Back to Journals » Lung Cancer: Targets and Therapy » Volume 15

Spotlight on Patritumab Deruxtecan (HER3-DXd) from HERTHENA Lung01. Is a Median PFS of 5.5 Months Enough in Light of FLAURA-2 and MARIPOSA?

Authors Arter ZL , Nagasaka M 

Received 1 May 2024

Accepted for publication 2 July 2024

Published 9 July 2024 Volume 2024:15 Pages 115—121

DOI https://doi.org/10.2147/LCTT.S467169

Checked for plagiarism Yes

Review by Single anonymous peer review

Peer reviewer comments 2

Editor who approved publication: Professor Fengying Wu



Zhaohui Liao Arter,1,2 Misako Nagasaka1– 3

1Division of Hematology/Oncology, University of California Irvine School of Medicine, Orange, CA, USA; 2Chao Family Comprehensive Cancer Center, Orange, CA, USA; 3St. Marianna University School of Medicine, Kawasaki, Japan

Correspondence: Misako Nagasaka, Department of Medicine, Division of Hematology/Oncology, UC Irvine, Orange, CA, 92602, USA, Email [email protected]

Abstract: On December 22, 2023, the US Food and Drug Administration (FDA) approved the biologics license application for patritumab deruxtecan (HER3-DXd) for priority review. This treatment is aimed at adult patients with locally advanced or metastatic NSCLC with EGFR mutations, who have received at least two prior systemic therapies. Approval of patritumab deruxtecan would mark it as the first HER3 targeted therapy in the United States. This prioritization by the FDA is grounded in compelling results from the global Phase II HERTHENA-Lung01 trial, wherein HER3-DXd exhibited clinically meaningful efficacy, achieving a median progression-free survival (mPFS) of 5.5 months in patients with heavily treated EGFR-mutated NSCLC. A pivotal question remains: Is a mPFS of 5.5 months sufficient in the context of the evolving first-line landscape observed in the FLAURA-2 and MARIPOSA trials?

Keywords: EGFR, NSCLC, post-osimertinib, targeted therapy

Background

Treatment landscape for NSCLC with EGFR mutations and resistance to EGFR TKIs

Significant progress has been made in NSCLC management through the development of targeted therapeutic approaches, particularly in the context of patients harboring EGFR mutations. These mutations are present in a significant percentage of NSCLC cases, varying by population and demographic factors. As a third-generation EGFR tyrosine kinase inhibitor (TKI), osimertinib has transformed the first-line treatment landscape for patients with NSCLC. However, despite initial great responses, a majority of tumors ultimately become resistant to osimertinib, necessitating the exploration of subsequent therapeutic strategies. This resistance can emerge through multiple pathways, such as EGFR-dependent and EGFR-independent routes. Salvage treatments following disease advancement on EGFR TKs and platinum-based chemotherapy (PBC) reported mPFS in the range of 2.8–3.3 months.1

To effectively manage progression following osimertinib treatment, a comprehensive and multifaceted strategy is essential. This includes proactively delaying resistance through intensified initial treatments. Choices involve pairing an EGFR TKI (such as osimertinib) with chemotherapy, as seen in the FLAURA2 trial,2 or integrating an EGFR-MET bispecific agent, as explored in the MARIPOSA study.3,4 Additionally, directly addressing specific resistance mechanisms is crucial. For instance, if a patient acquires a C797S mutation causing resistance to osimertinib, considering a novel fourth-generation EGFR inhibitor being tested in clinical trials might be a practical choice. Alternatively, broader approaches like chemotherapy, antibody-drug conjugates (ADCs), or a combination of chemotherapy and immunotherapy, potentially augmented with a VEGF inhibitor, may be employed.

Treatment approaches incorporating both chemotherapy and immune checkpoint inhibitors (ICI) were tested against PBC in the Phase 3 KEYNOTE-789 and CheckMate 722 trials. However, these investigations failed to show any notable disparities in survival results among patients unresponsive to previous EGFR TKI therapy.5,6 The ORIENT-31 and IMpower150 trials enhanced their regimen by including an anti-VEGF monoclonal antibody (either bevacizumab or its biosimilar) to the existing combination of ICIs and chemotherapy. ORIENT-31, however, failed to reveal a beneficial effect on overall survival (OS), and its study was specifically confined to China.7 The final results from the IMpower151 trial demonstrated that the inclusion of bevacizumab to the combined therapy of chemotherapy and immunotherapy did not yield any statistically significant therapeutic advantage for patients with EGFR mutated NSCLC.8

HER3 ADC

The human epidermal growth factor receptor 3 (HER3), also called receptor tyrosine-protein kinase ErbB3, belongs to the ErbB receptor family, encompassing EGFR (ErbB1), Human Epidermal Growth Factor Receptor (HER)-2 (ErbB2), and HER4 (ErbB4). HER3 activation contributes to the enhanced growth and advancement of cancer cells due to its role in activating the PI3K/AKT/mTOR and JAK–STAT signaling pathways. HER3 has been reported to be expressed in 83% of NSCLC tumors9 and in 85%–100% of tumors with a concurrent activating EGFR mutation.10,11 It is vital in the development of resistance to EGFR TKIs in EGFR-mutated NSCLC. For instance, aberrant mechanisms, such as MET amplification, can interact with HER3 to activate the PI3K/AKT/mTOR signaling pathway in tumor cells during EGFR TKI therapy.11 Furthermore, the ligand for HER3, heregulin, can induce the coupling and signaling of HER2/HER3, promoting cancer cell survival independently of EGFR.12 This independent pathway underscores the importance of HER3 as a target for oncological therapy, especially in EGFR-mutated NSCLC.13,14 It is reported that HER3 levels are elevated in patient tumor samples following progression from initial EGFR TKIs.11,15

Patritumab deruxtecan is a conjugate that combines patritumab, a monoclonal antibody targeting HER3, with deruxtecan - A payload of topoisomerase I inhibitor connected through a tetrapeptide-based cleavable linker. This design enables the targeted delivery of the chemotherapeutic agent directly to cancer cells, thus enhancing efficacy while minimizing systemic toxicity. A Phase I study (U31402-A-U102) demonstrated that 39% of heavily pretreated patients with EGFR-mutated NSCLC achieved an objective response to HER3-DXd.10 The mPFS reached 8.2 months (Table 1). HER3-DXd demonstrated anticancer effects across different resistance mechanisms to EGFR TKI, such as EGFR C797S mutation and MET. These encouraging results from the phase I trial prompted the start of the phase II HERTHENA-Lung01 trial.

Table 1 Efficacy and Safety of HER3-DXd Treating EGFR Mutated NSCLC Patients

HERTHENA-Lung01 Trial

In this global phase II trial,16 225 patients who had previously undergone at least one EGFR TKI and one PBC were treated with patritumab deruxtecan. At 18.9 months, the ORR was observed to be 29.8%, and the median duration of response was 6.4 months.16 The trial reported a mPFS of 5.5 months and a median overall survival (OS) of 11.9 months.16 Anticancer effects were noted across various subgroups as well. Notably, the outcomes for patients with central nervous system (CNS) metastases (N = 115) were comparable to those without CNS involvement. Tumor tissue analysis from 197 patients showed no significant variations in ORR based on HER3 expression. Specifically, 2 out of 7 patients (28.6%) with undetectable HER3 expression experienced confirmed partial responses. Sustained responses to HER3-DXd were evident in patients whose tumors exhibited EGFR C797X or MET amplification, representing the predominant resistance mechanisms to third-generation EGFR TKIs.17 The occurrence of treatment-related adverse events (TRAEs) of Grade 3 or higher was 64.9%, and 28.9% of patients encountered at least one Grade 4 TRAE (Table 1).

Discussion

Results from this phase II trial highlight the promising safety profile and effectiveness of the HER3-targeted ADC, patritumab deruxtecan, in EGFR-mutated NSCLC population who failed TKIs and PBCs. Both the HERTHENA-Lung01 and U102 Phase I study10 demonstrated clinical benefits across various subgroups, regardless of tumor HER3 expression, different resistance mechanisms to EGFR TKIs, and whether or not there were brain metastases. Despite these similarities, notable variations in the ORR and PFS were noted between the two studies (Table 1). The investigators suspect that these variations might be attributed to several factors, such as the smaller number of patients in the phase I study, differences in the demographic distribution of Asian versus non-Asian patients, and the number of previous treatment lines administered to the patients.16 CNS metastases are observed in approximately 70% of patients with advanced EGFR-mutated NSCLC.18 In this context, HER3-DXd has been effective, showing clinically meaningful responses in treating NSCLC with brain metastases. Increasing evidence is highlighting the potential for intracranial responses with large-molecule therapies.19

Ongoing Trials for HER3-DXd

The Phase 3 HERTHENALung02 trial is currently in progress, enrolling approximately 560 patients with locally advanced or metastatic non-squamous NSCLC harboring an EGFR-activating mutation. These patients, having undergone one or two lines of approved EGFR TKI treatment (including a third-generation EGFR TKI), are being randomized in a 1:1 ratio to receive either HER3-DXd or PBC. The primary endpoint is PFS, and the main secondary endpoint is OS. The trial commenced enrollment in August 2022, is expected to be completed by August 2026.20

Preclinical studies indicate that while resistance mechanisms to EGFR TKIs may not result in changes to HER3, the inhibition of EGFR can provoke a compensatory increase in HER3 membrane expression. Consequently, targeting HER3 could potentially enhance the efficacy of EGFR TKI therapy.21 A Phase 1 trial is currently exploring the combination of HER3-DXd and osimertinib as a first-line therapy for NSCLC patients with EGFR mutations.22

FLAURA-2, MARIPOSA, and MARIPOSA-2

The treatment landscape for EGFR-mutated NSCLC has significantly evolved over recent decades. Initially, standard chemotherapy provided a PFS of 4–6 months. The introduction of first- and second-generation EGFR TKIs extended PFS to 10–15 months.23,24 The latest advancement, the third-generation EGFR TKI osimertinib, further improved PFS to 18.9 months.25

Current clinical trials, including HERTHENA-Lung01 and HERTHENA-Lung02 studies, primarily focus on overcoming EGFR resistance after first-line treatment failure. In contrast, trials like FLAURA-2 and MARIPOSA aim to enhance first-line therapy to improve initial PFS (Table 2). The FLAURA-2 trial2 (a global Phase III trial) has shown that first-line therapy combining osimertinib with chemotherapy significantly extends PFS compared to osimertinib monotherapy in advanced NSCLC patients with EGFR mutations (29.4 vs 19.9 months, hazard ratio [HR] = 0.62). The regimen involves four cycles of chemotherapy (pemetrexed and either cisplatin or carboplatin) with osimertinib followed by maintenance therapy with pemetrexed and osimertinib, which suggests that early integration and consistent administration of chemotherapy enhance clinical outcomes by delaying disease progression and improving overall response rates. The FLAURA-2 regimen has subsequently been FDA approved in February 2024. MARIPOSA is another global phase III study, where EGFR mutated NSCLC patients were randomized in a 2:2:1 ratio, to receive a combination of amivantamab + lazertinib, osimertinib, or lazertinib.4 Lazertinib, an integral part of this trial, is a highly selective, third-generation TKI with strong CNS penetration, known for its effectiveness in addressing activating EGFR mutations and overcoming T790M resistance.26,27 Meanwhile, amivantamab, a bispecific antibody targeting EGFR and MET with activity directed at immune cells,28–30 has been approved for treating patients with EGFR exon 20 insertion mutations. PFS was substantially extended with amivantamab + lazertinib versus osimertinib (23.7 vs 16.6 months). The combination of amivantamab and lazertinib was associated with a higher incidence of treatment-emergent adverse events compared to osimertinib alone. For instance, 15% of patients in the combination group experienced grade 3 or higher rash, while only 1% of patients in the osimertinib group faced the same severity. Additionally, treatment-related adverse events leading to discontinuation occurred in 10% of patients treated with amivantamab and lazertinib, compared to 3% with osimertinib. These findings underscore the importance of carefully managing and understanding adverse reactions in combination therapies to optimize patient outcomes. FLAURA-2 and MARIPOSA trials affirm the effectiveness of augmenting initial therapy to delay EGFR-TKI resistance in NSCLC. Data show that incorporating agents like amivantamab or chemotherapy with third-generation EGFR TKIs extends the pre-resistance period by 8.8–9.5 months in FLAURA-2 and 7.1 months in MARIPOSA, highlighting a significant enhancement in disease management.

Table 2 Efficacy and Safety Comparison Among FLAURA2 and MARIPOSA

Not to be confused with MARIPOSA is the MARIPOSA-231 trial, a study focusing on second-line treatment for advanced NSCLC with EGFR mutation post-osimertinib, revealed that PFS was significantly longer for amivantamab + chemotherapy and amivantamab + lazertinib+ chemotherapy versus chemotherapy (median of 6.3 and 8.3 versus 4.2 months, respectively). Patients with a history of brain metastasis who had not previously undergone brain radiation also experienced comparable improvements. The augmentation in intracranial PFS observed with amivantamab treatment might be ascribed to its direct antitumor effects or, alternatively, to its ability to modulate the immune system, thereby exerting an indirect therapeutic impact.

Is a Median PFS of 5.5 Months in HERTHENA-LUNG 01 Sufficient in Light of FLAURA-2 and MARIPOSA Studies?

The FLAURA 2 trial demonstrated a notable PFS of 29.4 months, whereas the combination therapy of osimertinib with lazertinib and PBC in the MARIPOSA 2 trial yielded an aggregated PFS of 27.2 months (calculated as 18.9 months for osimertinib monotherapy plus an additional 8.3 months from amivantamab + lazertinib +PBC). A significant consideration in this therapeutic landscape is the potential early exhaustion of PBC options. The FLAURA 2 and MARIPOSA 2 regimens, while effective, may deplete PBC as a viable treatment option in subsequent lines of therapy. In contrast, the strategy of employing osimertinib in combination with a HER3 Dxd offers a notable advantage by sparing patients from immediate PBC utilization. This approach is especially pertinent for patients ineligible for platinum therapy, or when the preference would be to preserve PBC for future lines of treatment (ie, low tumor burden) when resistance to other agents becomes predominant.

It is important to note that in this phase II trial (HERTHENA-LUNG 01), HER3-Dxd exhibited a PFS of 5.5 months in a cohort that had previously been treated with osimertinib and chemotherapy. Transitioning directly to HER3 Dxd following osimertinib treatment in a phase III setting (HERTHENA-Lung02) presents a substantial opportunity to surpass this PFS benchmark. Achieving a PFS exceeding 5.5 months with HER3 Dxd alone could potentially align with, or even exceed, outcomes observed in other trials. Specifically, to surpass the PFS outcomes of FLAURA 2, an extension of 9–10 months is required; for MARIPOSA, an improvement of 4–5 months; and for MARIPOSA 2, an increase of 8–9 months. Notably, even an incremental improvement in PFS over the FLAURA 2 or MARIPOSA-2 results could be clinically significant, offering a treatment option that conserves PBC for later use.

Optimizing Strategies During the Interim: Awaiting Further Data in Clinical Research

As oncologists, while awaiting for more comprehensive data, we can still make effective use of current research to guide our treatment approaches. Based on insights from the FLAURA-2 trial, integrating osimertinib with chemotherapy as a first-line treatment appears to enhance patient outcomes significantly. In cases where disease progression occurs, and specific resistance mutations such as the C797S mutations are identified, shifting to a 4th-generation EGFR inhibitor on a clinical trial could be a strategic choice. For other situations, considering combinations such as amivantamab+ lazertinib+ chemotherapy, or exploring the use of HER3-DXd, may offer viable alternatives. Nevertheless, a deeper understanding and additional data, especially from the ongoing HERTHENA-Lung02 will be crucial in further refining these treatment strategies and ensuring the best possible care for our patients.

Conclusion

The evolving landscape of EGFR-mutated NSCLC treatment, particularly in the context of resistance to EGFR TKIs, is marked by significant advancements and challenges. Although osimertinib is very effective as an initial treatment, the eventual development of resistance necessitates innovative therapeutic strategies. The integration of intensified initial treatments, for instance, integrating a third-generation EGFR TKI with chemotherapy (FLAURA2) or using an EGFR-MET bispecific agent (MARIPOSA) has shown promise. Moreover, the exploration of broader approaches, including chemotherapy, ADCs, and combination therapies, continues to be vital.

The role of HER3 in EGFR-TKI resistance in EGFR-mutated NSCLC, highlighted by the increased HER3 expression following EGFR TKI progression, has opened new therapeutic avenues. Patritumab deruxtecan, an ADC targeting HER3, has exhibited notable efficacy in heavily pretreated EGFR-mutated NSCLC patients as demonstrated in HERTHENA-Lung01 trial. The ongoing HERTHENALung02 trial is set to provide further insights, particularly in assessing the potential of HER3-DXd in extending PFS compared to standard treatments. This study, along with others like FLAURA-2, MARIPOSA, and MARIPOSA 2, will help to refine and optimize treatment strategies for EGFR-mutated NSCLC.

Abbreviations

ADC, Antibody–Drug Conjugate; BLA, Biologics License Application; CI, Confidence Interval; CNS, Central Nervous System; EGFR, Epidermal Growth Factor Receptor; FDA, US Food and Drug Administration; HER, Human Epidermal Growth Factor Receptor; HR, Hazard Ratio; ICI, Immune Checkpoint Inhibitor; mPFS, Median Progression-Free Survival; NSCLC, Non-Small Cell Lung Cancer; ORR, Objective Response Rate; OS, Overall Survival; PBC, Platinum-Based Chemotherapy; TKI, Tyrosine Kinase Inhibitor; TRAE, Treatment-Related Adverse Event.

Ethics Statement

This commentary is based on publicly available, de-identified clinical trial data and does not require IRB approval.

Funding

No funding was received.

Disclosure

MN reports honoraria from AstraZeneca, BMS, Daiichi Sankyo, Novartis, Lilly, Pfizer, EMD Serono, Regeneron, Genentech, Mirati, Takeda, Janssen, and Blueprint Medicine; consulting fees from Caris Life Sciences; travel support from AnHeart Therapeutics. The authors report no other conflicts of interest in this work.

References

1. Yang CJ, Hung JY, Tsai MJ, et al. The salvage therapy in lung adenocarcinoma initially harbored susceptible EGFR mutation and acquired resistance occurred to the first-line gefitinib and second-line cytotoxic chemotherapy. BMC Pharmacol Toxicol. 2017;18(1):21. doi:10.1186/s40360-017-0130-0

2. Planchard D, Janne PA, Cheng Y, et al. Osimertinib with or without Chemotherapy in EGFR-Mutated Advanced NSCLC. N Engl J Med. 2023;389(21):1935–1948. doi:10.1056/NEJMoa2306434

3. Cho BC, Felip E, Hayashi H, et al. MARIPOSA: phase 3 study of first-line amivantamab + lazertinib versus Osimertinib in EGFR-mutant non-small-cell lung cancer. Future Oncol. 2022;18(6):639–647. doi:10.2217/fon-2021-0923

4. Cho BC, Felip E, Spira A, et al. Amivantamab plus lazertinib versus osimertinib as first-line treatment in EGFR-mutated advanced NSCLC: primary results from MARIPOSA, a Phase 3, global, randomized, controlled trial. ESMO Congress Oct; Madrid, Spain; 2023.

5. Yang JC, Lee DH, Lee JS, et al. Pemetrexed and platinum with or without pembrolizumab for tyrosine kinase inhibitor (TKI)-resistant, EGFR-mutant, metastatic nonsquamous NSCLC: phase 3 KEYNOTE-789 study. J Clin Oncol. 2023;17(41):abstrLBA9000.

6. Mok T, Nakagawa K, Park K, et al. Nivolumab plus chemotherapy in epidermal growth factor receptor-mutated metastatic non-small-cell lung cancer after disease progression on epidermal growth factor receptor tyrosine kinase inhibitors: final results of checkMate 722. J Clin Oncol. 2024;2:JCO2301017.

7. Lu S, Wu L, Jian H, et al. Sintilimab plus chemotherapy for patients with EGFR-mutated non-squamous non-small-cell lung cancer with disease progression after EGFR tyrosine-kinase inhibitor therapy (ORIENT-31): second interim analysis from a double-blind, randomised, placebo-controlled, phase 3 trial. Lancet Respir Med. 2023;11(7):624–636. doi:10.1016/S2213-2600(23)00135-2

8. Chen G. IMpower151: phase III study of atezolizumab+bevacizumab+ chemotherapy in first-line metastatic nonsquamous NSCLC. Presented at: 2023 IASLC World Conference on Lung Cancer. Republic of Singapore, Singapore; 2023.

9. Scharpenseel H, Hanssen A, Loges S, et al. EGFR and HER3 expression in circulating tumor cells and tumor tissue from non-small cell lung cancer patients. Sci Rep. 2019;9(1):7406. doi:10.1038/s41598-019-43678-6

10. Janne PA, Baik C, Su WC, et al. Efficacy and Safety of Patritumab Deruxtecan (HER3-DXd) in EGFR Inhibitor-Resistant, EGFR-Mutated Non-Small Cell Lung Cancer. Cancer Discov. 2022;12(1):74–89. doi:10.1158/2159-8290.CD-21-0715

11. Yonesaka K, Tanizaki J, Maenishi O, et al. HER3 augmentation via Blockade of EGFR/AKT signaling enhances anticancer activity of HER3-targeting patritumab deruxtecan in EGFR-mutated non-small cell lung cancer. Clin Cancer Res. 2022;28(2):390–403. doi:10.1158/1078-0432.CCR-21-3359

12. Yonesaka K, Hirotani K, Kawakami H, et al. Anti-HER3 monoclonal antibody patritumab sensitizes refractory non-small cell lung cancer to the epidermal growth factor receptor inhibitor erlotinib. Oncogene. 2016;35(7):878–886. doi:10.1038/onc.2015.142

13. Haikala HM, Janne PA. Thirty Years of HER3: from basic biology to therapeutic interventions. Clin Cancer Res. 2021;27(13):3528–3539. doi:10.1158/1078-0432.CCR-20-4465

14. Kawakami H, Yonesaka K. HER3 and its ligand, heregulin, as targets for cancer therapy. Recent Pat Anticancer Drug Discov. 2016;11(3):267–274. doi:10.2174/1574892811666160418123221

15. Engelman JA, Zejnullahu K, Mitsudomi T, et al. MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling. Science. 2007;316(5827):1039–1043. doi:10.1126/science.1141478

16. Yu HA, Goto Y, Hayashi H, et al. HERTHENA-Lung01, a Phase II Trial of Patritumab Deruxtecan (HER3-DXd) in epidermal growth factor receptor-mutated non-small-cell lung cancer after epidermal growth factor receptor tyrosine kinase inhibitor therapy and platinum-based chemotherapy. J Clin Oncol. 2023;41(35):5363–5375. doi:10.1200/JCO.23.01476

17. Ramalingam SS, Cheng Y, Zhou C, Mechanisms of acquired resistance to first-line Osimertinib: preliminary data from the phase III FLAURA study. Ann Oncol. 2018;29:740.

18. Ge M, Zhuang Y, Zhou X, Huang R, Liang X, Zhan Q. High probability and frequency of EGFR mutations in non-small cell lung cancer with brain metastases. J Neurooncol. 2017;135(2):413–418. doi:10.1007/s11060-017-2590-x

19. Jerusalem G, Park YH, Yamashita T, et al. Trastuzumab deruxtecan in HER2-positive metastatic breast cancer patients with brain metastases: a DESTINY-Breast01 Subgroup Analysis. Cancer Discov. 2022;12(12):2754–2762. doi:10.1158/2159-8290.CD-22-0837

20. Mok T, Janne PA, Nishio M, et al. HERTHENA-Lung02: phase III study of patritumab deruxtecan in advanced EGFR-mutated NSCLC after a third-generation EGFR TKI. Future Oncol. 2023. doi:10.2217/fon-2023-0602

21. Haikala HM, Lopez T, Kohler J, et al. EGFR inhibition enhances the cellular uptake and antitumor-activity of the HER3 antibody-drug conjugate HER3-DXd. Cancer Res. 2022;82(1):130–141. doi:10.1158/0008-5472.CAN-21-2426

22. A phase 1 open-label study of patritumab deruxtecan in combination with osimertinib in subjects with locally advanced or metastatic EGFR-mutated non-small cell lung cancer. Report No.: NCT04676477; 2023. Available from: https://clinicaltrials.gov/ct2/show/NCT04676477. Accessed July 3, 2024.

23. Zhou C, Wu YL, Chen G, et al. Erlotinib versus chemotherapy as first-line treatment for patients with advanced EGFR mutation-positive non-small-cell lung cancer (OPTIMAL, CTONG-0802): a multicentre, open-label, randomised, phase 3 study. Lancet Oncol. 2011;12(8):735–742. doi:10.1016/S1470-2045(11)70184-X

24. Sequist LV, Yang JC, Yamamoto N, et al. Phase III study of Afatinib or cisplatin plus pemetrexed in patients with metastatic lung adenocarcinoma with EGFR mutations. J Clin Oncol. 2013;31(27):3327–3334. doi:10.1200/JCO.2012.44.2806

25. Soria JC, Ohe Y, Vansteenkiste J, et al. Osimertinib in untreated EGFR-mutated advanced non-small-cell lung cancer. N Engl J Med. 2018;378(2):113–125. doi:10.1056/NEJMoa1713137

26. Yun J, Hong MH, Kim SY, et al. YH25448, an Irreversible EGFR-TKI with potent intracranial activity in EGFR mutant non-small cell lung cancer. Clin Cancer Res. 2019;25(8):2575–2587. doi:10.1158/1078-0432.CCR-18-2906

27. Ahn MJ, Han JY, Lee KH, et al. Lazertinib in patients with EGFR mutation-positive advanced non-small-cell lung cancer: results from the dose escalation and dose expansion parts of a first-in-human, open-label, multicentre, phase 1–2 study. Lancet Oncol. 2019;20(12):1681–1690. doi:10.1016/S1470-2045(19)30504-2

28. Moores SL, Chiu ML, Bushey BS, et al. A novel bispecific antibody targeting EGFR and cMet Is Effective against EGFR inhibitor-resistant lung tumors. Cancer Res. 2016;76(13):3942–3953. doi:10.1158/0008-5472.CAN-15-2833

29. Vijayaraghavan S, Lipfert L, Chevalier K, et al. Amivantamab (JNJ-61186372), an Fc Enhanced EGFR/cMet bispecific antibody, induces receptor downmodulation and antitumor activity by monocyte/macrophage trogocytosis. Mol Cancer Ther. 2020;19(10):2044–2056. doi:10.1158/1535-7163.MCT-20-0071

30. Yun J, Lee SH, Kim SY, et al. Antitumor activity of amivantamab (JNJ-61186372), an EGFR-MET bispecific antibody, in diverse models of EGFR Exon 20 insertion-driven NSCLC. Cancer Discov. 2020;10(8):1194–1209. doi:10.1158/2159-8290.CD-20-0116

31. Passaro A, Wang J, Wang Y, et al. Amivantamab plus chemotherapy with and without lazertinib in EGFR-mutant advanced NSCLC after disease progression on Osimertinib: primary results from the phase III MARIPOSA-2 study. Ann Oncol. 2023;34:S1307. doi:10.1016/j.annonc.2023.10.063

Creative Commons License © 2024 The Author(s). This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution - Non Commercial (unported, v3.0) License. By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.