Yonsei Med J. 2022 Sep;63(9):799-805. English.
Published online Aug 18, 2022.
© Copyright: Yonsei University College of Medicine 2022
Review

Lazertinib: on the Way to Its Throne

Jiyun Lee, Min Hee Hong and Byoung Chul Cho
    • Lung Cancer Center, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea.
Received April 06, 2022; Revised June 26, 2022; Accepted July 29, 2022.

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (https://creativecommons.org/licenses/by-nc/4.0) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Abstract

Lazertinib is an oral, irreversible, third-generation epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI) that forms an irreversible covalent bond to the Cys797 residue in the ATP-binding site of the EGFR kinase domain and exhibits a high selectivity for sensitizing and T790M EGFR mutations. In January 2021, it was first approved for the treatment of advanced or metastatic non-small cell lung cancer (NSCLC) patients with EGFR T790M who had previously received EGFR TKI therapy based on LASER201, a phase I/II trial. At a recommended dose of 240 mg, lazertinib achieved an encouraging anti-tumor activity in both extra- and intracranial lesions. With a high half-maximal inhibitory concentration for EGFR wildtype tumors, it is anticipated to pose a lower risk of skin and cardiac adverse events compared to osimertinib. Lazertinib is currently being investigated as a monotherapy in first-line treatment and in combination with amivantamab under various settings. In this review, we systematically summarize the preclinical and clinical data of lazertinib and discuss future perspectives on the treatment of EGFR-mutant NSCLC.

Keywords
Lazertinib; non-small cell lung cancer; epidermal growth factor receptor; targeted therapy

INTRODUCTION

Treatment strategies for non-small cell lung cancer (NSCLC) haves transformed tremendously over the past decades. Surgical resection, radiation therapy, cytotoxic chemotherapies, immunotherapies, and targeted therapies have been adopted at different stages and according to the tumor biology. Among these advances, targeted therapy is the one which brought a paradigm shift in the treatment scheme for NSCLC patients with molecular alterations. It significantly improved response rates and survival outcomes for selected patients, compared to conventional cytotoxic chemotherapeutic agents.

Epidermal growth factor receptor (EGFR) is a transmembrane protein with cytoplasmic kinase activity that transduces signals leading to increased cell proliferation, angiogenesis, metastasis, and decreased apoptosis.1 Mutations in the EGFR tyrosine kinase domain lead to constitutive activation of its kinase activity and activation of downstream signaling pathways, including PI3K/AKT/mTOR and Ras/Raf/MEK/ERK, which result in the apoptosis prevention or cell cycle progression.1, 2

Despite the advent of EGFR tyrosine kinase inhibitors (TKI), early clinical trials without patient selection based on biomarkers have failed to demonstrate any superiority in survival outcomes over cytotoxic chemotherapy.3 Their therapeutic capabilities were only visible after the identification of patients harboring activating EGFR mutations who demonstrate a unique susceptibility to EGFR TKIs.4, 5

Resistance to first-[erlotinib, gefitinib, and icotinib (icotinib is approved only in China)] or second-generation (afatinib, dacomitinib) EGFR TKIs typically develops in 50%–80% of initially responding patients after a median progression-free survival (PFS) of 9–13 months.6 Among the many resistance mechanisms involved, the emergence of a secondary mutation in the gatekeeper residue T790M is the most commonly encountered, accounting for 50%–60% of patients who are resistant to first/second-generation EGFR-TKIs.7, 8 The T790M mutation leads to impaired binding of TKI via steric hindrance.9, 10 As such, targeted therapy in EGFR-mutant NSCLC patients has been dominated by the third-generation TKI osimertinib,11, 12, 13 which irreversibly and selectively inhibits both EGFR-sensitizing and T790M mutations.14 Nevertheless, the emergence of treatment resistance, a higher risk of cardiac toxicity, and the “financial burden” of osimertinib dictated a need for the development of novel third-generation EGFR TKI, which leads us to lazertinib.

INTRODUCTION TO LAZERTINIB

Lazertinib (YH-25448, JNJ-73841937, Leclaza™) is an oral, irreversible, third-generation EGFR-TKI developed for the treatment of EGFR-mutant NSCLC (Fig. 1). It irreversibly binds to the Cys797 residue in the ATP-binding site of the EGFR kinase domain via covalent bond formation and exhibits high selectivity for sensitizing and T790M EGFR mutations while sparing EGFR WT tumors.15

Fig. 1
Chemical structures of lazertinib and osimertinib.

In January 2021, lazertinib was first approved in Korea for the treatment of advanced or metastatic NSCLC patients with EGFR T790M who had previously received EGFR TKI therapy at a recommended dose of 240 mg, taken orally once daily as tablets and without regard to food.16

PRECLINICAL STUDY

In its preclinical study, lazertinib significantly reduced the viability of Ba/F3 cells harboring Del19, L858R, Del19/T790M, and L858R/T790M mutations, with mean half-maximal inhibitory concentration (IC50) values ranging from 3.3–5.7 nmol/L,15 similar to those obtained from osimertinib (IC50 3.5–4.3) but lower than those obtained from gefitinib (IC50 10.2–7625.2). Additionally, lazertinib demonstrated a higher IC50 for EGFR WT, compared to osimertinib (722.7 nmol/L vs. 519.1 nmol/L).

Compatibly, a cell-free in vitro kinase inhibition assay has demonstrated that lazertinib inhibits the kinase activities of mutant EGFRs, including Del19, L858R, and T790M, with IC50 values ranging between 1.7–20.6 nmol/L, and also uncommon EGFR mutants, such as G719X and L861Q. WT EGFR was less affected by lazertinib than osimertinib (IC50 60 nmol/L vs. 20 nmol/L).15

The strong tumor inhibitory capability of lazertinib was robustly replicated in PDX models.15 Lazertinib demonstrated a superior tumor regression percentage than osimertinib (86.85% vs. 7.24% using 3 mg/kg lazertinib and osimertinib, respectively) in H1975 tumor-bearing mice models without any compromise in body weight. At a higher dose of 10 mg/kg (clinically equivalent to 240 mg once daily), lazertinib achieved a near-complete tumor regression (90%).

Lazertinib suppressed EGFR downstream pathways, including phospho-EGFR, phospho-AKT, and phospho-ERK, with an inhibitory capacity comparable to or higher than that of osimertinib.15 Once again, lazertinib did not inhibit EGFR signaling in a WT EGFR cell line.

Lastly, mouse models with intracranial implantation of H1975 cells were used to evaluate the efficacy of lazertinib in tumors with brain metastases.15 Intracranial tumors demonstrated a higher uptake of lazertinib than plasma or brain tissue at a single dose of 10 mg/kg. The relative exposure ratios of brain-to-plasma, intracranial tumor-to-plasma, and intracranial tumor-to-brain were 0.9, 7.0, and 7.9, respectively. Lazertinib was determined not to be a BCRP substrate, but only a weak substrate of MDR1 and was less affected by efflux transporters and maintained higher drug concentrations in intracranial tumors. In in vivo models, lazertinib more effectively inhibited intratumor growth than osimertinib, with a significantly longer survival duration (the median survival duration of mice, 124 days vs. 65 days at 10 mg/kg).

CLINICAL STUDY

LASER201: a first-in-human phase I/II study

LASER201 (ClinicalTrials.gov Identifier: NCT03046992), a multicenter, open-label, phase I/II study, was conducted to evaluate the efficacy of lazertinib in advanced NSCLC patients with activating EGFR mutations (L858R, 19del, G719X, and L861Q) who had progressed on first- or second-generation EGFR TKI.17, 18 The study consisted of three parts: part A (dose-escalation), part B (dose-expansion), and part C (dose-extension in first- and second-line cohorts). It was conducted across 17 centers in South Korea (Part D is being conducted in countries other than Korea, including Caucasians).

No dose-limiting toxicity occurred in the dose-escalation part (doses of 20, 40, 80, 120, 160, 240, and 320 mg), and the dose-expansion cohorts were opened for five doses between 40 and 240 mg. Ultimately, a dose of 160 mg or higher was considered necessary to achieve sufficient and consistent target inhibition, and a dose of 240 mg once daily was selected for dose extension.18

In pooled analysis of 76 patients with T790M mutation in the second-line cohort, the median duration of lazertinib treatment was 13.3 months (range, 0.3–37.4).17 The objective response rate (ORR) was 55.3% [95% confidence interval (CI), 44.1–66.4], including one complete response (1.3%) and 41 partial responses (53.9%), and the disease control rate (DCR) was 89.5% (95% CI, 82.6–96.4) with 26 patients achieving stable disease (Table 1). The median duration of response (DoR) was 17.7 months [95% CI, 9.9–not reached (NR)]. The median PFS and overall survival were 11.1 months (95% CI, 5.5–16.4) and NR, respectively, at a median follow-up of 22.0 months.

Table 1
Clinical Efficacies of Lazertinib and Osimertinib

Additionally, although the patient numbers were limited, seven patients with measurable intracranial lesions demonstrated a confirmed intracranial ORR and DCR of 85.7% and 100.0%, respectively, with a median intracranial DoR of 15.1 months (95% CI, 2.8–NR) and intracranial PFS of 26.0 months (95% CI, 5.4–NR).17

The safety profiles were largely compatible with previous EGFR TKI studies.11, 19, 20 The most common treatment-emergent adverse events of any grade were rash (37.2%), pruritus (34.6%), and paresthesia (33.3%),17 mostly consistent with those reported from the AURA3 trial (Table 2).21 The incidence of paresthesia was higher than that reported for osimertinib, although the underlying cause remains to be determined. A comparison of adverse events between lazertinib and osimertinib is discussed below.

Table 2
Incidence of Treatment-Related Adverse Events

Ongoing studies

LASER301 is a global, phase III trial aiming to compare the efficacy of lazertinib to gefitinib as a first-line treatment for locally advanced or metastatic EGFR-mutant NSCLC patients (NCT04248829).22 The results are highly anticipated to uncover whether lazertinib can demonstrate superiority over first-generation TKI to secure its place in the first-line treatment (Table 3).

Table 3
Ongoing Studies with Lazertinib as a Single Targeted Agent

ABLATE (NCT05167851) is a two-arm, phase II trial which evaluates the safety and efficacy of lazertinib with or without early local ablative radiation therapy in metastatic EGFR-mutant NSCLC patients with synchronous oligometastatic lesions.23 LU21-16 (NCT05277701) is another phase II trial aimed to discover the efficacy of lazertinib in NSCLC patients with uncommon EGFR mutations.24

Several studies are ongoing to evaluate the intracranial efficacy of lazertinib based on its favorable blood-brain barrier (BBB) penetrating efficacy observed in preclinical and clinical studies. LU20-15 is recruiting EGFR-mutant NSCLC patients with brain metastases who had progressed on prior EGFR TKI treatments.25 LU21-17 is assessing the efficacy of lazertinib combined with pemetrexed/carboplatin on EGFR-mutant NSCLC patients with asymptomatic brain metastases who had failed on osimertinib.26 LU21-01 is designed to combine lazertinib with pemetrexed for EGFR-mutant NSCLC patients with leptomeningeal carcinomatosis.27

Combination approaches: amivantamab

Amivantamab (amivantamab-vmjw; Rybrevant™) is a bispecific monoclonal antibody targeting EGFR and mesenchymal-epithelial transition factor (MET).28 By binding to extracellular domains of each receptor, amivantamab can inhibit ligand binding, promote receptor-antibody complex endocytosis and degradation, and induce Fc-dependent trogocytosis by macrophages and antibody-dependent cellular cytotoxicity by natural killer cells.29 Synergistic inhibition of the EGFR by targeting the receptor’s extracellular domains with amivantamab and the kinase domain with lazertinib hypothetically could elicit more potent inhibition of the EGFR pathway and could hold the potential to delay resistance.30

Based on this rationale, the efficacy of lazertinib in combination with amivantamab for EGFR-mutant NSCLC patients is being evaluated in several studies.

CHRYSALIS-1 (NCT02609776) has recruited patients with EGFR-mutant NSCLC who had progressed on available standard treatments. Part 1 dose-escalation determined the recommended phase 2 combination dose of 1050 mg amivantamab (1400 mg if bodyweight ≥80 kg) and 240 mg lazertinib.31 A distinct adverse event related to amivantamab was infusion-related reactions, mostly occurring on C1D1, which were managed by splitting the first dose (half dose on C1D1 and remainder on C1D2) and pre-medication with steroids, anti-histamine, and antipyretics.32 The combination therapy group reported higher incidences of rash (78%) and paronychia (42%), compared to the LASER201 group.17, 31 An interim analysis reported an improved outcome in the amivantamab+lazertinib combination group, compared to the amivantamab monotherapy group, for patients who had progressed on osimertinib but were chemotherapy-naïve: ORR of 36% (95% CI, 22–51) vs. 19% (95% CI, 12–27) and DoR of 9.6 months (95% CI, 5.3–NR) vs. 5.9 months (95% CI, 4.2–12.6), at a median follow-up duration of 11.1 and 6.9 months, respectively.33 Patients with osimertinib-resistance mutations or amplifications in EGFR/MET identified by next-generation sequencing in either ctDNA or tumor biopsy or high immunohistochemistry staining for EGFR/MET were more likely to benefit from combination therapy.34 However, further investigations are warranted on biomarker studies.

CHRYSALIS-2 (NCT04077463) is a phase I/Ib trial assessing the efficacy and safety of lazertinib alone (dose escalation) or in combination with amivantamab or combination with amivantamab and platinum doublet chemotherapy (dose expansion).30 Interim results have been published for cohort A, EGFR-mutant NSCLC patients who had progressed on osimertinib and platinum-based chemotherapy.35 Combination treatment with lazertinib plus amivantamab led to an ORR of 32% (95% CI, 16–52), and safety profiles were consistent with previous reports.33

MARIPOSA-1 (NCT04487080) is a multicenter, randomized, phase III trial comparing three experimental arms in treatment-naïve, EGFR-mutant NSCLC patients: amivantamab+lazertinib (arm A) vs. osimertinib (arm B) vs. lazertinib (arm C).36, 37 MARIPOSA-2 (NCT04988295) is combining platinum doublet chemotherapy to targeted agents in EGFR-mutant NSCLC patients who had progressed after osimertinib: amivantamab+lazertinib+pemetrexed/carboplatin (arm A) vs. pemetrexed/carboplatin (arm B) vs. amivantamab+pemetrexed/carboplatin (arm C).38, 39 The primary endpoint is the PFS in both studies, with planned sample sizes of 1000 and 500 patients, respectively.

The combination of lazertinib and amivantamab has shown encouraging activity in EGFR-mutant NSCLC patients in various settings. Final analyses on the combination approach (CHRYSALIS-1,2 and MARIPOSA-1,2) are highly anticipated.

OSIMERTINIB–A FRIEND OR FOE?

EGFR is abundant in keratinocytes in the basal layer of the epidermis and undifferentiated keratinocytes proliferating in the external root sheath of hair follicles.40 Hence, EGFR inhibition with TKI treatments can severely disrupt the skin homeostasis and ultimately lead to cutaneous toxicity, such as xerosis, pruritus, acneiform or papulopustular eruption, nail changes, and paronychia.41 Such adverse events have been reported with EGFR TKI treatments, including osimertinib and lazertinib.

In a preclinical study, lazertinib demonstrated a high IC50 for EGFR WT cells, implying that it poses a lower likelihood of manifesting off-target toxicities, including rash and paronychia.15 Lazertinib-treated mice showed minimal skin manifestation, while osimertinib-treated mice showed conspicuous skin changes, including keratosis on the face, neck, and abdomen.

Nonetheless, the actual incidence of skin-related adverse events, such as rash, pruritus, and paronychia, reported from patients did not differ between the two drugs (Table 2). Considering the interpretative challenges of cross-trial comparisons, further research on the incidence and management of adverse events related to lazertinib is warranted.

In addition, there are also concerns for cardiotoxicities, such as cardiac failure, atrial fibrillation, QT prolongation, myocardial infarction, and pericardial effusion, regarding osimertinib and other EGFR TKIs. Specifically, the risk of cardiac adverse events associated with osimertinib is a concern for clinicians.42, 43 The reported odds ratio for cardiac failure, atrial fibrillation, QT prolongation, myocardial infarction, and pericardial effusion when comparing osimertinib and other EGFR-TKIs were 2.2 (95% CI, 1.5–3.2), 2.1 (95% CI, 1.3–3.5), 6.6 (95% CI, 3.4–12.8), 1.2 (95% CI, 0.6–2.3), and 1.6 (95% CI, 0.8–3.3), respectively.44

Notwithstanding, the causal relationship between osimertinib and cardiac failure and the underlying mechanism thereof are not explicitly understood.45 A proposed mechanism suggests that the inhibition of ERBB2 or HER2 and the AMPK pathway leads to reduced contractility, mitochondrial energy depletion, and cardiomyocyte apoptosis. Meanwhile, researchers have shown that lazertinib selectivity for HER2 over EGFR was 275-fold, compared to 6.7-fold for osimertinib, which implies that lazertinib has lower cardiotoxicity.46

Consistent with this background, lazertinib has been found to exert minimal effects on cardiac function in both preclinical and clinical studies.46 No patients experienced clinically significant QT prolongation or decreased left ventricular ejection fraction. Considering that lazertinib demonstrated comparable anti-tumor efficacy to osimertinib, it may be considered a relatively safer option in terms of cardiotoxicity.47

Lastly, NSCLC patients with oncogenic-driven mutations such as EGFR are characterized by a high predilection for brain metastases, compared to unselected patients.48, 49 Brain metastasis is associated with deleterious effects on the patient’s quality of life and survival,50, 51 highlighting a need for novel TKI with improved intracranial efficacy. Although osimertinib has an improved BBB penetration, compared to early-generation EGFR TKIs,52 a significant proportion of patients (17%–20%) still experience disease progression in the brain after osimertinib treatment.53, 54 Lazertinib is not a substrate of BCRP and only a weak substrate of MDR1,55 as opposed to osimertinib, which is a substrate of both BCRP and MDR1.56 In LASER201, lazertinib demonstrated an encouraging intracranial activity with ORR of 87.5% and DCR of 100.0%, comparable to osimertinib from the AURA3 trial, which reported an intracranial ORR of 70.0% and DCR of 93.9%.52 Nonetheless, the sample sizes in these studies were limited, and real-world data with a larger number of patients on lazertinib should be investigated.

CONCLUSIONS

Despite the remarkable advances in targeted therapies for NSCLC with oncogenic mutations, the area of unmet need still remains. Lazertinib, a novel third-generation EGFR TKI, effectively inhibited extracranial lesions and demonstrated encouraging intracranial penetration and favorable safety profiles, including reduced skin or cardiac adverse events compared to osimertinib. Accordingly, it could be considered a sound option for the treatment of EGFR-mutant NSCLC patients. However, confirmatory phase 3 data or direct head-to-head comparisons with other EGFR TKIs are needed to finalize its position in the treatment scheme of EGFR-mutant NSCLC patients. The results from ongoing studies on front-line settings and in combination with amivantamab or cytotoxic chemotherapies are highly anticipated.

Notes

Byoung Chul Cho reports grants from Novartis, Bayer, AstraZeneca, MOGAM Institute, Dong-A ST, Champions Oncology, Janssen, Yuhan, Ono, Dizal Pharma, MSD, AbbVie, Medpacto, GI Innovation, Eli Lilly, Blueprint Medicines, and Interpark Bio Convergence Corp.; personal fees from Novartis, AstraZeneca, Boehringer-Ingelheim, Roche, BMS, Ono, Yuhan, Pfizer, Eli Lilly, Janssen, Takeda, MSD, Medpacto, Blueprint Medicines, TheraCanVac Inc, Gencurix Inc, Bridgebio Therapeutics, Kanaph Therapeutics Inc, Cyrus Therapeutics, Interpark Bio Convergence Corp., Guardant Health, Joseah BIO, and Champions Oncology; and other support from DAAN Biotherapeutics outside the submitted work. The remaining authors declare no conflicts of interest.

AUTHOR CONTRIBUTIONS:

  • Conceptualization: all authors.

  • Data curation: all authors.

  • Formal analysis: all authors.

  • Investigation: all authors.

  • Methodology: all authors.

  • Project administration: Byoung Chul Cho.

  • Resources: all authors.

  • Software: all authors.

  • Supervision: Min Hee Hong and Byoung Chul Cho.

  • Validation: all authors.

  • Writing—original draft: Jiyun Lee.

  • Writing—review & editing: all authors.

  • Approval of final manuscript: all authors.

References

    1. da Cunha Santos G, Shepherd FA, Tsao MS. EGFR mutations and lung cancer. Annu Rev Pathol 2011;6:49–69.
    1. Chang F, Steelman LS, Lee JT, Shelton JG, Navolanic PM, Blalock WL, et al. Signal transduction mediated by the Ras/Raf/MEK/ERK pathway from cytokine receptors to transcription factors: potential targeting for therapeutic intervention. Leukemia 2003;17:1263–1293.
    1. Maruyama R, Nishiwaki Y, Tamura T, Yamamoto N, Tsuboi M, Nakagawa K, et al. Phase III study, V-15-32, of gefitinib versus docetaxel in previously treated Japanese patients with non-small-cell lung cancer. J Clin Oncol 2008;26:4244–4252.
    1. Lynch TJ, Bell DW, Sordella R, Gurubhagavatula S, Okimoto RA, Brannigan BW, et al. Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. N Engl J Med 2004;350:2129–2139.
    1. Paez JG, Jänne PA, Lee JC, Tracy S, Greulich H, Gabriel S, et al. EGFR mutations in lung cancer: correlation with clinical response to gefitinib therapy. Science 2004;304:1497–1500.
    1. Karachaliou N, Fernandez-Bruno M, Bracht JWP, Rosell R. EGFR first- and second-generation TKIs-there is still place for them in EGFR-mutant NSCLC patients. Transl Cancer Res 2019;8 Suppl 1:S23–S47.
    1. Westover D, Zugazagoitia J, Cho BC, Lovly CM, Paz-Ares L. Mechanisms of acquired resistance to first- and second-generation EGFR tyrosine kinase inhibitors. Ann Oncol 2018;29 suppl_1:i10–i19.
    1. Takeda M, Nakagawa K. First- and second-generation EGFR-TKIs are all replaced to osimertinib in chemo-naive EGFR mutation-positive non-small cell lung cancer? Int J Mol Sci 2019;20:146
    1. Kobayashi S, Boggon TJ, Dayaram T, Jänne PA, Kocher O, Meyerson M, et al. EGFR mutation and resistance of non-small-cell lung cancer to gefitinib. N Engl J Med 2005;352:786–792.
    1. Pao W, Miller VA, Politi KA, Riely GJ, Somwar R, Zakowski MF, et al. Acquired resistance of lung adenocarcinomas to gefitinib or erlotinib is associated with a second mutation in the EGFR kinase domain. PLoS Med 2005;2:e73
    1. Mok TS, Wu YL, Ahn MJ, Garassino MC, Kim HR, Ramalingam SS, et al. Osimertinib or platinum-pemetrexed in EGFR T790M-positive lung cancer. N Engl J Med 2017;376:629–640.
    1. Soria JC, Ohe Y, Vansteenkiste J, Reungwetwattana T, Chewaskulyong B, Lee KH, et al. Osimertinib in untreated EGFR-mutated advanced non-small-cell lung cancer. N Engl J Med 2018;378:113–125.
    1. Wu YL, Tsuboi M, He J, John T, Grohe C, Majem M, et al. Osimertinib in resected EGFR-mutated non-small-cell lung cancer. N Engl J Med 2020;383:1711–1723.
    1. Cross DA, Ashton SE, Ghiorghiu S, Eberlein C, Nebhan CA, Spitzler PJ, et al. AZD9291, an irreversible EGFR TKI, overcomes T790M-mediated resistance to EGFR inhibitors in lung cancer. Cancer Discov 2014;4:1046–1061.
    1. Yun J, Hong MH, Kim SY, Park CW, Kim S, Yun MR, et al. YH25448, an irreversible EGFR-TKI with potent intracranial activity in EGFR mutant non-small cell lung cancer. Clin Cancer Res 2019;25:2575–2587.
    1. Dhillon S. Lazertinib: first approval. Drugs 2021;81:1107–1113.
    1. Cho BC, Han JY, Kim SW, Lee KH, Cho EK, Lee YG, et al. A phase 1/2 study of lazertinib 240 mg in patients with advanced EGFR T790M-positive NSCLC after previous EGFR tyrosine kinase inhibitors. J Thorac Oncol 2022;17:558–567.
    1. Ahn MJ, Han JY, Lee KH, Kim SW, Kim DW, Lee YG, 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;20:1681–1690.
    1. Maemondo M, Inoue A, Kobayashi K, Sugawara S, Oizumi S, Isobe H, et al. Gefitinib or chemotherapy for non-small-cell lung cancer with mutated EGFR. N Engl J Med 2010;362:2380–2388.
    1. Sequist LV, Yang JC, Yamamoto N, O’Byrne K, Hirsh V, Mok T, et al. Phase III study of afatinib or cisplatin plus pemetrexed in patients with metastatic lung adenocarcinoma with EGFR mutations. J Clin Oncol 2013;31:3327–3334.
    1. Papadimitrakopoulou VA, Mok TS, Han JY, Ahn MJ, Delmonte A, Ramalingam SS, et al. Osimertinib versus platinum-pemetrexed for patients with EGFR T790M advanced NSCLC and progression on a prior EGFR-tyrosine kinase inhibitor: AURA3 overall survival analysis. Ann Oncol 2020;31:1536–1544.
    1. Clinical trial of YH25448(lazertinib) as the first-line treatment in patients with EGFR mutation positive locally advanced or metastatic NSCLC (LASER301) [Internet]. [accessed on 2022 March 28].
    1. The safety and efficacy of first-line lazertinib and locally ablative radiotherapy in patients with synchronous oligo-metastatic EGFR-mutant non-small cell lung cancer [Internet]. [accessed on 2022 March 28].
    1. Lazertinib for patients with NSCLC harboring uncommon EGFR mutations (LU21-16) [Internet]. [accessed on 2022 March 28].
    1. Phase II trial of lazertinib in patients with epidermal growth factor receptor sensitizing mutation positive, metastatic non-small cell lung cancer with asymptomatic or mild symptomatic brain metastases after failure of EGFR tyrosine kinase inhibitor [Internet]. [accessed on 2022 March 28].
    1. Phase II trial of lazertinib and pemetrexed/carboplatin combination in patients with epidermal growth factor receptor sensitizing mutation positive, metastatic non-small cell lung cancer with asymptomatic or mild symptomatic brain metastases after failure of osimertinib [Internet]. [accessed on 2022 March 28].
    1. Phase II study of lazertinib and pemetrexed in EGFR mutant NSCLC patients with leptomeningeal carcinomatosis [Internet]. [accessed on 2022 March 28].
    1. Syed YY. Amivantamab: first approval. Drugs 2021;81:1349–1353.
    1. Park K, Haura EB, Leighl NB, Mitchell P, Shu CA, Girard N, et al. Amivantamab in EGFR exon 20 insertion–mutated non–small-cell lung cancer progressing on platinum chemotherapy: initial results from the CHRYSALIS phase I study. J Clin Oncol 2021;39:3391–3402.
    1. Shu CA, Goto K, Cho BC, Griesinger F, Yang JCH, Felip E, et al. CHRYSALIS-2: a phase 1/1b study of lazertinib as monotherapy and in combination with amivantamab in patients with EGFR-mutant NSCLC. J Clin Oncol 2021;39 15_suppl:TPS9132
    1. Cho BC, Lee KH, Cho EK, Kim DW, Lee JS, Han JY, et al. 1258O Amivantamab (JNJ-61186372), an EGFR-MET bispecific antibody, in combination with lazertinib, a 3rd-generation tyrosine kinase inhibitor (TKI), in advanced EGFR NSCLC. Ann Oncol 2020;31 supplement 4:S813
    1. Park K, Sabari JK, Haura EB, Shu CA, Spira A, Salgia R, et al. 1247P Management of infusion-related reactions (IRRs) in patients receiving amivantamab. Ann Oncol 2021;32 supplement 5:S981–S982.
    1. Leighl NB, Shu CA, Minchom A, Felip E, Cousin S, Cho BC, et al. 1192MO Amivantamab monotherapy and in combination with lazertinib in post-osimertinib EGFR-mutant NSCLC: analysis from the CHRYSALIS study. Ann Oncol 2021;32 supplement 5:S951–S952.
    1. Bauml J, Cho BC, Park K, Lee KH, Cho EK, Kim DW, et al. Amivantamab in combination with lazertinib for the treatment of osimertinib-relapsed, chemotherapy-naïve EGFR mutant (EGFRm) non-small cell lung cancer (NSCLC) and potential biomarkers for response. J Clin Oncol 2021;39 15_suppl:9006
    1. Shu CA, Goto K, Ohe Y, Besse B, Park K, Wang Y, et al. 1193MO Amivantamab plus lazertinib in post-osimertinib, post-platinum chemotherapy EGFR-mutant non-small cell lung cancer (NSCLC): preliminary results from CHRYSALIS-2. Ann Oncol 2021;32 supplement 5:S952–S953.
    1. Cho BC, Felip E, Hayashi H, Thomas M, Lu S, Besse B, et al. MARIPOSA: phase 3 study of first-line amivantamab + lazertinib versus osimertinib in EGFR-mutant non-small-cell lung cancer. Future Oncol 2022;18:639–647.
    1. A study of amivantamab and lazertinib combination therapy versus osimertinib in locally advanced or metastatic non-small cell lung cancer (MARIPOSA) [Internet]. [accessed on 2022 March 28].
    1. Shreeve SM, Martinez M, Verheijen RB, Xie J, Sun T, Haddish-Berhane N, et al. P76.73 MARIPOSA: randomized phase 3 study of first-line amivantamab+lazertinib vs osimertinib vs lazertinib in EGFR-mutant NSCLC. J Thorac Oncol 2021;16 supplement 3:S620–S621.
    1. A study of amivantamab and lazertinib in combination with platinum-based chemotherapy compared with platinum-based chemotherapy in patients with epidermal growth factor receptor (EGFR)-mutated locally advanced or metastatic non-small cell lung cancer after osimertinib failure (MARIPOSA-2) [Internet]. [accessed on 2022 March 28].
    1. Li Y, Fu R, Jiang T, Duan D, Wu Y, Li C, et al. Mechanism of lethal skin toxicities induced by epidermal growth factor receptor inhibitors and related treatment strategies. Front Oncol 2022;12:804212
    1. Tseng LC, Chen KH, Wang CL, Weng LC. Effects of tyrosine kinase inhibitor therapy on skin toxicity and skin-related quality of life in patients with lung cancer: an observational study. Medicine (Baltimore) 2020;99:e20510
    1. Grela-Wojewoda A, Pacholczak-Madej R, Adamczyk A, Korman M, Püsküllüoğlu M. Cardiotoxicity induced by protein kinase inhibitors in patients with cancer. Int J Mol Sci 2022;23:2815
    1. Kunimasa K, Kamada R, Oka T, Oboshi M, Kimura M, Inoue T, et al. Cardiac adverse events in EGFR-mutated non-small cell lung cancer treated with osimertinib. JACC CardioOncol 2020;2:1–10.
    1. Anand K, Ensor J, Trachtenberg B, Bernicker EH. Osimertinib-induced cardiotoxicity: a retrospective review of the FDA adverse events reporting system (FAERS). JACC CardioOncol 2019;1:172–178.
    1. Ewer MS, Tekumalla SH, Walding A, Atuah KN. Cardiac safety of osimertinib: a review of data. J Clin Oncol 2021;39:328–337.
    1. Jang SB, Kim KB, Sim S, Cho BC, Ahn MJ, Han JY, et al. Cardiac safety assessment of lazertinib: findings from patients with EGFR mutation-positive advanced NSCLC and preclinical studies. JTO Clin Res Rep 2021;2:100224
    1. Ewer MS, Ewer SM. Perspective on the cardiotoxicity of third-generation targeted EGFRs in the treatment of NSCLC. JTO Clin Res Rep 2021;2:100233
    1. Ng DZ, Tan WL, Ong WS, Gan EM, Jain A, Tan EH, et al. 139PD: lifetime incidence of brain metastases (BM) in EGFR-mutant (M+) lung cancer treated with first-line EGFR TKIs. J Thorac Oncol 2016;11 supplement 4:S117
    1. Shin DY, Na II, Kim CH, Park S, Baek H, Yang SH. EGFR mutation and brain metastasis in pulmonary adenocarcinomas. J Thorac Oncol 2014;9:195–199.
    1. Rodrigus P, de Brouwer P, Raaymakers E. Brain metastases and non-small cell lung cancer. Prognostic factors and correlation with survival after irradiation. Lung Cancer 2001;32:129–136.
    1. Sperduto PW, Mesko S, Li J, Cagney D, Aizer A, Lin NU, et al. Survival in patients with brain metastases: summary report on the updated diagnosis-specific graded prognostic assessment and definition of the eligibility quotient. J Clin Oncol 2020;38:3773–3784.
    1. Wu YL, Ahn MJ, Garassino MC, Han JY, Katakami N, Kim HR, et al. CNS efficacy of osimertinib in patients with T790M-positive advanced non–small-cell lung cancer: data from a randomized phase III trial (AURA3). J Clin Oncol 2018;36:2702–2709.
    1. Schmid S, Klingbiel D, Aeppli S, Britschgi C, Gautschi O, Pless M, et al. Patterns of progression on osimertinib in EGFR T790M positive NSCLC: a Swiss cohort study. Lung Cancer 2019;130:149–155.
    1. Kim H, Han J. P51.04 Pattern of disease progression on osimertinib and subsequent treatment in patients with EGFR-mutated non-small cell lung cancer. J Thorac Oncol 2021;16 supplement 10:S1120
    1. Wang F, Zhou Q. The challenges of third-generation EGFR tyrosine kinase inhibitors in the therapy of advanced NSCLC. J Thorac Oncol 2022;17:481–486.
    1. Ballard P, Yates JW, Yang Z, Kim DW, Yang JC, Cantarini M, et al. Preclinical comparison of osimertinib with other EGFR-TKIs in EGFR-mutant NSCLC brain metastases models, and early evidence of clinical brain metastases activity. Clin Cancer Res 2016;22:5130–5140.

Metrics
Share
Figures

1 / 1

Tables

1 / 3

PERMALINK