Abstract
Background
The proto‐oncogene ROS1 encodes an intrinsic type I membrane protein of the tyrosine kinase/insulin receptor family. ROS1 facilitates the progression of various malignancies via self‐mutations or rearrangements. Studies on ROS1‐directed tyrosine kinase inhibitors have been conducted, and some have been approved by the FDA for clinical use. However, the adverse effects and mechanisms of resistance associated with ROS1 inhibitors remain unknown. In addition, next‐generation ROS1 inhibitors, which have the advantage of treating central nervous system metastases and alleviating endogenous drug resistance, are still in the clinical trial stage.
Method
In this study, we searched relevant articles reporting the mechanism and clinical application of ROS1 in recent years; systematically reviewed the biological mechanisms, diagnostic methods, and research progress on ROS1 inhibitors; and provided perspectives for the future of ROS1‐targeted therapy.
Results
ROS1 is most expressed in malignant tumours. Only a few ROS1 kinase inhibitors are currently approved for use in NSCLC, the efficacy of other TKIs for NSCLC and other malignancies has not been ascertained. There is no effective standard treatment for adverse events or resistance to ROS1‐targeted therapy. Next‐generation TKIs appear capable of overcoming resistance and delaying central nervous system metastasis, but with a greater incidence of adverse effects.
Conclusions
Further research on next‐generation TKIs regarding the localization of ROS1 and its fusion partners, binding sites for targeted drugs, and coadministration with other drugs is required. The correlation between TKIs and chemotherapy or immunotherapy in clinical practice requires further study.
Keywords: gene fusion, ROS1 gene, tumor‐targeted therapy, tyrosine kinase inhibitor
This study searched for relevant articles on the mechanism and clinical application of the ROS1 gene in recent years, systematically reviewed the biological mechanisms, diagnostic methods, and research progress on ROS1 inhibitors, and prospected the future of ROS1 targeted therapy to provide new ideas for clinical application and treatment. We believe that our study makes a significant contribution to the literature because our review revealed the following findings: the proto‐oncogene ROS1 is mostly expressed in malignant tumours, such as non‐small cell lung cancer (NSCLC). Only a few ROS1 kinase inhibitors are currently approved for use in NSCLC. At the same time, the efficacy of other tyrosine kinase inhibitors (TKIs) for NSCLC and other malignancies has not been ascertained as they are still being tested in clinical trials. There is no standard effective treatment for adverse events or resistance to ROS1‐targeted therapy. Finally, next‐generation TKIs appear capable of overcoming resistance and delaying central nervous system metastasis owing to their high affinity, but with a greater incidence of adverse effects.

1. INTRODUCTION
The proto‐oncogene ROS1 (c‐ROS) was first identified in glioblastoma cells in 1987 1 and is located at position 6q22 on the long arm of chromosome 6. 2 The ROS1 protein encoded by the ROS1 gene is a type I intrinsic transmembrane tyrosine kinase receptor belonging to the insulin receptor family. 3 The self‐mutation or rearrangement of ROS1 can activate tyrosine kinase, which promotes the proliferation of various malignant tumors. 4 ROS1 was subsequently detected in non‐small cell lung cancer (NSCLC) in 2007 as a chromosomal rearrangement 5 and contains fusion genes, such as SLC34A2‐ROS1, 6 SDC4‐ROS1, and CD74‐ROS1. 7 The fusion of ROS1 can lead to autophosphorylation, mediating tumor progression through the mitogen‐activated protein kinase (MAPK) pathway or RAS phosphorylation. 8 , 9
Clinical trials of ROS1‐directed tyrosine kinase inhibitors (TKIs) have been conducted, showing considerable results. Currently, several ROS1‐targeted drugs, such as crizotinib 10 and ceritinib, 11 have been approved by the FDA for use in a variety of malignancies, especially ROS1‐positive NSCLC. 3 Although progress has been made with the use of ROS1 inhibitors in clinical practice, presistant challenges such as drug resistance and adverse reactions (including central nervous system [CNS] metastases) remain unresolved. Remon et al. 12 suggested that next‐generation TKIs with high affinity and selectivity could be effective against drug resistance and CNS metastasis in ROS1‐targeted therapy. However, these drugs have not been utilized in clinical practice, as they are still undergoing clinical trials. Therefore, elucidation of the biological mechanism of the ROS1 gene, including the mechanism of ROS1 gene mutation, clarification of the concept of targeted therapy, and development of next‐generation inhibitors are crucial. In this study, ROS1‐related articles were retrieved from PubMed/MEDLINE and Web of Science (search keywords: “ROS1” and “malignant tumors”) to review the biological mechanisms, diagnostic modalities, and treatment effectiveness of ROS1 inhibitors to provide new insights into their clinical applications, therapeutic strategies, and directions for future development.
2. BIOLOGICAL MECHANISMS OF THE ROS1 GENE
2.1. Nonmalignant biological mechanisms of the ROS1 gene
ROS1 contains 47 exons, of which exons 1–34 encode the largest extracellular N‐terminal domain of the human receptor tyrosine family. 13 Springer et al. 14 found that this domain is composed of three YWTD domains and scattered type III fibronectin (FN3). The YWTD domain has a round and folded “propeller” shape, which can closely interact with surrounding structures, and change the structure of ROS1. In addition, ROS1 and the anaplastic lymphoma kinase (ALK) domains are very similar at the amino acid level, sharing 77% identity at the ATP‐binding site. 15 Considering the similarity between ROS1 and ALK, ALK inhibitors are also commonly used in ROS1‐mutant tumors. 16 The signaling pathways involving ROS1 are related to the differentiation and proliferation of normal cells. ROS1 can activate and bind to the SH2 domain through the intracellular autophosphorylation of specific tyrosines, acting with specific adaptor proteins to mediate the RAS/RAF/MEK/ERK, 17 PI3K/AKT/mTOR, 18 and JAK/STAT3 19 signaling pathways (Figure 1). ROS1 can activate both PTPN6 (SHP1) and PTPN11 (SHP2). Despite their structural homology, PTPN6 often inhibits cell activity through a negative regulatory pathway, whereas PTPN11 mainly acts as a positive signal transducer. 20 , 21
FIGURE 1.

Nonmalignant biological signaling pathways involving the ROS1 gene.
2.2. Malignant biological mechanisms of the ROS1 gene
ROS1 can facilitate the progression of various tumors through its overexpression, mutation amplification, or gene fusion. The overexpression of ROS1 was initially discovered in neurological tumors, such as primary gliomas 22 and meningiomas. 23 Cheng et al. 24 found that ROS1 gene expression in the cytoplasm was positively correlated with the development of oral squamous cell carcinoma (OSCC). It has been suggested that ROS1 can mediate the invasiveness of OSCC by enhancing mitochondrial bioactivity and increasing cellular ATP levels. 25 Boyard et al. 26 found that the rearrangement and overexpression of ROS1 can activate the JAK/STAT pathway in inflammatory hepatocellular adenoma and promote its progression. Bajrami et al. suggested that ROS1 is highly expressed in E‐cadherin (CDH1)‐deficient breast cancer, and its inhibitors (such as GSK1363089 and crizotinib) can destroy tumor cells via a synergistic lethal pathway. 27 A clinical trial (NCT04551495) investigating this is currently underway. 28
Hou et al. 29 studied the cBioPortal for Cancer Genomics and next‐generation sequencing (NGS) test results of 177 patients with lung adenocarcinoma and found the frequent occurrence of ROS1 gene rearrangements in younger patients (<40 years old; p = 0.035); however, the significance of these mutations was unclear. Consistently, Wang et al. 30 , 31 also suggested that ROS1 gene amplification or rearrangement mutations occurred more frequently in malignant tumors, such as gastrointestinal stromal tumors, gallbladder cancer, and soft tissue sarcoma. However, the significance and impact of these mutations require further investigation.
Compared to the overexpression and amplification of the ROS1 gene, researchers believe that ROS1 gene fusion is the primary driving force for tumorigenesis and disease progression. 4 , 13 , 32 , 33 Several ROS1 fusion genes have been identified in NSCLC, Spitz neoplasms, and glioblastomas (Figure 2). In NSCLC, CD74 is the most common fusion partner of the ROS1 gene, accounting for approximately 38% of cases, followed by EZR (13%), SDC4 (13%), and SLC34A2 (10%). 7 Gerami et al. 34 found that the PWWP2A‐ROS1 fusion had the highest frequency among 16 cases of Spitz neoplasm. In addition, GOPC‐ROS1 is the most common fusion in glioblastoma (75%) 35 ; Richardson et al. 36 suggested that this fusion may be caused by a microdeletion in chromosome 6q22.1. In conclusion, the high‐frequency ROS1 fusion partners differ among cancer types and are related to the intrinsic levels of fusion gene activation and fusion sites. For example, unlike in glioblastomas, ROS1 gene fusion in NSCLC happens via interchromosomal translocations. 37 , 38 Differences in fusion partners lead to changes in subcellular localization, which affect downstream signaling pathways. For example, compared to CD74‐ROS1, which acts on the endoplasmic reticulum, SDC4‐ROS1 and SLC34A2‐ROS1 have a stronger ability to activate the MAPK pathway. 39 Furthermore, there exists a mutually exclusive relationship between ROS1 fusions and other oncogenic mutations, a trend observed in NSCLC, 40 gliomas, 41 and Spitz neoplasms. 42 ROS1 is rarely co‐mutated with the ALK, EGFR, or KRAS genes. 40 Despite the similarity and high homology of the ROS1 and ALK domains, ROS1/ALK double fusions rarely coexist. 43
FIGURE 2.

Pie chart of ROS1 fusion partner frequencies in non‐small cell lung cancer (NSCLC), Spitz neoplasms, and glioblastoma (percentages shown).
3. DIAGNOSIS OF ROS1 GENE MUTATIONS
ROS1 gene mutations can occur in a variety of tumors, including NSCLC (1%–2%), 44 gliomas (6%–7%), 45 and cholangiocarcinomas (1.1%), 46 among which ROS1 fusion NSCLC is the most common. A meta‐analysis of 9898 patients with NSCLC indicated that ROS1 fusion occurred in younger female patients with no smoking history and was more prevalent in patients with advanced disease (stage III–IV) than those with early disease (p < 0.001). 47 Therefore, the prompt and effective diagnosis of ROS1 mutations is crucial. Several techniques have been reported for detecting ROS1 mutations that can be used for clinical screening or definitive diagnosis, as discussed in the succeeding sections.
3.1. Immunohistochemical staining
Immunohistochemical staining (IHC) is a useful screening tool for ROS1 fusions. ROS1 IHC can be performed using cellular specimens, paraffin‐embedded tissue specimens, and cell blocks for lung cancer detection. 48 The most commonly used antibody for detecting ROS1 is the D4D6 rabbit monoclonal antibody (Cell Signaling Technology, Danvers, MA, USA), which has a sensitivity and specificity of 89% and 98%, respectively. 49 A multicenter evaluation by Conde et al. 50 found the SP384 antibody (Ventana, Tuscon, Arizona, USA) to be more sensitive (93%) in detecting ROS1 gene rearrangements than the D4D6 antibody. However, Hofman's experimental results suggested that D4D6 is more accurate for ROS1 rearrangements. 51 Therefore, the use of these antibodies in IHC remains controversial. IHC results are mainly reported in the literature as 1+ (weak cytoplasmic staining), 2+ (moderate staining), or 3+ (strong staining) with positive microscopic manifestations of granular cytoplasmic staining. 52 IHC is an inexpensive and highly accurate screening method; however, it lacks clear scoring criteria and can be expressed at different levels in different tumor cells. In addition, normal tissues, such as type II alveolar and osteoclast‐type giant cells, can also weakly express ROS1, which interferes with diagnosis. 53
3.2. Reverse transcription‐polymerase chain reaction
Reverse transcription‐polymerase chain reaction (RT‐PCR) detects ROS1 rearrangements by identifying fusion mRNA and distinguishing fusion partners. 54 RT‐PCR can be used to extract RNA from formalin‐fixed paraffin‐embedded tissue samples and cellular specimens, 55 requires fewer tissue samples, and can be easily performed within a shorter detection period. However, RT‐PCR can only detect known fusion partners; it cannot identify unknown species of fusion genes. 56 Owing to the large number of unknown ROS1 fusion partners, 57 use of RT‐PCR has certain limitations in clinical applications.
3.3. Fluorescence in situ hybridization
Fluorescence in situ hybridization (FISH) is currently the only technique that can detect almost all ROS1 mutations. 58 It involves using two probes labeled with different fluorescent colors (usually red, green, or orange) to target the 5′ and 3′ ends of ROS1. 59 ROS1 fusion manifests in FISH as a loss of the probe at the 5′ end or a break in the signal, separating the two colors. 60 An advantage of FISH is its ability to detect unknown fusion partners of ROS1. Furthermore, the cycle time, specificity, and sensitivity of FISH are clinically satisfactory. 59 However, FISH is expensive, and the requirements for tissue specimens are stringent, such as tissue sections from infants under 6 months of age. 56 Furthermore, FISH must be detected in more than 50 cell nuclei for diagnostic significance. 61 In addition, some fusion partners of ROS1 (GOPC‐ROS1) cannot be detected by FISH owing to the limitations in the probe design 62 ; self‐designed probes can overcome this limitation. 63 , 64 However, FISH remains the gold standard for detecting ROS1 gene mutations.
3.4. Next‐generation sequencing
The range of NGS technologies, from analysis of mutant gene regions to whole‐genome sequencing, and their ability to detect multiple variant forms in parallel 65 save time and tissue samples compared to single‐target detection. NGS can detect single nucleotide mutations, gene insertions/deletions, and genomic rearrangements, and both DNA and RNA can be used as samples. 66 NGS is currently used to detect ROS1 gene rearrangements, particularly for ROS1 fusion genes with negative FISH results, such as GOPC‐ROS1. 67 Additionally, NGS can be performed using cell‐free DNA (cfDNA) or circulating tumor DNA (ctDNA), which are valid tools for early diagnosis. 68 However, limitations exist in employing NGS technology for large‐scale clinical applications. First, NGS is expensive when analyzing only a few genes. 69 Second, DNA‐only detection is less accurate and does not cover intronic breakpoints with large amounts of repetitive nucleotides, which whole‐genome RNA sequencing can overcome. 70 Benayed et al. 71 used an RNA‐based genome sequencer to test samples from 2522 patients with lung adenocarcinoma and found an improvement in the detection rate of ROS1 rearrangements compared to that of DNA samples. Finally, no standardized NGS process exists for the different types and stages of tumor progression, so additional clinical information is needed to define rare variants.
In summary, each of these four detection methods has advantages and disadvantages (Table 1), and there is no definitive conclusion regarding which assay should be used to detect ROS1 mutations. Owing to the tendency of ROS1 to be mutually exclusive with genes such as ALK, this study suggests that IHC can be used to screen tumor specimens negative for ALK expression before conducting FISH on specimens with positive IHC results. By contrast, NGS may be more appropriate for cases requiring early diagnosis and guidance on drug use.
TABLE 1.
Advantages and disadvantages of ROS1 mutation detection technology.
| Diagnostic techniques | Advantages | Limitations | Quotes |
|---|---|---|---|
| IHC |
High sensitivity and specificity Wide range of selected materials Low cost |
Absence of definitive diagnostic criteria Some normal tissues are also positively expressed, interfering with diagnosis |
[48–53] |
| RT‐PCR |
Low tissue requirements. Short testing cycles and simplicity in operation. |
Unable to detect unknown types of fusion partners | [54–57] |
| FISH |
Known or unknown fusion partners can be detected Short testing period High sensitivity and specificity |
High cost Stringent requirements for tissue specimens Undetectable partial fusion genes |
[56,58–64] |
| NGS |
Possibility of parallel testing, saving time and tissue samples Partial detection of FISH‐negative fusion types Detectable ctDNA for early diagnosis |
High cost Requires combined RNA sequencing to complement the range of detection No standardized process |
[65–71] |
Abbreviations: ctDNA, circulating tumor DNA; FISH, fluorescence in situ hybridisation; IHC, immunohistochemical staining; NGS, next‐generation sequencing; RT‐PCR, reverse transcription polymerase chain reaction.
4. ROS1 KINASE INHIBITORS
All ROS1 inhibitors are multi‐kinase inhibitors that can inhibit ALK, MET, and other kinases (such as EGFR, JAK2, and TrkA) in addition to ROS. 72 Early‐stage ROS1 TKIs, such as crizotinib, ceritinib, and entrectinib, can be used in patients with ROS1‐positive NSCLC who have not received TKI treatment (TKI‐naïve). 10 , 73 Considering resistance mutations are inevitable during treatment with early‐stage TKIs, next‐generation TKIs such as cabozantinib and taletrectinib have been tested in clinical trials. However, most of these are not approved for ROS1‐positive tumors. 74 , 75 , 76
ROS1 kinase inhibitors primarily affect the kinase domain of ROS1. 77 Conventional kinases can be classified into DFG‐in (active, type I) and DFG‐out (inactive, type II) kinases, depending on the domain conformation. DFG refers to the activation loop that regulates kinase activity, consisting of aspartic acid (D), phenylalanine (F), and glycine (G). 78 When the domain is in the DFG‐in state, aspartate rotates inward to expose the ATP‐binding site. Most TKIs (crizotinib, ceritinib, entrectinib, taletrectinib, and repotrectinib) compete for the ATP‐binding site, preventing further phosphorylation of the kinase by ATP, thus inhibiting kinase activity. 79 In the DFG‐out state, the aspartic acid in the domain is rotated outward and exposes a hydrophobic site that can be occupied by a few TKIs (such as cabozantinib and foretinib) to inhibit tumor activity. 79 As the DFG‐out conformation does not need to participate in the catalytic reaction, resulting in less restricted structure and providing greater selectivity in the design of inhibitors. However, the development of type II kinase inhibitors remains challenging because most kinases lack the DFG‐out domain conformation.
Both type I and type II ROS1 kinase inhibitors have inhibitory effects on ROS1 fusion‐positive tumors, 80 including crizotinib in CD74‐ROS1 and SDC4‐ROS1 fusion NSCLC, 81 ceritinib in TFG‐ROS1 fusion inflammatory myofibroblastoma, 82 and repotrectinib in G2032R‐ROS1 and D2033N‐ROS1 fusion tumors. 83 Furthermore, studies by Davare and Facchinetti have shown slight differences in the inhibitory effects of TKIs on different ROS1 fusion phenotypes. 84 , 85 However, a statistical review of clinical trial data by Drilon et al. 13 revealed that differences in the level of inhibition did not affect treatment efficacy.
5. TREATMENT OF ROS1‐MUTANT TUMORS
5.1. NSCLC
NSCLC accounts for approximately 80% of all lung cancers, and ROS1 fusion‐positive NSCLC accounts for 1%–2% of NSCLC cases. 86 Currently, ROS1 kinase inhibitors are the first‐line treatment for advanced ROS1 fusion‐positive NSCLC 87 (Table 2 and Figure 3).
TABLE 2.
Summary of clinical trials for TKIs in ROS1 rearrangement‐positive NSCLC.
| Drug | Clinical trial identifier/Study | Study design | ORR | Median DOR (months) | Median PFS (months) |
|---|---|---|---|---|---|
| Crizotinib | NCT00585195 |
Interventional, parallel‐cohort, Open‐label Phase I trial |
72% | 24.7 | 19.3 |
| NCT01945021 |
Interventional Single‐group assignment Open‐label Phase II Single‐arm trial |
71.7% | 19.7 | 15.9 | |
| EUROS1 |
Single‐group assignment Open‐label |
80% | – | 9.1 | |
| NCT02183870/EUCROSS |
Interventional, Single‐group assignment Open‐label Multicentre Phase II trial |
70% | – | 20.0 | |
| Ceritinib | NCT01964157 |
Interventional Open‐label Multicentre Phase II trial |
62% | 21.0 | 9.3 |
| Entrectinib | ALKA‐372‐001, STARTRK‐1, STARTRK‐2 |
Interventional Open‐label Multicentre Phase I/II trial |
67.1% 79.2% (CNS) |
15.7 12.9 (CNS) |
15.7 12.0 (CNS) |
| Lorlatinib | NCT01970865 |
Interventional Non‐randomized Open‐label Multicentre Phase I/II Single‐arm trial |
41% | 25.3 |
21.0 (TKI‐naïve) 8.5 (crizotinib‐treated) |
| Repotrectinib | NCT03093116 |
Interventional Single‐group assignment Open‐label Multicentre Phase I/II trial |
79% (TKI‐naïve) 38% (TKI‐treated) |
34.1 (TKI‐naïve) 14.8 (TKI‐treated) |
35.7 (TKI‐naïve) 9.0 (TKI‐treated) |
| Taletrectinib | NCT02279433, NCT02675491 |
Interventional Non‐randomized Single‐group assignment Open‐label Multicentre Phase I/IB trial |
66.7% (TKI‐naïve) 33.3% (TKI‐refractory) |
23.5 (TKI‐naïve) 14.0 (TKI‐refractory) |
29.1 (TKI‐naïve) 14.2 (TKI‐refractory) |
| TQ‐B3139 | NCT03099330 |
Interventional Single‐group assignment Phase I trial Open‐label |
66.7% | – |
20.2 (partial response) 27.0 (complete response) |
| Cabozantinib | Sun et al. | – | 25% | – | 7.4 |
| Brigatinib | Dudnik et al. | – | 29% (crizotinib‐resistant) | – |
21.6 (TKI‐naïve) 2.5 (crizotinib‐resistant) |
FIGURE 3.

Activity of tyrosine kinase inhibitors (TKIs) in clinical trials involving ROS1 rearrangement‐positive non‐small cell lung cancer (NSCLC), comparing the objective remission rates (ORRs) and progression‐free survival (PFS). Unclear ORRs or PFS are indicated with dashed lines.
In the phase I PROFILE 1001 clinical trial, 50 patients with advanced NSCLC treated with standard doses of crizotinib had an objective remission rate (ORR) of 72%, a median duration of remission (DOR) of 17.6 months, and a mean progression‐free survival (PFS) of 19.2 months. 88 Based on the significant antitumor activity observed in this trial, crizotinib was the first drug approved by the FDA for advanced ROS1 rearrangement‐positive NSCLC. 88 Recent reports from this trial showed an increased DOR of 24.7 months and PFS of 19.3 months after crizotinib treatment, with similar adverse effects, including visual impairment, edema, vomiting, and diarrhea. 89 A phase II single‐arm trial (NCT01945021) of 127 patients with ROS1 rearrangement‐positive NSCLC in East Asia by Wu et al. 57 reported an ORR of 71.7% and a mean PFS of 15.9 months for crizotinib. Another result from the EUROS1 cohort demonstrated an ORR of 80% and a mean PFS of 9.1 months in patients treated with crizotinib. 90 In the follow‐up EUCROSS cohort phase II study (NCT02183870), the ORR was 70% in 30 patients, and PFS improved to 20.0 months. 91 This suggests that crizotinib is highly effective in NSCLC patients of different ethnicities. However, most patients with advanced NSCLC treated with crizotinib eventually experience disease progression owing to ROS1 resistance mutations or CNS metastases.
Ceritinib is currently approved for patients with ALK rearrangement‐positive NSCLC resistant to crizotinib 92 , 93 ; however, it is also effective in patients with ROS1 rearrangement‐positive NSCLC. A phase II trial (NCT01964157) comprising 30 patients with advanced ROS1 rearrangement‐positive NSCLC not treated with crizotinib indicated that after a standard dose of ceritinib treatment (750 mg once daily), patients exhibited an ORR of 62%, a DOR of 21 months, and a mean PFS of 9.3 months; the common adverse events reported were gastrointestinal symptoms (78% diarrhea, 59% nausea, and 56% anorexia). 94 As a selective ROS1 inhibitor, ceritinib is 20 times more effective than crizotinib. 95 However, despite promising trial data, ceritinib may not work against crizotinib‐resistant mutations, including G2032R, D2033N, and L2086F. 80 , 96 In addition, ceritinib has a higher incidence of adverse effects than crizotinib. 94 , 97 , 98
Entrectinib is a multi‐kinase inhibitor that inhibits ROS1, ALK, and TRK. 99 According to an analysis of three phase I and II clinical trials (ALKA‐372‐001, STARTRK‐1, and STARTRK‐2), entrectinib had an ORR of 67.1% and a DOR and PFS of 15.7 months. For those who developed CNS metastases, the ORR was 79.2%, DOR was 12.9 months, and PFS was 12.0 months, with primary adverse effects including weight gain and neutropenia. 100 , 101 Unlike crizotinib, entrectinib has a weaker interaction with P‐glycoprotein (P‐gp) and can thus cross the blood–brain barrier, yielding ideal blood concentrations in the CNS and efficacy in patients with NSCLC who develop intracranial metastases. 102 , 103 Based on the positive clinical trial results and the specificity of the drug itself, entrectinib has become the second targeted agent approved by the FDA for treating patients with advanced ROS1 rearrangement‐positive NSCLC. 104 However, similar to ceritinib, entrectinib appears to be ineffective in crizotinib‐resistant cases, particularly in the absence of activity in G2032R‐ and L2026M‐mutant tumor cells. 73 , 105
Other next‐generation TKIs also play an indispensable role in the treatment of ROS1 rearrangement‐positive NSCLC, including lorlatinib, repotrectinib, taletrectinib, TQ‐B3139, cabozantinib, and brigatinib. The ORR of lorlatinib in a single‐arm phase I‐II trials (NCT01970865) was 62% for TKI‐naïve cases and 35% for crizotinib‐treated cases, PFS was 21.0 months for TKI‐naïve cases compared to 8.5 months for crizotinib‐treated cases; the common adverse effects include hypertriglyceridemia (19%) and hypercholesterolemia (14%). 106 A phase I–II trial of repotrectinib (NCT03093116) showed an ORR of 79%, PFS of 35.7 months, and DOR of 34.1 months in TKI‐naïve cases. Repotrectinib was also shown to be effective in patients who had previously received ROS1 TKI and had never received chemotherapy, with ORR of 38%, PFS of 9.0 months, and DOR of 14.8 months. The main adverse reactions were dizziness (58%), dysgeusia (50%), and paresthesia (30%). 107 Phase I studies of taletrectinib in the United States (NCT02279433) and Japan (NCT02675491) showed an ORR of 66.7% and a mean PFS of 29.1 months in patients with TKI‐primary ROS1‐mutated NSCLC compared with an ORR of 33.3% and a PFS of 14.2 months in crizotinib‐resistant cases; its adverse effects included abnormal liver function (72.7%) and gastrointestinal symptoms (50%). 108 The clinical activity of TQ‐B3139 as a novel TKI was validated. A phase I trial involving four patients with ROS1 fusion NSCLC (NCT03099330) showed that TQ‐B3139 had an ORR of 66.7%, along with a PFS of 20.2 months (partial response) or 27.0 months (complete response). 109 The results of trials with cabozantinib (ORR, 25%; PFS: 4.9–13.8 months) 110 and brigatinib (ORR, 29%) 111 have also demonstrated potent antitumor activity. Next‐generation TKIs also performed well in crizotinib‐resistant NSCLC, such as repotrectinib for the G2032R/D2033N fusion mutation, 83 lorlatinib and taletrectinib for the G2032R fusion mutation, 112 , 113 and cabozantinib and brigatinib for the CD74 fusion mutation. 114 , 115 Despite these promising results, only repotrectinib has been approved by the FDA as a first‐line agent for ROS1 rearrangement‐positive NSCLC. 103 Further trials are necessary to demonstrate the efficacy and safety of next‐generation TKIs for clinical use.
The role of immunotherapy in ROS1‐positive NSCLC is not well‐defined. Choudhury et al. found that ROS1 regulates the expression of programmed cell death 1 ligand 1 (PD‐L1) by activating the MEK‐ERK and ROS1‐SHP2 pathways. However, most ROS1‐positive NSCLC cells do not express PD‐L1 and have a low mutation load. 116 Immunotherapy combined with chemotherapy elicited a higher ORR in patients with ROS1‐positive NSCLC compared with immunotherapy alone. 117 , 118 In addition, a phase II clinical trial of Atezolizumab (NCT04042558) showed that Atezolizumab with or without bevacizumab and the “platinum‐Pemetrexed” chemotherapy regimen are effective in NSCLC patients with metastatic EGFR mutation or ALK/ROS1 rearrangement after TKI failure. 119 This suggests that immunotherapy combined with chemotherapy is a potential treatment strategy for patients with ROS1‐positive NSCLC, especially those for whom TKI therapy has failed.
5.2. Non‐NSCLC tumors
ROS1 kinase inhibitors are also effective in non‐NSCLC tumors; however, owing to the low incidence of ROS1 rearrangements, no relevant TKIs are currently approved for treating non‐NSCLC tumors. This has also been mentioned in the literature as a case report (Table 3). For example, crizotinib has shown good or partial remission in YWHAE1‐ROS1 fusion inflammatory myofibroblastoma (IMT), 120 GOPC‐ROS1 fusion Spitz naevi, high‐grade serous ovarian cancer, 67 , 121 RDX‐ROS1 fusion intrahepatic cholangiocarcinoma, 122 and TJP1‐ROS1 fusion malignant peripheral nerve sheath tumors(MPNST). 123 Cases on the use of entrectinib for ER+/HER2‐breast cancer, ARCN1‐ROS1 and ZCCHC8‐ROS1 fusion pediatric glioma, GOPC‐ROS1 fusion limbal melanoma, and SLC4A4‐ROS1 fusion metastatic pancreatic cancer have also been reported. 28 , 124 , 125 , 126 , 127 Among the next‐generation TKIs, lorlatinib has been suggested to be potentially effective in ROS1 p.L1950F point‐mutated pancreatic cancer and TFG‐ROS1 fusion IMT of the chest wall, 128 , 129 with no relevant case reports for other TKIs.
TABLE 3.
Overview of ROS1 kinase inhibitor therapy in non‐NSCLC tumors.
| Drug | Study | Tumor histology | Fusion/Mutation | Response |
|---|---|---|---|---|
| Crizotinib | Comandini et al. | IMT | YWHAE1‐ROS1 | PR |
| Robertson et al. | Spitz nevi | GOPC‐ROS1 | PR | |
| Dong et al. | High‐grade serous ovarian cancer | GOPC‐ROS1 | PR | |
| Jakubowski et al. | Intrahepatic cholangiocarcinoma | RDX‐ROS1 | PR | |
| Li et al. | MPNST | TJP1‐ROS1 | SD | |
| Entrectinib | Agostinetto et al. | ER+/HER2‐breast cancer (invasive lobular carcinoma) | E‐cadherin defect | PR |
| Mayr et al. | Pediatric high‐grade glioma | ARCN1‐ROS1 | PR | |
| Papusha et al. | Infant hemispheric glioma | ZCCHC8‐ROS1 | PR | |
| Couts et al. | Acral lentiginous melanoma | GOPC‐ROS1 | PR | |
| Pishvaian et al. | Metastatic pancreatic cancer | SLC4A4‐ROS1 | SD | |
| Lorlatinib | Velthaus et al. | Pancreatic cancer | ROS1 p.L1950F mutation | SD |
| Carcamo et al. | IMT of the chest wall | TFG‐ROS1 | PR |
Abbreviations: IMT, inflammatory myofibroblastoma; MPNST, malignant peripheral nerve sheath tumor; PR, partial response; SD, stable disease.
6. CHALLENGES OF ROS1‐TARGETED THERAPY
6.1. Adverse effects related to ROS1 inhibitors
Adverse events (AEs) are a major challenge in ROS1‐targeted therapy. The use of most ROS1 kinase inhibitors in clinical practice has been limited because of the AEs associated with their mechanism of action (Table 4), such as edema, visual disorders, gastrointestinal symptoms, and, in severe cases, liver function abnormalities, neutropenia, and even life‐threatening effects. 130 , 131 Some AEs are related to the unique systems in which TKIs act. For example, entrectinib inhibits TRK and acts on the nervous system, resulting in dizziness, weight gain, and cognitive impairment during targeted therapy. 132 Moreover, crizotinib can cause peripheral edema in patients because of MET kinase inhibition. 133 , 134 However, other adverse reactions still lack clear mechanisms to explain their occurrences, such as abnormal liver function, 135 manifestations of ocular toxicity associated with crizotinib treatment, 136 hyperlipidemia, and peripheral neuropathy associated with lorlatinib treatment. 131 There is currently no standardized clinical treatment process to mitigate the damage caused by AEs. Bauer et al. 131 suggested that adjusting drug doses or symptomatic supportive treatment could manage mild to moderate AEs (grade <3). Although next‐generation TKIs are considered to have potential value in reducing the incidence of AEs owing to their high affinity, 13 recent studies have demonstrated that next‐generation TKIs do not have an absolute advantage in treating AEs. 137 , 138
TABLE 4.
Summary of AEs associated with clinical trials of ROS1 inhibitors.
| Drug | Study | Overall AEs in ≥20% of patients | Treatment‐related AEs (≥ grade 3) |
|---|---|---|---|
| Crizotinib | PROFILE 1001 |
Vision disorder (87%) Nausea (51%) Oedema (47%) Diarrhea (45%) Vomiting (38%) Elevated transaminases (36%) Constipation (34%) Bradycardia (21%) Fatigue (21%) |
Hypophosphatemia (15%) Neutropenia (9%) Vomiting (4%) Elevated transaminases (4%) |
| NCT01945021 |
Elevated transaminases (55.1%) Vision disorder (48%) Nausea (40.9%) Diarrhea (38.6%) Vomiting (32.3%) Constipation (29.9%) Neutropenia (29.1%) Leukopenia (22.8%) Oedema (22.8%) |
Neutropenia (10.2%) Elevated transaminases (5.5%) Leukopenia (2.4%) Nausea (1.6%) |
|
| EUCROSS |
Vision disorder (65%) Diarrhea (56%) Oedema (50%) Bradycardia (47%) Nausea (41%) Increased ALT (35%) Vomiting (32%) Leukopenia/neutropenia (32%) Increased AST (26%) Increased blood creatinine (21%) |
Leukopenia/neutropenia (9%) Nausea (3%) Vomiting (3%) ALT increased (3%) Dysgeusia (3%) Pulmonary embolism (3%) |
|
| Ceritinib | NCT01964157 |
Diarrhea (78%) Nausea (62%) Anorexia (59%) Vomiting (53%) Cough (47%) Pain (41%) Fatigue (38%) Dyspnoea (25%) |
Fatigue (16%) Pneumonia (12%) Dry mouth (3%) Pleural effusion (3%) |
| Entrectinib | NCT02097810, NCT02568267 |
Dysgeusia (43.4%) Dizziness (34.8%) Constipation (31.4%) Fatigue (30%) Weight gain (28.6%) Diarrhea (26.7%) |
Weight gain (8.1%) Increased ALT (3.3%) Diarrhea (2.9%) Increased AST (2.4%) Decreased neutrophil count (2.4%) Neutropenia (1.9%) Rash (1.4%) |
| Lorlatinib | NCT01970865 |
Hypercholesterolaemia (72%) Hypertriglyceridemia (39%) Peripheral oedema (39%) Peripheral neuropathy (39%) Cognitive effects (22%) |
Hypercholesterolaemia (13%) Hypertriglyceridemia (6%) Weight gain (6%) Increased lipase (4%) Cognitive effects (2%) Increased AST (2%) |
| Repotrectinib | NCT03093116 |
Dizziness (58%) Dysgeusia (50%) Paresthesia (30%) Constipation (26%) Anemia (26%) Ataxia (20%) |
Anemia (4%) Increased blood creatine kinase leve (4%) Dizziness (3%) Weight increase (2%) |
| Taletrectinib | NCT02279433, NCT02675491 |
Increased ALT (72.7%) Increased AST (72.7%) Nausea (59.1%) Diarrhea (54.5%) Vomiting (36.4%) Increased blood creatinine (31.8%) |
Increased ALT (22.7%) Increased AST (13.6%) Diarrhea (4.5%) |
6.2. Resistance to ROS1 inhibitors
The prolonged use of ROS1 inhibitors often leads to the development of drug resistance in tumor cells during disease progression. The mechanisms leading to the development of drug resistance in tumors mainly include mutations in the kinase domain and activation of collateral signaling. 139 , 140 Point mutations in the structural domain of the ROS1 kinase can transform the drug target and reduce its inhibitory effect, leading to resistance to targeted therapies. 141 G2032R was the first identified and most common drug resistance mutation in ROS1. 142 The mutation results in the substitution of glycine with an arginine that still enables ATP binding but conflicts with the piperidine ring structure of crizotinib, causing resistance to ROS1 kinase inhibition, mediating epithelial‐mesenchymal transition, and upregulating Twist 1, contributing to tumor progression. 141 , 143 , 144 Another mutation, D2033N, causes drug resistance by affecting the solvent front region of the ATP‐binding site. 145 In addition, other drug resistance mutations, such as L2155S, affect protein function to enhance drug resistance, 143 and S1986F/Y (parallel mutation) and L2026M, promote kinase activity by blocking the critical binding site. 85 , 146 This mechanism of action accounts for 50%–60% of ROS1‐resistant tumors.
The activation of bypass signaling pathways (e.g., the EGFR pathway) has been shown to confer drug resistance by reducing tumor dependence on ROS1 activity and increasing its dependence on the self. 147 , 148 Including previously reported mutations in the KIT and MAPK pathways, autophosphorylation or alterations in the MEK/ERK pathway can render tumor cells resistant to crizotinib 8 , 149 (Figure 4).
FIGURE 4.

Molecular mechanisms of resistance to ROS1 tyrosine kinase inhibitors (TKIs), depicting the two main drug resistance patterns.
In addition, resistance to ROS1 inhibitors can lead to the phenotypic transformation of tumor cells. 150 Lin et al. reported a case of a patient with NSCLC whose pathological classification transformed into adenocarcinoma after ROS1 inhibitor resistance; this phenotypic change may be related to retinoblastoma‐1 (RB1) and TP53 inactivation. 151
The current mainstream response in response to kinase inhibitor resistance is to replace next‐generation TKIs or combine them with other kinase inhibitors. Yun et al. 76 found that repotrectinib has a targeted therapeutic effect against the G2032R mutation with a similar response observed with foretinib. 84 Lorlatinib has therapeutic effects against other mutations, such as L2026M and S1986F/Y. 152 In particular, combinations of kinase inhibitors appeared to be more effective against resistance due to the activation of bypass signaling pathways. Dziadziuszko et al. 149 discovered that combining crizotinib with ponatinib could overcome resistance caused by mutations in the KIT pathway in vitro. Vaishnavi et al. 140 detected the expression of the EGFR pathway in SLC34A2‐ROS1 fusion HCC78 cells, and the coadministration of crizotinib with the EGFR inhibitor gefitinib reduced drug resistance in HCC78 cells. 153 Aside from point mutations in the structural domain of ROS1 and the activation of bypass signaling pathways, some unknown drug resistance mechanisms exist, such as lorlatinib resistance in patients with ROS1 fusion‐positive NSCLC. 80 Therefore, when developing next‐generation TKIs, future studies should focus on epigenetics, RNA, and proteins to elucidate the mechanisms of ROS1 resistance.
6.3. Central nervous system metastases
CNS metastases are common in ROS1‐mutant malignancies. CNS metastases occur in approximately 36% of patients with stage IV ROS1‐positive NSCLC. 154 Li et al. 155 found a higher incidence of CNS metastasis in patients with NSCLC having CD74‐ROS1 fusion compared to non‐CD74‐ROS1 fusion cases (p = 0.020), suggesting that the type of fusion partner is associated with CNS metastasis. Jun et al. 156 discovered that CD74‐ROS1 confers a higher metastatic potential to tumor cells by activating the phosphorylation of the plasma membrane protein E‐Syt1. Due to its poor ability to cross the blood–brain barrier, crizotinib cannot be used as the first drug of choice for CNS metastasis. 157 However, next‐generation TKIs have exhibited great potential in cases of CNS metastases, including ceritinib, which showed an ORR of 25% in patients with CNS metastases in a phase II study, 94 and lorlatinib, which showed an ORR of 50%. 106 Among these, entrectinib is the most effective, with an ORR of 55% and a mean PFS of 13.6 months. 100 In contrast to TKI replacement, CNS metastases are sensitive to radiotherapy. Hence, they can be an option for controlling the progression of CNS metastases when patients cannot tolerate targeted therapy or have few lesions. 73
7. PROSPECTS AND CONCLUSIONS
As a proto‐oncogene, ROS1 is mostly expressed in malignant tumors such as NSCLC. Chromosomal rearrangements resulting in ROS1 fusions drive tumor progression, and an appropriate diagnosis should be made on a clinical basis. Only a few ROS1 kinase inhibitors are currently approved for use in NSCLC; however, the efficacy of other TKIs for NSCLC and other malignancies has not been ascertained, as they are still being tested in clinical trials. Next‐generation TKIs appear capable of overcoming resistance and delaying CNS metastasis owing to their high affinity; however, they are associated with a greater incidence of adverse effects. Further research on next‐generation TKIs regarding the localization of ROS1 and its fusion partners, binding sites for targeted drugs (type I vs. type II), and coadministration with other drugs is required. Moreover, the correlation between TKIs and chemotherapy or immunotherapy in clinical practice requires further study.
AUTHOR CONTRIBUTIONS
Shizhe Li: Writing – original draft (equal). He Zhang: Methodology (equal); software (equal). Ting Chen: Data curation (equal); supervision (equal). Xiaowen Zhang: Writing – review and editing (equal). Guanning Shang: Writing – review and editing (equal).
FUNDING INFORMATION
Shenyang Science and Technology Project (21–173–9‐24). China Postdoctoral Science Foundation (2021 M693912). 345 Talent Project of Shengjing Hospital (M0944). 345 Talent Project of Shengjing Hospital (M0744).
CONFLICT OF INTEREST STATEMENT
The authors declare that they have no conflict of interests.
Li S, Zhang H, Chen T, Zhang X, Shang G. Current treatment and novel insights regarding ROS1‐targeted therapy in malignant tumors. Cancer Med. 2024;13:e7201. doi: 10.1002/cam4.7201
Shizhe Li and He Zhang contributed equally to this work and share first authorship.
Xiaowen Zhang and Guanning Shang contributed equally to this work and share last authorship.
Contributor Information
Xiaowen Zhang, Email: zhangxiaowen@cmu.edu.cn.
Guanning Shang, Email: cmushanggn@126.com.
DATA AVAILABILITY STATEMENT
The materials that support the conclusion of this review have been included within the article.
REFERENCES
- 1. Birchmeier C, Sharma S, Wigler M. Expression and rearrangement of the ROS1 gene in human glioblastoma cells. Proc Natl Acad Sci USA. 1987;84(24):9270‐9274. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Hubbard‐Smith K, Patsalis P, Pardinas JR, Jha KK, Henderson AS, Ozer HL. Altered chromosome 6 in immortal human fibroblasts. Mol Cell Biol. 1992;12(5):2273‐2281. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Azelby CM, Sakamoto MR, Bowles DW. ROS1 targeted therapies: current status. Curr Oncol Rep. 2021;23(8):94. [DOI] [PubMed] [Google Scholar]
- 4. Uguen A, De Braekeleer M. ROS1 fusions in cancer: a review. Future Oncol. 2016;12(16):1911‐1928. [DOI] [PubMed] [Google Scholar]
- 5. Rikova K, Guo A, Zeng Q, et al. Global survey of phosphotyrosine signaling identifies oncogenic kinases in lung cancer. Cell. 2007;131(6):1190‐1203. [DOI] [PubMed] [Google Scholar]
- 6. Wu K, Liao X, Gong Y, et al. Circular RNA F‐circSR derived from SLC34A2‐ROS1 fusion gene promotes cell migration in non‐small cell lung cancer. Mol Cancer. 2019;18(1):98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Cui M, Han Y, Li P, et al. Molecular and clinicopathological characteristics of ROS1‐rearranged non‐small‐cell lung cancers identified by next‐generation sequencing. Mol Oncol. 2020;14(11):2787‐2795. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Sato H, Schoenfeld AJ, Siau E, et al. MAPK pathway alterations correlate with poor survival and drive resistance to therapy in patients with lung cancers driven by ROS1 fusions. Clin Cancer Res. 2020;26(12):2932‐2945. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Skoulidis F, Heymach JV. Co‐occurring genomic alterations in non‐small‐cell lung cancer biology and therapy. Nat Rev Cancer. 2019;19(9):495‐509. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. D'Angelo A, Sobhani N, Chapman R, 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(11):3293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Raedler LA. Zykadia (Ceritinib) approved for patients with Crizotinib‐resistant ALK ‐positive non‐small‐cell lung cancer. Am Health Drug Benefits. 2015;8(Spec Feature):163‐166. [PMC free article] [PubMed] [Google Scholar]
- 12. Remon J, Pignataro D, Novello S, Passiglia F. Current treatment and future challenges in ROS1‐ and ALK‐rearranged advanced non‐small cell lung cancer. Cancer Treat Rev. 2021;95:102178. [DOI] [PubMed] [Google Scholar]
- 13. 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(1):35‐55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. 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(4):837‐862. [DOI] [PubMed] [Google Scholar]
- 15. Shaw AT, Hsu PP, Awad MM, Engelman JA. Tyrosine kinase gene rearrangements in epithelial malignancies. Nat Rev Cancer. 2013;13(11):772‐787. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Tan AC, Tan DSW. Targeted therapies for lung cancer patients with oncogenic driver molecular alterations. J Clin Oncol. 2022;40(6):611‐625. [DOI] [PubMed] [Google Scholar]
- 17. Wang W, He B. MiR‐760 inhibits the progression of non‐small cell lung cancer through blocking ROS1/Ras/Raf/MEK/ERK pathway. Biosci Rep. 2020;40:BSR20182483. [DOI] [PubMed] [Google Scholar]
- 18. Zong CS, Chan JL, Yang SK, et al. Mutations of Ros differentially effecting signal transduction pathways leading to cell growth versus transformation. J Biol Chem. 1997;272(3):1500‐1506. [DOI] [PubMed] [Google Scholar]
- 19. Acquaviva J, Wong R, Charest A. The multifaceted roles of the receptor tyrosine kinase ROS in development and cancer. Biochim Biophys Acta. 2009;1795(1):37‐52. [DOI] [PubMed] [Google Scholar]
- 20. Keilhack H, Muller M, Bohmer SA, et al. Negative regulation of Ros receptor tyrosine kinase signaling. An epithelial function of the SH2 domain protein tyrosine phosphatase SHP‐1. J Cell Biol. 2001;152(2):325‐334. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Charest A, Wilker EW, McLaughlin ME, 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(15):7473‐7481. [DOI] [PubMed] [Google Scholar]
- 22. Maxwell M, Galanopoulos T, Nevillegolden J, et al. Overexpression of the ros1 gene in primary human gliomas may contribute to malignant progression. Int J Oncol. 1996;8(4):713‐718. [DOI] [PubMed] [Google Scholar]
- 23. Zhao JF, Sharma S. Expression of the ROS1 oncogene for tyrosine receptor kinase in adult human meningiomas. Cancer Genet Cytogenet. 1995;83(2):148‐154. [DOI] [PubMed] [Google Scholar]
- 24. Cheng Y, Sun Y, Wang LZ, et al. Cytoplasmic c‐ros oncogene 1 receptor tyrosine kinase expression may be associated with the development of human oral squamous cell carcinoma. Oncol Lett. 2015;10(2):934‐940. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Chang YJ, Chen KW, Chen L. Mitochondrial ROS1 increases mitochondrial fission and respiration in Oral squamous cancer carcinoma. Cancers (Basel). 2020;12(10):2845. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Bayard Q, Caruso S, Couchy G, et al. Recurrent chromosomal rearrangements of ROS1, FRK and IL6 activating JAK/STAT pathway in inflammatory hepatocellular adenomas. Gut. 2020;69(9):1667‐1676. [DOI] [PubMed] [Google Scholar]
- 27. Bajrami I, Marlow R, van de Ven M, et al. E‐cadherin/ROS1 inhibitor synthetic lethality in breast cancer. Cancer Discov. 2018;8(4):498‐515. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Agostinetto E, Nader‐Marta G, Paesmans M, et al. ROSALINE: a phase II, neoadjuvant study targeting ROS1 in combination with endocrine therapy in invasive lobular carcinoma of the breast. Future Oncol. 2022;18(22):2383‐2392. [DOI] [PubMed] [Google Scholar]
- 29. Hou H, Zhang C, Qi X, et al. Distinctive targetable genotypes of younger patients with lung adenocarcinoma: a cBioPortal for cancer genomics data base analysis. Cancer Biol Ther. 2020;21(1):26‐33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Wang S, Chen R, Tang Y, et al. Comprehensive genomic profiling of rare tumors: routes to targeted therapies. Front Oncol. 2020;10:536. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Chen HW, Chen TW. Genomic‐guided precision therapy for soft tissue sarcoma. ESMO Open. 2020;5(2):e000626. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Takeuchi K, Soda M, Togashi Y, et al. RET, ROS1 and ALK fusions in lung cancer. Nat Med. 2012;18(3):378‐381. [DOI] [PubMed] [Google Scholar]
- 33. Kohno T, Nakaoku T, Tsuta K, et al. Beyond ALK‐RET, ROS1 and other oncogene fusions in lung cancer. Transl Lung Cancer Res. 2015;4(2):156‐164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Gerami P, Kim D, Compres EV, et al. Clinical, morphologic, and genomic findings in ROS1 fusion Spitz neoplasms. Mod Pathol. 2021;34(2):348‐357. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Sievers P, Stichel D, Sill M, et al. GOPC:ROS1 and other ROS1 fusions represent a rare but recurrent drug target in a variety of glioma types. Acta Neuropathol. 2021;142(6):1065‐1069. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Richardson TE, Tang K, Vasudevaraja V, et al. GOPC‐ROS1 fusion due to microdeletion at 6q22 is an oncogenic driver in a subset of pediatric gliomas and glioneuronal tumors. J Neuropathol Exp Neurol. 2019;78(12):1089‐1099. [DOI] [PubMed] [Google Scholar]
- 37. Bergethon K, Shaw AT, Ou SH, et al. ROS1 rearrangements define a unique molecular class of lung cancers. J Clin Oncol. 2012;30(8):863‐870. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Ou SI, Nagasaka M. A catalog of 5' fusion partners in ROS1‐positive NSCLC circa 2020. JTO Clin Res Rep. 2020;1(3):100048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Neel DS, Allegakoen DV, Olivas V, et al. Differential subcellular localization regulates oncogenic signaling by ROS1 kinase fusion proteins. Cancer Res. 2019;79(3):546‐556. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Lin JJ, Ritterhouse LL, Ali SM, et al. ROS1 fusions rarely overlap with other oncogenic drivers in non‐small cell lung cancer. J Thorac Oncol. 2017;12(5):872‐877. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Clarke M, Mackay A, Ismer B, et al. Infant high‐grade gliomas comprise multiple subgroups characterized by novel targetable gene fusions and favorable outcomes. Cancer Discov. 2020;10(7):942‐963. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Donati M, Kastnerova L, Martinek P, et al. Spitz tumors with ROS1 fusions: a clinicopathological study of 6 cases, including FISH for chromosomal copy number alterations and mutation analysis using next‐generation sequencing. Am J Dermatopathol. 2020;42(2):92‐102. [DOI] [PubMed] [Google Scholar]
- 43. Song Z, Zheng Y, Wang X, Su H, Zhang Y, Song Y. ALK and ROS1 rearrangements, coexistence and treatment in epidermal growth factor receptor‐wild type lung adenocarcinoma: a multicenter study of 732 cases. J Thorac Dis. 2017;9(10):3919‐3926. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Alexander M, Kim SY, Cheng H. Update 2020: Management of non‐small cell lung cancer. Lung. 2020;198(6):897‐907. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Guerreiro Stucklin AS, Ryall S, Fukuoka K, et al. Alterations in ALK/ROS1/NTRK/MET drive a group of infantile hemispheric gliomas. Nat Commun. 2019;10(1):4343. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Lim SM, Yoo JE, Lim KH, et al. Rare incidence of ROS1 rearrangement in cholangiocarcinoma. Cancer Res Treat. 2017;49(1):185‐192. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Zhu Q, Zhan P, Zhang X, et al. Clinicopathologic characteristics of patients with ROS1 fusion gene in non‐small cell lung cancer: a meta‐analysis. Transl Lung Cancer Res. 2015;4(3):300‐309. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Wang W, Cheng G, Zhang G, Song Z. Evaluation of a new diagnostic immunohistochemistry approach for ROS1 rearrangement in non‐small cell lung cancer. Lung Cancer. 2020;146:224‐229. [DOI] [PubMed] [Google Scholar]
- 49. Yoshida A, Tsuta K, Wakai S, et al. Immunohistochemical detection of ROS1 is useful for identifying ROS1 rearrangements in lung cancers. Mod Pathol. 2014;27(5):711‐720. [DOI] [PubMed] [Google Scholar]
- 50. Conde E, Hernandez S, Martinez R, et al. Assessment of a new ROS1 immunohistochemistry clone (SP384) for the identification of ROS1 rearrangements in patients with non‐small cell lung carcinoma: the ROSING study. J Thorac Oncol. 2019;14(12):2120‐2132. [DOI] [PubMed] [Google Scholar]
- 51. Hofman V, Rouquette I, Long‐Mira E, 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(7):1204‐1212. [DOI] [PubMed] [Google Scholar]
- 52. Boyle TA, Masago K, Ellison KE, Yatabe Y, Hirsch FR. ROS1 immunohistochemistry among major genotypes of non‐small‐cell lung cancer. Clin Lung Cancer. 2015;16(2):106‐111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Pavlakis N, Cooper C, John T, et al. Australian consensus statement for best practice ROS1 testing in advanced non‐small cell lung cancer. Pathology. 2019;51(7):673‐680. [DOI] [PubMed] [Google Scholar]
- 54. Shan L, Lian F, Guo L, et al. Detection of ROS1 gene rearrangement in lung adenocarcinoma: comparison of IHC, FISH and real‐time RT‐PCR. PLoS One. 2015;10(3):e0120422. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Zhang L, Wang Y, Zhao C, et al. High feasibility of cytological specimens for detection of ROS1 fusion by reverse transcriptase PCR in Chinese patients with advanced non‐small‐cell lung cancer. Onco Targets Ther. 2019;12:3305‐3311. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Bubendorf L, Buttner R, Al‐Dayel F, et al. Testing for ROS1 in non‐small cell lung cancer: a review with recommendations. Virchows Arch. 2016;469(5):489‐503. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. 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(14):1405‐1411. [DOI] [PubMed] [Google Scholar]
- 58. Chrzanowska NM, Kowalewski J, Lewandowska MA. Use of fluorescence in situ hybridization (FISH) in diagnosis and tailored therapies in solid tumors. Molecules. 2020;25(8):1864. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Hieggelke L, Schultheis AM. Application of FISH in the diagnosis of lung cancer. Pathologe. 2020;41(6):582‐588. [DOI] [PubMed] [Google Scholar]
- 60. Xu Y, Chang H, Wu L, et al. High prevalence of ROS1 gene rearrangement detected by FISH in EGFR and ALK negative lung adenocarcinoma. Exp Mol Pathol. 2020;117:104548. [DOI] [PubMed] [Google Scholar]
- 61. Fielder T, Butler J, Tierney G, et al. ROS1 rearrangements in lung adenocarcinomas are defined by diffuse strong immunohistochemical expression of ROS1. Pathology. 2022;54(4):399‐403. [DOI] [PubMed] [Google Scholar]
- 62. Rimkunas VM, Crosby KE, Li D, 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(16):4449‐4457. [DOI] [PubMed] [Google Scholar]
- 63. Antonescu CR, Suurmeijer AJ, Zhang L, 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(7):957‐967. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Pisapia P, Lozano MD, Vigliar E, et al. ALK and ROS1 testing on lung cancer cytologic samples: perspectives. Cancer Cytopathol. 2017;125(11):817‐830. [DOI] [PubMed] [Google Scholar]
- 65. Hussen BM, Abdullah ST, Salihi A, et al. The emerging roles of NGS in clinical oncology and personalized medicine. Pathol Res Pract. 2022;230:153760. [DOI] [PubMed] [Google Scholar]
- 66. Morganti S, Tarantino P, Ferraro E, et al. Next generation sequencing (NGS): a revolutionary technology in pharmacogenomics and personalized medicine in cancer. Adv Exp Med Biol. 2019;1168:9‐30. [DOI] [PubMed] [Google Scholar]
- 67. Dong D, Shen G, Da Y, et al. Successful treatment of patients with refractory high‐grade serous ovarian cancer with GOPC‐ROS1 fusion using Crizotinib: a case report. Oncologist. 2020;25(11):e1720‐e1724. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Aggarwal C, Thompson JC, Black TA, et al. Clinical implications of plasma‐based genotyping with the delivery of personalized therapy in metastatic non‐small cell lung cancer. JAMA Oncol. 2019;5(2):173‐180. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Tan AC, Lai GGY, Tan GS, et al. Utility of incorporating next‐generation sequencing (NGS) in an Asian non‐small cell lung cancer (NSCLC) population: incremental yield of actionable alterations and cost‐effectiveness analysis. Lung Cancer. 2020;139:207‐215. [DOI] [PubMed] [Google Scholar]
- 70. Li W, Guo L, Liu Y, et al. Potential unreliability of uncommon ALK, ROS1, and RET genomic breakpoints in predicting the efficacy of targeted therapy in NSCLC. J Thorac Oncol. 2021;16(3):404‐418. [DOI] [PubMed] [Google Scholar]
- 71. Benayed R, Offin M, Mullaney K, et al. High yield of RNA sequencing for targetable kinase fusions in lung adenocarcinomas with no mitogenic driver alteration detected by DNA sequencing and low tumor mutation burden. Clin Cancer Res. 2019;25(15):4712‐4722. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Lovly CM, Heuckmann JM, de Stanchina E, et al. Insights into ALK‐driven cancers revealed through development of novel ALK tyrosine kinase inhibitors. Cancer Res. 2011;71(14):4920‐4931. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Roys A, Chang X, Liu Y, Xu X, Wu Y, Zuo D. Resistance mechanisms and potent‐targeted therapies of ROS1‐positive lung cancer. Cancer Chemother Pharmacol. 2019;84(4):679‐688. [DOI] [PubMed] [Google Scholar]
- 74. Kato Y, Ninomiya K, Ohashi K, et al. Combined effect of cabozantinib and gefitinib in crizotinib‐resistant lung tumors harboring ROS1 fusions. Cancer Sci. 2018;109(10):3149‐3158. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Papadopoulos KP, Borazanci E, Shaw AT, 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(18):4785‐4794. [DOI] [PubMed] [Google Scholar]
- 76. Yun MR, Kim DH, Kim SY, et al. Repotrectinib exhibits potent antitumor activity in treatment‐naive and solvent‐front‐mutant ROS1‐rearranged non‐small cell lung cancer. Clin Cancer Res. 2020;26(13):3287‐3295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Davare MA, Vellore NA, Wagner JP, et al. Structural insight into selectivity and resistance profiles of ROS1 tyrosine kinase inhibitors. Proc Natl Acad Sci USA. 2015;112(39):E5381‐E5390. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Tian Y, Yu Y, Shen Y, et al. Molecular simulation studies on the binding selectivity of type‐I inhibitors in the complexes with ROS1 versus ALK. J Chem Inf Model. 2017;57(4):977‐987. [DOI] [PubMed] [Google Scholar]
- 79. Keddy C, Shinde P, Jones K, et al. Resistance Profile and structural modeling of next‐generation ROS1 tyrosine kinase inhibitors. Mol Cancer Ther. 2022;21(2):336‐346. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Lin JJ, Choudhury NJ, Yoda S, et al. Spectrum of mechanisms of resistance to Crizotinib and Lorlatinib in ROS1 fusion‐positive lung cancer. Clin Cancer Res. 2021;27(10):2899‐2909. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Inoue M, Toki H, Matsui J, et al. Mouse models for ROS1‐fusion‐positive lung cancers and their application to the analysis of multikinase inhibitor efficiency. Carcinogenesis. 2016;37(5):452‐460. [DOI] [PubMed] [Google Scholar]
- 82. Li Y, Chen X, Qu Y, et al. Partial response to ceritinib in a patient with abdominal inflammatory Myofibroblastic tumor carrying a TFG‐ROS1 fusion. J Natl Compr Cancer Netw. 2019;17(12):1459‐1462. [DOI] [PubMed] [Google Scholar]
- 83. Drilon A, Ou SI, Cho BC, 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(10):1227‐1236. [DOI] [PubMed] [Google Scholar]
- 84. Davare MA, Saborowski A, Eide CA, et al. Foretinib is a potent inhibitor of oncogenic ROS1 fusion proteins. Proc Natl Acad Sci USA. 2013;110(48):19519‐19524. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Facchinetti F, Loriot Y, Kuo MS, 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(24):5983‐5991. [DOI] [PubMed] [Google Scholar]
- 86. Rodak O, Peris‐Diaz MD, Olbromski M, et al. Current landscape of non‐small cell lung cancer: epidemiology, histological classification, targeted therapies, and immunotherapy. Cancers (Basel). 2021;13(18):4705. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Yu ZQ, Wang M, Zhou W, et al. ROS1‐positive non‐small cell lung cancer (NSCLC): biology, diagnostics, therapeutics and resistance. J Drug Target. 2022;30(8):845‐857. [DOI] [PubMed] [Google Scholar]
- 88. Shaw AT, Ou SH, Bang YJ, et al. Crizotinib in ROS1‐rearranged non‐small‐cell lung cancer. N Engl J Med. 2014;371(21):1963‐1971. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. 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(7):1121‐1126. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Mazieres J, Zalcman G, Crino L, et al. Crizotinib therapy for advanced lung adenocarcinoma and a ROS1 rearrangement: results from the EUROS1 cohort. J Clin Oncol. 2015;33(9):992‐999. [DOI] [PubMed] [Google Scholar]
- 91. 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(7):1266‐1276. [DOI] [PubMed] [Google Scholar]
- 92. Patil T, Simons E, Mushtaq R, et al. Targeted therapies for ROS1‐rearranged non‐small cell lung cancer. Drugs Today (Barc). 2019;55(10):641‐652. [DOI] [PubMed] [Google Scholar]
- 93. Friboulet L, Li N, Katayama R, et al. The ALK inhibitor ceritinib overcomes crizotinib resistance in non‐small cell lung cancer. Cancer Discov. 2014;4(6):662‐673. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Lim SM, Kim HR, Lee JS, 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(23):2613‐2618. [DOI] [PubMed] [Google Scholar]
- 95. Morris TA, Khoo C, Solomon BJ. Targeting ROS1 rearrangements in non‐small cell lung cancer: Crizotinib and newer generation tyrosine kinase inhibitors. Drugs. 2019;79(12):1277‐1286. [DOI] [PubMed] [Google Scholar]
- 96. Yue D, Qian J, Chen Z, et al. Short‐term response to immune‐chemotherapy and immune features of a ceritinib‐resistant patient with ROS1‐rearranged lung adenocarcinoma. J Immunother Cancer. 2021;9(2):e001967. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Shaw AT, Kim DW, Mehra R, et al. Ceritinib in ALK‐rearranged non‐small‐cell lung cancer. N Engl J Med. 2014;370(13):1189‐1197. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Shaw AT, Kim TM, Crino L, et al. Ceritinib versus chemotherapy in patients with ALK‐rearranged non‐small‐cell lung cancer previously given chemotherapy and crizotinib (ASCEND‐5): a randomised, controlled, open‐label, phase 3 trial. Lancet Oncol. 2017;18(7):874‐886. [DOI] [PubMed] [Google Scholar]
- 99. Rolfo C, Ruiz R, Giovannetti E, et al. Entrectinib: a potent new TRK, ROS1, and ALK inhibitor. Expert Opin Investig Drugs. 2015;24(11):1493‐1500. [DOI] [PubMed] [Google Scholar]
- 100. Drilon A, Siena S, Dziadziuszko R, et al. Entrectinib in ROS1 fusion‐positive non‐small‐cell lung cancer: integrated analysis of three phase 1‐2 trials. Lancet Oncol. 2020;21(2):261‐270. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Dziadziuszko R, Krebs MG, De Braud F, et al. Updated integrated analysis of the efficacy and safety of entrectinib in locally advanced or metastatic ROS1 fusion‐positive non‐small‐cell lung cancer. J Clin Oncol. 2021;39(11):1253‐1263. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102. Fischer H, Ullah M, de la Cruz CC, et al. Entrectinib, a TRK/ROS1 inhibitor with anti‐CNS tumor activity: differentiation from other inhibitors in its class due to weak interaction with P‐glycoprotein. Neuro‐Oncology. 2020;22(6):819‐829. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Sehgal K, Piper‐Vallillo AJ, Viray H, et al. Cases of ROS1‐rearranged lung cancer: when to use crizotinib, entrectinib, lorlatinib, and beyond? Precis Cancer Med. 2020;3:17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Frampton JE. Entrectinib: a review in NTRK+ solid tumours and ROS1+NSCLC. Drugs. 2021;81(6):697‐708. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105. Chong CR, Bahcall M, Capelletti M, et al. Identification of existing drugs that effectively target NTRK1 and ROS1 rearrangements in lung cancer. Clin Cancer Res. 2017;23(1):204‐213. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Shaw AT, Solomon BJ, Chiari R, 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(12):1691‐1701. [DOI] [PubMed] [Google Scholar]
- 107. Drilon A, Camidge DR, Lin JJ, et al. Repotrectinib in ROS1 fusion‐positive non‐small‐cell lung cancer. N Engl J Med. 2024;390(2):118‐131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Ou SI, Fujiwara Y, Shaw AT, et al. Efficacy of taletrectinib (AB‐106/DS‐6051b) in ROS1+ NSCLC: an updated pooled analysis of U.S. and Japan phase 1 studies. JTO Clin Res Rep. 2021;2(1):100108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. Ma Y, Zhao H, Xue J, et al. First‐in‐human phase I study of TQ‐B3139 (CT‐711) in advanced non‐small cell lung cancer patients with ALK and ROS1 rearrangements. Eur J Cancer. 2022;173:238‐249. [DOI] [PubMed] [Google Scholar]
- 110. Sun TY, Niu X, Chakraborty A, Neal JW, Wakelee HA. Lengthy progression‐free survival and intracranial activity of Cabozantinib in patients with crizotinib and ceritinib‐resistant ROS1‐positive non‐small cell lung cancer. J Thorac Oncol. 2019;14(2):e21‐e24. [DOI] [PubMed] [Google Scholar]
- 111. Dudnik E, Agbarya A, Grinberg R, et al. Clinical activity of brigatinib in ROS1‐rearranged non‐small cell lung cancer. Clin Transl Oncol. 2020;22(12):2303‐2311. [DOI] [PubMed] [Google Scholar]
- 112. Solomon BJ, Besse B, Bauer TM, et al. Lorlatinib in patients with ALK‐positive non‐small‐cell lung cancer: results from a global phase 2 study. Lancet Oncol. 2018;19(12):1654‐1667. [DOI] [PubMed] [Google Scholar]
- 113. Katayama R, Gong B, Togashi N, et al. The new‐generation selective ROS1/NTRK inhibitor DS‐6051b overcomes crizotinib resistant ROS1‐G2032R mutation in preclinical models. Nat Commun. 2019;10(1):3604. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Katayama R, Kobayashi Y, Friboulet L, et al. Cabozantinib overcomes crizotinib resistance in ROS1 fusion‐positive cancer. Clin Cancer Res. 2015;21(1):166‐174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. Hegde A, Hong DS, Behrang A, et al. Activity of brigatinib in crizotinib and ceritinib‐resistant ROS1‐rearranged non‐small‐cell lung cancer. JCO Precis Oncol. 2019;3:1‐6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116. Choudhury NJ, Schneider JL, Patil T, et al. Response to immune checkpoint inhibition as monotherapy or in combination with chemotherapy in metastatic ROS1‐rearranged lung cancers. JTO Clin Res Rep. 2021;2(7):100187. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117. Guisier F, Dubos‐Arvis C, Viñas F, et al. Efficacy and safety of anti‐PD‐1 immunotherapy in patients with advanced NSCLC with BRAF, HER2, or MET mutations or RET translocation: GFPC 01‐2018. J Thorac Oncol. 2020;15(4):628‐636. [DOI] [PubMed] [Google Scholar]
- 118. Mazieres J, Drilon A, Lusque A, et al. Immune checkpoint inhibitors for patients with advanced lung cancer and oncogenic driver alterations: results from the IMMUNOTARGET registry. Ann Oncol. 2019;30(8):1321‐1328. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119. Bylicki O, Tomasini P, Radj G, et al. Atezolizumab with or without bevacizumab and platinum‐pemetrexed in patients with stage IIIB/IV non‐squamous non‐small cell lung cancer with EGFR mutation, ALK rearrangement or ROS1 fusion progressing after targeted therapies: a multicentre phase II open‐label non‐randomised study GFPC 06‐2018. Eur J Cancer. 2023;183:38‐48. [DOI] [PubMed] [Google Scholar]
- 120. Comandini D, Catalano F, Grassi M, et al. Outstanding response in a patient with ROS1‐rearranged inflammatory myofibroblastic tumor of soft tissues treated with Crizotinib: case report. Front Oncol. 2021;11:658327. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121. Robertson SJ, Orme L, Teixeira R, et al. Evaluation of Crizotinib treatment in a patient with unresectable GOPC‐ROS1 fusion agminated Spitz nevi. JAMA Dermatol. 2021;157(7):836‐841. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122. Jakubowski CD, Mohan AA, Kamel IR, Yarchoan M. Response to Crizotinib in ROS1 fusion‐positive intrahepatic Cholangiocarcinoma. JCO Precis Oncol. 2020;4:825‐828. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123. Li J, Liu L, Zhang Q, et al. A novel TJP1‐ROS1 fusion in malignant peripheral nerve sheath tumor responding to crizotinib: a case report. Medicine (Baltimore). 2020;99(26):e20725. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124. Mayr L, Guntner AS, Madlener S, et al. Cerebrospinal fluid penetration and combination therapy of entrectinib for disseminated ROS1/NTRK‐fusion positive pediatric high‐grade glioma. J Pers Med. 2020;10(4):290. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125. Papusha L, Zaytseva M, Panferova A, et al. Two clinically distinct cases of infant hemispheric glioma carrying ZCCHC8:ROS1 fusion and responding to entrectinib. Neuro‐Oncology. 2022;24(6):1029‐1031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126. Couts KL, McCoach CE, Murphy D, et al. Acral lentiginous melanoma harboring a ROS1 gene fusion with clinical response to entrectinib. JCO Precis Oncol. 2017;1:1‐7. [DOI] [PubMed] [Google Scholar]
- 127. Pishvaian MJ, Garrido‐Laguna I, Liu SV, Multani PS, Chow‐Maneval E, Rolfo C. Entrectinib in TRK and ROS1 fusion‐positive metastatic pancreatic cancer. JCO Precis Oncol. 2018;2:1‐7. [DOI] [PubMed] [Google Scholar]
- 128. Velthaus JL, Iglauer P, Simon R, et al. Lorlatinib induces durable disease stabilization in a pancreatic cancer patient with a ROS1 p.L1950F mutation: case report. Oncol Res Treat. 2021;44(9):495‐502. [DOI] [PubMed] [Google Scholar]
- 129. Carcamo B, Bista R, Wilson H, Reddy P, Pacheco J. Rapid response to lorlatinib in a patient with TFG‐ROS1 fusion positive inflammatory myofibroblastic tumor of the chest wall metastatic to the brain and refractory to first and second generation ROS1 inhibitors. J Pediatr Hematol Oncol. 2021;43(5):e718‐e722. [DOI] [PubMed] [Google Scholar]
- 130. Spigel DR, Reynolds C, Waterhouse D, et al. Phase 1/2 study of the safety and tolerability of Nivolumab plus Crizotinib for the first‐line treatment of anaplastic lymphoma kinase translocation ‐ positive advanced non‐small cell lung cancer (CheckMate 370). J Thorac Oncol. 2018;13(5):682‐688. [DOI] [PubMed] [Google Scholar]
- 131. Bauer TM, Felip E, Solomon BJ, et al. Clinical management of adverse events associated with lorlatinib. Oncologist. 2019;24(8):1103‐1110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132. Cocco E, Scaltriti M, Drilon A. NTRK fusion‐positive cancers and TRK inhibitor therapy. Nat Rev Clin Oncol. 2018;15(12):731‐747. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133. Drilon A, Clark JW, Weiss J, et al. Antitumor activity of crizotinib in lung cancers harboring a MET exon 14 alteration. Nat Med. 2020;26(1):47‐51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134. Camidge DR, Otterson GA, Clark JW, et al. Crizotinib in patients with MET‐amplified NSCLC. J Thorac Oncol. 2021;16(6):1017‐1029. [DOI] [PubMed] [Google Scholar]
- 135. Lin JJ, Chin E, Yeap BY, et al. Increased hepatotoxicity associated with sequential immune checkpoint inhibitor and Crizotinib therapy in patients with non‐small cell lung cancer. J Thorac Oncol. 2019;14(1):135‐140. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136. Fortes BH, Tailor PD, Dalvin LA. Ocular toxicity of targeted anticancer agents. Drugs. 2021;81(7):771‐823. [DOI] [PubMed] [Google Scholar]
- 137. Li YX, Yang JY, Xu YF, et al. A meta‐analysis of the comparing of the first‐generation and next‐generation TKIs in the treatment of NSCLC. Math Biosci Eng. 2019;16(5):5687‐5696. [DOI] [PubMed] [Google Scholar]
- 138. Zhang L, Ren HW, Wu QL, Wu YJ, Song X. The effect of next‐generation TKI in non‐small cell lung cancer after failure of first‐line treatment: a meta‐analysis. Pathol Oncol Res. 2020;26(2):1137‐1143. [DOI] [PubMed] [Google Scholar]
- 139. McCoach CE, Le AT, Gowan K, et al. Resistance mechanisms to targeted therapies in ROS1(+) and ALK(+) non‐small cell lung cancer. Clin Cancer Res. 2018;24(14):3334‐3347. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140. Vaishnavi A, Schubert L, Rix U, et al. EGFR mediates responses to small‐molecule drugs targeting oncogenic fusion kinases. Cancer Res. 2017;77(13):3551‐3563. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141. Guaitoli G, Bertolini F, Bettelli S, et al. Deepening the knowledge of ROS1 rearrangements in non‐small cell lung cancer: diagnosis, treatment, resistance and concomitant alterations. Int J Mol Sci. 2021;22(23):12867. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142. 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;2017:1‐13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143. Song A, Kim TM, Kim DW, et al. Molecular changes associated with acquired resistance to Crizotinib in ROS1‐rearranged non‐small cell lung cancer. Clin Cancer Res. 2015;21(10):2379‐2387. [DOI] [PubMed] [Google Scholar]
- 144. Awad MM, Engelman JA, Shaw AT. Acquired resistance to crizotinib from a mutation in CD74‐ROS1. N Engl J Med. 2013;369(12):1173. [DOI] [PubMed] [Google Scholar]
- 145. Drilon A, Somwar R, Wagner JP, et al. A novel Crizotinib‐resistant solvent‐front mutation responsive to Cabozantinib therapy in a patient with ROS1‐rearranged lung cancer. Clin Cancer Res. 2016;22(10):2351‐2358. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146. Wu X, Fu Y, Wang Y, Wan SH, Zhang JJ. Gaining insight into crizotinib resistance mechanisms caused by L2026M and G2032R mutations in ROS1 via molecular dynamics simulations and free‐energy calculations. J Mol Model. 2017;23(4):141. [DOI] [PubMed] [Google Scholar]
- 147. Sasaki T, Koivunen J, Ogino A, et al. A novel ALK secondary mutation and EGFR signaling cause resistance to ALK kinase inhibitors. Cancer Res. 2011;71(18):6051‐6060. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148. 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(5):1472‐1482. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149. Dziadziuszko R, Le AT, Wrona A, et al. An activating KIT mutation induces Crizotinib resistance in ROS1‐positive lung cancer. J Thorac Oncol. 2016;11(8):1273‐1281. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150. Yang X, Tang Z, Li J, Jiang J, Liu Y. Progress of non‐small‐cell lung cancer with ROS1 rearrangement. Front Mol Biosci. 2023;10:1238093. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151. Lin JJ, Langenbucher A, Gupta P, et al. Small cell transformation of ROS1 fusion‐positive lung cancer resistant to ROS1 inhibition. NPJ Precis Oncol. 2020;4:21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152. Killock D. Lorlatinib in ROS1‐positive NSCLC. Nat Rev Clin Oncol. 2020;17(1):7. [DOI] [PubMed] [Google Scholar]
- 153. Davies KD, Mahale S, Astling DP, et al. Resistance to ROS1 inhibition mediated by EGFR pathway activation in non‐small cell lung cancer. PLoS One. 2013;8(12):e82236. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154. 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(11):1717‐1726. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155. 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(7):987‐995. [DOI] [PubMed] [Google Scholar]
- 156. Jun HJ, Johnson H, Bronson RT, de Feraudy S, White F, Charest A. The oncogenic lung cancer fusion kinase CD74‐ROS activates a novel invasiveness pathway through E‐Syt1 phosphorylation. Cancer Res. 2012;72(15):3764‐3774. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157. Landi L, Cappuzzo F. How selecting best upfront therapy for metastatic disease?‐focus on ROS1‐rearranged disease. Transl Lung Cancer Res. 2020;9(6):2686‐2695. [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.
Data Availability Statement
The materials that support the conclusion of this review have been included within the article.
