59 STROKOVNI ČLANEK Liquid biopsy – a diagnostic and therapeutic tool in the treatment of non-small cell lung cancer Copyright (c) 2023 Slovenian Medical Journal. This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License. Liquid biopsy – a diagnostic and therapeutic tool in the treatment of non-small cell lung cancer Tekočinska biopsija – diagnostični in terapevtski pripomoček pri zdravljenju nedrobnoceličnega raka pljuč Maša Majcen,1,2 Aleš Rozman,2,3 Izidor Kern2,3 Abstract Lung cancer is one of the most prevalent cancers and the leading cause of cancer mortality worldwide. The past decade has brought important progress in drug treatments by discovering the driver mutations. The evolution of targeted on- cological treatments directed to the biological properties of lung cancer in the individual patient has led to a significant increase in survival. During treatment, new mutations accumulate in the tumour, which prevents the long-term success of the therapy. Liquid biopsy is a method that has established itself in recent years as a less invasive diagnostic procedure that allows monitoring the response to treatment and identifying the mechanisms of resistance. The circulating tumor DNA is the most prevalent biomarker in lung cancer, but research on other biomarkers is also active. In this review article, we present the use of liquid biopsy in the clinical treatment of patients with non-small cell lung cancer. Its use is increas- ingly recognized in early detection of lung cancer, identifying resistant mutations, potential assessment of disease burden, and longitudinal monitoring. Izvleček Rak pljuč je ena najbolj pogostih in smrtonosnih rakavih bolezni na svetu. V zadnjih 10 letih je z odkritjem vodilnih muta- cij prišlo do velikega napredka pri zdravljenju. Zdravljenje postaja vse bolj prilagojeno bolniku oz. biološkim lastnostim raka, proti katerim so usmerjena tarčna onkološka zdravila, ki so bolnikom z napredovalo rakavo boleznijo pomembno podaljšala preživetje. Med tarčnim zdravljenjem se v tumorju razvijajo nove genetske spremembe, ki vodijo v odpornost tarč v rakavem tkivu, s čimer dolgoročno omejujejo uspešnost zdravljenja. Tekočinska biopsija (angl. liquid biopsy) je metoda, ki se je v zadnjih letih uveljavila kot manj invazivni diagnostični postopek, ki omogoča spremljanje odziva na zdravljenje in prepoznavanje mehanizmov odpornosti. Pri raku pljuč se najbolj uveljavlja vloga cirkulirajoče tumorske DNK (ctDNK), aktivne pa so raziskave tudi drugih bioloških označevalcev. V preglednem članku predstavljamo uporabo 1 General hospital Celje, Celje, Slovenia 2 Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia 3 University Clinic of respiratory and allergic diseases Golnik, Golnik, Slovenia Correspondence / Korespondenca: Maša Majcen, e: masa.majcen@gmail.com Key words: lung cancer; liquid biopsy; circulating tumor DNA; PCR; NGS Ključne besede: rak pljuč; tekočinska biopsija; cirkulirajoča tumorska DNK; PCR; NGS Received / Prispelo: 3. 8. 2021 | Accepted / Sprejeto: 2. 3. 2022 Cite as / Citirajte kot: Majcen M, Rozman A, Kern I. Liquid biopsy – a diagnostic and therapeutic tool in the treatment of non-small cell lung cancer. Zdrav Vestn. 2023;92(1–2):59–69. DOI: https://doi.org/10.6016/ZdravVestn.3294 eng slo element en article-lang 10.6016/ZdravVestn.3294 doi 3.8.2021 date-received 2.3.2022 date-accepted Respiratory system Dihala discipline Professional article Strokovni članek article-type Liquid biopsy – a diagnostic and therapeutic tool in the treatment of non-small cell lung cancer Tekočinska biopsija – diagnostični in terapevtski pripomoček pri zdravljenju nedrobnoceličnega raka pljuč article-title Liquid biopsy Tekočinska biopsija alt-title lung cancer, liquid biopsy, circulating tumor DNA, PCR, NGS rak pljuč, tekočinska biopsija, cirkulirajoča tu- morska DNK, PCR, NGS kwd-group The authors declare that there are no conflicts of interest present. Avtorji so izjavili, da ne obstajajo nobeni konkurenčni interesi. conflict year volume first month last month first page last page 2023 92 1 2 59 69 name surname aff email Maša Majcen 1,2 masa.majcen@gmail.com name surname aff Aleš Rozman 2,3 Izidor Kern 2,3 eng slo aff-id General hospital Celje, Celje, Slovenia Splošna bolnišnica Celje, Celje, Slovenija 1 Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia Medicinska fakulteta Ljubljana, Univerza v Ljubljani, Ljubljana, Slovenija 2 University Clinic of respiratory and allergic diseases Golnik, Golnik, Slovenia Univerzitetna klinika za pljučne bolezni in alergijo Golnik, Golnik, Slovenija 3 Slovenian Medical Journallovenian Medical Journal 60 DIHALA Zdrav Vestn | January – February 2023 | Volume 92 | https://doi.org/10.6016/ZdravVestn.3294 1 Introduction In recent years, the development of medicine has been directed towards patient-focused, personalized treatment. It is an individual approach to disease pre- vention and treatment, based on genetic and molecular diversity and the influence of the environment on the individual’s health (1). The greatest progress can be seen in the field of oncology, which has been one of the most active branches of medicine in terms of research in the last decade. Cancer is a genetic disease that contains a patient-specific and unique mutation profile. With the development of increasingly sensitive, specific and af- fordable technologies for the isolation and analysis of a tumour’s genetic material, the role of personalized treat- ment is increasingly included in oncology treatment guidelines worldwide. With the discovery of mutations and thus new possibilities for effective targeted drugs, even in the treatment of patients with lung cancer, the successes of the personalized approach are already visi- ble with prolongation of survival (2). 2 Lung cancer Lung cancer is one of the most common and deadly cancers worldwide (3). In Slovenia, lung cancer ranks 3rd among cancer types in men and 4th in women, with the proportion of female cancer patients still increasing (4). It is important to suspect and diagnose lung cancer as soon as possible (5). Despite medical progress, in most cases, the disease is detected at an advanced stage with subsequent poor survival and limited curative treatment options. In the last 10 years, with the discovery of driv- er mutations, significant progress has been made with treatment, which is becoming more and more tailored to the biological properties of the patient’s cancer (6,7). Lung cancer is a heterogeneous disease with sever- al histological subtypes, according to the World Health Organization classification (8). Due to differences in treatment methods, the division into small cell and non- small cell lung cancer, which includes adenocarcinoma and squamous cell carcinoma, is particularly important. Due to the emergence of many targeted treatments, an active and targeted search for driver mutations is recom- mended in lung adenocarcinoma. In patients with small tekočinske biopsije pri klinični obravnavi bolnikov z nedrobnoceličnim rakom pljuč, ki se kaže v možnosti zgodnjega odkrivanja raka pljuč, identifikaciji vodilnih in odpornih mutacij, potencialni oceni bremena bolezni ter dolgoročnega spremljanja bolnika. cell or squamous cell carcinoma, specific targeted ther- apy is not available, so routine identification of driver mutations is not necessary (9). 2.1 Lung adenocarcinoma The most studied and common mutation in non- small cell lung cancer is a mutation in the epidermal growth factor receptor (EGFR) gene; other types of mu- tations, such as ALK (anaplastic lymphoma kinase) re- arrangements, V600E point mutation in the BRAF gene (v-raf murine sarcoma viral oncogene homolog B1), rearrangements of ROS1 (c-ros oncogene 1), RET (re- arranged during transfection), NTRK (neurotrophic ty- rosine receptor kinase), and a skipping mutation in exon 14 of the MET gene are rarer (6,7,10). When patients are diagnosed with lung adenocarcinoma in Slovenia, EG- FR, KRAS (Kirsten rat sarcoma viral oncogene homo- log), and BRAF mutations and ALK, ROS 1, and NTRK rearrangements are determined in the tumour sample (9). As a result of targeted therapy, the survival of pa- tients with advanced cancer and their quality of life have significantly improved (2). During targeted therapy, new genetic changes which prevent the long-term success of this treatment develop in the tumour. Extensive re- search is being conducted around the world to identify the mechanisms of treatment resistance as precisely and quickly as possible with minimal invasiveness. The need for repeat tumour biopsies represents a limitation; a new method, the liquid biopsy, is becoming available for this purpose (11). 2.2 Squamous cell lung carcinoma At the oncologist’s request, the aforementioned mu- tations, though rarely present, are also tested in selected younger and non-smoking patients with squamous cell carcinoma and in other rare histological types of non- small cell lung cancer. As a rule, in patients with squa- mous cell carcinoma (and adenocarcinoma with nega- tive markers), due to the possibility of immunotherapy, only immunohistochemical staining for PD-L1 (pro- grammed death-ligand 1) is performed (9). 61 STROKOVNI ČLANEK Liquid biopsy – a diagnostic and therapeutic tool in the treatment of non-small cell lung cancer Legend: CT – computed tomography. Tkivna biopsija Tekočinska biopsija Sample type tumour tissue biopsy (bronchoscopy with biopsy, CT-guided needle biopsy, surgical biopsy) blood sample (less frequently pleural effusion, saliva, urine, ascites, cerebrospinal fluid) Procedure invasiveness Invasive Minimally invasive, non-invasive Procedure difficulty Difficult Less difficult Complications Rare, potentially life-threatening Very rare, less dangerous Tumour heterogeneity detection No Yes Patient burden Yes No Longitudinal follow-up of tumour characteristics No Yes Monitoring treatment response No Yes Sensitivity High High, but it differs depending on the authors and the type of method used Table 1: Comparison of tissue and liquid biopsy in lung cancer (13-26). 3 Liquid biopsy 3.1 A comparison of liquid and tissue biopsy A liquid biopsy is a procedure for the detection of cancer and molecular tumour characteristics in body fluids (12). Unlike traditional tissue biopsy using bron- choscopy or other minimally invasive procedures, a liquid biopsy is much less burdensome for patients. Around the world, different types of body fluids are used for analysis (e.g. pleural effusion, urine, sputum); most frequently, samples of peripheral blood or plas- ma are used (13-16). The role of liquid biopsy in the treatment of cancer is mainly in the identification of driver mutations, which allow for targeted treatment, monitoring the treatment response and success, as well as early detection of disease recurrence (17). Tissue biopsy is still the gold standard for cancer diagnosis, defining the tumour type and its character- istics. By tumour tissue sampling and processing, driv- er mutations have been discovered in recent decades, allowing for the development of targeted therapy. By taking tissue samples before and after the appearance of resistance to targeted therapy, researchers identi- fied the mechanisms of new, secondary mutations. Despite the advantages offered by tissue biopsy, it is still a time-consuming invasive method that requires an experienced team of specialists to perform the procedure and process the samples. There are several contraindications for invasive tissue biopsy, limiting its use in patients with advanced cancer. Complica- tions are also possible (18). Time required for the bi- opsy and intra- and intertumour heterogeneity are the most significant limitations. Accurate identification of mutations with a tissue biopsy is frequently not possi- ble at all, as due to tumour heterogeneity, the biopsy might not capture the entire set of mutations (19). Ad- ditionally, tissue biopsy does not allow for monitoring changes in the tumour’s genetic profile over time and the possible discovery of new mutations that pose an increased risk of treatment resistance (20). On the other hand, as a newer method, liquid bi- opsy is more accessible, reproducible, easier and faster to perform, either minimally or even non-invasively, and less burdensome for the patient. Compared to tis- sue biopsy, the risk of complications is negligible (18). Repeat sample collection is possible, giving immediate insight into the tumour’s changes and allowing longitu- dinal follow-up (21,22). Despite the many advantages that place liquid biopsy alongside the established tissue biopsy (Table 1), its use in everyday practice is gaining ground slowly, mainly due to dilemmas regarding the reliability of the test to prove the presence of a muta- tion and current high costs when using highly sensitive techniques. Liquid biopsy has already found a place in routine clinical practice in non-small cell lung cancer. In international recommendations (23), liquid biopsy is already the method of choice for the detection of 62 DIHALA Zdrav Vestn | January – February 2023 | Volume 92 | https://doi.org/10.6016/ZdravVestn.3294 treatment-resistant T790M mutation in patients with EGFR-mutated lung cancer treated with EGFR tyro- sine kinase inhibitors. The latest recommendations of the IASLC (International Association for the Study of Lung Cancer) include an expanded use of liquid biopsy in patients with lung cancer (24). The test’s sensitivi- ty and specificity undoubtedly depend on the size of the tumour mass, the presence of metastases and the type of method used to prove or determine mutations (25,26). 4 Types of liquid biopsy Liquid biopsy is based on the knowledge that can- cer cells present in the bloodstream are identical to primary tumour cells, which was first demonstrated by Australian scientist Thomas R. Ashworth in 1869 (27). It is estimated that thousands of cells are released into the circulation daily with an average half-life of 1–2.5 hours. As early as 1948, researchers Mandel and Metais described circulating free DNA fragments (cfDNA) in plasma, but the association between cfDNA concentra- tion and disease states was not proven until many years later (28). cfDNA is the total freely circulating DNA originating from all cells in the organism, healthy and diseased. With the improvement of techniques for the isolation of genetic material, it is now possible to detect and monitor various types of mutations that serve as specific biological markers for the presence of cancer in the bloodstream. 4.1 Molecular technologies In liquid biopsy, several types of molecular tech- niques are used, which are roughly divided into two groups. The first, the target group, represents a fast, more affordable, highly sensitive and specific technique that can identify even a small number of point mutations in one gene. This group includes methods based on the use of polymerase chain reaction (PCR). As narrowly focused methods, prior knowledge about the type of tumour and mutation is required. The second group includes high-performance methods that identify mutations in several genes (next-generation sequenc- ing, NGS). Since they are aimed at identifying a large part of the genome and can identify both specific and non-specific mutations, prior knowledge about the type of tumour and mutation is not required. Their biggest limitations are high cost, long procedure time, increased technology demands, and analysis (23,29). According to data from the literature, the sensitivity of cfDNA detection methods using NGS is between 75% and 90% with high tissue biopsy concordance (30). 4.2 Types of biological markers Liquid biopsy involves different methods of circu- lating marker detection. From a clinical point of view, among these markers, cfDNA, of which tumour DNA is a component, has been by far the most widely re- searched and used for genotyping solid cancer types, including lung cancer (mainly non-small cell lung cancer), where it is already included in routine man- agement (24). There are several other biomarkers, but their clinical use in lung cancer is rare and currently limited to research. 4.2.1 Circulating tumour DNA Circulating tumour DNA (ctDNA) is a subtype of cfDNA that is released from tumour cells and con- tains a transcript of extracellular tumour DNA (dou- ble-stranded or mitochondrial). It represents only a part of the total cfDNA, which also originates from non-tumour cells. It is found in body fluids (most of- ten in blood), into which it is released through various mechanisms, such as necrosis, apoptosis, active secre- tion, or cancer cell senescence (31). The concentration of ctDNA in body fluids is not constant. Its amount de- pends on several factors, mainly on the size of the tu- mour. The half-life of ctDNA is short (from a few min- utes to two hours), allowing monitoring the tumour’s current state. Its concentration is unfortunately low, therefore highly sensitive and advanced isolation and detection molecular techniques are needed to identify ctDNA (32). An additional obstacle with peripheral blood anal- ysis is cfDNA of non-cancerous origin, released from leukocytes. It dilutes the ctDNA present, further re- ducing the detection method’s sensitivity. In practice, adapted test tubes for sample collection and centri- fuging the sample as soon as possible after collection are used to solve this problem (33). In addition to false negative results, record false positive results of liquid biopsy with ctDNA are also possible. In healthy sub- jects, cancer-unrelated somatic mutations can arise from haematopoietic cells through the process of clon- al haematopoiesis, which complicates the interpre- tation of liquid biopsy results. These mutations most frequently arise from genes associated with haema- tological malignancies, but less frequently, mutations associated with lung cancer can also arise through this 63 STROKOVNI ČLANEK Liquid biopsy – a diagnostic and therapeutic tool in the treatment of non-small cell lung cancer process, e.g. KRAS mutations. In patients undergoing liquid biopsy for suspected lung cancer, a KRAS muta- tion can thus be detected despite the absence of lung cancer (34,35). Some specific tumour mutations, e.g. EGRF and BRAF, do not have a haematopoietic origin, so detection of a false mutation due to clonal haemato- poiesis is not possible. Despite excellent prospects, before the expected wider clinical use of ctDNA in oncology, the result’s credibility will require a precise standardization of the entire procedure from sample collection to final analy- sis (12). In the literature, studies that use targeted and even more affordable PCR methods are introducing ctDNA into clinical practice. Due to increasing sensi- tivity and gradually more affordable NGS methods, the use of these methods in clinical practice is expected to expand in the future. 4.2.2 Circulating tumour cells Circulating tumour cells (CTCs) are individual tu- mour cells that leave their site of origin (primary tu- mour or metastasis). They travel throughout the body to distant locations via the bloodstream (36). Blood is therefore a suitable sample for their identification, cancer diagnosis and cancer monitoring. CTCs already play an important role in the diagnosis of metastatic breast, prostate, and colon cancer, and they already have a possible role in lung cancer management (37- 39). Similar to ctDNA, their blood concentration is unfortunately low (0.1-10 CTC/mL whole blood), and the half-life is short (1-2.4 hours). Detection requires highly sensitive techniques that distinguish CTCs from other circulating cells based on their physical, mor- phological, and biological characteristics (40). 4.2.3 Other biological markers There are more types of biomarkers, but compared to ctDNA, they are currently used in lung cancer only for research purposes. Due to the extensive research on their possible use and predicted important future role in the treatment of lung cancer, they are only men- tioned in this article. Since they are not currently used in clinical practice, a detailed presentation is beyond the scope of this article. Various biological markers with possible roles in cancer development are used for research purposes. Exomes are small, 30–100 nm membrane-bound ves- icles that are produced by endocytosis or release from cells. They contain proteins and nucleic acids that can be transported between cells. Due to their lipid mem- brane, they are protected from degradation, can pass through the membrane, and persist in the bloodstream for longer periods. They play many different roles in the development of cancer: in carcinogenesis, the for- mation of metastases and the transfer of oncogenic factors between cells. The use of TEP (tumour educat- ed platelets), tumour-associated autoantibodies (TA- Ab) and microRNA is also mentioned (36,41). 5 Use of liquid biopsy in patients with lung cancer in clinical practice Liquid biopsy has established itself in clinical prac- tice in patients with lung cancer. With the discovery of critical molecular and cellular mechanisms of lung cancer development and the emergence of new target- ed therapy methods, new techniques have been devel- oped and are being used, particularly in the treatment of non-small cell lung cancer. Liquid biopsy has es- tablished itself as a method to identify specific EGFR mutations in patients who develop targeted treatment resistance. Due to its clinical availability and cost-ef- fectiveness, the current most widely used method is the detection of ctDNA with PCR, hence the reason for this article’s focus upon it. However, it is import- ant to emphasize that in recent years, the number of scientific publications investigating its use in patients with cancer other than lung cancer is rapidly increas- ing (Figure 1). We expect that in the coming years, the field of liquid biopsy will advance professionally and technologically, particularly with the expansion of the NGS method. 5.1 Early lung cancer detection Due to its non-invasiveness and potential low cost, liquid biopsy has shown its potential for use in screen- ing and early detection of lung cancer. Currently, the only recognized and recommended method for lung cancer screening is the use of low-dose computed to- mography (CT) with confirmatory tissue biopsy. Due to the low sensitivity and specificity for early cancer detection, ideas on improving screening with a com- bination of CT imaging and a blood test (liquid biop- sy) have appeared in the literature (42). According to data from the TRACERx study, due to early genomic and chemical changes, ctDNA should be present in the bloodstream 6-12 months before the first radiological signs of lung cancer, and the concentration of cfDNA is higher in cancer patients compared to healthy controls 64 DIHALA Zdrav Vestn | January – February 2023 | Volume 92 | https://doi.org/10.6016/ZdravVestn.3294 Figure 1: Graphic representation of the temporal distribution of search results in the PubMed digital database up to 2021. The search keywords used were »lung cancer« and »liquid biopsy« with synonyms. Se ar ch h its (p er 1 00 ,0 00 ) Year 1945 1949 1953 1957 1961 1965 1969 1973 1977 1981 1985 1989 1993 1997 2001 2005 2009 2013 2017 2021 0 5 10 15 20 Časovna razporeditev rezultatov iskanja z izbranimi ključnimi besedami v zbirki PubMed (številke podane na 100.000) and patients with benign lung changes (43,44). Ma- ny studies in recent years have been concerned with obtaining the most favourable biological markers for screening use, e.g. ctDNA, CTC, microRNA, exomes, proteins, etc. (45-47). One of the first studies, conduct- ed in 2014, to isolate CTCs from patients with chronic obstructive pulmonary disease, who represent a group at higher risk of developing lung cancer, was the first to show promising results for detecting patients at high risk of developing lung cancer with a normal CT scan result (48). Some similar follow-up studies have failed to demonstrate similar statistical reliability for predict- ing lung cancer risk (49,50). Currently, the combina- tion of a low-dose CT and liquid biopsy with CTC and/ or ctDNA cells is an important research topic for the early detection of lung cancer (studies by Spira’s group, Lecia et al) (51). A recent larger study by Chen et al, which included more than 120,000 healthy, asymp- tomatic individuals, highlighted the potential for early detection of multiple specific ctDNA methylations in 477 regions, which research suggests may be specific to certain solid tumours, including lung cancer. They reported correct identification of 88% of solid tumours (including lung cancer) demonstrated over a 4-year follow-up and 95% specificity for identifying the type of cancer (52). Despite high expectations, a problem arises when using ctDNA as a biological marker due to the low frequency of mutated alleles in early, cur- able forms of lung cancer, as their detection exceeds the sensitivity of the currently available methods (53). Today, liquid biopsy as a screening method does not yet seem viable from an organizational and cost point of view. False-negative tests are an additional problem; currently, it would be reasonable to use the test only in patients with risk factors for the development of lung cancer. Considering the extensive research into liquid biopsy, it is expected that the method will establish it- self as a new approach to disease management in the future. 5.2 Identifying mutations and treatment resistance Several different types of mutations have been iden- tified that participate in lung cancer development. Identifying mutations in cancer cells is key to select- ing an appropriate and effective targeted therapy. Ac- cording to the recommendations of the European and 65 STROKOVNI ČLANEK Liquid biopsy – a diagnostic and therapeutic tool in the treatment of non-small cell lung cancer American professional guidelines, treatment should be directed according to the detected resistance mecha- nism; before first-line treatment, a tissue biopsy is still in place today (54,55). It is particularly important to identify the mutations for which targeted treatment is available. The biggest limitation in the use of targeted therapy are secondary mutations resulting in resistance to primary treatment, which require a repeat biopsy to change treatment. According to data from Nosaki et al, a repeat lung tumour biopsy is not feasible in as many as 20% of patients with treatment resistance, and many studies also report insufficient or unrepresentative tu- mour tissue samples (56,57). Based on the favourable study results and the afore- mentioned problems with repeat tissue biopsies in the event of resistance, the European Medicines Agency (EMA) was in 2015 the first to issue an approval for the use of plasma ctDNA in lung cancer; a year later, the US Food and Drug Administration (FDA) did the same (58,59). Its use is currently limited for EGFR sta- tus determination in patients with newly diagnosed non-small cell lung cancer where tissue biopsy is not possible or in the event of resistance during treatment with primary or second-generation tyrosine kinase inhibitors (TKIs) for the detection of the point mu- tation in exon 20 T790M. This is the most common treatment-resistant mutation (proven in approximate- ly 50% of patients) and is an indication for treatment with third-generation TKIs. Two standardized, high- ly sensitive and specific mutation tests (Cobas EGFR mutations Test v2, Roche; Therascreen, Qiagen) based on the use of qPCR have been approved for detection (20,60-62). However, according to the AURA study, caution is required when interpreting the results of T790M mutation detection (63). In the study, they found in- consistencies between the presence of the mutation in tissue and liquid samples as a consequence of tumour heterogeneity. In case of a positive result (i.e. proven resistance mutation), the type of treatment should be changed. Additionally, due to low method sensitivity, it is necessary to take into account the possibility of false negative results, so another sample should be taken in the event of a negative result (64-66). In the case of EGFR-independent mutations, can- cer cells acquire an EGFR-independent alternative pathway for cancer cell survival and proliferation by acquiring new mutations, e.g. KRAS, HER2, MET, PI3KCA, etc. Transformation from non-small cell to small cell lung cancer is rarely seen. With the ex- pansion of advanced techniques in everyday clinical practice, ctDNA analysis with NGS with which we can identify a wider range of cancer cell mutations is becoming more and more popular. Since the publica- tion of the latest IASLC recommendations, a number of additional studies have emerged to support the use of broad-spectrum NGS analysis (24). It can be used to demonstrate the presence of other important muta- tions for which we have available targeted treatments, e.g. ALK (67) and ROS 1 (68) rearrangements, BRAF mutations (69), etc. In 2020, the ESMO expert group already recommended NGS for the detection of ge- netic alterations (EGFR, ALK, etc.) in advanced lung cancer in regular clinical practice (70). At the end of 2020, the FDA was the first to issue approval for two commercial NGS tests (Guardant360, FoundationOne Liquid CDx) (24). So far, the use of NGS worldwide is still limit- ed compared to PCR methods. However, its role will undoubtedly increase significantly in the future with lower costs and better availability. Despite the increas- ing role of liquid biopsy compared to tissue biopsy, it should be taken into account that ctDNA analysis does not detect the emergence of some other resistance mechanisms, e.g. histological transformations of can- cer tissue (71). 5.3 Assessment of disease burden and long- term monitoring of treatment response With a liquid biopsy, adequate treatment response, disease progression, and assessment of patient survival could be determined early. Studies examining liquid biopsy as a method for quantitative and qualitative monitoring of tumour burden and molecular profile are emerging in the literature. The concentration of ctDNA in plasma changes, and depends on the type of cancer, its location, size, and tumour blood supply, ergo monitoring trends and not the absolute concen- tration is more appropriate (72). Due to this variability, different concentrations of ctDNA can be found in pa- tients with the same tumour type and the same disease stage. Despite the variability, in one large Chinese mul- ticentre prospective cohort study from 2020, signifi- cantly shorter non-small cell lung cancer survival was reported in patients with higher pre-treatment ctDNA concentrations and mutations (73,74). This was associ- ated with a higher baseline disease burden. However, on the other hand, data can be found in the literature that baseline ctDNA concentrations do not predict re- sponse to treatment (75). With the introduction of a new drug, sequential 66 DIHALA Zdrav Vestn | January – February 2023 | Volume 92 | https://doi.org/10.6016/ZdravVestn.3294 References 1. Jain KK. Personalized medicine. Curr Opin Mol Ther. 2002;4(6):548-58. PMID: 12596356 2. Mok TS, Wu YL, Thongprasert S, Yang CH, Chu DT, Saijo N, et al. Gefitinib or carboplatin-paclitaxel in pulmonary adenocarcinoma. N Engl J Med. 2009;361(10):947-57. DOI: 10.1056/NEJMoa0810699 PMID: 19692680 3. Boloker G, Wang C, Zhang J. Updated statistics of lung and bronchus cancer in United States (2018). J Thorac Dis. 2018;10(3):1158-61. DOI: 10.21037/jtd.2018.03.15 PMID: 29708136 4. Onkološki Inštitut Ljubljana. Register raka Republike Slovenije in drugi registri. Ljubljana: Onkološki inštitut; 2018. 5. Ost DE, Yeung SC, Tanoue LT, Gould MK. Clinical and organizational factors in the initial evaluation of patients with lungcancer: Diagnosis and management of lung cancer. Chest. 2013;143:e121S-e141S. DOI: 10.1378/chest.12-2352 PMID: 23649435 6. Li T, Kung HJ, Mack PC, Gandara DR. Genotyping and genomic profiling of non-small-cell lung cancer: implications for currentand future therapies. J Clin Oncol. 2013;31(8):1039-49. DOI: 10.1200/JCO.2012.45.3753 PMID: 23401433 blood analyses could be used to monitor the response to treatment and rate of decline in ctDNA concentra- tions to assess treatment effectiveness. In their 2016 study, Lee et al used regular plasma samples to mon- itor the presence of EGFR mutation in patients from the initiation of EGFR TKI therapy and compared this with the clinical response. They found a statisti- cally significant difference in median progression-free survival in patients with undetectable EGFR mutation after EGFR TKI treatment compared with those with recurrence within two months of starting treatment (10.1 and 6.3 months, respectively, P=0.006) (76). Song et al also found a longer time to disease progres- sion and increased overall survival in patients with in- creased ctDNA clearance, regardless of the treatment type and regimen. In addition to evaluating the effectiveness of the treatment type, monitoring mutation levels would al- so allow early detection of treatment resistance emer- gence (24). In a Slovenian study that was conducted between 2014 and 2017, Kern et al studied the level of EGFR mutations in patients with non-small cell lung cancer who were treated with first-generation EGFR TKIs and compared changes in levels with clinical and radiological signs of disease progression. Patients were followed up with imaging after the first eight weeks of treatment and then every 16 weeks or earlier, if signs of disease progression were noted. Blood samples for EG- FR mutation testing in peripheral blood were collected at each follow-up visit and processed with a standard- ized qPCR Cobas EGFR assay. In 12 out of 35 patients, an increase in EGFR mutation levels was observed an average of 10 weeks before clinical and radiological signs of disease progression (77). According to recent data, the speed of response to treatment is a possible indicator of the long-term treat- ment success and indirectly affects the disease outcome. A recent Italian study reported a worse outcome in patients with a KRAS mutation who maintained high plasma ctDNA concentrations 3–4 weeks after treat- ment initiation. When ctDNA concentrations had in- creased, the risk for disease progression increased by as much as sevenfold (78). By identifying these patients, it could be possible to replace treatment with a more appropriate and effective faster (e.g. immunotherapy, chemotherapy). 6 Conclusion Liquid biopsy is a method that is slowly taking an important place in diagnosis and monitoring the re- sponse to lung cancer treatment. Although its role in the treatment of lung cancer is currently still limited, it is gradually establishing itself in global guidelines (55,56). Currently, ctDNA is already used in clinical practice in patients with non-small cell lung cancer for the primary detection of driver mutations (mainly EG- FR) and resistance mechanisms during targeted thera- py. The method also shows great promise in the early detection of cancer and in monitoring the response to treatment (24,55). Due to the increasing sensitivity and more affordable methods, the use of the NGS molec- ular method is expected to expand into clinical prac- tice, and other types of biological markers also show great potential in research (24). Further technological advancements and research are needed, to improve the reliability and applicability of the method in regular clinical practice, and above all, the standardization of the entire procedure is essential - from sample collec- tion to reporting of results and their evaluation. Conflict of interest None declared. 67 STROKOVNI ČLANEK Liquid biopsy – a diagnostic and therapeutic tool in the treatment of non-small cell lung cancer 7. Kris MG, Johnson BE, Berry LD, Kwiatkowski DJ, Iafrate AJ, Wistuba II, et al. Using multiplexed assays of oncogenic drivers in lung cancers to select targeted drugs. JAMA. 2014;311(19):1998-2006. DOI: 10.1001/ jama.2014.3741 PMID: 24846037 8. World Health Organization. IARC monographsx on the Evaluation of the Carcinogenic Risk of Chemicals to Humans. Geneva: WHO; 1986. 9. Boc N, Kern I, Rozman A, et al. Priporočila za obravnavo bolnikov s pljučnim rakom. Ljubljana: Onkološki inštitut; 2019. 10. Lindeman NI, Cagle PT, Aisner DL, Arcila ME, Beasley MB, Bernicker EH, et al. Updated molecular testing guideline for the selection of lung cancer patients fortreatment with targeted tyrosine kinase inhibitors: Guideline from the College of American Pathologists, the International Association for the Study of Lung Cancer,and the Association for Molecular Pathology. J Mol Diagn. 2018;20(2):129-59. DOI: 10.1016/j.jmoldx.2017.11.004 PMID: 29398453 11. Johann DJ, Steliga M, Shin IJ, Yoon D, Arnaoutakis K, Hutchins L, et al. Liquid biopsy and its role in an advanced clinical trial for lung cancer. Exp Biol Med (Maywood). 2018;243(3):262-71. DOI: 10.1177/1535370217750087 PMID: 29405770 12. Jesenko T, Grašič Kuhar C, Čemažar M. Tekočinska biopsija pri raku. Radiol Oncol. 2018;2:26-31. 13. Franovic A, Raymond VM, Erlander MG, Reckamp KL. Urine test for EGFR analysis in patients with non-small cell lung cancer. J Thorac Dis. 2017;9(S13):S1323-31. DOI: 10.21037/jtd.2017.06.144 PMID: 29184671 14. Ying S, Ke H, Ding Y, Liu Y, Tang X, Yang D, et al. Unique genomic profiles obtained from cerebrospinal fluid cell-free DNA of non-smallcell lung cancer patients with leptomeningeal metastases. Cancer Biol Ther. 2019;20(4):562-70. DOI: 10.1080/15384047.2018.1538614 PMID: 30395779 15. Song Z, Wang W, Li M, Liu J, Zhang Y. Cytological-negative pleural effusion can be an alternative liquid biopsy media fordetection of EGFR mutation in NSCLC patients. Lung Cancer. 2019;136:23-9. DOI: 10.1016/j. lungcan.2019.08.004 PMID: 31421258 16. Gu X, He J, Ji G. Combined use of circulating tumor cells and salivary mRNA to detect non-small-celllung cancer. Medicine (Baltimore). 2020;99(8):e19097. DOI: 10.1097/MD.0000000000019097 PMID: 32080083 17. Sequist LV, Neal JW. Personalized, genotype-directed therapy for advanced non-small cell lung cancer, UptoDate. Alphen aan den Rijn: Wolters Kluwer; 2020 [cited 2021 Apr 20]. Available from: https://www. uptodate.com/contents/personalized-genotype-directed-therapy-for- advanced-non-small-cell-lung-cancer. 18. Diaz LA, Bardelli A. Liquid biopsies: genotyping circulating tumor DNA. J Clin Oncol. 2014;32(6):579-86. DOI: 10.1200/JCO.2012.45.2011 PMID: 24449238 19. Wang N, Zhang X, Wang F, Zhang M, Sun B, Yin W, et al. The diagnostic accuracy of liquid biopsy in EGFR-mutated NSCLC: A systematic reviewand meta-analysis of 40 studies. SLAS Technol. 2021;26(1):42-54. DOI: 10.1177/2472630320939565 PMID: 32659150 20. Saarenheimo J, Eigeliene N, Andersen H, Tiirola M, Jekunen A. The value of liquid biopsies for guiding therapy decisions in non-small cell lungcancer. Front Oncol. 2019;9:129. DOI: 10.3389/fonc.2019.00129 PMID: 30891428 21. Russano M, Napolitano A, Ribelli G, Iuliani M, Simonetti S, Citarella F, et al. Liquid biopsy and tumor heterogeneity in metastatic solid tumors: the potentialityof blood samples. J Exp Clin Cancer Res. 2020;39(1):95. DOI: 10.1186/s13046-020-01601-2 PMID: 32460897 22. Lee JY, Qing X, Xiumin W, Yali B, Chi S, Bak SH, et al. Longitudinal monitoring of EGFR mutations in plasma predicts outcomes of NSCLC patientstreated with EGFR TKIs: Korean Lung Cancer Consortium (KLCC- 12-02). Oncotarget. 2016;7(6):6984-93. DOI: 10.18632/oncotarget.6874 PMID: 26755650 23. Rolfo C, Mack PC, Scagliotti GV, Baas P, Barlesi F, Bivona TG, et al. Liquid biopsy for advanced non-small cell lung cancer (NSCLC): A statement paper fromthe IASLC. J Thorac Oncol. 2018;13(9):1248-68. DOI: 10.1016/j. jtho.2018.05.030 PMID: 29885479 24. Rolfo C, Mack P, Scagliotti GV, Aggarwal C, Arcila ME, et al. Liquid biopsy for advanced non-small cell lung cancer: a consensus statement fromthe International Association for the Study of Lung Cancer (IASLC). J Thorac Oncol. 2021;16(10):1647-62. DOI: 10.1016/j.jtho.2021.06.017 PMID: 34246791 25. Sozzi G, Conte D, Leon M, Ciricione R, Roz L, Ratcliffe C, et al. Quantification of free circulating DNA as a diagnostic marker in lung cancer. J Clin Oncol. 2003;21(21):3902-8. DOI: 10.1200/JCO.2003.02.006 PMID: 14507943 26. Elazezy M, Joosse SA. Techniques of using circulating tumor DNA as a liquid biopsy component in cancer management. Comput Struct Biotechnol J. 2018;16:370-8. DOI: 10.1016/j.csbj.2018.10.002 PMID: 30364656 27. Ashworth TR. A case of cancer in which cells similar to those in the tumours were seen in the bloodafter death. Med J Aust. 1869;14:146147. 28. Ozkumur E, Shah AM, Ciciliano JC, Emmink BL, Miyamoto DT, Brachtel E, et al. Inertial focusing for tumor antigen-dependent and -independent sorting of rare circulatingtumor cells. Sci Transl Med. 2013;5(179). DOI: 10.1126/scitranslmed.3005616 PMID: 23552373 29. Progar V, Petrovič U. Vpliv parametrov sekvenciranja naslednje generacije na zanesljivost rezultatov v metagenomskih študijah. Informativa Medica Slovenica. 2013;18:1-8. 30. Yang SR, Schultheis AM, Yu H, Mandelker D, Ladanyi M, Büttner R. Precision medicine in non-small cell lung cancer: current applications and futuredirections. Semin Cancer Biol. 2020;S1044-579X(20):30164-4. DOI: 10.1016/j.semcancer.2020.07.00 PMID: 32730814 31. Fernandez-Cuesta L, Perdomo S, Avogbe PH, Leblay N, Delhomme TM, Gaborieau V, et al. Identification of circulating tumor dna for the early detection of small-cell lungcancer. EBioMedicine. 2016;10:117-23. DOI: 10.1016/j.ebiom.2016.06.032 PMID: 27377626 32. Corcoran RB, Chabner BA. Application of cell-free DNA analysis to cancer treatment. N Engl J Med. 2018;379(18):1754-65. DOI: 10.1056/ NEJMra1706174 PMID: 30380390 33. El Messaoudi S, Rolet F, Mouliere F, Thierry AR. Circulating cell free DNA: preanalytical considerations. Clin Chim Acta. 2013;424:222-30. DOI: 10.1016/j.cca.2013.05.022 PMID: 23727028 34. Boettcher S, Ebert BL. Clonal Hematopoiesis of Indeterminate Potential. J Clin Oncol. 2019;37(5):419-22. DOI: 10.1200/JCO.2018.79.3588 PMID: 30589599 35. Liu J, Chen X, Wang J, Zhou S, Wang CL, Ye MZ, et al. Biological background of the genomic variations of cf-DNA in healthy individuals. Ann Oncol. 2019;30(3):464-70. DOI: 10.1093/annonc/mdy513 PMID: 30475948 36. Trombetta D, Sparaneo A, Fabrizio FP, Muscarella LA. Liquid biopsy and NSCLC. Lung Cancer Manag. 2016;5(2):91-104. DOI: 10.2217/lmt-2016- 0006 PMID: 30643553 37. Bao-Caamano A, Rodriguez-Casanova A, Diaz-Lagares A. Epigenetics of circulating tumor cells in breast cancer. Adv Exp Med Biol. 2020;1220:117- 34. DOI: 10.1007/978-3-030-35805-1_8 PMID: 32304083 38. Pantel K, Hille C, Scher HI. Circulating tumor cells in prostate cancer: from discovery to clinical utility. Clin Chem. 2019;65(1):87-99. DOI: 10.1373/ clinchem.2018.287102 PMID: 30602476 39. Sundling KE, Lowe AC. Circulating tumor cells: overview and opportunities in cytology. Adv Anat Pathol. 2019;26(1):56-63. DOI: 10.1097/PAP.0000000000000217 PMID: 30325755 40. Alix-Panabières C, Pantel K. Clinical applications of circulating tumor cells and circulating tumor DNA as liquidbiopsy. Cancer Discov. 2016;6(5):479- 91. DOI: 10.1158/2159-8290.CD-15-1483 PMID: 26969689 41. Kahlert C, Kalluri R. Exosomes in tumor microenvironment influence cancer progression and metastasis. J Mol Med (Berl). 2013;91(4):431-7. DOI: 10.1007/s00109-013-1020-6 PMID: 23519402 42. Oudkerk M, Devaraj A, Vliegenthart R, Henzler T, Prosch H, Heussel CP, et al. European position statement on lung cancer screening. Lancet Oncol. 2017;18(12):e754-66. DOI: 10.1016/S1470-2045(17)30861-6 PMID: 29208441 68 DIHALA Zdrav Vestn | January – February 2023 | Volume 92 | https://doi.org/10.6016/ZdravVestn.3294 43. Jamal-Hanjani M, Wilson GA, McGranahan N, Birkbak NJ, Watkins TB, Veeriah S, et al.; TRACERx Consortium. Tracking the evolution of non- small-cell lung cancer. N Engl J Med. 2017;376(22):2109-21. DOI: 10.1056/ NEJMoa1616288 PMID: 28445112 44. Szpechcinski A, Rudzinski P, Kupis W, Langfort R, Orlowski T, Chorostowska-Wynimko J. Plasma cell-free DNA levels and integrity in patients with chest radiological findings:NSCLC versus benign lung nodules. Cancer Lett. 2016;374(2):202-7. DOI: 10.1016/j. canlet.2016.02.002 PMID: 26854716 45. LeBleu VS, Kalluri R. Exosomes as a multicomponent biomarker platform in cancer. Trends Cancer. 2020;6(9):767-74. DOI: 10.1016/j. trecan.2020.03.007 PMID: 32307267 46. Sohel MM. Circulating microRNAs as biomarkers in cancer diagnosis. Life Sci. 2020;248:117473. DOI: 10.1016/j.lfs.2020.117473 PMID: 32114007 47. Tang Z, Li D, Hou S, Zhu X. The cancer exosomes: clinical implications, applications and challenges. Int J Cancer. 2020;146(11):2946-59. DOI: 10.1002/ijc.32762 PMID: 31671207 48. Ilie M, Hofman V, Long-Mira E, Selva E, Vignaud JM, Padovani B, et al. “Sentinel” circulating tumor cells allow early diagnosis of lung cancer in patientswith chronic obstructive pulmonary disease. PLoS One. 2014;9(10):e111597. DOI: 10.1371/journal.pone.0111597 PMID: 25360587 49. Leroy S, Benzaquen J, Mazzetta A, Marchand-Adam S, Padovani B, Israel-Biet D, et al.; AIR Project Study Group. Circulating tumour cells as a potential screening tool for lung cancer (the AIR study):protocol of a prospective multicentre cohort study in France. BMJ Open. 2017;7(12):e018884. DOI: 10.1136/bmjopen-2017-018884 PMID: 29282271 50. Marquette CH, Boutros J, Benzaquen J, Ferreira M, Pastre J, Pison C, et al.; AIR project Study Group. Circulating tumour cells as a potential biomarker for lung cancer screening: a prospectivecohort study. Lancet Respir Med. 2020;8(7):709-16. DOI: 10.1016/S2213-2600(20)30081-3 PMID: 32649919 51. Serrano MJ, Garrido-Navas MC, Diaz Mochon JJ, Cristofanilli M, Gil-Bazo I, Pauwels P, et al.; International Society of Liquid Biopsy. Precision Prevention and Cancer Interception: The New Challenges of Liquid Biopsy. Cancer Discov. 2020;10(11):1635-44. DOI: 10.1158/2159-8290.CD- 20-0466 PMID: 33037026 52. Chen X, Gole J, Gore A, He Q, Lu M, Min J, et al. Non-invasive early detection of cancer four years before conventional diagnosis usinga blood test. Nat Commun. 2020;11(1):3475. DOI: 10.1038/s41467-020- 17316-z PMID: 32694610 53. Abbosh C, Birkbak NJ, Swanton C. Early stage NSCLC - challenges to implementing ctDNA-based screening and MRD detection. Nat Rev Clin Oncol. 2018;15(9):577-86. DOI: 10.1038/s41571-018-0058-3 PMID: 29968853 54. Planchard D, Popat S, Kerr K, Novello S, Smit EF, Faivre-Finn C, et al.; ESMO Guidelines Committee. Metastatic non-small cell lung cancer: ESMO Clinical Practice Guidelines for diagnosis,treatment and follow-up. Ann Oncol. 2018;29:iv192-237. DOI: 10.1093/annonc/mdy275 55. Ettinger DS, Wood DE, Aggarwal C, Aisner DL, Akerley W, Bauman JR, et al.; OCN. NCCN Guidelines Insights: non-small cell lung cancer, Version 1.2020. J Natl Compr Canc Netw. 2019;17(12):1464-72. DOI: 10.6004/ jnccn.2019.0059 PMID: 31805526 56. Nosaki K, Satouchi M, Kurata T, Yoshida T, Okamoto I, Katakami N, et al. Re-biopsy status among non-small cell lung cancer patients in Japan: A retrospectivestudy. Lung Cancer. 2016;101:1-8. DOI: 10.1016/j. lungcan.2016.07.007 PMID: 27794396 57. Coghlin CL, Smith LJ, Bakar S, Stewart KN, Devereux GS, Nicolson MC, et al. Quantitative analysis of tumor in bronchial biopsy specimens. J Thorac Oncol. 2010;5(4):448-52. DOI: 10.1097/JTO.0b013e3181ca12c4 PMID: 20125040 58. Food and drug administration. List of Cleared or Approved Companion Diagnostic Devices (In Vitro and Imaging Tools). Silver Spring: FDA; 2021 [cited 2021 Apr 20]. Available from: http://www.fda.gov/MedicalDevices/ ProductsandMedicalProcedures/InVitroDiagnostics/ucm301431.htm. 59. European Medicines Agency. List of Cleared or Approved Companion Diagnostic Devices (In Vitro and Imaging Tools). Amsterdam: EMA; 2021 [cited 2021 Apr 20]. Available from: http://www.ema.europa.eu/docs/en_ GB/document_library/EPAR__Product_Information/human/004124/ WC500202022.pdf. 60. Chong CR, Jänne PA. The quest to overcome resistance to EGFR-targeted therapies in cancer. Nat Med. 2013;19(11):1389-400. DOI: 10.1038/ nm.3388 PMID: 24202392 61. Kwapisz D. The first liquid biopsy test approved. Is it a new era of mutation testing for non-smallcell lung cancer? Ann Transl Med. 2017;5(3):46. DOI: 10.21037/atm.2017.01.32 PMID: 28251125 62. Pisapia P, Pepe F, Smeraglio R, Russo M, Rocco D, Sgariglia R, et al. Cell free DNA analysis by SiRe® next generation sequencing panel in non small celllung cancer patients: focus on basal setting. J Thorac Dis. 2017;9(S13):S1383-90. DOI: 10.21037/jtd.2017.06.97 PMID: 29184677 63. Yang JC, Ahn MJ, Kim DW, Ramalingam SS, Sequist LV, Su WC, et al. Osimertinib in pretreated T790M-positive advanced non-small-cell lung cancer: AURAstudy phase II extension component. J Clin Oncol. 2017;35(12):1288-96. DOI: 10.1200/JCO.2016.70.3223 PMID: 28221867 64. Normanno N, Maiello MR, Chicchinelli N, Iannaccone A, Esposito C, De Cecio R, et al. Targeting the EGFR T790M mutation in non-small- cell lung cancer. Expert Opin Ther Targets. 2017;21(2):159-65. DOI: 10.1080/14728222.2017.1272582 PMID: 28002980 65. Piotrowska Z, Niederst MJ, Karlovich CA, Wakelee HA, Neal JW, Mino- Kenudson M, et al. Heterogeneity underlies the emergence of egfrt790 wild-type clones following treatmentof T790M-Positive cancers with a third-generation EGFR inhibitor. Cancer Discov. 2015;5(7):713-22. DOI: 10.1158/2159-8290.CD-15-0399 PMID: 25934077 66. Remon J, Menis J, Hasan B, Peric A, De Maio E, Novello S, et al. The APPLE Trial: feasibility and activity of AZD9291 (Osimertinib) treatment on positiveplasma T790M in EGFR-mutant NSCLC patients. EORTC 1613. Clin Lung Cancer. 2017;18(5):583-8. DOI: 10.1016/j.cllc.2017.02.005 PMID: 28341106 67. Shaw AT, Solomon BJ, Besse B, Bauer TM, Lin CC, Soo RA, et al. ALK resistance mutations and efficacy of lorlatinib in advanced anaplastic lymphomakinase-positive non-small-cell lung cancer. J Clin Oncol. 2019;37(16):1370-9. DOI: 10.1200/JCO.18.02236 PMID: 30892989 68. Mezquita L, Swalduz A, Jovelet C, Ortiz-Cuaran S, Howarth K, Planchard D, et al. Clinical relevance of an amplicon-based liquid biopsy for detecting ALK and ROS1 fusionand resistance mutations in patients with non-small-cell lung cancer. JCO Precis Oncol. 2020;4(4):4. DOI: 10.1200/ PO.19.00281 PMID: 32923908 69. Ortiz-Cuaran S, Mezquita L, Swalduz A, Aldea M, Mazieres J, Leonce C, et al. Circulating tumor DNA genomics reveal potential mechanisms of resistance to BRAF-targetedtherapies in patients with BRAF-mutant metastatic non-small cell lung cancer. Clin Cancer Res. 2020;26(23):6242- 53. DOI: 10.1158/1078-0432.CCR-20-1037 PMID: 32859654 70. Mosele F, Remon J, Mateo J, Westphalen CB, Barlesi F, Lolkema MP, et al. Recommendations for the use of next-generation sequencing (NGS) for patients withmetastatic cancers: a report from the ESMO Precision Medicine Working Group. Ann Oncol. 2020;31(11):1491-505. DOI: 10.1016/j.annonc.2020.07.014 PMID: 32853681 71. Clery E, Pisapia P, Feliciano S, Vigliar E, Marano A, De Luca C, et al. There is still a role for cytology in the ‘liquid biopsy’ era. A lesson from a TKI- treatedpatient showing adenocarcinoma to squamous cell carcinoma transition during diseaseprogression. J Clin Pathol. 2017;70(9):798-802. DOI: 10.1136/jclinpath-2017-204370 PMID: 28363898 72. Revelo AE, Martin A, Velasquez R, Kulandaisamy PC, Bustamante J, Keshishyan S, et al. Liquid biopsy for lung cancers: an update on recent developments. Ann Transl Med. 2019;7(15):349. DOI: 10.21037/ atm.2019.03.28 PMID: 31516895 73. Wan JC, Massie C, Garcia-Corbacho J, Mouliere F, Brenton JD, Caldas C, et al. Liquid biopsies come of age: towards implementation of circulating tumour DNA. Nat Rev Cancer. 2017;17(4):223-38. DOI: 10.1038/nrc.2017.7 PMID: 28233803 69 STROKOVNI ČLANEK Liquid biopsy – a diagnostic and therapeutic tool in the treatment of non-small cell lung cancer 74. Song Y, Hu C, Xie Z, Wu L, Zhu Z, Rao C, et al.; Written on behalf of AME Lung Cancer Collaborative Group. Circulating tumor DNA clearance predicts prognosis across treatment regimen in a largereal-world longitudinally monitored advanced non-small cell lung cancer cohort. Transl Lung Cancer Res. 2020;9(2):269-79. DOI: 10.21037/tlcr.2020.03.17 PMID: 32420066 75. Horn L, Whisenant JG, Wakelee H, Reckamp KL, Qiao H, Leal TA, et al. Monitoring therapeutic response and resistance: analysis of circulating tumor DNAin patients with ALK+ lung cancer. J Thorac Oncol. 2019;14(11):1901-11. DOI: 10.1016/j.jtho.2019.08.003 PMID: 31446141 76. Lee JY, Qing X, Xiumin W, Yali B, Chi S, Bak SH, et al. Longitudinal monitoring of EGFR mutations in plasma predicts outcomes of NSCLC patientstreated with EGFR TKIs: Korean Lung Cancer Consortium (KLCC- 12-02). Oncotarget. 2016;7(6):6984-93. DOI: 10.18632/oncotarget.6874 PMID: 26755650 77. Kern I, Čufer T, Rot M, Mohorčič K, Požek I, Palma JF, et al. Dynamic changes of egfr activating mutations as an early predictor of progressionin non–small cell lung cancer patients treated with EGFR tyrosine kinase inhibitors. J Mol Biomark Diagn. 2020;11(3):1-7. 78. Zulato E, Attili I, Pavan A, Nardo G, Del Bianco P, Boscolo Bragadin A, et al. Early assessment of KRAS mutation in cfDNA correlates with risk of progression anddeath in advanced non-small-cell lung cancer. Br J Cancer. 2020;123(1):81-91. DOI: 10.1038/s41416-020-0833-7 PMID: 32376889