Abstract
Background:
ALK-tyrosine kinase inhibitors (ALK-TKIs) are effective for treating non-small-cell lung cancer with ALK gene rearrangement; however, resistance is inevitable. Brigatinib is a unique ALK-TKI that is effective against many resistance mutations. However, data on factors associated with its efficacy and resistance mechanisms are limited.
Objectives:
This study will evaluate the efficacy and safety of brigatinib in the real world and explore factors related to its efficacy, safety, and resistance mechanisms.
Design:
Prospective observational study.
Ethics:
This study is approved by the Ethics Committee of Wakayama Medical University. Written informed consent will be obtained from all patients before study-related procedures.
Methods and analysis:
This study comprises three cohorts. Cohorts A, B, and 0 will enroll patients receiving alectinib as the first ALK-TKI, receiving alectinib as the first ALK-TKI and subsequently cytotoxic agents and/or lorlatinib after alectinib, and without a history of ALK-TKI, respectively. Overall, 100, 30, and 50 patients will be enrolled in Cohorts A, B, and 0, respectively. Circulating tumor DNA before starting brigatinib and at disease progression will be analyzed in all cohorts using a hypersensitive next-generation sequencing (NGS) PGDx Elio plasma resolve panel. Serum protein levels will be analyzed using the Milliplex xMAP assay system with a Luminex 200 (Luminex, Austin, USA). The enrollment period is 31 months and the patients will be observed for 2 years after enrollment. Archived tissues will be collected for NGS analysis, gene expression analysis, and immunohistochemistry staining 1 year after completion of registration. Quality of life and safety evaluation using electronic patient-reported outcomes will be investigated.
Discussion:
This study will elucidate predictors of ALK-TKI efficacy and resistance mechanisms and evaluate the efficacy and safety of brigatinib in a real-world setting. The results will provide crucial information for establishing treatment strategies, discovering novel biomarkers, and developing new therapeutic agents.
Trial registration:
UMIN000042439.
Keywords: alectinib, ALK inhibitors, brigatinib, non-small-cell lung cancer, observational study
Introduction
Lung cancer was the leading cause of all cancer-related deaths worldwide in 2018, accounting for 18.4% (1.8 million) deaths. 1 Rearrangement of the ALK gene accounts for approximately 3–5% of all non-small-cell lung cancer (NSCLC) cases and tends to be more prevalent in young people. 2 In 2011, the Food and Drug Administration approved crizotinib, the first ALK tyrosine kinase inhibitor (TKI), for the treatment of advanced ALK-positive NSCLC. In 2014, a phase III trial in patients with untreated ALK gene metastases in advanced stages showed progression-free survival (PFS) superiority over standard chemotherapy, 3 cementing ALK-TKI as an essential drug. Subsequently, alectinib, lorlatinib, ensartinib, and brigatinib showed superiority over crizotinib in PFS in further phase III trials4–7 and, currently, these three ALK-TKIs are widely used as first-line therapy. However, because most cases eventually lead to tumor progression, the development of sequencing strategies for ALK-TKI is an urgent issue.
Various reports exist on factors associated with the efficacy of ALK-TKI; for example, in 2018, Lin et al. 8 reported that the efficacy of lorlatinib differed between Variants 1 and 3 of the ALK gene and showed that Variant 3 led to more frequent G1202R resistance mutations than Variant 1 after progression on a second-generation ALK inhibitor. Other reports also suggest that lorlatinib and brigatinib had better PFS in variant 1 than in variant 3 9,10; however, reports also exist showing no such difference, including an analysis using data from the CROWN trial.11,12 Thus, the significance of this finding is unclear. Moreover, mutations in genes other than ALK also affect ALK-TKI efficacy. The presence of TP53 mutations has been shown to reduce the efficacy of ALK-TKI, and an analysis of the ALTA-1L trial showed a trend toward shorter PFS in patients with TP53 mutations who received either brigatinib or crizotinib. 10 An analysis of the CROWN trial also showed a similar trend for lorlatinib11,12; however, no statistically significant difference was shown in this analysis and the significance of such compound mutations in ALK-TKI therapy is not fully understood.
Mutations in the tyrosine kinase domain of the ALK gene have been known to be one of the main resistance mechanisms in ALK-TKIs. For alectinib, the most widely used second-generation ALK-TKIs, approximately 50–60% of cases, had mutations in the ALK gene as a resistance mechanism, with I1171T/NS, V1180L, and G1202R being relatively common. 13 Because the meaning of the mutation varies depending on the drug, I1171T/N/S and V1180L are sensitive to ceritinib while reported to be resistant to crizotinib. 14 Brigatinib also has been reported to show sensitivity to I1171T/N/S and V1180L13,15 and, among five patients with these mutations, one showed response and three achieved stable diseases. 16 Furthermore, brigatinib has limited sensitivity for G1202R. 14 However, the concentration of brigatinib that inhibited 50% of BA/F3 cells with G1202R mutation in Variant 3 was lower than that for cells with G1202R mutation in Variant 1, 15 and notably, G1202R is mostly found in Variant 3. 8 Considering these, such mutations may be predictive of brigatinib efficacy. Furthermore, it has been demonstrated that double and triple mutations occur, particularly when multiple ALK-TKIs are used17,18 and that the appropriate agent for each mutation is also effective in such cases. 19 The accumulation of such information from clinical samples would therefore be crucial in developing appropriate treatment strategies for ALK-gene translocated NSCLC. 19
Although varying by ALK-TKI, a significant proportion of cases with ALK-TKI resistance involve mechanisms other than ALK gene alterations, which are referred to as ‘off-target’ resistance. Various off-target resistance mechanisms exist, 14 including Met gene amplification, 20 Met signaling activation associated with hepatocyte growth factor (HGF)upregulation, 21 TP53 mutation, 22 the Src pathway, 23 and epidermal growth factor receptor (EGFR) pathway activation. 24 Such off-target resistance can occur in conjunction with on-target resistance, and this coexistence was reported in 5 of the 17 (29%) patients with multiple ALK-TKIs, which was higher than that in 2 of the 35 (6%) individuals with crizotinib alone. 25 In addition, the tumor microenvironment is associated with the efficacy of TKIs. 26 Hypoxia is reportedly involved in ALK-TKI resistance through the epithelial–mesenchymal transition. 27 In EGFR-TKIs, it has been recently reported that serum protein levels, including interleukin-6 (IL-6) and vascular endothelial growth factor (VEGF)28,29 and tumor protein expression, such as EGFR,30,31 programmed death-ligand 1 (PD-L1),32,33 and CD47 34 are associated with efficacy and resistance. Similar resistance mechanisms may be observed for ALK-TKIs.
There is no clear indication of whether alectinib, lorlatinib, or brigatinib has the greatest efficacy for first-line therapy. Hence, elucidating the predictors of efficacy and resistance mechanisms for each of these drugs is essential to establishing the best sequencing strategy. In addition, data on brigatinib are relatively high in demand. Given these circumstances, evaluating its efficacy and safety in clinical practice, as well as elucidating the factors associated with its efficacy and resistance mechanisms, will be a major step toward establishing therapeutic strategies to utilize multiple second- and third-generation ALK-TKIs. Therefore, we have planned a multicenter observational study of brigatinib in three cohorts of patients with advanced ALK-rearranged NSCLC.
Study protocol
This study is prospectively enrolling patients with advanced ALK gene-rearranged NSCLC who receive brigatinib. Through this study, we aim to evaluate the efficacy and safety of brigatinib in the real world and explore factors related to its efficacy, safety, and resistance mechanisms.
This study includes the following three cohorts: A, B, and 0. Cohort A will enroll patients who receive alectinib as the first ALK-TKI, followed by brigatinib as an immediate subsequent therapy. This is the main cohort, and 100 patients will be enrolled. Cohort B will include 30 patients who receive alectinib as the first ALK-TKI, followed by chemotherapy and/or lorlatinib, and then brigatinib. Cohort 0 will comprise 50 patients who receive brigatinib as their first ALK-TKI. One chemotherapy regimen is allowed in all cohorts before the first ALK-TKI treatment (Figure 1). The eligibility criteria are as follows: (1) patients with advanced stages (IIIB, IIIC, IVA, and IVB) or those experiencing postoperative recurrence and ALK gene-rearranged NSCLC for which curative treatment is not indicated; (2) patients scheduled to receive brigatinib; and (3) patients who have received previous treatment: In Cohort A (brigatinib as second- or third-line therapy), alectinib was administered as the first ALK-TKI and was the most recent prior therapy. In Cohort B (brigatinib as 3rd–5th line therapy), alectinib was administered as the first ALK-TKI, and a cytotoxic agent (up to one regimen) and/or lorlatinib was administered after alectinib. Cytotoxic agents and lorlatinib are the most recent therapies. Cohort 0 (brigatinib as first- or second-line therapy): no prior use of ALK-TKIs. Up to one regimen of cytotoxic agents before the first ALK-TKI is allowed in all cohorts; (4) patients evaluated using computed tomography (CT), including the chest, within 42 days and brain magnetic resonance imaging or CT within 56 days before the start of brigatinib; and (5) patients aged 20 years or older at the time consent was obtained.
Figure 1.
Protocol of this study.
ALK-TKI, ALK-tyrosine kinase inhibitor.
The primary endpoint is PFS. The secondary endpoints are overall response rate, overall survival, time to treatment failure, duration of response, disease control rate, central nervous system (CNS) disease overall response rate, CNS disease control rate, CNS PFS, cumulative incidence of CNS disease, cumulative incidence of CNS disease progression, and incidence and severity of adverse events. Exploratory endpoints include next-generation sequencing (NGS) analysis using plasma and tumor tissue DNA, tissue gene expression, and serum and tumor-expressing proteins to explore factors associated with efficacy, safety, and resistance mechanisms to brigatinib. Plasma DNA will be collected before initiation of brigatinib and disease progression, and serum will be collected before initiation of brigatinib. Tumor tissues will be evaluated to the extent that is possible from incidental biopsies in clinical practice. In addition, patient-reported outcomes (PRO) will be assessed electronically in an exploratory manner in cases where additional consent is obtained. Specifically, health-related quality of life, performance status (PS), and adverse events will be assessed periodically. The impact of these assessments on clinical outcomes and the factors that influence participation in electronic PRO (ePRO) will be evaluated.
Treatment
All patients will receive brigatinib orally once daily for 7 days. Subsequently, 180 mg will be orally administered once daily. The dosage may be reduced according to the patient’s condition and the investigators’ clinical judgment.
Regarding circulating tumor DNA (ctDNA) analysis, we will collect plasma samples before treatment with brigatinib and after progressive disease, and ctDNA will be analyzed using hypersensitive NGS. For serum protein analysis, serum samples will be collected before brigatinib treatment. Furthermore, we will collect tumor tissue before the first ALK-TKI, after alectinib – before brigatinib, and after brigatinib – before the next treatment; we will also collect, when possible, and examine the tumor tissue DNA using NGS, tumor-expressing proteins using immunohistochemistry, and the gene expression profile of the tumor.
Participants’ registration
After confirmation of eligibility and providing informed consent, eligible patients will be enrolled, and the investigator will initiate treatment with brigatinib. Recruitment will begin in May 2021 and continue until December 2023. Overall, 83 facilities, including community hospitals, academic hospitals, and cancer centers, in Japan will participate in this study.
DNA extraction from peripheral blood samples and tumor tissue
Regarding ctDNA analysis, blood samples will be collected using Streck Cell-Free DNA BCT blood collection tubes. Samples will be collected by SRL Inc. (Tokyo, Japan), and DNA will be extracted within 72 h of blood collection, according to the manufacturer’s instructions. Briefly, blood will be centrifuged at 300g for 20 min and 5000g for 10 min at room temperature. Plasma DNA will be isolated using a QIAamp MinElute ccfDNA Midi Kit (Qiagen, Hilden, Germany) according to the manufacturer’s protocol. Moreover, DNA concentration will be measured using a Qubit 3.0 Fluorometer (Life Technologies, Carlsbad, CA, USA) or NanoDrop 2000 (Fisher Scientific, Pittsburgh, USA). DNA and RNA will be extracted from formalin-fixed paraffin-embedded samples using SRL. Inc.
Next-generation sequencing
NGS will be performed using ctDNA extracted from the plasma. The PGDx Elio plasma resolve panel will be used to detect mutations and amplifications in 33 genes (Table 1). For tissue samples, the PGDx Elio tissue complete panel will be used to extract the mutations and amplifications of 507 genes when more than 50 ng of DNA is available (Supplemental Table S1).
Table 1.
Genetic analysis using circulating tumor DNA.
| Sequence mutation analyses (33 genes) |
|
AKT1, BRCA1, CSF1R, HRAS, NTRK1, RET, ALK, BRCA2, EGFR, KIT, PDGFRA, ROS1, APC, BRIP1, ERBB2, KRAS, PIK3CA, TP53, ARID1A, CCND1, EZH2, MET, POLD1, ATM, CD274, FGFR1, MYC, POLE, BRAF, CDH1, FGFR2, NRAS, and RAF1 |
| Copy number variation (eight genes) |
| CCND1, CD274, EGFR, ERBB2, KIT, MET, MYC, and FGFR2 |
| Translocation analyses (five genes) |
| ALK, FGFR2, NTRK1, RET, and RO1 |
Serum and tumor protein and gene expression analyses
Serum will be used to analyze the concentrations of 44 proteins (Table 2). The analysis will be performed using a Milliplex xMAP assay system with a Luminex 200. Gene expression in tissues will be analyzed using RNA extracted from tissues using the nCounter Analysis System (NanoString Technologies, Inc., Seattle, USA) Pancer IO360 panel. In the residual tissues, the expression of EGFR, Amphiregulin, CD47, PD-L1, CD8, and CD24 will be evaluated using immunostaining (Supplemental Table S2).
Table 2.
Cytokine analysis in the peripheral blood.
| IL-1α/β | IL-1Ra | IL-2 | IL-4 | IL-5 | IL-6 | IL-7 |
| IL-8 | IL-10 | IL-12 | IL-13 | IL-15 | IL-17 | EGF |
| CCL11 | G-CSF | GM-CSF | GRO | IFN-α2 | IFN-γ | CXCL10 |
| CCL2 | CCL7 | CCL22 | MIP-1α/β | TGF-α | MIP-1α/β | TNF-α/β |
| VEGF-A | Angiopoietin-2 | BMP-9 | EGF | FGF-2 | Follistatin | HB-EGF |
| HGF | Leptin | PLGF | VEGF-C/D | TGF-β1/2/3 |
Electronic patient-reported outcome
In patients who provide additional consent, adverse events, quality of life, and PS evaluations will be periodically conducted using Welby MyKarte ONC® (Welby, Tokyo, Japan). Adverse events will be assessed weekly using up to 21 questions on 11 items (anorexia, nausea, vomiting, diarrhea, shortness of breath, cough, swelling, rash, pruritus, fatigue, and anxiety) selected from the PRO version of the Common Terminology Criteria for Adverse Events 5.0. PS will be assessed every 4 weeks according to the Eastern Cooperative Oncology Group PS, and quality of life will be evaluated every 4 weeks using the European Organization for Research and Treatment QLQ-C30 and QLQ-LC13.
Statistical considerations
This clinical study aims to evaluate the efficacy and safety of brigatinib in advanced or recurrent ALK gene rearranged NSCLC after treatment with alectinib as the first ALK-TKI in the real world. The primary endpoint of this prospective observational study is PFS, and the number of patients in Cohort A is calculated according to the following hypothesis: a phase II study evaluating the efficacy of brigatinib in patients with resistance to second-generation ALK-TKIs, including alectinib, reported a median PFS of 6.4 months. 35 By contrast, a combination of platinum and pemetrexed in patients with second-generation ALK-TKI resistant, including alectinib, resulted in a median PFS of 4.3 months. 36 Assuming a PFS of 6.0 months for brigatinib, which is significantly higher than the 4.0 months that is expected for the platinum and pemetrexed combination, 90 patients would be the required number of individuals under the following conditions: 5% one-sided significance level, 80% power, 26 months of enrollment period, and 1 year of follow-up. Thus, the number of patients was set to 100 to allow for fewer dropouts.
Cohorts B and 0 are created to collect information in a clinical practice setting, and the necessary statistical number of cases is not calculated. As a realistic and feasible number of cases for exploratory analysis, we will enroll 30 and 50 patients in Cohorts B and 0, respectively.
The primary endpoint, PFS, is defined as the period beginning on the date of brigatinib initiation and ending on the date of death from any cause, on the date of images confirming disease progression, or on the date of clinical diagnosis of disease progression, whichever occurs first. Disease progression is defined as an apparent worsening of imaging data, patient symptoms, or physical examination findings. The principal investigator or study investigator will determine disease progression with reference to Response Evaluation Criteria in Solid Tumors v1.1. The analysis will be performed on the entire cohort 1 year after the start of the follow-up period. At the end of the follow-up period, all endpoints will be analyzed after the data are confirmed based on a final survey.
Ethical consideration
This study will be conducted in accordance with the provisions of the Declaration of Helsinki and approved by the Ethics Committee of Wakayama Medical University (No. 3073). Written informed consent will be obtained from all patients before study-related procedures are performed. The trial registration number for this study is UMIN000042439.
Discussion
This prospective observational study has three main objectives. First, it aims to evaluate the efficacy and safety of brigatinib in a real-world setting. Second, it aims to explore predictive factors associated with the efficacy of brigatinib. Third, it aims to explore resistance mechanisms to brigatinib in each situation. We believe that these findings will contribute to developing appropriate treatment strategies for ALK-gene rearranged, advanced NSCLC.
Regarding the safety and efficacy of brigatinib, available data on brigatinib treatment after alectinib as the first ALK-TKI, which is the focus of cohort A in the ABRAID trial, are limited. Ou et al. reported a response rate of 26% with a median PFS of 3.8 months in 103 patients enrolled in a phase II study of brigatinib in individuals previously treated with ceritinib or alectinib, in the ALTA-2 trial. However, only 42 patients received alectinib as the first ALK-TKI. 37 J-ALTA study, a phase II study on brigatinib that included 47 patients previously treated with alectinib, also showed that brigatinib resulted in a response rate and median PFS of 34% and 7.3 months, respectively. However, 12 of the patients also received prior crizotinib, and only 35 received brigatinib after alectinib, which was administered as the first ALK-TKI. 38 Thus, Cohort A in this ABRAID trial is the largest prospective study to evaluate the efficacy and safety of brigatinib as a second ALK-TKI administered immediately after alectinib. In addition, to the best of our knowledge, no prospective clinical trials exist focusing on the efficacy and safety of brigatinib after third-line treatment or after treatment with two second-generation ALK-TKIs; therefore, the results of Cohort B, which enrolls this population, should be valuable. Furthermore, the J-ALTA study, which was conducted in Japan and included mostly Japanese participants, on brigatinib in patients with no prior ALK-TKI treatment resulted in response and 1-year PFS rates of 96.9% and 93% in 32 patients, respectively. 39 Cohort 0, which enrolls 50 ALK-TKI-naïve patients, is expected to confirm the results in a real-world setting. An analysis of 148 patients with ALK-positive lung cancers diagnosed between 2009 and 2021 showed that 54% received crizotinib and 44% alectinib as the first ALK-TKI with ceritinib, brigatinib, and lorlatinib as the second and subsequent ALK-TKI in 23%, 17%, and 29% of participants, respectively, in a real-world setting. 40 This indicates a lack of real-world data on brigatinib as the first ALK-TKI, as well as the need for a marker to select the second and subsequent ALK-TKI.
To explore predictive factors of efficacy, 180 ctDNA samples before brigatinib administration will be collected from the three cohorts. Samples from Cohort A will enable us to evaluate the association between resistance genetic alterations against alectinib and the efficacy of brigatinib. Although new mutations in the ALK gene including I1171T/NS, V1180L, and G1202R, 13 P53 mutations, 22 MET gene amplifications, 20 PIK3CA mutations, 41 and KRAS G12C mutations 42 are expected to occur after treatment with alectinib, there are few data about their impact on the efficacy of brigatinib or other ALK-TKI. Cohort B samples can be used to find genetic changes after alectinib followed by lorlatinib and to explore their association with the efficacy of brigatinib. After treatment with two second- or third-generation ALK-TKI administrations, multiple resistance mechanisms would be more likely to exist, 25 and evaluation of gene alterations alone may not be sufficient. Comparisons with cohorts 0 and A may provide an edge in clarifying this. Analysis of the association of ALK variants 13 and compound mutations like TP53 mutations 22 with the efficacy of brigatinib using cohort 0 samples is important to confirm the results of similar analysis using samples from the ALTA-1L trial in the real-world setting. 10 It will also be of interest to explore whether an impact of ALK variants or compound mutations on ALK-TKI efficacy is similar when the ALK-TKI is administered as the second or third ALK-TKI. If it is, a treatment strategy that does not rely too much on ALK-TKI sequencing may be a preferred choice in such cases.
Moreover, this study will also evaluate protein levels in the pre-treatment serum and tumor tissue. Serum concentration of IL-6 27 and VEGF, 26 as well as expression of proteins such as EGFR,30,31 PD-L1,32,33 and CD47, 34 is associated with the efficacy of EGFR-TKI. Because these tumor microenvironment-related resistance mechanisms are not necessarily EGFR mutation-specific and have rarely been evaluated in ALK-TKI, such analysis may lead to the elucidation of new resistance mechanisms for ALK-TKI. The association between genetic abnormalities and the efficacy of ALK-TKI often differs between reports, suggesting the existence of important resistance mechanisms other than genetic abnormalities. Combined analysis of genetic abnormalities with serum proteins and/or tumor-expressing proteins may better predict the efficacy of ALK-TKI.
Data on the resistance mechanisms of brigatinib are similar to or less complete than those on other ALK-TKIs. This study explores resistance mechanisms when brigatinib is used as the first ALK-TKI, as the second ALK-TKI after alectinib, and as the third ALK-TKI after alectinib and lorlatinib, using ctDNA samples at progression or treatment discontinuation from each cohort. Notably, all 180 study participants have only these three patterns of pre-treatment history with ALK-TKIs. Therefore, this study will provide crucial information on how the pre-treatment history with ALK-TKIs affects the resistance mechanisms. It has been reported that the coexistence of ALK-resistant and TP53 mutations occurs more frequently when multiple ALK-TKIs are used 25 ; however, it is not well understood whether similar events occur with other off-target mechanisms. Furthermore, alterations in resistance genes to brigatinib may depend on genetic alterations that existed prior to the brigatinib administration. Accumulating such data would enable us to choose the best ALK-TKI for each case considering not only the efficacy of the first ALK-TKI but also the efficacy of the second and third ALK-TKIs. Such exploration will also contribute to the development of new combination therapies and new ALK-TKI in the future.
The patient’s assessment of the quality of life and adverse events using electronic devices is crucial, owing to their high sensitivity and the possibility of assessment over time. 43 However, such ePRO is technically difficult for older adults, and some patients may be reluctant to use such devices. In this observational study, we will use ePRO to evaluate the quality of life and adverse events in a distinctive population with ALK-positive lung cancer. Another important aspect of ePRO evaluation is the feasibility assessment. Little has been reported on what clinical context influences patient acceptance and continuation of ePRO. The results of this study will be useful for promoting the efficient use and dissemination of ePRO in clinical practice.
Conclusion
We evaluate the real-world clinical efficacy and safety of brigatinib in various treatment lines, including patients who received alectinib as their first ALK-TKI. Furthermore, we attempt to elucidate the efficacy-related and resistance mechanisms from multiple angles using genes and proteins in blood and tumor tissue. The results of this study are expected to be a major step forward in establishing therapeutic strategies for using multiple second- and third-generation ALK-TKIs, which is currently a major challenge in the treatment of ALK-positive lung cancers and may further lead to the development of novel biomarkers and therapeutic agents.
Supplemental Material
Supplemental material, sj-docx-1-tam-10.1177_17588359231225046 for Prospective observational study to explore genes and proteins predicting efficacy and safety of brigatinib for ALK-gene rearranged non-small-cell lung cancer: study protocol for ABRAID study (WJOG11919L) by Yuichi Ozawa, Yasuhiro Koh, Tetsunari Hase, Kenji Chibana, Kyoichi Kaira, Kyoichi Okishio, Eiki Ichihara, Shuji Murakami, Mototsugu Shimokawa and Nobuyuki Yamamoto in Therapeutic Advances in Medical Oncology
Acknowledgments
We thank the patients, their families, and all investigators participating in the study.
Footnotes
ORCID iD: Yuichi Ozawa
https://orcid.org/0000-0003-3798-3488
Supplemental material: Supplemental material for this article is available online.
Contributor Information
Yuichi Ozawa, Department of Respiratory Medicine, Hamamatsu Medical Center, 328 Tomitsuka-cho, Naka-ku, Hamamatsu, Shizuoka 432-8580, Japan; Internal Medicine III, Wakayama Medical University, Wakayama City, Wakayama 641-0012, Japan.
Yasuhiro Koh, Internal Medicine III, Wakayama Medical University, Wakayama City, Wakayama, Japan; Center for Biomedical Sciences, Wakayama Medical University, Wakayama City, Wakayama, Japan.
Tetsunari Hase, Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan.
Kenji Chibana, Department of Respiratory Medicine, National Hospital Organization Okinawa National Hospital, Ginowan, Okinawa, Japan.
Kyoichi Kaira, Department of Respiratory Medicine, Comprehensive Cancer Center, Saitama Medical University International Medical Center, Hidaka, Saitama, Japan.
Kyoichi Okishio, Department of Internal Medicine, National Hospital Organization Kinki-chuo Chest Medical Center, Sakai City, Osaka, Japan.
Eiki Ichihara, Center for Clinical Oncology, Okayama University Hospital, Okayama, Okayama, Japan.
Shuji Murakami, Department of Thoracic Oncology, Kanagawa Cancer Center, Yokohama, Kanagawa, Japan.
Mototsugu Shimokawa, Department of Biostatistics, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan.
Nobuyuki Yamamoto, Internal Medicine III, Wakayama Medical University, Wakayama City, Wakayama, Japan.
Declarations
Ethics approval and consent to participate: The study was approved by the Ethics Committee of the Wakayama Medical University (No. 3073) and all the participating facilities. All patients signed an informed consent form before enrollment.
Consent for publication: Not applicable.
Author contributions: Yuichi Ozawa: Project administration; Conceptualization; Methodology; Writing – original draft.
Yasuhiro Koh: Investigation; Conceptualization; Methodology; Resources; Writing – review & editing.
Tetsunari Hase: Resources; Writing – review & editing.
Kenji Chibana: Resources; Writing – review & editing.
Kyoichi Kaira: Resources; Writing – review & editing.
Kyoichi Okishio: Resources; Writing – review & editing.
Eiki Ichihara: Resources; Writing – review & editing.
Shuji Murakami: Resources; Writing – review & editing.
Mototsugu Shimokawa: Formal analysis; Methodology; Writing – review & editing.
Nobuyuki Yamamoto: Supervision; Funding acquisition; Comceptualization; Methodology; Writing – review & editing.
Funding: The authors disclosed receipt of the following financial support for the research, authorship, and/or publication of this article: This study was funded by Takeda Pharmaceutical Company Limited.
All authors have completed the ICMJE uniform disclosure form at www.icmje.org/coi_disclosure.pdf: YO reported receiving honoraria from Chugai Pharmaceutical Co. Ltd., Takeda Pharmaceutical Company Limited, and Novartis, Japan. YK reported receiving research grants from Takeda Pharmaceutical Company, Ltd. and Chugai Pharmaceutical Co., Ltd., and honoraria from Takeda Pharmaceutical Company, Ltd., and Chugai Pharmaceutical Co., Ltd. TS reported receiving research grants from Chugai Pharmaceutical Co., Ltd. and Novartis Pharma K.K., and honoraria from Chugai Pharmaceutical Co., Ltd., Pfizer Japan Inc., and Takeda Pharmaceutical Company Ltd. KO reported receiving honoraria from Chugai Pharmaceutical Co., Ltd. and Takeda Pharmaceutical Company Ltd. EI reported receiving research grants from Takeda Pharmaceutical Company Ltd. and Pfizer Japan Inc., and honoraria from Chugai Pharmaceutical Co., Ltd., Takeda Pharmaceutical Company Ltd., Novartis Pharma K.K., and Pfizer Japan Inc. SM reported receiving grants from Chugai Pharmaceutical Co., Ltd. and Takeda Pharmaceutical Company Ltd., and personal fees from Pfizer Japan Inc., Chugai Pharmaceutical, and Takeda Pharmaceutical Company Ltd. NY reported receiving grants from Chugai Pharmaceutical, Novartis Pharma K.K., and Takeda Pharmaceutical Company Ltd., and honoraria from Chugai Pharmaceutical Co., Ltd., Novartis Pharma K.K., Pfizer Japan Inc., and Takeda Pharmaceutical Company Ltd.
Availability of data and materials: All data generated or analyzed during this study are included in this published article.
References
- 1. Bray F, Ferlay J, Soerjomataram I, et al. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2018; 68: 394–424. [DOI] [PubMed] [Google Scholar]
- 2. Ou SH, Bartlett CH, Mino-Kenudson M, et al. Crizotinib for the treatment of ALK-rearranged non-small cell lung cancer: a success story to usher in the second decade of molecular targeted therapy in oncology. Oncologist 2012; 17: 1351–1375. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Solomon BJ, Mok T, Kim DW, et al. First-line Crizotinib versus chemotherapy in ALK-positive lung cancer. N Engl J Med 2014; 371: 2167–2177. [DOI] [PubMed] [Google Scholar]
- 4. Shaw AT, Bauer TM, de Marinis F, et al. First-line Lorlatinib or Crizotinib in advanced ALK-positive lung cancer. N Engl J Med 2020; 383: 2018–2029. [DOI] [PubMed] [Google Scholar]
- 5. Camidge DR, Kim HR, Ahn MJ, et al. Brigatinib versus Crizotinib in ALK-positive non-small-cell lung cancer. N Engl J Med 2018; 379: 2027–2039. [DOI] [PubMed] [Google Scholar]
- 6. Peters S, Camidge DR, Shaw AT, et al. Alectinib versus Crizotinib in untreated ALK-positive non-small-cell lung cancer. N Engl J Med 2017; 377: 829–838. [DOI] [PubMed] [Google Scholar]
- 7. Horn L, Wang Z, Wu G, et al. Ensartinib vs Crizotinib for patients with anaplastic lymphoma kinase-positive non-small cell lung cancer: a randomized clinical trial. JAMA Oncol 2021; 7: 1617–1625. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Lin JJ, Zhu VW, Yoda S, et al. Impact of EML4-ALK variant on resistance mechanisms and clinical outcomes in ALK-positive lung cancer. J Clin Oncol 2018; 36: 1199–1206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Camidge DR, Dziadziuszko R, Peters S, et al. Updated efficacy and safety data and impact of the EML4-ALK fusion variant on the efficacy of alectinib in untreated ALK-positive advanced non-small cell lung cancer in the global phase III ALEX study. J Thorac Oncol 2019; 14: 1233–1243. [DOI] [PubMed] [Google Scholar]
- 10. Camidge DR, Kim HR, Ahn MJ, et al. Brigatinib versus Crizotinib in ALK inhibitor-naive advanced ALK-positive NSCLC: final results of phase 3 ALTA-1L trial. J Thorac Oncol 2021; 16: 2091–2108. [DOI] [PubMed] [Google Scholar]
- 11. Soo RA, Huat Tan E, Hayashi H, et al. Efficacy and safety of lorlatinib in Asian and non-Asian patients with ALK-positive advanced non-small cell lung cancer: subgroup analysis of a global phase 2 trial. Lung Cancer 2022; 169: 67–76. [DOI] [PubMed] [Google Scholar]
- 12. Bearz A, Martini JF, Jassem J, et al. Efficacy of Lorlatinib in treatment-naive patients with ALK-positive advanced NSCLC in relation to EML4::ALK variant type and ALK with or without TP53 mutations. J Thorac Oncol 2023; 18: 1581–1593. [DOI] [PubMed] [Google Scholar]
- 13. Gainor JF, Dardaei L, Yoda S, et al. Molecular mechanisms of resistance to first- and second-generation ALK inhibitors in ALK-rearranged lung cancer. Cancer Discov 2016; 6: 1118–1133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Elshatlawy M, Sampson J, Clarke K, et al. EML4-ALK biology and drug resistance in non-small cell lung cancer: a new phase of discoveries. Mol Oncol 2023; 17: 950–963. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Horn L, Whisenant JG, Wakelee H, et al. Monitoring therapeutic response and resistance: analysis of circulating tumor DNA in patients with ALK+ lung cancer. J Thorac Oncol 2019; 14: 1901–1911. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Lin JJ, Zhu VW, Schoenfeld AJ, et al. Brigatinib in patients with alectinib-refractory ALK-positive NSCLC. J Thorac Oncol 2018; 13: 1530–1538. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Zhu VW, Nagasaka M, Madison R, et al. A novel sequentially evolved EML4-ALK variant 3 G1202R/S1206Y double mutation in cis confers resistance to Lorlatinib: a brief report and literature review. JTO Clin Res Rep 2021; 2: 100116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Yoda S, Lin JJ, Lawrence MS, et al. Sequential ALK inhibitors can select for Lorlatinib-resistant compound ALK mutations in ALK-positive lung cancer. Cancer Discov 2018; 8: 714–729. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Shiba-Ishii A, Johnson TW, Dagogo-Jack I, et al. Analysis of lorlatinib analogs reveals a roadmap for targeting diverse compound resistance mutations in ALK-positive lung cancer. Nat Cancer 2022; 3: 710–722. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Katayama R, Shaw AT, Khan TM, et al. Mechanisms of acquired Crizotinib resistance in ALK-rearranged lung Cancers. Sci Transl Med 2012; 4: 120ra117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Crystal AS, Shaw AT, Sequist LV, et al. Patient-derived models of acquired resistance can identify effective drug combinations for cancer. Science 2014; 346: 1480–1486. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Christopoulos P, Budczies J, Kirchner M, et al. Defining molecular risk in ALK(+) NSCLC. Oncotarget 2019; 10: 3093–3103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Yoshida R, Sasaki T, Minami Y, et al. Activation of Src signaling mediates acquired resistance to ALK inhibition in lung cancer. Int J Oncol 2017; 51: 1533–1540. [DOI] [PubMed] [Google Scholar]
- 24. Miyawaki M, Yasuda H, Tani T, et al. Overcoming EGFR bypass signal-induced acquired resistance to ALK tyrosine kinase inhibitors in ALK-translocated lung cancer. Mol Cancer Res 2017; 15: 106–114. [DOI] [PubMed] [Google Scholar]
- 25. Yu Y, Ou Q, Wu X, et al. Concomitant resistance mechanisms to multiple tyrosine kinase inhibitors in ALK-positive non-small cell lung cancer. Lung Cancer 2019; 127: 19–24. [DOI] [PubMed] [Google Scholar]
- 26. Rotow J, Bivona TG. Understanding and targeting resistance mechanisms in NSCLC. Nat Rev Cancer 2017; 17: 637–658. [DOI] [PubMed] [Google Scholar]
- 27. Kogita A, Togashi Y, Hayashi H, et al. Hypoxia induces resistance to ALK inhibitors in the H3122 non-small cell lung cancer cell line with an ALK rearrangement via epithelial-mesenchymal transition. Int J Oncol 2014; 45: 1430–1436. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Jia Y, Li X, Zhao C, et al. Impact of serum vascular endothelial growth factor and interleukin-6 on treatment response to epidermal growth factor receptor tyrosine kinase inhibitors in patients with non-small-cell lung cancer. Lung Cancer 2018; 125: 22–28. [DOI] [PubMed] [Google Scholar]
- 29. Patel SA, Nilsson MB, Yang Y, et al. IL-6 mediates suppression of T and NK cells function in EMT-associated TKI-resistant EGFR mutant NSCLC<. Clin Cancer Res 2023; 29: 1292–1304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Taniguchi H, Takeuchi S, Fukuda K, et al. Amphiregulin triggered epidermal growth factor receptor activation confers in vivo Crizotinib-resistance of EML4-ALK lung cancer and circumvention by epidermal growth factor receptor inhibitors. Cancer Sci 2017; 108: 53–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Arai S, Takeuchi S, Fukuda K, et al. Osimertinib overcomes alectinib resistance caused by amphiregulin in a leptomeningeal carcinomatosis model of ALK-rearranged lung cancer. J Thorac Oncol 2020; 15: 752–765. [DOI] [PubMed] [Google Scholar]
- 32. Liu SY, Dong ZY, Wu SP, et al. Clinical relevance of PD-L1 expression and CD8+ T cells infiltration in patients with EGFR-mutated and ALK-rearranged lung cancer. Lung Cancer 2018; 125: 86–92. [DOI] [PubMed] [Google Scholar]
- 33. Yoneshima Y, Ijichi K, Anai S, et al. PD-L1 expression in lung adenocarcinoma harboring EGFR mutations or ALK rearrangements. Lung Cancer 2018; 118: 36–40. [DOI] [PubMed] [Google Scholar]
- 34. Nigro A, Ricciardi L, Salvato I, et al. Enhanced expression of CD47 is associated with off-target resistance to tyrosine kinase inhibitor gefitinib in NSCLC. Front Immunol 2019; 10: 3135. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Stinchcombe T, Doebele RC, Wang XF, et al. Preliminary results of single arm phase 2 trial of brigatinib in patients (pts) with progression disease (PD) after next-generation (NG) anaplastic lymphoma kinase (ALK) tyrosine kinase inhibitors (TKIs) in ALK + non-small cell lung cancer (NSCLC). J Clin Oncol 2019; 37(suppl): 9027. [Google Scholar]
- 36. Lin JJ, Schoenfeld AJ, Zhu VW, et al. Efficacy of platinum/pemetrexed combination chemotherapy in ALK-positive NSCLC refractory to second-generation ALK inhibitors. J Thorac Oncol 2020; 15: 258–265. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Ou SI, Nishio M, Ahn MJ, et al. Efficacy of brigatinib in patients with advanced ALK-positive NSCLC who progressed on alectinib or ceritinib: ALK in lung cancer trial of brigatinib-2 (ALTA-2). J Thorac Oncol 2022; 17: 1404–1414. [DOI] [PubMed] [Google Scholar]
- 38. Nishio M, Yoshida T, Kumagai T, et al. Brigatinib in Japanese patients with ALK-positive non-small cell lung cancer previously treated with alectinib and other tyrosine kinase inhibitors: outcomes of the phase 2 J-ALTA trial. J Thorac Oncol 2021; 16: 452–463. [DOI] [PubMed] [Google Scholar]
- 39. Sugawara S, Kondo M, Yokoyama T, et al. Brigatinib in Japanese patients with tyrosine kinase inhibitor-naive ALK-positive non-small cell lung cancer: first results from the phase 2 J-ALTA study. Int J Clin Oncol 2022; 27: 1828–1838. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Schmid S, Cheng S, Chotai S, et al. Real-world treatment sequencing, toxicities, health utilities, and survival outcomes in patients with advanced ALK-rearranged non-small-cell lung cancer. Clin Lung Cancer 2023; 24: 40–50. [DOI] [PubMed] [Google Scholar]
- 41. Kunimasa K, Hirotsu Y, Kukita Y, et al. EML4-ALK fusion variant.3 and co-occurrent PIK3CA E542K mutation exhibiting primary resistance to three generations of ALK inhibitors. Cancer Genet 2021; 256–257: 131–135. [DOI] [PubMed] [Google Scholar]
- 42. Doebele RC, Pilling AB, Aisner DL, et al. Mechanisms of resistance to crizotinib in patients with ALK gene rearranged non-small cell lung cancer. Clin Cancer Res 2012; 18: 1472–1482. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Basch E. The missing voice of patients in drug-safety reporting. N Engl J Med 2010; 362: 865–869. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Supplemental material, sj-docx-1-tam-10.1177_17588359231225046 for Prospective observational study to explore genes and proteins predicting efficacy and safety of brigatinib for ALK-gene rearranged non-small-cell lung cancer: study protocol for ABRAID study (WJOG11919L) by Yuichi Ozawa, Yasuhiro Koh, Tetsunari Hase, Kenji Chibana, Kyoichi Kaira, Kyoichi Okishio, Eiki Ichihara, Shuji Murakami, Mototsugu Shimokawa and Nobuyuki Yamamoto in Therapeutic Advances in Medical Oncology

