Distinct metastatic spread and progression patterns in patients treated with crizotinib for ROS1- and ALK-rearranged non-small cell lung cancer: a single-center retrospective study
Original Article

Distinct metastatic spread and progression patterns in patients treated with crizotinib for ROS1- and ALK-rearranged non-small cell lung cancer: a single-center retrospective study

Tomoaki Nakamura1,2^, Tatsuya Yoshida1^, Yuki Takeyasu1,3, Ken Masuda1, Yuki Sinno1, Yuji Matsumoto1,4, Yusuke Okuma1, Yasushi Goto1, Hidehito Horinouchi1, Noboru Yamamoto1, Yuichiro Ohe1

1Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan; 2Department of Pulmonary Medicine, Thoracic Center, St. Luke’s International Hospital, Tokyo, Japan; 3Course of Advanced Clinical Research of Cancer, Juntendo University Graduate School of Medicine, Tokyo, Japan; 4Department of Endoscopy, Respiratory Endoscopy Division, National Cancer Center Hospital, Tokyo, Japan

Contributions: (I) Conception and design: T Nakamura, T Yoshida, Y Takeyasu; (II) Administrative support: T Nakamura, T Yoshida; (III) Provision of study materials or patients: All authors; (IV) Collection and assembly of data: T Nakamura, Y Takeyasu; (V) Data analysis and interpretation: T Nakamura, T Yoshida, Y Takeyasu; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors.

^ORCID: Tomoaki Nakamura, 0000-0002-4248-8601; Tatsuya Yoshida, 0000-0003-4896-5824.

Correspondence to: Tatsuya Yoshida, MD, PhD. Department of Thoracic Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo, Tokyo, Japan. Email: tatyoshi@ncc.go.jp.

Background: Crizotinib has been approved for C-ros oncogene 1 (ROS1)- and anaplastic lymphoma kinase (ALK)-rearranged non-small cell lung cancer (NSCLC) patients. Few studies have examined the differences in crizotinib treatment outcomes between these patients and the progression sites during treatment. We investigated the metastatic spread, crizotinib efficacy, and progression patterns during crizotinib treatment in ROS1- and ALK-rearranged NSCLC patients.

Methods: We retrospectively reviewed crizotinib-treated ROS1- and ALK-rearranged NSCLC patients between January 2011 and March 2021. Patient characteristics, clinical outcomes, and progression patterns during treatment were collected from medical records. The metastasis extent, crizotinib response, and progression patterns between the groups were compared.

Results: We identified 26 patients with ROS1- and 42 with ALK-positive NSCLC. The baseline proportion of central nervous system (CNS) metastases did not differ between the groups (12% vs. 29%, P=0.10), but the proportion of extrathoracic metastases, including CNS metastases, was significantly higher in ALK-positive than in ROS1-positive NSCLC patients (35% vs. 71%, P=0.003). Regarding the response to crizotinib, the objective response rate (ORR), progression-free survival (PFS), or overall survival (OS) did not significantly differ between the groups (ROS1 vs. ALK, ORR: 69% vs. 69%, P=0.987; PFS: median 10.9 vs. 10.7 months, P=0.232; median OS: not reached vs. 67.7 months, P=0.495). The CNS was the most common metastasis site in both groups [ROS1 vs. ALK, 69% (11/16) vs. 46% (17/37), P=0.127], and the cumulative incidence of CNS metastasis did not differ between the groups (P=0.914).

Conclusions: Crizotinib treatment outcomes, including progression patterns, were similar between ROS1- and ALK-positive NSCLC patients.

Keywords: ROS1-rearrangement; non-small cell lung cancer (NSCLC); crizotinib; progression pattern; CNS metastasis


Submitted Jan 10, 2023. Accepted for publication May 30, 2023. Published online Jun 30, 2023.

doi: 10.21037/tlcr-23-10


Highlight box

Key findings

• We evaluated the distinct clinical features in ROS1- and ALK- positive NSCLC patient.

What is known and what is new?

• Oncogenic rearrangement in ROS1 is structurally homologous to the tyrosine kinase region of ALK.

• We demonstrated that metastatic spread at initial diagnosis differed between patients with ROS1- and ALK-positive NSCLC, but the clinical outcomes with crizotinib treatment and progression patterns, including that of CNS progression, were similar.

What is the implication, and what should change now?

• Continued efforts to develop novel TKIs that can overcome resistance to crizotinib and exhibit better intracranial activity are needed.


Introduction

Oncogenic rearrangement in C-ros oncogene 1 (ROS1), which is structurally homologous to the tyrosine kinase region of anaplastic lymphoma kinase (ALK), occur in approximately 2% to 3% of NSCLC patients (1,2). Crizotinib, which was first approved for patients with advanced NSCLC with ALK rearrangements, binds with high affinity to ROS1, and potently inhibits ROS1 signaling and cell viability (3-5). In clinical setting, crizotinib produced median PFS duration of 15.8 to 22.8 month in patients with advanced ROS1-positive NSCLC, while the median PFS of 7.7 to 10.9 months in patients with advanced ALK-positive NSCLC, based on the results of several clinical trials (2,6-11). Crizotinib received approval for ROS1-positive NSCLC. Subsequently, entrectinib, a ROS1 tyrosine kinase inhibitor (TKI), was approved for the treatment of ROS1-rearranged NSCLC.

ROS1 rearrangements have been associated with a lower incidence of extrathoracic metastases, including CNS metastases at initial diagnosis and during treatment, compared to ALK rearrangements (12). In contrast, similar frequencies of CNS metastases have been reported in patients with ALK and ROS1 rearrangements (13). ALK rearrangements were associated with pericardial, pleural, and liver metastases compared to EGFR/KRAS/ALK wild-type alterations (14). Additionally, in ALK-positive NSCLC patients, the CNS was the most common metastatic site during crizotinib treatment (15). In ALK-positive NSCLC patients, next-generation ALK inhibitors have been designed to more strongly inhibit the ALK native kinase, be active on crizotinib-induced resistance mutations, and have better blood-brain barrier (BBB) penetration to control or prevent CNS involvement. The second-generation inhibitors, alectinib and brigatinib, have become the standard first-line treatment. In contrast, crizotinib and entrectinib are still the standard of care in ROS1 positive NSCLC patients, but due to the resistance of these TKIs, the development of next-generation ROS1 TKIs is underway.

In this study, to evaluate the distinct clinical features in ROS1- and ALK-positive NSCLC patients, we investigated the baseline metastatic spread, crizotinib efficacy, and progression patterns during crizotinib treatment. This article is presented in accordance with the STROBE reporting checklist (available at https://tlcr.amegroups.com/article/view/10.21037/tlcr-23-10/rc).


Methods

Patients

We retrospectively reviewed and included all advanced ROS1- and ALK-positive NSCLC patients who had been treated with crizotinib at the National Cancer Center Hospital from January 2011 to March 2021. There were no exclusion criteria. Data on patient characteristics, metastatic site at the baseline, tumor response to crizotinib, and progression patterns during crizotinib treatment were collected from medical records. ROS1 and ALK rearrangements were identified by fluorescence in situ hybridization, targeted next-generation sequencing, immunohistochemistry, and/or real-time polymerase chain reaction (ROS1 only), as described in previous study (12).

Statistical analysis

PFS was measured as the time from the start of crizotinib treatment to radiographic disease progression (as assessed by the investigator) or death if no disease progression was documented. Patients who survived without documented disease progression were censored at the last follow-up visit. Overall survival (OS) was measured from the initiation of crizotinib treatment to death. Patients with undocumented dates of death were censored from the data of the last visit. Tumor response was assessed according to the Response Evaluation Criteria in Solid Tumors (RECIST) version 1.1 (16). The objective response rate (ORR) was defined as the proportion of patients who had a complete response (CR) or partial response (PR). The disease control rate (DCR) was defined as the proportion of patients who achieved CR, PR, or stable disease. Descriptive statistics were used for categorical (frequency and proportion) and continuous variables (median and range). Kaplan-Meier curves and estimates were used to assess the OS, PFS, and cumulative incidence of CNS progression during crizotinib treatment for ROS1- or ALK-positive NSCLC. Differences in OS, PFS, and CNS progression were assessed using log-rank tests. All statistical analyses were conducted using the Statistical Package for Social Sciences (SPSS/Windows, Version 19.0, SPSS Inc., Chicago, USA). All tests were two-tailed, and statistical significance was set at P<0.05. This study was conducted in accordance with the Declaration of Helsinki (as revised in 2013). The study was approved by the Ethics Committee of the National Cancer Center Hospital (No. 2015-355) and individual consent for this retrospective analysis was waived.


Results

Patients characteristics at baseline

We identified 26 ROS1-positive NSCLC and 42 ALK-positive NSCLC patients who had been treated with crizotinib. Patient characteristics are presented in Table 1. The baseline characteristics were similar between both groups. In ROS1-positive NSCLC, crizotinib was administered to 10 patients (38%) as a first-line treatment, 6 patients (23%) as a second-line treatment, and 10 patients (38%) as a third-line or later treatment. In ALK-positive NSCLC, crizotinib was administered to 23 patients (55%) as a first-line treatment, 11 patients (26%) as a second-line treatment, and 8 patients (19%) as a third-line or later treatment (Table 1).

Table 1

Patient and disease characteristics

Characteristics ROS1, n=26 ALK, n=42 P value
Age, years, median at diagnosis of advanced disease [range] 56 [36–82] 56 [36–82]
Sex, n [%]
   Male/female 10 [38]/16 [62] 19 [45]/23 [55] 0.583
Smoking history, n [%]
   Never/light (<10 pack years)/heavy (≥10 pack years) 15 [58]/5 [19]/6 [23] 26 [62]/5 [12]/11 [26] 0.706
Histopathology, n [%]
   Adenocarcinoma/others 23 [88]/3 [12] 42 [100]/0 [0] 0.052
Stage at diagnosis, n [%]
   IIIa–IV/recurrence 20 [77]/6 [23] 30 [71]/12 [29] 0.618
ECOG performance states, n [%]
   0–1/≥2 24 [92]/2 [8] 37 [88]/5 [12] 0.618
Brain metastasis at diagnosis, n [%]
   Present/absent 3 [12]/23 [88] 12 [29]/30 [71] 0.100
Crizotinib treatment line, n [%]
   1/2/≥3 10 [38]/6 [23]/10 [38] 23 [55]/11 [26]/8 [19] 0.199

a, 5 patients in ROS1-positive NSCLC and 2 patients in ALK-positive NSCLC. ECOG, Eastern Cooperative Oncology Group.

At the baseline, patients with ROS1- and ALK-positive NSCLC had similar intrathoracic metastasis spread (Figure 1A). However, patients with ROS1-positive NSCLC had significantly lower rates of extrathoracic metastases, including CNS metastasis, especially that of extrathoracic lymph node metastases (ROS1, 35%; ALK, 71%; P=0.003; Figure 1B and ROS1, 0%; ALK, 17%; P=0.028; Figure 1C). Regarding CNS metastasis at baseline, three patients with ROS1-positive NSCLC had previously been treated with radiotherapy or surgery, and three patients had untreated brain metastases at the initiation of crizotinib. All of these patients achieved a reduction in brain metastasis with crizotinib. Among patients with ALK-positive NSCLC, 12 had been previously treated for brain metastases before receiving crizotinib, and three had untreated brain metastases at its initiation. The percentage of brain metastases was also lower in ROS1-positive patients, although the difference was not statistically significant (ROS1, 12%; ALK, 29%; P=0.100; Figure 1D).

Figure 1 Frequency of metastatic sites of malignant disease at initial metastatic diagnosis. Frequency of (A) intrathoracic sites, (B) extrathoracic sites including CNS metastases, (C) extrathoracic sites, and (D) brain metastases. ROS1, C-ros oncogene 1; ALK, anaplastic lymphoma kinase; CNS, central nervous system.

Efficacy of crizotinib in ROS1 and ALK-positive NSCLC patients

The ORR and DCR for crizotinib in ROS1-positive patients were 69% and 85%, respectively (Table 2). In ALK-positive patients, the ORR and DCR were 69% and 81%, respectively. There were no significant differences in tumor response and PFS with crizotinib between the two groups (ORR, P=0.987; DCR, P=0.484; median PFS: 10.9 vs. 10.7 months, P=0.232) (Figure 2A). The OS did not significantly differ between ROS1- and ALK-positive NSCLC patients [not reached (95% CI: NR–NR), and 67.7 months (95% CI: 47.6–87.8), respectively (P=0.495)] (Figure 2B).

Table 2

Best response to crizotinib therapy

Efficacy ROS1, n=26 ALK, n=42 P value
Response
   CR 2 (8%) 5 (12%)
   PR 16 (62%) 24 (57%)
   SD 4 (15%) 5 (12%)
   PD 2 (8%) 3 (7%)
   NE 2 (8%) 5 (12%)
Clinical benefit
   ORR 69% 69% 0.987
   DCR 85% 81% 0.484

ALK, anaplastic lymphoma kinase; CR, complete response; PR, partial response; SD, stable disease; PD, progressive disease; NE, not estimable; ORR, objective response rate; DCR, disease control rate.

Figure 2 Kaplan-Meier curves for PFS and OS of patients with ROS1- and ALK-positive NSCLC. (A) PFS with crizotinib treatment (any line of therapy). (B) OS with crizotinib treatment (any line of therapy). PFS, progression-free survival; OS, overall survival; ROS1, C-ros oncogene 1; ALK, anaplastic lymphoma kinase; NSCLC, non-small cell lung cancer.

Progression patterns during crizotinib treatment in ROS1 and ALK-positive NSCLC

Data on progression patterns were available for 16 patients with ROS1-positive and 37 with ALK-positive NSCLC (Figure 3). Progression patterns were similar between the two groups. Brain metastases, including isolated CNS metastasis, were the most common sites of progression in both groups (ROS1, 69%; ALK, 46%; P=0.127). The cumulative incidence of CNS progression in patients with ROS1- and ALK-positive NSCLC during crizotinib treatment is presented in Figure 4. There were no significant differences between the two groups for all patients (P=0.914) or for patients with no CNS metastasis at baseline (P=0.437).

Figure 3 Progression pattern of ROS1- and ALK-positive NSCLC during crizotinib treatment. Frequency of occurrence of (A) intrathoracic and (B) extrathoracic sites, and (C) brain metastases. ROS1, C-ros oncogene 1; ALK, anaplastic lymphoma kinase; CNS, central nervous system; NSCLC, non-small cell lung cancer.
Figure 4 Cumulative incidence of CNS progression (probability) in patients with ROS1- and ALK-positive NSCLC during crizotinib treatment. (A) Cumulative incidence of CNS progression in all patients. (B) Cumulative incidence of CNS progression in patients without brain metastasis at the time of crizotinib initiation. ROS1, C-ros oncogene 1; ALK, anaplastic lymphoma kinase; CNS, central nervous system; NSCLC, non-small cell lung cancer.

Discussion

In this study, we retrospectively evaluated the metastatic spread at baseline and progression patterns during crizotinib treatment in patients with advanced ROS1- and ALK-positive NSCLC. The clinical outcomes of crizotinib were similar in both groups. Additionally, CNS was the most common progression site, and progression patterns during crizotinib treatment in both patient groups were similar.

Two phase II studies evaluating the efficacy of crizotinib in ROS1 fusion-positive NSCLC patients showed that PFS was 15.9 to 22.8 months, while phase III trials revealed that it was 11 months in patients with treatment-naive advanced ALK fusion-positive NSCLC (7,10). Although direct comparison between clinical trials is challenging due to the varying trial designs, the differences in the clinical outcomes with crizotinib treatment between patients with ROS1- and ALK-positive NSCLC could be related to the difference in inhibitory effects of this drug. Indeed, crizotinib had better inhibitory effect on ROS1-dependent cell lines than on ALK-dependent cell lines (1,3). Another reason could be the differences in metastatic spread at baseline. Gainor et al. reported that patients with ROS1-positive disease had significantly lower rates of extrathoracic metastases, including lower rates of brain metastases at the initial metastatic diagnosis, which was consistent with the results of our study (12). Regarding progression patterns during treatment, patients with ROS1-positive NSCLC had a significantly lower cumulative incidence of brain metastases over time. In contrast, Patil et al. reported that there were no statistically significant differences in CNS progression in ROS1- and ALK-positive patients, who were taking crizotinib (13). Our study showed that the progression patterns during crizotinib treatment in ROS1- and ALK-positive NSCLC were similar, and there were no significant differences in the incidence of CNS progression. The difference in the clinical outcomes with crizotinib, including the progression sites, between patients with ROS1- and ALK-positive NSCLC remains controversial.

Crizotinib poorly penetrates the cerebrospinal fluid (CSF) (17). In ALK-positive NSCLC patients, the CNS is the common initial progression site, including isolated progression, and brain metastasis status was significantly associated with both PFS in crizotinib-treated patients and CNS progression. In patients with ROS1-positive NSCLC, it remained unclear how baseline brain metastasis affects the clinical outcomes with crizotinib because two phase II trials evaluating the efficacy of crizotinib treatment in ROS1-positive NSCLC excluded patients with untreated CNS metastases (2,10). The METROS study showed that among six ROS1-positive NSCLC patients with intracranial disease, the intracranial ORR was 33%, and five patients experienced intracranial progression as a pattern of failure (8). In our study, six patients with ROS1-positive NSCLC with brain metastases at baseline, regardless of prior radiotherapy for CNS metastases, and four patients developed CNS progression. Our study showed that the presence of CNS metastasis at baseline in ROS1 fusion-positive NSCLC affected the clinical outcome with crizotinib. Although the biology of brain metastasis is poorly understood, metastases to the CNS, unlike metastases to other distal organ sites, involve the breach of the BBB. BBB effectively prevents the free exchange of substances between the blood and the interstitial fluid of the brain (18-20). The CNS is considered a sanctuary site for metastatic cancer cells because many therapeutic agents cannot cross the BBB. A better understanding of molecular biology of lung cancer metastasis to the brain and efficient drug delivery across the BBB will be essential to find the appropriate treatment strategy for prevention of brain metastasis.

The present study had several limitations. The sample size was small, and the study was conducted in a single institute, which included potential confounders, such as the number of lines of prior treatment and treatment duration. Moreover, although the prognostic impact of distinct fusion genes on clinical outcomes has been reported, details of the fusion types were unavailable (21-23).


Conclusions

We demonstrated that metastatic spread at initial diagnosis differed between patients with ROS1- and ALK-positive NSCLC, but the clinical outcomes with crizotinib treatment and progression patterns, including that of CNS progression, were similar. Continued efforts to develop novel TKIs that can overcome resistance to crizotinib and exhibit better intracranial activity are needed.


Acknowledgments

The authors greatly appreciate the assistance of the staff of the Department of Thoracic Oncology and Division of Molecular Pharmacology of the National Cancer Center Hospital.

Funding: None.


Footnote

Reporting Checklist: The authors have completed the STROBE reporting checklist. Available at https://tlcr.amegroups.com/article/view/10.21037/tlcr-23-10/rc

Data Sharing Statement: Available at https://tlcr.amegroups.com/article/view/10.21037/tlcr-23-10/dss

Peer Review File: Available at https://tlcr.amegroups.com/article/view/10.21037/tlcr-23-10/prf

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://tlcr.amegroups.com/article/view/10.21037/tlcr-23-10/coif). TY reports receiving grants and personal fees from Amgen, AstraZeneca, Ono, Merck Sharp & Dohme, Novartis, Chugai, and Bristol-Myers Squibb; grants from Takeda, Daiichi Sankyo, and AbbVie; and personal fees from Taiho, Eli Lilly, Roche, and ArcherDX outside of the submitted work. KM reports receiving personal fees from Ono Pharmaceutical Co., Ltd., AstraZeneca, Chugai, and Bristol-Myers Squibb, outside of the submitted work. YS reports receiving personal fees from Bristol-Myers Squibb, Chugai, AstraZeneca, and Eli Lilly; grants and personal fees from Ono; and grants from Janssen and Japan Clinical Research Operations K.K. outside of the submitted work. YM reports receiving grants from the National Cancer Center Research and Development Fund, Grant-in-Aid for Scientific Research on Innovative Areas, and Hitachi, Ltd.; grants and personal fees from Olympus; and personal fees from AstraZeneca, Novartis, COOK, AMCO Inc., Thermo Fisher Scientific, Erbe Elektromedizin GmbH, Fujifilm, Chugai, and Eli Lilly outside of the submitted work. Yusuke Okuma reports receiving grants from Roche and AbbVie K.K.; and personal fees from AstraZeneca, Ely Lilly K.K., Bristol-Myers Squibb, Pfizer Taiho Pharma Co. Ltd., AstraZeneca Nippon Boehringer Ingelheim, Chugai Pharma Co. Ltd., Ono Pharma Co. Ltd., and Taiho Pharma Co. Ltd. outside of the submitted work. YG reports receiving grants from AZK, AbbVie, Kyorin, and Preferred Network; grants and personal fees from Pfizer, Eli Lilly, Bristol-Myers Squibb, Ono, Novartis, and Daiichi Sankyo; and personal fees from Chugai, Taiho, Boehringer Ingelheim, Merck Sharp & Dohme, Merck, Thermo Fisher, AstraZeneca, Chugai, Guardant Health Inc., and Illumina outside of the submitted work. HH reports receiving grants and personal fees from Merck Sharp & Dohme, AstraZeneca, Ono, Chugai, Roche, and Novartis; grants from AbbVie, Bristol-Myers Squibb, Merck Biopharma, Daiichi Sankyo, Janssen, and Genomic Health; and personal fees from Eli Lilly and Kyowa-Kirin, outside of the submitted work. NY reports receiving grants from Chugai, Taiho, Eisai, Eli Lilly, Quintiles, Astellas, Bristol-Myers Squibb, Novartis, Daiichi Sankyo, Pfizer, Boehringer Ingelheim, Kyowa-Hakko Kirin, Bayer, Ono Pharmaceutical Co., Ltd., Takeda, Janssen Pharma, Merck Sharp & Dohme, Merck, GlaxoSmithKline, Sumitomo Dainippon, Chiome Bioscience Inc., Otsuka, Carna Biosciences, Genmab, and Shionogi; and personal fees from Ono Pharmaceutical Co., Ltd., Chugai, AstraZeneca, Pfizer, Lilly, Bristol-Myers Squibb, Eisai, Otsuka, Takeda, Boehringer Ingelheim, Cimic, Sysmex, and Eisai, outside of the submitted work. Yuichiro Ohe reports receiving grants, personal fees, and nonfinancial support from AstraZeneca, Chugai, Ono Pharmaceutical Co., Ltd., and Bristol-Myers Squibb; grants and personal fees from Eli Lilly and Pfizer; grants and nonfinancial support from Kyorin; grants from Dainippon-Sumitomo, Taiho, Novartis, Takeda, Kissei, Daiichi Sankyo, Janssen, and LOXO; and personal fees from Boehringer Ingelheim, Bayer, Merck Sharp & Dohme, Taiho, Nippon Kayaku, Kyowa-Hakko Kirin, Celltrion, Amgen, and AnHeeart Therapeutics Inc. outside of the submitted work. The other 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. The study was conducted in accordance with the Declaration of Helsinki (as revised in 2013). The study was approved by institutional ethics board of National Cancer Center Hospital (No. 2015-355) and individual consent for this retrospective analysis was waived.

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. Ou SH, Tan J, Yen Y, et al. ROS1 as a 'druggable' receptor tyrosine kinase: lessons learned from inhibiting the ALK pathway. Expert Rev Anticancer Ther 2012;12:447-56. [Crossref] [PubMed]
  2. Shaw AT, Ou SH, Bang YJ, et al. Crizotinib in ROS1-rearranged non-small-cell lung cancer. N Engl J Med 2014;371:1963-71. [Crossref] [PubMed]
  3. Davare MA, Vellore NA, Wagner JP, et al. Structural insight into selectivity and resistance profiles of ROS1 tyrosine kinase inhibitors. Proc Natl Acad Sci U S A 2015;112:E5381-90. [Crossref] [PubMed]
  4. Huber KV, Salah E, Radic B, et al. Stereospecific targeting of MTH1 by (S)-crizotinib as an anticancer strategy. Nature 2014;508:222-7. [Crossref] [PubMed]
  5. Bergethon K, Shaw AT, Ou SH, et al. ROS1 rearrangements define a unique molecular class of lung cancers. J Clin Oncol 2012;30:863-70. [Crossref] [PubMed]
  6. Shaw AT, Riely GJ, Bang YJ, et al. Crizotinib in ROS1-rearranged advanced non-small-cell lung cancer (NSCLC): updated results, including overall survival, from PROFILE 1001. Ann Oncol 2019;30:1121-6. [Crossref] [PubMed]
  7. Solomon BJ, Mok T, Kim DW, et al. First-line crizotinib versus chemotherapy in ALK-positive lung cancer. N Engl J Med 2014;371:2167-77. [Crossref] [PubMed]
  8. Landi L, Chiari R, Tiseo M, et al. Crizotinib in MET-Deregulated or ROS1-Rearranged Pretreated Non-Small Cell Lung Cancer (METROS): A Phase II, Prospective, Multicenter, Two-Arms Trial. Clin Cancer Res 2019;25:7312-9. [Crossref] [PubMed]
  9. Michels S, Massutí B, Schildhaus HU, et al. Safety and Efficacy of Crizotinib in Patients With Advanced or Metastatic ROS1-Rearranged Lung Cancer (EUCROSS): A European Phase II Clinical Trial. J Thorac Oncol 2019;14:1266-76. [Crossref] [PubMed]
  10. Wu YL, Yang JC, Kim DW, et al. Phase II Study of Crizotinib in East Asian Patients With ROS1-Positive Advanced Non-Small-Cell Lung Cancer. J Clin Oncol 2018;36:1405-11. [Crossref] [PubMed]
  11. Shaw AT, Kim DW, Nakagawa K, et al. Crizotinib versus chemotherapy in advanced ALK-positive lung cancer. N Engl J Med 2013;368:2385-94. [Crossref] [PubMed]
  12. Gainor JF, Tseng D, Yoda S, et al. Patterns of Metastatic Spread and Mechanisms of Resistance to Crizotinib in ROS1-Positive Non-Small-Cell Lung Cancer. JCO Precis Oncol 2017;2017:PO.17.00063.
  13. Patil T, Smith DE, Bunn PA, et al. The Incidence of Brain Metastases in Stage IV ROS1-Rearranged Non-Small Cell Lung Cancer and Rate of Central Nervous System Progression on Crizotinib. J Thorac Oncol 2018;13:1717-26. [Crossref] [PubMed]
  14. Doebele RC, Lu X, Sumey C, et al. Oncogene status predicts patterns of metastatic spread in treatment-naive nonsmall cell lung cancer. Cancer 2012;118:4502-11. [Crossref] [PubMed]
  15. Yoshida T, Oya Y, Tanaka K, et al. Clinical impact of crizotinib on central nervous system progression in ALK-positive non-small lung cancer. Lung Cancer 2016;97:43-7. [Crossref] [PubMed]
  16. Eisenhauer EA, Therasse P, Bogaerts J, et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer 2009;45:228-47. [Crossref] [PubMed]
  17. Costa DB, Kobayashi S, Pandya SS, et al. CSF concentration of the anaplastic lymphoma kinase inhibitor crizotinib. J Clin Oncol 2011;29:e443-5. [Crossref] [PubMed]
  18. Hanibuchi M, Kim SJ, Fidler IJ, et al. The molecular biology of lung cancer brain metastasis: an overview of current comprehensions and future perspectives. J Med Invest 2014;61:241-53. [Crossref] [PubMed]
  19. Preusser M, Winkler F, Valiente M, et al. Recent advances in the biology and treatment of brain metastases of non-small cell lung cancer: summary of a multidisciplinary roundtable discussion. ESMO Open 2018;3:e000262. [Crossref] [PubMed]
  20. Huang RSP, Haberberger J, McGregor K, et al. Clinicopathologic and Genomic Landscape of Breast Carcinoma Brain Metastases. Oncologist 2021;26:835-44. [Crossref] [PubMed]
  21. Kang J, Zhang XC, Chen HJ, et al. Complex ALK Fusions Are Associated With Better Prognosis in Advanced Non-Small Cell Lung Cancer. Front Oncol 2020;10:596937. [Crossref] [PubMed]
  22. Li Z, Shen L, Ding D, et al. Efficacy of Crizotinib among Different Types of ROS1 Fusion Partners in Patients with ROS1-Rearranged Non-Small Cell Lung Cancer. J Thorac Oncol 2018;13:987-95. [Crossref] [PubMed]
  23. Park S, Ahn BC, Lim SW, et al. Characteristics and Outcome of ROS1-Positive Non-Small Cell Lung Cancer Patients in Routine Clinical Practice. J Thorac Oncol 2018;13:1373-82. [Crossref] [PubMed]
Cite this article as: Nakamura T, Yoshida T, Takeyasu Y, Masuda K, Sinno Y, Matsumoto Y, Okuma Y, Goto Y, Horinouchi H, Yamamoto N, Ohe Y. Distinct metastatic spread and progression patterns in patients treated with crizotinib for ROS1- and ALK-rearranged non-small cell lung cancer: a single-center retrospective study. Transl Lung Cancer Res 2023;12(7):1436-1444. doi: 10.21037/tlcr-23-10

Download Citation