The treatment of patients with non-small cell lung cancer carrying uncommon EGFR mutations, HER2 mutations, or brain metastases: a systematic review of pre-clinical and clinical findings for dacomitinib
Review Article

The treatment of patients with non-small cell lung cancer carrying uncommon EGFR mutations, HER2 mutations, or brain metastases: a systematic review of pre-clinical and clinical findings for dacomitinib

Li-Li Yang1#, Xiao-Zhen Luo1#, Ling-Ling Xie1, Xiao-Zhen Lei1, Jiang Zhu2

1Department of Medical Oncology, Chengdu Shangjinnanfu Hospital, West China Hospital of Sichuan University, Chengdu, China; 2Department of Medical Oncology, West China Hospital of Sichuan University, Chengdu, China

Contributions: (I) Conception and design: LL Yang, XZ Luo, J Zhu; (II) Administrative support: J Zhu; (III) Provision of study materials or patients: All authors; (IV) Collection and assembly of data: LL Yang, XZ Luo, LL Xie, XZ Lei; (V) Data analysis and interpretation: LL Yang, XZ Luo; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors.

#These authors contributed equally to this work.

Correspondence to: Jiang Zhu, MD, PhD. Department of Medical Oncology, West China Hospital of Sichuan University, No. 37, Guo Xue Xiang, Chengdu 610041, China. Email: zhujiang@wchscu.cn.

Background: Accumulating evidence has shown that dacomitinib has potential activities for patients with non-small cell lung cancer (NSCLC) harboring uncommon epidermal growth factor receptor (EGFR) mutations, human epidermal growth factor receptor 2 (HER2) mutations, or central nervous system (CNS) metastases.

Methods: This study aimed to give a systematic review on its potential applications in the above settings by searching MEDLINE/PubMed, Embase, Cochrane Library, American Society of Clinical Oncology.org, European Society for Medical Oncology.org, and ClinicalTrials.gov.

Results: The literature search yielded 649 publications in total. According to our findings, dacomitinib exhibited promising efficacy in patients with major uncommon EGFR mutations (including G719X, S768I, and L861Q). Both EGFR exon 20 insertional mutation (Ex20ins) and HER2 Ex20ins demonstrated significant internal heterogeneity in response to dacomitinib, among which specific subtypes (including EGFR D770delinsGY, A763_Y764insFQEA, and HER2 M774delinsWLV) were highly sensitive. Other uncommon EGFR mutations including 18del and L747P have also been shown responsive to dacomitinib. Interestingly, limited studies suggested dacomitinib application on certain first or third generation tyrosine kinase inhibitors (TKIs)’ resistant secondary mutations. Last but not least, both pre-clinical and clinical data indicated that dacomitinib has an encouraging intracranial tumor control ability, regardless of uncommon mutations.

Conclusions: Dacomitinib demonstrated good disease control on patients with NSCLC harboring major uncommon EGFR mutations and specific EGFR or HER2 mutation subtypes, and selective clinical application of dacomitinib is considerable in this setting, especially for those with intracranial metastases.

Keywords: Dacomitinib; lung cancer; uncommon mutations; brain metastases


Submitted Jan 23, 2023. Accepted for publication Jul 21, 2023. Published online Aug 22, 2023.

doi: 10.21037/tcr-23-95


Highlight box

Key findings

• The efficacy of dacomitinib against different uncommon EGFR and HER2 mutation subtypes is highly heterogeneous, and the highly selective clinical application of dacomitinib is necessary for this setting.

• Dacomitinib has demonstrated good intracranial tumor control and should be considered clinically.

What is known and what is new?

• Dacomitinib has been approved for first-line treatment for EGFR-mutated non-small cell lung cancer (NSCLC); however, evidence of dacomitinib for uncommon EGFR/HER2 mutations and brain metastases in NSCLC is currently limited.

• This study aimed to give a comprehensive review of its potential applications by compiling available publications.

What is the implication, and what should change now?

• Dacomitinib exhibited promising efficacy in patients with major uncommon EGFR mutations (including G719X, S768I, and L861Q).

• Dacomitinib showed an encouraging intracranial tumor control ability, regardless of uncommon mutations.


Introduction

Lung cancer remains one of the most common cancers with the second-highest morbidity and highest mortality worldwide (1), among which non-small cell lung cancer (NSCLC) accounts for nearly 80%. The development of tyrosine kinase inhibitors (TKIs) targeting epidermal growth factor receptor (EGFR) and other driver genes has greatly changed the landscape of advanced NSCLC treatment (2). Second-generation (2G) EGFR-TKIs play a distinctive role among EGFR inhibitors due to their broad inhibitory activity of human epidermal growth factor receptor (HER) family members including HER1 (also called EGFR), HER2, and HER4 (3-5). In the pooled analysis of Lux-Lung serial studies, afatinib demonstrated superior efficacy for major uncommon mutations, and it has been recommended as preferred choice for patients with NSCLC harboring major uncommon mutations by the national comprehensive cancer network guidelines (5,6). As another 2G TKI, dacomitinib also demonstrated promising efficacy on this subset of patients in pre-clinical studies and limited real-world studies (4,6,7). In addition, many studies have also shown the potential efficacy of dacomitinib for patients with HER2 mutations and brain metastases (4,8-11).

Dacomitinib is another broad-spectrum, irreversible, highly selective EGFR-TKI (12). The ARCHER 1050 study demonstrated the excellent efficacy of dacomitinib over gefitinib in the first-line treatment of patients with NSCLC harboring classic EGFR mutations (namely 19del and L858R) (13). However, patients with brain metastases were excluded from this experiment due to the poor penetration ability of the control group (gefitinib) into the blood-brain barrier and the lack of data on the inhibitory activity of dacomitinib on brain metastases (12). Moreover, patients with uncommon mutations were also excluded from this study. Hence, data on the efficacy of dacomitinib for patients with uncommon mutations or central nervous system (CNS) metastases are limited (4,11,14), which makes clinicians uncertain about using dacomitinib in these scenarios.

This systematic review intends to give a comprehensive summary on potential applications of dacomitinib for patients with NSCLC carrying uncommon mutations (including EGFR and HER2) or CNS metastases, hoping to be helpful for clinicians’ decision-making. We present this article in accordance with the PRISMA reporting checklist (available at https://tcr.amegroups.com/article/view/10.21037/tcr-23-95/rc).


Methods

Definition of study population and outcome

The study population was defined as advanced NSCLC patients with uncommon EGFR mutations, HER2 mutations, or CNS metastases. Uncommon EGFR mutations were defined as those mutations other than 19del and L858R in the EGFR domain as previously described in literature (2). CNS metastases referred to both parenchymal metastases and leptomeningeal metastases. The percentage of objective response rate (ORR), disease control rate (DCR), progression-free survival (PFS), and overall survival (OS) for each of the currently available publications.

Data sources and search strategy

A systematic literature review was performed on 1 August 2022 according to the PRISMA criteria of 2009. We reviewed MEDLINE/PubMed, Embase, Cochrane Library, the American Society of Clinical Oncology (ASCO) Meeting Library, the European Society of Medical Oncology (ESMO) Library, and ClinicalTrials.gov for citation or ongoing trials without time limitation. We also searched the Google Scholar (https://scholar.google.com) for additional reports. The terms used for the search were (lung cancer*[Title/Abstract]) AND (dacomitinib[Title/Abstract]) in PubMed, (‘lung cancer*’:ab,ti AND dacomitinib:ab,ti) in Embase, (lung cancer*):ti,ab,kw AND (dacomitinib):ti,ab,kw in Cochrane library and (dacomitinib, lung cancer) in ASCO and ESMO. The terms used for the search were (“lung cancer” as condition/disease) and (“dacomitinib” as other terms) in ClinicalTrials.gov. After removing duplicates, titles and abstracts were independently screened by two researchers (Yang LL and Luo XZ).

Selection criteria

The search criteria were limited to cell-line, animal, or human studies published in English language. Besides, to be included in this review, a publication had to fulfill the following inclusion criteria: studies performed in advanced NSCLC patients, or proper cell lines or animals, harboring uncommon EGFR mutations, HER2 mutations or CNS metastases, treated with dacomitinib monotherapy. We included studies and abstracts without time limitations.


Results

The literature search yielded 649 publications in total. After excluding duplicates and applying the selection criteria, 36 studies were included in this systematic review (Figure 1). Overall, the eligible reports included 7 prospective trials, 9 retrospective studies, 4 conference abstracts, and 16 preclinical studies (Tables 1-3). The main uncommon mutation sites involved in this systematic review were shown in the molecular simulation graph (Figures 2,3).

Figure 1 Flow chart of the study. ASCO, American Society of Clinical Oncology; ESMO, European Society for Medical Oncology.

Table 1

A summary of published clinical data on uncommon EGFR mutations

First author Type Year Region N Gender Age (year) Histology LOT Mutation subtype Method Dosage Best response ORR DCR PFS (months) OS (months)
Zhang (11) R 2021 China 1 Female 62 AC 1 G719A NGS 30 mg PR NA NA 6.6 NA
1 Female 64 AC 1 L861Q NGS 30 mg Non-CR/non-PD NA NA 10+ NA
Shen (15) R 2021 China 1 Female 64 AC 4 L858R + L718Q NGS 45 mg PR NA NA 5 NA
Li (16) R 2021 China 1 Female 38 AC 3 L858R + T790M + L792F + L792H NGS 30 mg PD NA NA 1 NA
1 Male 58 AC 3 L858R + L792V + L718Q NGS 30 mg PD NA NA 1 NA
1 Female 72 AC 3 L858R + L718Q NGS 30 mg PD NA NA 2 NA
1 Male 81 AC 4 L858R + T790M + L792H NGS 15 mg PD NA NA 1 NA
1 Male 45 AC 3 L858R + L718Q NGS 30 mg PD NA NA 1 NA
Chan (17) CA 2021 Singapore 1 Female 73 AC 5 L858R + L718Q NA 15 mg (occasionally 30 mg) Non-CR/non-PD NA NA 5+ NA
Reckamp (18) P 2014 USA 1 NA NA AC 1 G719C + S768I ARMS 45 mg PR NA NA 15.5 NA
Peng (14) R 2021 China 1 Female 50 AC 1 G719A + I706T NGS 45 mg PR NA NA 1+ NA
Park (19) P 2014 Korea 1 NA NA AC 2 G719X NA 45 mg PR NA NA NA NA
Morita (20) R 2021 Japan 1 Female 71 AC 6 G719A ARMS 45 mg→30 mg PR NA NA 7.8 10.8
Li (4) R 2022 China 11 NA NA AC 2–4 G719X, S768I, L861Q, L747P NGS NA NA 54.5% 81.8% 10.3 1-year OS rate of 90.0%
1 Male 46 AC 3 L858R + E709K NGS 30 mg PD NA NA 1.2 NA
1 Male 65 AC 2 19del + G724S NGS 30 mg PR NA NA 9.4+ NA
Kris (21) CA 2012 USA 7 NA NA NA 1 NA NA NA NA 28.6% 71.4% NA NA
Han (22) P 2021 China 30 NA NA NA NA NA NGS 45 mg NA NA NA NA NA
Choudhury (23) P 2021 USA 1 NA NA AC 2 G719A NGS 45 mg PR NA NA 17+ NA
2 NA NA AC 2 C797S + 19del, C797S + L858R NGS 45 mg SD NA NA NA NA
1 NA NA AC 2 19del + G724S NGS 45 mg SD NA NA 5+ NA
Biswas (24) CA 2021 India 2 NA NA NA NA G719X NGS NA PR NA NA NA NA
1 NA NA NA NA L861Q NGS NA PR NA NA NA NA
Jänne (25) P+ 2011 USA 1 NA NA AC NA delD770insGY ARMS 45 mg PR NA NA 12.4 NA
5 NA NA AC NA Ex20ins ARMS NA NA 20% 60% NA NA
Jänne (9) P 2014 USA 8 NA NA AC NA 1 E709A + G719S, 1 G719S + R776H, 1 S768I + L858R, 3 T790M, 2 Ex20ins ARMS NA NA 37.5% 75% 7.3 17.9
1 NA NA AC 1 E709A + G719S ARMS NA PR NA NA NA NA
2 NA NA AC 1 Ex20ins ARMS NA NA 0% 50% NA NA

EGFR, epidermal growth factor receptor; N, number; LOT, line of therapy; ORR, objective response rate; DCR, disease control rate; PFS, progression-free survival; OS, overall survival; R, retrospective study; AC, adenocarcinoma; NGS, next generation sequencing; PR, partial response; NA, not applicable/not available; CR, complete response; PD, progressive disease; CA, conference abstract; P, prospective trials; ARMS, amplification refractory mutation system; SD, stable disease; Ex20ins, exon 20 insertional mutation.

Table 2

A summary of published clinical data on HER2 alterations

First author Type Year Region N Gender Age (years) Histology LOT Mutation subtype Method Dosage Best response ORR DCR PFS (months) OS (months)
Reckamp (18) P 2014 USA 1 NA NA AC 2 HER2 amplication + T790M + 19del FISH 45 mg PD NA NA NA NA
1 NA NA AC 2 HER2 amplication + T790M + L858R FISH 45 mg SD NA NA NA NA
1 NA NA Non-AC 2 HER2 amplication FISH 45 mg PR NA NA 2.8 NA
Kris (10) P 2015 USA 26 15 were women NA AC 21 were pre-treated HER2-mutant NA 21 received 45 mg NA 11.5% 92.3% 3 9
1 NA NA AC NA M774delinsWLV NA NA PR NA NA 3+ 23+
1 NA NA AC NA P780_Y781insGSP NA NA PR NA NA 11 25+
1 NA NA AC NA P780_Y782insGSP NA NA PR NA NA 14 27
1 Male NA AC ≥1 HER2 amplication [17]* FISH 45 mg SD NA NA 5 22
1 Male NA AC ≥1 HER2 amplication [>2]* FISH 45 mg PD NA NA 1 15
1 Male NA AC ≥1 HER2 amplication [2]* FISH 45 mg SD NA NA 5 7
1 Male NA AC ≥1 HER2 amplication [2.4]* FISH 45 mg PD NA NA 1 5
Jänne (25) P 2011 USA 2 NA NA AC NA HER2 amplication NA NA SD NA NA NA NA
Kelly (26) R 2010 USA 1 Male 50 LCNEC 3 HER2 amplication [6.1]* FISH 45 mg PR NA NA 6 NA

*, the ratio of HER2/CEP17. HER2, human epidermal growth factor receptor 2; N, number; LOT, line of therapy; ORR, objective response rate; DCR, disease control rate; PFS, progression-free survival; OS, overall survival; P, prospective trials; NA, not applicable/not available; AC, adenocarcinoma; FISH, fluorescent in situ hybridization; PD, progressive disease; SD, stable disease; PR, partial response; R, retrospective study; LCNEC, large cell neuroendocrine carcinoma.

Table 3

A summary of published clinical data on CNS metastases

First author Type Year Region N Gender Age (years) Histology LOT Mutation subtype Method Dosage Best response for brain lesion(s) iORR iDCR ORR DCR PFS (months) OS (months)
Zhao (27) R 2021 China 1 Male 47 AC 1 19del NA 30 mg CR NA NA NA NA 11 NA
1 Male 55 AC 1 L858R NA 30 mg CR NA NA NA NA 8 NA
Zhang (11) R 2021 China 32 (30 were evaluable, 8 evaluable for brain) 19 were female Median: 57.5 31 were AC, 1 was ASC All TKI-naïve 25 L858R, 5 19del, 1 L861Q, 1 G719A 13 by ARMS, 19 by NGS Mostly (n=28) received 30 mg NA 87.5% (85.2%) 100% 66.70% 100% NR NA
1 Female 62 AC 1 G719A NGS 30 mg PR NA NA NA NA 6.6 NA
1 Female 64 AC 1 L861Q NGS 30 mg Non-CR/non-PD NA NA NA NA 10+ NA
Shen (15) R 2021 China 1 Female 64 AC 4 L858R + L718Q NGS 45 mg PR NA NA NA NA 5 NA
Chan (17) CA 2021 Singapore 1 patient with leptomeningeal metastases Female 73 AC 5 L858R + L718Q NA 15 mg (occasionally 30 mg) SD§ NA NA NA NA 5+ NA
Peng (14) R 2021 China 14 7 were female Median: 54 AC 1 5 L858R, 8 19del, 1 G719A + I706T NGS Mostly (n=9) received 30 mg NA 85.70% 100% 92.90% 100% NR NA
1 patient with leptomeningeal metastases Female 41 AC 1 L858R NGS 45 mg SD§ NA NA NA NA 4 NA
1 Female 50 AC 1 G719A + I706T NGS 45 mg PR NA NA NA NA 1+ NA
Mizusaki (28) R 2021 Japan 1 patient with leptomeningeal metastases Male 72 AC 3 19del NA 30 mg CR NA NA NA NA 2.1+ NA
Li (4) R 2022 China 23 (20 were evaluable) 14 were female Median: 57.5 AC 2–5 5 19del, 14 L858R, 4 G719X NGS Mostly (n=12) received 30 mg NA NA NA 15% 85% 6.5 (2.6–10.4) NA
Biswas (24) CA 2021 India 10 NA NA NA NA NA NA NA NA NA NA 80% NA NA NA

, this rate was evaluated by modified RECIST (1.1). In the modified RECIST (1.1), up to five intracranial and up to five extracranial target lesions were included; intracranial target lesions of between 5 and 40 mm in diameter were allowed; , as most patients with brain metastases in the later-line of therapy had received local therapy (e.g., radiotherapy), the assessment of iORR and iDCR was not performed; §, judged by RANO-LM criteria; , median (range). CNS, central nervous system; N, number; LOT, line of therapy; iORR, intracranial objective response rate; iDCR, intracranial disease control rate; ORR, objective response rate; DCR, disease control rate; PFS, progression-free survival; OS, overall survival; R, retrospective study; AC, adenocarcinoma; NA, not applicable/not available; CR, complete response; ASC, adenosquamous carcinoma; TKI, tyrosine kinase inhibitor; ARMS, amplification refractory mutation system; NGS, next generation sequencing; NR, not reached; PR, partial response; PD, progressive disease; CA, conference abstract; SD, stable disease; RECIST, Response Evaluation Criteria In Solid Tumors; RANO-LM, Response Assessment in Neuro-oncology, Leptomeningeal Metastasis.

Figure 2 Molecular simulation graphs and assessment of dacomitinib activity in pre-clinical setting (Ba/F3 cells) for major uncommon EGFR mutations (A,B), EGFR 20 exon insertion mutations (C,D), and other uncommon EGFR mutations (E,F). Molecular simulation graphs were remodeled in EGFR (PDB: 4I23) protein structure by PyMOL software (version 2.3.4, Schrödinger, Inc., New York, USA). The graphs only show the approximate spatial location of the mutations and do not represent the specific mutation structure. The graphs were colored according to the scheme indicated in the graph and the corresponding drug sensitivity was classified as highly sensitive (<1 nM), sensitive (1–10 nM), relatively sensitive (10–100 nM), resistant (100–1,000 nM), highly resistant (1,000 nM), and NA based on current literature data. *, data were shown as IC90; ^, cell model carrying L747P was A431. EGFR, epidermal growth factor receptor; NA, not available; PDB, Protein Data Bank; IC90, 90% inhibiting concentration.
Figure 3 Molecular simulation graphs and assessment of dacomitinib activity in pre-clinical setting (Ba/F3 cells) for HER2 alterations (A,B). Molecular simulation graphs were remodeled in HER2 (PDB: 3PP0) protein structure by PyMOL software (version 2.3.4, Schrödinger, Inc.). The graphs only show the approximate spatial location of the mutations and do not represent the specific mutation structure. The graphs were colored according to the scheme indicated in the graph and the corresponding drug sensitivity was classified as highly sensitive (<1 nM), sensitive (1–10 nM), relatively sensitive (10–100 nM), resistant (100–1,000 nM), highly resistant (1,000 nM), and NA based on current literature data. *, data were shown as IC90; ^, cell models carrying HER2 amplification, HER2 amplification, and M774_A775insYVMA were Calu-3, H1819, and H827, respectively. HER2, human epidermal growth factor receptor 2; PDB, Protein Data Bank; NA, not available; IC90, 90% inhibiting concentration.

Uncommon EGFR mutations

The most common mutations on the EGFR gene in NSCLC, including 19Del (49–72%) and L858R (28–43%), are called “common mutations”, while other mutations (10–20%) on EGFR are called “uncommon mutations” or “rare mutations”, with significantly heterogenic responses to EGFR-TKIs (2,29-32). According to the incidence and clinical significance, we mainly summarize EGFR mutations into the following three categories and a summary of the literature data is shown in Table 1.

Major uncommon mutations

Li et al. presented the later-line efficacy of dacomitinib on patients with NSCLC harboring uncommon EGFR mutations in a relatively small scale, and the findings confirmed ORRs of 57.1%, 0%, 33.3%, and median PFS (mPFS) of 10.3, 0.7, 6.5 months as to G719X (n=7), L861Q (n=2), and S768I (n=3) (Figure 2A), respectively (4). Other studies on patients carrying major uncommon mutations treated with dacomitinib were mostly case reports, of which G719X subtype was the most reported. We pooled 5 G719X-carrying cases with dacomitinib treatment responses and survival data in multi-line settings from the literature, and the results showed an ORR of 100% and an mPFS of 7.8 months (11,14,18,20,23). Consistent with clinical observations, the IC90 value of dacomitinib-treated Ba/F3 cell lines carrying the G719X mutation was only 6 nM, compared to that of 1.6 nM for classic mutation 19del (Figure 2B). Two cases carrying L861Q mutation treated with dacomitinib were reported in literature (11,24). Different from the two patients with primary drug resistance reported in Li et al.’s work (4), both the two patients had disease controlled, of which one patient achieved partial response (PR), and the other one with brain metastases had stable disease and achieved an PFS of more than 10 months.

Exon 20 insertion mutation (Ex20ins)

The Ex20ins is the second prevalent subtype (~16%) among uncommon EGFR mutations (2,33-37). In an open-label phase II clinical trial, two patients harboring Ex20ins (specific sites not known) were enrolled, with an ORR of 0% and a DCR of 50% (9). Results from a phase I clinical trial showed that one (carrying D770delinsGY) of the six patients with Ex20ins treated with dacomitinib achieved PR, and the overall ORR, DCR, and mPFS were 20%, 60%, and less than 3 months, respectively (25). Subsequent preclinical studies conducted by Kobayashi et al. (38) demonstrated that, though not sensitive to erlotinib and osimertinib, D770delinsGY and other insertions with a G770 equivalence were still responsive to 2G TKIs (especially for dacomitinib). Via establishing Ba/F3 pre-clinical models harboring Ex20ins, Kobayashi et al. revealed by the dose-response proliferation assays that D770delinsGY and A763_Y764insFQEA were two uniquely sensitive subtypes to dacomitinib [50% inhibitory concentration (IC50) <0.1 nM], but other subtypes [including A767_V769dupASV (namely V769_D770insASV), D770_N771insSVD, and H773_V774insH] were not (38), which were consistent with other preclinical findings (39-42) (Figure 2C,2D).

Secondary uncommon EGFR mutations

Secondary uncommon EGFR mutations after 1G/3G TKIs resistance mainly included C797S/G, L718Q/V, and L792F/H (Figure 2E), which were found to be more enriched in the 3G TKI osimertinib-resistant setting (43). Chan (17) and Shen et al. (15) respectively reported a case of an elderly woman, both of which were detected with L718Q mutation after osimertinib resistance. And their conditions were both controlled after receiving dacomitinib treatment (≥5 lines), among whom one of them achieved PR and both patients obtained the PFS of more than 5 months. An in vitro experiment conducted by Nishino et al. (44) also revealed that dacomitinib was effective against L718Q/V- or L792F/H-mutated (in cis with activating EGFR mutations and without T790 M) Ba/F3 cells with IC50 values <1 nM. Consistently, an in vitro study by Kobayashi et al. (45) demonstrated that L792F-mutated afatinib-resistant Ba/F3 cells were highly sensitive to dacomitinib (IC50 <10 nM) (Figure 2F). However, there are also conflicting results. In Li et al.’s case series study, five patients with later-line osimertinib-resistant lung cancer were treated with dacomitinib (≥3 lines), whose resistance mutations at EGFR L792 (in cis with T790 M) and/or L718 were detected by NGS; however, all patients progressed within 2 months. Dynamic molecular simulations showed that L792H (in cis with T790 M) and L718Q mutations may interfere with the binding of dacomitinib to EGFR, leading to primary drug resistance (16). We believe that complicated tumor heterogeneity after multi-line therapies, different drug dosages, and patient status may all affect dacomitinib performance, leading to different treatment outcomes.

Other uncommon EGFR mutations

Except for major uncommon mutations and Ex20ins, other uncommon mutations spread across all exons of EGFR, but mainly in exons 18–21. As the quite low incidence (~1%) and exceptionally broad and complex distribution, studies on these uncommon mutations are currently very limited (2). From a therapeutic point of view, this part of mutations can be divided into primary uncommon mutations [E709X, delE709_T710insD (also called 18del), L747P/S, etc.] and secondary uncommon mutations (C797S/G, L718Q/V, L792F/H, etc.) (Figure 2E).

The E709X mutation (~0.3%) generally occurs as part of a complex mutation (2). Preclinical studies by Kobayashi et al. (46) showed that this mutant site was most sensitive to afatinib (IC90: 0.7 nM), followed by neratinib (IC90: 6 nM) and dacomitinib (IC90: 16 nM), whereas resistant to osimertinib and 1G TKIs (Figure 2F). In the afatinib uncommon mutations database (https://www.uncommonegfrmutations.com/) reported by Yang et al., nine patients (all in complex form and four of them were treatment-naïve) with E709X were included, with an ORR of 33.3% and a DCR of 55.6% (47). Currently, clinical data on dacomitinib for E709X are scarce. Li et al. reported that a patient carrying E709K combined with L858R mutation developed primary drug resistance after third-line dacomitinib treatment, and the PFS lasted only 1.2 months (4). Another patient with E709A combined with G719S mutation reported by Jänne et al. received first-line dacomitinib and responded (9). Currently, clinical data on dacomitinib treatment for delE709_T710insD (namely 18del) are lacking. Preclinical data suggested that delE709_T710insD was potentially sensitive to dacomitinib (IC50: 1–10 nM) (Figure 2F) (41). In addition, 16 patients with delE709_T710insD mutation (14 of them were treatment-naïve) were identified in the afatinib uncommon mutations database, and 13 of them responded, which is consistent with preclinical findings (41).

The L747P missense mutation, located in exon 19 of EGFR, is rarely observed (~0.59%) in NSCLC (48,49). Current studies revealed that this mutation presented primary resistance to 1G-TKIs with the PFS ranging from 0.5–2.9 months, and its sensitivity to 3G TKI osimertinib remains unclear. However, numerous studies have demonstrated better therapeutic responsiveness and efficacy of 2G TKI (afatinib) for this rare mutation, with a much longer PFS ranging from 12 to 24 months (48,49). In Li et al.’s study, two patients carrying L747P who were treated with later-line dacomitinib (≥3 lines) were enrolled. Both patients were in remission (PR) and continued to benefit, with the PFS of 6.6 and 9.1 months, respectively (4). In line with Li et al.’s clinical results, in the study by Yang et al. (48) and Yoshizawa et al. (49), A431 cells or Ba/F3 cells carrying L747P also demonstrated high sensitivity to dacomitinib, with IC50 values of 1.8 and 5.2 nM for dacomitinib (Figure 2F) relative to IC50 values of 45.3 and 147.3 nM for gefitinib, respectively. Besides, via instructing an L747P-mutant patient-derived xenograft mice model, Yang et al. showed that both dacomitinib and afatinib showed potent anti-tumor activities compared with osimertinib and poziotinib (48). Interestingly, compared with afatinib, dacomitinib also significantly reduced mice weight (P<0.001) and induced severe skin destruction (48). Nevertheless, the two patients carrying L747P in Li et al.’s work who received dacomitinib developed only grade 1–2 rash and grade 1 oral ulcer (4).

HER2 mutations

Different from the increase of gene copy number in breast cancer, variations of HER2 (2–5%) in NSCLC mainly occur in the kinase region (most of them are HER2 Ex20ins, accounting for ~56%) (50). The most common HER2 Ex20ins include A775_G776insYVMA, G776delinsVC and P780_Y781insGSP (Figure 3A), which make up ~94% of HER2 Ex20ins according to previously published works (10,39).

The phase II clinical trial conducted by Kris et al. (10) disclosed that among 26 patients (17% of them were treat-naïve) carrying HER2 mutations (25 Ex20ins and 1 missense mutation) treated with dacomitinib, their ORR, DCR, mPFS, and median OS (mOS) were 11.5%, 92.3%, 3 and 9 months, respectively. Specifically, among three patients who obtained PR, two patients with P780_Y781insGSP subtype had the longest PFS (11 and 14 months) and OS (25+ and 27 months), and one patient with M774delinsWLV had a PFS of 3+ months and an OS of 23+ months. Consistent with the data of Kris et al., preclinical studies conducted by Koga et al. and Kosaka et al. suggested that Ba/f3 cells carrying P780_Y781insGSP or M774delinsWLV were highly sensitive to dacomitinib, with IC50 values of 1–10 and <1 nM (Figure 3B), respectively (39,51,52). In addition, despite the lack of clinical data, preclinical studies suggested that both G776delinsVC (39,51,52) and G778_S779insCPG (39) subtypes were sensitive to dacomitinib (IC50: 1–10 nM), suggesting the potential of dacomitinib for patients carrying these mutations. According to preclinical data, as the most common HER2 mutation, A775_G776insYVMA subtype was moderately sensitive to dacomitinib (IC50: 10–100 nM); however, no PRs (ORR: 0%, DCR: 92.3%) were observed in 13 pre-treated patients carrying A775_G776insYVMA reported by Kris’ et al. (10). More clinical data are needed to confirm the potential of dacomitinib for patients carrying this mutation (39,51,52).

For HER2 amplification, in Kris et al.’s study, four patients with HER2 amplification had an ORR of 0%, and a DCR of 75%, with the PFS ranging from 1–5 months and OS from 5–22 months (10). The HER2-amplified cell line constructed by Engelman et al. also suggested that this type of mutation was insensitive to dacomitinib (40). Interestingly, one patient with a higher degree of amplification (HER2/CEP17 ratio of 17) had a prolonged survival, indicating a correlation between the degree of HER2 amplification and the efficacy of dacomitinib (10). In addition, a significant decrease in the HER2 amplification level clinically may suggest a better therapeutic efficacy of dacomitinib, according to the two responded patients (both cases got PR) reported by Reckamp et al. (18) and Kelly et al. (26). A summary of the literature data is shown in Table 2.

CNS metastases

In Kim et al.’s studies, by utilizing cassette dosing in wild-type and Abcb1/Abcg2-deficient mice, osimertinib and dacomitinib were demonstrated to be consistently ranked with a comparatively high brain penetration, while erlotinib, gefitinib, and afatinib were categorized in the low brain penetration group (53). Pharmacokinetic studies revealed that dacomitinib concentrations in rat whole brain homogenates were similar to those in plasma (brain:plasma ratio of 1.2:1), and radiolabeling could be detected in most CNS tissues and cerebrospinal fluid of rats administered 14C-dacomitinib for up to 48 hours (53). A preclinical study conducted by Zahonero et al. demonstrated that dacomitinib could effectively cross the BBB and inhibit EGFR signaling in EGFR-amplified glioblastomas (GBM) brain xenografts, leading to a drastic impairment in tumor growth (54). Consistently, Chen et al.’s work indicated that metabolite ratios were significantly decreased while the apoptotic index was significantly elevated in the dacomitinib-treated group compared with the C6 glioma control group (55).

In the post-hoc analysis of the ARCHER 1050 trial, according to the independent review committee, only 1 (0.4%) patient in dacomitinib group but 9 (4.0%) patients in gefitinib group developed new brain metastatic lesions (odds ratio: 0.11, P=0.034). Since patients with brain metastases were excluded from the ARCHER 1050 trial, current data on the efficacy of dacomitinib in patients with brain metastases are mainly derived from real-world studies and case reports (4,11,14,15,17,24,27,28,56) (Table 3). Peng et al.’s work demonstrated 14 EGFR-mutant NSCLC patients carrying brain metastasis who were treated with first-line dacomitinib, among which measurable responses of CNS metastases (92.3% with brain parenchymal metastasis) were observed in 85.7% of patients, and they obtained an ORR of 92.9% and a DCR of 100% (14). In another larger real-world study, Zhang et al. included a total of 32 TKI-naïve NSCLC patients with brain metastases, all of whom received dacomitinib monotherapy (11). Among 8 CNS evaluable patients, the intracranial ORR (iORR) was 87.5% and the intracranial DCR (iDCR) was 100%, which was consistent with Peng et al.’s findings (14). Furthermore, in 30 evaluable patients, the iORR was 66.7% and the iDCR was 100%, but intracranial PFS (iPFS) was not reached (11). Different from the above studies, Li et al. described the efficacy of later-line dacomitinib in patients with brain metastases (4). As most patients with brain metastases in the later line of therapy had received local therapy (e.g., radiotherapy), the assessment of iORR and iDCR was not performed. A comparative evaluation (49 evaluable patients included) revealed a lower ORR (16.7% vs. 32.3%, P=0.233) but a significantly higher DCR (88.9% vs. 61.3%, P=0.039) of patients with brain metastases than those of patients without brain metastases, and they did not observe any significant differences as to mPFS (P=0.587) and mOS (P=0.647) between the two groups, reflecting the potent efficacy of dacomitinib in patients carrying brain metastases in later-line settings (4). On the other hand, 9 of 15 (60%) patients with symptomatic brain metastases had the relief of their symptoms, and only 3 patients (13.0% of all patients with brain metastases) re-progressed due to brain progression (4).

In particular, limited evidence showed that dacomitinib was effective not only for patients with brain metastases carrying common mutations but also for patients carrying uncommon mutations (11,14). In four brain-metastatic patients harboring G719X compound mutations, Li et al.’s study revealed an ORR of 25% and a DCR of 75% in the later-line settings (4). Chan (17) and Shen et al. (15) respectively reported a patient with CNS-metastatic NSCLC harboring L858R/L718Q compound mutations, and both patients got their intracranial lesions controlled, with the PFS of more than 5 months.


Discussion

Due to the exclusion of clinical trials and the scarcity of study population, evidence on dacomitinib for patients harboring uncommon mutations/CNS metastases is limited (3,57,58), though accumulating evidence have shown its potential in this setting (10,14,18,25,44). Herein, a comprehensive review of dacomitinib on its potential applications from both pre-clinical and clinical findings is presented, in hope of helping clinicians on decision-making, clinical trial design, and drug development.

Compared to common mutations, uncommon mutations are generally less sensitive to TKIs (59-64). Nevertheless, some uncommon mutations including G719X (~3%), S768I (~1%), and L861Q (~1%), also called “major uncommon mutations” are still sensitive to 1G EGFR-TKIs, with ORRs ranging from 41.6% to 53.8% and mPFS, 2.2 to 7.7 months (59,62-64). In the KCSG-LU15-09 trial conducted by Cho et al. (30), osimertinib achieved ORRs of 53%, 78%, 38% and mPFS of 8.2, 15.2, 12.3 months regarding G719X (n=19), L861Q (n=9), and S768I (n=8), respectively. Apart from osimertinib, evidence suggested that 2G TKI was also favorable for this subset of patients. A combined post-hoc analysis by Yang et al. demonstrated that for G719X (n=18), L861Q (n=16), and S768I (n=9), the ORRs were 77.8%, 56.3%, and 100%, and the mPFS were 13.8, 8.2, and 14.7 months, respectively (5). A previous study conducted by Yang et al. also showed that the median time-to-treatment failure (mTTF) was 14.7, 10.0, and 15.6 months in patients treated with first-line afatinib who had G719X, L861Q, and S768I mutations, respectively (47). However, there have been few studies on the efficacy and safety of dacomitinib for patients harboring major uncommon EGFR mutations (3,57,58).

Regarding the different therapeutic responses between 3G TKI osimertinib and 2G TKIs on major uncommon mutations, scholars have given some inspiration from the molecular structure perspective. Different from traditional classification according to exons, Robichaux et al. developed a new structure-based classification (65), and G719X and S768I subtypes are classified as the “P-loop C-helix compressing” type, where changes in the orientation of the P-loop can cause destabilization of osimertinib binding. However, 2G TKIs do not interact with the P-loop of EGFR and maintain interaction points in the hydrophobic cleft, thus keeping full effectiveness for G719X and S768I. The L861Q mutation, categorized as the “classical-like” type, is distal from the drug-binding pocket (BDP) and has low impact on the overall structure of EGFR, thus binding of osimertinib to the mutant EGFR was not blocked.

Interestingly, when the data in the literature were compared, it was found that 2G TKI dacomitinib and afatinib were less effective on L861Q than G719X and S768I (4,30,47), while osimertinib was the opposite (30). Robichaux et al. (65) revealed that G719X and S768I could change the orientation of the P-loop which may lead to the destabilization of osimertinib binding, but 2G TKIs do not interact with the P-loop of EGFR, thus keeping full effectiveness for G719X and S768I. This finding is reflective, suggesting that the concept of “major uncommon mutation” is a definition of incidence, and we should not simply treat it as a therapeutic whole. In this setting, the head-to-head data on dacomitinib versus afatinib or osimretinib in treatment of major uncommon mutations are very expecting. Interestingly, limited data suggest that dacomitinib also exhibits a potent brain-introducing effect in patients with uncommon mutations. Therefore, dacomitinib is worth looking forward to in patients with major uncommon mutations carrying brain metastases. It needs to note that some case reports have reported the clinical benefits of dacomitinib in patients with brain metastases who have non-common EGFR mutations; the usage of dacomitinib has been focused on posterior-line therapy (15,17). One study also suggests a clinical benefit after first-line dacomitinib intervention in patients with brain metastatic lung cancer who have non-common mutations (14). Due to the scarcity of patients with uncommon EGFR mutations, further studies are needed to determine whether dacomitinib can provide clinical benefit to patients with uncommon EGFR mutations in the settings of first-line therapy.

Currently, data in the literature demonstrate the potential application of dacomitinib for patients with major uncommon mutations, but no convincing conclusions can be drawn. A single-arm, open-label, phase II trial (NCT04504071) designed to investigate the safety and efficacy of dacomitinib in advanced NSCLC patients with uncommon EGFR mutations is ongoing in China. Hopefully, this prospective phase II clinical study conducted by Han et al. may provide us with more definitive data (22,66).

Previous studies demonstrated very limited efficacy of 1G TKIs (including gefitinib, erlotinib, and icotinib) for patients with EGFR Ex20ins, with an ORR of 0–6.9%, an mPFS of 1.4–3.0 months, and an mOS of 4.8–26 months (33,36,59). The 2G TKI afatinib seems to be more favorable under comparison with 1G TKIs, with an observed ORR of 8.7–20%, an mPFS of ~2.7 months, and an mOS of ~9.2 months (18). HER2 EX20ins significantly tilt the C-helix toward the BDP of HER2 protein, resulting in a significant spatial blockage at the entrance of the BDP, which in turn prevents most conventional TKIs from binding to this particular conformation-altered BDP and acting (10,39). Nevertheless, as pan-HER2 inhibitors, studies have shown that 2G TKIs have certain efficacy for this subset of patients. Data from a small retrospective study showed that the mPFS and mOS for patients harboring P780_Y781insGSP subtype treated with afatinib were 10.0 and 19.7 months, respectively (67), while those for patients harboring other subtypes were only 3.3 and 7.0 months, respectively. Liu et al. revealed that afatinib obtained an ORR of 19% and an mTTF of 2.9 months in treating HER2-mutant NSCLC, among which the A775_G776ins YVMA subtype achieved an ORR of 33% and an mTTF of 9.6 months (67).

According to our findings, pre-clinical and clinical data revealed that both EGFR and HER2 Ex20ins exhibited significant internal heterogeneity in response to dacomitinib, among which EGFR D770delinsGY, EGFR A763_Y764insFQEA, and HER2 M774delinsWLV were uniquely sensitive to dacomitinib, indicating the feasible clinical applications of dacomitinib on this subset of patients. Interestingly, for HER2 amplification, despite pre-clinical and partial clinical evidence suggesting a less favourable efficacy (40), case reports revealed a correlation between high levels of HER2 amplification and dacomitinib efficacy, and rapid declines in amplification levels may predict better treatment outcomes (10,16,26). In addition, due to controversial results and data paucity, the clinical efficacy of dacomitinib on A767_V769dupASV and D770_N771insNPG still needs to be further confirmed. However, realizing that more and more potent TKIs specifically targeting EGFR and/or HER2 Ex20ins (such as amivantamab/mobocertinib/pyrotinib) are emerging, future research on dacomitinib should focus more on combination therapies and precision populations (58,68).

Some uncommon EGFR mutations including 18del and L747P have also been shown responsive to dacomitinib (41,48). In addition, pre-clinical studies suggested that dacomitinib also showed sensitivity to certain TKI-resistant (mainly osimertinib) secondary mutations (including L792X, L718X, and G724S). Given that osimertinib has been widely used, and there is currently no recommended standard treatment (mainly chemotherapy) for uncommon secondary mutations after drug resistance, the role of dacomitinib in the setting appears to be interesting, especially for the elderly and those who are not willing to receive intensive chemotherapy.

Concerning the side effects of dacomitinib, the common side effects of dacomitinib in NSCLC patients with common EGFR mutations include diarrhea, paronychia, dermatitis acneiform, stomatitis, decreased appetite, and so on in the clinical trial (13). Nevertheless, data on the side effect of dacomitinib for NSCLC patients harboring uncommon mutations is limited. A phase II trial demonstrated the common side of dacomitinib for NSCLC patients harboring uncommon mutations may include rash, diarrhea, oral mucositis, oral mucositis, paronychia, dry skin, and so on (69). Data suggest that patients with uncommon mutations have similar common side effects of gastrointestinal and skin disorders when treated with dacomitinib compared to common mutations. However, those have yet to be proven due to limited data.

Whether the efficacy of dacomitinib is affected by ethnic differences in NSCLC populations with uncommon EGFR mutations is a worthwhile consideration for clinical drug selection. Due to the scarcity of the study population, evidence of response rates on dacomitinib for diverse ethnicities is still limited. Further efforts should focus more on the efficacy differences in diverse populations.

A limitation of this paper is that the complexity of the type of literature collected and the large heterogeneity between the data made it difficult to combine the data, so we did not conduct a meta-analysis.

With the increasingly widespread and ubiquitous application of NGS technology, more and more uncommon mutations have been discovered, which is further rapidly changing the landscape of precision therapy of NSCLC. The challenge, however, is a smaller and smaller target population that corresponds to an increasingly complex mutational spectrum. Consistent with Kris et al. (10), we believe that efforts to build open, searchable databases to share precise molecular signatures of tumors to connect hospitals, institutes, and pharmaceutical companies could go a long way toward confronting these complexities.


Conclusions

Accumulating data have demonstrated promising efficacy of dacomitinib in patients with major uncommon EGFR mutations. The efficacy of dacomitinib against different uncommon EGFR and HER2 mutation subtypes is highly heterogeneous, among which some are clinically applicable. Finally, dacomitinib has demonstrated good intracranial tumor control and should be considered for specific individuals clinically.


Acknowledgments

Funding: None.


Footnote

Reporting Checklist: The authors have completed the PRISMA reporting checklist. Available at https://tcr.amegroups.com/article/view/10.21037/tcr-23-95/rc

Peer Review File: Available at https://tcr.amegroups.com/article/view/10.21037/tcr-23-95/prf

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://tcr.amegroups.com/article/view/10.21037/tcr-23-95/coif). The authors have no conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. Sung H, Ferlay J, Siegel RL, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin 2021;71:209-49. [Crossref] [PubMed]
  2. Passaro A, Mok T, Peters S, et al. Recent Advances on the Role of EGFR Tyrosine Kinase Inhibitors in the Management of NSCLC With Uncommon, Non Exon 20 Insertions, EGFR Mutations. J Thorac Oncol 2021;16:764-73. [Crossref] [PubMed]
  3. Wang S, Li J. Second-generation EGFR and ErbB tyrosine kinase inhibitors as first-line treatments for non-small cell lung cancer. Onco Targets Ther 2019;12:6535-48. [Crossref] [PubMed]
  4. Li HS, Zhang JY, Yan X, et al. A real-world study of dacomitinib in later-line settings for advanced non-small cell lung cancer patients harboring EGFR mutations. Cancer Med 2022;11:1026-36. [Crossref] [PubMed]
  5. Yang JC, Sequist LV, Geater SL, et al. Clinical activity of afatinib in patients with advanced non-small-cell lung cancer harbouring uncommon EGFR mutations: a combined post-hoc analysis of LUX-Lung 2, LUX-Lung 3, and LUX-Lung 6. Lancet Oncol 2015;16:830-8. [Crossref] [PubMed]
  6. NCCN. NCCN Guidelines Version 3.2022. Non-Small Cell Lung Cancer. 2022. Available online: https://www.nccn.org/professionals/physician_gls/pdf/nscl.pdf
  7. Nishio M, Kato T, Niho S, et al. Safety and efficacy of first-line dacomitinib in Japanese patients with advanced non-small cell lung cancer. Cancer Sci 2020;111:1724-38. [Crossref] [PubMed]
  8. Li T, Wang S, Ying J, et al. Afatinib treatment response in advanced lung adenocarcinomas harboring uncommon mutations. Thorac Cancer 2021;12:2924-32. [Crossref] [PubMed]
  9. Jänne PA, Ou SI, Kim DW, et al. Dacomitinib as first-line treatment in patients with clinically or molecularly selected advanced non-small-cell lung cancer: a multicentre, open-label, phase 2 trial. Lancet Oncol 2014;15:1433-41. [Crossref] [PubMed]
  10. Kris MG, Camidge DR, Giaccone G, et al. Targeting HER2 aberrations as actionable drivers in lung cancers: phase II trial of the pan-HER tyrosine kinase inhibitor dacomitinib in patients with HER2-mutant or amplified tumors. Ann Oncol 2015;26:1421-7. [Crossref] [PubMed]
  11. Zhang J, Wang Y, Liu Z, et al. Efficacy of dacomitinib in patients with EGFR-mutated NSCLC and brain metastases. Thorac Cancer 2021;12:3407-15. [Crossref] [PubMed]
  12. Bergonzini C, Leonetti A, Tiseo M, et al. Is there a role for dacomitinib, a second-generation irreversible inhibitor of the epidermal-growth factor receptor tyrosine kinase, in advanced non-small cell lung cancer? Expert Opin Pharmacother 2020;21:1287-98. [Crossref] [PubMed]
  13. Wu YL, Cheng Y, Zhou X, et al. Dacomitinib versus gefitinib as first-line treatment for patients with EGFR-mutation-positive non-small-cell lung cancer (ARCHER 1050): a randomised, open-label, phase 3 trial. Lancet Oncol 2017;18:1454-66. [Crossref] [PubMed]
  14. Peng W, Pu X, Jiang M, et al. Dacomitinib induces objective responses in metastatic brain lesions of patients with EGFR-mutant non-small-cell lung cancer: A brief report. Lung Cancer 2021;152:66-70. [Crossref] [PubMed]
  15. Shen Q, Qu J, Chen Z, et al. Case Report: Dacomitinib Overcomes Osimertinib Resistance in NSCLC Patient Harboring L718Q Mutation: A Case Report. Front Oncol 2021;11:760097. [Crossref] [PubMed]
  16. Li HS, Yang GJ, Wang Y. Case Report: Dacomitinib May Not Benefit Patients Who Develop Rare Compound Mutations After Later-Line Osimertinib Treatment. Front Oncol 2021;11:649843. [Crossref] [PubMed]
  17. Chan D. P76.87 Efficacy of Dacomitinib in EGFR TKI Refractory Metastatic Non-Small Cell Lung Cancer (EGFR Mutant) with Leptomeningeal Metastases. J Thorac Oncol 2021;16:abstr S627.
  18. Reckamp KL, Giaccone G, Camidge DR, et al. A phase 2 trial of dacomitinib (PF-00299804), an oral, irreversible pan-HER (human epidermal growth factor receptor) inhibitor, in patients with advanced non-small cell lung cancer after failure of prior chemotherapy and erlotinib. Cancer 2014;120:1145-54. [Crossref] [PubMed]
  19. Park K, Cho BC, Kim DW, et al. Safety and efficacy of dacomitinib in korean patients with KRAS wild-type advanced non-small-cell lung cancer refractory to chemotherapy and erlotinib or gefitinib: a phase I/II trial. J Thorac Oncol 2014;9:1523-31. [Crossref] [PubMed]
  20. Morita A, Hosokawa S, Yamada K, et al. Dacomitinib as a retreatment for advanced non-small cell lung cancer patient with an uncommon EGFR mutation. Thorac Cancer 2021;12:1248-51. [Crossref] [PubMed]
  21. Kris MG, Mok T, Ou SHI, et al. First-line dacomitinib (PF-00299804), an irreversible pan-her tyrosine kinase inhibitor, for patients with EGFR-mutant lung cancers. Ann Oncol 2012;23:abstr xi34.
  22. Han B, Zhang B, Shi C, et al. P76.59 Rationale and Design of a Phase II Trial of Dacomitinib in Advanced NSCLC Patients with Uncommon EGFR Mutations. J Thorac Oncol 2021;16:S613. [Crossref]
  23. Choudhury NJ, Makhnin A, Tobi YY, et al. Pilot Study of Dacomitinib for Patients With Metastatic EGFR-Mutant Lung Cancers With Disease Progression After Initial Treatment With Osimertinib. JCO Precis Oncol 2021;5:PO.21.00005.
  24. Biswas B, Ganguly S, Ghosh J, et al. Real-world experience of dacomitinib in EGFR mutated advanced NSCLC: A single center experience from India. J Clin Oncol 2021;39:abstr e21043.
  25. Jänne PA, Boss DS, Camidge DR, et al. Phase I dose-escalation study of the pan-HER inhibitor, PF299804, in patients with advanced malignant solid tumors. Clin Cancer Res 2011;17:1131-9. [Crossref] [PubMed]
  26. Kelly RJ, Carter C, Giaccone G. Personalizing therapy in an epidermal growth factor receptor-tyrosine kinase inhibitor-resistant non-small-cell lung cancer using PF-00299804 and trastuzumab. J Clin Oncol 2010;28:e507-10. [Crossref] [PubMed]
  27. Zhao S. Medicine (Baltimore) 2021;100:e26680. [Crossref] [PubMed]
  28. Mizusaki S, Otsubo K, Ninomiya T, et al. Remarkable response to dacomitinib in a patient with leptomeningeal carcinomatosis due to EGFR-mutant non-small cell lung cancer. Thorac Cancer 2021;12:114-6. [Crossref] [PubMed]
  29. Wu SG, Yu CJ, Yang JC, et al. The effectiveness of afatinib in patients with lung adenocarcinoma harboring complex epidermal growth factor receptor mutation. Ther Adv Med Oncol 2020;12:1758835920946156. [Crossref] [PubMed]
  30. Cho JH, Lim SH, An HJ, et al. Osimertinib for Patients With Non-Small-Cell Lung Cancer Harboring Uncommon EGFR Mutations: A Multicenter, Open-Label, Phase II Trial (KCSG-LU15-09). J Clin Oncol 2020;38:488-95. [Crossref] [PubMed]
  31. Tan J, Hu C, Deng P, et al. The Predictive Values of Advanced Non-Small Cell Lung Cancer Patients Harboring Uncommon EGFR Mutations-The Mutation Patterns, Use of Different Generations of EGFR-TKIs, and Concurrent Genetic Alterations. Front Oncol 2021;11:646577. [Crossref] [PubMed]
  32. Gristina V, Malapelle U, Galvano A, et al. The significance of epidermal growth factor receptor uncommon mutations in non-small cell lung cancer: A systematic review and critical appraisal. Cancer Treat Rev 2020;85:101994. [Crossref] [PubMed]
  33. Vyse S, Huang PH. Targeting EGFR exon 20 insertion mutations in non-small cell lung cancer. Signal Transduct Target Ther 2019;4:5. [Crossref] [PubMed]
  34. Attili I, Passaro A, Pisapia P, et al. Uncommon EGFR Compound Mutations in Non-Small Cell Lung Cancer (NSCLC): A Systematic Review of Available Evidence. Curr Oncol 2022;29:255-66. [Crossref] [PubMed]
  35. Remon J, Hendriks LEL, Cardona AF, et al. EGFR exon 20 insertions in advanced non-small cell lung cancer: A new history begins. Cancer Treat Rev 2020;90:102105. [Crossref] [PubMed]
  36. Burnett H, Emich H, Carroll C, et al. Epidemiological and clinical burden of EGFR Exon 20 insertion in advanced non-small cell lung cancer: A systematic literature review. PLoS One 2021;16:e0247620. [Crossref] [PubMed]
  37. Shigematsu H, Lin L, Takahashi T, et al. Clinical and biological features associated with epidermal growth factor receptor gene mutations in lung cancers. J Natl Cancer Inst 2005;97:339-46. [Crossref] [PubMed]
  38. Kobayashi IS, Viray H, Rangachari D, et al. EGFR-D770>GY and Other Rare EGFR Exon 20 Insertion Mutations with a G770 Equivalence Are Sensitive to Dacomitinib or Afatinib and Responsive to EGFR Exon 20 Insertion Mutant-Active Inhibitors in Preclinical Models and Clinical Scenarios. Cells 2021;10:3561. [Crossref] [PubMed]
  39. Kosaka T, Tanizaki J, Paranal RM, et al. Response Heterogeneity of EGFR and HER2 Exon 20 Insertions to Covalent EGFR and HER2 Inhibitors. Cancer Res 2017;77:2712-21. [Crossref] [PubMed]
  40. Engelman JA, Zejnullahu K, Gale CM, et al. PF00299804, an irreversible pan-ERBB inhibitor, is effective in lung cancer models with EGFR and ERBB2 mutations that are resistant to gefitinib. Cancer Res 2007;67:11924-32. [Crossref] [PubMed]
  41. Kobayashi Y, Mitsudomi T. Not all epidermal growth factor receptor mutations in lung cancer are created equal: Perspectives for individualized treatment strategy. Cancer Sci 2016;107:1179-86. [Crossref] [PubMed]
  42. Gergis C, Rangachari D, Fujii M, et al. EGFR-A763-Y764insFQEA: A unique exon 20 insertion mutation that displays sensitivity to all classes of approved lung cancer EGFR tyrosine kinase inhibitors. J Clin Oncol 2019;37:abstr e20593.
  43. Lin L, Lu Q, Cao R, et al. Acquired rare recurrent EGFR mutations as mechanisms of resistance to Osimertinib in lung cancer and in silico structural modelling. Am J Cancer Res 2020;10:4005-15. [PubMed]
  44. Nishino M, Suda K, Kobayashi Y, et al. Effects of secondary EGFR mutations on resistance against upfront osimertinib in cells with EGFR-activating mutations in vitro. Lung Cancer 2018;126:149-55. [Crossref] [PubMed]
  45. Kobayashi Y, Azuma K, Nagai H, et al. Characterization of EGFR T790M, L792F, and C797S Mutations as Mechanisms of Acquired Resistance to Afatinib in Lung Cancer. Mol Cancer Ther 2017;16:357-64. [Crossref] [PubMed]
  46. Kobayashi Y, Togashi Y, Yatabe Y, et al. EGFR Exon 18 Mutations in Lung Cancer: Molecular Predictors of Augmented Sensitivity to Afatinib or Neratinib as Compared with First- or Third-Generation TKIs. Clin Cancer Res 2015;21:5305-13. [Crossref] [PubMed]
  47. Yang JC, Schuler M, Popat S, et al. Afatinib for the Treatment of NSCLC Harboring Uncommon EGFR Mutations: A Database of 693 Cases. J Thorac Oncol 2020;15:803-15. [Crossref] [PubMed]
  48. Yang G, Liu C, Hu J, et al. The Lifted Veil of Uncommon EGFR Mutation p.L747P in Non-Small Cell Lung Cancer: Molecular Feature and Targeting Sensitivity to Tyrosine Kinase Inhibitors. Front Oncol 2022;12:843299. [Crossref] [PubMed]
  49. Yoshizawa T, Uchibori K, Araki M, et al. Microsecond-timescale MD simulation of EGFR minor mutation predicts the structural flexibility of EGFR kinase core that reflects EGFR inhibitor sensitivity. NPJ Precis Oncol 2021;5:32. [Crossref] [PubMed]
  50. Li BT, Ross DS, Aisner DL, et al. HER2 Amplification and HER2 Mutation Are Distinct Molecular Targets in Lung Cancers. J Thorac Oncol 2016;11:414-9. [Crossref] [PubMed]
  51. Koga T, Kobayashi Y, Tomizawa K, et al. Activity of a novel HER2 inhibitor, poziotinib, for HER2 exon 20 mutations in lung cancer and mechanism of acquired resistance: An in vitro study. Lung Cancer 2018;126:72-9. [Crossref] [PubMed]
  52. Robichaux JP, Elamin YY, Tan Z, et al. Mechanisms and clinical activity of an EGFR and HER2 exon 20-selective kinase inhibitor in non-small cell lung cancer. Nat Med 2018;24:638-46. [Crossref] [PubMed]
  53. Kim M, Laramy JK, Mohammad AS, et al. Brain Distribution of a Panel of Epidermal Growth Factor Receptor Inhibitors Using Cassette Dosing in Wild-Type and Abcb1/Abcg2-Deficient Mice. Drug Metab Dispos 2019;47:393-404. [Crossref] [PubMed]
  54. Zahonero C, Aguilera P, Ramírez-Castillejo C, et al. Preclinical Test of Dacomitinib, an Irreversible EGFR Inhibitor, Confirms Its Effectiveness for Glioblastoma. Mol Cancer Ther 2015;14:1548-58. [Crossref] [PubMed]
  55. Chen WS, Hong L, Wang F, et al. Investigation of dacomitinib on reducing cell necrosis and enhancing cell apoptosis in C6 glioma rat model by MRI. Biosci Rep 2019;39:BSR20190006. [Crossref] [PubMed]
  56. Zhou Q, Wu Y, Lin S, et al. P76.78 Evaluation of the Development of Brain Metastases in Patients Treated with Dacomitinib or Gefitinib from ARCHER 1050 Study. J Thorac Oncol 2021;16:S622-3. [Crossref]
  57. Lau SCM, Batra U, Mok TSK, et al. Dacomitinib in the Management of Advanced Non-Small-Cell Lung Cancer. Drugs 2019;79:823-31. [Crossref] [PubMed]
  58. Lavacchi D, Mazzoni F, Giaccone G. Clinical evaluation of dacomitinib for the treatment of metastatic non-small cell lung cancer (NSCLC): current perspectives. Drug Des Devel Ther 2019;13:3187-98. [Crossref] [PubMed]
  59. Wu JY, Yu CJ, Chang YC, et al. Effectiveness of tyrosine kinase inhibitors on "uncommon" epidermal growth factor receptor mutations of unknown clinical significance in non-small cell lung cancer. Clin Cancer Res 2011;17:3812-21. [Crossref] [PubMed]
  60. Watanabe S, Minegishi Y, Yoshizawa H, et al. Effectiveness of gefitinib against non-small-cell lung cancer with the uncommon EGFR mutations G719X and L861Q. J Thorac Oncol 2014;9:189-94. [Crossref] [PubMed]
  61. Baek JH, Sun JM, Min YJ, et al. Efficacy of EGFR tyrosine kinase inhibitors in patients with EGFR-mutated non-small cell lung cancer except both exon 19 deletion and exon 21 L858R: a retrospective analysis in Korea. Lung Cancer 2015;87:148-54. [Crossref] [PubMed]
  62. Chiu CH, Yang CT, Shih JY, et al. Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitor Treatment Response in Advanced Lung Adenocarcinomas with G719X/L861Q/S768I Mutations. J Thorac Oncol 2015;10:793-9. [Crossref] [PubMed]
  63. Kuiper JL, Hashemi SM, Thunnissen E, et al. Non-classic EGFR mutations in a cohort of Dutch EGFR-mutated NSCLC patients and outcomes following EGFR-TKI treatment. Br J Cancer 2016;115:1504-12. [Crossref] [PubMed]
  64. Krawczyk P, Kowalski DM, Ramlau R, et al. Comparison of the effectiveness of erlotinib, gefitinib, and afatinib for treatment of non-small cell lung cancer in patients with common and rare EGFR gene mutations. Oncol Lett 2017;13:4433-44. [Crossref] [PubMed]
  65. Robichaux JP, Le X, Vijayan RSK, et al. Structure-based classification predicts drug response in EGFR-mutant NSCLC. Nature 2021;597:732-7. [Crossref] [PubMed]
  66. Zhang B, Shi C, Gao Z, et al. Rationale and design of a phase II trial of dacomitinib in advanced non-small cell lung cancer patients with uncommon epidermal growth factor receptor mutations: a prospective and single arm study (DANCE study). BMC Cancer 2022;22:294. [Crossref] [PubMed]
  67. Liu Z, Wu L, Cao J, et al. Clinical characterization of ERBB2 exon 20 insertions and heterogeneity of outcomes responding to afatinib in Chinese lung cancer patients. Onco Targets Ther 2018;11:7323-31. [Crossref] [PubMed]
  68. Friedlaender A, Subbiah V, Russo A, et al. EGFR and HER2 exon 20 insertions in solid tumours: from biology to treatment. Nat Rev Clin Oncol 2022;19:51-69. [Crossref] [PubMed]
  69. Li HS, Wang SZ, Xu HY, et al. Afatinib and Dacomitinib Efficacy, Safety, Progression Patterns, and Resistance Mechanisms in Patients with Non-Small Cell Lung Cancer Carrying Uncommon EGFR Mutations: A Comparative Cohort Study in China (AFANDA Study). Cancers (Basel) 2022;14:5307. [Crossref] [PubMed]
Cite this article as: Yang LL, Luo XZ, Xie LL, Lei XZ, Zhu J. The treatment of patients with non-small cell lung cancer carrying uncommon EGFR mutations, HER2 mutations, or brain metastases: a systematic review of pre-clinical and clinical findings for dacomitinib. Transl Cancer Res 2023;12(8):2197-2211. doi: 10.21037/tcr-23-95

Download Citation