Next Article in Journal
Emerging Role of IGF-1 in Prostate Cancer: A Promising Biomarker and Therapeutic Target
Next Article in Special Issue
Chemo-Radio-Immunotherapy for NSCLC III: ESR/ATS Thresholds for DLCO Correlate with Radiation Dosimetry and Pneumonitis Rate
Previous Article in Journal
Endoscopic Ultrasound Advanced Techniques for Diagnosis of Gastrointestinal Stromal Tumours
Previous Article in Special Issue
Labyrinthin Expression Is Associated with Poor Prognosis in Patients with Non-Small-Cell Lung Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Evolving Treatment Landscape of HER2-mutant Non-Small Cell Lung Cancer: Trastuzumab Deruxtecan and Beyond

by
Ioannis A. Vathiotis
1,
Dimitrios Bafaloukos
2,
Konstantinos N. Syrigos
1 and
George Samonis
2,*
1
Third Department of Internal Medicine, Sotiria General Hospital for Chest Diseases, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
2
First Oncology Department, Metropolitan Hospital, 18547 Athens, Greece
*
Author to whom correspondence should be addressed.
Cancers 2023, 15(4), 1286; https://doi.org/10.3390/cancers15041286
Submission received: 19 January 2023 / Revised: 14 February 2023 / Accepted: 15 February 2023 / Published: 17 February 2023
(This article belongs to the Special Issue World Lung Cancer Awareness Month)

Abstract

:

Simple Summary

By coupling the potency of cytotoxic chemotherapy with the selectivity of targeted therapy, antibody–drug conjugates represent a unique and rapidly growing class of antitumor agents. In this review, we aim to outline the unanswered questions that have emerged after the approval of trastuzumab deruxtecan in HER2-mutant non-small cell lung cancer. We also summarize data on novel therapies that are currently being investigated in the same patient population.

Abstract

Successful targeting of HER2-activating mutations in DESTINY-Lung02 phase II study has led to the approval of the antibody–drug conjugate (ADC) trastuzumab deruxtecan (T-DXd) as second-line treatment in patients with non-small cell lung cancer (NSCLC). Despite the impressive results, several matters need to be addressed, including the clinical activity of T-DXd in patients with disease in the central nervous system as well as the role of T-DXd in the context of HER2 overexpression. Additionally, data regarding novel agents used to target HER2 continue to accumulate. This review highlights the challenges and unanswered questions that have emerged after the approval of T-DXd in patients with HER2-mutant NSCLC.
Keywords:
NSCLC; HER2; trastuzumab

1. Introduction

On 11 August 2022, the Food and Drug Administration (FDA) gave accelerated approval to trastuzumab deruxtecan (T-DXd) for adults with previously treated, unresectable or metastatic non-small cell lung cancer (NSCLC) harboring activating mutations in the gene encoding human epidermal growth factor receptor 2 (HER2), based on an interim efficacy analysis of DESTINY-Lung02 phase II clinical trial [1]. In the latter, a total of 80 patients with HER2-mutant NSCLC who had received at least one prior line of treatment, mostly platinum-based chemotherapy, were randomized in a 2:1 ratio to receive T-DXd at a dose of 5.4 mg/kg or 6.4 mg/kg, respectively. Patients receiving T-DXd at the 5.4 mg/kg dose level demonstrated a numerically superior objective response rate (ORR; 57.7%), confirmed by a blinded independent central review (BICR), along with a more favorable toxicity profile; any-grade adjudicated drug-related interstitial lung disease (ILD) was seen in 5.9% and 14.0% of patients receiving T-DXd at 5.4 mg/kg and 6.4 mg/kg, respectively.
T-DXd is an antibody–drug conjugate (ADC) consisting of a humanized monoclonal IgG1 antibody against HER2 (trastuzumab), linked to a topoisomerase I inhibitor (deruxtecan) payload through a cleavable tetrapeptide-based linker [2]. Compared to other drugs of the same class, this system incorporates a high drug-to-antibody ratio (DAR; 8 molecules of topoisomerase inhibitor per antibody), with high stability in plasma, exhibiting predictable pharmacokinetics.
Since the first approval of a targeted therapy in patients with NSCLC (erlotinib in epidermal growth factor receptor [EGFR]-positive disease) in 2013, molecular testing has evolved into an essential component of patient care with the National Comprehensive Cancer Network currently advising for broad molecular profiling to assess nine predictive biomarkers, at least, at baseline [3]. Among these, activating HER2 mutations are identified in 1–3% and 1.4–6.7% of NSCLC patients of European/American and Asian descent, respectively; in-frame insertion mutations occurring in exon 20 represent the most common subtype, seen in approximately 48% of the cases [4,5,6,7]. In the vast majority, de novo HER2 mutations are mutually exclusive with other driver alterations. They are most commonly present in females and never-smokers with pure adenocarcinoma histology or an adenocarcinoma component. Interestingly, NSCLC patients with HER2 mutations tend to exhibit a more aggressive disease phenotype with increased incidence of brain metastases during treatment; this association is significantly higher in individuals harboring the exon 20 A775_G776insYVMA insertion compared to non-YVMA cases [8,9]. Activating HER2 mutations have been shown to enhance receptor internalization in preclinical models, providing a potential explanation for the observed clinical efficacy of T-DXd regardless of the quantity of HER2 protein expression [10,11]. Previous studies have also confirmed the clinical efficacy of T-DXd across HER2 mutation subtypes [12].
In the subsequent sections, we will review the evidence on current challenges and unanswered questions regarding the implementation of T-DXd in the treatment algorithm of HER2-mutant NSCLC. Furthermore, we will summarize available data on novel strategies to target HER2 in patients with NSCLC.

2. Central Nervous System Involvement

Patients with HER2-driven tumors have a propensity to develop central nervous system (CNS) disease, which bears a negative impact on overall survival (OS) as well as quality of life [13]. Accumulating data support the clinical efficacy of T-DXd in breast cancer patients with CNS involvement, which may provide an opportunity for transition to tumor types with fewer data on the intracranial efficacy of T-DXd, including NSCLC. In the phase III DESTINY-Breast03 clinical trial, T-DXd conferred a significant improvement in progression-free survival (PFS), ORR and intracranial response rate compared with T-DM1 in 82 patients with HER2-positive breast cancer and stable brain metastases at baseline; the overall intracranial response rate was 63.8% for T-DXd compared with 33.3% for T-DM1 with the percentage of patients who achieved a complete intracranial response being 27.8% and 2.8%, respectively [14]. Notably, patients treated with T-DXd had a median PFS of 15 months. In addition, a recent subgroup analysis of DESTINY-Breast01 confirmed the durable clinical effect of T-DXd in patients with stable brain metastases [15].
The CNS activity of T-DXd has been proven in patient-derived xenograft models of HER2-positive as well as HER2-low breast cancer brain metastases [16]. The DEBBRAH phase II study assessed the efficacy of T-DXd in pretreated patients with either HER2-positive or HER2-low breast cancer and involvement of the CNS [17]. The intracranial ORR in patients with asymptomatic untreated brain metastases and those with brain metastases progressing after local therapy was 50% and 44.4%, respectively. In the TUXEDO-1 phase II study, treatment with T-DXd resulted in an intracranial response rate of 73.3% (by RANO-BM; 11/15 patients) and a median PFS of 14 months in patients with HER2-positive breast cancer and newly diagnosed or progressive brain metastases; the response was 100% in patients with de novo brain metastases and 66.7% in patients with brain metastases that had progressed after previous local therapy [18].

3. HER2-Amplified/Overexpressing Disease

Following the successful implementation of HER2-directed therapies in patients with breast cancer, strategies targeting HER2 have been investigated in other tumor types, including NSCLC [19]. Although the addition of trastuzumab did not confer survival benefit in patients with HER2-amplified/overexpressing NSCLC, promising results were seen with trastuzumab-emtansine (T-DM1), with responses correlating with the levels of HER2 protein expression by immunohistochemistry (IHC), but the latter was still regarded as a rather insufficient predictive biomarker [20,21,22]. In the case of T-DXd, the first signs of antitumor activity were documented by Tsurutani et al., in a mixed population of heavily pretreated HER2-expressing or HER2-mutant solid tumors [23]. Subsequently, the efficacy of T-DXd in HER2-overexpressing NSCLC was evaluated in a DESTINY-Lung01 phase II clinical trial [11]. In this study, a cohort of 49 patients with HER2 IHC 2+ (n = 39) or 3+ (n = 10) received T-DXd at 6.4 mg/kg after disease progression to prior anticancer therapy (median number of prior regimens was three). Confirmed ORR was 24.5%, including 25.6% in patients with IHC 2+ and 20.0% in patients with IHC 3+; the estimated median PFS reached 5.4 months. As a result, an additional cohort has opened to assess matters of safety and efficacy of T-DXd given at 5.4 mg/kg in the same population.
Based on the results of DESTINY-Breast04, the benefit from T-DXd administration has been extended to patients with HER2-low breast cancer, defined as IHC 2+ with negative results on in situ hybridization (ISH) or IHC 1+ [24]. Unlike other anti-HER2 agents, HER2 overexpression may not be required for the efficacy of T-DXd; as a matter of fact, T-DXd has demonstrated signs of antitumor activity across a full range of HER2 expression, essentially redefining HER2-positive disease [25]. Given the preanalytical as well as analytical factors that may explain, to some degree, patient misclassification when HER2 testing is performed by IHC, several novel quantitative assays with the potential to enhance the sensitivity of HER2 assessment have emerged [26,27,28]. As suggested by Tarantino et al., successful targeting of HER2 in nonamplified tumors has the potential to alter the current treatment landscape in several ways, including mitigation of oncogene dependency (as novel ADCs may exert their antitumor activity regardless of tumor cell dependency on the HER2 pathway), higher DAR/alternative payload mechanisms of action, and the use of modern cleavable linkers that have the potential to facilitate the “bystander effect”. Recently, the antitumor efficacy of T-DXd in HER2-low disease was also observed in gastric or gastroesophageal junction adenocarcinoma, with confirmed ORR equal to 26.3% and 9.5% in patients with IHC 2+/ISH-negative and IHC 1+ tumors, respectively [29].

4. Other ADCs

ADCs represent an innovative class of anticancer drugs that combine the selectivity of targeted therapies with the potency of cytotoxic chemotherapy. HER2 has long represented an attractive target for ADCs in NSCLC (Table 1) [30,31]. Composed of the anti-HER2 antibody trastuzumab linked to the microtubule inhibitor emtansine (DM1) via a non-cleavable linker, T-DM1 was the first ADC to be approved for the treatment of HER2-positive breast cancer and the first one to be evaluated in patients with NSCLC; T-DM1 has a DAR of 3.5 [30,32]. In a phase II basket trial, treatment with T-DM1 resulted in an ORR of 44.4%, meeting the primary study endpoint, and a median PFS of 5 months in heavily pretreated patients with HER2-mutant NSCLC [21]. Subsequently, Li et al. enrolled more patients to confirm the efficacy (ORR equal to 51%) and tolerable toxicity profile of T-DM1, with only one incidence of grade 3 or higher treatment-related adverse events (TRAEs) [10]. Lately, treatment with T-DM1 showed slightly inferior results in the Japanese population [33].
A166 is a novel ADC consisting of trastuzumab and monomethyl auristatin F derivative duostatin-5, linked with a cleavable linker. A166 exhibited clinical efficacy in 35 heavily pretreated patients with HER2 amplified/overexpressing advanced solid tumors [34]. The ORR of A166 was 36% with responses seen only at the 3.6 mg/kg and the 4.8 mg/kg dose level; ophthalmic toxicities of any grade, including keratitis, dry eye, blurred vision, etc., appeared in more than 80% of study participants at efficacious dose levels. In a second phase I study, A166 demonstrated a manageable toxicity profile with grade 3 or higher treatment-related adverse events (TRAEs) recorded in 18 out of 57 study participants (31.6%) [35]. Moreover, it showed high stability in plasma and promising antitumor activity. Among 36 HER2-positive breast cancer patients eligible for efficacy assessment, the best ORR was 59.1% (13/22) and 71.4% (10/14) at the 4.8 mg/kg and 6.0 mg/kg dose level, respectively; the median PFS was not reached at the time of data cutoff.
XMT-1522 is an ADC that consists of a human IgG1 anti-HER2 monoclonal antibody (HT-19) that binds to a distinct epitope of HER2, compared with trastuzumab, conjugated to auristatin F-hydroxypropylamide via a cleavable linker; this ADC has a DAR of 12. Preclinical evidence shows increased potency of XMT-1522 compared with T-DM1, as well as antitumor activity in the case of primary or acquired resistance to T-DM1 in vitro [36]. Preliminary results of a phase I study indicated that XMT-1522 was well tolerated up to the dose of 21.3 mg/m2 (all TRAEs were grade 1 and 2, most commonly elevated liver function tests and fatigue) and demonstrated early signs of antitumor activity [37].
ARX788 is a next-generation, site-specific ADC, composed of an anti-HER2 monoclonal antibody and the microtubulin inhibitor amberstatin (AS269; DAR, 1.9), that has already produced promising preliminary results in patients with HER2-positive metastatic breast cancer and is currently being evaluated in multiple solid tumors, including breast and gastric/gastroesophageal junction carcinoma (NCT03255070) [38,39]. Finally, NCT04235101 is investigating matters of safety for the combination of SYD985 (trastuzumab duocarmazine) with niraparib in patients with solid tumors.

5. New-Generation Tyrosine Kinase Inhibitors

Given the small size, flexible structure and covalent binding properties, novel tyrosine kinase inhibitors (TKIs; i.e., poziotinib) have demonstrated increased potency compared with the previous generation. Results from two phase II studies were highly concordant for poziotinib, indicating modest antitumor activity in previously treated patients with NSCLC and HER2 exon 20 insertion mutations, with an ORR of about 27% [40,41]. The median PFS was 5.5 months and the median overall survival (OS) reached 15 months. It should be noted that these response rates are well below compared with TKIs used in EGFR-driven NSCLC. The most common grade 3 or higher TRAE was rash, seen in nearly half of the patients, followed by diarrhea and stomatitis. Importantly, dose reductions were required in 76.7% of study participants, while treatment discontinuation occurred in 13.3%. On the basis of increased toxicity as well as lack of phase III data, the Food and Drug Administration (FDA) denied approval of poziotinib for this indication.
Showing activity against secondary C805S mutations, which represent the most common mechanism of acquired resistance to poziotinib, pyrotinib has also been evaluated in HER2-mutant NSCLC. In a phase II study, pyrotinib exhibited more promising antitumor activity (ORR was 30% with efficacy seen across HER2 mutation subtypes, median PFS was 6.9 months and median OS was 14.4 months), along with a rather acceptable toxicity profile (grade 3 or higher TRAEs were documented in 28.3% of patients, with the most common being diarrhea) [42].
Further attempts to target exon 20 insertions have included mobocertinib (TAK-788), a novel EGFR/HER2 TKI with high selectivity (low HER2 ex20ins IC50/wildtype EGFR IC50 ratio), increased efficacy against the G776 > VC subtype, and potential for synergy with T-DM1, with the latter being largely driven by M1 macrophage infiltration and CD4+ T cell activation [43]. Mobocertinib has already been FDA-approved in patients with NSCLC harboring EGFR exon 20 insertions (NCT02716116), resulting in an expansion cohort of patients with HER2 exon 20 insertions [44,45].
The high homology among ERBB family members has made it problematic to target HER2 while sparing other tyrosine kinases. Lack of selectivity has, thus, resulted in decreased potency and dose-limiting toxicity, hampering most efforts to target this disease in the clinic [46]. To circumvent the on-target toxicity caused by wild-type EGFR inhibition, tarloxitinib is a prodrug that can be converted to its active form, tarloxitinib-E, in the hypoxic tumor microenvironment [47]. Tarloxitinib-E has been shown to directly inhibit the phosphorylation and activation of EGFR, HER2 and HER2/HER3 heterodimers in vitro, resulting in tumor regression in vivo. Furthermore, tarloxitinib displays an IC50 about 180 times higher for wild type HER2 than the active metabolite, demonstrating a wide therapeutic index [48]. Importantly, secondary C805S HER2 mutations, as well as HER3 overexpression, have been identified as mechanisms of acquired resistance to tarloxotinib-E. In cohort B of the RAIN-701 phase I study (NCT03805841), tarloxitinib-E showed antitumor activity with an ORR of 22% and acceptable toxicity profile; the most common grade 3 toxicity was QTc interval prolongation, which appeared in 34.8% of patients [49].
Further attempts to address this particular issue led by Neumüller and colleagues have resulted in the development of selective HER2 inhibitors with a large therapeutic window (>50-fold for HER2 inhibition compared to wild-type EGFR); the presence of a hydrogen bond to serine 783 in the back pocket of HER2, which is not formed with the corresponding cysteine 775 in EGFR, establishes high selectivity [50]. Interestingly, selective HER2 YVMA inhibition was shown, which suffices to suppress tumor growth, as inhibition of wild-type EGFR did not appear to confer a meaningful therapeutic effect. JBJ-08–178-01 is a novel TKI effective against HER2-activating mutations as well as amplification both in vitro and in vivo [51]. It exhibits increased selectivity in comparison with other TKIs, inhibiting HER2 over wild-type EGFR; moreover, it has been shown to reduce the levels of HER2 by inducing proteasomal degradation of the receptor. Finally, BI 1810631 is a HER2-selective TKI that engages both wild-type and mutated HER2 receptors but spares EGFR signaling and is currently being evaluated in the context of a phase I study (NCT04886804) [52].

6. Bispecific Antibodies

Bispecific antibodies (bsAbs) are molecules designed to recognize two different epitopes or antigens [53]. Since the first mention of the concept in 1960, dual targeting enabled by bsAbs has sparked research interest leading to novel features and functionalities that cannot be accomplished with a simple mixture of the reference clones; physical linkage of two binding sites may in fact create temporal or spatial dependency [54]. As a result, the bsAb landscape is rapidly expanding, providing opportunities for enhanced epitope targeting and drug development.
As such, zenocutuzumab (MCLA-128) is a humanized IgG1 bsAb that is designed to target the extracellular domains of HER2 and HER3 using a unique “dock and block” mechanism [55]. The HER2-targeting arm binds to HER2 on the cell surface and in turn sets the HER3-targeting arm in place to prevent neuregulin 1 (NRG1) binding to HER3; the latter has been shown to induce a conformational change that is required for HER2:HER3 heterodimerization and downstream signaling. In addition, IgG1 glycosylation results in enhanced antibody-dependent cellular cytotoxicity (ADCC). Treatment with zenocutuzumab has been shown to decrease phosphorylation of HER2, HER3, EGFR, and HER4 and reduce NRG1 fusion-dependent oncogenic signaling, thereby suppressing tumor growth in preclinical models [56]. Moreover, zenocutuzumab has demonstrated promising antitumor activity leading to durable responses in heavily pretreated patients with NRG1 fusion-positive disease and a tolerable toxicity profile [57]. Based on these, it is currently being investigated in the context of a phase I/II study (NCT02912949).

7. Immune Checkpoint Inhibitors

Checkpoint inhibitor-based immunotherapy targeting the programmed cell death protein 1 (PD-1)/programmed death-ligand 1 (PD-L1) axis has acquired a central role in the management of patients with locally advanced or metastatic driver-negative NSCLC. However, the presence of driver alterations has oftentimes been associated with a poor response to immunotherapy. In particular, EGFR-mutant tumors harbor a low tumor mutational burden (TMB) and a “cold” tumor microenvironment, characterized by restricted T-cell clonality, and these features appear culpable to lack of benefit from PD-(L)1 inhibitors [58,59,60,61,62]. Moreover, the predictive value of PD-L1 expression in epidermal growth factor receptor (EGFR)-positive disease has not been established yet; preclinical data have suggested that PD-L1 expression may reflect EGFR signaling rather than T-cell activity. However, this may not be the case for every single actionable alteration in NSCLC [63].
The presence of activating HER2 mutations has been associated with low prevalence of PD-L1 positivity (47.6%) as well as low TMB (median <3 mut/Mb) [63]. Although multiple studies have investigated the role of PD-(L)1 inhibition in patients with NSCLC and activating HER2 mutations, current evidence remains retrospective in nature (Table 2). Lai et al. documented an ORR of 11.5% and median PFS of 1.9 months among 26 such patients treated with single-agent PD-(L)1 inhibitors [64]. Similar results were obtained by the IMMUNOTARGET registry, where 29 patients that received anti-PD-(L)1 therapy beyond the first line of treatment demonstrated an ORR of 7.4% and median PFS of 2.5 months [65]. Although the French Lung Cancer Group (GFPC) noted objective responses in six out of 23 (27.3%) patients with HER2-mutant tumors treated with PD-1/PD-L1 axis inhibitors, the median PFS did not exceed 2.2 months [66]. Negrao et al. reported on two different cohorts of unselected NSCLC patients with actionable alterations including activating HER2 mutations (MD Anderson Cancer Center [MDACC], n = 15; Flatiron Health-Foundation Medicine Clinico-Genomic Database [CGDB], n = 28) [63]. Again, patients with HER2-mutant tumors demonstrated poor outcomes with median PFS of 1.9 months and 3.0 months for the MDACC and CGDB cohort, respectively. Still, NSCLC patients harboring activating mutations in HER2 exhibit significantly better outcomes compared with those harboring sensitizing EGFR mutations, except for EGFR exon 20 insertions [67].
Given the limited benefit seen with checkpoint inhibitor monotherapy, recent efforts have focused on chemoimmunotherapy combinations. As such, among 13 NSCLC patients with HER2 exon 20 insertions the ORR was 30.8% with the combination of chemotherapy and immunotherapy [68]. In addition, Saalfeld et al. reported an ORR of 52.4% and median PFS of 6 months in treatment-naive patients that received pembrolizumab-based chemoimmunotherapy, as opposed to 16.1% and 4 months, respectively, for patients that received immune checkpoint inhibitor monotherapy beyond the first line of treatment [69]. Real-world data from the POLISH study indicated that first-line chemoimmunotherapy may achieve comparable results to chemotherapy plus angiogenesis inhibition with bevacizumab in the Chinese population; among 46 patients with actionable HER2 alterations, chemoimmunotherapy combinations achieved an ORR of 29% and median PFS of 5.2 months [70].
Currently, several clinical trials are testing combinations of PD-(L)1 inhibitors with anti-HER2 agents in patients with locally advanced or metastatic NSCLC. As such, NCT04042701 is a phase Ib study that will assess matters of safety and efficacy for the combination of T-DXd and pembrolizumab in HER2-expressing or HER2-mutant disease. Moreover, in the NCT04144569 phase II study, 30 patients with HER2 insertion mutation-positive advanced NSCLC will receive a PD-1 inhibitor plus pyrotinib after failure of first-line chemotherapy.

8. Conclusions

HER2 alterations represent important oncogenic drivers in NSCLC. During the past couple of years, progress has been made toward defining HER2-driven disease and determining the benefit from different classes of agents targeting HER2. Currently, platinum-based chemotherapy with/without immunotherapy is the preferred first-line treatment in patients with advanced or metastatic HER2-mutant NSCLC; T-DXd has been approved for the same subgroup of patients after receipt of prior systemic therapy. The European Society of Medical Oncology (ESMO) recommends upfront HER2 mutation testing (preferentially sequencing of exon 20) as part of a larger routine testing panel using next-generation sequencing (NGS), in patients with unresectable stage III and IV NSCLC who meet two or three of the following criteria, (i) lung adenocarcinoma or adenosquamous carcinoma; (ii) never-smoker status; (iii) female [4]. In addition, testing for HER2 amplification (by NGS or FISH), as well as HER2 expression (by IHC) can be considered for individuals in clinical studies and, in the case of EGFR TKI resistance, to explore the related resistance mechanisms.
With more agents in the pipeline, ongoing and future studies will need to provide answers to several questions including the role of T-DXd in first-line treatment (DESTINY-Lung04; NCT05048797), and the potential combination (NCT05048797 and NCT04686305) or sequencing of T-DXd with immunotherapy. In the case of TKIs, an important challenge lies in balancing treatment benefit with treatment-related toxicities that emanate from unselective inhibition of other ERBB family members; the latest efforts to target HER2-mutant NSCLC have employed highly selective HER2 inhibitors. Although we are in dire need of mature OS data as well as phase III data, the low incidence of HER2 mutations hampers the conducting of randomized controlled clinical trials. Finally, the impact of searching for and targeting HER2 mutations in early disease stages, as is the case in HER2-positive breast cancer, remains yet to be elucidated.

Author Contributions

Conceptualization, I.A.V., G.S.; writing—original draft preparation, I.A.V., G.S.; writing—review and editing, I.A.V., D.B., K.N.S., G.S.; supervision, G.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Goto, K.; Sang-We, K.; Kubo, T.; Goto, Y.; Ahn, M.-J.; Planchard, D.; Kim, D.-W.; Yang, J.-H.; Yang, T.-Y.; Pereira, K.; et al. LBA55 Trastuzumab deruxtecan (T-DXd) in patients (Pts) with HER2-mutant metastatic non-small cell lung cancer (NSCLC): Interim results from the phase 2 DESTINY-Lung02 trial. Ann. Oncol. 2022, 33, S1422. [Google Scholar] [CrossRef]
  2. Ogitani, Y.; Aida, T.; Hagihara, K.; Yamaguchi, J.; Ishii, C.; Harada, N.; Soma, M.; Okamoto, H.; Oitate, M.; Arakawa, S.; et al. DS-8201a, A Novel HER2-Targeting ADC with a Novel DNA Topoisomerase I Inhibitor, Demonstrates a Promising Antitumor Efficacy with Differentiation from T-DM1. Clin. Cancer Res. 2016, 22, 5097–5108. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. National Comprehensive Cancer Network. Non-Small Cell Lung Cancer (Version 1.2023). Available online: https://www.nccn.org/professionals/physician_gls/pdf/nscl.pdf (accessed on 18 January 2023).
  4. Ren, S.; Wang, J.; Ying, J.; Mitsudomi, T.; Lee, D.; Wang, Z.; Chu, Q.; Mack, P.; Cheng, Y.; Duan, J.; et al. Consensus for HER2 alterations testing in non-small-cell lung cancer. ESMO Open 2022, 7, 100395. [Google Scholar] [CrossRef] [PubMed]
  5. Robichaux, J.P.; Elamin, Y.Y.; Vijayan, R.; Nilsson, M.B.; Hu, L.; He, J.; Zhang, F.; Pisegna, M.; Poteete, A.; Sun, H.; et al. Pan-Cancer Landscape and Analysis of ERBB2 Mutations Identifies Poziotinib as a Clinically Active Inhibitor and Enhancer of T-DM1 Activity. Cancer Cell 2019, 36, 444–457.e7. [Google Scholar] [CrossRef]
  6. Mazières, J.; Peters, S.; Lepage, B.; Cortot, A.B.; Barlesi, F.; Beau-Faller, M.; Besse, B.; Blons, H.; Mansuet-Lupo, A.; Urban, T.; et al. Lung Cancer That Harbors an HER2 Mutation: Epidemiologic Characteristics and Therapeutic Perspectives. J. Clin. Oncol. 2013, 31, 1997–2003. [Google Scholar] [CrossRef] [Green Version]
  7. Tan, A.C.; Saw, S.P.; Chen, J.; Lai, G.G.; Oo, H.N.; Takano, A.; Lau, D.P.; Yeong, J.P.; Tan, G.S.; Lim, K.H.; et al. Clinical and Genomic Features of HER2 Exon 20 Insertion Mutations and Characterization of HER2 Expression by Immunohistochemistry in East Asian Non–Small-Cell Lung Cancer. JCO Precis. Oncol. 2022, 6, e2200278. [Google Scholar] [CrossRef]
  8. Offin, M.; Feldman, D.; Ni, A.; Myers, M.L.; Lai, W.V.; Pentsova, E.; Boire, A.; Daras, M.; Jordan, E.J.; Solit, D.B.; et al. Frequency and outcomes of brain metastases in patients with HER2-mutant lung cancers. Cancer 2019, 125, 4380–4387. [Google Scholar] [CrossRef]
  9. Yang, S.; Wang, Y.; Zhao, C.; Li, X.; Liu, Q.; Mao, S.; Liu, Y.; Yu, X.; Wang, W.; Tian, Q.; et al. Exon 20 YVMA insertion is associated with high incidence of brain metastasis and inferior outcome of chemotherapy in advanced non-small cell lung cancer patients with HER2 kinase domain mutations. Transl. Lung Cancer Res. 2021, 10, 753–765. [Google Scholar] [CrossRef]
  10. Li, B.T.; Michelini, F.; Misale, S.; Cocco, E.; Baldino, L.; Cai, Y.; Shifman, S.; Tu, H.-Y.; Myers, M.L.; Xu, C.; et al. HER2-Mediated Internalization of Cytotoxic Agents in ERBB2 Amplified or Mutant Lung Cancers. Cancer Discov. 2020, 10, 674–687. [Google Scholar] [CrossRef] [Green Version]
  11. Nakagawa, K.; Nagasaka, M.; Felip, E.; Pacheco, J.; Baik, C.; Goto, Y.; Saltos, A.; Li, B.; Udagawa, H.; Gadgeel, S.; et al. OA04.05 Trastuzumab Deruxtecan in HER2-Overexpressing Metastatic Non-Small Cell Lung Cancer: Interim Results of DESTINY-Lung01. J. Thorac. Oncol. 2021, 16, S109–S110. [Google Scholar] [CrossRef]
  12. Li, B.T.; Smit, E.F.; Goto, Y.; Nakagawa, K.; Udagawa, H.; Mazières, J.; Nagasaka, M.; Bazhenova, L.; Saltos, A.N.; Felip, E.; et al. Trastuzumab Deruxtecan in HER2-Mutant Non-Small-Cell Lung Cancer. N. Engl. J. Med. 2022, 386, 241–251. [Google Scholar] [CrossRef]
  13. Tarantino, P.; Prat, A.; Curigliano, G. New anti-HER2 agents for brain metastasis: Histology-agnostic weapons? Breast Cancer Res. Treat. 2021, 185, 879–881. [Google Scholar] [CrossRef]
  14. Jacobson, A. Trastuzumab Deruxtecan Improves Progression-Free Survival and Intracranial Response in Patients with HER2-Positive Metastatic Breast Cancer and Brain Metastases. Oncologist 2022, 27, S3–S4. [Google Scholar] [CrossRef]
  15. Jerusalem, G.; Park, Y.H.; Yamashita, T.; Hurvitz, S.A.; Modi, S.; Andre, F.; Krop, I.E.; Farré, X.G.; You, B.; Saura, C.; et al. Trastuzumab Deruxtecan in HER2-Positive Metastatic Breast Cancer Patients with Brain Metastases: A DESTINY-Breast01 Subgroup Analysis. Cancer Discov. 2022, 12, 2754–2762. [Google Scholar] [CrossRef]
  16. Kabraji, S.; Ni, J.; Sammons, S.; Li, T.; Van Swearingen, A.E.; Wang, Y.; Pereslete, A.; Hsu, L.; DiPiro, P.J.; Lascola, C.; et al. Preclinical and Clinical Efficacy of Trastuzumab Deruxtecan in Breast Cancer Brain Metastases. Clin. Cancer Res. 2022, 29, 174–182. [Google Scholar] [CrossRef]
  17. Pérez-García, J.M.; Batista, M.V.; Cortez, P.; Ruiz-Borrego, M.; Cejalvo, J.M.; de la Haba-Rodriguez, J.; Garrigós, L.; Racca, F.; Servitja, S.; Blanch, S.; et al. Trastuzumab deruxtecan in patients with central nervous system involvement from HER2-positive breast cancer: The DEBBRAH trial. Neuro-Oncology 2022, 25, 157–166. [Google Scholar] [CrossRef]
  18. Bartsch, R.; Berghoff, A.S.; Furtner, J.; Marhold, M.; Bergen, E.S.; Roider-Schur, S.; Starzer, A.M.; Forstner, H.; Rottenmanner, B.; Dieckmann, K.; et al. Trastuzumab deruxtecan in HER2-positive breast cancer with brain metastases: A single-arm, phase 2 trial. Nat. Med. 2022, 28, 1840–1847. [Google Scholar] [CrossRef]
  19. Vathiotis, I.A.; Charpidou, A.; Gavrielatou, N.; Syrigos, K.N. HER2 Aberrations in Non-Small Cell Lung Cancer: From Pathophysiology to Targeted Therapy. Pharmaceuticals 2021, 14, 1300. [Google Scholar] [CrossRef]
  20. Hotta, K.; Aoe, K.; Kozuki, T.; Ohashi, K.; Ninomiya, K.; Ichihara, E.; Kubo, T.; Ninomiya, T.; Chikamori, K.; Harada, D.; et al. A Phase II Study of Trastuzumab Emtansine in HER2-Positive Non–Small Cell Lung Cancer. J. Thorac. Oncol. 2018, 13, 273–279. [Google Scholar] [CrossRef] [Green Version]
  21. Li, B.T.; Shen, R.; Buonocore, D.; Olah, Z.T.; Ni, A.; Ginsberg, M.S.; Ulaner, G.A.; Offin, M.; Feldman, D.; Hembrough, T.; et al. Ado-Trastuzumab Emtansine for Patients With HER2-Mutant Lung Cancers: Results From a Phase II Basket Trial. J. Clin. Oncol. 2018, 36, 2532–2537. [Google Scholar] [CrossRef]
  22. Peters, S.; Stahel, R.; Bubendorf, L.; Bonomi, P.; Villegas, A.; Kowalski, D.M.; Baik, C.S.; Isla, D.; Carpeno, J.C.; Garrido, P.; et al. Trastuzumab Emtansine (T-DM1) in Patients with Previously Treated HER2-Overexpressing Metastatic Non-Small Cell Lung Cancer: Efficacy, Safety, and Biomarkers. Clin. Cancer Res. 2019, 25, 64–72. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Tsurutani, J.; Iwata, H.; Krop, I.; Jänne, P.A.; Doi, T.; Takahashi, S.; Park, H.; Redfern, C.; Tamura, K.; Wise-Draper, T.M.; et al. Targeting HER2 with Trastuzumab Deruxtecan: A Dose-Expansion, Phase I Study in Multiple Advanced Solid Tumors. Cancer Discov. 2020, 10, 688–701. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Modi, S.; Jacot, W.; Yamashita, T.; Sohn, J.; Vidal, M.; Tokunaga, E.; Tsurutani, J.; Ueno, N.T.; Prat, A.; Chae, Y.S.; et al. Trastuzumab Deruxtecan in Previously Treated HER2-Low Advanced Breast Cancer. New Engl. J. Med. 2022, 387, 9–20. [Google Scholar] [CrossRef] [PubMed]
  25. Diéras, V.; Deluche, E.; Lusque, A.; Pistilli, B.; Bachelot, T.; Pierga, J.-Y.; Viret, F.; Levy, C.; Salabert, L.; Le Du, F.; et al. Abstract PD8-02: Trastuzumab deruxtecan (T-DXd) for advanced breast cancer patients (ABC), regardless HER2 status: A phase II study with biomarkers analysis (DAISY). Cancer Res. 2022, 82, PD8-02. [Google Scholar] [CrossRef]
  26. Fernandez, A.I.; Liu, M.; Bellizzi, A.; Brock, J.; Fadare, O.; Hanley, K.; Harigopal, M.; Jorns, J.M.; Kuba, M.G.; Ly, A.; et al. Examination of Low ERBB2 Protein Expression in Breast Cancer Tissue. JAMA Oncol. 2022, 8, 607. [Google Scholar] [CrossRef]
  27. Tarantino, P.; Hamilton, E.; Tolaney, S.M.; Cortes, J.; Morganti, S.; Ferraro, E.; Marra, A.; Viale, G.; Trapani, D.; Cardoso, F.; et al. HER2-Low Breast Cancer: Pathological and Clinical Landscape. J. Clin. Oncol. 2020, 38, 1951–1962. [Google Scholar] [CrossRef]
  28. Moutafi, M.; Robbins, C.J.; Yaghoobi, V.; Fernandez, A.I.; Martinez-Morilla, S.; Xirou, V.; Bai, Y.; Song, Y.; Gaule, P.; Krueger, J.; et al. Quantitative measurement of HER2 expression to subclassify ERBB2 unamplified breast cancer. Lab. Investig. 2022, 102, 1101–1108. [Google Scholar] [CrossRef]
  29. Yamaguchi, K.; Bang, Y.-J.; Iwasa, S.; Sugimoto, N.; Ryu, M.-H.; Sakai, D.; Chung, H.C.; Kawakami, H.; Yabusaki, H.; Lee, J.; et al. Trastuzumab Deruxtecan in Anti–Human Epidermal Growth Factor Receptor 2 Treatment–Naive Patients With Human Epidermal Growth Factor Receptor 2–Low Gastric or Gastroesophageal Junction Adenocarcinoma: Exploratory Cohort Results in a Phase II Trial. J. Clin. Oncol. 2023, 41, 816–825. [Google Scholar] [CrossRef]
  30. Abuhelwa, Z.; Alloghbi, A.; Nagasaka, M. A comprehensive review on antibody-drug conjugates (ADCs) in the treatment landscape of non-small cell lung cancer (NSCLC). Cancer Treat. Rev. 2022, 106, 102393. [Google Scholar] [CrossRef]
  31. Coleman, N.; Yap, T.A.; Heymach, J.V.; Meric-Bernstam, F.; Le, X. Antibody-drug conjugates in lung cancer: Dawn of a new era? NPJ Precis. Oncol. 2023, 7, 5. [Google Scholar] [CrossRef]
  32. Amiri-Kordestani, L.; Blumenthal, G.M.; Xu, Q.C.; Zhang, L.; Tang, S.W.; Ha, L.; Weinberg, W.C.; Chi, B.; Candau-Chacon, R.; Hughes, P.; et al. FDA Approval: Ado-Trastuzumab Emtansine for the Treatment of Patients with HER2-Positive Metastatic Breast Cancer. Clin. Cancer Res. 2014, 20, 4436–4441. [Google Scholar] [CrossRef] [Green Version]
  33. Iwama, E.; Zenke, Y.; Sugawara, S.; Daga, H.; Morise, M.; Yanagitani, N.; Sakamoto, T.; Murakami, H.; Kishimoto, J.; Matsumoto, S.; et al. Trastuzumab emtansine for patients with non–small cell lung cancer positive for human epidermal growth factor receptor 2 exon-20 insertion mutations. Eur. J. Cancer 2021, 162, 99–106. [Google Scholar] [CrossRef]
  34. Liu, Y.; Lian, W.; Zhao, X.; Qi, W.; Xu, J.; Xiao, L.; Qing, Y.; Xue, T.; Wang, J. A first in-human study of A166 in patients with locally advanced/metastatic solid tumors which are HER2-positive or HER2-amplified who did not respond or stopped responding to approved therapies. J. Clin. Oncol. 2020, 38, 1049. [Google Scholar] [CrossRef]
  35. Hu, X.; Zhang, J.; Liu, R.; Gao, S.; Qing, Y.; Yi, S.; Yuan, J.; Chen, H.; Fan, B.; Zheng, H.; et al. Phase I study of A166 in patients with HER2-expressing locally advanced or metastatic solid tumors. J. Clin. Oncol. 2021, 39, 1024. [Google Scholar] [CrossRef]
  36. Le Joncour, V.; Martins, A.; Puhka, M.; Isola, J.; Salmikangas, M.; Laakkonen, P.; Joensuu, H.; Barok, M. A Novel Anti-HER2 Antibody–Drug Conjugate XMT-1522 for HER2-Positive Breast and Gastric Cancers Resistant to Trastuzumab Emtansine. Mol. Cancer Ther. 2019, 18, 1721–1730. [Google Scholar] [CrossRef] [Green Version]
  37. Hamilton, E.P.; Barve, M.A.; Bardia, A.; Beeram, M.; Bendell, J.C.; Mosher, R.; Hailman, E.; Bergstrom, D.A.; Burris, H.A.; Soliman, H.H. Phase 1 dose escalation of XMT-1522, a novel HER2-targeting antibody-drug conjugate (ADC), in patients (pts) with HER2-expressing breast, lung and gastric tumors. J. Clin. Oncol. 2018, 36, 2546. [Google Scholar] [CrossRef] [Green Version]
  38. Zhang, J.; Ji, D.; Shen, W.; Xiao, Q.; Gu, Y.; O’Shaughnessy, J.; Hu, X. Phase I Trial of a Novel Anti-HER2 Antibody–Drug Conjugate, ARX788, for the Treatment of HER2-Positive Metastatic Breast Cancer. Clin. Cancer Res. 2022, 28, 4212–4221. [Google Scholar] [CrossRef]
  39. Skidmore, L.; Sakamuri, S.; Knudsen, N.A.; Hewet, A.G.; Milutinovic, S.; Barkho, W.; Biroc, S.L.; Kirtley, J.; Marsden, R.; Storey, K.; et al. ARX788, a Site-specific Anti-HER2 Antibody–Drug Conjugate, Demonstrates Potent and Selective Activity in HER2-low and T-DM1–resistant Breast and Gastric Cancers. Mol. Cancer Ther. 2020, 19, 1833–1843. [Google Scholar] [CrossRef]
  40. Elamin, Y.Y.; Robichaux, J.P.; Carter, B.W.; Altan, M.; Gibbons, D.L.; Fossella, F.V.; Lam, V.K.; Patel, A.B.; Negrao, M.V.; Le, X.; et al. Poziotinib for Patients With HER2 Exon 20 Mutant Non–Small-Cell Lung Cancer: Results From a Phase II Trial. J. Clin. Oncol. 2022, 40, 702–709. [Google Scholar] [CrossRef]
  41. Le, X.; Cornelissen, R.; Garassino, M.; Clarke, J.M.; Tchekmedyian, N.; Goldman, J.W.; Leu, S.-Y.; Bhat, G.; Lebel, F.; Heymach, J.V.; et al. Poziotinib in Non–Small-Cell Lung Cancer Harboring HER2 Exon 20 Insertion Mutations After Prior Therapies: ZENITH20-2 Trial. J. Clin. Oncol. 2022, 40, 710–718. [Google Scholar] [CrossRef]
  42. Zhou, C.; Li, X.; Wang, Q.; Gao, G.; Zhang, Y.; Chen, J.; Shu, Y.; Hu, Y.; Fan, Y.; Fang, J.; et al. Pyrotinib in HER2-Mutant Advanced Lung Adenocarcinoma After Platinum-Based Chemotherapy: A Multicenter, Open-Label, Single-Arm, Phase II Study. J. Clin. Oncol. 2020, 38, 2753–2761. [Google Scholar] [CrossRef] [PubMed]
  43. Han, H.; Li, S.; Chen, T.; Fitzgerald, M.; Liu, S.; Peng, C.; Tang, K.H.; Cao, S.; Chouitar, J.; Wu, J.; et al. Targeting HER2 Exon 20 Insertion–Mutant Lung Adenocarcinoma with a Novel Tyrosine Kinase Inhibitor Mobocertinib. Cancer Res. 2021, 81, 5311–5324. [Google Scholar] [CrossRef] [PubMed]
  44. Neal, J.; Doebele, R.; Riely, G.; Spira, A.; Horn, L.; Piotrowska, Z.; Costa, D.; Zhang, S.; Bottino, D.; Zhu, J.; et al. P1.13–44 Safety, PK, and Preliminary Antitumor Activity of the Oral EGFR/HER2 Exon 20 Inhibitor TAK-788 in NSCLC. J. Thorac. Oncol. 2018, 13, S599. [Google Scholar] [CrossRef] [Green Version]
  45. Riely, G.; Neal, J.; Camidge, D.; Spira, A.; Piotrowska, Z.; Horn, L.; Costa, D.; Tsao, A.; Patel, J.; Gadgeel, S.; et al. 1261MO Updated results from a phase I/II study of mobocertinib (TAK-788) in NSCLC with EGFR exon 20 insertions (exon20ins). Ann. Oncol. 2020, 31, S815–S816. [Google Scholar] [CrossRef]
  46. Katayama, R. Hitting the target in HER2 mutant cancers. Nat. Cancer 2022, 3, 785–786. [Google Scholar] [CrossRef]
  47. Estrada-Bernal, A.; Le, A.T.; Doak, A.E.; Tirunagaru, V.G.; Silva, S.; Bull, M.R.; Smaill, J.B.; Patterson, A.V.; Kim, C.; Liu, S.V.; et al. Tarloxotinib Is a Hypoxia-Activated Pan-HER Kinase Inhibitor Active Against a Broad Range of HER-Family Oncogenes. Clin. Cancer Res. 2021, 27, 1463–1475. [Google Scholar] [CrossRef]
  48. Koga, T.; Suda, K.; Nishino, M.; Fujino, T.; Ohara, S.; Hamada, A.; Soh, J.; Tirunagaru, V.; Vellanki, A.; Doebele, R.C.; et al. Activity and mechanism of acquired resistance to tarloxotinib in HER2 mutant lung cancer: An in vitro study. Transl. Lung Cancer Res. 2021, 10, 3659–3670. [Google Scholar] [CrossRef]
  49. Liu, S.V.; Villaruz, L.C.; Lee, V.H.F.; Zhu, V.W.; Baik, C.S.; Sacher, A.; McCoach, C.E.; Nguyen, D.; Li, J.Y.-C.; Pacheco, J.M.; et al. LBA61 First analysis of RAIN-701: Study of tarloxotinib in patients with non-small cell lung cancer (NSCLC) EGFR Exon 20 insertion, HER2-activating mutations & other solid tumours with NRG1/ERBB gene fusions. Ann. Oncol. 2020, 31, S1189. [Google Scholar] [CrossRef]
  50. Wilding, B.; Scharn, D.; Böse, D.; Baum, A.; Santoro, V.; Chetta, P.; Schnitzer, R.; Botesteanu, D.A.; Reiser, C.; Kornigg, S.; et al. Discovery of potent and selective HER2 inhibitors with efficacy against HER2 exon 20 insertion-driven tumors, which preserve wild-type EGFR signaling. Nat. Cancer 2022, 3, 821–836. [Google Scholar] [CrossRef]
  51. Son, J.; Jang, J.; Beyett, T.S.; Eum, Y.; Haikala, H.M.; Verano, A.; Lin, M.; Hatcher, J.M.; Kwiatkowski, N.P.; Eser, P.; et al. A Novel HER2-Selective Kinase Inhibitor Is Effective in HER2 Mutant and Amplified Non–Small Cell Lung Cancer. Cancer Res. 2022, 82, 1633–1645. [Google Scholar] [CrossRef]
  52. Heymach, J.; Opdam, F.; Barve, M.; Gibson, N.; Sadrolhefazi, B.; Serra, J.; Yamamoto, N. A Phase I, Open-Label, Dose Confirmation, Escalation, and Expansion Trial of BI 1810631 as Monotherapy in Patients with Advanced or Metastatic Solid Tumors With HER2 Aberrations. Clin. Lung Cancer 2022. [Google Scholar] [CrossRef]
  53. Riethmüller, G. Symmetry breaking: Bispecific antibodies, the beginnings, and 50 years on. Cancer Immun. 2012, 12, 12. [Google Scholar]
  54. Labrijn, A.F.; Janmaat, M.L.; Reichert, J.M.; Parren, P.W.H.I. Bispecific antibodies: A mechanistic review of the pipeline. Nat. Rev. Drug Discov. 2019, 18, 585–608. [Google Scholar] [CrossRef]
  55. Geuijen, C.A.W.; De Nardis, C.; Maussang, D.; Rovers, E.; Gallenne, T.; Hendriks, L.J.A.; Visser, T.; Nijhuis, R.; Logtenberg, T.; de Kruif, J.; et al. Unbiased Combinatorial Screening Identifies a Bispecific IgG1 that Potently Inhibits HER3 Signaling via HER2-Guided Ligand Blockade. Cancer Cell 2018, 33, 922–936e10. [Google Scholar] [CrossRef] [Green Version]
  56. Schram, A.M.; Odintsov, I.; Espinosa-Cotton, M.; Khodos, I.; Sisso, W.J.; Mattar, M.S.; Lui, A.J.W.; Vojnic, M.; Shameem, S.H.; Chauhan, T.; et al. Zenocutuzumab, a HER2xHER3 Bispecific Antibody, Is Effective Therapy for Tumors Driven by NRG1 Gene Rearrangements. Cancer Discov. 2022, 12, 1233–1247. [Google Scholar] [CrossRef]
  57. Alsina, M.; Boni, V.; Schellens, J.H.; Moreno, V.; Bol, K.; Westendorp, M.; Sirulnik, L.A.; Tabernero, J.; Calvo, E. First-in-human phase 1/2 study of MCLA-128, a full length IgG1 bispecific antibody targeting HER2 and HER3: Final phase 1 data and preliminary activity in HER2+ metastatic breast cancer (MBC). J. Clin. Oncol. 2017, 35, 2522. [Google Scholar] [CrossRef]
  58. Dong, Z.-Y.; Zhang, J.-T.; Liu, S.-Y.; Su, J.; Zhang, C.; Xie, Z.; Zhou, Q.; Tu, H.-Y.; Xu, C.-R.; Yan, L.-X.; et al. EGFR mutation correlates with uninflamed phenotype and weak immunogenicity, causing impaired response to PD-1 blockade in non-small cell lung cancer. OncoImmunology 2017, 6, e1356145. [Google Scholar] [CrossRef] [Green Version]
  59. Toki, M.I.; Mani, N.; Smithy, J.W.; Liu, Y.; Altan, M.; Wasserman, B.; Tuktamyshov, R.; Schalper, K.; Syrigos, K.N.; Rimm, D.L. Immune Marker Profiling and Programmed Death Ligand 1 Expression Across NSCLC Mutations. J. Thorac. Oncol. 2018, 13, 1884–1896. [Google Scholar] [CrossRef] [Green Version]
  60. Reuben, A.; Zhang, J.; Chiou, S.-H.; Gittelman, R.M.; Li, J.; Lee, W.-C.; Fujimoto, J.; Behrens, C.; Liu, X.; Wang, F.; et al. Comprehensive T cell repertoire characterization of non-small cell lung cancer. Nat. Commun. 2020, 11, 603. [Google Scholar] [CrossRef] [Green Version]
  61. Qiao, M.; Jiang, T.; Liu, X.; Mao, S.; Zhou, F.; Li, X.; Zhao, C.; Chen, X.; Su, C.; Ren, S.; et al. Immune Checkpoint Inhibitors in EGFR-Mutated NSCLC: Dusk or Dawn? J. Thorac. Oncol. 2021, 16, 1267–1288. [Google Scholar] [CrossRef]
  62. Benjamin, D.J.; Chen, S.; Eldredge, J.B.; Schokrpur, S.; Li, D.; Quan, Z.; Chan, J.W.; Cummings, A.L.; Daly, M.E.; Goldman, J.W.; et al. The Role of Chemotherapy Plus Immune Checkpoint Inhibitors in Oncogenic-Driven NSCLC: A University of California Lung Cancer Consortium Retrospective Study. JTO Clin. Res. Rep. 2022, 3, 100427. [Google Scholar] [CrossRef] [PubMed]
  63. Negrao, M.V.; Skoulidis, F.; Montesion, M.; Schulze, K.; Bara, I.; Shen, V.; Xu, H.; Hu, S.; Sui, D.; Elamin, Y.Y.; et al. Oncogene-specific differences in tumor mutational burden, PD-L1 expression, and outcomes from immunotherapy in non-small cell lung cancer. J. Immunother. Cancer 2021, 9, e002891. [Google Scholar] [CrossRef] [PubMed]
  64. Lai, W.-C.V.; Feldman, D.L.; Buonocore, D.J.; Brzostowski, E.B.; Rizvi, H.; Plodkowski, A.J.; Ni, A.; Sabari, J.K.; Offin, M.D.; Kris, M.G.; et al. PD-L1 expression, tumor mutation burden and response to immune checkpoint blockade in patients with HER2-mutant lung cancers. J. Clin. Oncol. 2018, 36, 9060. [Google Scholar] [CrossRef]
  65. Mazieres, J.; Drilon, A.; Lusque, A.B.; Mhanna, L.; Cortot, A.; Mezquita, L.; Thai, A.A.; Mascaux, C.; Couraud, S.; Veillon, R.; et al. Immune checkpoint inhibitors for patients with advanced lung cancer and oncogenic driver alterations: Results from the IMMUNOTARGET registry. Ann. Oncol. 2019, 30, 1321–1328. [Google Scholar] [CrossRef] [PubMed]
  66. Guisier, F.; Dubos-Arvis, C.; Viñas, F.; Doubre, H.; Ricordel, C.; Ropert, S.; Janicot, H.; Bernardi, M.; Fournel, P.; Lamy, R.; et al. Efficacy and Safety of Anti-PD-1 Immunotherapy in Patients With Advanced NSCLC With BRAF, HER2, or MET Mutations or RET Translocation: GFPC 01-2018. J. Thorac. Oncol. 2020, 15, 628–636. [Google Scholar] [CrossRef]
  67. Lau, S.C.; Fares, A.F.; Le, L.W.; Mackay, K.M.; Soberano, S.; Chan, S.W.; Smith, E.; Ryan, M.; Tsao, M.S.; Bradbury, P.A.; et al. Subtypes of EGFR- and HER2-Mutant Metastatic NSCLC Influence Response to Immune Checkpoint Inhibitors. Clin. Lung Cancer 2021, 22, 253–259. [Google Scholar] [CrossRef]
  68. Tian, P.; Zeng, H.; Ji, L.; Ding, Z.; Ren, L.; Gao, W.; Fan, Z.; Li, L.; Le, X.; Li, P.; et al. Lung adenocarcinoma with ERBB2 exon 20 insertions: Comutations and immunogenomic features related to chemoimmunotherapy. Lung Cancer 2021, 160, 50–58. [Google Scholar] [CrossRef]
  69. Saalfeld, F.C.; Wenzel, C.; Christopoulos, P.; Merkelbach-Bruse, S.; Reissig, T.M.; Laßmann, S.; Thiel, S.; Stratmann, J.A.; Marienfeld, R.; Berger, J.; et al. Efficacy of Immune Checkpoint Inhibitors Alone or in Combination With Chemotherapy in NSCLC Harboring ERBB2 Mutations. J. Thorac. Oncol. 2021, 16, 1952–1958. [Google Scholar] [CrossRef]
  70. Yang, G.; Yang, Y.; Liu, R.; Li, W.; Xu, H.; Hao, X.; Li, J.; Xing, P.; Zhang, S.; Ai, X.; et al. First-line immunotherapy or angiogenesis inhibitor plus chemotherapy for HER2-altered NSCLC: A retrospective real-world POLISH study. Ther. Adv. Med. Oncol. 2022, 14, 17588359221082339. [Google Scholar] [CrossRef]
Table 1. Clinical trials evaluating antibody–drug conjugates in patients with NSCLC harboring HER2 aberrations. NSCLC, non-small cell lung cancer; HER2, human epidermal growth factor receptor 2; DAR, drug-to-antibody ratio; ORR, objective response rate; PFS, progression-free survival; CI, confidence intervals; OS, overall survival; TRAE, treatment-related adverse event; IHC, immunohistochemistry; FISH, fluorescence in situ hybridization; NE, not evaluable; NA, not available; NR, not reached.
Table 1. Clinical trials evaluating antibody–drug conjugates in patients with NSCLC harboring HER2 aberrations. NSCLC, non-small cell lung cancer; HER2, human epidermal growth factor receptor 2; DAR, drug-to-antibody ratio; ORR, objective response rate; PFS, progression-free survival; CI, confidence intervals; OS, overall survival; TRAE, treatment-related adverse event; IHC, immunohistochemistry; FISH, fluorescence in situ hybridization; NE, not evaluable; NA, not available; NR, not reached.
AgentAntibody; ChemotherapeuticStudyPhasenPrior
Systemic Therapy
HER2
Positivity Definition
ORR
n (%)
PFS in Months
Median (95% CI)
OS in Months
Median (95% CI)
Most
Common Grade 3 or Higher TRAE
(%)
T-DM1Trastuzumab; emtansineHotta et al. [20].215YesIHC 3+, 2+/FISH+, or exon 20 mutation1 (6.7)2.0 (1.4–4.0)10.9 (4.4–12.0)Thrombocytopenia (40.0)
T-DM1Trastuzumab; emtansinePeters et al. [22].229; 20YesIHC 2+; IHC 3+0 (0); 4 (20)2.6 (1.4–2.8); 2.7 (1.4–8.3)12.2 (3.8–23.3); 15.3 (4.1-NE)NA
T-DM1Trastuzumab; emtansineLi et al. [10]249YesActivating mutation or amplification25 (51.0)5.0 (3.5–5.9)NAThrombocytopenia (6.0),
Anemia (6.0)
T-DM1Trastuzumab; emtansineIwama et al. [32]222YesExon 20 insertion mutation8 (38.1)2.8 (1.4–4.4)8.1 (3.5–13.2)Thrombocytopenia (18.2)
T-DXdTrastuzumab; deruxtecanTsurutani et al. [23]118YesIHC 1+, 2+, 3+ or amplification10 (55.6)11.3 (5.5–14.1)NR (17.3-NE)NA
T-DXdTrastuzumab; deruxtecanLi et al. [12].191YesMutation50 (54.9)8.2 (6.0–11.9)17.8 (13.8–22.1)Neutropenia (19.0)
T-DXdTrastuzumab; deruxtecanNakagawa et al. [11]149YesIHC 2+, 3+12 (24.5)5.4 (2.8–7.0)NANeutropenia (20.4)
T-DXdTrastuzumab; deruxtecanGoto et al. [1]252 (5.4 mg/kg); 28 (6.4 mg/kg)YesMutation28 (53.8); 12 (42.9)NANANA
Table 2. Studies evaluating PD-1 axis blockade in patients with NSCLC harboring HER2 aberrations. NSCLC, non-small cell lung cancer; HER2, human epidermal growth factor receptor 2; PD-1, programmed cell death protein 1; IO, immunotherapy; PD-L1 TPS, programmed death-ligand 1 tumor proportion score; ORR, objective response rate; PFS, progression-free survival; CI, confidence intervals; OS, overall survival; NA, not available; NR, not reached.
Table 2. Studies evaluating PD-1 axis blockade in patients with NSCLC harboring HER2 aberrations. NSCLC, non-small cell lung cancer; HER2, human epidermal growth factor receptor 2; PD-1, programmed cell death protein 1; IO, immunotherapy; PD-L1 TPS, programmed death-ligand 1 tumor proportion score; ORR, objective response rate; PFS, progression-free survival; CI, confidence intervals; OS, overall survival; NA, not available; NR, not reached.
StudyDesignnPrior
Systemic Therapy
IO RegimenPD-L1 TPS ≥ 50 (%)ORR
n (%)
PFS in Months
Median (95% CI)
OS in Months
Median (95% CI)
Lai et al. [64]Retrospective26NANANA3 (11.5)1.9 (1.5–4.0)10.4 (5.9–NA)
Mazieres et al. [64]Retrospective29YesMonotherapy02 (7.4)2.1 (1.3–4.7)21.3 (3.8–28.0)
Guisier et al. [65]Retrospective23YesMonotherapy4.36 (27.3)2.2 (1.7–15.2)20.4 (9.3–NR)
Negrao et al. [63]Retrospective15; 28NA; NAMonotherapy; monotherapyNA; NANA; NA1.9 (1.6–2.1); 3.0 (1.8–NA)16.8 (3.1–30.6); 10.8 (5.6–NA)
Lau et al. [63]Retrospective14Yes (78.6)Monotherapy, combination with anti-CTLA-43 (23.1)4 (28.6)3.6 (1.6–NR)NA
Tian et al. [63]Retrospective13No (76.9)Combination with chemotherapyNA4 (30.8)8.0 (5.2–NR)NA
Saalfeld et al. [63]Retrospective61No (44.3)Monotherapy, combination with chemotherapy15.511 (52.4), 1st line chemoimmunotherapy; 5 (16.1), >1st line monotherapy6.0 (6.0–14.0) 1st line chemoimmunotherapy; 4.0 (4.0–6.0), >1st line monotherapyNR, (NA–NA) 1st line chemoimmunotherapy; 10.0 (6.0–NA), >1st line monotherapy
Yang et al. [63]Retrospective46NoCombination with chemotherapy6.513 (28.9)5.2 (3.6–6.8)NR (NA–NA)
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Vathiotis, I.A.; Bafaloukos, D.; Syrigos, K.N.; Samonis, G. Evolving Treatment Landscape of HER2-mutant Non-Small Cell Lung Cancer: Trastuzumab Deruxtecan and Beyond. Cancers 2023, 15, 1286. https://doi.org/10.3390/cancers15041286

AMA Style

Vathiotis IA, Bafaloukos D, Syrigos KN, Samonis G. Evolving Treatment Landscape of HER2-mutant Non-Small Cell Lung Cancer: Trastuzumab Deruxtecan and Beyond. Cancers. 2023; 15(4):1286. https://doi.org/10.3390/cancers15041286

Chicago/Turabian Style

Vathiotis, Ioannis A., Dimitrios Bafaloukos, Konstantinos N. Syrigos, and George Samonis. 2023. "Evolving Treatment Landscape of HER2-mutant Non-Small Cell Lung Cancer: Trastuzumab Deruxtecan and Beyond" Cancers 15, no. 4: 1286. https://doi.org/10.3390/cancers15041286

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop