Next Article in Journal
Unprocessed snRNAs Are a Prognostic Biomarker and Correlate with a Poorer Prognosis in Colorectal Cancer
Previous Article in Journal
Prognostic Role of CA-125 Elimination Rate Constant (KELIM) in Patients with Advanced Epithelial Ovarian Cancer Who Received PARP Inhibitors
Previous Article in Special Issue
Baseline Cell-Free DNA Can Predict Malignancy of Nodules Observed in the ITALUNG Screening Trial
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Systematic Review

Identification and Application of Emerging Biomarkers in Treatment of Non-Small-Cell Lung Cancer: Systematic Review

by
Juan Carlos Restrepo
1,
Darly Martínez Guevara
1,
Andrés Pareja López
2,
John Fernando Montenegro Palacios
3 and
Yamil Liscano
1,*
1
Grupo de Investigación en Salud Integral (GISI), Departamento Facultad de Salud, Universidad Santiago de Cali, Cali 760035, Colombia
2
Grupo de Investigación Unidad de Toxicidad In Vitro—UTi, Facultad de Ciencias, Universidad CES, Medellin 050021, Colombia
3
Specialization in Internal Medicine, Department of Health, Universidad Santiago de Cali, Cali 760035, Colombia
*
Author to whom correspondence should be addressed.
Cancers 2024, 16(13), 2338; https://doi.org/10.3390/cancers16132338
Submission received: 14 May 2024 / Revised: 15 June 2024 / Accepted: 17 June 2024 / Published: 26 June 2024
(This article belongs to the Special Issue Predictive Biomarkers for Lung Cancer)

Abstract

:

Simple Summary

Lung cancer remains a leading cause of cancer-related mortality globally, requiring new diagnostic and therapeutic approaches. This research arises from the need to improve diagnostic accuracy and treatment effectiveness for patients with non-small cell lung cancer. The authors aim to systematically evaluate the potential of emerging biomarkers, including circulating tumor DNA, microRNAs and mutational load of blood tumors and their relationship with different treatments. The findings of this study could have a significant impact on the research community by providing a basis for integrating these biomarkers into clinical practice, thereby improving personalized treatment strategies and patient outcomes in non-small cell lung cancer.

Abstract

Non-small-cell lung cancer (NSCLC) comprises approximately 85% of all lung cancer cases, often diagnosed at advanced stages, which diminishes the effective treatment options and survival rates. This systematic review assesses the utility of emerging biomarkers—circulating tumor DNA (ctDNA), microRNAs (miRNAs), and the blood tumor mutational burden (bTMB)—enhanced by next-generation sequencing (NGS) to improve the diagnostic accuracy, prognostic evaluation, and treatment strategies in NSCLC. Analyzing data from 37 studies involving 10,332 patients from 2020 to 2024, the review highlights how biomarkers like ctDNA and PD-L1 expression critically inform the selection of personalized therapies, particularly beneficial in the advanced stages of NSCLC. These biomarkers are critical for prognostic assessments and in dynamically adapting treatment plans, where high PD-L1 expression and specific genetic mutations (e.g., ALK fusions, EGFR mutations) significantly guide the use of targeted therapies and immunotherapies. The findings recommend integrating these biomarkers into standardized clinical pathways to maximize their potential in enhancing the treatment precision, ultimately fostering significant advancements in oncology and improving patient outcomes and quality of life. This review substantiates the prognostic and predictive value of these biomarkers and emphasizes the need for ongoing innovation in biomarker research.

1. Introduction

Non-small-cell lung cancer (NSCLC) represents approximately 85% of all lung cancer cases and necessitates early detection to improve clinical outcomes [1,2,3]. Traditionally, NSCLC management includes surgery, chemotherapy, and radiation therapy, the effectiveness of which varies significantly based on the stage at diagnosis. Unfortunately, about 60% of patients are diagnosed in advanced stages [4], reducing the likelihood of curative treatment and increasing the mortality rate [5,6].
In 2020, lung cancer accounted for 2.2 million new cases and 1.8 million deaths globally, constituting 11.4% and 18% of all cancer cases and deaths, respectively. Approximately 65.33% of male cases were diagnosed at advanced stages (III or IV), contributing to the high mortality rates, particularly in regions with significant exposure to carcinogens like tobacco smoke in Central Europe and air pollution in China [7,8]. Notably, China experienced about 820,000 new cases and 715,000 deaths alone, indicating a rise in both incidence and mortality since 2015. In contrast, the U.S. reported 57% of cases at the metastatic stage, while the late-stage diagnosis rates were extraordinarily high in Mexico (98–99%), Brazil (70%), and Peru (85.5%), highlighting the need for early detection programs [9]. Colombia, dealing with environmental and occupational exposure, sees 6876 new cases yearly and considers lung cancer as its sixth most prevalent cancer, although recent trends suggest declining rates. Despite the common association with smoking, 12% of patients worldwide have never smoked, underscoring the influence of other risks, such as environmental pollutants [9].
NSCLC is a highly heterogeneous disease, exhibiting significant variability at both the molecular and clinical levels. This heterogeneity, which encompasses genetic mutations, histological subtypes, and tumor microenvironments, critically impacts treatment responses and overall prognosis [10,11]. Adenocarcinomas (ADCs) and squamous cell carcinomas (SCCs) are the predominant histological types of NSCLC, each associated with distinct genetic profiles. ADCs frequently harbor mutations in genes such as KRAS, EGFR, and ALK, while SCCs often exhibit alterations in DDR2, FGFR1, and the PI3K pathway [10,11,12]. The recognition of these genetic diversities has led to the development of targeted therapies, although their effectiveness is often short-lived due to acquired resistance. Additionally, the tumor microenvironment, comprising fibroblasts, immune cells, and extracellular matrix components, further complicates the disease landscape by influencing tumor progression and treatment responses [10,13,14,15]. The effective management of NSCLC requires well-documented diagnostic comparisons to tailor treatment strategies. Incorporating advanced biomarkers into diagnostic protocols, such as circulating tumor DNA (ctDNA), microRNAs (miRNA), and the blood tumor mutational burden (bTMB), is essential in identifying actionable mutations and predicting therapeutic responses [16,17].
ctDNA is crucial in detecting actionable mutations and monitoring tumor progression non-invasively, serving as both a predictive and prognostic biomarker [18,19]. PD-L1 is another relevant biomarker, as its expression in tumor cells can predict the response to immunotherapies, such as checkpoint inhibitors, making it a key predictive biomarker [20]. miRNAs play a vital role in NSCLC by acting as key regulators in cancer progression and as potential biomarkers for prognosis and treatment responses. Some, like miRNA-21, have been shown to be important predictors of the response to immunotherapy [21,22,23]. Finally, bTMB can be an important predictive indicator of the response to immunotherapies. Together, these biomarkers enable the more precise detection of NSCLC and the personalization of treatments, thereby improving the clinical outcomes.
The clinical outcomes for lung cancer are closely related to the stage at which the cancer is diagnosed. For example, patients diagnosed at stage I have a five-year survival rate of 68.4%, while those diagnosed at stage IV have a significantly lower survival rate of just 5.8% [24]. Unfortunately, most lung cancers are diagnosed at stage IV, associated with lower survival rates and a higher symptom burden. Early screening and detection significantly reduce lung cancer mortality [25]. The standard of care for NSCLC patients at stages I and II, and some at stage IIIA, is surgical resection, followed by adjuvant systemic therapy if needed. The introduction of osimertinib, a third-generation oral tyrosine kinase inhibitor, for adjuvant therapy in completely resected NSCLC at stages II and III has shown promising results [24]. Systemic treatment options for metastatic NSCLC include chemotherapy, targeted therapy, and immunotherapy, with specific tests for driver mutations in non-squamous tumors for more effective and less toxic treatment [24]. While the development and integration of osimertinib and other systemic treatments have marked a significant improvement in the care for patients with early and metastatic NSCLC, the role of molecular diagnostics has become increasingly central in the management of this disease [26].
Next-generation sequencing technologies have transformed NSCLC treatment by providing a comprehensive genetic snapshot of individual tumors. This advanced genomic profiling helps to identify actionable mutations and biomarkers, critical in customizing patient care [27,28]. By pinpointing specific genetic alterations, clinicians can select targeted therapies that are more likely to be effective, improving treatment outcomes and reducing unnecessary side effects [29,30].
Identifying specific genetic alterations, such as epidermal growth factor receptor (EGFR) mutations, NGS facilitates the development of targeted treatment strategies [31,32]. These strategies have significantly evolved to enhance the progression-free survival (PFS) and overall survival (OS) rates, particularly for NSCLC patients with EGFR mutations. EGFR mutations, primarily the exon 19 deletions (ex19del) and the L858R point mutation in exon 21, represent 85–90% of all EGFR mutations and are highly sensitive to EGFR tyrosine kinase inhibitors (EGFR-TKIs). However, the T790M mutation in exon 20, less common at diagnosis, is associated with about half of the cases of resistance to first- and second-generation EGFR-TKIs [31,33]. EGFR mutations, which occur in approximately 10% of cases in North America and Western Europe and between 30 and 50% in East Asia, are predictive of a good response to EGFR-TKIs. Clinical trials have demonstrated a 54% improvement in progression-free survival and a 6.8-month absolute benefit in overall survival with this medication [9,34].
Genes such as ALK, ROS1, BRAFV600E, and NTRK have also been identified as critical in developing targeted treatment strategies [35,36]. Immunotherapy with agents such as nivolumab, pembrolizumab, and atezolizumab, which inhibit PD-1 and PD-L1, has been shown to increase survival in NSCLC. However, resistance to these therapies persists, prompting ongoing research to develop combinations of targeted therapies and immunotherapies that address these resistance challenges [37,38].
ROS1 rearrangements, present in approximately 1–2% of NSCLC patients, are sensitive to the ROS1/MET inhibitor crizotinib, which has shown a response rate of 72% [9,39]. Lorlatinib has been effective against acquired resistance to crizotinib in NSCLC positive for ROS1, and entrectinib has been approved for these cases. KRAS mutations, the most common type of driver mutation in lung cancer, often found in individuals with a history of smoking, have recently been targeted by sotorasib, a new agent directed at KRAS G12C mutations, showing a response rate of 32% in clinical trials [9].
These advancements underscore the importance of biomarkers in personalizing NSCLC treatment, offering hope for significant improvements in patient outcomes through more targeted therapies. This review systematically examines recent studies on the identification and application of emerging biomarkers in NSCLC treatment, focusing on their utility in informing therapeutic decisions and improving survival outcomes. By contributing to the existing body of knowledge, this review emphasizes the impact of biomarkers on personalizing oncological treatment and enhancing the clinical outcomes in NSCLC.

2. Materials and Methods

2.1. Study Protocol

This systematic review was conducted in accordance with the guidelines of the Cochrane Collaboration Handbook and reported considering the recommendations for systematic reviews and meta-analyses of the PRISMA statement [40]. The research was formulated considering the PICO strategy (Population, Intervention, Comparison, Outcomes) [1].

2.2. Research Question

In patients with non-small-cell lung cancer (NSCLC) (P), how have emerging biomarkers (I), compared to non-specific or previously established biomarkers (C), impacted the diagnostic accuracy and prognostic value and informed treatment strategies, including predictive monitoring and treatment maintenance strategies, particularly in the context of immunotherapy and palliative care, over the last five years, in terms of reducing symptomatology or improving quality of life (O)?

2.3. Eligibility Criteria

2.3.1. Inclusion Criteria

The inclusion criteria were as follows:
  • Manuscripts published between 2020 and 2024;
  • Studies including predictive biomarkers;
  • Studies including diagnostic biomarkers;
  • Studies including monitoring biomarkers;
  • Clinical follow-up studies with biomarkers for oncological treatments (predictive biomarkers);
  • Full-access articles;
  • Articles in English;
  • Clinical trials.

2.3.2. Exclusion Criteria

  • Articles in pre-print mode or letters to the editor;
  • Studies with biomarkers not specific to NSCLC;
  • Studies published >5 years ago;
  • Studies with inconclusive experimental designs;
  • Studies of selected population analysis;
  • Quality of life studies;
  • Studies on treatment maintenance strategies and palliative care management.

2.4. Data Sources and Search Strategy

The search was conducted in the following databases: PubMed, Cochrane Clinical Trials, SCOPUS, ScienceDirect, Biomed Central (BMC), Web of Science, Springer, and the Virtual Health Library (VHL). No language filters were applied, and the date range was set between 2020 and 2024. The search strategy was designed and executed from November 2023 to January 2024 by two researchers independently (D.M. and J.-C.R.L.). Terms (keywords) were combined using the Boolean operators AND and OR (see search details in Appendix A). The references of relevant articles were reviewed, and additional web searches were conducted to identify studies not evident through initial tracking. When necessary to confirm the clinical trial or expand the information of a study, access to ClinicalTrials.gov (https://clinicaltrials.gov/ (accessed on 18 September 2023)) was sought, if applicable. Data were stored using Zotero version 6.0 (accessed on 23 November 2023).

2.5. Selection and Data Extraction

The selection of potentially eligible studies was carried out independently by the two researchers, initially examining the title, abstract, and subsequently the full text. Studies whose relevance was unclear were discussed thoroughly, and the decision to include them in the review was reached by consensus. Two reviewers (J-C.R.L. and Y.L.) extracted information from the primary studies considering the details of the clinical trial (first author, year of publication), country, type of design, biological sample, number of participants, age of participants, sex of participants, biomarker evaluated, method of biomarker analysis, cut-off value, sensitivity and specificity of biomarkers, predictive and prognostic value, diagnostic performance, overall survival (OS) and progression-free survival (PFS), hazard ratios (HR) and 95% confidence intervals (CI) for tumor size, TNM stage, survival rates, treatment, statistical methods used for biomarker evaluation, fold change, p-value, and conclusions. Subsequently, a third and fourth reviewer (A.P. and D.M.G.) verified the integrity and accuracy of the recorded information.

2.6. Risk of Bias Assessment

The risk of bias in the primary studies was assessed by independent reviewers utilizing standardized instruments attuned to the core design elements of clinical trials. Data for this assessment were entered into Review Manager version 5.4® (RevMan, accessed on 23 January 2024), considering the following criteria: generation of random sequences, allocation concealment, blinding of participants and personnel, blinding of outcome assessments, completeness of outcome data, and avoidance of selective reporting. The RCTs were judged to have a low or high risk of bias based on their conformity to pre-established guidelines.
For each domain, the risk was classified as “low”, “high”, or “unclear”, in line with the evaluative scales provided by the Cochrane Risk of Bias tool (accessed on 9 June 2023) for randomized trials and the ROBINS-I tool for non-randomized trials [41,42]. Any discrepancies found during the risk of bias assessment were addressed and resolved through discussions between the reviewers until a consensus was reached.

2.7. Ethical Considerations

During the development of this study, no interventions were made regarding the demographic and physiological variables of the participants. Therefore, this research work was considered of minimal risk according to Resolution No. 8430 of 1993 of the Colombian legislation and the Declaration of Helsinki.

3. Results

3.1. Characteristics of the Included Studies

Following an extensive search across the referenced databases, a total of 625 articles were initially retrieved from eight databases. Of these, 114 were identified as duplicates and subsequently removed. The remaining articles underwent a title and abstract screening process, resulting in 394 articles being assessed. This screening led to the exclusion of 247 articles, and a Cohen’s kappa coefficient of 0.92 was calculated at this stage, suggesting a high level of agreement between the reviewers. Further evaluation was carried out on the full texts of the remaining 147 articles to determine their eligibility for inclusion in the review. During this phase, 31 articles were excluded due to their lack of relevance to the review topic, 54 were discarded due to having insufficient data, 9 were identified as additional duplicates within the dataset, and 14 were excluded as they did not match the required study type, and a Cohen’s Kappa coefficient of 0.823 was obtained, evidencing substantial agreement, slightly below that in the initial screening. Ultimately, 37 studies satisfied all inclusion criteria and were included in the systematic review. The details of the study selection process are depicted in the PRISMA [43] flowchart illustrated in Figure 1.

3.2. Findings of the Studies

Table 1 summarizes the characteristics of the 37 studies reviewed, which included both randomized clinical trials (RCTs) (29 studies) and non-randomized trials (eight studies). These studies involved a total of 10,332 patients with NSCLC, examining a variety of emerging biomarkers through the analysis of peripheral blood samples for ctDNA and tumor biopsies. The studies predominantly focused on patients with advanced stages of NSCLC (IIIB-IV), ranging in age from 18 to 91 years, with a median age of 59 ± 20 years. The gender distribution was approximately 54.9% male and 45.1% female.
Most of the research focused on ctDNA as a biomarker, analyzing specific mutations such as ALK fusions, EGFR exon 20 insertions, bTMB, PD-L1 expression, and other genomic alterations, including mutations in EGFR, BRCA2, BRINP3, FBXW7, KIT, and RB1 and the EGFR T790M mutation [44,45,46,47]. Biomarkers related to the immune response were also evaluated, including the responses of circulating immune cells and gene expression profiles.
Table 1. Synthesis of studies included in systematic review.
Table 1. Synthesis of studies included in systematic review.
Author and
Publication Year
CountryType of DesignBiological SampleNumber of
Participants
Age of ParticipantsSex of ParticipantsBiomarker Evaluated
Ren et al., 2022
[48]
ChinaRandomized Clinical TrialPeripheral blood ctDNA samples38918 to 75 years.Men and women participated, but most patients were menctDNA dynamics
Yang et al., 2023
[49]
ChinaRandomized Clinical TrialPeripheral blood ctDNA samples248Median age 54.0 years (brigatinib group), 53.0 years (alectinib group)54% female (brigatinib), 55% female (alectinib)ALK fusions in plasma ctDNA
Riess et al., 2021
[50]
USARandomized Clinical TrialPeripheral blood ctDNA samples1160 (51–70)27% male, 73% femaleEGFR exon 20 insertions
Lo Russo et al., 2022 [51]USARandomized Clinical TrialPeripheral blood ctDNA samples65Median age was 70.9 years (Q1–Q3: 63.7–77.1 years)21 (32.3%) were female and the rest were male36 immunobiomarkers like CD14, CD15, CD16, CD33, CD56, CD19, CD3, HLA-DR
Garon et al., 2023
[52]
USA, JapanRandomized Clinical TrialPeripheral blood ctDNA samples449NSNSThe biomarkers evaluated were EGFR and co-occurring/treatment-emergent (TE) genomic alterations in ctDNA
Han et al., 2023
[53]
ChinaNon-Randomized Clinical TrialPeripheral blood ctDNA samples4018 to 75 yearsNSbTMB through ctDNA profiling
Si et al., 2021
[54]
USARandomized Clinical TrialPeripheral blood ctDNA samples809NSNSbTMB
Jiang et al., 2021
[55]
ChinaRandomized Clinical TrialTumor biopsies for whole-exome and transcriptome sequencing and plasma samples for ctDNA analysis4018 to 75 years oldIncluded both male (19, 47.5%) and female (21, 52.5%) patientsPD-L1, TMB, CD8+ TIL density, DSPP
Zhang et al., 2024
[56]
ChinaRandomized Clinical TrialPeripheral blood ctDNA samples47Median age was 65 years (range: 52–76)Mostly male (45/46, 97.8%); females (1/46, 2.2%)ctDNA dynamics, BRCA2, BRINP3, FBXW7, KIT, RB1
Tan et al., 2024
[44]
AustraliaNon-Randomized Clinical TrialPeripheral blood ctDNA samples47Median age was 60 years (range: 32–86)62% female, 38% maleEGFR T790M, EGFR exon 19 deletion, L858R mutation
Kim et al., 2022
[57]
USARandomized Clinical TrialPeripheral blood ctDNA samples152NSNSbTMB
Peters et al., 2022
[58]
MultinationalRandomized Clinical TrialPeripheral blood ctDNA samples471NSNSbTMB
Chaft et al., 2022
[59]
USANon-Randomized Clinical TrialTumor samples181Median age of 65 years (range: 37–83)93 females (51%)Major pathological response (MPR), PD-L1 tumor proportion score (TPS)
Shi et al., 2022
[60]
ChinaRandomized Clinical TrialPeripheral blood ctDNA samples29018 to 75 yearsMajority male (93.8% sintilimab, 90.4% docetaxel)PD-L1, OVOL2, CTCF via tissue/blood sequencing
Papadimitrakopoulou et al., 2020 [45]MultinationalRandomized Clinical TrialPeripheral blood ctDNA samplesNSNSNSEGFR mutations
Sakai et al., 2021a
[46]
JapanRandomized Clinical TrialFormalin-fixed paraffin-embedded tumor tissue389NSNSTumor mutation burden (TMB)
Park et al., 2023
[61]
South KoreaNon-Randomized Clinical TrialPeripheral blood ctDNA samples100NS84 male, 16 femalebTMB, cfDNA concentration, hVAF, VAFSD
Park et al., 2021
[47]
South KoreaRandomized Clinical TrialPeripheral blood ctDNA samples19Median age of 70 years (range: 32–84)13 females (68%) and 6 males (32%)Activating EGFR mutations in ctDNA and tumor DNA
Gu et al., 2023
[62]
ChinaRandomized Clinical TrialPeripheral blood ctDNA samples92Median age 65 years34% male and 66% femaleEGFR mutations, ctDNA for minimal residual disease (MRD)
Han et al., 2022
[63]
ChinaNon-Randomized Clinical TrialPeripheral blood ctDNA samples33Median age 56 years (range: 31–71)26 males (78.79%) and 7 females (21.21%)ctDNA analysis with 448-gene panel for short-term dynamics
Zhong et al., 2023
[64]
ChinaRandomized Clinical TrialPeripheral blood ctDNA samples69Median age was 58 years (range: 33–76 years)38 males (55.1%) and 31 females (44.9%)EGFR mutation detection via ctDNA
García-Pardo et al., 2023 [65]CanadaNon-Randomized Clinical TrialPeripheral blood ctDNA samples150Median age at diagnosis 68 years (range: 33–91 years)80 men (53%), 70 women (47%)ctDNA genotyping
Nomura et al., 2020 [66]JapanRandomized Clinical TrialPeripheral blood ctDNA samples216NSBoth males and females (NS)ctDNA (Guardant360® ancillary study)
Martini et al., 2022
[67]
ItalyNon-Randomized Clinical TrialBasal fecal samples and peripheral blood samples for ctDNA analysis14NSNSGut microbiota species and ctDNA RAS/BRAF WT MSS disease
Provencio et al., 2022 [68]SpainNon-Randomized Clinical TrialPeripheral blood samples for ctDNA analysis and formalin-fixed paraffin-embedded tissue samples46Median age was 67.6 yearsMajority male (66.5%)ctDNA analysis for prognosis and predictive value
West et al., 2022
[69]
Multinational studyRandomized Clinical TrialPeripheral blood ctDNA samples920NSBoth males and females included, exact distribution not providedKRAS, STK11, KEAP1, TP53 mutations
Lo Russo et al., 2023 [70]ItalyRandomized Clinical TrialBlood and stool samples for circulating immune profiling and gut bacterial taxonomic abundance analysis65Median age was 70 years, with a range of 47–87 years44 men (68%) and 21 women (32%)Immune circulating cell subsets and gene expression levels
Zhou et al., 2023
[71]
Multinational studyRandomized Clinical TrialFormalin-fixed paraffin-embedded tumor tissue for PD-L1 expression assessmentNSNSBoth males and females included, but the exact distribution is not providedPD-L1 expression on tumor cells
Sakai et al., 2021b
[72]
JapanRandomized Clinical TrialPeripheral blood ctDNA samples52Median age 67 (range: 37–82 years)17 male (32.7%), 35 female (67.3%)EGFR genomic alterations including the T790M mutation
Redman et al., 2020 [73]USARandomized Clinical TrialFormalin-fixed paraffin-embedded tumor specimens for genomic DNA extraction1864NSBoth males and females includedMultiple biomarkers defined by the FoundationOne® NGS assay
Hirsch et al., 2022
[74]
USARandomized Clinical TrialFormalin-fixed paraffin-embedded tumor specimens1313NSBoth males and females includedEGFR copy number and protein expression
Schuler et al., 2020
[75]
Multinational studyRandomized Clinical TrialTumor samples55Median age was 60 yearsBoth males (60%) and females (40%)MET dysregulation
Gadgeel et al., 2022 [76]Multinational studyRandomized Clinical TrialTumor samples577NSBoth males and females includedPD-L1 expression
Ramalingam et al., 2021 [77]Multinational studyRandomized Clinical TrialFormalin-fixed paraffin-embedded tumor samples970NSBoth males (82%) and females included52-gene expression histology classifier (LP52)
Song et al., 2022
[78]
ChinaRandomized Clinical TrialPeripheral blood ctDNA samples78Median age 62 yearsBoth male (47.4%) and female (52.6%) participantsHER2 mutations
Anagnostou et al., 2023 [79]Multinational studyRandomized Clinical TrialPeripheral blood ctDNA samples50NSBoth males and females includedctDNA dynamics
Park et al., 2021
[80]
South KoreaRandomized Clinical TrialPeripheral blood ctDNA samples21Mean age of 68.5 years17 females and 4 malesEGFR exon 19 deletions, exon 21 point mutations (ctDNA)
ctDNA: Circulating Tumor DNA; ALK: Anaplastic Lymphoma Kinase; EGFR: Epidermal Growth Factor Receptor; bTMB: Blood Tumor Mutational Burden; TMB: Tumor Mutational Burden; PD-L1: Programmed Death Ligand 1; MPR: Major Pathological Response; TPS: Tumor Proportion Score; MRD: Minimal Residual Disease; cfDNA: Circulating Free DNA; hVAF: High Variant Allele Frequency; VAFSD: Variant Allele Frequency Standard Deviation; NGS: Next-Generation Sequencing; IHC: Immunohistochemistry; HR: Hazard Ratio; CI: Confidence Interval; OS: Overall Survival; PFS: Progression-Free Survival; RECIST: Response Evaluation Criteria In Solid Tumors; ORR: Objective Response Rate; NSCLC: Non-Small-Cell Lung Cancer; Mb: Megabase; BRCA2: Breast Cancer 2, Early Onset; DSPP: Dentin Sialophosphoprotein; CTCF: CCCTC-Binding Factor; OVOL2: Ovo-Like Transcriptional Repressor 2; FFPE: Formalin-Fixed Paraffin-Embedded; KRAS: Kirsten Rat Sarcoma Viral Oncogene Homolog; ABCP: Atezolizumab, Bevacizumab, Carboplatin, Paclitaxel; BCP: Bevacizumab, Carboplatin, Paclitaxel; KEAP1: Kelch-Like ECH-Associated Protein 1; STK11: Serine/Threonine Kinase 11; TP53: Tumor Protein p53; LASSO: Least Absolute Shrinkage and Selection Operator; ddPCR: Droplet Digital Polymerase Chain Reaction; miR: MicroRNA; LP52: 52-Gene Expression Histology Classifier; Mo: Month. NS: Not Specified.
The methodologies of these studies varied widely (see Table 2), employing techniques such as NGS, IHC, digital PCR, and specific assays like GuardantOMNI ctDNA and Foundation Medicine bTMB, reflecting a comprehensive approach to biomarker assessment through genetic and immunological analysis [37,54,56,61,74]. The differentiation between residual and non-residual ctDNA, along with the interpretation of PD-L1 expression and the density of infiltrating CD8+ T lymphocytes, highlights the importance of integrating genetic and immunological findings into NSCLC treatment [55,59,60]. Specifying the mutations per megabase to distinguish between high and low bTMB evidences the precision needed to assess the tumor’s mutational load, enhancing the predictive and prognostic value of these biomarkers [54,81].
Basal activating alterations in EGFR, associated with shorter median PFS, served as prognostic markers; however, treatment with ramucirumab plus erlotinib improved the outcomes regardless of these alterations, indicating their predictive value. The reduction in bTMB was established as a predictive biomarker for treatment with sintilimab plus docetaxel, while a high bTMB indicated a clinical benefit with durvalumab plus tremelimumab compared to chemotherapy [52]. The DSPP mutation was identified as a predictive biomarker of longer PFS, as was the clearance of ctDNA, which was associated with more durable outcomes [55]. The decrease in and elimination of EGFRm and T790M in post-treatment ctDNA were linked to longer progression-free survival and overall survival, highlighting the efficacy of sequential therapies [44]. High levels of PD-L1 expression were related to a greater pathological response, and the levels of expression of OVOL2 and CTCF were associated with PFS outcomes, highlighting their potential as prognostic markers [60].
In the study conducted by Ren et al., in 2021 [48], it was observed that the combined treatment of camrelizumab and chemotherapy significantly extended the OS and PFS compared to a placebo combined with chemotherapy. The results indicated median PFS of 8.5 months versus 4.9 months, and median OS not reached versus 14.5 months, for the groups treated with camrelizumab–chemotherapy and placebo–chemotherapy, respectively. Similar findings were reported for treatments with brigatinib and alectinib, both with a median PFS of approximately 19 months [49]. Additionally, patients who presented detectable alterations of monoclonal antibody against epidermal growth factor (aEGFR) had a median PFS of 12.7 months, contrasting with the 22.0 months for those without these alterations, although specific OS data were not provided [52].
This systematic review also identified variations in PFS and OS associated with different molecular biomarkers, such as CD14, CD16, HLA-DR, CD3, CD56, and NKT, evidencing their impact on treatment outcomes. It was noted that different combinations of these markers were related to significant variations in OS and PFS [51].
In Han et al.’s 2022 [53] study, various therapies were compared, noting significant improvements in the median OS and PFS with sintilimab over docetaxel. Notably, for patients with mKRAS mutations and squamous cell carcinoma, targeted treatment regimens and the addition of cetuximab, respectively, showed significant enhancements in survival rates, regardless of the KRAS mutation status [69,74].
The analysis of the HR and 95% CI shed light on the OS and PFS across different scenarios. An HR of 0.55 for OS suggested a 45% reduction in mortality risk, while the HR for PFS of 0.97 between brigatinib and alectinib indicated no significant difference in disease progression between these treatments [49]. Detectable alterations of aEGFR with an HR of 1.87 suggested faster disease progression. Meanwhile, a positive molecular pathological response (MPR) status indicated a favorable prognosis, with the HRs showing significant improvements in disease-free survival (DFS) and OS [52]. A high tumor mutational burden (TMB) and low or undetectable levels of ctDNA following neoadjuvant treatment were associated with significant improvements in PFS and OS, highlighting their prognostic value [82,83].
For patients with a TMB of 12 mutations per megabase or higher, an HR of 0.477 for improved recurrence-free survival (RFS) underscores the TMB’s potential as a predictive biomarker for the treatment response [57]. The pre-treatment levels of ctDNA were found to be significant, with low levels correlating with improved PFS and OS outcomes, suggesting ctDNA’s prognostic importance after therapy [59].
The assessment of the biomarker-based treatment efficacy has unveiled a range of therapeutic approaches. These include monotherapy with immune checkpoint inhibitors (ICIs) such as pembrolizumab, as well as targeted therapy combinations with chemotherapy. The treatment regimens have spanned from specific targeted therapies, like ALK and EGFR inhibitors, to immunotherapies and their integration with chemotherapy [49,59,70].
The Kaplan–Meier methodology and the Cox proportional hazards model have been extensively used to analyze the OS and PFS across studies, comparing treatments such as camrelizumab with carboplatin and paclitaxel, brigatinib versus alectinib, and durvalumab with or without tremelimumab versus chemotherapy. These methods provide precise estimates of the survival time and assess the relative risks of events such as disease progression or death [46,47,48,62,67].
The Clopper–Pearson method for the estimation of objective response rates and disease control, and the use of two-sided tests at the 0.05 significance level, as in the study of docetaxel plus sintilimab, highlight the rigor in comparing treatments’ efficacy [63]. Furthermore, advanced techniques like cross-validation for the determination of the optimal cut-off for the bTMB and the minimum p-value approach reflect the ongoing evolution of statistical methods in oncological research [54].
In addition to these statistical analyses, some studies have incorporated innovative approaches such as multivariable Cox regression, competitive risk analysis, and corrections for multiple comparisons, showcasing the complexity and need for precision in interpreting data on survival and treatment responses [54,68].
In the context of immunotherapy, the studies by G. Lo Russo et al. (2022) [51] and Chaft et al. (2022) [59] suggest that the circulating immune biomarkers and innate immune cells in the peripheral blood before treatment can predict the pathological response following neoadjuvant atezolizumab, indicating a need for further validation studies.
Meanwhile, the research by Yang et al. (2023) [49] found that brigatinib did not show superiority over alectinib for PFS in patients with ALK-positive NSCLC previously treated with crizotinib, highlighting the consistent safety profiles of both drugs and reaffirming their positions as standard treatments post-crizotinib.
In 2022, Martini et al. [67] conducted a pivotal study exploring the impact of the gut microbiota on the antitumor efficacy of cetuximab and avelumab treatments in NSCLC and mCRC patients. Utilizing 16S rRNA sequencing, the research analyzed fecal samples to identify microbial compositions, particularly highlighting two butyrate-producing bacteria, Agathobacter M104/1 and Blautia SR1/5. Their findings revealed a significant correlation between the presence of these bacteria and extended PFS, suggesting these microbial species as promising biomarkers for the prediction of treatment success. The study illuminated the potential mechanism of action, where butyrate production by these bacteria modulates the immune response, thereby potentially enhancing the antitumor activity of the immunotherapy regimen.
In Table 3, the summarized findings are presented along with their clinical implications, the treatment response, the heterogeneity in NSCLC, and their clinical and demographic characteristics. For example, in the case of ctDNA, it is identified as a biomarker with detection correlated to the tumor burden and disease progression. A reduction in levels during treatment is indicative of better PFS and OS. Its clinical implication is highlighted as a non-invasive biomarker for the monitoring of the response and the adjustment of therapeutic strategies. The predictive response to treatment is noted, with ctDNA being predictive of the response to camrelizumab, carboplatin, paclitaxel, osimertinib, sintilimab, nab-paclitaxel, and gefitinib. The studies evaluated show that its heterogeneity varies with the mutation type (EGFR, ALK), tumor stage, distribution of histological subtypes, and frequency of co-alterations. The patient demographic is predominantly men with a history of heavy smoking, ECOG 1 status, and stage IV disease.
PD-L1 is identified as a biomarker for tumor progression and prognosis. High levels are associated with better responses to ICIs and longer survival. Its clinical implication is its importance in selecting patients for immunotherapy. Regarding the treatment response, PD-L1 is predictive of the response to atezolizumab, pembrolizumab, and toripalimab. The heterogeneity of PD-L1 varies with the expression levels, tumor microenvironment, distribution of histological subtypes, and frequency of co-alterations. The patient demographic is predominantly men with a history of heavy smoking, ECOG 1 status, and stage IV disease.
miRNA is related to the response to immunotherapy and survival outcomes. miRNA-21 and miRNA-155 are correlated with the responses to immunotherapy and survival outcomes. Additionally, the plasma miR-32 levels correlate with the chemotherapy response and prognosis. The clinical implications of miRNA include patient stratification and treatment personalization based on molecular profiles. Xu et al., 2019 shows that it is predictive of the chemotherapy efficacy and prognosis with platinum-based chemotherapy. The heterogeneity of miRNA varies with the miRNA type, interaction with other molecular pathways, and frequency of co-alterations. The patient demographic includes those aged 45–78, predominantly men and 81.4% smokers, with ECOG 1–2 status and stage II–IV disease.
Finally, the bTMB is identified as a biomarker associated with tumor progression and prognosis. A high bTMB is associated with better outcomes in combined immunotherapy and chemotherapy. Accurate measurement predicts the immunotherapy efficacy and guides treatment selection. The studies by Han et al., 2023 [53] and Si et al., 2021 [54] highlight that it is predictive of the response to sintilimab + docetaxel and durvalumab + tremelimumab. The heterogeneity of the bTMB varies with the mutation burden, specific gene mutations, distribution of histological subtypes, and frequency of co-alterations. The patient demographic is predominantly men with a history of heavy smoking, ECOG 1 status, and stage IV disease.

3.3. Risk of Bias Assessment

Using the risk of bias graph created with RevMan 5.4® (accessed on 20 February 2024), the risk of bias assessment for the included studies is outlined as follows.
The assessment of the risk of bias in these studies was carried out using several criteria, as illustrated in Figure 2. Most of the studies, including those by Ren et al., 2021; Yang et al., 2023; G. Lo Russo et al., 2022; Garon et al., 2023; Si et al., 2021; Jiang et al., 2021; Zhang et al., 2024; Kim et al., 2022; Peters et al., 2022; Shi et al., 2022; Papadimitrakopoulou et al., 2020; Park et al., 2021; Gu et al., 2023; Zhong et al., 2023; Nomura et al., 2020; West et al., 2022; Lo Russo et al., 2023; Zhou et al., 2023; Sakai et al., 2020a; Redman et al., 2020; Hirsch et al., 2022; Schuler et al., 2020; Gadgeel et al., 2022; Ramalingam et al., 2022; Song et al., 2022; Anagnostou et al., 2023; and Park et al., 2020, were evaluated as having a low risk for random sequence generation (selection bias), suggesting thorough and well-documented randomization procedures [45,47,48,49,51,52,54,62,64,66,69,70,73,74,75,76,77,78,79].
Allocation concealment (selection bias) was primarily rated as low risk, indicating that the allocation process was sufficiently concealed to reduce selection bias in most studies. However, some studies, such as G. Lo Russo et al., 2022 [51], presented an unclear risk, indicating the need for more explicit documentation or reporting.
Regarding the blinding of participants and personnel (performance bias), many studies were evaluated as having a high risk due to the open nature of many clinical trials, which exposes them to possible performance bias. This includes the studies by Garon et al., 2023; Jiang et al., 2021; Kim et al., 2022; Peters et al., 2022; Shi et al., 2022; Papadimitrakopoulou et al., 2020; Park et al., 2021; Gu et al., 2023; Zhong et al., 2023; Nomura et al., 2020; West et al., 2022; Lo Russo et al., 2023; Zhou et al., 2023; Sakai et al., 2020b; Schuler et al., 2020; Gadgeel et al., 2022; Ramalingam et al., 2022; Song et al., 2022; Anagnostou et al., 2023; and Park et al., 2020 [45,47,48,49,51,52,54,62,64,66,69,70,73,74,75,76,77,78,79]. The concern around unblinding in open clinical trials underscores a significant issue, as both the researchers and participants are aware of the assigned treatments, potentially influencing the outcomes and adherence to treatment.
The blinding of the outcome assessment (detection bias) was mainly classified as low risk, suggesting that the outcome assessors were likely unaware of the intervention groups, thereby reducing the detection bias. This practice was consistent across most studies, reflecting a standard in the objective evaluation of clinical outcomes.
However, the domain concerning incomplete outcome data (attrition bias) exhibited a range of risks—low, unclear, and high—across the studies. A low risk indicates the transparent reporting of participant dropouts and proper handling of missing data. Conversely, studies with an unclear risk lacked detailed reporting, while a high risk indicated insufficient transparency, potentially undermining the study’s validity.
Selective reporting (reporting bias) varied, with most studies classified as low risk, implying that all predefined outcomes were likely reported and the study protocol was registered. However, some studies had an unclear risk due to insufficient information regarding the reporting of all expected outcomes.
Overall, the studies demonstrated a predominantly low risk of bias across most domains, indicating high methodological quality. Nevertheless, the variability in incomplete outcome data and selective reporting call for the cautious interpretation of the findings.
Figure 3 illustrates the bias risk analysis of the non-randomized clinical studies using the ROBINS-I tool (ROBINS-I tool | Cochrane Methods, accessed 18 September 2023) The results revealed that a significant proportion of the studies demonstrated high methodological quality. However, the studies by Han et al., 2022b [63]; Martini et al., 2022 [67]; Park et al., 2023 [61]; and Provencio et al., 2023 [68] showed an uncertain risk in domains 5 and 7 (bias due to missing data and bias in the selection of the reported result). This uncertainty underscores the need for the cautious interpretation of the findings. Although the general prevalence of a low risk reinforces the confidence in the accumulated evidence, the lack of transparency in these domains could influence the robustness of the final conclusions. Therefore, detailed assessment in future research is recommended to ensure robust clinical results.

4. Discussion

4.1. Most Significant Findings

In this study, a systematic review was conducted focusing on the identification and application of emerging biomarkers for the treatment of NSCLC. The most significant findings include the identification of ctDNA, bTMB, miRNAs, and PD-L1 as valuable tools for early diagnosis, treatment response prediction, and disease monitoring.
It has been demonstrated that ctDNA, due to its non-invasive nature and high sensitivity, can accurately detect genetic alterations, providing an effective biomarker for early intervention and personalized treatment [44,48,49,52,56]. miRNAs, despite facing specificity challenges, have shown potential as independent prognostic factors [84]. The bTMB has emerged as a key predictor of the response to immunotherapy, although its utility depends on the standardization and cost of sequencing technologies [53,54,57,58]. Additionally, PD-L1 has been identified as essential in predicting the efficacy of immune checkpoint inhibitors, facilitating the selection of patients who may benefit most from immunotherapy [55,71,76].
The economic impact and accessibility of these biomarkers are crucial factors that greatly affect their use in clinical practice, particularly when evaluating their cost-effectiveness in various healthcare environments. Implementing widespread ctDNA screening, for example, involves assessing not only the direct costs associated with the screening technologies themselves but also the potential healthcare savings from early detection and personalized treatment strategies [85,86,87]. These savings can be substantial, as early intervention typically leads to better patient outcomes and reduced treatment costs over time. A comprehensive analysis of the cost-effectiveness of these biomarkers is essential. By understanding the economic benefits and constraints associated with ctDNA, bTMB, and miRNA technologies, healthcare providers can make informed decisions about incorporating these tools into practice. This not only optimizes health outcomes but also ensures that the benefits of precision medicine are accessible to a broader range of patients, thereby reducing disparities in care and advancing public health goals [88,89,90].
Table 4 provides a comparative analysis of these biomarkers in NSCLC. ctDNA, known for its non-invasiveness and high sensitivity, is proposed as an effective tool for early diagnosis and treatment monitoring. However, its high cost and the complexity of interpretation limit its widespread use. The study by Postel et al., 2017 [91] highlighted the significant role of ddPCR and optimized NGS in the detection and monitoring of ctDNA. It elucidates how these advanced methodologies have markedly enhanced the ability to identify ctDNA with high sensitivity and specificity.
Ren et al., 2021 [48] showed that the dynamics of ctDNA during treatment could predict the efficacy of camrelizumab combined with chemotherapy in advanced squamous NSCLC, significantly improving the PFS and OS. Yang et al., 2023 [49] found that the detectability of ALK fusions in ctDNA was associated with the prognosis, with specific variants like EML4-ALK linked to poorer PFS. Garon et al., 2023 [52] reported that baseline alterations in ctDNA were prognostic indicators in patients with metastatic NSCLC harboring active EGFR mutations, with improvements in PFS observed when treated with ramucirumab plus erlotinib. Zhang et al., 2024 [56] highlighted that ctDNA clearance after two cycles of treatment was associated with longer PFS, emphasizing the predictive value of ctDNA in the combined treatment of sintilimab with nab-paclitaxel/carboplatin. Tan et al., 2024 [44] demonstrated that the reduction and clearance of ctDNA mutations, including EGFR and T790M, post-therapy were linked to longer PFS and OS in the alternating treatment of osimertinib and gefitinib. The studies by Goh et al., 2023 [101] and Cao et al., 2023 [102] have consistently demonstrated that reductions in ctDNA levels post-treatment are indicative of better PFS and OS. Tostes et al., 2023 [103] emphasized ctDNA as a dynamic biomarker reflecting changes in tumor burden and predicting therapy responses. Cao et al., 2023 [102] advocated for a multi-biomarker strategy, validated by the systematic review’s findings, where the integration of ctDNA with other biomarkers like the bTMB provided a more reliable prediction model for treatment responses. Comprehensive profiling aids in customizing therapy, as exemplified by Martini et al. (2022) [67], who found that specific gut microbiota species identified through ctDNA analysis were associated with longer PFS.
The significance of genetic mutations within ctDNA, particularly those in the EGFR or ALK genes, has been highlighted for their impact on clinical outcomes. Goh et al., 2023 [101] noted these mutations as crucial prognostic markers, a finding supported by Garon et al., 2023 [52], who observed that activating EGFR mutations correlated with shorter PFS.
These findings illustrate that ctDNA serves as a non-invasive biomarker for the monitoring of treatment responses and adjustment of therapeutic strategies in real time. It can also act as a predictive indicator of the treatment efficacy, particularly in targeted therapies and combinations of chemotherapy and immunotherapy [44,72,104].
The predictive efficacy of ctDNA varies according to the mutation type (e.g., EGFR, ALK), tumor stage, and histological subtype distribution. Additionally, the presence of co-alterations in genes and the clinical contexts of patients, such as their smoking history and ECOG performance status, can influence the outcomes. Elevated levels of ctDNA in serum are associated with a lower probability of a response to immunotherapy and generally poorer survival outcomes compared to patients with lower ctDNA levels, suggesting the potential of ctDNA as both a predictive and prognostic biomarker in treatment, including immunotherapy [18,105].
Regarding PD-L1, this biomarker is crucial in predicting the efficacy of checkpoint inhibitors such as anti-PD-1 and anti-PD-L1 antibodies. Studies have shown that patients with high PD-L1 expression generally respond better to these treatments, leading to higher survival rates. This marker aids in the selection of patients who are likely to benefit most from immunotherapy, thus personalizing the therapeutic options and improving the clinical outcomes [20,106].
Several key studies illustrate the clinical utility of PD-L1. Jiang et al., 2021 [55] found that PD-L1 expression was not predictive of PFS in treatment with toripalimab plus chemotherapy in EGFR-mutant NSCLC. Zhou et al., 2023 [71] observed high concordance between the SP263 and 22C3 assays for PD-L1 expression, with both predicting the efficacy of adjuvant atezolizumab in early NSCLC. Gadgeel et al., 2022 [76] reported that atezolizumab showed better survival compared to docetaxel, regardless of the PD-L1 status, with a greater benefit observed in patients with high PD-L1 expression.
PD-L1 is crucial in selecting patients for immunotherapy, helping to identify those who may benefit most from ICIs. The accurate measurement of PD-L1 can guide therapeutic decisions and optimize clinical outcomes. However, the PD-L1 expression and its association with the treatment response vary with the tumor microenvironment and histological subtype distribution, influencing the treatment response and survival. The expression of PD-L1 as a predictive biomarker for immunotherapy is complicated by issues such as variable detection antibodies and differing immunohistochemistry (IHC) cut-offs. While PD-L1 protein detection by IHC can enrich the response to anti-PD-L1 blockade, it is not absolute [107,108,109].
Studies have shown that patients with NSCLC who overexpress PD-L1 tend to respond more favorably to immunotherapy and demonstrate higher survival rates compared to those whose tumors do not express or have low expression of PD-L1. This underscores the importance of evaluating PD-L1 expression as a predictive biomarker for the selection of candidates for immunotherapy [71,83,110]. However, its sensitivity varies depending on the dynamics of the immune and tumor microenvironments, which can change over time and with treatment. Despite its high specificity in identifying patients likely to respond to ICIs, PD-L1 expression alone is not always indicative of treatment success [103,110].
miRNAs have emerged as significant biomarkers in NSCLC. Particular miRNAs, like miRNA-21 and miRNA-155, are notable predictors of the response to immunotherapy and survival. Xu et al., 2019 [84] found that the plasma levels of miR-32 correlated with the chemotherapy response and prognosis in NSCLC patients treated with platinum-based chemotherapy. Similarly, Ren et al., 2021 [48] observed correlations between miRNA-21 and miRNA-155 and the immunotherapy response and survival outcomes. Li et al., 2017 [96] found that miR-21-5p and miR-30d-5p were independent prognostic factors for overall survival in NSCLC, while Liu et al., 2012 [94] indicated that elevated levels of serum miR-21 and tumor miR-200c were associated with a poor prognosis in NSCLC patients. Wu et al., 2014 [95] demonstrated that high miR-19b and low miR-146a expression in NSCLC tissue correlated with a higher TNM stage, lymph node metastasis, and decreased survival rates.
Their assessment provides valuable information for patient stratification and the optimization of treatment regimens based on specific molecular profiles. Consequently, miRNAs have the potential to serve as non-invasive and useful markers for the stratification of patients and prediction of outcomes in the context of immunotherapy [22,104,111,112]. However, the quantification of miRNAs presents challenges due to their low abundance and high susceptibility to degradation. This complexity is further compounded by the need to normalize their levels against stable reference miRNAs and the significant discrepancies caused by the different detection platforms, such as qRT-PCR and microarrays. Standardizing the protocols for the selection of reference miRNAs based on the tissue origin and pathological conditions is crucial to increase the reliability of these analyses [21,111,113,114].
The interaction between the microbiota, particularly the gut microbiota, and the patient’s immune system plays a critical role in modulating the responses to treatments such as immunotherapy [115,116]. Recent research has highlighted that the composition and diversity of the microbiota can directly influence therapeutic outcomes, affecting both the efficacy and toxicity of the administered treatments in patients with NSCLC [117,118]. In the analysis of the gut microbiome, miRNAs, and their interaction with NSCLC, an interesting perspective emerges on the potential of these biological entities as biomarkers and their modulatory effects on the treatment efficacy, especially in targeted therapies and immunotherapies [119,120].
For instance, Martini et al., 2022 [67] demonstrated that certain intestinal bacteria, such as butyrate producers (Agathobacter and Blautia), were associated with improved PFS in NSCLC patients treated with combined therapies including cetuximab and avelumab. This relationship suggests that the microbiota not only affects carcinogenesis but also modulates the immune response, which can enhance or inhibit the efficacy of treatments based on ICIs. Comparatively, Shah and Ng, 2023 [121] emphasize how the gut microbiota influences the response to ICIs in NSCLC, associating microbiota diversity with greater treatment efficacy. These findings support the notion that modifying the microbiota could increase the sensitivity of patients who are initially resistant to ICIs, thus opening new avenues for personalized therapies.
Additionally, the role of miRNAs in this microbiota–NSCLC dynamic is crucial. miRNAs play a significant role in modulating immunity, inflammation, and cellular stress responses—all processes that are dysregulated in NSCLC. Casciaro et al., 2020 [122] suggest that miRNAs can influence the composition of the microbiota and vice versa, where changes in the microbiota might alter the expression of host cell miRNAs, affecting cancer progression and treatment responses. For example, alterations in specific miRNAs could result in changes in intestinal permeability and antigen presentation capacity, directly influencing the efficacy of immunotherapy in NSCLC patients, as discussed by Martini et al., 2022 [67] and supported by Chrysostomou et al., 2023 [123], who explore how miRNAs derived from the microbiota can be crucial in regulating the immune response and tumor progression.
Understanding the interaction between miRNA, the microbiota, and NSCLC offers considerable potential to improve outcomes in NSCLC, particularly in personalized therapies. The use of probiotics, prebiotics, or dietary changes to alter the microbiota, along with interventions that modify the expression of specific miRNAs, could not only enhance the response to immunotherapy but also reduce the toxicity associated with conventional treatments [123,124]. The individual variability in the microbiota composition and miRNA profiles suggests that personalized treatments could be developed around these differences, as the same intervention might have varied effects on different patients [21,125]. To develop effective and personalized interventions, a deeper understanding of the interactions among the microbiota, miRNAs, and NSCLC is essential.
Casciaro et al., 2020 [122] and Shah and Ng, 2023 [121] recommend that future studies focus on identifying combined signatures of miRNAs and the microbial composition that could serve as predictive biomarkers and guides for personalized therapies. Allegra et al., 2020 [126] explore how the microbiota can impact miRNA expression (miR-21, miR-155, miR-146a, miR-223), which, in turn, regulates gene expression, which affects cancer progression and the response to treatment. The study highlights that miR-21 and miR-155 are often associated with promoting inflammation and cancer progression, making them targets for therapies aimed at reducing their expression to suppress tumor growth. Meanwhile, miR-146a and miR-223 typically act as tumor suppressors by regulating genes involved in inflammatory processes; thus, enhancing their activity could be beneficial in controlling NSCLC’s progression. Allegra et al. suggest that antagomirs (anti-miRNA oligonucleotides) could specifically inhibit these dysregulated miRNAs, providing a direct method to enhance the treatment responses, particularly in personalizing therapy based on individual microbiota and miRNA profiles.
Finally, the bTMB has emerged as a significant biomarker in predicting the efficacy of immunotherapy combined with chemotherapy in NSCLC. Several key studies have highlighted its clinical utility. Han et al., 2022 [63] found that a reduction in the bTMB at six weeks could serve as a potential predictive biomarker for the regimen of sintilimab plus docetaxel in advanced NSCLC. Si et al., 2021 [54] reported that a high bTMB (≥20 mutations/Mb) predicted clinical benefits with durvalumab plus tremelimumab compared to chemotherapy. Kim et al., 2022 [57] showed that a bTMB ≥ 16 was associated with a higher objective response rate (ORR) and, in exploratory analyses, greater overall survival (OS) compared to a bTMB < 16. Conversely, Peters et al., 2022 [58] noted that a primary endpoint benefit in PFS was not achieved with a bTMB ≥ 16 for atezolizumab versus chemotherapy.
The bTMB can predict the efficacy of combined immunotherapy and chemotherapy, aiding in the selection of patients for specific treatments [52]. However, the mutational burden and its impact on the treatment response vary depending on the mutation type, the frequency of co-alterations, and the histological subtype distribution. This heterogeneity in the bTMB can influence treatment response prediction and clinical outcomes. In patients treated with specific therapies, such as sintilimab plus docetaxel and durvalumab plus tremelimumab, the bTMB has been identified as a predictive and prognostic indicator. A high bTMB indicates a clinical benefit with durvalumab plus tremelimumab compared to conventional chemotherapy. Recent studies have highlighted the need for precision in measuring the bTMB, influenced by technical and sample factors, underlining the importance of standardizing and validating the assessment methods to ensure their clinical utility [53,54,63,81].
The precision of TMB assessment is crucial in predicting and prognosticating tumor behavior. Nie et al., 2020 [81] found that a low bTMB was associated with a survival advantage in NSCLC patients treated with docetaxel, indicating the potential of the bTMB as a prognostic and predictive biomarker. Si et al., 2020 [54] further validated this by identifying a bTMB cut-off of ≥20 mutations per megabase as predictive of a clinical benefit with durvalumab plus tremelimumab in first-line metastatic NSCLC treatment. Schuurbiers et al., 2022 [97] highlighted the challenges in assessing the bTMB, including the need for a minimally sized bTMB panel and the influence of technical and sample-related factors. Raiber-Moreau et al., 2023 [98] emphasized the importance of developing and validating bTMB reference standards to ensure the accuracy and reproducibility of bTMB measurement. These studies collectively underscore the need for precision in assessing the bTMB to enhance the predictive and prognostic value of this biomarker.
Harmonizing the gene panels used for the sequencing of the bTMB by standardizing the size and the genes included is suggested to decrease the results’ variability and refine the classification of mutations as either drivers or passengers.

4.2. Quality of Evidence

The quality of the evidence obtained from these studies was critical in validating the reliability of the biomarkers in a clinical setting. Most RCTs showed a low risk of bias in the generation of random sequences and concealment of allocation, indicating robust randomization processes that minimized selection bias. However, the risk of performance bias was significantly high due to the open nature of many trials, where the participants and investigators knew the treatment assignments, which could influence the outcomes. Although most studies ensured a low probability of bias in the outcome assessment, inconsistencies in terms of incomplete outcome data and selective reporting underline the need for more transparent reporting practices and rigorous methodologies to ensure the applicability and accuracy of these biomarkers in medical practice. This includes a strong focus on the validation of microRNAs and other emerging biomarkers, such as those related to the gut microbiota, which could offer new perspectives in the personalized treatment of NSCLC.
A critical issue is the inconsistency of biomarkers as measurable indicators used to distinguish precisely and objectively between a normal biological state, pathological condition, and response to a specific therapeutic intervention. The development of biomarkers presents a significant challenge in cancer due to its complexity, sensitivity, and therapy resistance. The complex interplay of molecular pathways makes it necessary to use multiple markers to create sensitive and specific biomarkers that can represent a type of cancer pathogenesis and predict treatment responses and outcomes [127].
Despite the potential of ctDNA to effectively monitor therapeutic responses and disease progression, many studies, like those in phase II clinical trials by Han et al., 2022 [63], are not yet concluded. These studies suggest that the state of ctDNA and the elimination of ctDNA mutations serve as predictive biomarkers for treatments combining sintilimab with docetaxel in patients with advanced, pre-treated NSCLC [60].
The heterogeneity of the study designs and evaluation methods, as seen in the research by Goh et al., 2023 [101], indicates that the responses are highly variable and influenced by individual tumor characteristics, such as PD-L1 expression (also known as the tumor proportion score; TPS), the tumor mutational burden (TMB), tumor-infiltrating lymphocytes (TIL), the host’s immune system, and molecular signatures [124]. Similarly, the studies by Cao et al., 2023 [102] found that patients with resected NSCLC and high levels of PD-L1 exhibited lower survival rates compared to those with low levels. Conversely, inhibitors of PD-L1 or PD-1 can substantially increase patient survival, and detectable perioperative ctDNA is also correlated with poorer survival outcomes. Elevated CEA levels in circulation before and after surgery predicted significantly reduced survival outcomes [102].
This diversity in the evaluation methods underscores the need for a more standardized approach in research. Defining and standardizing the key parameters of the most promising biomarkers is essential to enable all stakeholders to make meaningful observations and inferences about the efficacy of seemingly similar agents and combinations in various settings. Several studies have demonstrated that a higher TMB is associated with better outcomes in various types of tumors, including NSCLC, colorectal cancer, bladder cancer, and melanoma [128].
Predicting the response to immunotherapy is crucial to identify patients who are likely to benefit from costly therapy and to avoid subjecting those who do not benefit from ICIs to unnecessary adverse effects. However, the FDA has not approved any circulating biomarker to predict the response to immunotherapy. Patients with high sPD-L1, low PD1+ CD8+, and low NK cell counts were significantly associated with poorer PFS [101].
Researchers must strive for transparency and thoroughness in reporting their methodologies. Standard practices should include detailed descriptions of the randomization processes and blinding methods and the complete reporting of outcome data. Integrating these biomarkers into standard clinical pathways could maximize their potential in enhancing the treatment precision, thereby fostering significant advances in oncology and improving patient outcomes and quality of life.

4.3. Future Directions and Clinical Implications

Advances in biomarker research are fundamental in enhancing diagnostic procedures and treatment protocols in oncology, particularly for complex conditions such as NSCLC. Future research should focus on establishing standardized protocols for the use of biomarkers and their integration into clinical pathways, which promises to significantly elevate the accuracy of diagnostics and the personalization of treatment strategies, thereby improving patient outcomes [129].
These studies must encompass all stages of biomarker use, from detection methods and machine calibration to data interpretation and report formatting. Technologies such as digital PCR and NGS offer profound insights into genetic and epigenetic modifications; however, their results are only as reliable as the standards used to calibrate them. Developing universal calibration standards that are uniformly applied across different platforms and laboratories can minimize the variability and enhance the reproducibility of the results [129,130].
Moreover, integrating biomarkers into clinical pathways also requires the exploration of their systemic implications. For instance, understanding how the early detection of mutations through ctDNA impacts long-term survival rates and quality of life requires longitudinal studies that track these outcomes over extended periods. Future research should focus on standardizing analytical methods and evaluating ctDNA in various clinical contexts and treatment types to validate its utility as a universal biomarker [131]. Similarly, personalizing chemotherapy and immunotherapy regimens based on the bTMB and microRNA profiles could revolutionize treatment paradigms [97,112]. Additional studies are needed to refine the cut-off points for the bTMB and evaluate its predictive efficacy in different treatment combinations, with an emphasis on genetic heterogeneity and the impact on treatment responses [132,133]. Future research on miRNAs should focus on identifying specific profiles associated with the treatment response and disease progression, as well as developing targeted therapies that modulate miRNA levels to enhance the treatment efficacy [134,135]. Regarding PD-L1, further research is needed to understand the heterogeneity in PD-L1 expression and its impact on the treatment response, exploring therapeutic combinations that can overcome resistance in specific subgroups [99,110].
The incorporation of nanotechnology, biotechnology, and AI further enhances the potential of biomarkers in NSCLC. Nanotechnology can improve drug delivery systems, making them more effective by targeting specific cells and reducing side effects. For instance, nanoparticles can be designed to release drugs in response to specific biological signals or environmental conditions, thus improving the precision of drug delivery at tumor sites [129,136,137,138]. Meanwhile, biotechnology plays a crucial role in the development of biomarkers and therapeutic targets. Techniques like CRISPR and gene editing allow for the manipulation of genetic material in cells, aiding in the creation of more effective models for the study of cancer and development of targeted therapies. For example, gene editing could be used to modify immune cells to enhance their ability to recognize and destroy cancer cells, a method currently being explored with CAR-T cell therapies [139,140,141].
AI complements these technologies by providing advanced data analysis capabilities that can predict treatment outcomes, optimize treatment plans, and even identify new potential biomarkers from vast datasets [129]. AI algorithms are capable of analyzing complex patterns in data that would be impossible to efficiently process by humans. For instance, AI can analyze imaging data to distinguish between benign and malignant lesions with high precision or predict which patients are more likely to respond to certain therapies based on historical data [129,142].
These technologies not only refine but also expand the current landscape of NSCLC treatment by facilitating the development of new targets and therapeutic strategies. This harmonious integration of biomarkers with cutting-edge technologies promises to revolutionize oncological practice by improving the precision and personalization of care. Eventually, this will lead to more effective treatments, the better management of treatment side effects, and improved survival rates for patients. By combining detailed genetic, molecular, and imaging data with innovative treatment methods, clinicians can provide more targeted, efficient, and patient-friendly cancer care. This approach underscores the potential of modern oncology to transform the nature of cancer treatment, ensuring that patients receive the most effective interventions specifically tailored to their unique disease profiles.

5. Conclusions

This systematic review emphasizes the transformative role of ctDNA and other biomarkers in the management of NSCLC, integrating the findings from 37 diverse studies to substantiate their effectiveness in personalized medicine. It highlights ctDNA’s utility in prognostic evaluations and real-time therapy monitoring, allowing for adaptive treatment strategies, particularly with immunotherapies and targeted therapies, where PD-L1 expression and specific genetic alterations such as ALK fusions and EGFR mutations are pivotal. The review also points to emerging biomarkers like microRNA profiles and the gut microbiota, which broaden the scope of immunotherapy’s efficacy. Collectively, these biomarkers not only refine the diagnostic and therapeutic protocols but also underscore the necessity for their standardized integration into clinical practice to optimize outcomes and reduce therapeutic redundancies. This synthesis confirms their prognostic and predictive value, advocating for ongoing innovation in biomarker research to improve NSCLC management, patient survival, and quality of life.

Author Contributions

Conceptualization, Y.L. and J.C.R.; methodology, Y.L. and J.C.R.; validation, Y.L., D.M.G. and A.P.L.; formal analysis, Y.L. and J.C.R.; investigation, Y.L. and J.C.R.; resources, Y.L.; data curation, Y.L. and J.C.R.; writing—original draft preparation Y.L., J.F.M.P. and J.C.R.; writing—review and editing, Y.L., D.M.G., A.P.L. and J.F.M.P.; visualization J.C.R.; supervision Y.L.; project administration, Y.L.; funding acquisition Y.L. All authors have read and agreed to the published version of the manuscript.

Funding

This research has been funded by Dirección General de Investigaciones de la Universidad Santiago de Cali, Convocatoria Interna No. 01-2024, Proyecto de fortalecimiento de grupo de investigación en salud integral, and grant number DGI-COCEIN No. 934-621121-3384.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We thank the Universidad Santiago de Cali for their financial and academic support in the preparation of this manuscript and give its consent for recognition in this study and publication.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

NSCLCNon-Small-Cell Lung Cancer
SCLCSmall Cell Lung Cancer
ctDNACirculating Tumor DNA
miRNAmicroRNA
bTMBBlood Tumor Mutational Burden
NGSNext-Generation Sequencing
EGFREpidermal Growth Factor Receptor
ALKAnaplastic Lymphoma Kinase
ROS1ROS Proto-Oncogene 1, Receptor Tyrosine Kinase
KRASKirsten Rat Sarcoma Viral Oncogene Homolog
PD-1Programmed Death-1
PD-L1Programmed Death Ligand 1
CTComputed Tomography
MRIMagnetic Resonance Imaging
PETPositron Emission Tomography
FISHFluorescence In Situ Hybridization
PCRPolymerase Chain Reaction
IHCImmunohistochemistry
TKIsTyrosine Kinase Inhibitors
EMTEpithelial–Mesenchymal Transition
METMesenchymal Epithelial Transition
HER2Human Epidermal Growth Factor Receptor 2
VEGFVascular Endothelial Growth Factor
FDAU.S. Food and Drug Administration
NIHNational Institutes of Health
ICBsImmune Checkpoint Inhibitors
NTRKNeurotrophic Tyrosine Kinase Receptor
TNMTumor, Node, Metastasis
TMETumor Microenvironment
CEACarcinoembryonic Antigen
CA125Carbohydrate Antigen 125
STK11Serine/Threonine Kinase 11
DDR2Discoidin Domain Receptor 2
RETRearranged during Transfection
JAK-STATJanus Kinase Signal Transducer and Activator of Transcription
RAS/MAPKRat Sarcoma/Mitogen-Activated Protein Kinase
PI3K/AKT/mTORPhosphoinositide 3-Kinase/Protein Kinase B/Mammalian Target of Rapamycin
MEK/ERKMitogen-Activated Protein Kinase Kinase/Extracellular Signal-Regulated Kinase
BRAFB-Raf Proto-Oncogene, Serine/Threonine Kinase
TUBB3Tubulin Beta-3
TGF-βTransforming Growth Factor Beta
LAG-3Lymphocyte Activation Gene-3
MDSMultidimensional Scaling
UMAPUniform Manifold Approximation and Projection
GEOGene Expression Omnibus
TCGAThe Cancer Genome Atlas
AMPAssociation for Molecular Pathology
CAPCollege of American Pathologists
IASLCInternational Association for the Study of Lung Cancer
ASCOAmerican Society of Clinical Oncology
ESMOEuropean Society for Medical Oncology
NCCNNational Comprehensive Cancer Network
FGFR1Fibroblast Growth Factor Receptor 1
TIM-3T-Cell Immunoglobulin and Mucin Domain Containing-3
TMBTumor Mutational Burden
OPNOsteopontin
DNADeoxyribonucleic Acid
SNPsSingle-Nucleotide Polymorphisms
FDA-NIHFDA-NIH Biomarker Working Group
NKX2-1Thyroid Transcription Factor-1
TTF-1Thyroid Transcription Factor-1
NAPSANapsin A
CYFRA 21-1Cytokeratin 19 Fragment
SCCASquamous Cell Carcinoma Antigen
m6AN6-Methyladenine
SFTA2Surfactant Protein A2
KIAA1522KIAA1522 Protein
PFSProgression-Free Survival
NLRNeutrophil-to-Lymphocyte Ratio
RCTsRandomized Clinical Trials
MPRMajor Pathological Response
TPSTumor Proportion Score
MDRMinimal Residual Disease
hVAFHigh Variant Allele Frequency
VAFSDVariant Allele Frequency Standard Deviation
RECISTResponse Evaluation Criteria In Solid Tumors
ORRObjective Response Rate
BRCA2Breast Cancer 2, Early Onset
DSPPDentin Sialophosphoprotein
CTCFCCCTC-Binding Factor
OVOL2Ovo-Like Transcriptional Repressor 2
FFPEFormalin-Fixed Paraffin-Embedded
ABCPAtezolizumab Bevacizumab Carboplatin Paclitaxel
BCPBevacizumab Carboplatin Paclitaxel
KEAP1Kelch-Like ECH-Associated Protein
TP53Tumor Protein p53
LASSOLeast Absolute Shrinkage and Selection Operator
ddPCRDroplet Digital Polymerase Chain Reaction
LP5252-Gene Expression Histology Classifier
BRAFV600EBRAF Gene Mutation V600E
ASOsAntisense Oligonucleotides
siRNAsSmall Interfering RNAs

Appendix A

Reference search algorithms:
(Non small cell lung cancer) AND (Biomarker); ((Biomarker) AND (Predictive)) AND (Non small cell lung cancer); Non small cell lung cancer biomarker monitoring; ((Non small cell lung cancer) AND (biomarker)) AND (monitoring); (((non small cell lung cancer) AND (biomarkers)) AND (immunotherapy)) AND (chemotherapy).

References

  1. Molina, J.R.; Yang, P.; Cassivi, S.D.; Schild, S.E.; Adjei, A.A. Non–Small Cell Lung Cancer: Epidemiology, Risk Factors, Treatment, and Survivorship. Mayo Clin. Proc. 2008, 83, 584–594. [Google Scholar] [CrossRef] [PubMed]
  2. Gridelli, C.; Rossi, A.; Carbone, D.P.; Guarize, J.; Karachaliou, N.; Mok, T.; Petrella, F.; Spaggiari, L.; Rosell, R. Non-Small-Cell Lung Cancer. Nat. Rev. Dis. Primers 2015, 1, 15009. [Google Scholar] [CrossRef] [PubMed]
  3. Knight, S.; Crosbie, P.A.; Balata, H.; Chudziak, J.; Hussell, T.; Dive, C. Progress and Prospects of Early Detection in Lung Cancer. Open Biol. 2017, 7, 170070. [Google Scholar] [CrossRef]
  4. Polanco, D.; Pinilla, L.; Gracia-Lavedan, E.; Mas, A.; Bertran, S.; Fierro, G.; Seminario, A.; Gómez, S.; Barbé, F. Prognostic Value of Symptoms at Lung Cancer Diagnosis: A Three-Year Observational Study. J. Thorac. Dis. 2021, 13, 1485–1494. [Google Scholar] [CrossRef] [PubMed]
  5. Alduais, Y.; Zhang, H.; Fan, F.; Chen, J.; Chen, B. Non-Small Cell Lung Cancer (NSCLC): A Review of Risk Factors, Diagnosis, and Treatment. Medicine 2023, 102, e32899. [Google Scholar] [CrossRef] [PubMed]
  6. Petrella, F.; Rizzo, S.; Attili, I.; Passaro, A.; Zilli, T.; Martucci, F.; Bonomo, L.; Del Grande, F.; Casiraghi, M.; De Marinis, F.; et al. Stage III Non-Small-Cell Lung Cancer: An Overview of Treatment Options. Curr. Oncol. 2023, 30, 3160–3175. [Google Scholar] [CrossRef] [PubMed]
  7. Rodak, O.; Peris-Díaz, M.D.; Olbromski, M.; Podhorska-Okołów, M.; Dzięgiel, P. Current Landscape of Non-Small Cell Lung Cancer: Epidemiology, Histological Classification, Targeted Therapies, and Immunotherapy. Cancers 2021, 13, 4705. [Google Scholar] [CrossRef] [PubMed]
  8. Chen, P.; Liu, Y.; Wen, Y.; Zhou, C. Non-small Cell Lung Cancer in China. Cancer Commun. 2022, 42, 937–970. [Google Scholar] [CrossRef] [PubMed]
  9. Cardona, A.F.; Mejía, S.A.; Viola, L.; Chamorro, D.F.; Rojas, L.; Ruíz-Patiño, A.; Serna, A.; Martínez, S.; Muñoz, Á.; Rodríguez, J.; et al. Lung Cancer in Colombia. J. Thorac. Oncol. 2022, 17, 953–960. [Google Scholar] [CrossRef] [PubMed]
  10. Chen, Z.; Fillmore, C.M.; Hammerman, P.S.; Kim, C.F.; Wong, K.-K. Non-Small-Cell Lung Cancers: A Heterogeneous Set of Diseases. Nat. Rev. Cancer 2014, 14, 535–546. [Google Scholar] [CrossRef] [PubMed]
  11. Senosain, M.-F.; Massion, P.P. Intratumor Heterogeneity in Early Lung Adenocarcinoma. Front. Oncol. 2020, 10, 349. [Google Scholar] [CrossRef] [PubMed]
  12. Flury, D.V.; Minervini, F.; Kocher, G.J. Heterogeneity of Stage IIIA Non-Small Cell Lung Cancer—Different Tumours, Different Nodal Status, Different Treatment, Different Prognosis: A Narrative Review. Curr. Chall Thorac. Surg. 2022, 4, 13. [Google Scholar] [CrossRef]
  13. Palumbo, A.; De Oliveira Meireles Da Costa, N.; Bonamino, M.H.; Ribeiro Pinto, L.F.; Nasciutti, L.E. Genetic Instability in the Tumor Microenvironment: A New Look at an Old Neighbor. Mol. Cancer 2015, 14, 145. [Google Scholar] [CrossRef] [PubMed]
  14. Siveke, J.T. Fibroblast-Activating Protein: Targeting the Roots of the Tumor Microenvironment. J. Nucl. Med. 2018, 59, 1412–1414. [Google Scholar] [CrossRef] [PubMed]
  15. Wu, T.; Dai, Y. Tumor Microenvironment and Therapeutic Response. Cancer Lett. 2017, 387, 61–68. [Google Scholar] [CrossRef] [PubMed]
  16. Bevere, M.; Masetto, F.; Carazzolo, M.E.; Bettega, A.; Gkountakos, A.; Scarpa, A.; Simbolo, M. An Overview of Circulating Biomarkers in Neuroendocrine Neoplasms: A Clinical Guide. Diagnostics 2023, 13, 2820. [Google Scholar] [CrossRef] [PubMed]
  17. Planchard, D. Identification of Driver Mutations in Lung Cancer: First Step in Personalized Cancer. Targ. Oncol. 2013, 8, 3–14. [Google Scholar] [CrossRef]
  18. Zografos, E.; Dimitrakopoulos, F.-I.; Koutras, A. Prognostic Value of Circulating Tumor DNA (ctDNA) in Oncogene-Driven NSCLC: Current Knowledge and Future Perspectives. Cancers 2022, 14, 4954. [Google Scholar] [CrossRef] [PubMed]
  19. Fenizia, F.; Rachiglio, A.M.; Iannaccone, A.; Chicchinelli, N.; Normanno, N. Circulating Tumor Cells and ctDNA in NSCLC. In Oncogenomics; Elsevier: Amsterdam, The Netherlands, 2019; pp. 465–475. ISBN 978-0-12-811785-9. [Google Scholar]
  20. Patel, S.P.; Kurzrock, R. PD-L1 Expression as a Predictive Biomarker in Cancer Immunotherapy. Mol. Cancer Ther. 2015, 14, 847–856. [Google Scholar] [CrossRef] [PubMed]
  21. Latini, A.; Borgiani, P.; Novelli, G.; Ciccacci, C. miRNAs in Drug Response Variability: Potential Utility as Biomarkers for Personalized Medicine. Pharmacogenomics 2019, 20, 1049–1059. [Google Scholar] [CrossRef] [PubMed]
  22. Wang, Z.; Han, J.; Cui, Y.; Fan, K.; Zhou, X. Circulating microRNA-21 as Noninvasive Predictive Biomarker for Response in Cancer Immunotherapy. Med. Hypotheses 2013, 81, 41–43. [Google Scholar] [CrossRef] [PubMed]
  23. Wang, W.; Li, X.; Liu, C.; Zhang, X.; Wu, Y.; Diao, M.; Tan, S.; Huang, S.; Cheng, Y.; You, T. MicroRNA-21 as a Diagnostic and Prognostic Biomarker of Lung Cancer: A Systematic Review and Meta-Analysis. Biosci. Rep. 2022, 42, BSR20211653. [Google Scholar] [CrossRef] [PubMed]
  24. Mithoowani, H.; Febbraro, M. Non-Small-Cell Lung Cancer in 2022: A Review for General Practitioners in Oncology. Curr. Oncol. 2022, 29, 1828–1839. [Google Scholar] [CrossRef] [PubMed]
  25. Qian, F.; Yang, W.; Chen, Q.; Zhang, X.; Han, B. Screening for Early Stage Lung Cancer and Its Correlation with Lung Nodule Detection. J. Thorac. Dis. 2018, 10, S846–S859. [Google Scholar] [CrossRef] [PubMed]
  26. Marchetti, A.; Buttitta, F.; D’Angelo, E. Adjuvant Osimertinib Treatment in Patients with Early Stage NSCLC (IB-IIIA): Pathological Pathway Adaptations. Oncotarget 2022, 13, 456–463. [Google Scholar] [CrossRef] [PubMed]
  27. Sabari, J.K.; Santini, F.; Bergagnini, I.; Lai, W.V.; Arbour, K.C.; Drilon, A. Changing the Therapeutic Landscape in Non-Small Cell Lung Cancers: The Evolution of Comprehensive Molecular Profiling Improves Access to Therapy. Curr. Oncol. Rep. 2017, 19, 24. [Google Scholar] [CrossRef] [PubMed]
  28. Coco, S.; Truini, A.; Vanni, I.; Dal Bello, M.; Alama, A.; Rijavec, E.; Genova, C.; Barletta, G.; Sini, C.; Burrafato, G.; et al. Next Generation Sequencing in Non-Small Cell Lung Cancer: New Avenues Toward the Personalized Medicine. CDT 2015, 16, 47–59. [Google Scholar] [CrossRef] [PubMed]
  29. Das, S.; Dey, M.K.; Devireddy, R.; Gartia, M.R. Biomarkers in Cancer Detection, Diagnosis, and Prognosis. Sensors 2023, 24, 37. [Google Scholar] [CrossRef] [PubMed]
  30. Liberini, V.; Mariniello, A.; Righi, L.; Capozza, M.; Delcuratolo, M.D.; Terreno, E.; Farsad, M.; Volante, M.; Novello, S.; Deandreis, D. NSCLC Biomarkers to Predict Response to Immunotherapy with Checkpoint Inhibitors (ICI): From the Cells to In Vivo Images. Cancers 2021, 13, 4543. [Google Scholar] [CrossRef] [PubMed]
  31. Pretelli, G.; Spagnolo, C.C.; Ciappina, G.; Santarpia, M.; Pasello, G. Overview on Therapeutic Options in Uncommon EGFR Mutant Non-Small Cell Lung Cancer (NSCLC): New Lights for an Unmet Medical Need. Int. J. Mol. Sci. 2023, 24, 8878. [Google Scholar] [CrossRef] [PubMed]
  32. Simarro, J.; Pérez-Simó, G.; Mancheño, N.; Ansotegui, E.; Muñoz-Núñez, C.F.; Gómez-Codina, J.; Juan, Ó.; Palanca, S. Impact of Molecular Testing Using Next-Generation Sequencing in the Clinical Management of Patients with Non-Small Cell Lung Cancer in a Public Healthcare Hospital. Cancers 2023, 15, 1705. [Google Scholar] [CrossRef] [PubMed]
  33. Yang, Z.; Tam, K.Y. Combination Strategies Using EGFR-TKi in NSCLC Therapy: Learning from the Gap between Pre-Clinical Results and Clinical Outcomes. Int. J. Biol. Sci. 2018, 14, 204–216. [Google Scholar] [CrossRef] [PubMed]
  34. Melosky, B.; Kambartel, K.; Häntschel, M.; Bennetts, M.; Nickens, D.J.; Brinkmann, J.; Kayser, A.; Moran, M.; Cappuzzo, F. Worldwide Prevalence of Epidermal Growth Factor Receptor Mutations in Non-Small Cell Lung Cancer: A Meta-Analysis. Mol. Diagn. Ther. 2022, 26, 7–18. [Google Scholar] [CrossRef] [PubMed]
  35. Araghi, M.; Mannani, R.; Heidarnejad Maleki, A.; Hamidi, A.; Rostami, S.; Safa, S.H.; Faramarzi, F.; Khorasani, S.; Alimohammadi, M.; Tahmasebi, S.; et al. Recent Advances in Non-Small Cell Lung Cancer Targeted Therapy; an Update Review. Cancer Cell Int. 2023, 23, 162. [Google Scholar] [CrossRef] [PubMed]
  36. Wu, J.; Lin, Z. Non-Small Cell Lung Cancer Targeted Therapy: Drugs and Mechanisms of Drug Resistance. Int. J. Mol. Sci. 2022, 23, 15056. [Google Scholar] [CrossRef] [PubMed]
  37. Guo, H.; Zhang, J.; Qin, C.; Yan, H.; Liu, T.; Hu, H.; Tang, S.; Tang, S.; Zhou, H. Biomarker-Targeted Therapies in Non–Small Cell Lung Cancer: Current Status and Perspectives. Cells 2022, 11, 3200. [Google Scholar] [CrossRef] [PubMed]
  38. Saller, J.J.; Boyle, T.A. Molecular Pathology of Lung Cancer. Cold Spring Harb. Perspect. Med. 2022, 12, a037812. [Google Scholar] [CrossRef] [PubMed]
  39. Mehta, A.; Saifi, M.; Batra, U.; Suryavanshi, M.; Gupta, K. Incidence of ROS1-Rearranged Non-Small-Cell Lung Carcinoma in India and Efficacy of Crizotinib in Lung Adenocarcinoma Patients. LCTT 2020, 11, 19–25. [Google Scholar] [CrossRef] [PubMed]
  40. Moher, D.; Liberati, A.; Tetzlaff, J.; Altman, D.G. Preferred Reporting Items for Systematic Reviews and Meta-Analyses: The PRISMA Statement. PLoS. Med. 2009, 15, 264–269. [Google Scholar] [CrossRef]
  41. Cochrane Cochrane Handbook for Systematic Reviews of Interventions. Available online: https://training.cochrane.org/handbook (accessed on 9 June 2023).
  42. Sterne, J.A.; Hernán, M.A.; Reeves, B.C.; Savović, J.; Berkman, N.D.; Viswanathan, M.; Henry, D.; Altman, D.G.; Ansari, M.T.; Boutron, I.; et al. ROBINS-I: A Tool for Assessing Risk of Bias in Non-Randomised Studies of Interventions. BMJ 2016, 355, i4919. [Google Scholar] [CrossRef] [PubMed]
  43. Haddaway, N.R.; Page, M.J.; Pritchard, C.C.; McGuinness, L.A. PRISMA2020: An R Package and Shiny App for Producing PRISMA 2020-Compliant Flow Diagrams, with Interactivity for Optimised Digital Transparency and Open Synthesis. Campbell Syst. Rev. 2022, 18, e1230. [Google Scholar] [CrossRef] [PubMed]
  44. Tan, L.; Brown, C.; Mersiades, A.; Lee, C.K.; John, T.; Kao, S.; Newnham, G.; O’Byrne, K.; Parakh, S.; Bray, V.; et al. A Phase II Trial of Alternating Osimertinib and Gefitinib Therapy in Advanced EGFR-T790M Positive Non-Small Cell Lung Cancer: OSCILLATE. Nat. Commun. 2024, 15, 1823. [Google Scholar] [CrossRef] [PubMed]
  45. Papadimitrakopoulou, V.A.; Han, J.; Ahn, M.; Ramalingam, S.S.; Delmonte, A.; Hsia, T.; Laskin, J.; Kim, S.; He, Y.; Tsai, C.; et al. Epidermal Growth Factor Receptor Mutation Analysis in Tissue and Plasma from the AURA3 Trial: Osimertinib versus Platinum-pemetrexed for T790M Mutation-positive Advanced Non–Small Cell Lung Cancer. Cancer 2020, 126, 373–380. [Google Scholar] [CrossRef] [PubMed]
  46. Sakai, K.; Tsuboi, M.; Kenmotsu, H.; Yamanaka, T.; Takahashi, T.; Goto, K.; Daga, H.; Ohira, T.; Ueno, T.; Aoki, T.; et al. Tumor Mutation Burden as a Biomarker for Lung Cancer Patients Treated with Pemetrexed and Cisplatin (the JIPANG-TR). Cancer Sci. 2021, 112, 388–396. [Google Scholar] [CrossRef] [PubMed]
  47. Park, C.-K.; Cho, H.-J.; Choi, Y.-D.; Oh, I.-J.; Kim, Y.-C. A Phase II Trial of Osimertinib as the First-Line Treatment of Non–Small Cell Lung Cancer Harboring Activating EGFR Mutations in Circulating Tumor DNA: LiquidLung-O-Cohort 1. Cancer Res. Treat. 2021, 53, 93–103. [Google Scholar] [CrossRef] [PubMed]
  48. Ren, S.; Chen, J.; Xu, X.; Jiang, T.; Cheng, Y.; Chen, G.; Pan, Y.; Fang, Y.; Wang, Q.; Huang, Y.; et al. Camrelizumab Plus Carboplatin and Paclitaxel as First-Line Treatment for Advanced Squamous NSCLC (CameL-Sq): A Phase 3 Trial. J. Thorac. Oncol. 2022, 17, 544–557. [Google Scholar] [CrossRef] [PubMed]
  49. Yang, J.C.-H.; Liu, G.; Lu, S.; He, J.; Burotto, M.; Ahn, M.-J.; Kim, D.-W.; Liu, X.; Zhao, Y.; Vincent, S.; et al. Brigatinib Versus Alectinib in ALK-Positive NSCLC After Disease Progression on Crizotinib: Results of Phase 3 ALTA-3 Trial. J. Thorac. Oncol. 2023, 18, 1743–1755. [Google Scholar] [CrossRef]
  50. Riess, J.W.; Reckamp, K.L.; Frankel, P.; Longmate, J.; Kelly, K.A.; Gandara, D.R.; Weipert, C.M.; Raymond, V.M.; Keer, H.N.; Mack, P.C.; et al. Erlotinib and Onalespib Lactate Focused on EGFR Exon 20 Insertion Non-Small Cell Lung Cancer (NSCLC): A California Cancer Consortium Phase I/II Trial (NCI 9878). Clin. Lung Cancer 2021, 22, 541–548. [Google Scholar] [CrossRef] [PubMed]
  51. Lo Russo, G.; Sgambelluri, F.; Prelaj, A.; Galli, F.; Manglaviti, S.; Bottiglieri, A.; Di Mauro, R.M.; Ferrara, R.; Galli, G.; Signorelli, D.; et al. PEOPLE (NCT03447678), a First-Line Phase II Pembrolizumab Trial, in Negative and Low PD-L1 Advanced NSCLC: Clinical Outcomes and Association with Circulating Immune Biomarkers. ESMO Open 2022, 7, 100645. [Google Scholar] [CrossRef] [PubMed]
  52. Garon, E.B.; Reck, M.; Nishio, K.; Heymach, J.V.; Nishio, M.; Novello, S.; Paz-Ares, L.; Popat, S.; Aix, S.P.; Graham, H.; et al. Ramucirumab plus Erlotinib versus Placebo plus Erlotinib in Previously Untreated EGFR-Mutated Metastatic Non-Small-Cell Lung Cancer (RELAY): Exploratory Analysis of next-Generation Sequencing Results. ESMO Open 2023, 8, 101580. [Google Scholar] [CrossRef] [PubMed]
  53. Han, X.; Guo, J.; Tang, X.; Zhu, H.; Zhu, D.; Zhang, X.; Meng, X.; Hua, Y.; Wang, Z.; Zhang, Y.; et al. Efficacy and Safety of Sintilimab plus Docetaxel in Patients with Previously Treated Advanced Non-Small Cell Lung Cancer: A Prospective, Single-Arm, Phase II Study in China. J. Cancer Res. Clin. Oncol. 2023, 149, 1443–1451. [Google Scholar] [CrossRef] [PubMed]
  54. Si, H.; Kuziora, M.; Quinn, K.J.; Helman, E.; Ye, J.; Liu, F.; Scheuring, U.; Peters, S.; Rizvi, N.A.; Brohawn, P.Z.; et al. A Blood-Based Assay for Assessment of Tumor Mutational Burden in First-Line Metastatic NSCLC Treatment: Results from the MYSTIC Study. Clin. Cancer Res. 2021, 27, 1631–1640. [Google Scholar] [CrossRef] [PubMed]
  55. Jiang, T.; Wang, P.; Zhang, J.; Zhao, Y.; Zhou, J.; Fan, Y.; Shu, Y.; Liu, X.; Zhang, H.; He, J.; et al. Toripalimab plus Chemotherapy as Second-Line Treatment in Previously EGFR-TKI Treated Patients with EGFR-Mutant-Advanced NSCLC: A Multicenter Phase-II Trial. Sig. Transduct. Target Ther. 2021, 6, 355. [Google Scholar] [CrossRef] [PubMed]
  56. Zhang, M.; Zhang, G.; Niu, Y.; Zhang, G.; Ji, Y.; Yan, X.; Zhang, X.; Wang, Q.; Jing, X.; Wang, J.; et al. Sintilimab with Two Cycles of Chemotherapy for the Treatment of Advanced Squamous Non-Small Cell Lung Cancer: A Phase 2 Clinical Trial. Nat. Commun. 2024, 15, 1512. [Google Scholar] [CrossRef] [PubMed]
  57. Kim, E.S.; Velcheti, V.; Mekhail, T.; Yun, C.; Shagan, S.M.; Hu, S.; Chae, Y.K.; Leal, T.A.; Dowell, J.E.; Tsai, M.L.; et al. Blood-Based Tumor Mutational Burden as a Biomarker for Atezolizumab in Non-Small Cell Lung Cancer: The Phase 2 B-F1RST Trial. Nat. Med. 2022, 28, 939–945. [Google Scholar] [CrossRef] [PubMed]
  58. Peters, S.; Dziadziuszko, R.; Morabito, A.; Felip, E.; Gadgeel, S.M.; Cheema, P.; Cobo, M.; Andric, Z.; Barrios, C.H.; Yamaguchi, M.; et al. Atezolizumab versus Chemotherapy in Advanced or Metastatic NSCLC with High Blood-Based Tumor Mutational Burden: Primary Analysis of BFAST Cohort C Randomized Phase 3 Trial. Nat. Med. 2022, 28, 1831–1839. [Google Scholar] [CrossRef] [PubMed]
  59. Chaft, J.E.; Oezkan, F.; Kris, M.G.; Bunn, P.A.; Wistuba, I.I.; Kwiatkowski, D.J.; Owen, D.H.; Tang, Y.; Johnson, B.E.; Lee, J.M.; et al. Neoadjuvant Atezolizumab for Resectable Non-Small Cell Lung Cancer: An Open-Label, Single-Arm Phase II Trial. Nat. Med. 2022, 28, 2155–2161. [Google Scholar] [CrossRef] [PubMed]
  60. Shi, Y.; Wu, L.; Yu, X.; Xing, P.; Wang, Y.; Zhou, J.; Wang, A.; Shi, J.; Hu, Y.; Wang, Z.; et al. Sintilimab versus Docetaxel as Second-line Treatment in Advanced or Metastatic Squamous Non-small-cell Lung Cancer: An Open-label, Randomized Controlled Phase 3 Trial (ORIENT-3). Cancer Commun. 2022, 42, 1314–1330. [Google Scholar] [CrossRef] [PubMed]
  61. Park, C.-K.; Jun, H.R.; Oh, H.-J.; Lee, J.-Y.; Cho, H.-J.; Kim, Y.-C.; Lee, J.E.; Yoon, S.H.; Choi, C.M.; Lee, J.C.; et al. Evaluation of Blood Tumor Mutation Burden for the Efficacy of Second-Line Atezolizumab Treatment in Non-Small Cell Lung Cancer: BUDDY Trial. Cells 2023, 12, 1246. [Google Scholar] [CrossRef] [PubMed]
  62. Gu, W.; Zhang, H.; Lu, Y.; Li, M.; Yang, S.; Liang, J.; Ye, Z.; Li, Z.; He, M.; Shi, X.; et al. EGFR-TKI Combined with Pemetrexed versus EGFR-TKI Monotherapy in Advanced EGFR-Mutated NSCLC: A Prospective, Randomized, Exploratory Study. Cancer Res. Treat. 2023, 55, 841–850. [Google Scholar] [CrossRef] [PubMed]
  63. Han, X.; Tang, X.; Zhu, H.; Zhu, D.; Zhang, X.; Meng, X.; Hua, Y.; Wang, Z.; Zhang, Y.; Huang, W.; et al. Short-Term Dynamics of Circulating Tumor DNA Predicting Efficacy of Sintilimab plus Docetaxel in Second-Line Treatment of Advanced NSCLC: Biomarker Analysis from a Single-Arm, Phase 2 Trial. J. Immunother. Cancer 2022, 10, e004952. [Google Scholar] [CrossRef] [PubMed]
  64. Zhong, H.; Zhang, X.; Tian, P.; Chu, T.; Guo, Q.; Yu, X.; Yu, Z.; Li, Y.; Chen, L.; Liu, J.; et al. Tislelizumab plus Chemotherapy for Patients with EGFR -Mutated Non-Squamous Non-Small Cell Lung Cancer Who Progressed on EGFR Tyrosine Kinase Inhibitor Therapy. J. Immunother. Cancer 2023, 11, e006887. [Google Scholar] [CrossRef]
  65. García-Pardo, M.; Czarnecka-Kujawa, K.; Law, J.H.; Salvarrey, A.M.; Fernandes, R.; Fan, Z.J.; Waddell, T.K.; Yasufuku, K.; Liu, G.; Donahoe, L.L.; et al. Association of Circulating Tumor DNA Testing Before Tissue Diagnosis with Time to Treatment Among Patients With Suspected Advanced Lung Cancer: The ACCELERATE Nonrandomized Clinical Trial. JAMA Netw. Open 2023, 6, e2325332. [Google Scholar] [CrossRef] [PubMed]
  66. Nomura, S.; Goto, Y.; Mizutani, T.; Kataoka, T.; Kawai, S.; Okuma, Y.; Murakami, H.; Tanaka, K.; Ohe, Y. A Randomized Phase III Study Comparing Continuation and Discontinuation of PD-1 Pathway Inhibitors for Patients with Advanced Non-Small-Cell Lung Cancer (JCOG1701, SAVE Study). Jpn. J. Clin. Oncol. 2020, 50, 821–825. [Google Scholar] [CrossRef] [PubMed]
  67. Martini, G.; Ciardiello, D.; Dallio, M.; Famiglietti, V.; Esposito, L.; Corte, C.M.D.; Napolitano, S.; Fasano, M.; Gravina, A.G.; Romano, M.; et al. Gut Microbiota Correlates with Antitumor Activity in Patients with mCRC and NSCLC Treated with Cetuximab plus Avelumab. Int. J. Cancer 2022, 151, 473–480. [Google Scholar] [CrossRef] [PubMed]
  68. Provencio, M.; Serna-Blasco, R.; Nadal, E.; Insa, A.; García-Campelo, M.R.; Casal Rubio, J.; Dómine, M.; Majem, M.; Rodríguez-Abreu, D.; Martínez-Martí, A.; et al. Overall Survival and Biomarker Analysis of Neoadjuvant Nivolumab Plus Chemotherapy in Operable Stage IIIA Non–Small-Cell Lung Cancer (NADIM Phase II Trial). JCO 2022, 40, 2924–2933. [Google Scholar] [CrossRef] [PubMed]
  69. West, H.J.; McCleland, M.; Cappuzzo, F.; Reck, M.; Mok, T.S.; Jotte, R.M.; Nishio, M.; Kim, E.; Morris, S.; Zou, W.; et al. Clinical Efficacy of Atezolizumab plus Bevacizumab and Chemotherapy in KRAS- Mutated Non-Small Cell Lung Cancer with STK11, KEAP1, or TP53 Comutations: Subgroup Results from the Phase III IMpower150 Trial. J. Immunother. Cancer 2022, 10, e003027. [Google Scholar] [CrossRef] [PubMed]
  70. Lo Russo, G.; Prelaj, A.; Dolezal, J.; Beninato, T.; Agnelli, L.; Triulzi, T.; Fabbri, A.; Lorenzini, D.; Ferrara, R.; Brambilla, M.; et al. PEOPLE (NTC03447678), a Phase II Trial to Test Pembrolizumab as First-Line Treatment in Patients with Advanced NSCLC with PD-L1 <50%: A Multiomics Analysis. J. Immunother. Cancer 2023, 11, e006833. [Google Scholar] [CrossRef] [PubMed]
  71. Zhou, C.; Srivastava, M.K.; Xu, H.; Felip, E.; Wakelee, H.; Altorki, N.; Reck, M.; Liersch, R.; Kryzhanivska, A.; Oizumi, S.; et al. Comparison of SP263 and 22C3 Immunohistochemistry PD-L1 Assays for Clinical Efficacy of Adjuvant Atezolizumab in Non-Small Cell Lung Cancer: Results from the Randomized Phase III IMpower010 Trial. J. Immunother. Cancer 2023, 11, e007047. [Google Scholar] [CrossRef] [PubMed]
  72. Sakai, K.; Takahama, T.; Shimokawa, M.; Azuma, K.; Takeda, M.; Kato, T.; Daga, H.; Okamoto, I.; Akamatsu, H.; Teraoka, S.; et al. Predicting Osimertinib-treatment Outcomes through EGFR Mutant-fraction Monitoring in the Circulating Tumor DNA of EGFR T790M-positive Patients with Non-small Cell Lung Cancer (WJOG8815L). Mol. Oncol. 2021, 15, 126–137. [Google Scholar] [CrossRef]
  73. Redman, M.W.; Papadimitrakopoulou, V.A.; Minichiello, K.; Hirsch, F.R.; Mack, P.C.; Schwartz, L.H.; Vokes, E.; Ramalingam, S.; Leighl, N.; Bradley, J.; et al. Biomarker-Driven Therapies for Previously Treated Squamous Non-Small-Cell Lung Cancer (Lung-MAP SWOG S1400): A Biomarker-Driven Master Protocol. Lancet Oncol. 2020, 21, 1589–1601. [Google Scholar] [CrossRef] [PubMed]
  74. Hirsch, F.R.; Redman, M.W.; Moon, J.; Agustoni, F.; Herbst, R.S.; Semrad, T.J.; Varella-Garcia, M.; Rivard, C.J.; Kelly, K.; Gandara, D.R.; et al. EGFR High Copy Number Together With High EGFR Protein Expression Predicts Improved Outcome for Cetuximab-Based Therapy in Squamous Cell Lung Cancer: Analysis From SWOG S0819, a Phase III Trial of Chemotherapy With or Without Cetuximab in Advanced NSCLC. Clin. Lung Cancer 2022, 23, 60–71. [Google Scholar] [CrossRef] [PubMed]
  75. Schuler, M.; Berardi, R.; Lim, W.-T.; De Jonge, M.; Bauer, T.M.; Azaro, A.; Gottfried, M.; Han, J.-Y.; Lee, D.H.; Wollner, M.; et al. Molecular Correlates of Response to Capmatinib in Advanced Non-Small-Cell Lung Cancer: Clinical and Biomarker Results from a Phase I Trial. Ann. Oncol. 2020, 31, 789–797. [Google Scholar] [CrossRef] [PubMed]
  76. Gadgeel, S.; Hirsch, F.R.; Kerr, K.; Barlesi, F.; Park, K.; Rittmeyer, A.; Zou, W.; Bhatia, N.; Koeppen, H.; Paul, S.M.; et al. Comparison of SP142 and 22C3 Immunohistochemistry PD-L1 Assays for Clinical Efficacy of Atezolizumab in Non–Small Cell Lung Cancer: Results From the Randomized OAK Trial. Clin. Lung Cancer 2022, 23, 21–33. [Google Scholar] [CrossRef] [PubMed]
  77. Ramalingam, S.S.; Novello, S.; Guclu, S.Z.; Bentsion, D.; Zvirbule, Z.; Szilasi, M.; Bernabe, R.; Syrigos, K.; Byers, L.A.; Clingan, P.; et al. Veliparib in Combination With Platinum-Based Chemotherapy for First-Line Treatment of Advanced Squamous Cell Lung Cancer: A Randomized, Multicenter Phase III Study. JCO 2021, 39, 3633–3644. [Google Scholar] [CrossRef] [PubMed]
  78. Song, Z.; Li, Y.; Chen, S.; Ying, S.; Xu, S.; Huang, J.; Wu, D.; Lv, D.; Bei, T.; Liu, S.; et al. Efficacy and Safety of Pyrotinib in Advanced Lung Adenocarcinoma with HER2 Mutations: A Multicenter, Single-Arm, Phase II Trial. BMC Med. 2022, 20, 42. [Google Scholar] [CrossRef] [PubMed]
  79. Anagnostou, V.; Ho, C.; Nicholas, G.; Juergens, R.A.; Sacher, A.; Fung, A.S.; Wheatley-Price, P.; Laurie, S.A.; Levy, B.; Brahmer, J.R.; et al. ctDNA Response after Pembrolizumab in Non-Small Cell Lung Cancer: Phase 2 Adaptive Trial Results. Nat. Med. 2023, 29, 2559–2569. [Google Scholar] [CrossRef] [PubMed]
  80. Park, C.; Lee, S.; Lee, J.C.; Choi, C.; Lee, S.Y.; Jang, T.; Oh, I.; Kim, Y. Phase II OPEN-LABEL Multicenter Study to Assess the Antitumor Activity of Afatinib in Lung Cancer Patients with Activating Epidermal Growth Factor Receptor Mutation from Circulating Tumor DNA: LIQUID-LUNG-A. Thoracic. Cancer 2021, 12, 444–452. [Google Scholar] [CrossRef] [PubMed]
  81. Nie, W.; Qian, J.; Xu, M.-D.; Gu, K.; Qian, F.-F.; Lu, J.; Zhang, X.-Y.; Wang, H.-M.; Yan, B.; Zhang, B.; et al. Prognostic and Predictive Value of Blood Tumor Mutational Burden in Patients With Lung Cancer Treated With Docetaxel. J. Natl. Compr. Cancer Netw. 2020, 18, 582–589. [Google Scholar] [CrossRef] [PubMed]
  82. Willis, C.; Bauer, H.; Au, T.H.; Menon, J.; Unni, S.; Tran, D.; Rivers, Z.; Akerley, W.; Schabath, M.B.; Badin, F.; et al. Real-World Survival Analysis by Tumor Mutational Burden in Non-Small Cell Lung Cancer: A Multisite U.S. Study. Oncotarget 2022, 13, 257–270. [Google Scholar] [CrossRef] [PubMed]
  83. Zheng, Y.; Yao, M.; Yang, Y. Higher Tumor Mutation Burden Was a Predictor for Better Outcome for NSCLC Patients Treated with PD-1 Antibodies: A Systematic Review and Meta-Analysis. SLAS Technol. 2021, 26, 605–614. [Google Scholar] [CrossRef] [PubMed]
  84. Xu, S.; Li, J.; Chen, L.; Guo, L.; Ye, M.; Wu, Y.; Ji, Q. Plasma miR-32 Levels in Non-Small Cell Lung Cancer Patients Receiving Platinum-Based Chemotherapy Can Predict the Effectiveness and Prognosis of Chemotherapy. Medicine 2019, 98, e17335. [Google Scholar] [CrossRef] [PubMed]
  85. Simeonidis, S.; Koutsilieri, S.; Vozikis, A.; Cooper, D.N.; Mitropoulou, C.; Patrinos, G.P. Application of Economic Evaluation to Assess Feasibility for Reimbursement of Genomic Testing as Part of Personalized Medicine Interventions. Front. Pharmacol. 2019, 10, 830. [Google Scholar] [CrossRef] [PubMed]
  86. Fagery, M.; Khorshidi, H.A.; Wong, S.Q.; Vu, M.; IJzerman, M. Health Economic Evidence and Modeling Challenges for Liquid Biopsy Assays in Cancer Management: A Systematic Literature Review. PharmacoEconomics 2023, 41, 1229–1248. [Google Scholar] [CrossRef] [PubMed]
  87. Graham, D.M.; Leighl, N.B. Economic Impact of Tissue Testing and Treatments of Metastatic NSCLC in the Era of Personalized Medicine. Front. Oncol. 2014, 4, 258. [Google Scholar] [CrossRef] [PubMed]
  88. Hofmarcher, T.; Malmberg, C.; Lindgren, P. A Global Analysis of the Value of Precision Medicine in Oncology—The Case of Non-Small Cell Lung Cancer. Front. Med. 2023, 10, 1119506. [Google Scholar] [CrossRef] [PubMed]
  89. Chen, W.; Anothaisintawee, T.; Butani, D.; Wang, Y.; Zemlyanska, Y.; Wong, C.B.N.; Virabhak, S.; Hrishikesh, M.A.; Teerawattananon, Y. Assessing the Cost-Effectiveness of Precision Medicine: Protocol for a Systematic Review and Meta-Analysis. BMJ Open 2022, 12, e057537. [Google Scholar] [CrossRef] [PubMed]
  90. Rolfo, C.; Mack, P.C.; Scagliotti, G.V.; Baas, P.; Barlesi, F.; Bivona, T.G.; Herbst, R.S.; Mok, T.S.; Peled, N.; Pirker, R.; et al. Liquid Biopsy for Advanced Non-Small Cell Lung Cancer (NSCLC): A Statement Paper from the IASLC. J. Thorac. Oncol. 2018, 13, 1248–1268. [Google Scholar] [CrossRef] [PubMed]
  91. Postel, M.; Roosen, A.; Laurent-Puig, P.; Taly, V.; Wang-Renault, S.-F. Droplet-Based Digital PCR and next Generation Sequencing for Monitoring Circulating Tumor DNA: A Cancer Diagnostic Perspective. Expert Rev. Mol. Diagn. 2018, 18, 7–17. [Google Scholar] [CrossRef]
  92. Valpione, S.; Campana, L. Detection of Circulating Tumor DNA (ctDNA) by Digital Droplet Polymerase Chain Reaction (Dd-PCR) in Liquid Biopsies. Methods Enzymol. 2019, 629, 1–15. [Google Scholar] [CrossRef] [PubMed]
  93. Heitzer, E.; Van Den Broek, D.; Denis, M.G.; Hofman, P.; Hubank, M.; Mouliere, F.; Paz-Ares, L.; Schuuring, E.; Sültmann, H.; Vainer, G.; et al. Recommendations for a Practical Implementation of Circulating Tumor DNA Mutation Testing in Metastatic Non-Small-Cell Lung Cancer. ESMO Open 2022, 7, 100399. [Google Scholar] [CrossRef] [PubMed]
  94. Liu, X.-G.; Zhu, W.-Y.; Huang, Y.-Y.; Ma, L.-N.; Zhou, S.-Q.; Wang, Y.-K.; Zeng, F.; Zhou, J.-H.; Zhang, Y.-K. High Expression of Serum miR-21 and Tumor miR-200c Associated with Poor Prognosis in Patients with Lung Cancer. Med. Oncol. 2012, 29, 618–626. [Google Scholar] [CrossRef] [PubMed]
  95. Wu, C.; Cao, Y.; He, Z.; He, J.; Hu, C.; Duan, H.; Jiang, J. Serum Levels of miR-19b and miR-146a as Prognostic Biomarkers for Non-Small Cell Lung Cancer. Tohoku J. Exp. Med. 2014, 232, 85–95. [Google Scholar] [CrossRef] [PubMed]
  96. Li, C.; Yin, Y.; Liu, X.; Xi, X.; Xue, W.; Qu, Y. Non-Small Cell Lung Cancer Associated microRNA Expression Signature: Integrated Bioinformatics Analysis, Validation and Clinical Significance. Oncotarget 2017, 8, 24564–24578. [Google Scholar] [CrossRef] [PubMed]
  97. Schuurbiers, M.; Huang, Z.; Saelee, S.; Javey, M.; de Visser, L.; van den Broek, D.; van den Heuvel, M.; Lovejoy, A.F.; Monkhorst, K.; Klass, D. Biological and Technical Factors in the Assessment of Blood-Based Tumor Mutational Burden (bTMB) in Patients with NSCLC. J. Immunother. Cancer 2022, 10, e004064. [Google Scholar] [CrossRef] [PubMed]
  98. Raiber-Moreau, E.-A.; Portella, G.; Butler, M.G.; Clement, O.; Konigshofer, Y.; Hadfield, J. Development and Validation of Blood Tumor Mutational Burden Reference Standards. Genes Chromosomes Cancer 2023, 62, 121–130. [Google Scholar] [CrossRef] [PubMed]
  99. Cottrell, T.R.; Taube, J.M. PD-L1 and Emerging Biomarkers in Immune Checkpoint Blockade Therapy. Cancer J. 2018, 24, 41–46. [Google Scholar] [CrossRef] [PubMed]
  100. Domblides, C.; Ruppert, A.-M.; Antoine, M.; Wislez, M. Immunotherapy’s New Challenge: Identification of Predictive Biomarkers for Tumor Response. Transl. Cancer Res. 2017, 6, S306–S308. [Google Scholar] [CrossRef]
  101. Goh, K.Y.; Cheng, T.Y.D.; Tham, S.C.; Lim, D.W.-T. Circulating Biomarkers for Prediction of Immunotherapy Response in NSCLC. Biomedicines 2023, 11, 508. [Google Scholar] [CrossRef]
  102. Cao, W.; Tang, Q.; Zeng, J.; Jin, X.; Zu, L.; Xu, S. A Review of Biomarkers and Their Clinical Impact in Resected Early-Stage Non-Small-Cell Lung Cancer. Cancers 2023, 15, 4561. [Google Scholar] [CrossRef]
  103. Tostes, K.; Siqueira, A.P.; Reis, R.M.; Leal, L.F.; Arantes, L.M.R.B. Biomarkers for Immune Checkpoint Inhibitor Response in NSCLC: Current Developments and Applicability. Int. J. Mol. Sci. 2023, 24, 11887. [Google Scholar] [CrossRef] [PubMed]
  104. Leidinger, P.; Backes, C.; Blatt, M.; Keller, A.; Huwer, H.; Lepper, P.; Bals, R.; Meese, E. The Blood-Borne miRNA Signature of Lung Cancer Patients Is Independent of Histology but Influenced by Metastases. Mol. Cancer 2014, 13, 202. [Google Scholar] [CrossRef] [PubMed]
  105. Zhang, Q.; Luo, J.; Wu, S.; Si, H.; Gao, C.; Xu, W.; Abdullah, S.E.; Higgs, B.W.; Dennis, P.A.; Van Der Heijden, M.S.; et al. Prognostic and Predictive Impact of Circulating Tumor DNA in Patients with Advanced Cancers Treated with Immune Checkpoint Blockade. Cancer Discov. 2020, 10, 1842–1853. [Google Scholar] [CrossRef] [PubMed]
  106. Doroshow, D.B.; Bhalla, S.; Beasley, M.B.; Sholl, L.M.; Kerr, K.M.; Gnjatic, S.; Wistuba, I.I.; Rimm, D.L.; Tsao, M.S.; Hirsch, F.R. PD-L1 as a Biomarker of Response to Immune-Checkpoint Inhibitors. Nat. Rev. Clin. Oncol. 2021, 18, 345–362. [Google Scholar] [CrossRef] [PubMed]
  107. Wang, Y.; Niu, X.; Cheng, Y.; Zhang, Y.; Xia, L.; Xia, W.; Lu, S. Exosomal PD-L1 Predicts Response with Immunotherapy in NSCLC Patients. Clin. Exp. Immunol. 2022, 208, 316–322. [Google Scholar] [CrossRef]
  108. Massafra, M.; Passalacqua, M.I.; Gebbia, V.; Macrì, P.; Lazzari, C.; Gregorc, V.; Buda, C.; Altavilla, G.; Santarpia, M. Immunotherapeutic Advances for NSCLC. BTT 2021, 15, 399–417. [Google Scholar] [CrossRef] [PubMed]
  109. Incorvaia, L.; Fanale, D.; Badalamenti, G.; Barraco, N.; Bono, M.; Corsini, L.R.; Galvano, A.; Gristina, V.; Listì, A.; Vieni, S.; et al. Programmed Death Ligand 1 (PD-L1) as a Predictive Biomarker for Pembrolizumab Therapy in Patients with Advanced Non-Small-Cell Lung Cancer (NSCLC). Adv. Ther. 2019, 36, 2600–2617. [Google Scholar] [CrossRef] [PubMed]
  110. Ullah, A.; Pulliam, S.; Karki, N.R.; Khan, J.; Jogezai, S.; Sultan, S.; Muhammad, L.; Khan, M.; Jamil, N.; Waheed, A.; et al. PD-L1 Over-Expression Varies in Different Subtypes of Lung Cancer: Will This Affect Future Therapies? Clin. Pract. 2022, 12, 653–671. [Google Scholar] [CrossRef] [PubMed]
  111. Romano, G.; Kwong, L.N. Diagnostic and Therapeutic Applications of miRNA-Based Strategies to Cancer Immunotherapy. Cancer Metastasis Rev. 2018, 37, 45–53. [Google Scholar] [CrossRef] [PubMed]
  112. Xing, Y.; Wang, Z.; Lu, Z.; Xia, J.; Xie, Z.; Jiao, M.; Liu, R.; Chu, Y. MicroRNAs: Immune Modulators in Cancer Immunotherapy. Immunother. Adv. 2021, 1, ltab006. [Google Scholar] [CrossRef]
  113. Sirker, M.; Liang, W.; Zhang, L.; Fimmers, R.; Rothschild, M.A.; Gomes, I.; Schneider, P.M. Impact of Using Validated or Standard Reference Genes for miRNA qPCR Data Normalization in Cell Type Identification. Forensic Sci. Int. Genet. Suppl. Ser. 2015, 5, e199–e201. [Google Scholar] [CrossRef]
  114. Nuzziello, N.; Liguori, M. Seeking a Standardized Normalization Method for the Quantification of microRNA Expression. Muscle Nerve 2019, 59, E39. [Google Scholar] [CrossRef] [PubMed]
  115. Mosquera, F.E.C.; Guevara-Montoya, M.C.; Serna-Ramirez, V.; Liscano, Y. Neuroinflammation and Schizophrenia: New Therapeutic Strategies through Psychobiotics, Nanotechnology, and Artificial Intelligence (AI). JPM 2024, 14, 391. [Google Scholar] [CrossRef] [PubMed]
  116. Mosquera, F.E.C.; Lizcano Martinez, S.; Liscano, Y. Effectiveness of Psychobiotics in the Treatment of Psychiatric and Cognitive Disorders: A Systematic Review of Randomized Clinical Trials. Nutrients 2024, 16, 1352. [Google Scholar] [CrossRef] [PubMed]
  117. Jin, Y.; Dong, H.; Xia, L.; Yang, Y.; Zhu, Y.; Shen, Y.; Zheng, H.; Yao, C.; Wang, Y.; Lu, S. The Diversity of Gut Microbiome Is Associated with Favorable Responses to Anti–Programmed Death 1 Immunotherapy in Chinese Patients with NSCLC. J. Thorac. Oncol. 2019, 14, 1378–1389. [Google Scholar] [CrossRef] [PubMed]
  118. Zhang, F.; Ferrero, M.; Dong, N.; D’Auria, G.; Reyes-Prieto, M.; Herreros-Pomares, A.; Calabuig-Fariñas, S.; Duréndez, E.; Aparisi, F.; Blasco, A.; et al. Analysis of the Gut Microbiota: An Emerging Source of Biomarkers for Immune Checkpoint Blockade Therapy in Non-Small Cell Lung Cancer. Cancers 2021, 13, 2514. [Google Scholar] [CrossRef] [PubMed]
  119. Roviello, G.; Iannone, L.F.; Bersanelli, M.; Mini, E.; Catalano, M. The Gut Microbiome and Efficacy of Cancer Immunotherapy. Pharmacol. Ther. 2022, 231, 107973. [Google Scholar] [CrossRef]
  120. Katayama, Y.; Yamada, T.; Shimamoto, T.; Iwasaku, M.; Kaneko, Y.; Uchino, J.; Takayama, K. The Role of the Gut Microbiome on the Efficacy of Immune Checkpoint Inhibitors in Japanese Responder Patients with Advanced Non-Small Cell Lung Cancer. Transl. Lung. Cancer Res. 2019, 8, 847–853. [Google Scholar] [CrossRef] [PubMed]
  121. Shah, H.; Ng, T.L. A Narrative Review from Gut to Lungs: Non-Small Cell Lung Cancer and the Gastrointestinal Microbiome. Transl. Lung. Cancer Res. 2023, 12, 909–926. [Google Scholar] [CrossRef] [PubMed]
  122. Casciaro, M.; Salvo, E.D.; Pioggia, G.; Gangemi, S. Microbiota and microRNAs in Lung Diseases: Mutual Influence and Role Insights. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 13000–13008. [Google Scholar] [PubMed]
  123. Chrysostomou, D.; Roberts, L.A.; Marchesi, J.R.; Kinross, J.M. Gut Microbiota Modulation of Efficacy and Toxicity of Cancer Chemotherapy and Immunotherapy. Gastroenterology 2023, 164, 198–213. [Google Scholar] [CrossRef] [PubMed]
  124. Rescigno, M. Training the Microbiota to Increase Immune Checkpoint Blockade and to Reduce Toxicity. Eur. J. Immunol. 2023, 53, 2250183. [Google Scholar] [CrossRef] [PubMed]
  125. Ryu, G.; Kim, H.; Koh, A. Approaching Precision Medicine by Tailoring the Microbiota. Mamm. Genome 2021, 32, 206–222. [Google Scholar] [CrossRef] [PubMed]
  126. Allegra, A.; Musolino, C.; Tonacci, A.; Pioggia, G.; Gangemi, S. Interactions between the MicroRNAs and Microbiota in Cancer Development: Roles and Therapeutic Opportunities. Cancers 2020, 12, 805. [Google Scholar] [CrossRef] [PubMed]
  127. Purkayastha, K.; Dhar, R.; Pethusamy, K.; Srivastava, T.; Shankar, A.; Rath, G.K.; Karmakar, S. The Issues and Challenges with Cancer Biomarkers. J. Cancer Res. Ther. 2023, 19, S20–S35. [Google Scholar] [CrossRef]
  128. Truesdell, J.; Miller, V.A.; Fabrizio, D. Approach to Evaluating Tumor Mutational Burden in Routine Clinical Practice. Transl. Lung. Cancer Res. 2018, 7, 678–681. [Google Scholar] [CrossRef] [PubMed]
  129. Restrepo, J.C.; Dueñas, D.; Corredor, Z.; Liscano, Y. Advances in Genomic Data and Biomarkers: Revolutionizing NSCLC Diagnosis and Treatment. Cancers 2023, 15, 3474. [Google Scholar] [CrossRef] [PubMed]
  130. Van Den Broek, D.; Hiltermann, T.J.N.; Biesma, B.; Dinjens, W.N.M.; ‘T Hart, N.A.; Hinrichs, J.W.J.; Leers, M.P.G.; Monkhorst, K.; Van Oosterhout, M.; Scharnhorst, V.; et al. Implementation of Novel Molecular Biomarkers for Non-Small Cell Lung Cancer in the Netherlands: How to Deal with Increasing Complexity. Front. Oncol. 2020, 9, 1521. [Google Scholar] [CrossRef] [PubMed]
  131. Hofman, P. The Challenges of Evaluating Predictive Biomarkers Using Small Biopsy Tissue Samples and Liquid Biopsies from Non-Small Cell Lung Cancer Patients. J. Thorac. Dis. 2019, 11, S57–S64. [Google Scholar] [CrossRef] [PubMed]
  132. Ahmed, J.; Das, B.; Shin, S.; Chen, A. Challenges and Future Directions in the Management of Tumor Mutational Burden-High (TMB-H) Advanced Solid Malignancies. Cancers 2023, 15, 5841. [Google Scholar] [CrossRef] [PubMed]
  133. Zhang, N.; Zhang, J.; Wang, G.; He, X.; Mi, Y.; Cao, Y.; Yu, X. Predictive Efficacy of Blood-Based Tumor Mutation Burden Assay for Immune Checkpoint Inhibitors Therapy in Non-Small Cell Lung Cancer: A Systematic Review and Meta-Analysis. Front. Oncol. 2022, 12, 795933. [Google Scholar] [CrossRef] [PubMed]
  134. Arghiani, N.; Shah, K. Modulating microRNAs in Cancer: Next-Generation Therapies. Cancer Biol. Med. 2021, 19, 289–304. [Google Scholar] [CrossRef] [PubMed]
  135. Tadesse, K.; Benhamou, R.I. Targeting MicroRNAs with Small Molecules. ncRNA 2024, 10, 17. [Google Scholar] [CrossRef] [PubMed]
  136. Pimienta, D.A.; Cruz Mosquera, F.E.; Palacios Velasco, I.; Giraldo Rodas, M.; Oñate-Garzón, J.; Liscano, Y. Specific Focus on Antifungal Peptides against Azole Resistant Aspergillus Fumigatus: Current Status, Challenges, and Future Perspectives. J. Fungi 2022, 9, 42. [Google Scholar] [CrossRef] [PubMed]
  137. Qiao, Y.; Wan, J.; Zhou, L.; Ma, W.; Yang, Y.; Luo, W.; Yu, Z.; Wang, H. Stimuli-responsive Nanotherapeutics for Precision Drug Delivery and Cancer Therapy. WIREs Nanomed. Nanobiotechnol. 2019, 11, e1527. [Google Scholar] [CrossRef] [PubMed]
  138. Holder, J.E.; Ferguson, C.; Oliveira, E.; Lodeiro, C.; Trim, C.M.; Byrne, L.J.; Bertolo, E.; Wilson, C.M. The Use of Nanoparticles for Targeted Drug Delivery in Non-Small Cell Lung Cancer. Front. Oncol. 2023, 13, 1154318. [Google Scholar] [CrossRef] [PubMed]
  139. Macarrón Palacios, A.; Korus, P.; Wilkens, B.G.C.; Heshmatpour, N.; Patnaik, S.R. Revolutionizing in Vivo Therapy with CRISPR/Cas Genome Editing: Breakthroughs, Opportunities and Challenges. Front. Genome Ed. 2024, 6, 1342193. [Google Scholar] [CrossRef] [PubMed]
  140. Ou, X.; Ma, Q.; Yin, W.; Ma, X.; He, Z. CRISPR/Cas9 Gene-Editing in Cancer Immunotherapy: Promoting the Present Revolution in Cancer Therapy and Exploring More. Front. Cell Dev. Biol. 2021, 9, 674467. [Google Scholar] [CrossRef] [PubMed]
  141. Yahya, S.; Abdelmenym Mohamed, S.I.; Yahya, S. Gene Editing: A Powerful Tool for Cancer Immunotherapy. Biointerface Res. Appl. Chem. 2022, 13, 98. [Google Scholar] [CrossRef]
  142. Mandair, D.; Reis-Filho, J.S.; Ashworth, A. Biological Insights and Novel Biomarker Discovery through Deep Learning Approaches in Breast Cancer Histopathology. NPJ. Breast. Cancer 2023, 9, 21. [Google Scholar] [CrossRef]
Figure 1. PRISMA flow diagram with the search and study selection strategy. Cohen Kappa = 0.92 y Cohen Kappa = 0.82 shows a high level of agreement between the reviewers, which gives reliability and validity to the results.
Figure 1. PRISMA flow diagram with the search and study selection strategy. Cohen Kappa = 0.92 y Cohen Kappa = 0.82 shows a high level of agreement between the reviewers, which gives reliability and validity to the results.
Cancers 16 02338 g001
Figure 2. Cochrane Risk of Bias assessment for randomized studies of interventions in the systematic review. (A). Risk of bias summary: review authors’ judgements about each risk of bias item for each included study. The symbol “+” indicates a low risk of bias, the symbol “?” indicates an unclear risk of bias, and the symbol “−” indicates a high risk of bias. The colors used are green for low risk of bias, yellow for unclear risk of bias, and red for high risk of bias [45,47,48,49,51,52,54,62,64,66,69,70,73,74,75,76,77,78,79]. (B). Risk of bias graph: review authors’ judgements about each risk of bias item presented as percentages across all included studies. Figure created by RevMan 5 (accessed on 20 February 2024).
Figure 2. Cochrane Risk of Bias assessment for randomized studies of interventions in the systematic review. (A). Risk of bias summary: review authors’ judgements about each risk of bias item for each included study. The symbol “+” indicates a low risk of bias, the symbol “?” indicates an unclear risk of bias, and the symbol “−” indicates a high risk of bias. The colors used are green for low risk of bias, yellow for unclear risk of bias, and red for high risk of bias [45,47,48,49,51,52,54,62,64,66,69,70,73,74,75,76,77,78,79]. (B). Risk of bias graph: review authors’ judgements about each risk of bias item presented as percentages across all included studies. Figure created by RevMan 5 (accessed on 20 February 2024).
Cancers 16 02338 g002
Figure 3. ROBINS-I in the systematic review. (A). Summary of risk of bias: judgements of review authors regarding each risk of bias item for each included study [15,44,53,59,61,63,65,68]. The symbol “+” indicates a low risk of bias, the symbol “?” indicates an unclear risk of bias, and the symbol “−” indicates a high risk of bias. The colors used are green for low risk of bias, yellow for unclear risk of bias, and red for high risk of bias. (B). Graph of risk of bias: judgements of review authors on each risk of bias item, presented as percentages across all included studies. Figure created by RevMan 5 (accessed on 20 February 2024).
Figure 3. ROBINS-I in the systematic review. (A). Summary of risk of bias: judgements of review authors regarding each risk of bias item for each included study [15,44,53,59,61,63,65,68]. The symbol “+” indicates a low risk of bias, the symbol “?” indicates an unclear risk of bias, and the symbol “−” indicates a high risk of bias. The colors used are green for low risk of bias, yellow for unclear risk of bias, and red for high risk of bias. (B). Graph of risk of bias: judgements of review authors on each risk of bias item, presented as percentages across all included studies. Figure created by RevMan 5 (accessed on 20 February 2024).
Cancers 16 02338 g003
Table 2. Synthesis of studies included in systematic review by biomarker evaluated.
Table 2. Synthesis of studies included in systematic review by biomarker evaluated.
Author and Publication YearBiomarker EvaluatedMethod
of Biomarker Analysis
Cut-Off-ValuePredictive and Prognostic ValueOverall Survival (OS) and
Progression-Free Survival (PFS)
Hazard Ratios (HRs) and 95% Confidence
Intervals (CIs) for Tumor Size
TreatmentStatistical Methods Used for
Biomarker Evaluation
Conclusions
Ren et al., 2022 [48]ctDNA
dynamics
Dynamic
monitoring
ctDNA clearance after two cyclesPredicts camrelizumab plus chemotherapy efficacyProlonged OS and PFSOS HR = 0.55Camrelizumab + carboplatin and paclitaxelKaplan–Meier, Cox modelsctDNA dynamics useful in predicting treatment efficacy in advanced squamous NSCLC
Yang et al., 2023
[49]
ALK fusions in ctDNANGS of plasma ctDNANSALK fusion detectability related to prognosisPFS similar for brigatinib (19.3 mo) and alectinib (19.2 mo)PFS HR = 0.97 (95% CI: 0.66–1.42)Brigatinib vs. alectinibKaplan–Meier, Cox regressionNo superiority of brigatinib over alectinib in ALK-positive NSCLC previously treated with crizotinib
Riess et al., 2021
[50]
EGFR exon 20 insertionsNext-generation sequencing (NGS)NSNot predictivePFS: 5.4 monthsNSErlotinib and onalespibStandard 3 + 3 dose escalation, RECIST 1.1Limited activity of erlotinib and onalespib in EGFR exon 20 insertion NSCLC
Lo Russo et al., 2022 [51]36 immunobiomarkers like CD14, CD16Multiparametric flow cytometryNSImproved PFS and OSCD14 = 0.018 OS significant improvementsNSPembrolizumabOrthogonal component clustering, multivariable Cox regressionPredictive outcomes in PD-L1-low NSCLC with pembrolizumab
Garon et al., 2023
[52]
EGFR, TE genomic alterations in ctDNANGS of ctDNANSBaseline EGFR alterations indicate shorter PFSMedian PFS: 12.7 mo (with aEGFR), 22 mo (without)HR for PFS = 1.87 (95% CI: 1.42–2.51)Ramucirumab + erlotinibKaplan–MeierBaseline EGFR mutations linked to shorter PFS, predictive of ramucirumab + erlotinib efficacy
Han et al., 2023
[53]
bTMBNGS with 448 gene panel≥1.72 mutations/MbPredictive of sintilimab + docetaxel efficacyMedian PFS: 5.8 mo; Median OS: 12.6 moNSDocetaxel + sintilimabKaplan–Meier, Clopper–PearsonSintilimab + docetaxel improves PFS, OS in advanced NSCLC
Si et al., 2021
[54]
bTMBGuardantOMNI ctDNA assay≥20 mutations/MbPredictive of benefit with durvalumab + tremelimumab vs. chemotherapyNSNSDurvalumab ± tremelimumab or chemotherapyCox models, minimal p-value cross-validationHigh bTMB predicts clinical benefit with durvalumab + tremelimumab vs. chemotherapy
Jiang et al., 2021
[55]
PD-L1, TMB, CD8+ TIL density, DSPP mutationIHC and whole-exome sequencingPD-L1 ≥ 1% expressionDSPP mutation associated with longer PFSMedian PFS: 7.0 mo; Median OS: 23.5 moNSToripalimab + carboplatin and pemetrexedKaplan–MeierDSPP mutation suggested as potential biomarker for toripalimab efficacy in second-line treatment
Zhang et al., 2024
[56]
ctDNA dynamics, BRCA2, BRINP3, FBXW7, KIT, RB1Comprehensive NGS ctDNA profilingClearance after 2 cyclesBRCA2, others shorten PFS; clearance associated with longer PFSMedian PFS: 11.4 months, OS: 27.2 monthsNSSintilimab, nab-paclitaxel, carboplatinKaplan–Meier, hazard ratiosEffective first-line treatment with promising PFS; genetic markers as predictive values
Tan et al., 2024
[44]
EGFR T790M, EGFRmDroplet digital PCR, targeted sequencingNSDecrease in/clearance of EGFRm and T790M predicts longer PFS/OSMedian PFS: 9.4 months, OS: 26 monthsNSAlternating osimertinib and gefitinibKaplan–Meier, Cox regressionFeasible therapy with profound impact on clonal dynamics, though primary endpoint unmet
Kim et al., 2022
[57]
bTMBFoundation Medicine bTMB assay≥16 mutations/MbHigher ORR, possibly longer OS for bTMB ≥ 16Longer OS for bTMB ≥ 16, PFS and OS not significant at cut-offNSFirst-line atezolizumab monotherapyKaplan–Meier, log-rank, Cox modelbTMB ≥ 16 linked to higher ORR; further studies needed for bTMB as a standalone predictive marker
Peters et al., 2022
[58]
bTMBFoundation Medicine bTMB CTA≥16 for analysisbTMB ≥ 16 did not meet primary endpoint for PFS benefitNo significant difference in PFS or OSHR: 0.77 (95% CI: 0.59–1.00,
p = 0.053)
Atezolizumab vs. chemotherapyKaplan–Meier, Cox modelbTMB ≥ 16 not supported as standalone predictive marker; potential for optimization
Chaft et al., 2022
[59]
MPR, PD-L1 TPSBlood immunophenotype, exome sequencingMPR ≤ 10% viable cells, PD-L1 high TPSHigh PD-L1 TPS predicts MPR3-year DFS: 72%, OS: 80%DFS HR = 0.373, OS HR = 0.273Neoadjuvant atezolizumab monotherapyKaplan–MeierNeoadjuvant atezolizumab met primary endpoint of MPR; further validation needed
Shi et al., 2022.
[60]
PD-L1, OVOL2, CTCFDeep sequencingNSOVOL2 high = better PFS, CTCF high = worse PFSMedian OS significantly longer in sintilimab armNSSintilimab vs. docetaxelCox regressionSintilimab improved survival, response rates; potential new treatment option for advanced NSCLC
Papadimitrakopoulou et al., 2020 [45]EGFR T790M, EGFRmCobas, ddPCR, NGST790M-negative predicts longer PFST790M status predictiveMedian PFS: 12.5 vs. 8.3 months (osimertinib)NSOsimertinib, platinum-pemetrexedKaplan–MeierT790M-negative plasma predicts longer PFS
Sakai et al., 2021a
[46]
TMBTargeted deep sequencing≥12–16 mut/MbHigh TMB predicts better RFSTMB ≥ 12 mut/Mb: improved RFSHR = 0.477Pemetrexed/cisplatin vs. vinorelbine/cisplatinKaplan–Meier, Cox modelHigh TMB beneficial for pemetrexed/cisplatin in NS-NSCLC
Park et al., 2023
[61]
bTMB, cfDNA, hVAF, VAFSDCT-ULTRA, targeted NGSbTMB ≥ 11.5 mut/Mb at baselinecfDNA, bTMB dynamics predictiveMedian OS: 13.1 months, PFS: 2.1 monthsNS1200 mg of atezolizumab every three weeksKaplan–Meier, Cox modelBaseline biomarkers predict atezolizumab efficacy
Park et al., 2021
[47]
Activating EGFR mutations in ctDNA and DNAMutyper and Cobas v2 assaysNSctDNA sensitivity for EGFR mutationsMedian PFS: 11.1 monthsNSOsimertinib 80 mg dailyKaplan–MeierctDNA assays effective in detecting EGFR mutations
Gu et al., 2023
[62]
EGFR mutations, ctDNA MRDNGSNSMRD predicts therapeutic efficacyPFS longer with TKI + pemetrexed in altered genesNSEGFR-TKI monotherapy or combined with pemetrexedKaplan–Meier, log-rank testsctDNA MRD as a biomarker for therapy efficacy in NSCLC
Han et al., 2022
[63]
ctDNA dynamicsNGS with 448-gene panel≥2 mutations positive, ≤1 mutation negativectDNA clearance at 6 weeks predictiveNSNSSintilimab + docetaxel, maintenance with sintilimabKaplan–Meier, Cox regressionctDNA dynamics predict sintilimab efficacy
Zhong et al., 2023
[64]
EGFR mutations, ctDNA mutation detectionNGS targeting 425 genesPositive ctDNA at baseline predicts shorter PFSctDNA as a predictive biomarkerNSNSTislelizumab + carboplatin and nab-paclitaxelKaplan–Meier, Cox regressionctDNA predicts progression in tislelizumab therapy
García-Pardo et al., 2023 [65]ctDNA genotypingPlasma ctDNA testing with NGS before diagnosisNSEarly ctDNA testing accelerates treatment initiationNSNSAdvanced nonsquamous NSCLC treatmentsStandard genotyping comparisonEarly ctDNA genotyping shortens treatment initiation
Nomura et al., 2020 [66]ctDNA (Guardant360®)Guardant360®NSNon-inferiority of discontinuing PD-1 inhibitorsNSNSContinue or discontinue PD-1 inhibitorsKaplan–Meier, Cox model, non-inferiority testStudy supports possible discontinuation of PD-1 inhibitors without affecting survival
Martini et al., 2022
[67]
Gut microbiota, ctDNA RAS/BRAF WT MSS16S rRNA sequencing, targeted NGSNSCertain gut bacteria species linked to longer PFSNSNSCetuximab + avelumab combination therapyKendall Tau-b, Kaplan–Meier, log-rankGut microbiota as potential biomarker for cetuximab + avelumab efficacy
Provencio et al., 2022 [68]ctDNA for prognosis and predictive valueNGS of FFPE and plasma samplesMAF ≥ 1% at baselineLow ctDNA levels associated with better survival outcomes3-year OS: 81.9% (ITT), 91.0% (per protocol)HR for PFS: 0.20, OS: 0.07Neoadjuvant paclitaxel, carboplatin, and nivolumabKaplan–Meier, Cox regression, competing riskctDNA levels predict success in chemoimmunotherapy
West et al., 2022
[69]
KRAS, STK11, KEAP1, TP53 mutationsBlood-based ctDNA sequencingNSKRAS, STK11, KEAP1 mutations affect treatment efficacymKRAS: improved OS and PFS with ABCP vs. BCPOS: HR = 0.50 (0.34–0.72); PFS: HR = 0.42 (0.29–0.61)Atezolizumab, bevacizumab, carboplatin, paclitaxelKaplan–Meier, Cox modelImproved outcomes in mKRAS with ABCP; ctDNA levels after treatment correlate with PFS and OS
Lo Russo et al., 2023 [70]Immune cell subsets, gene expression levelsFlow cytometry, gene expression, metagenomic sequencingNSImmune profiles correlate with PFSMedian PFS: 2.9 monthsNSPembrolizumab as first-line treatmentSequential Cox regression, Benjamini–Hochberg, LASSOMultiomic markers predict PFS; NK cells at baseline may determine pembrolizumab benefit
Zhou et al., 2023
[71]
PD-L1 on tumor cellsSP263 and 22C3 assaysNSHigh concordance between SP263 and 22C3 assaysNSNSAdjuvant atezolizumab vs. supportive careConcordance assessment, survival evaluationSP263 and 22C3 assays effectively predict adjuvant atezolizumab benefit in early-stage NSCLC
Sakai et al., 2021b
[72]
EGFR T790M mutationCobas v2, ddPCR, deep sequencingNSMonitors EGFR T790M for treatment responseNSNSOsimertinib administrationUnivariate regression, Cox modelEffective monitoring of EGFR T790M with ctDNA enhances osimertinib treatment decisions
Redman et al., 2020 [73]Multiple biomarkersFoundationOne® NGS assayNSMultiple biomarkers assessed for targeted therapiesMedian OS varied by treatment arm, no PFS providedNSTargeted therapies, immunotherapies in sqNSCLCKaplan–Meier, Cox regression, other methodsThe study validates the use of molecularly targeted therapies in genomically defined sqNSCLC subgroups
Hirsch et al., 2022
[74]
EGFR copy number, protein expressionEGFR FISH, IHCTC ≥ 50% or IC ≥ 10% for SP142 and TPS ≥ 50% for 22C3High EGFR copy number and protein expression predictiveImproved OS in SCC with cetuximab addition (12.6 vs. 4.6 months)HR: 0.32 (0.18–0.59), p = 0.0002Chemotherapy with or without cetuximabCox model for OS and PFSEGFR FISH and IHC are predictive of cetuximab benefit in SCC, independently of KRAS status
Schuler et al., 2020
[75]
MET dysregulationImmunohistochemistry, FISH, NGSNSCapmatinib’s antitumor activity based on MET dysregulationNSNSCapmatinib administrationSafety and activity assessmentCapmatinib shows promise in NSCLC with MET dependency, particularly with high MET GCN or METex14 mutations
Gadgeel et al., 2022 [76]PD-L1 expressionSP142 and 22C3 IHC assaysTC ≥ 50% or IC ≥ 10% for SP142 and TPS ≥ 50% for 22C3Atezolizumab improves survival over docetaxelAtezolizumab benefits across all PD-L1 subgroups, especially high PD-L1OS and PFS: HR varied by assay, greater in high PD-L1 groupsAtezolizumab or docetaxelAnalysis in PD-L1 subgroups, assay selectionSP142 and 22C3 assays effectively predict atezolizumab efficacy in metastatic NSCLC across PD-L1 thresholds
Ramalingam et al., 2021 [77]LP52 gene expressionWhole-transcriptome sequencingNSVeliparib’s efficacy in sqNSCLCNo significant OS benefit in smokers; slight benefit in general populationOS: HR = 0.853 (0.747 to 0.974), PFS not differentVeliparib or placebo with carboplatin and paclitaxelKaplan–Meier, log-rank test, biomarker analysisVeliparib shows a marginal OS benefit; LP52 may help to identify responsive patients
Song et al., 2022
[78]
HER2 mutationsNGSNSPyrotinib’s efficacy based on HER2 mutation types6-month PFS rate: 49.5%, median PFS: 5.6 months, OS: 10.5 monthsNSPyrotinib treatmentKaplan–Meier, Cox regression, Fisher’s exact testPyrotinib effective in HER2-mutant NSCLC; potential for ctDNA to aid in disease monitoring
Anagnostou et al., 2023 [79]ctDNA dynamicsSerial quantitative ctDNA assessmentsMaximal mutant allele fraction clearance at cycle 3ctDNA clearance after two cycles predictiveMedian PFS significantly longer with molecular response, OS not reached vs. 7.23 monthsNSPembrolizumab treatmentKaplan–Meier, exploratory biomarker analysesctDNA dynamics correlate with pembrolizumab efficacy, guiding treatment adjustments
Park et al., 2021
[80]
Activating EGFRmEGFRm ctDNA analysis using PANA MutypeNSEfficacy of afatinib in EGFRm-positive lung cancerMedian PFS: 12.0 months, OS data immatureNSAfatinib 40 mg dailyMann–Whitney U, Pearson’s χ2, Fisher’s exact, Kaplan–MeierAfatinib shows favorable ORR and PFS in treatment-naïve patients with detectable EGFRm in ctDNA
ctDNA: Circulating Tumor DNA; ALK: Anaplastic Lymphoma Kinase; EGFR: Epidermal Growth Factor Receptor; bTMB: Blood Tumor Mutational Burden; TMB: Tumor Mutational Burden; PD-L1: Programmed Death Ligand 1; MPR: Major Pathological Response; TPS: Tumor Proportion Score; MRD: Minimal Residual Disease; cfDNA: Circulating Free DNA; hVAF: High Variant Allele Frequency; VAFSD: Variant Allele Frequency Standard Deviation; NGS: Next-Generation Sequencing; IHC: Immunohistochemistry; HR: Hazard Ratio; CI: Confidence Interval; OS: Overall Survival; PFS: Progression-Free Survival; RECIST: Response Evaluation Criteria In Solid Tumors; ORR: Objective Response Rate; NSCLC: Non-Small-Cell Lung Cancer; Mb: Megabase; BRCA2: Breast Cancer 2, Early Onset; DSPP: Dentin Sialophosphoprotein; CTCF: CCCTC-Binding Factor; OVOL2: Ovo-Like Transcriptional Repressor 2; FFPE: Formalin-Fixed Paraffin-Embedded; KRAS: Kirsten Rat Sarcoma Viral Oncogene Homolog; ABCP: Atezolizumab, Bevacizumab, Carboplatin, Paclitaxel; BCP: Bevacizumab, Carboplatin, Paclitaxel; KEAP1: Kelch-Like ECH-Associated Protein 1; STK11: Serine/Threonine Kinase 11; TP53: Tumor Protein p53; LASSO: Least Absolute Shrinkage and Selection Operator; ddPCR: Droplet Digital Polymerase Chain Reaction; miR: MicroRNA; LP52: 52-Gene Expression Histology Classifier; Mo: Month. NS: Not Specified.
Table 3. Summary of biomarker findings with heterogeneity in NSCLC.
Table 3. Summary of biomarker findings with heterogeneity in NSCLC.
BiomarkerRole of BiomarkerMain FindingsClinical ImplicationsAuthorsTreatment ResponseHeterogeneity in NSCLCClinical and Demographic Characteristics
ctDNATumor burden and disease progression- Detection correlated with tumor burden and disease progression.
- Reduction in levels during treatment indicative of better PFS and OS.
Non-invasive biomarker for monitoring of response and adjustment of therapeutic strategies.Ren et al., 2022 [48]; Yang et al., 2023 [49]; Garon et al., 2023 [52]; Zhang et al., 2024 [56]; Tan et al., 2024 [44]Predictive of response to camrelizumab, carboplatin, paclitaxel, osimertinib, sintilimab, nab-paclitaxel, gefitinibVaries with mutation type (EGFR, ALK), tumor stage, distribution of histological subtypes, and frequency of co-alterations.Predominantly men, history of heavy smoking, ECOG 1, stage IV disease.
PD-L1Tumor progression and prognosisHigh levels associated with better response to ICIs and longer survival.Crucial in selecting patients for immunotherapy.Jiang et al., 2021 [55]; Zhou et al., 2023 [71]; Gadgeel et al., 2022 [76]Predictive of response to atezolizumab, pembrolizumab, toripalimabVaries with expression levels, tumor microenvironment, distribution of histological subtypes, and frequency of co-alterations.Predominantly men, history of heavy smoking, ECOG 1, stage IV disease.
miRNA *Response to immunotherapy and survival outcomesmiRNA-21 and miRNA-155 correlated with response to immunotherapy and survival outcomes.
Plasma miR-32 levels correlated with chemotherapy response and prognosis.
Patient stratification and treatment personalization based on molecular profiles.Xu et al., 2019 [84]Predictive of chemotherapy efficacy and prognosis with platinum-based chemotherapyVaries with miRNA type, interaction with other molecular pathways, and frequency of co-alterations.Patients aged 45–78, predominantly men, 81.4% smokers, ECOG 1–2, stage II–IV disease.
bTMBTumor progression and prognosisHigh bTMB associated with better outcomes in combined immunotherapy and chemotherapy.Accurate measurement predicts immunotherapy efficacy and guides treatment selection.Han et al., 2023 [53];
Si et al., 2021 [54];
Kim et al., 2022 [57];
Peters et al., 2022 [58]
Predictive of response to sintilimab + docetaxel, durvalumab + tremelimumab, atezolizumabVaries with mutation burden, specific gene mutations, distribution of histological subtypes, and frequency of co-alterations.Predominantly men, history of heavy smoking, ECOG 1, stage IV disease.
* Currently, no specific miRNA studies are included in the provided data. Xu et al., 2019 [84], a cohort study, was added for the comparison of biomarkers in NSCLC.
Table 4. Comparative analysis of biomarkers in NSCLC.
Table 4. Comparative analysis of biomarkers in NSCLC.
VariablectDNA
[91,92,93]
miRNA
[94,95,96]
bTMB
[54,81,97,98]
Immunological Markers [20,99,100]
Type of BiomarkerGenetic (circulating DNA)Genetic (non-coding RNA)Genetic (mutational burden)Protein (immune proteins)
Detection MethodNGS, digital PCRReal-time PCR, microarraysNGS, digital PCRIHC, flow cytometry
Clinical UtilityDiagnosis, prognosis, monitoringPrognosis, monitoringPrognostic, predictiveDiagnostic, predictive
Prognostic and Predictive AspectsHigh sensitivity for early detectionCorrelates with immunotherapy responsePredicts response to specific immunotherapiesExpression correlated with survival and response
Variability FactorsInfluenced by tumor burden, detection techniquesInfluenced by sample conditionsRequires standardization in measurementSensitive to detection methods and immune status
AdvantagesNon-invasive, high sensitivityNon-invasive, easily quantifiableInformation on tumor heterogeneityDirectly related to mechanisms of action of therapies
LimitationsCost, need for sequencingInter- and intra-individual variabilityInfluenced by technical and biological factorsRequires validation for specific interpretation
Cost-EffectivenessModerate–highLow–moderateHigh due to sequencing technologiesModerate, depends on the marker and method
Usage RecommendationsWidely recommended in clinical guidelinesIn research, some clinical applicationsRecommended in specific contextsEmerging use, supported by recent studies
Recent InnovationsAdvances in digital PCR technologyNew miRNAs associated with NSCLCImprovements in accuracy and cost of NGSNew predictive markers of response to PD-1/PD-L1
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Restrepo, J.C.; Martínez Guevara, D.; Pareja López, A.; Montenegro Palacios, J.F.; Liscano, Y. Identification and Application of Emerging Biomarkers in Treatment of Non-Small-Cell Lung Cancer: Systematic Review. Cancers 2024, 16, 2338. https://doi.org/10.3390/cancers16132338

AMA Style

Restrepo JC, Martínez Guevara D, Pareja López A, Montenegro Palacios JF, Liscano Y. Identification and Application of Emerging Biomarkers in Treatment of Non-Small-Cell Lung Cancer: Systematic Review. Cancers. 2024; 16(13):2338. https://doi.org/10.3390/cancers16132338

Chicago/Turabian Style

Restrepo, Juan Carlos, Darly Martínez Guevara, Andrés Pareja López, John Fernando Montenegro Palacios, and Yamil Liscano. 2024. "Identification and Application of Emerging Biomarkers in Treatment of Non-Small-Cell Lung Cancer: Systematic Review" Cancers 16, no. 13: 2338. https://doi.org/10.3390/cancers16132338

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop