Response to EGFR/NTRK/MET Co-Inhibition Guided by Paired NGS in Advanced NSCLC With Acquired EGFR L858R/T790M/C797S Mutations

Authors:
Xue Yang Division of Thoracic Tumor Multimodality Treatment, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China

Search for other papers by Xue Yang in
Current site
Google Scholar
PubMed
Close
 MD, PhD
,
Xintong Li Division of Thoracic Tumor Multimodality Treatment, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China

Search for other papers by Xintong Li in
Current site
Google Scholar
PubMed
Close
 MD, PhD
,
Jiaqi Yan Department of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan, China

Search for other papers by Jiaqi Yan in
Current site
Google Scholar
PubMed
Close
 MD, PhD
,
Yuanxin Liu Division of Thoracic Tumor Multimodality Treatment, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China

Search for other papers by Yuanxin Liu in
Current site
Google Scholar
PubMed
Close
 MD
,
Jie Yin School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China

Search for other papers by Jie Yin in
Current site
Google Scholar
PubMed
Close
 MD
,
Weikang Shao Genecast Biotechnology Co., Ltd., Wuxi, Jiangsu, China

Search for other papers by Weikang Shao in
Current site
Google Scholar
PubMed
Close
 MD
,
You Lu Division of Thoracic Tumor Multimodality Treatment, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
Laboratory of Clinical Cell Therapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China

Search for other papers by You Lu in
Current site
Google Scholar
PubMed
Close
 MD, PhD
, and
Jianxin Xue Division of Thoracic Tumor Multimodality Treatment, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
Laboratory of Clinical Cell Therapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China

Search for other papers by Jianxin Xue in
Current site
Google Scholar
PubMed
Close
 MD, PhD
Full access

EGFR tyrosine kinase inhibitors (TKIs) have significantly improved clinical outcomes for patients with non–small cell lung cancer (NSCLC) harboring EGFR-activating mutations. However, resistance to TKI therapy often develops due to secondary EGFR mutations or the activation of bypass signalling pathways. Next-generation sequencing (NGS) can efficiently identify actionable genetic alterations throughout treatment. MET amplification is a well-established off-target resistance mechanism. Additionally, rarer mechanisms, such as NTRK1 gene fusions, have been reported. This report highlights a case of a 58-year-old male diagnosed with bone-metastatic NSCLC harboring the EGFR L858R mutation. After receiving dacomitinib and almonertinib sequentially, plasma-based NGS revealed the emergence of EGFR T790M-trans-C797S mutations, prompting a switch to a combination therapy of almonertinib and gefitinib. Upon disease progression, repeat NGS identified EGFR T790M-cis&trans-C797S mutations and a novel POT1::NTRK3 fusion in the blood. The fusion retained a complete NTRK kinase domain without frameshift variants, making it a target for treatment. Larotrectinib was incorporated into the dual EGFR-TKI regimen, forming a triplet therapy. Although this resulted in grade 3 dermatitis, the condition resolved after discontinuing gefitinib. At multiorgan progression, matched tissue- and plasma-based NGS identified MET amplification. Subsequently, the patient was started on a triple-inhibition regimen targeting EGFR, NTRK, and MET, which achieved a partial response with favorable tolerability. This is the first reported case of a novel, targetable POT1::NTRK3 fusion as a potential off-target mechanism mediating EGFR-TKI resistance, occurring alongside MET amplification in a patient with NSCLC harboring acquired EGFR L858R/T790M/C797S mutations. Concomitant inhibition of EGFR, NTRK, and MET was safe and resulted in a significant response, underscoring the importance of precision medicine guided by matched NGS.

Background

EGFR-activating mutations are pivotal therapeutic targets in non–small cell lung cancer (NSCLC).1 EGFR-targeting tyrosine kinase inhibitors (TKIs) have significantly improved patient outcomes.1 However, resistance to these therapies can develop through various on- and off-target mechanisms.2,3 Next-generation sequencing (NGS) enables high-throughput genomic profiling, facilitating the identification of actionable genetic alterations. EGFR T790M and C797S are common EGFR secondary mutations. EGFR T790M is the primary cause of resistance to first- and second-generation EGFR-TKIs, which can often be effectively addressed with third-generation TKIs.4 In contrast, EGFR C797S, either in cis (on the same allele as T790M) or in trans (on a different allele), is prevalent in patients who progress on third-generation TKIs, posing additional challenges for treatment.5,6

Activation of bypass signaling pathways is another significant cause of resistance to EGFR-TKIs.7,8 MET amplification has long been recognized as a key acquired resistance mechanism that can lead to the failure of EGFR-targeted therapies.9 Studies such as SAVANNAH, TATTON, and INSIGHT 2 have demonstrated the efficacy of co-inhibiting EGFR and MET pathways in overcoming resistance. This strategy is recommended for patients with MET amplification whose disease progressed on EGFR-TKIs.1012 Several EGFR-MET bispecific antibodies have shown promise in addressing MET-driven resistance in preclinical and clinical studies. Among these, amivantamab has recently been approved for NSCLC based on results of the phase III PAPILLON study.13 NTRK gene fusions are rare oncogenic drivers in solid tumors, with a reported prevalence of only 0.2% to 3.3% in NSCLC.14 First-generation tropomyosin receptor kinase (TRK) TKIs, such as larotrectinib and entrectinib, have proven effective in treating solid tumors with NTRK fusions and have received FDA approval for tumor-agnostic use.15,16 Next-generation TRK inhibitors have demonstrated effectiveness in inhibiting TRK receptor signaling and overcoming resistance to first-generation TRK inhibitors. Among these, repotrectinib has received FDA approval for NTRK fusion–positive solid tumors.17 Recent studies provide early insights into the potential correlation between NTRK fusions and lack of response to EGFR-TKIs.18,19 The diverse mechanisms complicate the treatment landscape for patients with NSCLC that fails to respond to EGFR-TKIs, underscoring the need for standard clinical management of EGFR-TKI resistance involving multiple pathways.7,8

This report presents a challenging case of bone-metastatic NSCLC featuring the concurrence of a novel POT1::NTRK3 fusion and MET amplification, which rendered EGFR-TKIs ineffective. To our knowledge, this is the first reported case of a patient with advanced NSCLC and acquired EGFR-TKI resistance achieving a response to EGFR/NTRK/MET triple inhibition. This case highlights the value of NGS-guided precision treatment in tailoring effective treatment strategies for complex resistance mechanisms.

Case Description

In May 2020, a 58-year-old Asian male with no history of smoking or familial cancer presented with left hip pain lasting 1 year and a cough for 2 months. The severity of localized hip pain was assessed using the Numeric Rating Scale (NRS; scale 0–10), a simple and commonly used tool for evaluating pain intensity, with an initial rating of 3.20 Serum CEA and cytokeratin 19 fragment 21-1 (CYFRA21-1) levels were significantly elevated (see Supplementary Figure S1, available online in the supplementary material). Enhanced CT scans revealed a mass in the lower lobe of right lung causing atelectasis, enlarged mediastinal lymph nodes, and bony destruction in the T5 and T11 vertebrae, left ilium, and right acetabulum. A biopsy of the left ilium showed immunochemical positivity for thyroid transcription factor 1 (TTF-1), Napsin A, and cytokeratin 7 (CK7), indicating metastatic adenocarcinoma (Figure 1). The patient was diagnosed with stage IV right lung adenocarcinoma (cT2N3M1c) and had an ECOG performance status of 2. At the time of diagnosis, the patient also had deep vein thrombosis and pulmonary embolism. Two additional pulmonary embolism events were confirmed in May 2022 and April 2023, each treated appropriately. Given multiple occurrences, the patient has been taking rivaroxaban for prevention as of the time of writing. The patient underwent tissue and liquid biopsies analyzed with NGS targeting 769 cancer-related genes (Genecast Biotechnology Co., Ltd.). Before NGS results were available, the patient received 1 cycle of pemetrexed (500 mg/m2, intravenously guttae, day 1, every 3 weeks) and cisplatin (25 mg/m2, intravenously guttae, days 1–3, every 3 weeks). NGS identified an EGFR L858R mutation, with variant allele frequencies (VAFs) of 44.93% in tissue and 13.32% in plasma. In July 2020, the patient began treatment with dacomitinib (30 mg, orally, daily), which was later reduced to 15 mg due to grade 2 paronychia. Denosumab and zoledronic acid were administered to mitigate the risk of skeletal-related events. The patient reported symptom relief and achieved partial response (PR). Follow-up CT scans showed significant reduction of the primary lung lesion and osteoblastic transformation of the osteolytic lesion in the left ilium (Figure 2).

Figure 1.
Figure 1.

Histopathologic findings of the left ilium biopsy. (A) Hematoxylin-eosin staining, and immunochemical stains of (B) Napsin A, (C) thyroid transcription factor 1 (TTF-1), and (D) cytokeratin 7 (CK7) (original magnification ×200 for all). All consistent with metastatic adenocarcinoma.

Citation: Journal of the National Comprehensive Cancer Network 2025; 10.6004/jnccn.2024.7070

Figure 2.
Figure 2.

Clinical treatment history and serial imaging data of the patient. Timeline of events, including systemic and localized treatments and disease progression, is shown at the top. Cross-sectional CT images at the bottom indicate the development, progression, and shrinkage of representative lesions in lung and pelvis during the treatment course.

Abbreviations: OR, overall response; PFS, progression-free survival; PR, partial response; SD, stable disease.

Citation: Journal of the National Comprehensive Cancer Network 2025; 10.6004/jnccn.2024.7070

In October 2021, the patient reported worsening left hip pain. CT scans revealed enlarged extraosseous soft tissue around the left ilium (Figure 2). Palliative radiotherapy (30 Gy/10 fractions) was administered, resulting in slight pain improvement. However, follow-up CT in March 2022 showed further growth of the left iliac lesion and new metastases to sacrum and right ilium, indicating a progression-free survival (PFS) of 20.2 months with dacomitinib. To address the progression, CT-guided 125I implantation was performed in the left pelvis. Despite this, the pain persisted. Plasma-based NGS detected the emergence of EGFR T790M (VAF 19.11%). Treatment was switched to third-generation EGFR-TKI almonertinib (110 mg, orally, daily), and CT re-evaluation suggested stable disease (SD).

In October 2022, the patient experienced new soreness in the right hip. CT scans revealed gradual progression of pelvic metastases. Another round of radiotherapy (30 Gy/10 fractions) was administered, but symptoms did not improve. Plasma-based NGS detected EGFR T790M-trans-C797S (VAF 0.83%; PFS, 6.4 months). Gefitinib (250 mg, orally, daily) was added to the regimen, and the overall response was subsequently evaluated as SD once again.

By May 2023, the patient reported increased hip pain (NRS score 4). CT revealed a new osteolytic lesion in the right ilium (Figure 2; PFS, 4.4 months). Plasma-based NGS identified EGFR T790M-cis&trans-C797S mutations (c.2389T>A VAF 0.35%, c.2390G>C VAF 0.6%) and a POT1::NTRK3 fusion (VAF, 0.54%) (Figure 3). In June 2023, larotrectinib (100 mg, orally, twice daily) was added to the previous regimen. However, the patient developed grade 3 dermatitis, leading to a reduction of gefitinib to twice weekly and eventual discontinuation after 1 month. Thereafter, the dermatitis gradually resolved to grade 1, the NRS score decreased from 4 to 2, and therapeutic efficacy was evaluated as SD.

Figure 3.
Figure 3.

Dynamic changes of the variant allele frequencies of mutations over 6 time points. Fish plots at top and bottom show the evolving frequencies of gene alterations of interest in blood and tissue, respectively. To enhance visualization, we applied logarithmic transformation to the actual frequencies. The middle panel illustrates the relative frequencies by the sizes of circles, and the numbers within circles represent the actual frequencies detected by NGS. Filled circles and outlined circles indicate results from plasma- and tissue-based NGS analysis, respectively.

Abbreviations: delins, deletion-insertion; NGS, next-generation sequencing.

*Copy number of MET amplification.

Citation: Journal of the National Comprehensive Cancer Network 2025; 10.6004/jnccn.2024.7070

In July 2023, the patient reported persistent hip pain and underwent bilateral internal iliac artery chemoembolization with gemcitabine (60 mg). Despite this, he experienced pain exacerbation (NRS score 6) and left hip swelling. In September 2023, CT scans revealed new metastases near the left ilium and in the left hilum and left erector spinae muscle (Figure 2), indicating multiorgan progression (PFS, 3.2 months). NGS analysis of the nodule near the left ilium and paired plasma revealed the emergence of MET amplification (copy number 4.21 in tissue, 3.83 in plasma), and the POT1::NTRK3 fusion was again detected in blood (VAF, 0.59%). Thus, the patient was started on a combination therapy of osimertinib (80 mg, orally, daily), larotrectinib (100 mg, orally, twice daily), and capmatinib (400 mg, orally, twice daily). Despite grade 2 paronychia, immediate symptom relief was observed within 2 days, with dramatic shrinkage of new measurable metastases, evaluated as a PR (Figure 2). Plasma-based NGS performed after treatment initiation showed that both MET amplification and the NTRK3 fusion had become undetectable, and the frequencies of EGFR L858R and T790M had also decreased significantly. Additionally, a new MET insertion-deletion mutation (p.D1228_Y1230delinsHLN) (VAF, 0.79%) was identified. At the last follow-up in January 2024, after being monitored for 44 months, the patient continued to demonstrate a PR.

Discussion

EGFR C797S, which can occur either in cis or trans with EGFR T790M, is an important on-target resistance mechanism to third-generation EGFR-TKIs. EGFR T790M-trans-C797S is sensitive to combined first- and third-generation EGFR-TKIs, whereas patients harboring EGFR T790M-cis-C797S may benefit from combination therapy with brigatinib and cetuximab.2125 In our case, we first detected the trans configuration and started combined treatment with gefitinib and almonertinib. Notably, the allelic configuration of EGFR T790M/C797S can shift during treatment, which is associated with the development of drug resistance.23,26 After dual EGFR-TKI therapy, we detected a conversion from EGFR T790M-trans-C797S to T790M-cis&trans-C797S (Figure 3). The cis&trans configuration accounts for approximately 10% of C797S-mutated cases and correlates with the worst prognosis compared with other subtypes.26 Currently, there are no effective therapies for this patient subgroup.27,28 In our case, an NTRK fusion was identified concomitantly. Given the patient’s tolerability, only larotrectinib was added to the previous treatment regimen. However, severe adverse events occurred during the combined regimen. Therefore, the safety profile of dual EGFR-TKIs in combination with other targeted agents warrants attention.

The novel POT1::NTRK3 fusion we detected has not been previously documented in the literature. Both DNA- and RNA-based NGS offer the advantage of identifying the fusion partner with high specificity and are recommended for detecting NTRK rearrangements.29 In our case, despite the low frequency of the fusion, DNA-based NGS successfully identified the POT1::NTRK3 fusion event (Figure 4A). To further confirm expression, we performed a tissue-based RNA sequencing; however, it did not detect the fusion, which we propose could be due to the spatiotemporal heterogeneity of tumor. Recently, based on acquired NTRK1 fusions detected after patients progressed on EGFR-TKIs, preliminary evidence suggests that NTRK1 fusions can emerge as acquired resistance mechanisms to both first- and third-generation EGFR-TKIs, even at a low frequencies.18,19,30 In patients with EGFR-mutated NSCLC harboring NTRK1 fusions, 4 of 7 NTRK1 fusion genes were detected with a frequency of ≤1%.19 However, current reports on NTRK3-related EGFR-TKI resistance are scarce. In our case, the NTRK3 fusion was detected after progression on combined first- and third-generation EGFR-TKIs, indicating its potential role in EGFR-TKI resistance. Although the underlying mechanism remains unclear, dysregulation of receptor tyrosine kinases may compensate for the inhibited EGFR signaling through the abnormal activation of convergent downstream signaling pathways, such as those mediated by MET amplification.31 Similarly, given that NTRK signaling and EGFR signaling converge on the PI3K/Akt and MAPK pathways, NTRK fusions may induce EGFR-TKI resistance via compensatory activation of downstream molecules.3234 Despite the overall low frequency of NTRK fusions in patients with lung cancer, a substantial proportion of detected fusions co-occur with primary and secondary EGFR mutations.19,35 In this clinical context, co-targeting EGFR and the acquired fusion may offer clinical benefits.30,36 The novel fusion we detected contains the full-length tyrosine kinase domain of NTRK3 and conforms to an in-framestructure (Figure 4B,C), suggesting its sensitivity to first-generation TRK-TKIs. Larotrectinib induces a response in 75% of patients in a pan-cancer cohort.15 However, its efficacy remains uncertain in patients with an EGFR mutation, particularly those harboring secondary EGFR mutations and resistant to EGFR-TKIs. In our case, although the addition of larotrectinib resulted in pain relief and improved mobility, it showed suboptimal efficacy in reducing tumor burden. We speculate that this could be due to progression driven by untargeted activated pathways, such as EGFR T790M-cis&trans-C797S, or other undetected genetic alterations. However, the possibility that the NTRK3 fusion did not drive the major resistance events cannot be ruled out.

Figure 4.
Figure 4.

Illustration of the POT1::NTRK3 fusion event. (A) Identification of POT1::NTRK3 fusion in plasma-based next-generation sequencing analysis. (B) Circos plot showing interchromosomal translocations between POT1 and NTRK3 represented by a colored line linking 2 genes. (C) Schematic diagram depicting the in-frame POT1::NTRK3 (P7;N15) fusion event. The fusion contains the full-length kinase domain of NTRK3 (exons 15–19).

Citation: Journal of the National Comprehensive Cancer Network 2025; 10.6004/jnccn.2024.7070

After the multiorgan progression, MET amplification was identified as a crucial contributing factor through matched NGS. Because continued inhibition of EGFR and NTRK was required, the patient was treated with concomitant EGFR/NTRK/MET inhibition. This triplet therapy induced a robust response and rapid symptom relief. Subsequent NGS revealed >80% reductions in EGFR L858R and T790M mutations and undetectable levels of NTRK3 fusion and MET amplification (Figure 3). As previously noted, genetic alterations of EGFR, MET, and NTRK promote cancer initiation and progression by engaging multiple common downstream signaling pathways.32,34,37 MET amplification can mediate resistance to EGFR-TKIs and TRK inhibitors via convergent activation of the PI3K/Akt and MAPK pathways, respectively.9,38 Thus, it is possible that the triplet therapy synergistically suppressed these downstream molecules, contributing to its remarkable efficacy. Overall, this case highlights the efficacy and safety of concurrent multipathway inhibition in heavily treated patients with TKI-resistant NSCLC. Further research is needed to explore the interaction between pathways and its impact on combination therapy. Notably, an atypical insertion-deletion at MET D1228_Y1230 was detected in the latest plasma-based NGS analysis. MET D1228 and Y1230 point mutations are common resistance mechanisms to type I MET inhibitors while sensitive to type II MET inhibitors.39,40 The clinical significance of this insertion-deletion variant deserves increased focus.

Bone metastases are detected in >60% of patients with NSCLC harboring EGFR L858R mutations.41 In this case, the patient experienced multiple metastases to major weight-bearing bones, with gradual progression causing worsening pain and elevated tumor markers. The efficacy of localized therapy is limited. Currently, systemic targeted therapy remains the primary treatment for bone metastases. In the detection of treatable targets, tissue-based NGS demonstrates higher sensitivity compared with plasma-based NGS. As a complementary approach, liquid biopsy better captures tumor heterogeneity and offers the advantages of being minimally invasive and real-time.4244 However, concordance rates between liquid and tissue biopsies for molecular genotyping vary across studies and mutation types.7,45,46 Therefore, matched NGS may provide a more comprehensive characterization of the cancer genome, facilitating the identification of targetable genetic alterations. In our case, emerging MET amplification was identified by matched NGS, suggesting its critical role in disease progression. Plasma-based NGS expanded the range of potentially actionable targets and revealed the presence of EGFR C797S and NTRK3 fusion (Figure 5), which may exist in subclonal-resistant populations. Overall, in the framework of precision medicine, matched NGS represents an informative strategy to aid optimal clinical decision-making.

Figure 5.
Figure 5.

Mutation profiles over 6 time points. Matched plasma- and tissue-based next-generation sequencing revealed the complex and evolving mutational landscape of the patient during treatment. Paired results of tissue and plasma are shown at timepoint 1 and timepoint 5. Numbers within the squares represent frequencies of the mutation.

*Copy number.

Citation: Journal of the National Comprehensive Cancer Network 2025; 10.6004/jnccn.2024.7070

Conclusions

This case reports a novel, targetable POT1::NTRK3 fusion in a patient with NSCLC and acquired EGFR L858R/T790M/C797S mutations, which may serve as a resistance mechanism to EGFR-TKIs. It also highlights the rare co-occurrence of the POT1::NTRK3 fusion and MET amplification, which may contribute to disease progression. The triple inhibition of EGFR, NTRK, and MET resulted in a dramatic therapeutic response and was well tolerated. This case underscores the importance of real-time, matched NGS in complex clinical scenarios. Further studies are expected to explore the role of NTRK fusions in EGFR-TKI resistance and to determine the optimal treatment combinations and follow-up strategies for patients with EGFR-mutated NSCLC who develop resistance in a stepwise manner.

Acknowledgments

The authors thank the patient and his family. They also extend thanks to Genecast Biotechnology Co., Ltd. (Wuxi), which participated in gene sequencing and data bioinformatic analysis.

References

  • 1.

    Sharma SV, Bell DW, Settleman J, et al. Epidermal growth factor receptor mutations in lung cancer. Nat Rev Cancer 2007;7:169181.

  • 2.

    Westover D, Zugazagoitia J, Cho BC, et al. Mechanisms of acquired resistance to first- and second-generation EGFR tyrosine kinase inhibitors. Ann Oncol 2018;29:I1019.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 3.

    Cooper AJ, Sequist L, Lin JJ. Third-generation EGFR and ALK inhibitors: mechanisms of resistance and management. Nat Rev Clin Oncol 2022;19:499514.

  • 4.

    Chakravarty D, Solit DB. Clinical cancer genomic profiling. Nat Rev Genet 2021;22:483501.

  • 5.

    Jänne PA, Yang JCH, Kim DW, et al. AZD9291 in EGFR inhibitor-resistant non-small-cell lung cancer. N Engl J Med 2015;372:16891699.

  • 6.

    Camidge DR, Pao W, Sequist LV. Acquired resistance to TKIs in solid tumours: learning from lung cancer. Nat Rev Clin Oncol 2014;11:473481.

  • 7.

    Leonetti A, Verze M, Minari R, et al. Resistance to osimertinib in advanced EGFR-mutated NSCLC: a prospective study of molecular genotyping on tissue and liquid biopsies. Br J Cancer 2024;130:135142.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 8.

    Rotow JK, Lee JK, Madison RW, et al. Real-world genomic profile of EGFR second-site mutations and other osimertinib resistance mechanisms and clinical landscape of NSCLC post-osimertinib. J Thorac Oncol 2024;19:227239.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 9.

    Zhang Z, Yang S, Wang QM. Impact of MET alterations on targeted therapy with EGFR-tyrosine kinase inhibitors for EGFR-mutant lung cancer. Biomark Res 2019;7:27.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 10.

    Ahn MJ, De Marinis F, Bonanno L, et al. MET biomarker-based preliminary efficacy analysis in SAVANNAH: savolitinib plus osimertinib in EGFRm NSCLC post-osimertinib. J Thorac Oncol 2022;17:S469470.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 11.

    Hartmaier RJ, Markovets AA, Ahn MJ, et al. Osimertinib plus savolitinib to overcome acquired MET-mediated resistance in epidermal growth factor receptor-mutated, MET-amplified non-small cell lung cancer: TATTON. Cancer Discov 2023;13:98113.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 12.

    Kim TM, Guarneri V, Jye VP, et al. Tepotinib plus osimertinib in EGFR-mutant NSCLC with MET amplification following 1L osimertinib: INSIGHT 2 primary analysis. J Thorac Oncol 2023;18:S94.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 13.

    Zhou CC, Tang KJ, Cho BC, et al. Amivantamab plus chemotherapy in NSCLC with EGFR exon 20 insertions. N Engl J Med 2023;389:20392051.

  • 14.

    Liu FF, Wei YX, Zhang H, et al. NTRK fusion in non-small cell lung cancer: diagnosis, therapy, and TRK inhibitor resistance. Front Oncol 2022;12:864666.

  • 15.

    Hong DS, DuBois SG, Kummar S, et al. Larotrectinib in patients with TRK fusion-positive solid tumours: a pooled analysis of three phase 1/2 clinical trials. Lancet Oncol 2020;21:531540.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 16.

    Doebele RC, Drilon A, Paz-Ares L, et al. Entrectinib in patients with advanced or metastatic NTRK fusion-positive solid tumours: integrated analysis of three phase 1-2 trials. Lancet Oncol 2020;21:271282.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 17.

    Drilon A, Camidge DR, Lin JJ, et al. Repotrectinib in ROS1 fusion-positive non-small-cell lung cancer. N Engl J Med 2024;390:118131.

  • 18.

    Rolfo C, Drilon A, Hong D, et al. NTRK1 fusions identified by non-invasive plasma next-generation sequencing (NGS) across 9 cancer types. Br J Cancer 2022;126:514520.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 19.

    Xia H, Xue XY, Ding HX, et al. Evidence of NTRK1 fusion as resistance mechanism to EGFR TKI in EGFR+ NSCLC: results from a large-scale survey of NTRK1 fusions in Chinese patients with lung cancer. Clin Lung Cancer 2020;21:247254.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 20.

    Euasobhon P, Atisook R, Bumrungchatudom K, et al. Reliability and responsivity of pain intensity scales in individuals with chronic pain. Pain 2022;163:E11841191.

  • 21.

    Arulananda S, Do H, Musafer A, et al. Combination osimertinib and gefitinib in C797S and T790M EGFR-mutated non-small cell lung cancer. J Thorac Oncol 2017;12:17281732.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 22.

    Zhou Z, Zhao Y, Shen SP, et al. Durable clinical response of lung adenocarcinoma harboring EGFR 19Del/T790M/in trans-C797S to combination therapy of first- and third-generation EGFR tyrosine kinase inhibitors. J Thorac Oncol 2019;14:E157159.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 23.

    Wang Z, Yang JJ, Huang J, et al. Lung adenocarcinoma harboring EGFR T790M and in trans C797S responds to combination therapy of first- and third-generation EGFR TKIs and shifts allelic configuration at resistance. J Thorac Oncol 2017;12:17231727.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 24.

    Yang YN, Xu HY, Ma L, et al. Possibility of brigatinib-based therapy, or chemotherapy plus anti-angiogenic treatment after resistance of osimertinib harboring EGFR T790M-cis-C797S mutations in lung adenocarcinoma patients. Cancer Med 2021;10:83288337.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 25.

    Wang YB, Yang N, Zhang YC, et al. Effective treatment of lung adenocarcinoma harboring EGFR-activating mutation, T790M, and cis-C797S triple mutations by brigatinib and cetuximab combination therapy. J Thorac Oncol 2020;15:13691375.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 26.

    Lu C, Wei XW, Wang Z, et al. Allelic context of EGFR C797X-mutant lung cancer defines four subtypes with heterogeneous genomic landscape and distinct clinical outcomes. J Thorac Oncol 2024;19:601612.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 27.

    Liu CL, Li JD, Liu HL, et al. The concomitant EGFR T790M/C797S in trans and cis in three osimertinib-resistant lung adenocarcinoma patients. J Clin Oncol 2019;37(Suppl):Abstract e20105.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 28.

    Zhou RG, Song L, Zhang WW, et al. Combination of osimertinib and anlotinib may overcome the resistance mediated by in cis EGFR T790M-C797S in NSCLC: a case report. Onco Targets Ther 2021;14:28472851.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 29.

    Marchiò C, Scaltriti M, Ladanyi M, et al. ESMO recommendations on the standard methods to detect NTRK fusions in daily practice and clinical research. Ann Oncol 2019;30:14171427.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 30.

    Schrock AB, Zhu VW, Hsieh WS, et al. Receptor tyrosine kinase fusions and BRAF kinase fusions are rare but actionable resistance mechanisms to EGFR tyrosine kinase inhibitors. J Thorac Oncol 2018;13:13121323.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 31.

    Liu Q, Yu SN, Zhao WH, et al. EGFR-TKIs resistance via EGFR-independent signaling pathways. Mol Cancer 2018;17:53.

  • 32.

    Amatu A, Sartore-Bianchi A, Bencardino K, et al. Tropomyosin receptor kinase (TRK) biology and the role of NTRK gene fusions in cancer. Ann Oncol 2019;30(Suppl 8):viii515.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 33.

    Cocco E, Scaltriti M, Drilon A. NTRK fusion-positive cancers and TRK inhibitor therapy. Nat Rev Clin Oncol 2018;15:731747.

  • 34.

    Levantini E, Maroni G, Del Re M, et al. EGFR signaling pathway as therapeutic target in human cancers. Semin Cancer Biol 2022;85:253275.

  • 35.

    Farago AF, Taylor MS, Doebele RC, et al. Clinicopathologic features of non-small-cell lung cancer harboring an NTRK gene fusion. JCO Precis Oncol 2018;2018:PO.18.00037.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 36.

    Piotrowska Z, Isozaki H, Lennerz JK, et al. Landscape of acquired resistance to osimertinib in EGFR-mutant NSCLC and clinical validation of combined EGFR and RET inhibition with osimertinib and BLU-667 for acquired RET fusion. Cancer Discov 2018;8:15291539.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 37.

    Zhang YZ, Xia MF, Jin K, et al. Function of the c-Met receptor tyrosine kinase in carcinogenesis and associated therapeutic opportunities. Mol Cancer 2018;17:45.

  • 38.

    Cocco E, Schram AM, Kulick A, et al. Resistance to TRK inhibition mediated by convergent MAPK pathway activation. Nat Med 2019;25:14221427.

  • 39.

    Tsai JM, Hata AN, Lennerz JK. MET D1228N and D1246N are the same resistance mutation in MET exon 14 skipping. Oncologist 2021;26:E22972301.

  • 40.

    Bahcall M, Sim T, Paweletz CP, et al. Acquired METD1228V mutation and resistance to MET inhibition in lung cancer. Cancer Discov 2016;6:13341341.

  • 41.

    Kuijpers CCHJ, Hendriks LEL, Derks JL, et al. Association of molecular status and metastatic organs at diagnosis in patients with stage IV non-squamous non-small cell lung cancer. Lung Cancer 2018;121:7681.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 42.

    Huang HM, Li HX. Tumor heterogeneity and the potential role of liquid biopsy in bladder cancer. Cancer Commun (Lond) 2021;41:91108.

  • 43.

    Russano M, Napolitano A, Ribelli G, et al. Liquid biopsy and tumor heterogeneity in metastatic solid tumors: the potentiality of blood samples. J Exp Clin Canc Res 2020;39:95.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 44.

    Pascual J, Attard G, Bidard FC, et al. ESMO recommendations on the use of circulating tumour DNA assays for patients with cancer: a report from the ESMO Precision Medicine Working Group. Ann Oncol 2022;33:750768.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 45.

    Sacher AG, Paweletz C, Dahlberg SE, et al. Prospective validation of rapid plasma genotyping for the detection of EGFR and KRAS mutations in advanced lung cancer. JAMA Oncol 2016;2:10141022.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 46.

    Odegaard JI, Vincent JJ, Mortimer S, et al. Validation of a plasma-based comprehensive cancer genotyping assay utilizing orthogonal tissue- and plasma-based methodologies. Clin Cancer Res 2018;24:35393549.

    • PubMed
    • Search Google Scholar
    • Export Citation

Submitted June 21, 2024; final revision received July 25, 2024; accepted for publication August 26, 2024. Published online December 27, 2024.

X. Yang, X. Li, and J. Yan contributed equally.

Disclosures: The authors have disclosed that they have not received any financial considerations from any person or organization to support the preparation, analysis, results, or discussion of this article.

Funding: This work was supported by funding from the National Natural Science Foundation of China (82373021; J. Xue); 1.3.5 Project for Disciplines of Excellence, West China Hospital, Sichuan University (ZYYC23010 to J. Xue; ZYJC21003 to Y. Lu); and Science and Technology Department of Sichuan Province (00402053A29Z1).

Supplementary material: Supplementary material associated with this article is available online at https://doi.org/10.6004/jnccn.2024.7070. The supplementary material has been supplied by the author(s) and appears in its originally submitted form. It has not been edited or vetted by JNCCN. All contents and opinions are solely those of the author. Any comments or questions related to the supplementary materials should be directed to the corresponding author.

Correspondence: Jianxin Xue, MD, PhD, West China Hospital, Sichuan University, No. 37, Guoxue Lane, Chengdu, Sichuan, 610041, China. Email: radjianxin@163.com

Supplementary Materials

  • Collapse
  • Expand
  • Figure 1.

    Histopathologic findings of the left ilium biopsy. (A) Hematoxylin-eosin staining, and immunochemical stains of (B) Napsin A, (C) thyroid transcription factor 1 (TTF-1), and (D) cytokeratin 7 (CK7) (original magnification ×200 for all). All consistent with metastatic adenocarcinoma.

  • Figure 2.

    Clinical treatment history and serial imaging data of the patient. Timeline of events, including systemic and localized treatments and disease progression, is shown at the top. Cross-sectional CT images at the bottom indicate the development, progression, and shrinkage of representative lesions in lung and pelvis during the treatment course.

    Abbreviations: OR, overall response; PFS, progression-free survival; PR, partial response; SD, stable disease.

  • Figure 3.

    Dynamic changes of the variant allele frequencies of mutations over 6 time points. Fish plots at top and bottom show the evolving frequencies of gene alterations of interest in blood and tissue, respectively. To enhance visualization, we applied logarithmic transformation to the actual frequencies. The middle panel illustrates the relative frequencies by the sizes of circles, and the numbers within circles represent the actual frequencies detected by NGS. Filled circles and outlined circles indicate results from plasma- and tissue-based NGS analysis, respectively.

    Abbreviations: delins, deletion-insertion; NGS, next-generation sequencing.

    *Copy number of MET amplification.

  • Figure 4.

    Illustration of the POT1::NTRK3 fusion event. (A) Identification of POT1::NTRK3 fusion in plasma-based next-generation sequencing analysis. (B) Circos plot showing interchromosomal translocations between POT1 and NTRK3 represented by a colored line linking 2 genes. (C) Schematic diagram depicting the in-frame POT1::NTRK3 (P7;N15) fusion event. The fusion contains the full-length kinase domain of NTRK3 (exons 15–19).

  • Figure 5.

    Mutation profiles over 6 time points. Matched plasma- and tissue-based next-generation sequencing revealed the complex and evolving mutational landscape of the patient during treatment. Paired results of tissue and plasma are shown at timepoint 1 and timepoint 5. Numbers within the squares represent frequencies of the mutation.

    *Copy number.

  • 1.

    Sharma SV, Bell DW, Settleman J, et al. Epidermal growth factor receptor mutations in lung cancer. Nat Rev Cancer 2007;7:169181.

  • 2.

    Westover D, Zugazagoitia J, Cho BC, et al. Mechanisms of acquired resistance to first- and second-generation EGFR tyrosine kinase inhibitors. Ann Oncol 2018;29:I1019.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 3.

    Cooper AJ, Sequist L, Lin JJ. Third-generation EGFR and ALK inhibitors: mechanisms of resistance and management. Nat Rev Clin Oncol 2022;19:499514.

  • 4.

    Chakravarty D, Solit DB. Clinical cancer genomic profiling. Nat Rev Genet 2021;22:483501.

  • 5.

    Jänne PA, Yang JCH, Kim DW, et al. AZD9291 in EGFR inhibitor-resistant non-small-cell lung cancer. N Engl J Med 2015;372:16891699.

  • 6.

    Camidge DR, Pao W, Sequist LV. Acquired resistance to TKIs in solid tumours: learning from lung cancer. Nat Rev Clin Oncol 2014;11:473481.

  • 7.

    Leonetti A, Verze M, Minari R, et al. Resistance to osimertinib in advanced EGFR-mutated NSCLC: a prospective study of molecular genotyping on tissue and liquid biopsies. Br J Cancer 2024;130:135142.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 8.

    Rotow JK, Lee JK, Madison RW, et al. Real-world genomic profile of EGFR second-site mutations and other osimertinib resistance mechanisms and clinical landscape of NSCLC post-osimertinib. J Thorac Oncol 2024;19:227239.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 9.

    Zhang Z, Yang S, Wang QM. Impact of MET alterations on targeted therapy with EGFR-tyrosine kinase inhibitors for EGFR-mutant lung cancer. Biomark Res 2019;7:27.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 10.

    Ahn MJ, De Marinis F, Bonanno L, et al. MET biomarker-based preliminary efficacy analysis in SAVANNAH: savolitinib plus osimertinib in EGFRm NSCLC post-osimertinib. J Thorac Oncol 2022;17:S469470.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 11.

    Hartmaier RJ, Markovets AA, Ahn MJ, et al. Osimertinib plus savolitinib to overcome acquired MET-mediated resistance in epidermal growth factor receptor-mutated, MET-amplified non-small cell lung cancer: TATTON. Cancer Discov 2023;13:98113.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 12.

    Kim TM, Guarneri V, Jye VP, et al. Tepotinib plus osimertinib in EGFR-mutant NSCLC with MET amplification following 1L osimertinib: INSIGHT 2 primary analysis. J Thorac Oncol 2023;18:S94.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 13.

    Zhou CC, Tang KJ, Cho BC, et al. Amivantamab plus chemotherapy in NSCLC with EGFR exon 20 insertions. N Engl J Med 2023;389:20392051.

  • 14.

    Liu FF, Wei YX, Zhang H, et al. NTRK fusion in non-small cell lung cancer: diagnosis, therapy, and TRK inhibitor resistance. Front Oncol 2022;12:864666.

  • 15.

    Hong DS, DuBois SG, Kummar S, et al. Larotrectinib in patients with TRK fusion-positive solid tumours: a pooled analysis of three phase 1/2 clinical trials. Lancet Oncol 2020;21:531540.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 16.

    Doebele RC, Drilon A, Paz-Ares L, et al. Entrectinib in patients with advanced or metastatic NTRK fusion-positive solid tumours: integrated analysis of three phase 1-2 trials. Lancet Oncol 2020;21:271282.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 17.

    Drilon A, Camidge DR, Lin JJ, et al. Repotrectinib in ROS1 fusion-positive non-small-cell lung cancer. N Engl J Med 2024;390:118131.

  • 18.

    Rolfo C, Drilon A, Hong D, et al. NTRK1 fusions identified by non-invasive plasma next-generation sequencing (NGS) across 9 cancer types. Br J Cancer 2022;126:514520.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 19.

    Xia H, Xue XY, Ding HX, et al. Evidence of NTRK1 fusion as resistance mechanism to EGFR TKI in EGFR+ NSCLC: results from a large-scale survey of NTRK1 fusions in Chinese patients with lung cancer. Clin Lung Cancer 2020;21:247254.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 20.

    Euasobhon P, Atisook R, Bumrungchatudom K, et al. Reliability and responsivity of pain intensity scales in individuals with chronic pain. Pain 2022;163:E11841191.

  • 21.

    Arulananda S, Do H, Musafer A, et al. Combination osimertinib and gefitinib in C797S and T790M EGFR-mutated non-small cell lung cancer. J Thorac Oncol 2017;12:17281732.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 22.

    Zhou Z, Zhao Y, Shen SP, et al. Durable clinical response of lung adenocarcinoma harboring EGFR 19Del/T790M/in trans-C797S to combination therapy of first- and third-generation EGFR tyrosine kinase inhibitors. J Thorac Oncol 2019;14:E157159.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 23.

    Wang Z, Yang JJ, Huang J, et al. Lung adenocarcinoma harboring EGFR T790M and in trans C797S responds to combination therapy of first- and third-generation EGFR TKIs and shifts allelic configuration at resistance. J Thorac Oncol 2017;12:17231727.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 24.

    Yang YN, Xu HY, Ma L, et al. Possibility of brigatinib-based therapy, or chemotherapy plus anti-angiogenic treatment after resistance of osimertinib harboring EGFR T790M-cis-C797S mutations in lung adenocarcinoma patients. Cancer Med 2021;10:83288337.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 25.

    Wang YB, Yang N, Zhang YC, et al. Effective treatment of lung adenocarcinoma harboring EGFR-activating mutation, T790M, and cis-C797S triple mutations by brigatinib and cetuximab combination therapy. J Thorac Oncol 2020;15:13691375.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 26.

    Lu C, Wei XW, Wang Z, et al. Allelic context of EGFR C797X-mutant lung cancer defines four subtypes with heterogeneous genomic landscape and distinct clinical outcomes. J Thorac Oncol 2024;19:601612.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 27.

    Liu CL, Li JD, Liu HL, et al. The concomitant EGFR T790M/C797S in trans and cis in three osimertinib-resistant lung adenocarcinoma patients. J Clin Oncol 2019;37(Suppl):Abstract e20105.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 28.

    Zhou RG, Song L, Zhang WW, et al. Combination of osimertinib and anlotinib may overcome the resistance mediated by in cis EGFR T790M-C797S in NSCLC: a case report. Onco Targets Ther 2021;14:28472851.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 29.

    Marchiò C, Scaltriti M, Ladanyi M, et al. ESMO recommendations on the standard methods to detect NTRK fusions in daily practice and clinical research. Ann Oncol 2019;30:14171427.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 30.

    Schrock AB, Zhu VW, Hsieh WS, et al. Receptor tyrosine kinase fusions and BRAF kinase fusions are rare but actionable resistance mechanisms to EGFR tyrosine kinase inhibitors. J Thorac Oncol 2018;13:13121323.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 31.

    Liu Q, Yu SN, Zhao WH, et al. EGFR-TKIs resistance via EGFR-independent signaling pathways. Mol Cancer 2018;17:53.

  • 32.

    Amatu A, Sartore-Bianchi A, Bencardino K, et al. Tropomyosin receptor kinase (TRK) biology and the role of NTRK gene fusions in cancer. Ann Oncol 2019;30(Suppl 8):viii515.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 33.

    Cocco E, Scaltriti M, Drilon A. NTRK fusion-positive cancers and TRK inhibitor therapy. Nat Rev Clin Oncol 2018;15:731747.

  • 34.

    Levantini E, Maroni G, Del Re M, et al. EGFR signaling pathway as therapeutic target in human cancers. Semin Cancer Biol 2022;85:253275.

  • 35.

    Farago AF, Taylor MS, Doebele RC, et al. Clinicopathologic features of non-small-cell lung cancer harboring an NTRK gene fusion. JCO Precis Oncol 2018;2018:PO.18.00037.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 36.

    Piotrowska Z, Isozaki H, Lennerz JK, et al. Landscape of acquired resistance to osimertinib in EGFR-mutant NSCLC and clinical validation of combined EGFR and RET inhibition with osimertinib and BLU-667 for acquired RET fusion. Cancer Discov 2018;8:15291539.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 37.

    Zhang YZ, Xia MF, Jin K, et al. Function of the c-Met receptor tyrosine kinase in carcinogenesis and associated therapeutic opportunities. Mol Cancer 2018;17:45.

  • 38.

    Cocco E, Schram AM, Kulick A, et al. Resistance to TRK inhibition mediated by convergent MAPK pathway activation. Nat Med 2019;25:14221427.

  • 39.

    Tsai JM, Hata AN, Lennerz JK. MET D1228N and D1246N are the same resistance mutation in MET exon 14 skipping. Oncologist 2021;26:E22972301.

  • 40.

    Bahcall M, Sim T, Paweletz CP, et al. Acquired METD1228V mutation and resistance to MET inhibition in lung cancer. Cancer Discov 2016;6:13341341.

  • 41.

    Kuijpers CCHJ, Hendriks LEL, Derks JL, et al. Association of molecular status and metastatic organs at diagnosis in patients with stage IV non-squamous non-small cell lung cancer. Lung Cancer 2018;121:7681.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 42.

    Huang HM, Li HX. Tumor heterogeneity and the potential role of liquid biopsy in bladder cancer. Cancer Commun (Lond) 2021;41:91108.

  • 43.

    Russano M, Napolitano A, Ribelli G, et al. Liquid biopsy and tumor heterogeneity in metastatic solid tumors: the potentiality of blood samples. J Exp Clin Canc Res 2020;39:95.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 44.

    Pascual J, Attard G, Bidard FC, et al. ESMO recommendations on the use of circulating tumour DNA assays for patients with cancer: a report from the ESMO Precision Medicine Working Group. Ann Oncol 2022;33:750768.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 45.

    Sacher AG, Paweletz C, Dahlberg SE, et al. Prospective validation of rapid plasma genotyping for the detection of EGFR and KRAS mutations in advanced lung cancer. JAMA Oncol 2016;2:10141022.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 46.

    Odegaard JI, Vincent JJ, Mortimer S, et al. Validation of a plasma-based comprehensive cancer genotyping assay utilizing orthogonal tissue- and plasma-based methodologies. Clin Cancer Res 2018;24:35393549.

    • PubMed
    • Search Google Scholar
    • Export Citation

Metrics

All Time Past Year Past 30 Days
Abstract Views 0 0 0
Full Text Views 2221 2222 2221
PDF Downloads 503 503 503
EPUB Downloads 0 0 0