INTRODUCTION
The therapeutic landscape of non-small-cell lung cancer (NSCLC) has witnessed a dramatic revolution since the discovery of targetable molecular alterations and the rapid development of small molecule inhibitors. ROS1 (c-ros oncogene) is one of the well-known alterations. ROS1 alterations are mainly genomic rearrangements present in up to 2% of cases of NSCLC.[1] They can be targeted by various tyrosine kinase inhibitors (TKIs) like crizotinib, the most widely used TKI for this alteration.[2] However, after an initial response, resistance develops because of various on-target and off-target mechanisms. This phenomenon has been widely elucidated in the literature. In this review article, we aim to describe the molecular biology, alterations, clinical and pathological profiles, diagnostic methods, treatment options, and outcomes of ROS1 rearranged NSCLC.
METHODS
In this narrative review, we identified the relevant articles by using various search engines and searching in scientific databases like PubMed, Scopus, Embase, and MyCancerGenome using the keywords “ROS1”, “NSCLC”, “ROS1 crizotinib”, “Oncogene NSCLC,” and “ROS1 resistance”. Since this was not intended to be a systematic review or a meta-analysis, we did not perform a formal statistical analysis and did not pre-specify any inclusion or exclusion criteria for the articles to be included in the review. We included 72 articles for the preparation of this review.
HISTORICAL PERSPECTIVE
ROS1 was first identified in 1986 during pre-clinical research involving the chicken sarcoma ribonucleic acid (RNA) UR2 tumor virus.[3] The first ROS1 rearrangement was detected in the U118MG glioblastoma cell line.[3,4] Subsequently, the oncogenic potential of these rearrangements has been widely elucidated in many pre-clinical studies. Finally, in 2007, the first case of ROS1 rearranged NSCLC was identified in a proteomic screen.[5]
MOLECULAR BIOLOGY
ROS1 or c-ROS (cellular ROS) is a receptor tyrosine kinase encoded by ROS1, which maps to chromosome 6q22.1.[6] The structure and details of signaling are discussed below.
STRUCTURE
ROS1 exists in two alternative isoforms, one with 43 exons and one with 44 exons.[6,7] The extracellular N terminal domain contains exons 1-34, with 9 fibronectin type III repeats and 3 beta-propeller domains.[8,9] The tyrosine kinase domain is intracellular and along with the C terminal, it is connected to the extracellular domain via a single transmembrane domain, homologous to that seen in other receptor tyrosine kinases like anaplastic lymphoma kinase (ALK).[6,10,11] ALK and ROS1 are considerably homologous in structure. The structure is depicted in Figure 1a.
Figure 1: ROS1 domain structure and normal signaling. (a) The ROS1 receptor comprises nine fibronectin type III motifs (blue rectangle), three beta propeller domains (yellow circle) with YWTD repeats, a single transmembrane domain (black line) and an intracellular kinase domain (purple rectangle). (b) The downstream signaling pathways activated by ROS1 include the RAS-RAF-MEK-ERK pathway, PI3K-AKT-mTOR pathway, and the JAK-STAT pathway. Autophosphorylation occurs in the tyrosine resides in the intracellular domain of ROS1 which results in recruitment of docking proteins namely GAB, GRB2, SHC, SOS, and SHP2, thus activating downstream pathways. (ROS1: cellular ROS, YWTD: tyrosine, tryptophan, threonine, aspartate: JAK: Janus kinase, RAS: rous sarcoma, RAF: rapidly accelerated fibrosarcoma, MEK: Mitogen-activated protein kinase kinase, ERK: Extracellular signal-regulated kinase, STAT3: signal transducer and activator of transcription 3, mTOR: mammalian target of rapamycin, PI3K: phosphatidylinositol 3-kinase, AKT: Ak strain transforming, P: phosphate, GAB: GRB2-associated binding protein, GRB2: growth factor receptor bound protein 2, SHP2: Src homology region 2 domain-containing phosphatase-2, SOS: son of sevenless, PDK1: pyruvate dehydrogenase kinase 1)
SIGNALING
The ligand for ROS1 is neural epidermal growth factor-like like 2 (NEFLL2), which is a testicular germ cell-secreted lumicrine factor.[12] This was discovered initially in the testes, and additional validation reports for other organs are awaited. It has been postulated that ROS1 is activated by the binding of specific tyrosine residues in the intracellular domain, which serve as docking sites for adaptor proteins like SHP2.[11] This stimulates the RAS/RAF/MEK/ERK, PI3K/AKT/mTOR, and JAK/STAT pathways, thus regulating cell survival, cell growth, and proliferation.[6,10,13,14] The signaling schema is depicted in Figure 1b.
ROS1 ALTERATIONS IN CANCER
Various types of alterations can occur in the ROS1 gene, which may cause oncogenesis. These are described in detail below.
ROS1 fusions/gene rearrangements
ROS1, analogous to ALK, is a promiscuous gene, with at least 55 different 5’ fusion partners described in the literature, across all malignancies.[15,16] The first partner reported was in glioblastoma, the so-called “FIG-ROS1” (Fused in glioblastoma).[4] The other partners are listed in Table 1.[7,17] The frequency or prevalence of each partner varies with the different cancer types and a schematic depiction is provided in Figure 2.[7,17–19] In NSCLC, the most common is CD74-ROS1, reported in 44% of cases of ROS1 rearranged NSCLC, followed by EZR in 16%, SDC4 in 14%, and SLC34A2 in 10% of cases. On the contrary, GOPC is the most common partner in glioblastoma cases, reported in approximately 77% of cases of ROS1-positive glioblastoma [Figure 3].[7,17,19,20]
Table 1: Various fusion partners reported for ROS1 along with their prevalence
Figure 2: Frequency of various fusion partners for ROS1 across different malignancies. (IMT: inflammatory myofibroblastic tumor, NSCLC: non-small-cell lung carcinoma)
Figure 3: Frequency of ROS1 rearrangement across various cancer types. (IMT: inflammatory myofibroblastic tumor, NSCLC: non-small-cell lung carcinoma, GBM: glioblastoma, ALCL: Anaplastic large cell lymphoma; GIT: gastrointestinal tract)
The fusions in ROS1 can occur through intrachromosomal or interchromosomal mechanisms. The majority of ROS1 fusions in NSCLC occur as a result of interchromosomal alterations, in contrast to glioblastoma, where most fusions occur as a result of microdeletions of 6q, and thus are intrachromosomal.[7,21] This distinction is important as it suggests that a distinct variation exists in both the dynamics as well as the chromatin architecture between different tumor types with respect to ROS1 rearrangement. Commonly, the extracellular domain is lost and there is an in-frame fusion between the N terminal of the partner gene and the intracellular tyrosine kinase domain of ROS1. The ROS1 fusion eventually leads to ligand-independent constitutive catalytic activity with unabated downstream signaling through RAS/RAF/MEK/ERK, PI3K/AKT/mTOR, or the JAK/STAT pathways.
ROS1 mutations and amplifications
Mutations in ROS1 have been reported throughout the gene, as noted in cBioportal,[18] however, they have not been deemed pathogenic by any functional assays. On the other hand, missense mutations in the tyrosine kinase domain have been reported to be important, as they confer resistance to ROS1-tyrosine kinase inhibitors (TKIs). Likewise, splice variants have been reported resulting in exon skipping,[15,22] analogous to the alteration that occurs in the MET gene, however, the clinical significance of these is yet to be established. Amplification and copy number gains in ROS1 have been reported in NSCLC as well as in other malignancies like breast cancer, and sarcomas[7,23]; their effect on pathogenesis and the treatment efficacy of ROS1-TKIs is unclear.
Concomitant alterations with ROS1
ROS1 rearranged NSCLC is a distinct molecular entity, and is, by and large mutually exclusive of the other known driver alterations like EGFR and ALK[24]; some exceptions have been reported as anecdotal cases.[25,26] In a study by Zhang et al.[27] on 235 patients with ROS1 rearranged NSCLC, next-generation sequencing (NGS) done in 54 (22.9%) patients revealed more than one genomic alteration in 38.9% (n = 21) cases, with the commonest alteration being TP53 mutations in 13% (n = 7) cases. Other co-mutations included KRAS and EGFR mutations and MET amplification. However, the number of patients harboring co-mutations was too small to draw definitive conclusions.
CLINICAL AND PATHOLOGIC FEATURES OF ROS1 REARRANGED NSCLC
Although rare, ROS1 rearranged NSCLC has been studied in both controlled trials as well as in real-world series. A meta-analysis of 9898[28] patients with NSCLC, concluded that ROS1 is associated with young age, female sex, never smoking status, and adenocarcinoma histology. A study by Joshi et al.[29] from India, reported a 4% prevalence (22 cases positive of the 535 tested) of ROS1 rearranged NSCLC. The median age of these 22 patients with ROS1 rearranged NSCLC was 53 years (range, 45-70), similar to that reported in the PROFILE 1001 (NCT00585195) registration trial.[30] The proportion of never-smokers was 75% (n = 40) in the ROS1 positive group. Similar findings were reported in another study from the Indian subcontinent by Mehta et al.[31] in 20 patients with ROS1 rearranged NSCLC. Other clinical aspects which were reported included a higher incidence of venous thromboembolism.[32] This occurs owing to the mucin present in both the extracellular and intracellular compartments which binds the P or L selectins, thus resulting in platelet activation and subsequently embolization.[33] Brain metastases at diagnosis have been reported in 35% (n = 12) of ROS1 rearranged NSCLC in a study by Patil et al.[34] Histologically, tumors with ROS1 rearrangement are noted to have adenocarcinoma morphology with predominantly acinar and lepidic patterns, with more than 90% of cases expressing TTF1. Imaging may show lymph node tropism characterized by a higher predilection for lymph node metastases, with fewer extrathoracic metastases when compared to ALK-positive cancers.[35]
Alterations in ROS1 have been reported across various tumor types in almost 20 different adult and pediatric malignancies. The reported frequencies are provided in Figure 2.[7,35–40]
DETECTION OF ROS1 ALTERATIONS
Fusions
Various methods have been described for the detection of ROS1 fusions. A detailed comparison of each is shown in Table 2 along with the advantages and disadvantages of each.[41–48]
Table 2: Detection methods for ROS1 fusions
Kinase domain mutations
Kinase domain mutations are usually missense mutations in the tyrosine kinase domain of ROS1. The canonical G2032R (G: glycine, R: Arginine) has been well documented.[51] This mutation can be detected using any specifically designed single-gene polymerase chain reaction (PCR). However, since resistance is an ever-evolving phenomenon with the ongoing discovery of new mutations and variants, NGS-based assays designed to cover the entire tyrosine kinase domain of the gene are the preferred modality.
TREATMENT OF ROS1 REARRANGED NSCLC
Conventional chemotherapy and response
Systematic studies have assessed the responsiveness to chemotherapy of ROS1-rearranged NSCLC. In a study by Xu et al.[52] first-line platinum-based chemotherapy was administered to 46 patients with ROS1-rearranged tumors (detected and confirmed by FISH), of which 35 (76.1%) patients also received pemetrexed; the median progression-free survival (PFS) was 8.8 months (95% CI, 6.8-10.3). Another study by Chen et al.[53] reported an objective response rate (ORR) of 47% (95% CI, 29.8-57.4) with a median PFS of 8.6 months (95% CI, 6.9-10.3) in patients with ROS1 rearranged NSCLC who were treated with first-line platinum-pemetrexed combination chemotherapy (n = 47). With pemetrexed monotherapy, Park et al.,[54] reported an ORR of 53.3% in 90 cases, and a PFS of 8 months (95% CI, 6.4-11.7). ROS1 rearranged cancers have low thymidylate synthase messenger RNA (mRNA) levels, which may explain the benefit noted from pemetrexed-based therapy,[55] when compared to that in other oncogene-addicted tumors.
ROS1 inhibition
Crizotinib
In the PROFILE 1001 (NCT00585195)[30] trial, crizotinib, a first-generation ALK-TKI, showed efficacy in 53 pretreated patients with ROS1 rearranged NSCLC; the reported ORR was 72% (95% CI, 58-83), the median PFS was 19 months (95% CI, 15.2-45.3), and the median overall survival (OS) was 51.4 months (95% CI, 29.3-not reached [NR]). In the AcSé study on 37 patients,[56] the ORR was 47.2% (95% CI, 30.4-64.5) with median PFS and OS of 5.5 months (95% CI, 4.2-9.1) and 17.2 months (95% CI, 6.8-32.8), respectively. Thus, the AcSé results were strikingly worse as compared to those of PROFILE 1001[30] owing to a higher proportion of patients with Eastern Cooperative Oncology Group (ECOG) performance status (PS) 2 in the AcSé study. The other prospective studies, including the European EUROCROSS[57] (NCT02183870) and METROS (NCT02499614)[32] reported results that were more similar to those of the PROFILE[30] study with ORRs of 70% (95% CI, 51-85) and 65% (95% CI, 44-82), respectively. However, despite the remarkably high reported responses, there was poor central nervous system (CNS) penetration, especially considering the fact that brain metastases have been reported in approximately 35% of cases of ROS1 rearranged NSCLC at diagnosis. Newer TKIs have been developed to tackle this problem. The commonly reported adverse events with crizotinib include vision disturbances (82%), diarrhea (44%), nausea and peripheral edema (40% each), deranged liver function tests (22%), fatigue (20%), and dysgeusia (18%), mainly grade 1 and 2.[30,32,57]
Ceritinib
This is a second-generation TKI which has been approved for patients with TKI-naive and crizotinib-resistant ALK-positive NSCLC. Ceritinib is also a selective inhibitor of ROS1. In the Pan-Korean study, the ORR was 62% (95% CI, 45-77) with a median PFS of 19.3 months.[58] The intracranial ORR was 25% (95% CI, 7-59). However, ceritinib is not active against several mutations like G2032R, D2033N, L1951R, and S1986F/Y that confer resistance to crizotinib. The adverse events reported include diarrhea (78%), nausea (59%), anorexia (56%), and vomiting (53%).[58]
Entrectinib
This is a multi-kinase inhibitor which the targets ALK, ROS1, and pan-tropomyosin receptor kinase (TRK).[59] Entrectinib can penetrate the blood-brain barrier. In vitro studies have shown that the anti-ROS1 activity of entrectinib is 40 times greater than that of crizotinib. Two landmark trials that investigated the efficacy and outcomes of entrectinib were STARTRK-1/ALKA-372-001 (NCT02097810)[60] and STARTRK-2 (NCT02568267).[61] In 53 evaluable patients, the reported ORR was 67% (95% CI, 59-74) with a median PFS of 16 months (95% CI, 11-21) and a one-year survival rate of 81%. Among the 17 patients with brain metastases at diagnosis, the intracranial response rate to entrectinib was 55% (95% CI, 32-68). Thus, due to favorable CNS responses, entrectinib gained FDA (United States Food and Drug Administration) approval in August 2019.[62] However, owing to activity against pan-TRK, it can lead to peculiar side effects like fatigue (46%), dizziness (16%), dysgeusia (42%), paresthesia (29%), nausea (28%), and myalgia (23%).[60,61]
Lorlatinib
Lorlatinib is a third-generation ALK inhibitor with anti-ROS1 activity, specifically developed for CNS penetration by decreasing the efflux mediated by P-glycoprotein 1. In a phase I/II trial (NCT101970865)[63] the anti-tumor activity in ROS1 rearranged NSCLC was demonstrated in 21 TKI treatment-naive and 40 crizotinib-pretreated patients. In the TKI-naive group, the ORR was 62% (95% CI, 38-82) with a median PFS of 21 months (95% CI, 4.2-31.9) and an intracranial ORR of 64% (95% CI, 31-89). The median intracranial PFS was not reached, thus endorsing the remarkable efficacy of lorlatinib against CNS metastases. In the crizotinib-pretreated patients, the ORR was 35% (95% CI, 21-52). Grade 3 and 4 adverse events included hypercholesterolemia (65%), hypertriglyceridemia (42%), peripheral edema (39%), peripheral neuropathies (35%), weight gain (16%), and mood disturbances (16%).[63] In the presence of resistance mutations in ROS1, p.K1991E and p.S1986F, lorlatinib has been proven to have superior efficacy after pretreatment with crizotinib.[63] However, patients with the canonical G2032R mutation do not show a response to lorlatinib.[7,63]
Repotrectinib
Also known as TPX-0005, this is a next-generation low molecular weight macrocyclic TKI which can target ROS1-TKI, and ALK and has the ability to cross the blood-brain barrier.[64] The FDA has granted a breakthrough therapy designation to repotrectinib for the treatment of patients with ROS1-positive metastatic NSCLC who have previously been treated with one ROS1 TKI and have not received prior platinum-based chemotherapy. Repotrectinib has shown activity against a variety of solvent-front substitutions, including p.G2032R and D2033N. Repotrectinib is being investigated in a dose-escalation phase I/II clinical trial TRIDENT-1 (NCT03093116)[65] in patients with advanced ALK-, ROS1-, or NTRK1–3-rearranged cancers. This trial included 33 patients. In the TKI-naive subgroup (n = 11), the ORR was 82% (95% CI, 44-97), and the intracranial ORR (n = 3 of those with brain metastases) was 100% (95% CI, 29-100).
Taletrectinib
This is a dual inhibitor of ROS1 and pan-TRK, with activity against L1951R, L2026M, S1986F, and G2032R mutations proved in pre-clinical studies.[66] A phase I study is ongoing (DS-6051b [NCT02279433])[66,67] and the preliminary ORR reported was 58% (95% CI, NR).
Drugs in development
Preliminary data from the phase I portion of the ARROS-1 phase I/II trial (NCT05118789)[68] presented at the 34th European Organisation for Research and Treatment of Cancer (EORTC)-National Cancer Institute (NCI)-American Association for Cancer Research (AACR) Symposium on Molecular Targets and Cancer Therapeutics, reported that NVL-520, a new drug in development, showed early signs of activity in the 35 patients enrolled in the study, with good CNS penetration as well as commendable activity against solvent-front mutations, with no dose-limiting toxicities (DLTs) or side effects that led to dose reductions or discontinuation of treatment.
Immunotherapy
In the IMMUNOTARGET study[69] seven patients with ROS1-positive NSCLC were included, and the reported ORR was 17%. In a Japanese retrospective study, 15 patients with ROS1-altered NSCLC were enrolled. High expression of programmed death ligand 1 (PD-L1), i.e., >50% of tumor cells stained by 22C3 was observed in 53% of cases, however, no responses to immunotherapy were noted.[70] Therefore, there is not much evidence to support the use and efficacy of immunotherapy in this group of patients.
Resistance mechanisms to ROS1 TKIs
Despite remarkable responses, resistance eventually develops to targeted ROS1 inhibition. Almost 60% of crizotinib-resistant mutations are on-target and occur in the kinase domain. Of these, G2032R is a canonical mutation, which is analogous to G1202R of ALK. This is a solvent-front alteration and the arginine results in steric hindrance with the crizotinib piperidine ring. This mutation is also capable of inducing an epithelial-mesenchymal transition. Second-generation TKIs like ceritinib, entrectinib, brigatinib, and even lorlatinib are not active against this mutation. Repotrectinib is one of the few drugs that has shown activity against this.[71,72] Other resistance mutations described include D2033, L1951, S1986F/Y, and the gatekeeper mutation L2026M. Rarer mutations include L1982F, E1990G, and F1994L. Activation of bypass pathways with mutations in EGFR, ERBB2, and PIK3CA has been described as well. Concomitant alterations in TP53 and other tumor suppressors like PTEN and RB1 have also been reported in anecdotal cases. The effect of these mutations requires further validation and larger studies.[7,30,71,72]
CONCLUSIONS AND FUTURE DIRECTIONS
ROS1 rearranged NSCLC is a distinct molecular entity, with available targeted therapy resulting in remarkable outcomes. Myriad detection methods are available which are all well-established and validated. It is necessary for clinical laboratories to include ROS1 in the initial testing battery. Additionally, the use of liquid biopsy can be investigated in future trials for monitoring of response to therapy and developing a BCR-ABL1-like treatment response parameter for the same.
Financial support and sponsorship
Nil.
Conflicts of interest
There are no conflicts of interest.
REFERENCES
1. D'Angelo A, Sobhani N, Chapman R, Bagby S, Bortoletti C, Traversini M, et al. Focus on
ROS1-positive non-small cell lung cancer (NSCLC):Crizotinib, resistance mechanisms and the newer generation of targeted therapies. Cancers (Basel) 2020;12:3293.
2. Shaw AT, Ou SH, Bang YJ, Camidge DR, Solomon BJ, Salgia R, et al. Crizotinib in
ROS1-rearranged non-small-cell lung cancer. N Engl J Med 2014;371:1963–71.
3. Birchmeier C, Sharma S, Wigler M Expression and rearrangement of the
ROS1 gene in human glioblastoma cells. Proc Natl Acad Sci U S A 1987;84:9270–4.
4. Charest A, Lane K, McMahon K, Park J, Preisinger E, Conroy H, et al. Fusion of FIG to the receptor tyrosine kinase ROS in a glioblastoma with an interstitial del (6)(q21q21). Genes Chromosomes Cancer 2003;37:58–71.
5. Odintsov I, Somwar R, Davare MA A blast from the past:
ROS1 on the brain. Oncotarget 2019;10:1664–6.
6. Davies KD, Doebele RC Molecular pathways:
ROS1 fusion proteins in cancer. Clin Cancer Res 2013;19:4040–5.
7. Drilon A, Jenkins C, Iyer S, Schoenfeld A, Keddy C, Davare MA
ROS1-dependent cancers-biology, diagnostics and therapeutics. Nat Rev Clin Oncol 2021;18:35–55.
8. Matsushime H, Shibuya M Tissue-specific expression of rat c-ros-1 gene and partial structural similarity of its predicted products with sev protein of Drosophila melanogaster. J Virol 1990;64:2117–25.
9. Springer TA An extracellular beta-propeller module predicted in lipoprotein and scavenger receptors, tyrosine kinases, epidermal growth factor precursor, and extracellular matrix components. J Mol Biol 1998;283:837–62.
10. Neckameyer WS, Shibuya M, Hsu MT, Wang LH Proto-oncogene c-ros codes for a molecule with structural features common to those of growth factor receptors and displays tissue specific and developmentally regulated expression. Mol Cell Biol 1986;6:1478–86.
11. Shibuya M, Matsushime H, Yamazaki H, Wang LH, Fukui Y, Ueyama Y, et al. Analysis of structure and activation of some receptor-type tyrosine kinase oncogenes. Princess Takamatsu Symp 1986;17:195–202.
12. Kiyozumi D, Noda T, Yamaguchi R, Tobita T, Matsumura T, Shimada K, et al. NELL2-mediated lumicrine signaling through OVCH2 is required for male fertility. Science 2020;368:1132–5.
13. Zong CS, Zeng L, Jiang Y, Sadowski HB, Wang LH Stat3 plays an important role in oncogenic Ros- and insulin-like growth factor I receptor-induced anchorage-independent growth. J Biol Chem 1998;273:28065–72.
14. Charest A, Wilker EW, McLaughlin ME, Lane K, Gowda R, Coven S, et al. ROS fusion tyrosine kinase activates a SH2 domain-containing phosphatase-2/phosphatidylinositol 3-kinase/mammalian target of rapamycin signaling axis to form glioblastoma in mice. Cancer Res 2006;66:7473–81.
15. Rose-John S, Heinrich PC Soluble receptors for cytokines and growth factors:Generation and biological function. Biochem J 1994;300:281–90.
16. Cerami E, Gao J, Dogrusoz U, Gross BE, Sumer SO, Aksoy BA, et al. The cBio cancer genomics portal:An open platform for exploring multidimensional cancer genomics data. Cancer Discov 2012;2:401–4.
17. Ou SI, Nagasaka M A catalog of 5'fusion partners in
ROS1-positive NSCLC circa 2020. JTO Clin Res Rep 2020;1:100048.
18. Gao J, Aksoy BA, Dogrusoz U, Dresdner G, Gross B, Sumer SO, et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal 2013;6:l1.
19. Rimkunas VM, Crosby KE, Li D, Hu Y, Kelly ME, Gu TL, et al. Analysis of receptor tyrosine kinase
ROS1-positive tumors in non-small cell lung cancer:Identification of a FIG-
ROS1 fusion Clin Cancer Res 2012;18:4449–57.
20. Seo JS, Ju YS, Lee WC, Shin JY, Lee JK, Bleazard T, et al. The transcriptional landscape and mutational profile of lung adenocarcinoma. Genome Res 2012;22:2109–19.
21. Akhoundova D, Hussung S, Sivakumar S, Töpfer A, Rechsteiner M, Kahraman A, et al.
ROS1 genomic rearrangements are rare actionable drivers in microsatellite stable colorectal cancer. Int J Cancer 2022;151:2161–71.
22. Kalla C, Gruber K, Rosenwald A, Kimmich M, Friedel G, Kohlhaufl MJ, et al.
ROS1 gene rearrangement and expression of splice isoforms in lung cancer, diagnosed by a novel quantitative RT-PCR assay. J Mod Hum Pathol 2016;1:25–34.
23. Gounder MM, Agaram NP, Trabucco SE, Robinson V, Ferraro RA, Millis SZ, et al. Clinical genomic profiling in the management of patients with soft tissue and bone sarcoma. Nat Commun 2022;13:3406.
24. Korpanty GJ, Graham DM, Vincent MD, Leighl NB Biomarkers that currently affect clinical practice in lung cancer:EGFR, ALK, MET, ROS-1, and KRAS. Front Oncol 2014;4:204.
25. Mao Y, Wu S ALK and
ROS1 concurrent with EGFR mutation in patients with lung adenocarcinoma. Onco Targets Ther 2017;10:3399–404.
26. Zhu YC, Xu CW, Ye XQ, Yin MX, Zhang JX, Du KQ, et al. Lung cancer with concurrent EGFR mutation and
ROS1 rearrangement:A case report and review of the literature. Onco Targets Ther 2016;9:4301–5.
27. Zhang Y, Zhang X, Zhang R, Xu Q, Yang H, Lizaso A, et al. Clinical and molecular factors that impact the efficacy of first-line crizotinib in
ROS1-rearranged non-small-cell lung cancer:A large multicenter retrospective study. BMC Med 2021;19:206.
28. Zhu Q, Zhan P, Zhang X, Lv T, Song Y Clinicopathologic characteristics of patients with
ROS1 fusion gene in non-small cell lung cancer:A meta-analysis. Transl Lung Cancer Res 2015;4:300–9.
29. Joshi A, Pande N, Noronha V, Patil V, Kumar R, Chougule A, et al.
ROS1 mutation non-small cell lung cancer-access to optimal treatment and outcomes. Ecancermedicalscience 2019;13:900.
30. Shaw AT, Riely GJ, Bang YJ, Kim DW, Camidge DR, Solomon BJ, 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.
31. Mehta A, Saifi M, Batra U, Suryavanshi M, Gupta K Incidence of
ROS1-rearranged non-small-cell lung carcinoma in India and efficacy of crizotinib in lung adenocarcinoma patients. Lung Cancer (Auckl) 2020;11:19–25.
32. Chiari R, Ricciuti B, Landi L, Morelli AM, Delmonte A, Spitaleri G, et al.
ROS1-rearranged non-small-cell lung cancer is associated with a high rate of venous thromboembolism:Analysis from a phase II, prospective, multicenter, two-arms trial (METROS). Clin Lung Cancer 2020;21:15–20.
33. Yi J, Chen H, Li J, Jiang X, Xu Y, Wang M, et al. The association between
ROS1 rearrangement and risk of thromboembolic events in patients with advanced non-small cell lung cancer:A multicenter study in China. Thromb J 2022;20:56.
34. Patil T, Smith DE, Bunn PA, Aisner DL, Le AT, Hancock M, 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.
35. Ji X, Xie H, Zhu R, Chen B, Jiang S, Luo J Different clinical features between patients with
ROS1-positive and ALK-positive advanced non-small cell lung cancer. J Int Med Res 2021;49:300060521993643.
36. Antonescu CR, Suurmeijer AJ, Zhang L, Sung YS, Jungbluth AA, Travis WD, et al. Molecular characterization of inflammatory myofibroblastic tumors with frequent ALK and
ROS1 gene fusions and rare novel RET rearrangement. Am J Surg Pathol 2015;39:957–67.
37. Lovly CM, Gupta A, Lipson D, Otto G, Brennan T, Chung CT, et al. Inflammatory myofibroblastic tumors harbor multiple potentially actionable kinase fusions. Cancer Discov 2014;4:889–95.
38. Wiesner T, He J, Yelensky R, Esteve-Puig R, Botton T, Yeh I, et al. Kinase fusions are frequent in Spitz tumours and spitzoid melanomas. Nat Commun 2014;5:3116.
39. Stucklin AS, Ryall S, Fukuoka K, Zapotocky M, Lassaletta A, Li C, et al. Alterations in ALK/
ROS1/NTRK/MET drive a group of infantile hemispheric gliomas. Nat Commun 2019;10:4343.
40. Parikh DA, Walia G, Freeman-Daily J, Hennink M, Tomalia T, Buonanno L, et al. Characteristics of patients with
ROS1+Cancers:Results from the first patient designed, global, pan-cancer
ROS1 data repository. JCO Oncol Pract 2020;16:e183–9.
41. AmoyDx
ROS1 kit approval in Taiwan, China. Amoy Diagnostics; 18 December 2018 Available from:
http://www.amoydiagnostics.com/newDetail/44 [Last accessed on 2022 Dec 07].
42. Davies KD, Le AT, Theodoro MF, Skokan MC, Aisner DL, Berge EM, et al. Identifying and targeting
ROS1 gene fusions in non-small cell lung cancer. Clin Cancer Res 2012;18:4570–9.
43. Dagogo-Jack I, Rooney M, Nagy RJ, Lin JJ, Chin E, Ferris LA, et al. Molecular analysis of plasma from patients with
ROS1-positive NSCLC. J Thorac Oncol 2019;14:816–24.
44. US Food and Drug Administration. Premarket approval of the Oncomine Dx Target Test; 22 June 2017 Available from:
https://www.accessdata.fda.gov/cdrh_docs/pdf16/P160045A.pdf [Last accessed on 2022 Dec 07].
45. Sholl LM, Sun H, Butaney M, Zhang C, Lee C, Jänne PA, et al.
ROS1 immunohistochemistry for detection of
ROS1-rearranged lung adenocarcinomas. Am J Surg Pathol 2013;37:1441–9.
46. Pavlakis N, Cooper C, John T, Kao S, Klebe S, Lee CK, et al. Australian consensus statement for best practice
ROS1 testing in advanced non-small cell lung cancer. Pathology 2019;51:673–80.
47. Hofman V, Rouquette I, Long-Mira E, Piton N, Chamorey E, Heeke S, et al. Multicenter evaluation of a novel
ROS1 immunohistochemistry assay (SP384) for detection of
ROS1 rearrangements in a large cohort of lung adenocarcinoma patients. J Thorac Oncol 2019;14:1204–12.
48. Shan L, Lian F, Guo L, Qiu T, Ling Y, Ying J, et al. Detection of
ROS1 gene rearrangement in lung adenocarcinoma:Comparison of IHC, FISH and real-time RT-PCR. PLoS One 2015;10:e0120422.
49. Lin JJ, Choudhury NJ, Yoda S, Zhu VW, Johnson TW, Sakhtemani R Spectrum of Mechanisms of resistance to crizotinib and lorlatinib in
ROS1 fusion-positive lung cancer. Clin Cancer Res 2021;27:2899–909.
50. Xu H, Zhang Q, Liang L, Li J, Liu Z, Li W, et al. Crizotinib vs platinum-based chemotherapy as first-line treatment for advanced non-small cell lung cancer with different
ROS1 fusion variants. Cancer Med 2020;9:3328–36.
51. Chen YF, Hsieh MS, Wu SG, Chang YL, Yu CJ, Yang JC, et al. Efficacy of pemetrexed-based chemotherapy in patients with
ROS1 fusion-positive lung adenocarcinoma compared with in patients harboring other driver mutations in east Asian populations. J Thorac Oncol 2016;11:1140–52.
52. Park S, Ahn BC, Lim SW, Sun JM, Kim HR, Hong MH, 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.
53. Song Z, Su H, Zhang Y Patients with
ROS1 rearrangement-positive non-small-cell lung cancer benefit from pemetrexed-based chemotherapy. Cancer Med 2016;5:2688–93.
54. Moro-Sibilot D, Cozic N, Pérol M, Mazières J, Otto J, Souquet PJ, et al. Crizotinib in c-MET- or
ROS1-positive NSCLC:Results of the AcSéphase II trial. Ann Oncol 2019;30:1985–91.
55. Michels S, Massutí B, Schildhaus HU, Franklin J, Sebastian M, Felip E, 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.
56. Lim SM, Kim HR, Lee JS, Lee KH, Lee YG, Min YJ, et al. Open-label, multicenter, phase ii study of ceritinib in patients with non-small-cell lung cancer harboring
ROS1 rearrangement. J Clin Oncol 2017;35:2613–8.
57. Menichincheri M, Ardini E, Magnaghi P, Avanzi N, Banfi P, Bossi R, et al. Discovery of entrectinib:A new 3-aminoindazole as a potent anaplastic lymphoma kinase (ALK), c-ros oncogene 1 kinase (
ROS1), and pan-tropomyosin receptor kinases (pan-TRKs) inhibitor. J Med Chem 2016;59:3392–408.
58. Drilon A, Siena S, Ou SI, Patel M, Ahn MJ, Lee J, et al. Safety and antitumor activity of the multitargeted pan-TRK,
ROS1, and ALK inhibitor entrectinib:Combined results from two phase I trials (ALKA-372-001 and STARTRK-1). Cancer Discov 2017;7:400–9.
59. Drilon A, Siena S, Dziadziuszko R, Barlesi F, Krebs MG, Shaw AT, et al. Entrectinib in
ROS1 fusion-positive non-small-cell lung cancer:Integrated analysis of three phase 1–2 trials. Lancet Oncol 2020;21:261–70.
61. Shaw AT, Solomon BJ, Chiari R, Riely GJ, Besse B, Soo RA, et al. Lorlatinib in advanced
ROS1-positive non-small-cell lung cancer:A multicentre, open-label, single-arm, phase 1-2 trial. Lancet Oncol 2019;20:1691–701.
62. Drilon A, Nagasubramanian R, Blake JF, Ku N, Tuch BB, Ebata K, et al. A next-generation TRK kinase inhibitor overcomes acquired resistance to prior TRK kinase inhibition in patients with TRK fusion-positive solid tumors. Cancer Discov 2017;7:963–72.
63. Drilon A, Ou SI, Cho BC, Kim DW, Lee J, Lin JJ, et al. Repotrectinib (TPX-0005) is a next-generation
ROS1/TRK/ALK inhibitor that potently inhibits
ROS1/TRK/ALK solvent-front mutations. Cancer Discov 2018;8:1227–36.
64. Fujiwara Y, Takeda M, Yamamoto N, Nakagawa K, Nosaki K, Toyozawa R, et al. Safety and pharmacokinetics of DS-6051b in Japanese patients with non-small cell lung cancer harboring
ROS1 fusions:A phase I study. Oncotarget 2018;9:23729–37.
65. Papadopoulos KP, Borazanci E, Shaw AT, Katayama R, Shimizu Y, Zhu VW, et al. U. S. Phase I First-in-human study of taletrectinib (DS-6051b/AB-106), a
ROS1/TRK inhibitor, in patients with advanced solid tumors. Clin Cancer Res 2020;26:4785–94.
66. Drilon A, Besse B, Camidge DR, Ou SHI, Gadgeel SN, Johnson ML, et al. Safety and preliminary clinical activity of NVL-520, a highly selective
ROS1 inhibitor, in patients with advanced
ROS1 fusion positive solid tumors. Eur J Can 2022;174:S6–7.
67. Mazieres J, Drilon A, Lusque A, Mhanna L, Cortot AB, Mezquita L, et al. Immune checkpoint inhibitors for patients with advanced lung cancer and oncogenic driver alterations:Results from the IMMUNOTARGET registry. Ann Oncol 2019;30:1321–8.
68. Dudnik E, Bshara E, Grubstein A, Fridel L, Shochat T, Roisman LC, et al. Rare targetable drivers (RTDs) in non-small cell lung cancer (NSCLC):Outcomes with immune check-point inhibitors (ICPi). Lung Cancer 2018;124:117–24.
69. Dagogo-Jack I, Shaw AT Crizotinib resistance:Implications for therapeutic strategies. Ann Oncol 2016;27 (Suppl 3) iii42–50.
70. Facchinetti F, Loriot Y, Kuo MS, Mahjoubi L, Lacroix L, Planchard D, et al. Crizotinib-resistant
ROS1 mutations reveal a predictive kinase inhibitor sensitivity model for
ROS1- and ALK-rearranged lung cancers. Clin Cancer Res 2016;22:5983–91.
71. Choughule A, D'Souza H
ROS1 rearrangement testing:Is immunohistochemistry changing the horizon?. Can Res Stat Treat 2019;2:66–8.
72. Chadha KG, Barodawala SM, Patkar AJ Reviewing
ROS1 immunohistochemistry vis-à-vis fluorescence
in situ hybridization in NSCLC. Cancer Res Stat Treat 2019;2:259–60.