Ltk mutations responsible for resistance to lorlatinib in non-small cell lung cancer harboring clip1-ltk fusion

Ltk mutations responsible for resistance to lorlatinib in non-small cell lung cancer harboring clip1-ltk fusion

Play all audios:

Loading...

ABSTRACT The _CLIP1-LTK_ fusion was recently discovered as a novel oncogenic driver in non-small cell lung cancer (NSCLC). Lorlatinib, a third-generation ALK inhibitor, exhibited a dramatic


clinical response in a NSCLC patient harboring _CLIP1-LTK_ fusion. However, it is expected that acquired resistance will inevitably develop, particularly by _LTK_ mutations, as observed in


NSCLC induced by oncogenic tyrosine kinases treated with corresponding tyrosine kinase inhibitors (TKIs). In this study, we evaluate eight _LTK_ mutations corresponding to _ALK_ mutations


that lead to on-target resistance to lorlatinib. All _LTK_ mutations show resistance to lorlatinib with the L650F mutation being the highest. In vitro and in vivo analyses demonstrate that


gilteritinib can overcome the L650F-mediated resistance to lorlatinib. In silico analysis suggests that introduction of the L650F mutation may attenuate lorlatinib-LTK binding. Our study


provides preclinical evaluations of potential on-target resistance mutations to lorlatinib, and a novel strategy to overcome the resistance. SIMILAR CONTENT BEING VIEWED BY OTHERS ANALYSIS


OF LORLATINIB ANALOGS REVEALS A ROADMAP FOR TARGETING DIVERSE COMPOUND RESISTANCE MUTATIONS IN ALK-POSITIVE LUNG CANCER Article 20 June 2022 ANTI-EGFR THERAPY CAN OVERCOME ACQUIRED


RESISTANCE TO THE THIRD-GENERATION ALK-TYROSINE KINASE INHIBITOR LORLATINIB MEDIATED BY ACTIVATION OF EGFR Article 21 March 2025 THIRD-GENERATION EGFR AND ALK INHIBITORS: MECHANISMS OF


RESISTANCE AND MANAGEMENT Article 09 May 2022 INTRODUCTION The discovery of several actionable oncogenic drivers in non-small cell lung cancer (NSCLC) and the development of corresponding


targeted therapies have changed the treatment strategy, leading to great improvement in patient outcome1. The _CLIP1- LTK_ fusion is identified as a novel oncogenic driver in NSCLC using a


large-scale lung cancer genome screening platform (LC-SCRUM-Asia; UMIN000036871)2,3. The CLIP1-LTK fusion protein constitutively activates LTK and its downstream signaling molecules,


including AKT and ERK4, resulting in cell proliferation and the suppression of apoptosis. This fusion gene is present in 0.4% of NSCLC and is mutually exclusive of other known oncogenic


drivers. Interestingly, ALK-tyrosine kinase inhibitors (TKIs), especially lorlatinib, a third-generation ALK-TKI, were effective in cells expressing the _CLIP1-LTK_ fusion in vitro and in


vivo. The rationale for the use of ALK-TKIs is based on the fact that LTK and ALK share nearly 80% protein sequence identity in their kinase domains, and most ALK-TKIs demonstrate LTK


inhibition at ALK-inhibitory concentrations5,6. Notably, we also demonstrated that lorlatinib exhibits a dramatic and durable response in a patient with NSCLC harboring _CLIP1-LTK_ fusion.


However, despite the remarkable efficacy of lorlatinib against NSCLC patients harboring _CLIP1-LTK_, acquired resistance to lorlatinib will inevitably develop, as observed in NSCLC induced


by oncogenic tyrosine kinases treated with corresponding TKIs7,8,9,10,11. Therefore, it is essential to identify the potential resistance mechanisms of lorlatinib in _LTK_ fusion-positive


NSCLC and establish effective treatment strategies to overcome this resistance. The mechanism of lorlatinib resistance in _LTK_ fusion-positive NSCLC is yet to be elucidated. In general,


resistance mechanisms against targeted therapies are divided into three groups: (1) on-target gene alterations12,13, (2) off-target mechanisms such as the upregulation of alternative bypass


pathways, including _MET_ amplifications14,15, and (3) histological transformations16,17. Among these resistance mechanisms, on-target gene alterations account for 50-70% of patients treated


with respective targeted therapies13,18. In this study, we demonstrated that _LTK_ mutations, especially the L650F mutation, potentially confer resistance to lorlatinib treatment, and that


L650F-mediated resistance to lorlatinib can be overcome by gilteritinib. RESULTS LORLATINIB IS PREDICTED TO BIND TO LTK AND ALK _LTK_ and _ALK_ belong to the insulin receptor subfamily of


receptor tyrosine kinases, which consist of an extracellular region, transmembrane region, and intracellular region. The kinase domain of LTK and ALK contains 268 amino acids (Fig. 1a).


Intriguingly, LTK and ALK exhibit 79% amino acid homology in their respective kinase domains19, and lorlatinib inhibits LTK at ALK-inhibitory concentrations5. We first evaluated the binding


affinity of lorlatinib to LTK proteins using in silico approaches to further support the efficacy of lorlatinib in _CLIP1-LTK_ fusion-expressing cells2. Molecular dynamics (MD) simulations


indicated that lorlatinib fitted into the ATP-binding pocket of LTK and was stabilized by hydrogen bonds with backbone amides of E591 (corresponding to E1197 in ALK) and M593 (corresponding


to M1199 in ALK) (Fig. 1b). In addition, the estimated LTK-lorlatinib binding free energy (ΔG) of −11.6 ± 0.8 kcal/mol is similar to that between ALK and lorlatinib (−14.3 ± 1.3 kcal/mol)9.


These results suggest that lorlatinib binds to LTK in a manner similar to ALK, further supporting the efficacy of lorlatinib in tumors expressing _CLIP1-LTK_ fusion2. CONSERVED AND


HOMOLOGOUS SEQUENCE OF LTK/ALK IN THE KINASE DOMAIN RESPONSIBLE FOR TKI RESISTANCE The most common resistance mechanism to genotype-matched therapy is caused by acquired genetic alterations


in the on-target gene, including gatekeeper or solvent-front mutations12,13,20,21. Various ALK-TKI resistance mutations have been identified in _ALK_ fusion-positive NSCLC, most of which


occur in the ALK kinase domain. Among these kinase domains, eight residues (I1171, F1174, L1196, L1198, G1202, D1203, L1256, G1269) are responsible for lorlatinib resistance in _ALK_


fusion-positive NSCLC in clinical setting and/or experimental models7,9,13,22. As LTK and ALK exhibit 79% amino acid homology in their respective kinase domains (Fig. 2a), we hypothesized


that corresponding mutations to these _ALK_-acquired mutations may emerge in _LTK_ fusion-positive cells treated with lorlatinib. Indeed, all these residues are conserved in the LTK protein


(Fig. 2a). Thus, we hypothesized that _LTK_ mutations analogous to _ALK_ mutations could emerge, resulting in lorlatinib resistance (Fig. 2b). ANALOGOUS _LTK_ MUTATIONS SHOW LORLATINIB


RESISTANCE We then established Ba/F3 and NIH3T3 cells expressing CLIP1-LTK fusion proteins with the aforementioned _LTK_ mutations to clarify the impact of these mutations on sensitivity to


lorlatinib, as well as other targeted agents. Cell viability assays using Ba/F3 cells expressing WT _CLIP1-LTK_ or each _CLIP1-LTK_ mutation revealed that Ba/F3 cells expressing mutant


_CLIP1-LTK_ were less sensitive to lorlatinib compared with those expressing WT _CLIP1-LTK_ (Fig. 3a). The western blotting assay also showed that the effect of lorlatinib on LTK tyrosine


phosphorylation was attenuated in Ba/F3 cells expressing mutant _CLIP1-LTK_ compared with Ba/F3 cells expressing WT _CLIP1-LTK_. Ten nM of lorlatinib or higher inhibited the LTK tyrosine


phosphorylation of Ba/F3 cells expressing WT _CLIP1-LTK_, whereas that of _CLIP1-LTK_ with kinase mutations was not inhibited by 10 nM of lorlatinib (Fig. 3b). In particular, the L650F


mutation was the most resistant to lorlatinib in terms of inhibition of cell proliferation with IC50 value as well as LTK phosphorylation (Fig. 3a, b). Furthermore, cell apoptosis was


evaluated in Ba/F3 cells expressing WT or mutant _CLIP1-LTK_ treated with lorlatinib. Ba/F3 cells expressing _CLIP1-LTK_-L592F were more susceptible to lorlatinib than those expressing other


_CLIP1-LTK_ mutations possibly due to differences in cell growth rates. However, apoptosis was significantly suppressed in all _CLIP1-LTK_ mutant Ba/F3 cells compared to those expressing WT


_CLIP1-LTK_ (Fig. 3c). These results suggested that these _LTK_ mutations are resistant to lorlatinib-induced LTK kinase inhibition and cell apoptosis. CELL VIABILITY PROFILES OF ALK


INHIBITORS Next, we explored potential compounds that could overcome lorlatinib resistance mediated by these _LTK_ mutations and evaluated the sensitivity of the following compounds:


lorlatinib, crizotinib, alectinib, ceritinib, brigatinib, entrectinib, repotrectinib, and gilteritinib. To compare the sensitivity to each compound in Ba/F3 cells expressing WT or mutant


_CLIP1-LTK_, the IC50 values of the compounds were determined in a cell viability assay. The IC50 values of these eight compounds to Ba/F3 cells expressing WT _CLIP1-LTK_ or other mutations


are shown in Fig. 4. All _LTK_ mutations showed resistance to lorlatinib, with IC50 values of lorlatinib ranging 2.0 to 11,070 nM, which were higher than that of WT _CLIP-LTK_ (1.0 nM).


Notably, the IC50 of lorlatinib against Ba/F3 cells expressing _CLIP1-LTK_-L650F was 11,070 nM, which was the highest among the _LTK_ mutations tested in this study, while the IC50 of


gilteritinib against Ba/F3 cells expressing _CLIP1-LTK_-L650F was 23.7 nM, which was the lowest among the tested compounds. Moreover, gilteritinib can also inhibit Ba/F3 expressing WT


_CLIP1-LTK_, with IC50 value of 0.3 nM, which is consistent with a previous report demonstrating gilteritinib shows LTK inhibition at a similar concentration23. We also explored potential


compounds that could overcome resistance by G596R, L650F, and G663A, which may induce high-level resistance to lorlatinib. The western blotting assay showed that repotrectinib inhibited LTK


phosphorylation of CLIP1-LTK-G596R at a concentration of 100 nM or higher, whereas lorlatinib at a concentration of 1 µM or higher was required to achieve this (Supplementary Fig. 1).


Similarly, gilteritinib successfully inhibited LTK phosphorylation of CLIP1-LTK-L650F or G663A at 100 nM, whereas lorlatinib did so at concentrations of 1 µM or higher (Supplementary Fig. 


1). In addition, we evaluated a combination of repotrectinib and gilteritinib using Ba/F3 cells expressing WT or mutant _CLIP1-LTK_. The combination significantly enhanced suppression of


cell viability compared to repotrectinib or gilteritinib alone in most cells. However, such effect was not observed in Ba/F3 cells expressing WT _CLIP1-LTK_ or L650F (Supplementary Fig. 2).


GILTERITINIB OVERCOMES LORLATINIB RESISTANCE BY _LTK_ L650F MUTATION IN VITRO AND IN VIVO We further focused on _CLIP1-LTK_-L650F, which was the most resistant strain to lorlatinib in this


study. Among the tested compounds, gilteritinib was the most potent in Ba/F3 cells expressing _CLIP1-LTK_-L650F (Fig. 5a). Therefore, we investigated whether gilteritinib could overcome


resistance to lorlatinib induced by _CLIP1-LTK_-L650F. The western blotting assay showed that gilteritinib successfully inhibited LTK phosphorylation in Ba/F3 cells expressing


_CLIP1-LTK_-L650F. Indeed, at 100 nM, gilteritinib strongly attenuated AKT and ERK phosphorylation, whereas lorlatinib did not. In addition, gilteritinib increased the levels of the


stabilized form of BIM and cleaved caspase-3, the hallmark of apoptosis (Fig. 5b). Fluorescence-activated cell sorting (FACS) analysis using annexin V/propidium iodide (PI) staining also


confirmed that gilteritinib induced apoptosis in Ba/F3 cells carrying _CLIP1-LTK_-L650F (Fig. 5c). An increase in caspase activity by gilteritinib, but not lorlatinib, also supported the


successful induction of apoptosis by gilteritinib (Supplementary Fig. 3). We subsequently investigated the inhibitory effect of gilteritinib in another cell model, NIH3T3 cells carrying


_CLIP1-LTK_-L650F, using a soft agar colony formation assay. The diameter of colonies treated with gilteritinib was significantly smaller than that treated with lorlatinib or


dimethylsulfoxide (DMSO), whereas lorlatinib did not inhibit colony formation compared with DMSO (Fig. 5d). Finally, we tested the activity of gilteritinib against _CLIP1-LTK_-L650F cells


using a xenograft model. Consistent with the results of the in vitro experiments, there was no significant difference in tumor size between the lorlatinib and vehicle control groups,


suggesting the robust resistance of _CLIP1-LTK_-L650F to lorlatinib. In contrast, gilteritinib significantly inhibited tumor growth compared with the vehicle control or lorlatinib (Fig. 5e).


Notably, no significant difference in body weight was detected among these three groups, suggesting that gilteritinib showed less toxicity (Fig. 5f). Collectively, gilteritinib potentially


overcame the L650F-mediated resistance to lorlatinib in tumors expressing _CLIP1-LTK_-L650F. L650F MUTATION DISTURB LORLATINIB BINDING TO LTK We further explored how these _LTK_ mutations


affect sensitivity to lorlatinib and gilteritinib. As lorlatinib failed to inhibit LTK phosphorylation in cells with _LTK_ mutations, we speculated that these mutations affected


LTK-lorlatinib binding. We estimated the binding affinity of lorlatinib against WT CLIP1-LTK and its mutants using the Massively Parallel Computation of Absolute binding Free Energy with


well-equilibrated states (MP-CAFEE) method24 and found that the IC50 of lorlatinib showed in Fig. 4 was well correlated with the LTK-lorlatinib ΔG, with a correlation coefficient (R) of


0.509 (Fig. 6a). We also observed a moderate correlation between the IC50 and ΔG values of gilteritinib, with an R of 0.597 (Fig. 6b). These results suggested that decreased LTK-drug binding


affinity is a major contributor to mutation-induced drug resistance. For example, the binding affinity of lorlatinib to CLIP1-LTK-L650F (ΔG, −5.4 ± 1.4) was significantly lower than that of


WT CLIP1-LTK (ΔG, −11.6 ± 0.8) due to the loss of intermolecular van der Waals interactions, leading to a large displacement of the drug in the pocket (Fig. 6c). In contrast, no significant


difference in the binding affinity of gilteritinib was observed between WT CLIP1-LTK and CLIP1-LTK-L650F, suggesting that this mutation has little effect on gilteritinib binding (Fig. 6d).


DISCUSSION This study was the first to explore potential _LTK_ resistance alternations against lorlatinib in tumors expressing the CLIP1-LTK fusion protein. We found that all eight _LTK_


tested mutations were responsible for lorlatinib resistance, among which the L650F mutation showed the most robust resistance to lorlatinib. We also demonstrated that gilteritinib was an


exquisite and potent inhibitor of _CLIP1-LTK_-L650F in in vivo and in vitro experiments. _LTK_ fusion is a rare but actionable oncogenic driver in NSCLC2. To clinically develop an effective


targeted therapy, an investigator-initiated clinical trial (IIT) of lorlatinib for advanced NSCLC with _LTK_ fusion is ongoing using the LC-SCRUM-Asia (jRCT2031220600). However, acquired


resistance to lorlatinib inevitably develops despite the expected initial favorable efficacy. To develop more efficient resistance mechanism-matched therapies, we explored the potential


mechanism of resistance to lorlatinib in preclinical models. In general, resistance mechanisms against targeted therapies are divided into three groups: 1) on-target gene alterations7,13, 2)


off-target mechanisms, such as the upregulation of alternative bypass pathways, including _MET_ amplifications14,15, and 3) histological transformations16,17 Among these resistance


mechanisms, on-target gene alterations account for 50-70% of patients treated with respective targeted therapies13. In this study, we focused on _LTK_ mutations among the various resistance


mechanisms against ALK-TKIs. However, it is certainly possible that other resistance mechanisms, including off-target mechanisms, such as the upregulation of alternative bypass pathways, can


emerge. Along with LC-SCRUM-Asia, we conduct genomic screening for treatment-resistant patients with advanced NSCLC (LC-SCRUM-TRY; UMIN000041957) to identify the resistance mechanisms and


support the clinical development of resistance mechanism-matched therapies. We will explore the mechanism of lorlatinib resistance using clinical samples obtained from patients enrolled in


the IIT. In this study, we showed that gilteritinib can inhibit kinase activity of CLIP1-LTK with _LTK_ mutations including L650F, as well as WT _CLIP1-LTK_. Gilteritinib may be an


alternative option for _LTK_ fusion-positive NSCLC as either a first TKI treatment or second TKI after lorlatinib treatment. Considering similar pattern of drug sensitivity between _ALK_ and


_LTK_, and the rarity of patients harboring specific resistance mechanisms, basket-type trials of targeted therapy for patients with specific resistance mutations might be useful, for


example for L1256F/L650F mutated _ALK/LTK_ fusion-positive NSCLC to efficiently develop targeted therapy for rare fusion-positive NSCLC resistant to prior targeted therapies. In summary,


_LTK_ mutations analogous to _ALK_ mutations were resistant to lorlatinib, with the L650F mutation being the most potent. Our preclinical models demonstrate that gilteritinib may be a


promising strategy to overcome L650F-mediated resistance. MATERIALS AND METHODS CELL LINES AND REAGENTS NIH3T3 cells were purchased from American Type Culture Collection (ATCC). Ba/F3, WEHI,


and BOSC23 cells were kindly provided by Dr. Daniel G. Tenen (Harvard Medical School). Crizotinib, ceritinib, alectinib, brigatinib, lorlatinib, entrectinib, gilteritinib, and repotrectinib


were purchased from Selleck. NIH3T3 cells were maintained in Dulbecco’s Modified Eagle Medium (DMEM) supplemented with 10% fetal bovine serum (FBS), 100 units/ml penicillin, and 100 mg/ml


streptomycin (P/S). Parental Ba/F3 cells were maintained in RPMI1640 supplemented with 5% WEHI (as a source of IL-3), 10% FBS, and P/S. Ba/F3 cells expressing _CLIP1-LTK_ mutants were


maintained in RPMI1640 supplemented with 10% FBS and P/S. All cell lines were routinely tested for mycoplasma infection and negative for mycoplasma infection. CONSTRUCTION OF PLASMID The


MIGR1 retroviral vector harboring CLIP1-LTK fusion protein was constructed as previously described2. Plasmids expressing each mutant _CLIP1-LTK_ (I565N, F568C, L590M, L592F, G596R, D597N,


L650F, and G663A) were generated using the Quick Change Lightning Site-Directed Mutagenesis Kit (Agilent). All the primers used are listed in Supplementary Table 1. The integrity of all


constructs was confirmed by Sanger sequencing. VIRAL TRANSDUCTION Ba/F3 and NIH3T3 cells expressing WT _CLIP1-LTK_ fusion or various mutant _CLIP1-LTK_ fusions were generated by retroviral


transduction as previously described2. WESTERN BLOTTING Cells were lysed in sodium dodecyl sulfate (SDS) sample buffer and boiled for 5 min. Lysates were subjected to SDS polyacrylamide gel


electrophoresis and blotted onto poly (vinylidene fluoride) (PVDF) membranes (Millipore). The antibodies and dilutions used are listed in Supplementary Table 2. Images were captured using


ImageQuant LAS 4000 (GE Healthcare) and analyzed using the ImageJ software (ver. 1.53). All images were assembled, and figures were generated using the Affinity Designer (ver. 1.10.5), and


Microsoft PowerPoint 2016 (ver. 2108). CELL VIABILITY ASSAY Ba/F3 cells (10,000 cells per well) were seeded in 96-well plates and treated with inhibitors of interest for 48 h, and viability


was evaluated using the Cell Counting Kit-8 (Fujifilm). Data were captured using the Spectra Max Paradigm (Molecular Devices) with SoftMax Pro software (ver.7.10). Absorbance was measured at


a wavelength of 450 nm. The IC50 value was determined using a nonlinear regression model (four parameters) using the GraphPad Prism software (ver. 9.3.1). SOFT AGER FORMATION ASSAY NIH3T3


cells expressing _CLIP1-LTK_-L650F (30,000 cells per well) were seeded in 6-well plates and treated with the inhibitors of interest for 14 days. Cell images were captured using the BZ-II


Viewer software (v. 2.10), and the diameter of the colonies was measured using ImageJ software (ver. 1.53). APOPTOSIS ASSAY Ba/F3 cells (100,000 cells/well) were seeded in 6-well plates.


After they were treated with the inhibitors of interest for 24 h, they were stained with annexin-V and PI using the Annexin V-FITC Apoptosis Detection Kit (Nacalai Tesque). A total of 10,000


cells were captured using FACSDiva software (v. 9.0). FACS data were analyzed using FlowJo software (v. 10.7.1). Gating was conducted to detect single cells and then determined so that


there were no annexin V-positive cells in untreated Ba/F3 cells. Cells undergoing apoptosis were defined as annexin V-positive cells. The Caspase-Glo3/7 Assay System (Promega) was used to


evaluate cell apoptosis. Ba/F3 cells (5000 cells/well) were seeded in 96-well plates and treated with the indicated drugs for 12 h. Data were captured using the Spectra Max Paradigm


(Molecular Devices) with SoftMax Pro software (ver.7.10). Absorbance was measured at 490 nm. MOLECULAR DOCKING Molecular docking of alectinib, gilteritinib, and lorlatinib with the


LTK-tyrosine kinase domain was performed using GOLD 5.5. Standard default settings for the genetic algorithm were used. The structure of the LTK kinase domain was predicted using


AlphaFold225. The dominant protonation state at pH 7.0 was assigned to titratable residues. The ATP-binding site was defined to include all atoms within 10 Å of the midpoint of the Leu516 Cα


and Gly596 Cα atoms. Alectinib, gilteritinib, and lorlatinib, whose 3D structures were obtained from the crystal structures of ALK-alectinib (PDBID:3AOX), FLT3-gilteritinib (PDBID:6JQR),


and ALK-lorlatinib complexes (PDBID:4CLI), respectively, were protonated to form ionization states in solution (net charges of +1, +2, and 0, respectively). After the backbone Cα atoms in


LTK were structurally aligned with those in each crystal structure, alectinib, gilteritinib, and lorlatinib were docked into the ATP-binding site in LTK with positional restraints on the


benzocarbazole, pyrazinamide, and cyclotetradecine moieties, respectively, assuming that these drugs had a similar binding geometry between LTK and ALK/FLT3. The top-ranked docking pose was


extracted and used as the initial structure for MD simulations of the LTK drug complexes. MD SIMULATION OF WILD-TYPE LTK OR ITS MUTANTS IN COMPLEX WITH DRUGS Each of I565N, F568C, L590M,


L592F, G596R, D597N, L650F, and G663A mutations were introduced into the structural model of WT LTK using the MODELER program26. According to a previously described procedure27,


computational systems of LTK-drug complexes were prepared, and MD simulations were carried out. For each LTK mutant, five independent production runs of 50 ns (with different atomic


velocities) were performed in a constant number of molecules, pressure, and temperature (NPT) ensemble, where the temperature was maintained at 298 K by stochastic velocity rescaling28. A


Parrinello-Rahman barostat was used to maintain the pressure at 1 bar29, with the temperature and pressure time constants set to 0.1 and 2 ps, respectively. Three sets of 20 ns production


runs were performed for the solvated drug system. The LTK-drug ΔG was calculated using MP-CAFEE, which is one of the chemical free energy perturbation methods24. The ΔG for each LTK mutant


was computed according to a protocol described in a previous study30. The GROMACS 2019 and 2021 programs31 were used for the free energy simulations and preceding production runs,


respectively. XENOGRAFT EXPERIMENTS The Institutional Animal Care and Use Committee of the National Cancer Center (K20-009) approved all the animal experiments. We have complied with all


relevant ethical regulations for animal use. To establish tumor xenografts, NIH3T3 cells transduced with _CLIP1-LTK-_L650F were transplanted into the flanks of athymic nude mice (female,


8-weeks old BALB/cAJcl-_Foxn1__nu_, CLEA Japan). The mice were housed on a 12:12 light/dark cycle, and the temperature was maintained at 24 °C (23–25 °C) and humidity at 49% (40–60%). When


the mean tumor volume reached 100-200 mm3, mice were randomized into three groups and treated with lorlatinib (10 mg/kg once daily), gilteritinib (30 mg/kg once daily), or vehicle control by


oral gavage. Lorlatinib was formulated in 2% DMSO and 30% polyethylene glycol 300 in H2O. Gilteritinib was formulated using 0.5% methylcellulose in H2O. Tumor volumes (six tumors per group)


were calculated using the following formula32: 1/2(length × width2). STATISTICS AND REPRODUCIBILITY The group size was based on previous experience. Unless otherwise noted, each experiment


was repeated three or more times with similar results. One-way ANOVA and post-hoc analysis, including Dunnett’s test and Tukey’s test, were used to determine statistical significance among


more than three groups. All statistical analyses were conducted on data from three or more biologically independent experimental replicates using the GraphPad Prism software (ver. 9.3.1).


Statistical significance was set at _p_ < 0.05. REPORTING SUMMARY Further information on research design is available in the Nature Portfolio Reporting Summary linked to this article.


DATA AVAILABILITY The sequence data used in this study are publicly available in the National Center for Biotechnology Information (https://www.ncbi.nlm.nih.gov/). The protein structure data


are publicly available in RCSB Protein Data Bank (https://www.rcsb.org/). The uncropped western blotting images were exhibited in Supplementary Fig. 4. The gating strategy was exhibited in


Supplementary Fig. 5. Source data behind the graphs can be found in the Supplementary Data file. All other data are available through the corresponding author (Susumu S. Kobayashi:


[email protected]). REFERENCES * Konig, D., Savic Prince, S. & Rothschild, S. I. Targeted Therapy in Advanced and Metastatic Non-Small Cell Lung Cancer. An Update on Treatment


of the Most Important Actionable Oncogenic Driver Alterations. _Cancers_ 13, 804 (2021). Article  PubMed  PubMed Central  Google Scholar  * Izumi, H. et al. The CLIP1-LTK fusion is an


oncogenic driver in non-small-cell lung cancer. _Nature_ 600, 319–323 (2021). Article  CAS  PubMed  PubMed Central  Google Scholar  * Harada, G., Yang, S. R., Cocco, E. & Drilon, A. Rare


molecular subtypes of lung cancer. _Nat. Rev. Clin. Oncol._ 20, 229–249 (2023). Article  CAS  PubMed  PubMed Central  Google Scholar  * Yamada, S. et al. Expression of a chimeric CSF1R-LTK


mediates ligand-dependent neurite outgrowth. _Neuroreport_ 19, 1733–1738 (2008). Article  CAS  PubMed  Google Scholar  * Johnson, T. W. et al. Discovery of


(10R)-7-amino-12-fluoro-2,10,16-trimethyl-15-oxo-10,15,16,17-tetrahydro-2H-8,4-(metheno)pyrazolo[4,3-h][2,5,11]-benzoxadiazacyclotetradecine-3-carbonitrile (PF-06463922), a macrocyclic


inhibitor of anaplastic lymphoma kinase (ALK) and c-ros oncogene 1 (ROS1) with preclinical brain exposure and broad-spectrum potency against ALK-resistant mutations. _J. Med Chem._ 57,


4720–4744 (2014). Article  CAS  PubMed  Google Scholar  * Sakamoto, H. et al. CH5424802, a selective ALK inhibitor capable of blocking the resistant gatekeeper mutant. _Cancer Cell_ 19,


679–690 (2011). Article  CAS  PubMed  Google Scholar  * Yoda, S. et al. Sequential ALK inhibitors can select for Lorlatinib-resistant compound ALK mutations in ALK-positive lung. _Cancer


Discov._ 8, 714–729 (2018). Article  CAS  PubMed  PubMed Central  Google Scholar  * Recondo, G. et al. Diverse resistance mechanisms to the third-generation ALK inhibitor Lorlatinib in


ALK-rearranged lung cancer. _Clin. Cancer Res._ 26, 242–255 (2020). Article  CAS  PubMed  Google Scholar  * Okada, K. et al. Prediction of ALK mutations mediating ALK-TKIs resistance and


drug re-purposing to overcome the resistance. _EBioMedicine_ 41, 105–119 (2019). Article  PubMed  PubMed Central  Google Scholar  * Redaelli, S. et al. Lorlatinib treatment elicits multiple


on- and off-target mechanisms of resistance in ALK-driven cancer. _Cancer Res._ 78, 6866–6880 (2018). Article  CAS  PubMed  Google Scholar  * Zhu, V. W. et al. A novel sequentially evolved


EML4-ALK variant 3 G1202R/S1206Y double mutation In Cis confers resistance to Lorlatinib: A brief report and literature review. _JTO Clin. Res. Rep._ 2, 100116 (2021). PubMed  Google Scholar


  * Kobayashi, S. et al. EGFR mutation and resistance of non-small-cell lung cancer to gefitinib. _N. Engl. J. Med_. 352, 786–792 (2005). Article  CAS  PubMed  Google Scholar  * Gainor, J.


F. et al. Molecular mechanisms of resistance to first- and second-generation ALK inhibitors in ALK-rearranged lung. _Cancer Discov._ 6, 1118–1133 (2016). Article  CAS  PubMed  PubMed Central


  Google Scholar  * Engelman, J. A. et al. MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling. _Science_ 316, 1039–1043 (2007). Article  CAS  PubMed


  Google Scholar  * Isozaki, H. et al. Non-small cell lung cancer cells acquire resistance to the ALK inhibitor alectinib by activating alternative receptor Tyrosine Kinases. _Cancer Res._


76, 1506–1516 (2016). Article  CAS  PubMed  Google Scholar  * Sequist, L. V. et al. Genotypic and histological evolution of lung cancers acquiring resistance to EGFR inhibitors. _Sci.


Transl. Med._ 3, 75ra26 (2011). Article  PubMed  PubMed Central  Google Scholar  * Katayama, R. Drug resistance in anaplastic lymphoma kinase-rearranged lung cancer. _Cancer Sci._ 109,


572–580 (2018). Article  CAS  PubMed  PubMed Central  Google Scholar  * Yu, H. A. et al. Analysis of tumor specimens at the time of acquired resistance to EGFR-TKI therapy in 155 patients


with EGFR-mutant lung cancers. _Clin. Cancer Res._ 19, 2240–2247 (2013). Article  CAS  PubMed  PubMed Central  Google Scholar  * Morris, S. W. et al. ALK, the chromosome 2 gene locus altered


by the t(2;5) in non-Hodgkin’s lymphoma, encodes a novel neural receptor tyrosine kinase that is highly related to leukocyte tyrosine kinase (LTK). _Oncogene_ 14, 2175–2188 (1997). Article


  CAS  PubMed  Google Scholar  * Cocco, E., Scaltriti, M. & Drilon, A. NTRK fusion-positive cancers and TRK inhibitor therapy. _Nat. Rev. Clin. Oncol._ 15, 731–747 (2018). Article  CAS 


PubMed  PubMed Central  Google Scholar  * Awad, M. M. et al. Acquired resistance to crizotinib from a mutation in CD74-ROS1. _N. Engl. J. Med_ 368, 2395–2401 (2013). Article  CAS  PubMed 


Google Scholar  * Mizuta, H. et al. Gilteritinib overcomes lorlatinib resistance in ALK-rearranged cancer. _Nat. Commun._ 12, 1261 (2021). Article  CAS  PubMed  PubMed Central  Google


Scholar  * Mori, M. et al. Gilteritinib, a FLT3/AXL inhibitor, shows antileukemic activity in mouse models of FLT3 mutated acute myeloid leukemia. _Investig. N. Drugs_ 35, 556–565 (2017).


Article  CAS  Google Scholar  * Fujitani, H., Tanida, Y. & Matsuura, A. Massively parallel computation of absolute binding free energy with well-equilibrated states. _Phys. Rev. E Stat.


Nonlin. Soft Matter Phys._ 79, 021914 (2009). Article  PubMed  Google Scholar  * Jumper, J. et al. Highly accurate protein structure prediction with AlphaFold. _Nature_ 596, 583–589 (2021).


Article  CAS  PubMed  PubMed Central  Google Scholar  * Sali, A. & Blundell, T. L. Comparative protein modelling by satisfaction of spatial restraints. _J. Mol. Biol._ 234, 779–815


(1993). Article  CAS  PubMed  Google Scholar  * Uchibori, K. et al. Brigatinib combined with anti-EGFR antibody overcomes osimertinib resistance in EGFR-mutated non-small-cell lung cancer.


_Nat. Commun._ 8, 14768 (2017). Article  CAS  PubMed  PubMed Central  Google Scholar  * Bussi, G., Donadio, D. & Parrinello, M. Canonical sampling through velocity rescaling. _J. Chem.


Phys._ 126, 014101 (2007). Article  PubMed  Google Scholar  * Parrinello, M. & Rahman, A. Polymorphic transitions in single crystals: A new molecular dynamics method. _J. Appl. Phys._


52, 7182–7190 (1981). Article  CAS  Google Scholar  * Araki, M. et al. The effect of conformational flexibility on binding free energy estimation between kinases and their inhibitors. _J.


Chem. Inf. Model_ 56, 2445–2456 (2016). Article  CAS  PubMed  Google Scholar  * Abraham, M. J. et al. GROMACS: High performance molecular simulations through multi-level parallelism from


laptops to supercomputers. _SoftwareX_ 1-2, 19–25 (2015). Article  Google Scholar  * Tomayko, M. M. & Reynolds, C. P. Determination of subcutaneous tumor size in athymic (nude) mice.


_Cancer Chemother. Pharm._ 24, 148–154 (1989). Article  CAS  Google Scholar  Download references ACKNOWLEDGEMENTS We thank Dr. Takashi Kohno at National Cancer Research Institute for helpful


discussion. We also thank Ms. Yuri Murata and PREMIA for administrative assistance with managing clinical samples, molecular screening and the clinico-genomic database in LC-SCRUM-Asia and


the members of the Division of Translational Genomics, Exploratory Oncology Research and Clinical Trial Center, and National Cancer Center for helpful discussion for valuable comments on the


manuscript. This study was supported by MEXT/JSPS KAKENHI (JP20K17215 to H.I., JP21K06510 to M.A., 16K21746 to S.S.K., and 22H03084 to S.S.K.), JSPS Bilateral Joint Research Projects grant


number 120207408 (S.S.K.), Princess Takamatsu Cancer Research Fund 18-250 (S.S.K.), the National Cancer Center Research and Development Fund 31-A-6 (S.S.K) and National Institute of Health 


1R01CA240257 (S.S.K.). This study was also supported by MEXT as “Program for Promoting Researches on the Supercomputer Fugaku (Application of Molecular Dynamics Simulation to Precision


Medicine Using Big Data Integration System for Drug Discovery)” (Y.O.), and FOCUS Establishing Supercomputing Center of Excellence (Y.O.). This research used computational resources of the


supercomputer Fugaku provided by the RIKEN Center for Computational Science through the HPCI System Research Project (Project ID: hp210172 and hp220164). AUTHOR INFORMATION Author notes *


These authors contributed equally: Shunta Mori, Hiroki Izumi. AUTHORS AND AFFILIATIONS * Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, 277-8577, Japan


Shunta Mori, Hiroki Izumi, Yu Tanaka, Yosuke Kagawa, Yuji Shibata, Hibiki Udagawa, Shingo Matsumoto, Kiyotaka Yoh & Koichi Goto * Graduate School of Medicine, Kyoto University,


Shogoin-Kawaharacho, Sakyo-ku, Kyoto, 606-8507, Japan Mitsugu Araki, Yukari Sagae & Yasushi Okuno * Division of Translational Genomics, Exploratory Oncology Research and Clinical Trial


Center, National Cancer Center, Kashiwa, 277-8577, Japan Jie Liu, Yuta Sakae, Kosuke Tanaka & Susumu S. Kobayashi * RIKEN Center for Computational Science, Kobe, Hyogo, 650-0047, Japan


Biao Ma, Yuta Isaka & Yoko Sasakura * Division of Cancer Immunology, Research Institute/Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa,


277-8577, Japan Shogo Kumagai * Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, 277-8561, Japan Susumu S. Kobayashi * Department


of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA Susumu S. Kobayashi Authors * Shunta Mori View author publications You can also search for


this author inPubMed Google Scholar * Hiroki Izumi View author publications You can also search for this author inPubMed Google Scholar * Mitsugu Araki View author publications You can also


search for this author inPubMed Google Scholar * Jie Liu View author publications You can also search for this author inPubMed Google Scholar * Yu Tanaka View author publications You can


also search for this author inPubMed Google Scholar * Yosuke Kagawa View author publications You can also search for this author inPubMed Google Scholar * Yukari Sagae View author


publications You can also search for this author inPubMed Google Scholar * Biao Ma View author publications You can also search for this author inPubMed Google Scholar * Yuta Isaka View


author publications You can also search for this author inPubMed Google Scholar * Yoko Sasakura View author publications You can also search for this author inPubMed Google Scholar * Shogo


Kumagai View author publications You can also search for this author inPubMed Google Scholar * Yuta Sakae View author publications You can also search for this author inPubMed Google Scholar


* Kosuke Tanaka View author publications You can also search for this author inPubMed Google Scholar * Yuji Shibata View author publications You can also search for this author inPubMed 


Google Scholar * Hibiki Udagawa View author publications You can also search for this author inPubMed Google Scholar * Shingo Matsumoto View author publications You can also search for this


author inPubMed Google Scholar * Kiyotaka Yoh View author publications You can also search for this author inPubMed Google Scholar * Yasushi Okuno View author publications You can also


search for this author inPubMed Google Scholar * Koichi Goto View author publications You can also search for this author inPubMed Google Scholar * Susumu S. Kobayashi View author


publications You can also search for this author inPubMed Google Scholar CONTRIBUTIONS S.Mo., H.I., S.Ma., and S.S.K conceived the study and designed the experiments. M.A. and Y.O. designed


and supervised the simulation. Y.I. modeled the LTK structure, B.M. and Y.Sas. performed molecular docking, and Y.Sas. performed molecular dynamics simulation (MP-CAFEE). S.Mo., H.I., and


J.L. performed cloning and mutagenesis of the expression constructs for in vitro analysis. S.M, J.L., Y.K. and S.K. generated stable cell lines. S.Mo., H.I., J.L., Y.K., Y.Sak., K.T., S.Y.,


Y.T. H.U. and S.S.K. performed biochemical analysis. K.Y., K.G. and S.S.K. supervised this project. S.Mo., H.I., S.Ma., Y.O. and S.S.K wrote the manuscript with input from all the authors.


CORRESPONDING AUTHOR Correspondence to Susumu S. Kobayashi. ETHICS DECLARATIONS COMPETING INTERESTS The authors declare the following competing interests. S.Mo. reports no conflicts of


interest in this study. Y.T. reports personal fees (honoraria) from Chugai, Eli Lilly, AstraZeneca, Taiho. H.I. reports research support from Amgen, Ono, Takeda, Eisai and personal fees


(honoraria) from Ono, Chugai, AstraZeneca, Merck. Y.S. reports research support from Ono, MSD, and personal fees (honoraria) from Ono, Chugai, AstraZeneca, Eli Lilly, Bristol-Myers Squibb,


Pfizer. H.U. reports research support from Takeda, Boehringer Ingelheim, Taiho and personal fees (honoraria) from Taiho. S.Ma. reports research support from Chugai, Novartis, Eli Lilly,


Merck, MSD, and personal fees (honoraria) from AstraZeneca, Chugai, Novartis, Pfizer and Eli Lilly. K.Y. reports research support from AstraZeneca, Eli Lilly, Phizer, Diichi sankyo, Abbvie,


Taiho, MSD, Takeda, Chugai, and personal fees (honoraria) from Chugai, AstraZeneca, Bristol-Myers Squibb, Daiichi sankyo, Janssen, Eli Lilly, Taiho, Novaritis, Kyowa kirin, Boehringer


Ingelheim. G.K. reports research support from Amgen, Amgen Astellas BioPharma, AstraZeneca, Bayer, Boehringer Ingelheim Japan, Bristol-Myers Squibb, Blueprint Medicines, Chugai, Daiichi


sankyo, Eisai, Eli Lilly, Haihe Biopharma, Ignyta, Janssen, KISSEI, Kyowa Kirin, Life Technologies, Loxo Oncology., Medical & Biological Laboratories, Merck, Merus, MSD, NEC Corporation,


Novartis, Ono, Pfizer, Sumitomo Dainippon, Spectrum Pharmaceuticals, Sysmex Corporation, Taiho, Takeda, Turning Point Therapeutics, and personal fees (honoraria) from Amgen, Amoy


Diagnosties, Amgen Astellas BioPharma, AstraZeneca, Bayer, Boehringer Ingelheim, Bristol-Myers Squibb, Chugai, Daiichi sankyo, Eisai, Eli Lilly Japan, Guardant Health, Janssen, Thermo Fisher


Scientifi, Medpace, Merck, MSD, Novartis Pharma, Ono, Otsuka, Taiho, and Takeda. SSK reports grants from Boehringer Ingelheim, MiRXES, Johnson&Johnson, and Taiho Therapeutics, as well


as personal fees from AstraZeneca, Boehringer Ingelheim, Bristol Meyers Squibb, Chugai Pharmaceutical, and Takeda Pharmaceuticals plus royalties from Life Technologies. Other authors declare


no conflicts of interest. PEER REVIEW PEER REVIEW INFORMATION _Communications Biology_ thanks the anonymous reviewers for their contribution to the peer review of this work. Primary


Handling Editors: Toril Holien and Christina Karlsson Rosenthal. ADDITIONAL INFORMATION PUBLISHER’S NOTE Springer Nature remains neutral with regard to jurisdictional claims in published


maps and institutional affiliations. SUPPLEMENTARY INFORMATION SUPPLEMENTARY INFORMATION DESCRIPTION OF ADDITIONAL SUPPLEMENTARY FILES SUPPLEMENTARY DATA 1 REPORTING SUMMARY RIGHTS AND


PERMISSIONS OPEN ACCESS This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any


medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The


images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not


included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly


from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. Reprints and permissions ABOUT THIS ARTICLE CITE THIS ARTICLE Mori, S., Izumi,


H., Araki, M. _et al._ _LTK_ mutations responsible for resistance to lorlatinib in non-small cell lung cancer harboring _CLIP1-LTK_ fusion. _Commun Biol_ 7, 412 (2024).


https://doi.org/10.1038/s42003-024-06116-6 Download citation * Received: 08 June 2023 * Accepted: 27 March 2024 * Published: 04 April 2024 * DOI: https://doi.org/10.1038/s42003-024-06116-6


SHARE THIS ARTICLE Anyone you share the following link with will be able to read this content: Get shareable link Sorry, a shareable link is not currently available for this article. Copy to


clipboard Provided by the Springer Nature SharedIt content-sharing initiative