Impacts of co-mutations in oligometastatic and oligoprogressive non-small cell lung cancer with EGFR/ALK mutations—a narrative review of the current literature
Review Article

Impacts of co-mutations in oligometastatic and oligoprogressive non-small cell lung cancer with EGFR/ALK mutations—a narrative review of the current literature

Jeong Uk Lim1 ORCID logo, Ae Lee Jang1, Jayoung Lee2, Sonya Youngju Park3, Tai Joon An1, Young Jo Sa4, Hyo Rim Kim5, Tae-Jung Kim6, Byoung Hyuck Kim7, Chan Kwon Park1

1Division of Pulmonology and Critical Care Medicine, Department of Internal Medicine, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; 2Department of Radiation Oncology, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; 3Division of Nuclear Medicine, Department of Radiology, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; 4Department of Thoracic and Cardiovascular Surgery, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; 5Department of Radiology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; 6Department of Hospital Pathology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; 7Department of Radiation Oncology, Seoul Metropolitan Government Seoul National University Boramae Medical Center, Seoul, Republic of Korea

Contributions: (I) Conception and design: JU Lim, CK Park; (II) Administrative support: JU Lim, AL Jang; (III) Provision of study materials or patients: All authors; (IV) Collection and assembly of data: All authors; (V) Data analysis and interpretation: JU Lim; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors.

Correspondence to: Chan Kwon Park, MD, PhD. Division of Pulmonology and Critical Care Medicine, Department of Internal Medicine, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, 10, 63-ro, Yeongdeungpo-gu, Seoul 07345, Republic of Korea. Email: ckpaul@catholic.ac.kr.

Background and Objective: Non-small cell lung cancer (NSCLC) remains one of the primary causes of cancer mortality globally, with an increasing focus on advanced targeted therapies. Despite these advancements, oligometastatic NSCLC, particularly cases with actionable mutations such as those in epidermal growth factor receptor (EGFR) and anaplastic lymphoma kinase (ALK), presents unique therapeutic challenges and opportunities for improved outcomes. Recent studies indicate that consolidative local ablative therapies (LAT) such as stereotactic body radiation therapy (SBRT) combined with tyrosine kinase inhibitors (TKIs) may enhance progression-free and overall survival for patients with oligometastatic NSCLC harboring these mutations. This narrative review aims to summarize current evidence on the clinical impact of co-mutations in EGFR/ALK-positive oligometastatic NSCLC.

Methods: The relevant literature was identified by using PubMed and ClinicalTrials.gov (last search phase November 2024) and was restricted to English language. Peer-reviewed manuscripts but also conference abstracts that did not undergo peer-review were included.

Key Content and Findings: Co-mutations complicate treatment by potentially influencing radiosensitivity and resistance to systemic therapies. This review discusses current findings on co-mutations in EGFR/ALK-positive oligometastatic NSCLC, examining their impact on LAT and systemic treatment outcomes, with a particular focus on synchronous and oligoprogressive disease states. Moreover, emerging biomarkers such as circulating tumor DNA may guide therapeutic strategies and optimize personalized treatment plans.

Conclusions: As clinical trials continue to investigate combinative and sequential LAT-TKI strategies, understanding the genomic landscape of co-mutations in oligometastatic NSCLC is important for refining treatment approaches and enhancing long-term survival.

Keywords: Non-small cell lung cancer (NSCLC); EGFR mutation; oligometastasis; co-mutations; stereotactic body radiation therapy (SBRT)


Submitted Nov 21, 2024. Accepted for publication Mar 13, 2025. Published online May 28, 2025.

doi: 10.21037/tlcr-2024-1121


Introduction

Non-small cell lung cancer (NSCLC) comprises approximately 80% of all lung cancers worldwide and is one of the leading causes of cancer-related mortality (1). Approximately 40% of NSCLC patients present with metastatic disease. With the advent of targeted therapies and immunotherapy, patients with advanced NSCLC can expect improved survival outcomes compared to those with non-mutation-driven NSCLC. Unfortunately, as these systemic treatments are typically palliative rather than curative, most cancers eventually progress (2).

An emerging focus in improving outcomes for NSCLC is recognizing oligometastatic NSCLC as a distinct disease entity. Oligometastatic NSCLC can be described as an intermediate metastatic state, indicating an early stage in the spread of cancer with limited metastatic potential. The term oligometastasis was first proposed in 1995 by Hellman and Weichselbaum (3). This stage describes a clinically significant condition characterized by limited metastatic burden in one or a few organs, while the generally accepted number of metastatic lesions is 1 to 5 (4). Moreover, more sensitive staging imaging modalities, such as positron emission tomography-computed tomography (PET-CT), have gained clinical importance in defining oligometastatic disease (5). In comparison to more disseminated metastatic states, oligometastatic disease appears to follow an indolent course, with better prognosis, and may therefore be considered for radical multimodality treatment. Systemic treatment combined with stereotactic ablative radiotherapy (SABR), resection of metastatic lesion or thermal ablation can achieve long-term survival in terms of progression-free survival (PFS) and overall survival (OS) rates in oligo­metastatic NSCLC (6-8).

When addressing oligometastases, clinical efforts traditionally focus on the size and number of metastatic lesions (9). However, mutational burden has received relatively little clinical attention. Clinicians should determine whether a radical treatment approach needs adjustment based on the presence of co-mutations. This consideration becomes particularly significant in oligometastatic NSCLC patients who possess actionable mutations, such as those in the epidermal growth factor receptor (EGFR) and anaplastic lymphoma kinase (ALK) genes. In a prospective phase 2 trial assessing SABR in stage IV NSCLC patients with ≤5 lesions, of the 47 participants, somatic genetic alterations (EGFR/ALK) were detected in 44.7%, with EGFR exon 19 deletions being the most prevalent in 38.3% of the cohort (10).

In metastatic NSCLC, including oligometastatic cases, patients undergo comprehensive mutational analyses, such as next-generation sequencing (NGS) from tissue or plasma or whole-exome sequencing (WES). When applied to oligometastatic NSCLC, this approach provides detailed information on mutational burden and potential targetable co-mutations in EGFR/ALK-mutated NSCLC and is not significantly different from strategies used in disseminated metastatic NSCLC. While the efficacy of local treatments like radiotherapy and surgical resection combined with systemic therapy is well-studied in oncogene-addicted oligometastatic NSCLC (6,7,11), now is an optimal time to explore how co-mutations influence the management of oligometastatic NSCLC with EGFR or ALK mutations and identify opportunities for further enhancing clinical outcomes.

In this review, we discuss the impact of co-mutations on oligometastatic NSCLC patients with targetable alterations undergoing targeted therapy. Among various oligometastatic categories (12), our primary discussion will focus on synchronous oligometastatic disease and oligoprogressive NSCLC in patients with targetable mutations. We present this article in accordance with the Narrative Review reporting checklist (available at https://tlcr.amegroups.com/article/view/10.21037/tlcr-2024-1121/rc).


Methods

The authors conducted an electronic literature search to identify relevant publications available in PubMed and ClinicalTrials.gov (search in November 2024). The search was restricted to articles in the English language. In addition to peer-reviewed manuscripts, conference abstracts were also included in this review. The titles and abstracts of the search results were screened, and irrelevant publications were excluded based on the authors’ evaluation following established criteria.

Only the pertinent manuscripts were further examined for their full content to extract relevant information. However, since this is intended as a narrative review, a systematic literature search was not conducted, and the included literature reflects the authors’ preferences. The literature reviewed for this manuscript covers the period from 2008 to 2024 (Table 1). J.U.L., J.L., and C.K.P. independently screened the studies for inclusion. In cases of disagreement, consensus was reached through discussion.

Table 1

The search strategy summary

Items Specification
Date of search November 2024
Databases and other sources searched PubMed, ClinicalTrials.gov
Search terms used ((NSCLC) OR (lung cancer)) AND ((oligometastases) OR (oligoprogressive) OR (metastases)) AND ((ALK) OR (EGFR) OR (co-mutation))
Timeframe 2008 to 2024
Inclusion criteria All study types written in English. We included peer-reviewed manuscripts but also conference abstracts that have not yet been published following peer review
Selection process Study selection was conducted by J.U.L., J.L., and C.K.P.

NSCLC, non-small cell lung cancer; ALK, anaplastic lymphoma kinase; EGFR, epidermal growth factor receptor.


Current treatment of synchronous oligometastatic NSCLC with targetable mutations

Existing research on managing oligometastatic NSCLC with actionable mutations is mostly retrospective and from single institutions. However, several key prospective studies and current guidelines provide necessary insights.

In a randomized trial involving patients with common EGFR mutations, the efficacy of a first-generation tyrosine kinase inhibitor (TKI) alone was compared with a regimen combining stereotactic body radiation therapy (SBRT) to both the primary tumor and all metastatic lesions followed by TKI therapy (13). The phase II ATOM study evaluated the effectiveness of local ablative therapy (LAT) in oligoresidual patients undergoing EGFR-TKI treatment. Preemptive LAT significantly decreased the progression risk [hazard ratio (HR) 0.41, P=0.0097]. Nonetheless, enrollment was halted prematurely due to sluggish accrual, possibly because of heterogeneity in oligometastatic cases and a shift in clinical practice and oligometastatic criteria (14).

In the SINDAS phase III randomized controlled trial (RCT), 133 patients with oligometastatic lung adenocarcinoma harboring an EGFR mutation received radiotherapy to all metastases and the primary tumor/involved regional lymphatics alongside first-generation EGFR-TKIs. This combination significantly enhanced median progression-free survival (mPFS) and overall survival (mOS), with mPFS at 20.2 versus 12.5 months and mOS at 25.5 versus 17.4 months, both showing HR of 0.68 with high statistical significance (P<0.001), but the exclusion of patients with brain metastases (BM) limits the applicability of the results to broader clinical practice challenging (13). Nevertheless, several retrospective studies have shown that the use of consolidative LAT, such as SBRT or surgery, to treat metastatic sites in patients with EGFR-mutant oligometastatic NSCLC during ongoing EGFR-TKI therapy results in improvements in both PFS and OS (15-17). Not only have early-generation TKIs demonstrated efficacy when combined with localized radiotherapy in oligometastatic NSCLC, but later-generation TKIs, such as osimertinib, have also shown positive results (18,19).

Current guidelines from major medical societies generally recommend integrating local treatment with targeted therapy for managing oligometastatic NSCLC that harbor actionable mutations. ASTRO/ESTRO Clinical Practice Guidelines recommend administering optimal systemic therapy including targeted therapy for metastatic disease for at least three months to evaluate response and treatment tolerance before considering definitive local therapy. This systemic approach is to ensure eligibility for local treatment of oligometastatic disease by assessing the potential for complete response or progression during the initial treatment period (20). Recent recommendations by the EORTC-ESTRO OligoCare consortium advise combining EGFR or ALK inhibitors with SBRT for eligible patients without reducing the radiation dose (21). The 2023 European Society for Medical Oncology (ESMO) guidelines, despite a lack of extensive prospective data, recommend integrating LAT with systemic treatment for oncogene-addicted synchronous oligometastatic and oligoprogressive NSCLC (22). The National Comprehensive Cancer Network (NCCN) guideline recommends integrating local treatment with EGFR or ALK inhibitors (23).

Ongoing studies involving patients with oligometastatic NSCLC and actionable mutations are expected to yield results that will enhance clinical strategies (Table 2).

Table 2

Ongoing studies related to oligometastatic diseases with targetable mutations

Trials Study design Patient eligibility Study arms Objectives Reported outcomes References
OMEGA (NCT03827577) Phase III randomized Metachronous and synchronous, 1–3 metastases Systemic therapy ± upfront LAT or LCT consisting of primary resection and OM LAT with surgery, SABR, or RFA OS up to 60 months N/A, ongoing (24)
OITROLC (NCT02076477) Multicenter phase III RCT Stage IV synchronous OM-NSCLC up to 5 metastases Upfront LAT arm: ChemoRT followed by 2 cycles of CTx vs. LCT arm receive 2 cycles of CTx followed by ChemoRT Response rate at 3 months, PFS N/A, ongoing (25)
NRG-LU002 (NCT03137771) Randomized phase II/III Oligometastatic NSCLC (≤3 sites), after 1st line systemic therapy Maintenance therapy plus SABR vs. maintenance therapy alone Phase II: PFS; phase III: OS 1-yr and 2-yr PFS and OS rates not significantly different between arms. The experimental arm exhibited higher incidence of grade 2 and more severe toxicities (26)
SARON, UK (NCT02417662) Randomized phase III Synchronous oligometastatic NSCLC (≤3 sites). EGFR, ALK and ROS1 mutation-negative Chemotherapy plus SABR vs. chemotherapy alone OS N/A, ongoing (27)
STEREO-OS (NCT03143322) Multicenter phase III RCT Solid tumors 1–3 bone-only metastases Systemic treatment and SBRT vs. systemic treatment + palliative RT PFS N/A, ongoing (28)
OUTRUN (NCT03497767) Randomized phase II Untreated brain metastases in patients with EGFR mutant NSCLC Osimertinib with or without SRS for EGFR mutated NSCLC with brain metastases Intracranial PFS, OS Pooled analysis found no sufficient evidence to support that upfront SRS improves intracranial PFS at 12 months or OS, possibly due to small sample size (29)
PROMISE (NCT03808662) Randomized phase II Oligoprogressive breast and NSCLC patients Early SBRT to all extracranial oligoprogressive sites compared to SOC PFS, OS The median PFS was higher for patients with NSCLC in the SBRT group compared to those in the standard-of-care group (10.0 vs. 2.2 months; HR 0.41, 95% CI: 0.22–0.75; P=0.0039) (30)
STOP (NCT02756793) Randomized, interventional (phase N/A) Oligoprogressive NSCLC SABR to all sites of progressive disease, compared to SOC arm with continuation of systemic treatment PFS, OS, etc. SABR achieved better control of individual lesions, but did not lead to improved PFS or OS. Could have been influenced by patients in SOC arm undergoing ablation (31)
NORTHSTAR (NCT03410043) Randomized phase II Stage IIIB/IV or recurrent NSCLC with EGFR mutations. Candidate for LCT After frontline osimertinib, patients undergo surgery and/or radiation for up to 8 weeks. Osimertinib continued after LCT PFS, OS, progression of target lesions, Ongoing, N/A (32)
HALT (NCT03256981) Randomized phase II/III Oligoprogressive NSCLC with actionable mutation Randomized to receive either SBRT or no SBRT at a ratio of 2:1 with continuation of TKI PFS Ongoing, N/A (33)
CORE (NCT02759783) Randomized phase II/III Synchronous or metachronous breast cancer, NSCLC, and prostate cancer SBRT followed by SOC treatment versus SOC treatment only PFS Overall PFS was not significantly different, with an HR of 0.79 (95% CI: 0.57–1.09, P=0.16) favoring SBRT + SOC; median PFS was 25.0 months for SBRT + SOC and 19.9 months for SOC (34)

ALK, anaplastic lymphoma kinase; ChemoRT, chemoradiotherapy; CI, confidence interval; CTx, chemotherapy; EGFR, epidermal growth factor receptor; HR, hazard ratio; LAT, local ablative therapy; LCT, local consolidative therapy; N/A, not available; NSCLC, non-small cell lung cancer; OM, oligometastases; OS, overall survival; PFS, progression-free survival; RCT, randomized controlled trial; RFA, radiofrequency ablation; RT, radiation therapy; SABR, stereotactic ablative radiotherapy; SBRT, stereotactic body radiation therapy; SOC, standard of care; SRS, stereotactic radiosurgery; TKI, tyrosine kinase inhibitor.

In the OMEGA study (NCT03827577), among the four subgroups classified based on targetable mutations and PD-L1 expression, patients with either EGFR, ALK, or ROS1 mutations were categorized separately as one group. Meanwhile, the phase II NORTHSTAR trial (NCT03410043) aims to evaluate the efficacy of initial osimertinib treatment, with consideration for adding local consolidative radiotherapy or surgical resection in patients diagnosed with stage IIIB-IV EGFR-mutant NSCLC following 6–12 weeks of induction with osimertinib (24,32,35). While earlier research primarily assessed the efficacy of combining local and systemic treatments, current studies are expanding their scope to include detailed treatment strategies such as the sequencing and timing of local and systemic interventions, and the identification of biomarkers (36-38).


Management of oligoprogressive oncogene‑driven NSCLC

While the related literature predominantly focuses on synchronous oligometastatic diseases, oligoprogressive disease frequently encountered in oncogene-addicted NSCLC requires a separate approach. Oligoprogression is characterized as the emergence of metachronous oligometastatic disease during active systemic treatment, predominantly seen in patients with oncogene-driven NSCLC undergoing targeted therapy. This concept defines a subset of metastatic disease where limited sites progress while the primary tumor and other metastases remain controlled under ongoing systemic treatment (39). In EGFR-mutant NSCLC, patients are more likely to show oligoprogressive progression patterns rather than systemic patterns (72% vs. 28%), as shown in a real-world data analysis of patients undergoing osimertinib treatment (40).

In a study by Weickhardt et al., disease control benefit was noted in patients with EGFR and ALK-positive NSCLC undergoing oligoprogression during ongoing targeted therapy. Of 51 patients, 49% (25 patients) received local therapy in addition to continuous crizotinib or erlotinib. Among those experiencing progression after the local therapy, the post-local therapy PFS was 6.2 months (41). Qiu et al. performed a retrospective study on 46 patients with oligoprogression, showing that EGFR mutation type, local treatment site, and the interval from progression to local treatment were independent prognostic factors. They demonstrated a median OS of 14 months, significantly longer than the 7 months (P=0.0009) observed in control group patients who did not receive local treatment (42). Patients with NSCLC harboring EGFR or ALK mutations have a higher incidence of BM at baseline compared to wild-type patients (43,44), highlighting the need for a special focus on BM. In a retrospective Swiss cohort study, 50 EGFR T790M-positive NSCLC patients were treated with osimertinib, revealing that 17% experienced brain progression, with 83% of these cases classified as oligoprogression (40). Another study analyzing 61 ALK/ROS1/RET-rearranged lung adenocarcinomas showed that local therapy (radiotherapy, surgery, and percutaneous thermal ablation) for progression on ALK, ROS1, or RET TKIs was associated with significant prolongation of continued TKI therapy beyond progression (45).

In cases of oligoprogression in NSCLC with EGFR mutations, several management strategies can be selected based on tumor burden, patient conditions, and characteristics of the available treatment modalities. For polymetastatic NSCLC with targetable mutations, clinical decision-making regarding the switch in systemic treatment modalities at the time of progression after prior targeted therapy is more straightforward when actionable co-mutations are identified, either through tissue-based or circulating markers, as mechanisms of acquired resistance (46-49). However, for oligoprogressive NSCLC patients, this decision should be more personalized, as discontinuing targeted therapy too early may prematurely remove future treatment options. Specifically, localized treatment, such as SBRT for an oligoprogressive lesion, can be a viable option.

One strategy is to continue existing systemic treatments, such as EGFR-TKIs, even after oligoprogression. This consideration is based on the possibility for tumor flare-ups, which are observed in approximately 20% of patients following the abrupt discontinuation of previously administered TKIs (50). The current NCCN guidelines recommend continuing first-line treatment and definitive local therapy for asymptomatic progressive lesions in patients with limited progression while on osimertinib or amivantamab-vmjw plus lazertinib in EGFR-mutant NSCLC (51). Different strategies combine first-line EGFR-TKIs in patients with chemotherapy or with another targeted agent (52). Acquiring tissue at the time of progression can be beneficial for optimal clinical decision-making in oligoprogressive NSCLC with EGFR mutations. Choudhury et al. demonstrated that patients treated with TKIs who underwent post-progression tissue-based genomic analysis to identify acquired resistance mechanisms had improved survival outcomes with subsequent treatments (53). For appropriate candidates, surgical resection of oligoprogressive lesions may be advantageous as it not only provides abundant tissue samples for precise pathological examination but also reduces tumor burden (54).


Genetic alterations and intertumoral heterogeneity in oligometastatic NSCLC

Data of oligometastatic NSCLC specifically with targetable mutations are scarce, but a number of previous studies have examined genetic alterations associated with oligometastatic NSCLC. The co-mutations should be considered in relation to other disease statuses, with resectable or locally advanced NSCLC indicating a disease burden smaller than oligometastasis, and polymetastatic NSCLC indicating a larger burden. Comparing genetic alterations among early-stage, oligometastatic, and polymetastatic NSCLC would be clinically significant in understanding disease progression and treatment implications. However, more real-data analyses based on in-depth pathological samples are essential for further validation. Nevertheless, recent studies provide some insights into the co-mutations of oligometastatic diseases.

Werner et al. reported that Kirsten rat sarcoma viral oncogene homolog (KRAS) mutations were the most prevalent genetic alterations in primary tumors, with variations such as G12C, G12V, G13C, G12A, and Q61H, listed in order of frequency. Other notable genetic alterations observed included EGFR mutations, CDK4 amplifications, NRAS mutation, amplifications of EGFR, ERBB2, PIK3CA, and an EML4-ALK fusion (55).

In a study by Liao et al., oligometastatic (n=77) and polymetastatic (n=21) NSCLC cases were compared for genetic alterations using NGS. Mutations in ERBB2, ALK, MLL4, PIK3CB, and TOP2A were significantly less frequent in the oligometastatic group compared to polymetastatic patients. Additionally, EGFR and Kelch-like ECH-associated protein 1 (KEAP1) alterations were found to be mutually exclusive in the oligometastatic group; however, this finding is specific to this study and is limited by the small sample size. They further found that mutations in high-frequency genes among oligometastatic patients included TP53 (64%), EGFR (56%), and others such as CDKN2A and KRAS, linked to critical pathways like p53, RTK/RAS, PI3K-Akt, and cell cycle regulation (56).

While oligometastatic NSCLC comprises multiple tumor sites, discrepancies in genetic alterations between primary tumors and metastatic sites can be observed. This intertumoral heterogeneity can be an obstacle in the effective treatment of oligometastatic NSCLC (57). However, few studies have specifically evaluated intertumoral heterogeneity between primary tumor and metastatic samples in oligometastatic NSCLC. Werner et al. (55) retrospectively analyzed genetic alterations in oligometastatic NSCLC by examining primary tumors and BM from 46 patients. While most primary tumor alterations (93.5%) were preserved in BM, about 34.8% of BMs showed additional oncogenic mutations, especially MYC amplifications found only in metastatic sites.

Intertumoral heterogeneity, marked by additional genetic alterations of metastatic sites, is an independent predictor of reduced OS in patients with oligometastatic NSCLC and synchronous or metachronous BM (55). Song et al. examined treatment-naïve lung cancer patients with oligometastatic BM for genetic alterations in primary and metastatic tumors. The most frequent mutations in primary tumors included EGFR (44%) and TP53 (40%), while BM showed a similar profile with minor differences including slightly higher co-mutations of EGFR and TP53. RNA Binding Motif Protein 10 (RBM10), a tumor suppressor gene involved in RNA splicing regulation, was more frequently observed in primary tumors. The analysis revealed only minor differences in genetic mutations between primary tumors and their corresponding metastases (58).

Future studies with sufficiently large, molecularly stratified analyses are needed to determine whether specific genetic alterations influence the response to localized therapies such as SBRT or combinatorial approaches in oligometastatic NSCLC. Identifying such associations could enhance personalized treatment strategies and improve patient outcomes.


Impact of co-mutations on management of oligometastatic NSCLC with targetable mutations

It is crucial for clinicians to consider information about co-mutations in oligometastatic NSCLC with targetable mutations, focusing on prognosis prediction and treatment strategy planning. The approach may vary depending on the type of oligometastasis (synchronous or oligoprogressive) and previous treatments received.

In a prospective study focusing on the surgical resection of oligopersistent NSCLC, where the majority of participants exhibited EGFR mutations, WES of tumors identified resistant clones in some patients. Importantly, patients without detected acquired resistance mutations in their tumor tissues demonstrated more favorable prognosis compared to those with such mutations [HR =3.67, 95% confidence interval (CI): 0.92–14.7, P=0.006] (59).

Although not all patients enrolled were oligometastatic, in a retrospective analysis of EGFR mutant NSCLC patients who underwent SBRT combined with TKI treatment, liquid NGS at baseline and at the time of progression indicated the presence of T790M mutations in 64.3% (18/28) of patients, followed by TP53 mutations in 28.6% (8/28), and BRAF mutations in 3.6% (60). These findings suggest that the genetic alteration landscape in patients eligible for local treatment closely mirrors that of individuals undergoing TKI therapy for advanced EGFR-mutant NSCLC (61).

Earlier studies often did not focus on co-mutations in EGFR/ALK-mutated oligometastatic NSCLC. However, ongoing studies are expected to more clearly show the impact of co-mutations on treatment outcomes (24,32,62), as they incorporate more comprehensive genetic analyses, including NGS.


Role of lung resection in detection of co-mutations

For synchronous oligometastatic NSCLC patients with actionable mutations, systemic targeted therapy is based on targetable mutations identified at the time of diagnosis. For patients with oligoprogressive NSCLC, when rebiopsy samples are available at the time of oligoprogression, the mutational profile is crucial for guiding subsequent treatment decisions, particularly information related to acquired resistance. In this context, obtaining an adequate biopsy sample after the initiation of first-line TKI treatment can significantly facilitate decisions regarding further therapeutic options. In this regard, lung resection is a clinical option to consider.

Lung resection following initial nonoperative management for initially unresectable NSCLC that later becomes resectable is a treatment option increasingly under consideration in oligometastatic NSCLC (63-65). Dunne et al. observed that 22% of patients underwent surgery for oligoprogressive disease and 38% for local control of oligometastatic disease; their study found that patients with stage IV NSCLC who underwent salvage lung resection had better OS compared to those who did not undergo surgery (66). In a study by Joosten et al., where 28 patients underwent surgery for an oligoprogressive lesion, it was shown that resecting oligoprogressive lesions after systemic treatment is a feasible option in NSCLC, yielding favorable outcomes post-resection (67). Besides improving prognosis in appropriate surgical candidates, resecting lung lesions also enables the detection of co-mutations using adequate sample amounts. For instance, a retrospective analysis involving 33 EGFR mutant NSCLC patients who underwent salvage surgery after EGFR-TKI treatment showed that 11 of the 33 patients had a T790M mutation in the resected sample (63). In another retrospective analysis of 29 salvage surgery cases following prior EGFR-TKI treatment, the majority of patients had M1b and oligometastatic M1c disease, possibly representing oligopersistent or oligoresidual cases. Resected lung samples showed a variety of co-mutations, including the T790M mutation and mutations in the APC, CTNNB1, PIK3CA, and PTEN genes (68).

However, a multidisciplinary discussion should precede surgical intervention to ensure that patients who are operable and have resectable target lesions undergo surgery, thereby avoiding unnecessary surgery-related adverse outcomes.


Impact of co-mutations on radiosensitivity of oligometastatic NSCLC with targetable mutations

Radiotherapy primarily induces tumor cell death by causing DNA damage. However, tumor cells respond by activating checkpoint signaling and DNA repair mechanisms. While the DNA damage response (DDR) typically initiates programmed cell death, co-mutations, intertumor heterogeneity, and the surrounding microenvironment allow cancer cells to evade death and develop resistance to radiation (69). For example, mutant EGFR can activate various downstream pathways including RAS/ERK, MAPK, JNK, or mTOR leading to tumor cell proliferation and anti-apoptosis (70). This acquired radioresistance is a key factor in treatment failure and recurrences.

Radiotherapy plays a central role in the management of oligometastatic NSCLC with targetable mutations, and predicting sensitivity to the treatment can be crucial for improving patient outcomes. Certain genetic alterations can impact radiosensitivity. For instance, KRAS mutations are associated with radioresistance (71,72), an association supported in several real-world data analyses (73,74). In a small SBRT case series, KRAS mutation was associated with cancer-specific survival in early-stage NSCLC (72). Mutations in other genes such as KEAP1 (75,76), and TP53 are also known to be correlated with radioresistance in NSCLC (77,78). In an analysis of patients who underwent SBRT for stage I lung adenocarcinoma, MET-amplification was associated with worse regional and distant disease control (74). While certain genetic alterations are associated with decreased responses to radiotherapy, future studies are needed to determine whether radiotherapy methods or dose adjustments can help overcome this resistance in oligometastatic NSCLC.

Combining radiotherapy with targeted therapy may improve radiosensitivity in oncogene-addicted oligometastatic NSCLC. Several preclinical studies have shown that combining TKI, anti-EGFR antibody and other oncogenic molecular inhibitors can help overcome radioresistance (79,80). Potential synergistic effects from combining targeted therapy and radiotherapy may be more pronounced in oncogene-addicted NSCLC. However, clinicians should exercise caution when combining high-dose radiotherapy with targeted therapy, as the risk of radiation pneumonitis may increase (81). In patients with identifiable risk factors such as severe pre-existing lung diseases, withholding systemic therapy during radiotherapy can be considered to mitigate this risk (82).

An important question for clinicians to consider is whether the radiotherapy approach should vary based on co-mutations detected. In an analysis of 580 patients with 1,487 BM who underwent radiotherapy, genomic alterations identified through NGS revealed that mutations in 11 genes (ATM, MYCL, PALB2, FAS, PRDM1, PAX5, CDKN1B, EZH2, NBN, DIS3, and MDM4) and 2 genes (FBXW7 and AURKA) were linked to decreased or increased risk of local recurrence, respectively. Although patients with EGFR/ALK-altered NSCLC comprised only a subset of the study population, these results suggest that genetic alterations are clinically significant for predicting radiotherapy failure (83). For patients with aggressive genomic profiles, studies validating whether more intensive local treatment for BM is necessary should be performed.


Role of liquid biopsy

Liquid biopsy, a minimally invasive diagnostic platform, was examined for its potential role as a prognostic biomarker in oligometastatic NSCLC by detecting oncogenic mutations and predicting disease courses (84,85). The detection of circulating tumor DNA (ctDNA) and its burden has been analyzed in oligometastatic NSCLC using both single-point and serial measurements. In patients with oligometastatic NSCLC, the presence of a driver alteration in ctDNA has been linked to reduced PFS (HR 2.62, 95% CI: 1.27–5.43, P=0.009) after receiving consolidative radiation therapy, regardless of mutation status (37). In a multi-institutional cohort study involving 1,487 patients with oligometastatic NSCLC, analyses of 1,880 liquid biopsies revealed a 20% detection rate of ctDNA before radiotherapy. Patients with undetectable ctDNA prior to radiation therapy exhibited significantly improved PFS (P=0.004) and OS (P=0.030). Pre-radiation ctDNA maximum variant allele frequency (VAF) and ctDNA mutational burden were independently inversely correlated with PFS and OS (86). Cell-free DNA genetic alterations detected include EGFR, KRAS, ERBB2, MET, BRAF, ALK, ROS1, and RET, listed in order of frequency. Liquid biopsy, though minimally invasive, can aid in identifying patients with oligometastatic NSCLC who may benefit from more aggressive local treatment. With advancements in assay sensitivity, particularly in the detection of minimal residual disease (MRD), the prognostic and predictive value of liquid biopsy as a biomarker continues to evolve.

Changes in ctDNA status before and after local treatment can serve as prognostic indicators. In a prospective observational study by Fu et al., perioperative ctDNA levels were monitored in patients, primarily with oligopersistent disease, who underwent surgical resection of residual lesions following initial systemic therapy. Of the 68 patients studied, EGFR mutations were present in 64.7% of cases. Preoperative ctDNA negativity was a significant predictor of improved outcomes (HR =3.42, P=0.001). Moreover, patients who maintained or shifted to a ctDNA-negative status one month after surgery experienced better survival compared to those who remained ctDNA-positive, with hazard ratios of 3.22 for maintaining negativity and 4.57 for transitioning to negativity (59).


Future perspectives

Future research should focus on identifying the optimal treatment strategy for oligometastatic NSCLC, including the best sequencing approach and patient selection criteria.

While detecting co-mutations is becoming increasingly clinically significant and detection techniques are advancing rapidly, this is particularly important for oligometastatic NSCLC with targetable mutations. As more patient data accumulates from ongoing studies, treatment strategies will continue to evolve. Additionally, whether certain genetic alterations are associated with treatment outcomes specifically in oligometastatic NSCLC should be closely investigated so that the findings can be reflected in treatment guidelines. Furthermore, it will be clinically significant to determine how genetic alterations contribute to differentiating oligometastatic from polymetastatic NSCLC. Moreover, biomarker-driven treatment decisions, including ctDNA monitoring, could help refine therapy selection and improve patient outcomes.


Conclusions

Oligometastatic disease remains an evolving field, and a consensus on the optimal treatment approach has yet to be established. In oligometastatic NSCLC with targetable mutations, active management combining systemic treatment and local therapy can improve clinical outcomes. Accounting for co-mutations is important to enhance personalized treatment, and when possible obtaining sufficient pathological samples may help clinicians to create better treatment plans.


Acknowledgments

None.


Footnote

Reporting Checklist: The authors have completed the Narrative Review reporting checklist. Available at https://tlcr.amegroups.com/article/view/10.21037/tlcr-2024-1121/rc

Peer Review File: Available at https://tlcr.amegroups.com/article/view/10.21037/tlcr-2024-1121/prf

Funding: This research was supported by the Nano & Material Technology Development Program through the National Research Foundation of Korea (NRF) funded by Ministry of Science and ICT (RS-2024-00403376).

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://tlcr.amegroups.com/article/view/10.21037/tlcr-2024-1121/coif). The authors have no conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. Postmus PE, Kerr KM, Oudkerk M, et al. Early and locally advanced non-small-cell lung cancer (NSCLC): ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol 2017;28:iv1-iv21. [Crossref] [PubMed]
  2. Rusthoven KE, Hammerman SF, Kavanagh BD, et al. Is there a role for consolidative stereotactic body radiation therapy following first-line systemic therapy for metastatic lung cancer? A patterns-of-failure analysis. Acta Oncol 2009;48:578-83. [Crossref] [PubMed]
  3. Hellman S, Weichselbaum RR. Oligometastases. J Clin Oncol 1995;13:8-10. [Crossref] [PubMed]
  4. Lievens Y, Guckenberger M, Gomez D, et al. Defining oligometastatic disease from a radiation oncology perspective: An ESTRO-ASTRO consensus document. Radiother Oncol 2020;148:157-66. [Crossref] [PubMed]
  5. Vietti Violi N, Hajri R, Haefliger L, et al. Imaging of Oligometastatic Disease. Cancers (Basel) 2022;14:1427. [Crossref] [PubMed]
  6. Iyengar P, Wardak Z, Gerber DE, et al. Consolidative Radiotherapy for Limited Metastatic Non-Small-Cell Lung Cancer: A Phase 2 Randomized Clinical Trial. JAMA Oncol 2018;4:e173501. [Crossref] [PubMed]
  7. Gomez DR, Tang C, Zhang J, et al. Local Consolidative Therapy Vs. Maintenance Therapy or Observation for Patients With Oligometastatic Non-Small-Cell Lung Cancer: Long-Term Results of a Multi-Institutional, Phase II, Randomized Study. J Clin Oncol 2019;37:1558-65. [Crossref] [PubMed]
  8. Rim CH, Cho WK, Lee JH, et al. Role of Local Treatment for Oligometastasis: A Comparability-Based Meta-Analysis. Cancer Res Treat 2022;54:953-69. [Crossref] [PubMed]
  9. Chang JY, Verma V. Optimize Local Therapy for Oligometastatic and Oligoprogressive Non-Small Cell Lung Cancer to Enhance Survival. J Natl Compr Canc Netw 2022;20:531-9. [Crossref] [PubMed]
  10. Blake-Cerda M, Lozano-Ruíz F, Maldonado-Magos F, et al. Consolidative stereotactic ablative radiotherapy (SABR) to intrapulmonary lesions is associated with prolonged progression-free survival and overall survival in oligometastatic NSCLC patients: A prospective phase 2 study. Lung Cancer 2021;152:119-26. [Crossref] [PubMed]
  11. Bauml JM, Mick R, Ciunci C, et al. Pembrolizumab After Completion of Locally Ablative Therapy for Oligometastatic Non-Small Cell Lung Cancer: A Phase 2 Trial. JAMA Oncol 2019;5:1283-90. [Crossref] [PubMed]
  12. Guckenberger M, Lievens Y, Bouma AB, et al. Characterisation and classification of oligometastatic disease: a European Society for Radiotherapy and Oncology and European Organisation for Research and Treatment of Cancer consensus recommendation. Lancet Oncol 2020;21:e18-28. [Crossref] [PubMed]
  13. Wang XS, Bai YF, Verma V, et al. Randomized Trial of First-Line Tyrosine Kinase Inhibitor With or Without Radiotherapy for Synchronous Oligometastatic EGFR-Mutated Non-Small Cell Lung Cancer. J Natl Cancer Inst 2023;115:742-8. [Crossref] [PubMed]
  14. Chan OSH, Lam KC, Li JYC, et al. ATOM: A phase II study to assess efficacy of preemptive local ablative therapy to residual oligometastases of NSCLC after EGFR TKI. Lung Cancer 2020;142:41-6. [Crossref] [PubMed]
  15. Xu Q, Zhou F, Liu H, et al. Consolidative Local Ablative Therapy Improves the Survival of Patients With Synchronous Oligometastatic NSCLC Harboring EGFR Activating Mutation Treated With First-Line EGFR-TKIs. J Thorac Oncol 2018;13:1383-92. [Crossref] [PubMed]
  16. Elamin YY, Gomez DR, Antonoff MB, et al. Local Consolidation Therapy (LCT) After First Line Tyrosine Kinase Inhibitor (TKI) for Patients With EGFR Mutant Metastatic Non-small-cell Lung Cancer (NSCLC). Clin Lung Cancer 2019;20:43-7. [Crossref] [PubMed]
  17. Jiang T, Chu Q, Wang H, et al. EGFR-TKIs plus local therapy demonstrated survival benefit than EGFR-TKIs alone in EGFR-mutant NSCLC patients with oligometastatic or oligoprogressive liver metastases. Int J Cancer 2019;144:2605-12. [Crossref] [PubMed]
  18. Zeng Y, Ni J, Yu F, et al. The value of local consolidative therapy in Osimertinib-treated non-small cell lung cancer with oligo-residual disease. Radiat Oncol 2020;15:207. [Crossref] [PubMed]
  19. Gandhi S. 20 Phase II randomized study of osimertinib (OSI) with or without local consolidative therapy (LCT) for metastatic EGFR mutant non-small cell lung cancer (NSCLC): Analysis of adverse events (AEs). J Thorac Oncol 2023;18:S36.
  20. Iyengar P, All S, Berry MF, et al. Treatment of Oligometastatic Non-Small Cell Lung Cancer: An ASTRO/ESTRO Clinical Practice Guideline. Pract Radiat Oncol 2023;13:393-412. [Crossref] [PubMed]
  21. Kroeze SGC, Pavic M, Stellamans K, et al. Metastases-directed stereotactic body radiotherapy in combination with targeted therapy or immunotherapy: systematic review and consensus recommendations by the EORTC-ESTRO OligoCare consortium. Lancet Oncol 2023;24:e121-32. [Crossref] [PubMed]
  22. Hendriks LE, Kerr KM, Menis J, et al. Oncogene-addicted metastatic non-small-cell lung cancer: ESMO Clinical Practice Guideline for diagnosis, treatment and follow-up. Ann Oncol 2023;34:339-57. [Crossref] [PubMed]
  23. National Comprehensive Cancer Network. NCCN guidelines non-small cell lung cancer. 2024. Available online: https://www.nccn.org/guidelines/guidelines-detail?category=1&id=1450
  24. National Library of Medicine. OMEGA, Local Ablative Therapy in Oligometastatic NSCLC (OMEGA) (NCT03827577) 2020. Available online: https://clinicaltrials.gov/study/NCT03827577?cond=NCT03827577&rank=1
  25. National Library of Medicine. The Optimal Intervention Time of Radiotherapy for Oligometastatic Stage IV Non-small Cell Lung Cancer (NSCLC) (OITROLC) 2020. Available online: https://clinicaltrials.gov/study/NCT02076477?a=5
  26. Iyengar P, Hu C, Gomez DR, et al. NRG-LU002: Randomized phase II/III trial of maintenance systemic therapy versus local consolidative therapy (LCT) plus maintenance systemic therapy for limited metastatic non-small cell lung cancer (NSCLC). J Clin Oncol 2024;42:8506.
  27. Conibear J, Chia B, Ngai Y, et al. Study protocol for the SARON trial: a multicentre, randomised controlled phase III trial comparing the addition of stereotactic ablative radiotherapy and radical radiotherapy with standard chemotherapy alone for oligometastatic non-small cell lung cancer. BMJ Open 2018;8:e020690. [Crossref] [PubMed]
  28. Thureau S, Marchesi V, Vieillard MH, et al. Efficacy of extracranial stereotactic body radiation therapy (SBRT) added to standard treatment in patients with solid tumors (breast, prostate and non-small cell lung cancer) with up to 3 bone-only metastases: study protocol for a randomised phase III trial (STEREO-OS). BMC Cancer 2021;21:117. [Crossref] [PubMed]
  29. Ho C, Soon YY, Nichol A, et al. Osimertinib with or without SRS for Brain Metastases from EGFRm NSCLC: Pooled Analysis of Two RCTs. J Thorac Oncol 2024;19:S28-9.
  30. Tsai CJ, Yang JT, Shaverdian N, et al. Standard-of-care systemic therapy with or without stereotactic body radiotherapy in patients with oligoprogressive breast cancer or non-small-cell lung cancer (Consolidative Use of Radiotherapy to Block [CURB] oligoprogression): an open-label, randomised, controlled, phase 2 study. Lancet 2024;403:171-82.
  31. Schellenberg D, Gabos Z, Duimering A, et al. Stereotactic Ablative Radiation for Oligoprogressive Cancers: Results of the Randomized Phase 2 STOP Trial. Int J Radiat Oncol Biol Phys 2025;121:28-38. [Crossref] [PubMed]
  32. National Library of Medicine. Osimertinib, Surgery, and Radiation Therapy in Treating Patients With Stage IIIB or IV Non-small Cell Lung Cancer With EGFR Mutations, NORTHSTAR Study (NCT03410043). 2024. Available online: https://clinicaltrials.gov/study/NCT03410043?cond=NCT03410043&rank=1
  33. National Library of Medicine. Stereotactic Body Radiotherapy for the Treatment of OPD (HALT). 2023. Available online: https://clinicaltrials.gov/study/NCT03256981
  34. National Library of Medicine. CORE - Standard of care +/- stereotactic body radiotherapy for oligometastases - primary results, ESTRO 2023, Late-breaking Papers 2023. Available online: https://www.estro.org//Congresses/ESTRO-2023/1418/late-breakingpapers/13175/core-standardofcare--stereotacticbodyradiotherapyf
  35. Milano MT, Salama JK, Chmura SJ. Should We Target Oligometastatic EGFR-Mutated Non-Small Cell Lung Cancer With Radiotherapy Before Administering Targeted Systemic Therapy? J Natl Cancer Inst 2023;115:605-7. [Crossref] [PubMed]
  36. Semenkovich NP, Samson P, Badiyan SN, et al. Circulating Tumor DNA for Early Risk Stratification of Oligometastatic Lung Cancer. Int J Radiat Oncol Biol Phys 2023;117:S29-S30.
  37. Lebow ES, Murciano-Goroff Y, Razavi P, et al. Circulating Tumor DNA as a Biomarker in Oligometastatic Non-small Cell Lung Cancer. Int J Radiat Oncol Biol Phys 2020;108:S174.
  38. Tachibana T, Matsuura Y, Ninomiya H, et al. Optimal Treatment Strategy for Oligo-Recurrence Lung Cancer Patients with Driver Mutations. Cancers (Basel) 2024;16:464. [Crossref] [PubMed]
  39. Schoenmaekers JJAO, Paats MS, Dingemans AC, et al. Central nervous system metastases and oligoprogression during treatment with tyrosine kinase inhibitors in oncogene-addicted non-small cell lung cancer: how to treat and when? Transl Lung Cancer Res 2020;9:2599-617. [Crossref] [PubMed]
  40. Schmid S, Klingbiel D, Aeppli S, et al. Patterns of progression on osimertinib in EGFR T790M positive NSCLC: A Swiss cohort study. Lung Cancer 2019;130:149-55. [Crossref] [PubMed]
  41. Weickhardt AJ, Scheier B, Burke JM, et al. Local ablative therapy of oligoprogressive disease prolongs disease control by tyrosine kinase inhibitors in oncogene-addicted non-small-cell lung cancer. J Thorac Oncol 2012;7:1807-14. [Crossref] [PubMed]
  42. Qiu B, Liang Y, Li Q, et al. Local Therapy for Oligoprogressive Disease in Patients With Advanced Stage Non-small-cell Lung Cancer Harboring Epidermal Growth Factor Receptor Mutation. Clin Lung Cancer 2017;18:e369-73. [Crossref] [PubMed]
  43. Li L, Luo S, Lin H, et al. Correlation between EGFR mutation status and the incidence of brain metastases in patients with non-small cell lung cancer. J Thorac Dis 2017;9:2510-20. [Crossref] [PubMed]
  44. Kim Y, Sudo A, Oyama R, et al. Isolated Central Nervous System Progression During Systemic Treatment With Brentuximab Vedotin Monotherapy in a Pediatric Patient With Recurrent ALK-negative Anaplastic Large Cell Lymphoma. J Pediatr Hematol Oncol 2021;43:e864-6. [Crossref] [PubMed]
  45. Hubbeling H, Choudhury N, Flynn J, et al. Outcomes With Local Therapy and Tyrosine Kinase Inhibition in Patients With ALK/ROS1/RET-Rearranged Lung Cancers. JCO Precis Oncol 2022;6:e2200024. [Crossref] [PubMed]
  46. Marin-Acevedo JA, Pellini B, Kimbrough EO, et al. Treatment Strategies for Non-Small Cell Lung Cancer with Common EGFR Mutations: A Review of the History of EGFR TKIs Approval and Emerging Data. Cancers (Basel) 2023;15:629. [Crossref] [PubMed]
  47. Hartmaier RJ, Markovets AA, Ahn MJ, et al. Osimertinib + Savolitinib to Overcome Acquired MET-Mediated Resistance in Epidermal Growth Factor Receptor-Mutated, MET-Amplified Non-Small Cell Lung Cancer: TATTON. Cancer Discov 2023;13:98-113. [Crossref] [PubMed]
  48. Saw SPL, Le X, Hendriks LEL, et al. New Treatment Options for Patients With Oncogene-Addicted Non-Small Cell Lung Cancer Focusing on EGFR-Mutant Tumors. Am Soc Clin Oncol Educ Book 2024;44:e432516. [Crossref] [PubMed]
  49. Gomatou G, Syrigos N, Kotteas E. Osimertinib Resistance: Molecular Mechanisms and Emerging Treatment Options. Cancers (Basel) 2023;15:841. [Crossref] [PubMed]
  50. Yap TA, Macklin-Doherty A, Popat S. Continuing EGFR inhibition beyond progression in advanced non-small cell lung cancer. Eur J Cancer 2017;70:12-21. [Crossref] [PubMed]
  51. National Comprehensive Cancer Network. National Comprehensive Cancer Network clinical practice guidelines in oncology (NCCN guidelines). Non-small cell lung cancer, version11.2024. 2024. Available online: https://www.nccn.org/professionals/physician_gls/pdf/nscl.pdf
  52. Rossi A, Galetta D. Systemic Therapy for Oligoprogression in Patients with Metastatic NSCLC Harboring Activating EGFR Mutations. Cancers (Basel) 2022;14:832. [Crossref] [PubMed]
  53. Choudhury NJ, Marra A, Sui JSY, et al. Molecular Biomarkers of Disease Outcomes and Mechanisms of Acquired Resistance to First-Line Osimertinib in Advanced EGFR-Mutant Lung Cancers. J Thorac Oncol 2023;18:463-75. [Crossref] [PubMed]
  54. Lim JU, Kang HS, Moon MH, et al. PD-L1 expression from surgically resected lung tumors predictive of early progression in patients previously treated with targeted therapy for initially unresectable non-small cell lung cancer. Transl Lung Cancer Res 2024;13:1463-80. [Crossref] [PubMed]
  55. Werner RS, Rechsteiner M, Moch H, et al. Genetic profiles of oligometastatic non-small-cell lung cancer and corresponding brain metastases. Eur J Cardiothorac Surg 2024;65:ezae217. [Crossref] [PubMed]
  56. Liao R, Chen K, Li J, et al. Genomic Characteristics and the Potential Clinical Implications in Oligometastatic Non-Small Cell Lung Cancer. Cancer Res Treat 2023;55:814-31. [Crossref] [PubMed]
  57. Lim JU, Lee E, Lee SY, et al. Current literature review on the tumor immune micro-environment, its heterogeneity and future perspectives in treatment of advanced non-small cell lung cancer. Transl Lung Cancer Res 2023;12:857-76. [Crossref] [PubMed]
  58. Song Z, Yang L, Zhou Z, et al. Genomic profiles and tumor immune microenvironment of primary lung carcinoma and brain oligo-metastasis. Cell Death Dis 2021;12:106. [Crossref] [PubMed]
  59. Fu R, Xiong YY, Cai M, et al. 49P Perioperative circulating tumor DNA redefines induced oligometastatic non-small cell lung cancer at the molecular level. ESMO Open 2024;9:102628.
  60. Wang X, Zeng Z, Cai J, et al. Efficacy and acquired resistance for EGFR-TKI plus thoracic SBRT in patients with advanced EGFR-mutant non-small-cell lung cancer: a propensity-matched retrospective study. BMC Cancer 2021;21:482. [Crossref] [PubMed]
  61. Chhouri H, Alexandre D, Grumolato L. Mechanisms of Acquired Resistance and Tolerance to EGFR Targeted Therapy in Non-Small Cell Lung Cancer. Cancers (Basel) 2023;15:504. [Crossref] [PubMed]
  62. Kim KH, Yoon S, Ahn HK, et al. A Multicenter Two-arm, Phase II Trial Assessing the Safety and Efficacy of First-line Lazertinib and Locally Ablative Radiotherapy in Patients With Synchronous Oligo-metastatic EGFR-mutant Non-small Cell Lung Cancer (ABLATE, KCSG-LU21-11). Clin Lung Cancer 2022;23:e536-9. [Crossref] [PubMed]
  63. Ohtaki Y, Shimizu K, Suzuki H, et al. Salvage surgery for non-small cell lung cancer after tyrosine kinase inhibitor treatment. Lung Cancer 2021;153:108-16. [Crossref] [PubMed]
  64. Suzuki S, Asakura K, Okui M, et al. Prognostic factors affecting survival in patients with non-small cell lung cancer treated with salvage surgery after drug therapy: a multi-institutional retrospective study. World J Surg Oncol 2023;21:290. [Crossref] [PubMed]
  65. Chien YN, Lin YC, Chang CL, et al. Thoracic surgery improved overall survival in patients with stage IIIB-IV epidermal growth factor receptor-mutant lung adenocarcinoma who received and responded to tyrosine kinase inhibitor treatment. Lung Cancer 2021;162:29-35. [Crossref] [PubMed]
  66. Dunne EG, Fick CN, Tan KS, et al. Lung resection after initial nonoperative treatment for non-small cell lung cancer. J Thorac Cardiovasc Surg 2024;168:364-373.e10. [Crossref] [PubMed]
  67. Joosten PJM, de Langen AJ, van der Noort V, et al. The role of surgery in the treatment of oligoprogression after systemic treatment for advanced non-small cell lung cancer. Lung Cancer 2021;161:141-51. [Crossref] [PubMed]
  68. Lin MW, Yu SL, Hsu YC, et al. Salvage Surgery for Advanced Lung Adenocarcinoma After Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitor Treatment. Ann Thorac Surg 2023;116:111-9. [Crossref] [PubMed]
  69. Wu Y, Song Y, Wang R, et al. Molecular mechanisms of tumor resistance to radiotherapy. Mol Cancer 2023;22:96. [Crossref] [PubMed]
  70. Zhou T, Zhang LY, He JZ, et al. Review: Mechanisms and perspective treatment of radioresistance in non-small cell lung cancer. Front Immunol 2023;14:1133899. [Crossref] [PubMed]
  71. Bernhard EJ, Stanbridge EJ, Gupta S, et al. Direct evidence for the contribution of activated N-ras and K-ras oncogenes to increased intrinsic radiation resistance in human tumor cell lines. Cancer Res 2000;60:6597-600.
  72. Mak RH, Hermann G, Lewis JH, et al. Outcomes by tumor histology and KRAS mutation status after lung stereotactic body radiation therapy for early-stage non-small-cell lung cancer. Clin Lung Cancer 2015;16:24-32. [Crossref] [PubMed]
  73. Zhu DQ, Liu Y, Yu ZJ, et al. The Diverse Analysis Identifies Mutated KRAS Associated With Radioresistance in Non-Small Cell Lung Cancer. World J Oncol 2022;13:84-95. [Crossref] [PubMed]
  74. Cassidy RJ, Zhang X, Patel PR, et al. Next-generation sequencing and clinical outcomes of patients with lung adenocarcinoma treated with stereotactic body radiotherapy. Cancer 2017;123:3681-90. [Crossref] [PubMed]
  75. Binkley MS, Jeon YJ, Nesselbush M, et al. KEAP1/NFE2L2 Mutations Predict Lung Cancer Radiation Resistance That Can Be Targeted by Glutaminase Inhibition. Cancer Discov 2020;10:1826-41. [Crossref] [PubMed]
  76. Kinslow CJ, Kumar P, Cai LL, et al. NRF2-pathway mutations predict radioresistance in non-small cell lung cancer. Transl Lung Cancer Res 2022;11:1510-3. [Crossref] [PubMed]
  77. Pustovalova M, Alhaddad L, Smetanina N, et al. The p53-53BP1-Related Survival of A549 and H1299 Human Lung Cancer Cells after Multifractionated Radiotherapy Demonstrated Different Response to Additional Acute X-ray Exposure. Int J Mol Sci 2020;21:3342. [Crossref] [PubMed]
  78. Gurtner K, Kryzmien Z, Koi L, et al. Radioresistance of KRAS/TP53-mutated lung cancer can be overcome by radiation dose escalation or EGFR tyrosine kinase inhibition in vivo. Int J Cancer 2020;147:472-7. [Crossref] [PubMed]
  79. Lin SH, Zhang J, Giri U, et al. A high content clonogenic survival drug screen identifies mek inhibitors as potent radiation sensitizers for KRAS mutant non-small-cell lung cancer. J Thorac Oncol 2014;9:965-73. [Crossref] [PubMed]
  80. Kim TW, Hong DW, Kang CM, et al. A novel PPARɣ ligand, PPZ023, overcomes radioresistance via ER stress and cell death in human non-small-cell lung cancer cells. Exp Mol Med 2020;52:1730-43. [Crossref] [PubMed]
  81. van Aken E, Branco DM, De Ruysscher D, et al. 1906P ESMO-ESTRO consensus recommendations regarding the safety of combining radiotherapy with targeted agents or immunotherapy. Ann Oncol 2024;35:S1110-1.
  82. Lim JU. Management of Oligometastasis and Oligoprogression in Patients with Epidermal Growth Factor Receptor Mutation-Positive NSCLC in the Era of Third-Generation Tyrosine Kinase Inhibitors. Clin Lung Cancer 2021;22:e786-92. [Crossref] [PubMed]
  83. Lamba N, Cagney DN, Catalano PJ, et al. A genomic score to predict local control among patients with brain metastases managed with radiation. Neuro Oncol 2023;25:1815-27. [Crossref] [PubMed]
  84. Tsai CJ, Yang JT, Guttmann DM, et al. Final Analysis of Consolidative Use of Radiotherapy to Block (CURB) Oligoprogression Trial - A Randomized Study of Stereotactic Body Radiotherapy for Oligoprogressive Metastatic Lung and Breast Cancers. Int J Radiat Oncol Biol Phys 2022;114:1061.
  85. Tang C, Lee WC, Reuben A, et al. Immune and Circulating Tumor DNA Profiling After Radiation Treatment for Oligometastatic Non-Small Cell Lung Cancer: Translational Correlatives from a Mature Randomized Phase II Trial. Int J Radiat Oncol Biol Phys 2020;106:349-57. [Crossref] [PubMed]
  86. Semenkovich NP, Badiyan SN, Samson PP, et al. Pre-radiotherapy ctDNA liquid biopsy for risk stratification of oligometastatic non-small cell lung cancer. NPJ Precis Oncol 2023;7:100. [Crossref] [PubMed]
Cite this article as: Lim JU, Jang AL, Lee J, Park SY, An TJ, Sa YJ, Kim HR, Kim TJ, Kim BH, Park CK. Impacts of co-mutations in oligometastatic and oligoprogressive non-small cell lung cancer with EGFR/ALK mutations—a narrative review of the current literature. Transl Lung Cancer Res 2025;14(5):1848-1861. doi: 10.21037/tlcr-2024-1121

Download Citation