Purpose:

We report updated clinical outcomes from a phase II study of pembrolizumab, trastuzumab, and chemotherapy (PTC) in metastatic esophagogastric cancer in conjunction with outcomes from an independent Memorial Sloan Kettering (MSK) cohort.

Patients and Methods:

The significance of pretreatment 89Zr-trastuzumab PET, plasma circulating tumor DNA (ctDNA) dynamics, and tumor HER2 expression and whole exome sequencing was evaluated to identify prognostic biomarkers and mechanisms of resistance in patients treated on-protocol with PTC. Additional prognostic features were evaluated using a multivariable Cox regression model of trastuzumab-treated MSK patients (n = 226). Single-cell RNA sequencing (scRNA-seq) data from MSK and Samsung were evaluated for mechanisms of therapy resistance.

Results:

89Zr-trastuzumab PET, scRNA-seq, and serial ctDNA with CT imaging identified how pre-treatment intrapatient genomic heterogeneity contributes to inferior progression-free survival (PFS). We demonstrated that the presence of intensely avid lesions by 89Zr-trastuzumab PET declines in tumor-matched ctDNA by 3 weeks, and clearance of tumor-matched ctDNA by 9 weeks were minimally invasive biomarkers of durable PFS. Paired pre- and on-treatment scRNA-seq identified rapid clearance of HER2-expressing tumor clones with expansion of clones expressing a transcriptional resistance program, which was associated with MT1H, MT1E, MT2A, and MSMB expression. Among trastuzumab-treated patients at MSK, ERBB2 amplification was associated with improved PFS, while alterations in MYC and CDKN2A/B were associated with inferior PFS.

Conclusions:

These findings highlight the clinical relevance of identifying baseline intrapatient heterogeneity and serial ctDNA monitoring of HER2-positive esophagogastric cancer patients to identify early evidence of treatment resistance, which could guide proactive therapy escalation or deescalation.

Translational Relevance

The addition of PD-1 inhibition to chemotherapy and trastuzumab demonstrates promising response rate and survival, and durable benefit can be predicted by evaluating ctDNA decline and clearance and high pretreatment 89Zr-trastuzumab PET uptake. Single-cell sequencing identified MT1H, MT1E, MT2A, and MSMB as transcriptionally regulating resistance, which may serve as future therapeutic targets.

Approximately 20% of esophagogastric cancer (EGC) patients’ tumors carry amplifications of ERBB2 or overexpress its product, human epidermal growth factor receptor 2 (HER2). For these patients, the anti-HER2 antibody trastuzumab and chemotherapy (TC) have been the standard treatment for over a decade (1). The combination of these agents with pembrolizumab, an anti-programmed death 1 (PD-1) antibody, gained fast-track approval in first-line HER2-positive esophagogastric cancer by the United States Food and Drug Administration based on a 74.4% objective response rate in the Phase III KEYNOTE 811 study with a 22.7% improvement in objective responses versus trastuzumab and chemotherapy alone (2).

In addition to providing updated survival results, which build upon concordant short follow-up results seen in INTEGA and PANTHERA, we present expanded biomarker analysis from our antecedent phase II trial (3–5). In the phase II cohort, more than half of the patients had developed new non-target “escape” lesions upon progression, either due to loss of HER2 expression or via genomically mediated resistance mechanisms. We hypothesized that escape lesions reflect preexisting tumor heterogeneity between disease sites (intrapatient) and within a disease site (intratumor) that eventually conferred resistance. Therefore, we sought to understand how we can better predict durable responders by using biomarkers derived from integrated clinical characteristics, tumor and plasma genomics, and molecular imaging features generated from patients treated in a phase II trial or in clinically annotated institutional cohorts of HER2-positive EGC patients treated with trastuzumab and chemotherapy with or without PD-1 inhibition (Supplementary Table S1) to identify putative biomarkers of intrapatient tumor heterogeneity and their prognostic significance. Finally, additional paired pre- and on-treatment TC single-cell RNA sequencing (scRNA-seq) was used to identify a transcriptional resistance program that allowed certain tumor clones to persist despite therapy and may serve as future therapeutic targets.

Study design, participants, interventions, and sample size

Patients with HER2+ [defined as IHC 3+ or IHC 2+ and HER2:CEP17 fluorescence in situ hybridization (FISH) ratio ≥ 2.0] metastatic esophageal, gastroesophageal junction, or gastric adenocarcinoma (n = 37) were recruited to an open-label, nonrandomized, single-arm, investigator-initiated, single-institution phase II trial of trastuzumab, pembrolizumab, capecitabine, and oxaliplatin. The study protocol and all amendments were approved by the Memorial Sloan Kettering (MSK) Institutional Review Board (IRB 16–937) and the trial was registered with ClinicalTrials.gov (NCT02954536). Treatment and procedure details have been published (3). In addition to standard response assessment (RECIST 1.1), up to 5 lesions from each affected disease site (i.e., up to 5 lymph nodes, 5 liver metastases, 5 lung nodules, etc.) were measured for “expanded assessment” to better account for response heterogeneity, including lesions smaller than 1 cm at baseline. The phase II protocol was performed in accordance with the protocol and its amendments and Good Clinical Practice guidelines and was overseen by the MSK Data and Safety Monitoring Committee. All patients provided written informed consent before enrollment.

An additional retrospective cohort of patients in the MSK esophagogastric database who received platinum chemotherapy with HER2-directed therapy (trastuzumab or zanidatamab) from 2010 to 2022 were manually abstracted for demographic, pathologic, radiographic, and genomic features under a separate IRB-approved retrospective protocol.

Immunohistochemistry (IHC) and FISH

HER2 clinical testing was performed by IHC using the PATHWAY anti-HER2/neu (4B5) assay (Ventana) and programmed death-ligand 1 (PD-L1) testing using the E1L3N clone (Cell Signaling Technology). MET IHC was performed, when relevant, using Ventana clone SP44. HER2 IHC was considered uniformly 3+ if all pretreatment testing at MSK or outside institutions was interpreted as 3+. Conversely, if pretreatment HER2 IHC was ever interpreted as ≤2+, the patient's tumor was considered 2+/heterogeneous.

HER2 FISH was performed in cases with equivocal (2+ intensity) HER2 immunohistochemistry staining using HER2 IQFISH pharmDx (Dako) with scoring per clinical guidelines (6). FISH for MET was performed on paraffin section using a 2-color MET/CEN7 probe consisting of BAC clones spanning the MET gene (RP11–39K12 and RP11–163L9 labeled with red dUTP) and a centromeric repeat plasmid for Chr 7 (P7t1 labeled with green dUTP) as control. MET amplification was defined as ≥ 2 MET/CEN7 ratio or ≥ 6 MET copies (discrete signal) or the presence of at least one MET cluster (≥ 4 copies; low-level amplicon resulting from tandem duplications) in >10% of cells.

89Zr-trastuzumab PET

A subset of phase II trial patients were additionally consented to investigational PET imaging on MSK IRB protocol 13–165. Whole-body PET/CT scans from mid skull to proximal thigh were performed on days 5 to 7 following intravenous injection of 5 mCi of 89Zr-trastuzumab. All scans were obtained on a dedicated PET/CT scanner (Discovery STE GE Healthcare) using low-dose CT scans for attenuation correction (10–80 mA current) and PET acquisition for 7 to 8 minutes per bed position in 3-dimensional mode and attenuation, scatter, and other standard corrections were applied to generate images using iterative reconstruction. Localization in the tumor was defined as positive if focal accumulation of activity was greater than adjacent background activity and distinct from areas of expected physiologic activity. Volumes of interest were drawn to include lesions on PET images using dedicated software (Hermes Medical Solutions; ref. 7), and images were independently assessed by a nuclear radiologist. Only lesions computed tomography (CT) measurements per “expanded assessment” as described above were evaluated. Lesion uptake was classified based upon uptake values (SUV) categorized as low (3 ≤ SUVmax < 5), moderate (5 ≤ SUVmax < 10), intense (10 ≤ SUVmax < 15), or very intense (15 ≤ SUVmax; ref. 8).

Next-generation sequencing (NGS)

Collection, processing, and analysis of tissue and plasma NGS samples are described in the Supplementary Methods section.

Single-cell RNA-seq

Fresh tissue was collected from a patient receiving capecitabine and oxaliplatin (CAPOX) and trastuzumab ± pembrolizumab at MSK and underwent processing and analysis as described in the Supplementary Methods section. Additionally, paired scRNA-seq data collected before cycles 1 and 3 from patients who received CAPOX with (n = 3) or without trastuzumab (n = 4) was provided by Samsung Medical Center and processed as previously described (9).

Statistical analysis

Patient demographic characteristics were summarized using descriptive summary statistics and compared between groups of interest using Fisher exact test for categorical data or Wilcoxon rank-sum test for continuous variables. Progression-free survival (PFS) and overall survival (OS) were calculated from the date of treatment until date of first progression or death, whichever occurred first for PFS or date of death for OS. Patients alive and without progression of disease (POD) were censored at their last available scan. PFS and OS were estimated using Kaplan–Meier methods and compared between subgroups using the log-rank test. Among patients treated with pembrolizumab, trastuzumab, and chemotherapy on-protocol, a landmark time of 9 weeks was used to examine the association between response status and clearance of all tumor-matched ctDNA at 9 weeks with OS and PFS. Patients without the 9-week follow-up were excluded. Univariate PFS analysis was done using Cox proportional hazards model and included all patients who received trastuzumab and chemotherapy with or without PD-1 inhibitor (n = 217). The final multivariable PFS model was created by including the main covariate of interest (trastuzumab and chemotherapy with or without PD-1 inhibitor) and adjusting for factors associated with PFS from univariate analysis at P < 0.05. TP53 alteration was not included in the final multivariable analysis since it was highly correlated with ERBB2 amplification status. All statistical analyses were done using R version 4.0.4. All P values were two-sided and P < 0.05 was considered statistically significant.

Code availability

Data were analyzed using publicly available computational packages as described in the NGS and scRNA-seq sections above.

Data availability

Clinical data and key genomic features for each patient are provided in the Supplementary Data. Targeted sequencing data generated for this study are available at the European Variation Archive (EVA) accession #PRJEB63072 (https://www.ebi.ac.uk/eva/?eva-study=PRJEB63072) and at https://www.cbioportal.org/study/summary?id=egc_trap_ccr_msk_2023.

Addition of PD-1 blockade is associated with increased survival and objective response rate

Clinical data for the phase II study was updated on April 1, 2022, providing nearly three additional years of follow-up since the prior publication. Six of 37 patients have not yet progressed, and 5 of these patients are still being treated on-study with a median follow-up time of 44 (range: 41–63) months. Median PFS was 13 [95% confidence interval (CI): 8.5–20] months, median OS was 27 (95% CI: 21–44) months, and 31 of 35 evaluable patients (89%; 95% CI: 73%–97%) achieved a RECIST response. Since study enrollment was completed, an additional 29 patients received PD-1 inhibition (nivolumab or pembrolizumab) in combination with TC in standard practice, and achieved a median 14.7-month [95% CI, 11.2–not reached (NR)] PFS, median OS was not reached, 80% (23/29; 95% CI: 60%–92%) objective response rate (ORR) with a median of 10.4 months of follow-up (Supplementary Figs. S1 and S2; Supplementary Tables S1 and S2; Supplementary Data S1).

Homogeneous HER2 NR, and overexpression by IHC correlates with benefit from trastuzumab and chemotherapy

Building upon prior findings that high ERBB2 gene copy number and homogeneity predict improved PFS (3, 10), we evaluated the correlation between intensity and homogeneity of pretreatment HER2 IHC overexpression and PFS in 193 patients with HER2-positive EGC treated with first-line chemotherapy with trastuzumab with or without PD-1 inhibitor. Patients with uniformly high pretreatment tumor HER2 overexpression [IHC 3+; n = 129/193 (67%)] who received either PD-1 inhibition and TC or TC alone had longer median PFS [15 (95% CI: 13–20) vs. 8.5 (95% CI: 6.4–13) months; CI, 0.43–0.86; Fig. 1A] compared with patients with indeterminate (HER2 IHC 2+) or heterogeneous (HER2 IHC 3+ and HER2 IHC 0–1+) HER2 expression within a disease site or between sites (n = 64/193) pretreatment. This correlation may result from greater and/or more uniform HER2 overexpression in ERBB2-amplified tumors, as ERBB2 was amplified in the tumors of 110 of 125 patients (88%) with HER2 3+ overexpression, versus 21 of 58 (36%) evaluable patients with HER2 2+/heterogeneous expression (P = 1.8e−12; Supplementary Data S1).

Figure 1.

IHC, ctDNA, and 89Zr-trastuzumab PET imaging predict durable clinical benefit from trastuzumab, pembrolizumab, and chemotherapy. A, PFS of patients stratified according to baseline testing into HER2+ (IHC 3+ expression) or “indeterminate/heterogeneous” (IHC 2+/FISH+ or heterogeneous expression between disease sites or between testing at MSK and another site). B, Timeline of disease evolution in a patient who remains on therapy for over 30 months after a rapid and durable response. The patient had homogeneous high pretreatment 89Zr-trastuzumab PET avidity in all sites (right) and clearance of ctDNA after pembrolizumab and trastuzumab induction. Tumor burden (cumulative unidimensional lesion measurements) declined in conjunction with ctDNA. Lesion-level unidimensional measurements and post-induction 89Zr-trastuzumab PET demonstrated response in all lesions. C, Timeline of a patient who had a HER2+ primary tumor and HER2 metastatic biopsy at baseline, and no ERBB2 amplification in tissue or ctDNA at any time. ctDNA maxVAF and tumor burden increased with pembrolizumab and trastuzumab induction, followed by decline once chemotherapy was added. Several lesions were trastuzumab-non-avid by 89Zr-trastuzumab PET (dashed lines), and a mixed response in a celiac lymph node and increasing ctDNA maxVAF at 4 months were identified prior to RECIST progression. FGFR2 amplification was detected in ctDNA at baseline, which likely contributed to resistance along with intertumoral HER2 heterogeneity. D, PFS of patients in whom ctDNA was cleared by 9 weeks versus those with persistent ctDNA. E, PFS of patients with uniform response at 9 weeks versus those with mixed responses.

Figure 1.

IHC, ctDNA, and 89Zr-trastuzumab PET imaging predict durable clinical benefit from trastuzumab, pembrolizumab, and chemotherapy. A, PFS of patients stratified according to baseline testing into HER2+ (IHC 3+ expression) or “indeterminate/heterogeneous” (IHC 2+/FISH+ or heterogeneous expression between disease sites or between testing at MSK and another site). B, Timeline of disease evolution in a patient who remains on therapy for over 30 months after a rapid and durable response. The patient had homogeneous high pretreatment 89Zr-trastuzumab PET avidity in all sites (right) and clearance of ctDNA after pembrolizumab and trastuzumab induction. Tumor burden (cumulative unidimensional lesion measurements) declined in conjunction with ctDNA. Lesion-level unidimensional measurements and post-induction 89Zr-trastuzumab PET demonstrated response in all lesions. C, Timeline of a patient who had a HER2+ primary tumor and HER2 metastatic biopsy at baseline, and no ERBB2 amplification in tissue or ctDNA at any time. ctDNA maxVAF and tumor burden increased with pembrolizumab and trastuzumab induction, followed by decline once chemotherapy was added. Several lesions were trastuzumab-non-avid by 89Zr-trastuzumab PET (dashed lines), and a mixed response in a celiac lymph node and increasing ctDNA maxVAF at 4 months were identified prior to RECIST progression. FGFR2 amplification was detected in ctDNA at baseline, which likely contributed to resistance along with intertumoral HER2 heterogeneity. D, PFS of patients in whom ctDNA was cleared by 9 weeks versus those with persistent ctDNA. E, PFS of patients with uniform response at 9 weeks versus those with mixed responses.

Close modal

HER2 expression by 89Zr-trastuzumab imaging correlates with lesion-level response

As pretreatment biopsies of multiple metastatic disease sites are not feasible in routine care, we examined whether noninvasive detection of HER2 expression heterogeneity via 89Zr-trastuzumab imaging could serve as a biomarker of treatment response. Among the 25 on-protocol patients who received a dose of pembrolizumab and trastuzumab “induction” 3 weeks before initiation of concurrent chemotherapy, 8 patients (32%) underwent 89Zr-trastuzumab PET prior to initiating therapy; 7 of 8 patients (88%) had at least 1 PET-avid lesion (Fig. 1B; Supplementary Table S3; Supplementary Data S2 and S3), including all evaluable patients with high HER2 expression. We observed significant variability in PET avidity between disease sites within patients pre-treatment. Of 47 assessable pretreatment lesions, 31 (66%) were PET-avid (SUV ≥ 3); of these, 48% (15/31) demonstrated intense 89Zr-trastuzumab avidity (SUV ≥ 10; Supplementary Data S2). All 15 intensely 89Zr-trastuzumab avid lesions decreased in size by CT after a single dose of pembrolizumab and trastuzumab, whereas only 9 of 32 non-intensely avid lesions (8%) showed reduction (P = 2.4 ×10−6 by Fisher exact test; Supplementary Fig. S3). All 4 patients with ≥ 1 intensively avid (SUV ≥ 10) lesion at baseline achieved 6-month PFS (Supplementary Data Table S2; example shown in Fig. 1C), versus only 2 of 4 patients without intensely avid lesions (SUV < 10; example in Fig. 1D). Three of these patients also had plasma ctDNA ERBB2 amplification, and exceeded a 6-month PFS, while the two patients lacking both 89Zr-trastuzumab intense avidity and plasma ctDNA ERBB2 amplification failed to achieve a 6-month PFS (Supplementary Data Table S2). However, 1 patient with low-volume lymph node–only disease lacked PET-avid lesions at baseline yet achieved an 8.7-month PFS. These results, though limited by the small sample size, suggest that 89Zr-trastuzumab PET can noninvasively reveal the intensity and uniformity of HER2 expression, which may correlate with clinical benefit from pembrolizumab and trastuzumab.

Early ctDNA decline and clearance of ctDNA correlate with PFS

We previously demonstrated that pretreatment plasma ERBB2 amplification predicts durable PFS (3, 11), and so we next explored the utility of early ctDNA decline as a biomarker of ≥6-month PFS on pembrolizumab with chemotherapy and trastuzumab. Of patients with tumor-matched (see Materials and Methods) mutations at baseline, 12 of 16 (75%) had a decline in maximum variant allelic frequency (maxVAF) by week 3, and 9 of these patients (75%) achieved 6-month PFS, versus 0 of 4 patients (0%) with rising maxVAF at week 3 (Supplementary Fig. S4; Supplementary Data S3). Plasma ctDNA dynamics were then compared with lesion-level CT scan dynamics. At 3-week CT, no responding lesions were identified in patients lacking ERBB2-amplified ctDNA (Supplementary Fig. S5A). In contrast, lesions from 5 of 9 patients (56%) lacking ERBB2 tissue amplification responded to pembrolizumab and trastuzumab by 3-week CT, suggesting intrapatient HER2 heterogeneity (Supplementary Fig. S5B; Supplementary Data S3). Only 3 of 10 patients (30%) who achieved both 6-month PFS and decline in ctDNA demonstrated a uniform CT response in all disease sites (P = 0.58), highlighting the discordance of early ctDNA with CT imaging.

As decline in VAF fails to account for copy number alteration events, we also evaluated clearance of all tumor-matched ctDNA alterations as a biomarker of survival. Of the 23 patients who had tumor-matched ctDNA alterations, the 17 in whom all were cleared by 9 weeks exhibited longer PFS than in the 6 patients who did not (HR 0.18; 95% CI, 0.06–0.53; Fig. 1D). While 13 of 17 patients (76%) who cleared ctDNA achieved 6-month PFS, only 1 of 6 (17%) who failed to clear ctDNA did. This performed similarly to follow-up imaging where phase II pembrolizumab, trastuzumab, and chemotherapy-treated patients with uniform CT responses (i.e., all lesions radiographically improving) at 9 weeks (21/35; 60%) had a decreased risk of progression or death (HR 0.38; 95% CI, 0.17–0.81; Fig. 1E) compared with patients with mixed responses (14/35; 40%). This suggests that ctDNA clearance may have a role in reducing the need for frequent cross-sectional imaging.

ctDNA reveals genomic mechanisms of resistance

Of the 31 pembrolizumab, trastuzumab, and chemotherapy-treated trial patients whose disease progressed, 19 (61%) developed escape lesions that were not present on baseline CT, including 3 (10%) who developed brain metastases. While all 37 protocol patients had HER2-positive tumors at baseline, only 11 of 21 patients (52%) remained HER2-positive upon progression, corresponding to 9 of 18 (50%) with ERBB2 amplification in tissue and 8 of 21 (38%) in plasma (variation in denominators reflects data availability; Supplementary Fig. S6). There was no significant change seen in tumor mutation burden post-treatment. As site-matched pre- and posttreatment biopsies were not available for these lesions, ctDNA evaluation improved sensitivity to detect baseline and acquired resistance mechanisms. Eighteen of 21 (86%) evaluable patients had tumor-matched ctDNA mutations upon progression (Supplementary Data S1). However, only 49% of plasma NGS alterations could be tumor-matched, as the nonmatched mutations presumably arose outside the sequenced tumor site or via clonal hematopoiesis (Supplementary Table S4). Tissue and/or ctDNA testing identified putative mechanisms of resistance in nearly all patients, including loss of HER2 expression and alterations in PIK3CA, KRAS, MET, and EGFR (Supplementary Fig. S6). One patient had pretreatment heterogeneity with a HER2−, PD-L1 combined positive score (CPS) 100 retroperitoneal lymph node (Fig. 2A; Supplementary Fig. S7) which resolved on-treatment, while their esophageal primary tumor was HER2 3+, PD-L1 CPS <1. Upon progression, their metastases demonstrated MET IHC expression and concurrent MET ctDNA amplification. We also demonstrated that PIK3CD S367L, currently a variant of undetermined significance, increases phosphorylation of AKT, suggesting functional relevance (Supplementary Fig. S8). These findings indicate that the predominant mechanisms of resistance were either loss of HER2 expression or selection for resistant subclones, similar to previous findings with trastuzumab and chemotherapy (10).

Figure 2.

Intratumoral and intertumoral heterogeneity associated with resistance. A, Therapeutic timeline in a patient demonstrating pretreatment HER2 heterogeneity with a HER2+ primary tumor and HER2 but high PD-L1 CPS retroperitoneal lymph node (Supplementary Fig. S7). ERBB2 amplification was also identified in the primary tumor and ctDNA pretreatment. This patient initially responded to induction pembrolizumab and trastuzumab induction by ctDNA and radiographic measurements, but ctDNA was never cleared. Serial ctDNA identified a reappearance of ERBB2 amplification, as well as a new MET coamplification. Imaging identified new liver lesions that were later confirmed as metastases, as well as non-radiographically evident scalp lesions. Both the liver and scalp lesions were found to have strong MET expression by IHC upon progression. B, scRNA-seq analysis of 1,116 viable cells from an otherwise mostly necrotic on-treatment biopsy. Uniform manifold approximation and projection (UMAP) plots; color scale in inset indicates single tumor cell (n = 51) ERBB2 and FGFR3 RNA expression. Serial ctDNA assessment identified a pretreatment subclonal FGFR3-TACC3 fusion that expanded over time (see Supplementary Fig. S9).

Figure 2.

Intratumoral and intertumoral heterogeneity associated with resistance. A, Therapeutic timeline in a patient demonstrating pretreatment HER2 heterogeneity with a HER2+ primary tumor and HER2 but high PD-L1 CPS retroperitoneal lymph node (Supplementary Fig. S7). ERBB2 amplification was also identified in the primary tumor and ctDNA pretreatment. This patient initially responded to induction pembrolizumab and trastuzumab induction by ctDNA and radiographic measurements, but ctDNA was never cleared. Serial ctDNA identified a reappearance of ERBB2 amplification, as well as a new MET coamplification. Imaging identified new liver lesions that were later confirmed as metastases, as well as non-radiographically evident scalp lesions. Both the liver and scalp lesions were found to have strong MET expression by IHC upon progression. B, scRNA-seq analysis of 1,116 viable cells from an otherwise mostly necrotic on-treatment biopsy. Uniform manifold approximation and projection (UMAP) plots; color scale in inset indicates single tumor cell (n = 51) ERBB2 and FGFR3 RNA expression. Serial ctDNA assessment identified a pretreatment subclonal FGFR3-TACC3 fusion that expanded over time (see Supplementary Fig. S9).

Close modal

scRNA-seq of on-treatment biopsy reveals putative mechanisms of resistance

To explore the contribution of intratumor heterogeneity to pre-existing subclonal resistance, we performed scRNA-seq on tumor tissue collected from a metastatic EGC patient treated with chemotherapy and trastuzumab ± pembrolizumab (Fig. 2B). At the single-cell level, we identified extensive ERBB2 expression heterogeneity and co-occuring high expression of FGFR3, a putative biomarker of trastuzumab resistance, in a subpopulation of highly ERBB2-expressing cells. Furthermore, pretreatment plasma NGS identified a subclonal FGFR3-TACC3 fusion event (not detected by targeted sequencing of a pretreatment tumor biopsy) that supports scRNA-seq findings and also identified EGFR, ERBB2, and PIK3CA amplifications (Fig. 2B; Supplementary Fig. S9).

scRNA-seq identifies transcriptional resistance program

We postulated that durable clinical benefit is inversely proportional to intratumor clonal diversity and evaluated on-treatment clonal selection using recently published paired scRNA-seq data collected before cycles 1 and 3 from patients who received CAPOX with (TC; n = 3) or without trastuzumab (C; n = 4; ref. 9). Using inferCNV (Supplementary Methods), we reconstructed the clonal composition of the pre- and on-treatment tumor biopsies (Supplementary Fig. S10A) and were successfully able to demonstrate rapid clearance of most highly ERBB2 expressing clones, which was consistent with our prior ctDNA findings. However, we also identified many clones that expanded despite treatment, suggesting resistance to both trastuzumab and chemotherapy (Supplementary Fig. S10B). To identify a trastuzumab resistance program, gene sets associated with clonal expansion versus contraction were devised using GeneVector on pretreatment expression from patients who received either TC or C (Supplementary Methods). Therefore, the equation [(CAPOX+T_ExpandedT0 − CAPOX+T_ContractedT0) − (CAPOX_ExpandedT0 − CAPOX_ContractedT0)], allowed us to identify that MT1H, MT1E, MT2A, and MSMB expression were associated with CAPOX+trastuzumab resistance (Supplementary Fig. S10C and S10D). Though exploratory and requiring orthogonal validation, these genes may serve as future therapeutic targets in overcoming resistance.

Identifying additional prognostic tumor features

The combined PD-1 inhibitor with trastuzumab and chemotherapy cohort (n = 66) was evaluated in conjunction with patients who received chemotherapy and HER2 inhibition without PD-1 inhibitor (n = 151). Another 9 patients were included, where able, whose concurrent PD-1 inhibitor receipt was unknown. We sought to identify additional prognostic features including sites of metastatic involvement and molecular alterations (Supplementary Tables S5 and S6; Supplementary Data S4), in patients with EGC treated with trastuzumab and chemotherapy with or without anti-PD-1 therapy. Univariate analysis identified that patients with fewer disease sites involved and with ERBB2 amplification had superior PFS, while those with oncogenic alterations in KRAS, MYC, CCND1, and CDKN2A/B had inferior PFS (Supplementary Table S7). Though no specific disease sites were associated with prognosis, the multivariable model identified ERBB2 amplification as a positive prognostic feature, and alterations in MYC, CDKN2A/B as negative prognostic features for PFS (Supplementary Table S8).

These results demonstrate that pretreatment tumor heterogeneity, in particular HER2 expression heterogeneity, is associated with shorter PFS among patients with HER2+ EGC treated with trastuzumab, pembrolizumab, and chemotherapy. Since our initial clinical publication (3), INTEGA and PANTHERA similarly demonstrated PD-L1 agnostic improved response and survival with the addition of immune checkpoint blockade to chemotherapy and trastuzumab, albeit with shorter follow-up (4, 5). Although combined PD-1 and HER2 blockade augments trastuzumab efficacy across all 3 studies, our long-term follow-up confirms that preexistent lesion-to-lesion and even intratumoral HER2 heterogeneity remains a major barrier to HER2-targeted therapy efficacy and a negative predictor of survival (12, 13).

While biopsy of multiple disease sites to assess for HER2 and other molecular heterogeneity is not feasible in clinical practice, we found that noninvasive ctDNA analysis and 89Zr-trastuzumab molecular imaging can identify patients most likely to have durable benefit on first-line pembrolizumab and trastuzumab therapy. However, these modalities have limitations and require relatively high disease burden for accurate assessment.

We found that declining tumor-matched ctDNA maxVAF at 3 weeks, after a single dose of pembrolizumab and trastuzumab, predicted 6-month PFS, which demonstrates ctDNA's ability to predict treatment response in EGC, mirroring results in other cancers (14–17). While uniformity of 3-week CT response predicted ≥ 6-month PFS, changes in individual lesions were often too small to reliably predict future response. However, several patients with progressive lesions after induction therapy responded to the addition of chemotherapy, highlighting the role for combination approaches in overcoming heterogeneity-driven drug resistance. This may explain why previous studies of targeted monotherapies failed to associate early ctDNA decline with improved PFS (18). The observation that ctDNA is an early prognostic biomarker of treatment response may inform future targeted and immunotherapy trials in which therapy escalation or alternate molecularly guided agents are used as salvage in patients with mixed responses resulting from tumor heterogeneity.

Upon progression, IHC and DNA sequencing of tumor and/or ctDNA identified low HER2 expression or lack of ERBB2 amplification in approximately half of tested patients. This was higher than in prior studies, although limited by sample size (19, 20). In patients with persistent HER2 expression, escape lesions had co-occurring alterations in KRAS, MET, EGFR, or PI3K-associated genes or were in reservoir sites with poor trastuzumab penetration, such as the central nervous system. scRNA-seq in conjunction with serial ctDNA sequencing illustrated de novo pretreatment subclonal coexpression of a FGFR3-TACC3 fusion and ERBB2, reiterating the correlation between genomic heterogeneity and therapeutic resistance, and the role of ctDNA testing in identifying tumor heterogeneity.

Furthermore, scRNA-seq suggested that MT1H, MT1E, MT2A, and MSMB expression were associated with trastuzumab resistance. MT1H, MT1E, and MT2A encode isoforms of metallothioneins, which are found in all tissues and are associated with trace element homeostasis and protection against oxidative stress. While MT2A has been associated with chemotherapy resistance (21, 22), MT1G expression contributes to P53 activation and improved oxaliplatin cytotoxicity, though this may not be relevant in HER2-expressing EGC as nearly all of these patients have TP53 mutations (23). Similarly, MSMB encodes microseminoprotein-beta. Microseminoprotein expression is stimulated by hypoxia, which leads to MAPK pathway activation and has been associated with vascular endothelial growth factor inhibitor resistance in ovarian cancer models (24). Further preclinical validation of metallothionein inhibition in overcoming chemotherapy resistance is merited.

Multivariable analysis of our larger institutional cohort of HER2-targeted patients did not reveal a difference in PFS with or without concurrent immune checkpoint blockade despite impressive results with adding PD-1 inhibitors in multiple trials. Survival outcomes from the phase 3 KEYNOTE-811 trial are expected shortly. Pre-clinically, combining trastuzumab and PD-1 blockade augments HER2-specific T-cell response, promotes T-cell and dendritic cell trafficking, and induces peripheral memory T-cell expansion (25–29). Further clinical validation of these mechanisms is needed. While we identified oncogenic MYC and CDKN2A/B alterations as prognostic of inferior PFS, PANTHERA suggested that pretreatment RTK/RAS pathway alterations were associated with improved PFS. This is contrary to our prior findings (3, 10) and likely reflects their liberal definition of oncogenic RTK-RAS alterations (61% vs. 28% of patients), and would benefit from validation in a larger cohort. Both studies identified that ERBB2 amplification was associated with improved PFS.

Promising initial phase III KEYNOTE-811 study findings suggest that the combination of pembrolizumab and trastuzumab is a transformative treatment for patients with EGC, and mechanistic evaluation of this combination remains ongoing. Here, we identified prognostic pre- and early-treatment biomarkers of durable response. We also confirmed that tumor heterogeneity remains the greatest barrier to precision oncology in EGC, and that preexistent molecular heterogeneity can be identified noninvasively through molecular imaging and ctDNA analysis. Future trials should assess whether noninvasive assessment of mixed response and molecular heterogeneity can dynamically inform adaptive treatment strategies prior to disease progression.

S.B. Maron reports grants from Society of MSK, Cycle for Survival, NIH, and non-financial support from Guardant Health during the conduct of the study, as well as personal fees from Natera, Bayer, Basilea, Daiichi Sankyo, Bicara, Novartis, Amgen, Elevation Oncology, and Purple Oncology outside the submitted work; S.B. Maron also previously owned stock in Calithera. N. Pandit-Taskar reports other support from Bayer, Fusion Pharma, Clarity Pharma, and Janssen, as well as personal fees and other support from Imaginab, Illumina-innervate, and Actinium Pharma outside the submitted work. J.S. Lewis reports grants from NIH during the conduct of the study. T. Biachi De Castria reports personal fees from Merck Sharp & Dohme Corp, Bristol Myers Squibb, Eli Lilly, AstraZeneca, and A2Bio during the conduct of the study. R.J. Nagy reports other support from Guardant Health outside the submitted work. G.Y. Ku reports grants and personal fees from AstraZeneca, Daiichi Sankyo, Merck, Pieris, and Zymeworks during the conduct of the study, as well as grants from Adaptimmune, CARsgen, and Oncolys outside the submitted work. T. Merghoub reports personal fees from Daichii Sankyo and Normunity during the conduct of the study, as well as grants from Bristol Myers Squibb, Surface Oncology, Kyn Therapeutics, Infinity Pharmaceuticals, Peregrine Pharmaceuticals, Adaptive Biotechnologies, Leap Therapeutics, and Aprea. T. Merghoub is a consultant for Daiichi Sankyo, Leap Therapeutics, Immunos Therapeutics, and Pfizer, and is co-founder of Imvaq Therapeutics with equity. In addition, T. Merghoub is an inventor on patent applications related to work on oncolytic viral therapy, alpha virus-based vaccine, neoantigen modeling, CD40, GITR, OX40, PD-1, and CTLA-4. S. Shah reports other support from Canexia Health Inc, as well as personal fees from AstraZeneca Inc outside the submitted work. D.B. Solit reports personal fees from Pfizer, Scorpion Therapeutics, FORE Therapeutics, Function Oncology, Fog Pharma, Elsie Biotechnologies, Rain Oncology, and BridgeBio outside the submitted work. Y.Y. Janjigian reports personal fees from Merck Serono, Micheal J. Hennessy Associates, Paradigm Medical Communications, PeerView Institute, Pfizer, Research to Practice, Geneos Therapeutics, GlaxoSmithKline, Imedex, Imugene, Lynx Health, Daiichi Sankyo, AbbVie, Amerisource Bergen, Ask-Gene Pharma, Inc., Arcus Biosciences, AstraZeneca, and Basilea Pharmaceutica; personal fees and other support from Rgenix, Seagen, Silverback Therapeutics, Zymeworks Inc., Merck, Eli Lilly, Bayer, and Bristol Myers Squibb; and other support from Cycle for Survival, Department of Defense, Fred's Team, Genentech/Roche, and NCI outside the submitted work. Y.Y. Janjigian also reports stock options from Rgenix. No disclosures were reported by the other authors.

S.B. Maron: Conceptualization, formal analysis, supervision, validation, investigation, visualization, methodology, writing–original draft, project administration, writing–review and editing. W. Chatila: Formal analysis, visualization, methodology. H. Walch: Formal analysis, visualization, methodology. J.F. Chou: Conceptualization, formal analysis, writing–original draft. N. Ceglia: Investigation, visualization, methodology. R. Ptashkin: Resources, data curation, methodology. R.K.G. Do: Data curation. V. Paroder: Data curation. N. Pandit-Taskar: Investigation, methodology, writing–review and editing. J.S. Lewis: Conceptualization, resources. T. Biachi De Castria: Data curation, validation. S. Sabwa: Data curation. F. Socolow: Data curation. L. Feder: Data curation, project administration. J. Thomas: Investigation. I. Schulze: Investigation. K. Kim: Investigation, methodology. A. Elzein: Investigation. V. Bojilova: Investigation, visualization. M. Zatzman: Validation, investigation. U. Bhanot: Supervision, investigation. R.J. Nagy: Resources, funding acquisition. J. Lee: Data curation, validation. M. Simmons: Data curation, supervision. M. Segal: Project administration. G.Y. Ku: Investigation, writing–review and editing. D.H. Ilson: Supervision, writing–review and editing. M. Capanu: Formal analysis, writing–original draft. J.F. Hechtman: Conceptualization, data curation, supervision, investigation. T. Merghoub: Conceptualization, writing–review and editing. S. Shah: Conceptualization, resources. N. Schultz: Conceptualization, resources, formal analysis, visualization, methodology, writing–review and editing. D.B. Solit: Conceptualization, validation, investigation, visualization, writing–original draft, writing–review and editing. Y.Y. Janjigian: Conceptualization, formal analysis, supervision, validation, investigation, writing–original draft, writing–review and editing.

The original trial was funded by Merck & Co, which provided pembrolizumab (ClinicalTrials.gov trial registration ID: NCT01522768). We acknowledge the use of the Integrated Genomics Operation Core (iGO) for sequencing and the Radiochemistry and Molecular Imaging Probe Core for the production of 89Zr-trastuzumab, both funded by the NCI Cancer Center Support Grant (CCSG, P30 CA08748). iGO is also supported by Cycle for Survival, and the Marie-Josée and Henry R. Kravis Center for Molecular Oncology. JSL was supported in part by NCI R35 CA232130. Other funding sources include the Robertson Foundation (grant to NS), the Department of Defense Congressionally Directed Medical Research Program (CA 150646 to YYJ), the MSK Paul Calabresi Career Development Award for Clinical Oncology (K12 CA184746 to SBM), Cycle for Survival (to YYJ) and the Society of Memorial Sloan Kettering (to SBM).

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 Clinical Cancer Research Online (http://clincancerres.aacrjournals.org/).

1.
Bang
Y-J
,
Van Cutsem
E
,
Feyereislova
A
,
Chung
HC
,
Shen
L
,
Sawaki
A
, et al
.
Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer (ToGA): a phase 3, open-label, randomised controlled trial
.
Lancet
2010
;
376
:
687
97
.
2.
Janjigian
YY
,
Kawazoe
A
,
Yanez
PE
,
Luo
S
,
Lonardi
S
,
Kolesnik
O
, et al
.
Pembrolizumab plus trastuzumab and chemotherapy for HER2+ metastatic gastric or gastroesophageal junction (G/GEJ) cancer: initial findings of the global phase 3 KEYNOTE-811 study
.
JCO
2021
;
39
:
4013
.
3.
Janjigian
YY
,
Maron
SB
,
Chatila
WK
,
Millang
B
,
Chavan
SS
,
Alterman
C
, et al
.
First-line pembrolizumab and trastuzumab in HER2-positive oesophageal, gastric, or gastro-oesophageal junction cancer: an open-label, single-arm, phase 2 trial
.
Lancet Oncol
2020
;
21
:
821
31
.
4.
Stein
A
,
Paschold
L
,
Tintelnot
J
,
Goekkurt
E
,
Henkes
S-S
,
Simnica
D
, et al
.
Efficacy of ipilimumab vs FOLFOX in combination with nivolumab and trastuzumab in patients with previously untreated ERBB2-positive esophagogastric adenocarcinoma: the AIO INTEGA randomized clinical trial
.
JAMA Oncol
2022
;
8
:
1150
8
.
5.
Lee
C-K
,
Rha
SY
,
Kim
HS
,
Jung
M
,
Kang
B
,
Che
J
, et al
.
A single arm phase Ib/II trial of first-line pembrolizumab, trastuzumab and chemotherapy for advanced HER2-positive gastric cancer
.
Nat Commun
2022
;
13
:
6002
.
6.
Bartley
AN
,
Washington
MK
,
Colasacco
C
,
Ventura
CB
,
Ismaila
N
,
Benson
AB
, et al
.
HER2 testing and clinical decision making in gastroesophageal adenocarcinoma: guideline from the College of American Pathologists, American Society for Clinical Pathology, and the American Society of Clinical Oncology
.
J Clin Oncol
2017
;
35
:
446
64
.
7.
O'Donoghue
JA
,
Lewis
JS
,
Pandit-Taskar
N
,
Fleming
SE
,
Schöder
H
,
Larson
SM
, et al
.
Pharmacokinetics, biodistribution, and radiation dosimetry for 89Zr-trastuzumab in patients with esophagogastric cancer
.
J Nucl Med
2018
;
59
:
161
6
.
8.
Hofman
MS
,
Hicks
RJ
.
How we read oncologic FDG PET/CT
.
Cancer Imaging
2016
;
16
:
35
.
9.
Kim
R
,
An
M
,
Lee
H
,
Mehta
A
,
Heo
YJ
,
Kim
K-M
, et al
.
Early tumor-immune microenvironmental remodeling and response to first-line fluoropyrimidine and platinum chemotherapy in advanced gastric cancer
.
Cancer Discov
2022
;
12
:
984
1001
.
10.
Janjigian
YY
,
Sanchez-Vega
F
,
Jonsson
P
,
Chatila
WK
,
Hechtman
JF
,
Ku
GY
, et al
.
Genetic predictors of response to systemic therapy in esophagogastric cancer
.
Cancer Discov
2018
;
8
:
49
58
.
11.
Maron
SB
,
Chase
LM
,
Lomnicki
S
,
Kochanny
S
,
Moore
KL
,
Joshi
SS
, et al
.
Circulating tumor DNA sequencing analysis of gastroesophageal adenocarcinoma
.
Clin Cancer Res
2019
;
25
:
7098
112
.
12.
Haffner
I
,
Schierle
K
,
Raimúndez
E
,
Geier
B
,
Maier
D
,
Hasenauer
J
, et al
.
HER2 expression, test deviations, and their impact on survival in metastatic gastric cancer: results from the prospective multicenter VARIANZ study
.
J Clin Oncol
2021
;
39
:
1468
78
.
13.
Filho
OM
,
Viale
G
,
Stein
S
,
Trippa
L
,
Yardley
DA
,
Mayer
IA
, et al
.
Impact of HER2 heterogeneity on treatment response of early-stage HER2-positive breast cancer: phase II neoadjuvant clinical trial of T-DM1 combined with pertuzumab
.
Cancer Discov
2021
;
11
:
2474
87
.
14.
Murciano-Goroff
YR
,
Hui
AB
,
Chabon
JJ
,
Moding
EJ
,
Lebow
ES
,
Araujo-Filho
JA
, et al
.
Early ctDNA response assessment for prediction of platinum sensitivity in small cell lung cancer
.
JCO
2020
;
38
:
9067
.
15.
Goldberg
SB
,
Narayan
A
,
Kole
AJ
,
Decker
RH
,
Teysir
J
,
Carriero
NJ
, et al
.
Early assessment of lung cancer immunotherapy response via circulating tumor DNA
.
Clin Cancer Res
2018
;
24
:
1872
80
.
16.
Tie
J
,
Kinde
I
,
Wang
Y
,
Wong
HL
,
Roebert
J
,
Christie
M
, et al
.
Circulating tumor DNA as an early marker of therapeutic response in patients with metastatic colorectal cancer
.
Ann Oncol
2015
;
26
:
1715
22
.
17.
Iijima
Y
,
Hirotsu
Y
,
Amemiya
K
,
Ooka
Y
,
Mochizuki
H
,
Oyama
T
, et al
.
Very early response of circulating tumour-derived DNA in plasma predicts efficacy of nivolumab treatment in patients with non-small cell lung cancer
.
Eur J Cancer
2017
;
86
:
349
57
.
18.
Hyman
DM
,
Smyth
LM
,
Donoghue
MTA
,
Westin
SN
,
Bedard
PL
,
Dean
EJ
, et al
.
AKT inhibition in solid tumors with AKT1 mutations
.
J Clin Oncol
2017
;
35
:
2251
9
.
19.
Seo
S
,
Ryu
M-H
,
Park
YS
,
Ahn
JY
,
Park
Y
,
Park
SR
, et al
.
Loss of HER2 positivity after anti-HER2 chemotherapy in HER2-positive gastric cancer patients: results of the GASTric cancer HER2 reassessment study 3 (GASTHER3)
.
Gastric Cancer
2019
;
22
:
527
35
.
20.
Wang
D-S
,
Liu
Z-X
,
Lu
Y-X
,
Bao
H
,
Wu
X
,
Zeng
Z-L
, et al
.
Liquid biopsies to track trastuzumab resistance in metastatic HER2-positive gastric cancer
.
Gut
2019
;
68
:
1152
61
.
21.
Mangelinck
A
,
da Costa
MEM
,
Stefanovska
B
,
Bawa
O
,
Polrot
M
,
Gaspar
N
, et al
.
MT2A is an early predictive biomarker of response to chemotherapy and a potential therapeutic target in osteosarcoma
.
Sci Rep
2019
;
9
:
12301
.
22.
Yap
TA
,
Gerlinger
M
,
Futreal
PA
,
Pusztai
L
,
Swanton
C
.
Intratumor heterogeneity: seeing the wood for the trees
.
Sci Transl Med
2012
;
4
:
127ps10
.
23.
Arriaga
JM
,
Greco
A
,
Mordoh
J
,
Bianchini
M
.
Metallothionein 1G and zinc sensitize human colorectal cancer cells to chemotherapy
.
Mol Cancer Ther
2014
;
13
:
1369
81
.
24.
Mitamura
T
,
Pradeep
S
,
McGuire
M
,
Wu
SY
,
Ma
S
,
Hatakeyama
H
, et al
.
Induction of anti-VEGF therapy resistance by upregulated expression of microseminoprotein (MSMP)
.
Oncogene
2018
;
37
:
722
31
.
25.
Gall
VA
,
Philips
AV
,
Qiao
N
,
Clise-Dwyer
K
,
Perakis
AA
,
Zhang
M
, et al
.
Trastuzumab increases HER2 uptake and cross-presentation by dendritic cells
.
Cancer Res
2017
;
77
:
5374
83
.
26.
Mortenson
ED
,
Park
S
,
Jiang
Z
,
Wang
S
,
Fu
Y-X
.
Effective anti-neu-initiated antitumor responses require the complex role of CD4+ T cells
.
Clin Cancer Res
2013
;
19
:
1476
86
.
27.
Park
S
,
Jiang
Z
,
Mortenson
ED
,
Deng
L
,
Radkevich-Brown
O
,
Yang
X
, et al
.
The therapeutic effect of anti-HER2/neu antibody depends on both innate and adaptive immunity
.
Cancer Cell
2010
;
18
:
160
70
.
28.
Taylor
C
,
Hershman
D
,
Shah
N
,
Suciu-Foca
N
,
Petrylak
DP
,
Taub
R
, et al
.
Augmented HER-2 specific immunity during treatment with trastuzumab and chemotherapy
.
Clin Cancer Res
2007
;
13
:
5133
43
.
29.
zum Büschenfelde
CM
,
Hermann
C
,
Schmidt
B
,
Peschel
C
,
Bernhard
H
.
Antihuman epidermal growth factor receptor 2 (HER2) monoclonal antibody trastuzumab enhances cytolytic activity of class I-restricted HER2-specific T lymphocytes against HER2-overexpressing tumor cells
.
Cancer Res
2002
;
62
:
2244
7
.
This open access article is distributed under the Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0) license.

Supplementary data