F generating other unpaired Cys residues involved in nearby disulfide bonds to form intermolecular disulfide bonds. To gain insight into potential long-range effects of S310F and other HER2 ECD mutations, we performed PRS analysis of the HER2 ECD (27). Simulated effects of perturbation-induced structural alterations of the HER2 ECD suggest that the S310 site and many residues near a putative ligand-binding site are effective transducers of long-range perturbation affecting the CR2 subdomain, which contains numerous disulfide bonds and forms the secondary ECD dimer interface (Supplementary Fig. S6). It also remains conceivable for the HER2 S310F mutation to facilitate HER2 activation through enhanced noncovalent dimerization, as previously proposed (36). Hence, our observations can be reconciled with previous studies by proposing that the HER2 S310F/Y mutation induces the covalent homodimerization of HER2 through the intermolecular disulfide bond formation facilitated by an intermediary noncovalent interaction.

To establish the general applicability of covalent homodimerization mechanism of HER2, we characterized two other oncogenic HER2 variants, p95-M611 and Δ16, with free cysteines present in the CR2 domain (Fig. 4A). We used U87MG human glioma cells to express HER2 (Fig. 4D; Supplementary Fig. S7). HER2 p95-M611 is generated through aberrant initiation of translation, producing an N-terminally truncated oncogenic form missing a significant portion of the ECD. HER2 Δ16 is generated by aberrant splicing, producing an oncogenic form that excludes exon 16 in the CR2 domain. As the CR2 domain directly participates in the ECD dimer interface, the majority of p95-M611 and Δ16 HER2 variants resulted in activated covalent dimers, involving C623 of HER2 p95-M611 and C626/C630 of HER2 Δ16 in the intermolecular disulfide bond formation (Fig. 4D).

We sought to determine the effect of HER2 oncogenic mutations that promote covalent homodimerization on the activity for the antibody trastuzumab. The trastuzumab binding site (HER2 residues 579–625) is located in the ECD CR2 (Fig. 4A). While trastuzumab is effective at reducing the amount of total HER2 WT and consequently interfering with the formation of phosphorylated dimers, it does not effectively inhibit HER2 dimer phosphorylation for each of the three HER2 mutants examined (HER2 S310F, HER2 p95-M611, and HER2 Δ16; Fig. 4E). Similarly, we observed trastuzumab markedly affected proliferation of BT474 cells expressing HER2 WT (64% inhibition at 1 µg/mL), but not of Ba/F3 cells individually expressing HER2 p95-M611 and HER2 Δ16 (p95-M611: 0% inhibition, Δ16: 11% inhibition; Fig. 4F). These results demonstrate how trastuzumab activity is lost either by alterations that directly affect the trastuzumab-binding site (HER2 p95-M611) or by alterations through allostery (HER2 Δ16). We therefore postulate that it should not be possible to optimally reposition trastuzumab to treat tumors expressing these HER2 oncogenic variants. Indeed, clinical data have shown reduced efficacy for trastuzumab in breast tumors exhibiting high HER2 p95-M611 expression (49–51) or in other tumor types such as endometrioid cancer where HER2 p95-M611 expression is also high (52).

Tyrosine kinase inhibitor–induced activation of HER2 covalent dimers results in altered pharmacology

Previous studies have shown that erlotinib, an effective tyrosine kinase inhibitor (TKI) against EGFR ATP-site mutations including the E746-A750del mutation, induces covalent dimerization and activation for select allosteric EGFR oncogenic mutants (45). This results in paradoxical stimulation of cells expressing these mutants. We sought to understand whether covalently activated HER2 oncogenes also exhibit altered pharmacology for small molecules binding to the ATP site. We tested the effect of the FDA-approved inhibitor lapatinib on the proliferation of cells driven by HER2 WT (BT474) or the panel of BaF3 transformants expressing HER2 S310F, HER2 p95-M611, or HER2 Δ16 (Fig. 5A; Supplementary Table S4). For each mutant, lapatinib exhibited reduced potency versus HER2 WT. Because reduced activity for select EGFR TKIs against covalently activated EGFR mutants is associated with the propensity of the TKI to induce covalent dimerization, we tested the effect of lapatinib on covalent homodimerization for the HER2 p95-M611 mutant. Here, we similarly observe a dose-dependent increase in covalent homodimerization of HER2 p95-M611 following lapatinib treatment (Fig. 5B and C). These observations suggest that HER2 allosteric mutants with enhanced covalent homodimerization have altered pharmacology that renders existing TKIs ineffective (Fig. 6).

Figure 5.

TKIs reveal altered pharmacology of HER2 variants S310F, p95-M611, and Δ16. A, Concentration-dependent inhibition of HER2 WT (cell line, BT474) and variants (S310F, p95-M611, and Δ16; cell line Ba/F3) by lapatinib. B and C, Lapatinib-induced dimerization of HER2 p95-M611 variant.

Figure 5.

TKIs reveal altered pharmacology of HER2 variants S310F, p95-M611, and Δ16. A, Concentration-dependent inhibition of HER2 WT (cell line, BT474) and variants (S310F, p95-M611, and Δ16; cell line Ba/F3) by lapatinib. B and C, Lapatinib-induced dimerization of HER2 p95-M611 variant.

Close modal
Figure 6.

Common mechanism of oncogenicity and unique pharmacology of HER2 allosteric mutations. HER2 WT monomers in an extended state can either homodimerize or heterodimerize with other ERBB family members in a dynamic manner to facilitate the regulation of normal ERBB signaling activities (yellow). HER2 dimer interfaces mainly involve the cysteine-rich domains, CR1 and CR2, of the ECD, the TMD, the JMD, and the intracellular KD. The asymmetrical dimerization of KD is required for HER2 activation. WT HER2 is targeted by the HER2-selective antibody trastuzumab, which readily binds to the ECD to reduce the cell surface expression of HER2, and the tyrosine kinase inhibitor lapatinib, which shows high efficacy toward WT HER2. In comparison, allosteric HER2 oncogenic mutants display one of several features, such as free cysteine and increased hydrophobicity (the S310F position and the dimer arm region are shown in purple), that promote covalent or stable HER2 homodimerization and constitutive activation of HER2 signaling (red and yellow circles). Intracellular KD mutations that increase its asymmetrical dimerization contribute to HER2 activation through inside-out signaling. Conformational changes induced by allosteric mutations have resulted in reduced pharmacologic effects due to altered antibody binding sites and the TKI-induced dimerization of HER2 mutants.

Figure 6.

Common mechanism of oncogenicity and unique pharmacology of HER2 allosteric mutations. HER2 WT monomers in an extended state can either homodimerize or heterodimerize with other ERBB family members in a dynamic manner to facilitate the regulation of normal ERBB signaling activities (yellow). HER2 dimer interfaces mainly involve the cysteine-rich domains, CR1 and CR2, of the ECD, the TMD, the JMD, and the intracellular KD. The asymmetrical dimerization of KD is required for HER2 activation. WT HER2 is targeted by the HER2-selective antibody trastuzumab, which readily binds to the ECD to reduce the cell surface expression of HER2, and the tyrosine kinase inhibitor lapatinib, which shows high efficacy toward WT HER2. In comparison, allosteric HER2 oncogenic mutants display one of several features, such as free cysteine and increased hydrophobicity (the S310F position and the dimer arm region are shown in purple), that promote covalent or stable HER2 homodimerization and constitutive activation of HER2 signaling (red and yellow circles). Intracellular KD mutations that increase its asymmetrical dimerization contribute to HER2 activation through inside-out signaling. Conformational changes induced by allosteric mutations have resulted in reduced pharmacologic effects due to altered antibody binding sites and the TKI-induced dimerization of HER2 mutants.

Close modal

The expanded use of next-generation sequencing testing has revealed hundreds of unique mutations affecting HER2, which are distributed throughout the amino acid sequence, including the ECD, the TMD, and the KD. We have developed a MAP-scoring algorithm based on machine learning that can be used to predict for oncogenic mutations in silico, followed by scaled biology to experimentally validate oncogenicity in isogenic cell panels. We applied this technology to HER2 to reveal a spectrum of 37 driver mutations and 74 neutral mutations. This spectrum of driver mutations consists of 15 previously characterized oncogenic mutations and 22 newly characterized mutations that markedly increased Ba/F3 cell proliferation. All validated mutations, except for T862A, which occurs at the orthosteric ATP-binding site of HER2, are located at allosteric positions throughout the amino acid sequence. Further characterization of validated mutations revealed distinct proteomic fingerprints of oncogenic mutations clustered together within the ECD and KD of HER2. Some mutations are prevalent across tumor types, while other mutations are very rare. Our data demonstrate how even rarely occurring mutations in HER2 can exhibit potent oncogenicity and possess similar molecular attributes as prevalent driver mutations. As the number of unique mutations is expected to rise over time, an effective means to identify oncogenic mutations, such as MAP technology, will be crucial for expanding the use of precision medicine to treat cancers driven by rare oncogenic mutations.

Our study showed that covalent homodimerization is a common mechanism of oncogenicity for HER2 mutations affecting the ECD (Fig. 6). While covalent dimerization of HER2 has been previously proposed to facilitate enhanced activation of a limited number of HER2 mutations (36), we demonstrated that covalent homodimerization mechanisms have a common oncogenic role among several HER2 ECD allosteric mutations. This mechanism parallels our previous characterization of oncogenic EGFR mutations in glioblastoma, where most ECD mutations are activated as covalently linked dimers, including the EGFR variant III forming covalent dimers through EGFR C307 that is structurally and functionally equivalent to HER2 C311 (45, 53). We demonstrate for the first time how the HER2 S310F mutation, the most common HER2 mutation, is activated as a covalently linked dimer. In silico analysis of the HER2 ECD structure suggests the S310F mutation induces both localized and long-range structural disorders to generate free cysteines that engage in the intermolecular disulfide bond formation. This key observation was overlooked in prior studies that failed to address the oligomeric state of the phosphorylated HER2 S310F mutant (36). Our analysis of the phosphorylated pool of HER2 under nonreducing conditions reveals that activated HER2 proteins are largely covalent dimers when the HER2 S310F mutation is present, and this is true for at least six other ECD mutants of HER2. Covalent homodimerization as a mechanism for oncogenic activation pertains not only to HER2 genomic mutations, but also to HER2 oncogenic variants produced by alternative initiation of translation (HER2 p95-M611) and by alternative splicing (HER2 Δ16). There are numerous examples of other oncogenes undergoing activation through mutation-induced covalent dimerization (54).

We showed that these allosteric HER2 mutations exhibit a unique pharmacology that renders existing anti-HER2 therapies ineffective toward HER2 allosteric mutants (Fig. 6). These HER2 allosteric mutants demonstrate resistance toward trastuzumab, the anti-HER2 antibody that recognizes the ECD of HER2 WT, consistent with clinical data that show reduced effectiveness of trastuzumab in patients expressing the ECD variant HER2 p95-M611 (50). We found that the small-molecule TKI lapatinib displayed reduced activity against allosteric HER2 oncogenes versus HER2 WT. This reduction of activity was associated with the TKI-induced propensity of these mutants to promote HER2 dimerization (Fig. 6). This finding also parallels our observations of paradoxical dimer induction first described for EGFR-mutant oncogenes expressed in glioblastoma (45). The unique pharmacology that characterizes this family of HER2 allosteric oncogenes means that existing HER2 WT directed therapies (including antibodies and small molecules) may not be optimally used to treat cancers expressing HER2 allosteric oncogenes. Because allosteric mutations can introduce global changes in protein conformation that ultimately impact the pharmacology of the ATP-binding site (Fig. 6), screening of HER2 oncogenes as intact proteins in whole-cell assays is necessary for the design of a potent inhibitor against this family of HER2 allosteric oncogenes. Indeed, we have recently deployed our unique screening strategy to design a potent and selective inhibitor of allosteric ERBB mutants, including the allosteric HER2-mutant family addressed in this study (55). Recognition of mutation-induced dimerization as a general mode of activation among RTK oncogenes beyond the ERBB family points to the necessity of novel inhibitor design approaches to target allosteric mutations currently unaddressed by existing RTK-directed therapies.

N. Ishiyama reports a patent for METHODS OF CHARACTERIZING AND CLASSIFYING SOMATIC VARIANTS pending to none and being an employee and shareholder of Black Diamond Therapeutics. A. Salomatov reports a patent for METHODS OF CHARACTERIZING AND CLASSIFYING SOMATIC VARIANTS pending to none. D. Romashko reports a patent for ASET-023 - Methods of Treating Cancers with Alkyne Substituted Quinazoline Derivatives pending. S. Thakur is an employee and shareholder of Black Diamond Therapeutics. A. Mentes is an employee and shareholder of Black Diamond Therapeutics. J.F. Hopkins is an employee of Foundation Medicine Inc. and a stockholder of Roche Holdings AG. G.M. Frampton is an employee of Foundation Medicine Inc. and shareholder of Roche AG. L.A. Albacker reports employment at Foundation Medicine, Inc.. and stockholder of Roche Holdings AG. C. Roberts reports personal fees from Black Diamond Therapeutics, Inc. during the conduct of the study and personal fees from Civetta Therapeutics, Outrun Therapeutics, and Stablix Therapeutics outside the submitted work. E. Buck reports a patent for Methods of Characterizing and Classifying Somatic Variants pending. No disclosures were reported by the other authors.

N. Ishiyama: Conceptualization, data curation, software, investigation, visualization, methodology, writing–original draft, writing–review and editing. M. O'Connor: Resources, investigation, visualization, methodology, writing–original draft. A. Salomatov: Conceptualization, data curation, software, investigation, methodology, writing–original draft. D. Romashko: Resources, investigation, visualization, methodology. S. Thakur: Data curation, investigation, visualization. A. Mentes: Software, investigation, visualization, methodology, writing–original draft. J.F. Hopkins: Data curation, investigation, writing–review and editing. G.M. Frampton: Data curation, investigation. L.A. Albacker: Data curation, investigation, writing–original draft. A. Kohlmann: Software, supervision, investigation, writing–original draft, project administration, writing–review and editing. C. Roberts: Supervision, investigation, project administration, writing–review and editing. E. Buck: Conceptualization, supervision, investigation, writing–original draft, project administration, writing–review and editing.

Professional writing assistance was provided by Emily Cullinan, PhD, and Francesca Balordi, PhD, of The Lockwood Group (Stamford, Connecticut) in accordance with Good Publication Practice (GPP3) guidelines, with funding by Black Diamond Therapeutics, Inc.

The publication costs of this article were defrayed in part by the payment of publication fees. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

Note: Supplementary data for this article are available at Cancer Research Online (http://cancerres.aacrjournals.org/).

1.
Pahuja
KB
,
Nguyen
TT
,
Jaiswal
BS
,
Prabhash
K
,
Thaker
TM
,
Senger
K
, et al
.
Actionable activating oncogenic ERBB2/HER2 transmembrane and juxtamembrane domain mutations
.
Cancer Cell
2018
;
34
:
792
806
.
2.
Rubin
I
,
Yarden
Y
.
The basic biology of HER2
.
Ann Oncol
2001
;
12
:
S3
S8
.
3.
Tai
W
,
Mahato
R
,
Cheng
K
.
The role of HER2 in cancer therapy and targeted drug delivery
.
J Control Release
2010
;
146
:
264
75
.
4.
Zhao
J
,
Xia
Y
.
Targeting HER2 alterations in non–small-cell lung cancer: a comprehensive review
.
JCO Precis Oncol
2020
;
4
:
411
25
.
5.
Gaibar
M
,
Beltran
L
,
Romero-Lorca
A
,
Fernandez-Santander
A
,
Novillo
A
.
Somatic mutations in HER2 and implications for current treatment paradigms in HER2-positive breast cancer
.
J Oncol
2020
;
2020
:
6375956
.
6.
Roskoski
R
Jr
.
Small molecule inhibitors targeting the EGFR/ErbB family of protein-tyrosine kinases in human cancers
.
Pharmacol Res
2019
;
139
:
395
411
.
7.
Kovacs
E
,
Zorn
JA
,
Huang
Y
,
Barros
T
,
Kuriyan
J
.
A structural perspective on the regulation of the epidermal growth factor receptor
.
Annu Rev Biochem
2015
;
84
:
739
64
.
8.
Chmielecki
J
,
Ross
JS
,
Wang
K
,
Frampton
GM
,
Palmer
GA
,
Ali
SM
, et al
.
Oncogenic alterations in ERBB2/HER2 represent potential therapeutic targets across tumors from diverse anatomic sites of origin
.
Oncologist
2015
;
20
:
7
12
.
9.
TUKYSA [prescribing information]
.
Bothell, WA
:
Seattle Genetics, Inc
;
2020
.
10.
NERLYNX [prescribing information]
.
Los Angeles, CA
:
Puma Biotechnology, Inc
;
2020
.
11.
TYKERB [prescribing information]
.
East Hanover, NJ
:
Novartis Pharmaceuticals Corp
;
2018
.
12.
ENHERTU [prescribing information]
.
Wilmington, DE
:
AstraZeneca Pharmaceuticals
;
2019
.
13.
KADCYLA [prescribing information]
.
South San Francisco, CA
:
Genentech, Inc
;
2020
.
14.
PERJETA [prescribing information]
.
South San Francisco, CA
:
Genentech, Inc
;
2020
.
15.
HERCEPTIN [prescribing information]
.
South San Francisco, CA
:
Genentech, Inc
;
2018
.
16.
MARGENZA [prescribing information]
.
Rockville, MD
:
MacroGenics, Inc
;
2020
.
17.
Riese
DJ
2nd
,
van Raaij
TM
,
Plowman
GD
,
Andrews
GC
,
Stern
DF
.
The cellular response to neuregulins is governed by complex interactions of the erbB receptor family
.
Mol Cell Biol
1995
;
15
:
5770
6
.
18.
Jacobson
MP
,
Pincus
DL
,
Rapp
CS
,
Day
TJ
,
Honig
B
,
Shaw
DE
, et al
.
A hierarchical approach to all-atom protein loop prediction
.
Proteins
2004
;
55
:
351
67
.
19.
Humphrey
W
,
Dalke
A
,
Schulten
K
.
VMD: visual molecular dynamics
.
J Mol Graph
1996
;
14
:
33
8
.
20.
Phillips
JC
,
Braun
R
,
Wang
W
,
Gumbart
J
,
Tajkhorshid
E
,
Villa
E
, et al
.
Scalable molecular dynamics with NAMD
.
J Comput Chem
2005
;
26
:
1781
802
.
21.
MacKerell
AD
,
Bashford
D
,
Bellott
M
,
Dunbrack
RL
,
Evanseck
JD
,
Field
MJ
, et al
.
All-atom empirical potential for molecular modeling and dynamics studies of proteins
.
J Phys Chem B
1998
;
102
:
3586
616
.
22.
MacKerell
AD
,
Feig
M
,
Brooks
CL
.
Improved treatment of the protein backbone in empirical force fields
.
J Am Chem Soc
2004
;
126
:
698
9
.
23.
Jorgensen
WL
,
Chandrasekhar
J
,
Madura
JD
,
Impey
RW
,
Klein
ML
.
Comparison of simple potential functions for simulating liquid water
.
J Chem Phys
1983
;
79
:
926
35
.
24.
Essmann
U
,
Perera
L
,
Berkowitz
ML
,
Darden
T
,
Lee
H
,
Pedersen
LG
.
A smooth particle mesh Ewald method
.
J Chem Phys
1995
;
103
:
8577
93
.
25.
Ryckaert
JP
,
Coccotti
G
,
Berendsen
HJC
.
Numerical integration of the cartesian equations of motion of a system with constraints: molecular dynamics of n-alkanes
.
J Comput Phys
1977
;
23
:
327
41
.
26.
Atilgan
C
,
Atilgan
AR
.
Perturbation-response scanning reveals ligand entry-exit mechanisms of ferric binding protein
.
PLoS Comput Biol
2009
;
5
:
e1000544
.
27.
General
IJ
,
Liu
Y
,
Blackburn
ME
,
Mao
W
,
Gierasch
LM
,
Bahar
I
.
ATPase subdomain IA is a mediator of interdomain allostery in Hsp70 molecular chaperones
.
PLoS Comput Biol
2014
;
10
:
e1003624
.
28.
Bakan
A
,
Dutta
A
,
Mao
W
,
Liu
Y
,
Chennubhotla
C
,
Lezon
TR
, et al
.
Evol and ProDy for bridging protein sequence evolution and structural dynamics
.
Bioinformatics
2014
;
30
:
2681
3
.
29.
Chakravarty
D
,
Gao
J
,
Phillips
SM
,
Kundra
R
,
Zhang
H
,
Wang
J
, et al
.
OncoKB: a precision oncology knowledge base
.
JCO Precis Oncol
2017
;
2017
:
PO.17.00011
.
30.
Armenia
J
,
Wankowicz
SAM
,
Liu
D
,
Gao
J
,
Kundra
R
,
Reznik
E
, et al
.
The long tail of oncogenic drivers in prostate cancer
.
Nat Genet
2018
;
50
:
645
51
.
31.
Cannataro
VL
,
Gaffney
SG
,
Townsend
JP
.
Effect sizes of somatic mutations in cancer
.
J Natl Cancer Inst
2018
;
110
:
1171
7
.
32.
Lemmon
MA
.
Ligand-induced ErbB receptor dimerization
.
Exp Cell Res
2009
;
315
:
638
48
.
33.
Aertgeerts
K
,
Skene
R
,
Yano
J
,
Sang
BC
,
Zou
H
,
Snell
G
, et al
.
Structural analysis of the mechanism of inhibition and allosteric activation of the kinase domain of HER2 protein
.
J Biol Chem
2011
;
286
:
18756
65
.
34.
Wang
SE
,
Narasanna
A
,
Perez-Torres
M
,
Xiang
B
,
Wu
FY
,
Yang
S
, et al
.
HER2 kinase domain mutation results in constitutive phosphorylation and activation of HER2 and EGFR and resistance to EGFR tyrosine kinase inhibitors
.
Cancer Cell
2006
;
10
:
25
38
.
35.
Bose
R
,
Kavuri
SM
,
Searleman
AC
,
Shen
W
,
Shen
D
,
Koboldt
DC
, et al
.
Activating HER2 mutations in HER2 gene amplification negative breast cancer
.
Cancer Discov
2013
;
3
:
224
37
.
36.
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
.
37.
Hyman
DM
,
Piha-Paul
SA
,
Won
H
,
Rodon
J
,
Saura
C
,
Shapiro
GI
, et al
.
HER kinase inhibition in patients with HER2- and HER3-mutant cancers
.
Nature
2018
;
554
:
189
94
.
38.
Kancha
RK
,
von Bubnoff
N
,
Bartosch
N
,
Peschel
C
,
Engh
RA
,
Duyster
J
.
Differential sensitivity of ERBB2 kinase domain mutations towards lapatinib
.
PLoS One
2011
;
6
:
e26760
.
39.
Nagano
M
,
Kohsaka
S
,
Ueno
T
,
Kojima
S
,
Saka
K
,
Iwase
H
, et al
.
High-throughput Functional evaluation of variants of unknown significance in ERBB2
.
Clin Cancer Res
2018
;
24
:
5112
22
.
40.
Yu
DH
,
Tang
L
,
Dong
H
,
Dong
Z
,
Zhang
L
,
Fu
J
, et al
.
Oncogenic HER2 fusions in gastric cancer
.
J Transl Med
2015
;
13
:
116
.
41.
de Martino
M
,
Zhuang
D
,
Klatte
T
,
Rieken
M
,
Roupret
M
,
Xylinas
E
, et al
.
Impact of ERBB2 mutations on in vitro sensitivity of bladder cancer to lapatinib
.
Cancer Biol Ther
2014
;
15
:
1239
47
.
42.
Nayar
U
,
Cohen
O
,
Kapstad
C
,
Cuoco
MS
,
Waks
AG
,
Wander
SA
, et al
.
Acquired HER2 mutations in ER+ metastatic breast cancer confer resistance to estrogen receptor-directed therapies
.
Nat Genet
2019
;
51
:
207
16
.
43.
Yamamoto
H
,
Higasa
K
,
Sakaguchi
M
,
Shien
K
,
Soh
J
,
Ichimura
K
, et al
.
Novel germline mutation in the transmembrane domain of HER2 in familial lung adenocarcinomas
.
J Natl Cancer Inst
2014
;
106
:
djt338
.
44.
Ng
PK
,
Li
J
,
Jeong
KJ
,
Shao
S
,
Chen
H
,
Tsang
YH
, et al
.
Systematic functional annotation of somatic mutations in cancer
.
Cancer Cell
2018
;
33
:
450
62
.
45.
O'Connor
M
,
Zhang
J
,
Markovic
S
,
Romashko
D
,
Salomatov
A
,
Ishiyama
N
, et al
.
EGFR oncogenes expressed in glioblastoma are activated as covalent dimers and paradoxically stimulated by erlotinib
.
bioRxiv
2019
;
810721
.
46.
Siegel
PM
,
Muller
WJ
.
Mutations affecting conserved cysteine residues within the extracellular domain of Neu promote receptor dimerization and activation
.
Proc Natl Acad Sci U S A
1996
;
93
:
8878
83
.
47.
Siegel
PM
,
Dankort
DL
,
Hardy
WR
,
Muller
WJ
.
Novel activating mutations in the neu proto-oncogene involved in induction of mammary tumors
.
Mol Cell Biol
1994
;
14
:
7068
77
.
48.
Lu
C
,
Mi
L-Z
,
Grey
MJ
,
Zhu
J
,
Graef
E
,
Yokoyama
S
, et al
.
Structural evidence for loose linkage between ligand binding and kinase activation in the epidermal growth factor receptor
.
Mol Cell Biol
2010
;
30
:
5432
43
.
49.
Duchnowska
R
,
Sperinde
J
,
Chenna
A
,
Haddad
M
,
Paquet
A
,
Lie
Y
, et al
.
Quantitative measurements of tumoral p95HER2 protein expression in metastatic breast cancer patients treated with trastuzumab: independent validation of the p95HER2 clinical cutoff
.
Clin Cancer Res
2014
;
20
:
2805
13
.
50.
Scaltriti
M
,
Rojo
F
,
Ocaña
A
,
Anido
J
,
Guzman
M
,
Cortes
J
, et al
.
Expression of p95HER2, a truncated form of the HER2 receptor, and response to anti-HER2 therapies in breast cancer
.
J Natl Cancer Inst
2007
;
99
:
628
38
.
51.
Sperinde
J
,
Jin
X
,
Banerjee
J
,
Penuel
E
,
Saha
A
,
Diedrich
G
, et al
.
Quantitation of p95HER2 in paraffin sections by using a p95-specific antibody and correlation with outcome in a cohort of trastuzumab-treated breast cancer patients
.
Clin Cancer Res
2010
;
16
:
4226
35
.
52.
Growdon
WB
,
Groeneweg
J
,
Byron
V
,
DiGloria
C
,
Borger
DR
,
Tambouret
R
, et al
.
HER2 over-expressing high grade endometrial cancer expresses high levels of p95HER2 variant
.
Gynecol Oncol
2015
;
137
:
160
6
.
53.
Casolari
DA
,
Nguyen
T
,
Butcher
CM
,
Iarossi
DG
,
Hahn
CN
,
Bray
SC
, et al
.
A novel, somatic, transforming mutation in the extracellular domain of epidermal growth factor receptor identified in myeloproliferative neoplasm
.
Sci Rep
2017
;
7
:
2467
.
54.
Robertson
SC
,
Meyer
AN
,
Hart
KC
,
Galvin
BD
,
Webster
MK
,
Donoghue
DJ
.
Activating mutations in the extracellular domain of the fibroblast growth factor receptor 2 function by disruption of the disulfide bond in the third immunoglobulin-like domain
.
Proc Natl Acad Sci U S A
1998
;
95
:
4567
72
.
55.
Flohr
A
,
O'Connor
M
,
Ottaviani
G
,
N.
W
,
Romashko
D
,
Weibel
F
, et al
.
BDTX-189, a potent and selective inhibitor against the family of allosteric EGFR and HER2 mutants
.
Cancer Res
2022
.
This open access article is distributed under the Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0) license.