Non-invasive diagnostic platforms in management of non-small cell lung cancer: opportunities and challenges
Review Article

Non-invasive diagnostic platforms in management of non-small cell lung cancer: opportunities and challenges

Vamsidhar Velcheti, Nathan A. Pennell

Department of Hematology and Medical Oncology, Cleveland Clinic Taussig Cancer Institute, Cleveland, OH, USA

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

Correspondence to: Vamsidhar Velcheti, MD. Department of Hematology and Medical Oncology, Cleveland Clinic Taussig Cancer Institute, 9500 Euclid Ave, CA6, Cleveland, OH 44195, USA. Email: velchev@ccf.org.

Abstract: Several non-invasive diagnostic platforms are already being incorporated in routine clinical practice in the work up and monitoring of patients with lung cancer. These approaches have great potential to improve patient selection and monitor patients while on therapy, however several challenges exist in clinical validation and standardization of such platforms. In this review, we summarize the current technologies available for non-invasive diagnostic evaluation from the blood of patients with non-small cell lung cancer (NSCLC), and discuss the technical and logistical challenges associated incorporating such testing in clinical practice.

Keywords: Non-invasive biopsies; biomarkers; non-small cell lung cancer (NSCLC)


Submitted Jul 20, 2017. Accepted for publication Aug 14, 2017.

doi: 10.21037/atm.2017.08.24


Introduction

Genomic analysis of non-small cell lung cancer (NSCLC) revealed high genomic mutational burden and recurrent alterations in several oncogenes (1,2). These driver oncogenic alterations are key molecular events and often have therapeutic implications. Testing for oncogenic alterations in EGFR, ALK, ROS1 and BRAF are considered standard of care for all patients with non-squamous NSCLC and in addition, more comprehensive genomic profiling including MET, ERBB2, RET, and TRK is often considered (3). There have been remarkable advances in our understanding of the genomic heterogeneity among patients with NSCLC leading to opportunities for targeted therapy for patients with clinically actionable genomic alterations. Interrogation of the genomic alterations in the tumor is thus critical in clinical decision-making for patients with NSCLC. Tissue biopsy is the gold standard for tumor genotyping. However, the challenges of tissue based biomarker analysis include complexity of molecular alterations and issues with adequate tumor tissue acquisition. In addition, spatial and temporal intra-patient heterogeneity of the genomic alterations can make tissue testing and interpretation of the results challenging. Efforts to understand the complex biology of cancers is largely limited by the inability to adequately capture the intra-patient heterogeneity of tumors. Moreover, exposure to treatments both cytotoxic chemotherapy and targeted therapy can create an adaptive biological process in the tumor and such clonal evolution can have therapeutic implications (4). Such key molecular insights are hard to obtain through tissue biopsies. Non-invasive diagnostic platforms popularly known as “liquid biopsies” can potentially provide useful insights into the genetic and epigenetic make-up of the tumor and can be used as complementary diagnostics in addition to the tissue biomarker analysis. Liquid biopsies can also address some of the issues around tumor heterogeneity and clonal evolution of the tumors particularly in the setting of acquired resistance to targeted therapies.

Liquid biopsies generally refer to genomic analysis of circulating tumor cells (CTCs) or circulating tumor nucleic acids (ctDNA and ctRNA). However, several novel techniques using microRNA, platelet-microvesicles, serum metabolites and exosomes are currently being validated. In this review, we summarize the liquid biopsy platforms currently available for clinical use in patients with NSCLC. We discuss the general approach, the clinical applications and methodological challenges of liquid biopsy testing in lung cancer.

Liquid biopsies use peripheral blood samples as a source of the bioanalytics like ctDNA, ctRNA, CTCs, exosomes, platelets or serum proteome. Liquid biopsies allow for rapid biomarker work up, can be used to follow emergence of acquired resistance to treatments and could potentially identify early response or lack of response.


Circulating tumor nucleic acids

Circulating tumor nucleic acid technologies were first developed to detect circulating fetal DNA in the circulation of pregnant women to develop non-invasive prenatal tests (5). Patients with cancer have higher levels of circulating free DNA (cfDNA) compared to normal healthy individuals (6). Recent technological innovations in massively parallel sequencing led to developments of molecular assays with ability to detect minute allelic imbalances in circulating nucleic acid. These platforms can now provide opportunity for interrogating the cfDNA with robust sensitivity and specificity.

Detection and analysis

Sample preparation for ctDNA analysis is very critical and is typically done using collection kits like cell-free DNA BCT® and PAXgene Blood DNA tubes that contain formaldehyde-free preservative reagents that prevent the nuclease-mediated degradation of ctDNA. These tubes also help stabilize nucleated blood cells, thus reducing the release of cellular germline cfDNA (7). A majority of the cfDNA in patients with cancer is not tumor DNA (ctDNA). There are varying amounts [0.01–10%, minor allele frequency (MAF)] of cfDNA in patients with cancer and only a fraction of the DNA represents tumor DNA (ctDNA). The proportion of the ctDNA is a factor of the tumor burden and the tumor biology (8). The clinical utility and reproducibility of the ctDNA assay is thus contingent upon adequately differentiating ctDNA and appropriate normalization. Recent studies suggest that ctDNA have shorter cell-free DNA fragment lengths compared to normal cell-free DNA (9-11). Several kits capturing the smaller fragment lengths from cell-free DNA are available and this may optimize the detection and amplification of ctDNA.

There are multiple sequencing platforms that are available for analysis of ctDNA and these platforms all have some various advantages and disadvantages (12). Platforms such has real-time quantitative PCR (qPCR), the Scorpion Amplification-Refractory Mutation System (ARMS) and droplet digital PCR (dPCR) can provide high sensitivity for detection of known genomic alterations and relatively easy to integrate into the clinical workflow with easy bioinformatic burden. Beads, Emulsion, Amplification and Magnetics (BEAMing) is another recent approach that can provide an extremely sensitive detection threshold of low MAF known genomic alterations in a high background of wild type cfDNA (13). Both BEAMing and dPCR use emulsion PCR based methodology with individual DNA fragments are in droplets allowing for DNA fragments to be amplified independently. Using fluorescent labeled probes mutant and wild-type alleles can be distinguished allowing more accurate quantification of the mutant allele fractions compared to RT-PCR (14,15).

The methods discussed so far allow for detection and quantification of known genomic alterations. However, sequencing using next generation sequencing (NGS) platforms allows for detecting novel alterations at a frequency as low as one mutant copy in several thousand wild-type copies. NGS based approach can have high sensitivity and can screen not only for known mutations, but entire breadth of the targeted genome for previously unknown mutations and alterations. In addition to mutations gene rearrangements and copy number alterations can also be detected. Using hybrid capture methodology portions of the genomic areas of interest are identified among the amplified cfDNA libraries by hybridization capture with oligonucleotides or “baits” complementary to these regions for enrichment and repeat PCR. In addition, tagged-amplicon deep NGS using multiplexed PCR can be used for a lager panel of target regions of interest. Using these approaches adequate sensitivity required for clinical multiplexing can be ensured with ctDNA profiling (16-18). However, NGS based approach is expensive, and is time intensive requiring more sophisticated bioinformatic support in interpretation of the results (Table 1).

Table 1
Table 1 Comparison of circulating tumor cells and circulating-tumor DNA platforms
Full table

Clinical utility of ctDNA assays

There are several potential opportunities for incorporating ctDNA assays into clinical practice. The most important indications for ctDNA assays would be in patients who have inadequate tissue for molecular diagnostic work up for predicting response to targeted treatment, and monitoring for the development of acquired resistance to targeted therapy. In addition, recent studies suggest possible role in early response monitoring (19) and monitoring minimal residual disease after definitive oncologic treatment (8,20-23).

Comprehensive genomic profiling using ctDNA assays can cover all forms of genomic alterations namely indels, point mutations, gene amplifications and rearrangements. This is invaluable for patients where biopsy at diagnosis or progression is not feasible or tissue is insufficient. In addition, ctDNA may capture a more comprehensive molecular summary involving all the metastatic burden of the patients’ cancer compared to a small biopsy which may not reflect the tumor heterogeneity (24).

Identification of clinically actionable genomic alterations

Most patients with advanced stage NSCLC have diagnostic tissue from bronchoscopic or CT-guided biopsies and often patients have limited material for molecular work up (25). Recent advances in NGS technology and optimizing these newer platforms for ctDNA assays, allow for multiplex screening of large panels of genes with high sensitivity often over 75% and high concordance with tissue based testing particularly when the tissue biopsy and the ctDNA testing are temporally concurrent (<1 month) to the ctDNA testing (18,26). In another meta-analysis of 20 studies ctDNA based platforms for detecting EGFR mutations had a pooled sensitivity of 62% and specificity of 95% when compared to tissue based testing (27). ctDNA is an effective and efficient method to detect EGFR mutation status in NSCLC and could complement tissue based testing to improve diagnostic accuracy and yield in conjunction with tissue based testing.

Detecting acquired resistance mechanism and defining sequencing of targeted therapies based on emergence of resistant clones

Acquired resistance to targeted therapy occurs through clonal evolution and dynamic changes in molecular make-up of the tumors. Treatment with targeted drugs causes selective pressures leading to temporal molecular heterogeneity which can have clinical and biological implications (28). There is increasing evidence that some of these acquired molecular events following therapeutic interventions have clinical implications. Most notable example is the emergence of a gatekeeper mutation in EGFR kinase domain, namely T790M in patients with EGFR mutant NSCLC on treatment with a first-generation EGFR targeted therapy. These secondary EGFR T790M mutations are seen in approximately 50% of EGFR-mutant tumors upon progression on a first-line EGFR inhibitor and predict response to newer generation EGFR inhibitors with higher specificity and potency against the T790M mutation (29,30). Unlike EGFR mutant NSCLC patients with ALK rearranged NSCLC appear to have a much broader array of on-target gatekeeper mutations (L1196M, G1269A, C1156Y, L1152R, I1151Tins, F1174C/L/V etc.) in the ALK domain and amplification of ALK (PMID, PMID, PMID, PMID) (31-34). In addition, on-target acquired genomic alterations tumors can develop alternate pathway activation like MET and KIT amplifications, BRAF, NRAS, FGFR2, PIK3CA, IGF1R and ERBB family of receptor mutations (35-38). This degree of heterogeneity, dynamic shifts in clonal cellular populations and polyclonal resistance mechanisms makes molecular work up for patients progressing on targeted therapy complicated. Despite advances in sequencing technologies for interrogating tissue biopsies there are operational challenges in acquiring biopsies at multiple time points during therapy. Moreover, the tissue based approaches do not capture the temporal or spatial heterogeneity accurately (39). Recently plasma ctDNA based assays for EGFR T790M were demonstrated to have high tissue concordance for EGFR T790M and shown to be predictive of response to osimertinib (40-42). In addition, EGFR C797S and L798I were identified as novel mechanisms of acquired resistance to osimertinib (43). Using more sensitive ctDNA NGS platforms one can identify sub-clonal mutations (like MET, PIK3CA, HER2, BRAF etc.) that may evolve into the dominate clone at progression. This could aide in designing potentially novel strategies for sequential targeted therapies targeting these resistant sub-clonal populations by using ctDNA monitoring resulting in prolonged benefit with lower risk of toxicity from combination approaches.


CTCs

Tumors shed cells into circulation and these cells have the potential to disseminate widely through the venous and arterial circulation with a potential to initiate a metastatic focus. These CTCs persist in circulation withstanding a variety of stresses both mechanical and metabolic. Generally epithelial cells in circulation lacking anchorage undergo apoptosis by a process called anoikis (44). Complex integrin-dependent mechanisms orchestrated by receptor tyrosine kinases like TrkB are essential to suppress anoikis and permit tumor invasion and metastasis (45). In addition the large size of the CTCs (20–30 µm) relative to the capillary lumens (~8 µm) allow for CTCs to get trapped in tissues and escape the circulation on their first pass (46). Recent advances in technology allow for accurate detection of CTCs with potential for clinical utility (47,48).

Detection and analysis

Two broad methodologies for CTC detection are label-based positive selection using tumor epithelial cell surface markers or label-independent negative selection approach based on biophysical or functional properties of CTCs (49). Label-based capture assumes that CTCs have the same phenotypic characteristics as the primary tumor. Specific markers like cytokeratin and EpCAM are used for positive selection techniques (50). CellSearch® system (Veridex, Raritan, NJ, USA) is a Food and Drug Administration (FDA) approved platform utilizing the positive selection approach for CTC enumeration for patients with castration-resistant prostate cancer, breast cancer and colon cancer (51-53). This method uses enrichment of cytokeratin and EpCAM positive cells by immunofluorescence staining. However, in NSCLC some of the CTCs may transform to mesenchymal cells and lack cytokeratin staining (54,55). There are no reliable surface markers that can be used to comprehensively sort and select NSCLC CTCs. Label-independent isolation of CTCs using biophysical and functional characteristics of CTCs appears to be an attractive alternative for patients with NSCLC. Isolation by Size of Epithelial Tumor cell (ISET®) platform allows for separation of CTCs using a filtration device which sorts the cells and permits cytological phenotyping of the cells (56,57). Some other recent platforms allow not just sorting of the morphologically distinct CTC populations but allow for live and functional CTC separation allowing opportunities for further molecular studies (58-61). In another non-labelled approach using micro-fluidic chip based platform CTCs are separated in laminar flow conditions due to physical interactions with EpCAM-coated microposts in the chip (47). These non-labelled approaches have superior CTC detection rates patients with NSCLC compared to EpCAM/Cytokeratin labelled sorting approaches. In addition to increased detection rates these platforms allow for several post-isolation downstream applications. These platforms that retain CTC viability can allow ex-vivo expansion and creating in-vitro and patient-derived xenograft models to investigate drug susceptibility and allow deeper understanding the molecular profiles and biologic behavior in response to therapeutic intervention (59,60). In addition to sequencing and gene expression studies such platforms have recently been used to evaluate expression of PD-L1 and could potentially be used as a biomarker and monitoring tool for PD1/PDL1 targeted drugs (61,62). However, these assays need further clinical validation for reproducibility and therapeutic utility prior to routine clinical use.

Clinical utility of CTC assays

Due to the lack of standardization of the methods, varying detection thresholds and lack of clear definition of CTCs across platforms the clinical utilization of CTC based assays has been somewhat limited.

Role as a prognostic biomarker

In a few studies done in patients with early stage NSCLC increased CTC were found to correlate with poor outcomes (63-65). This correlation with adverse outcomes was also seen in patients with advanced stage NSCLC having systemic chemotherapy (66,67). Though some studies failed to show the correlation with prognosis (67) a meta-analysis of 20 studies evaluating CTCs in NSCLC demonstrated a correlation with stage, lymph node status and the outcome (68). Despite these encouraging findings the use of CTCs enumeration in clinical practice is limited because of lack of proper prospective validation and need for standardization of cutoff values and isolation methods of CTCs.

Prediction of response to treatment

Like ctDNA based approaches CTC assays can be useful tools to detect and monitor oncogenic driver mutations particularly EGFR and ALK fusion genes. The concordance of EGFR mutations using CTC with tissue was reported to be over 80% (69,70). Beyond NGS sequencing CTCs have the potential for mRNA characterization and array-based comparative genomic hybridization studies (58,59).

Though both ctDNA and CTC platforms appear to be comparable and with equal clinical potential in serving the role of a “liquid biopsy” these platforms have some distinct differences in the clinical applications. The logistics of implementation of the liquid biopsy platforms are simpler with ctDNA based approaches because of ease of collection and storage for biobanking. Most importantly CTC approaches require specialized instrumentation for capture of the target cells and cut-offs of minimal CTC for the assays are yet to be standardized. The newer NGS platforms optimized for ctDNA assays are highly efficient with great degree of sensitivity in monitoring low levels of disease. In addition to potential role in complementing tissue based genotyping for patients with advanced stage NSCLC and ctDNA can be a very valuable tool for monitoring for minimal residual disease especially after surgical resection (8,20,22). However, utility of liquid biopsies in monitoring minimal residual disease in early stage NSCLC is yet to be demonstrated in a prospective trial. In addition to the technical differences with implementation of the ctDNA and CTC platforms there may be some differences in the biological distinctions that need to be noted. ctDNA could likely is derived from tumor cells actively shedding ctDNA or necrotic cells undergoing apoptosis compared to CTC which are tumor derived intact cells (71,72). Unlike ctDNA CTCs can provide more information regarding clonal evolution and allow single cell interrogation and truly representative of the level of intra-patient heterogeneity. Depending on the platform used CTCs could be intact viable tumor cells that can be invaluable for downstream analyses, allowing interrogation of the DNA, RNA or at the protein level (58-61). In addition, recent work demonstrating the utility of CTCs in developing models of in-vitro and in-vivo (patient derived xenografts) drug sensitivity hold great promise (59,60).


Conclusions

Despite significant advances in our understanding of the biology of lung cancer and improved scope of personalized therapy the overall improvement in outcomes of patients with NSCLC has been modest. There were significant advances in liquid biopsy technologies to interrogate the clonal evolution and heterogeneity of tumors. Having effective diagnostic tools like liquid biopsies can help monitor tumor clonal evolution of tumors during treatment and will enable more rationale disease monitoring and sequencing of targeted therapy.


Acknowledgements

None.


Footnote

Conflicts of Interest: The authors have no conflicts of interest to declare.


References

  1. Devarakonda S, Morgensztern D, Govindan R. Clinical applications of The Cancer Genome Atlas project (TCGA) for squamous cell lung carcinoma. Oncology (Williston Park) 2013;27:899-906. [PubMed]
  2. Gan TQ, Chen WJ, Qin H, et al. Clinical Value and Prospective Pathway Signaling of MicroRNA-375 in Lung Adenocarcinoma: A Study Based on the Cancer Genome Atlas (TCGA), Gene Expression Omnibus (GEO) and Bioinformatics Analysis. Med Sci Monit 2017;23:2453-64. [Crossref] [PubMed]
  3. Ettinger DS, Wood DE, Aisner DL, et al. Non-Small Cell Lung Cancer, Version 5.2017, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw 2017;15:504-35. [Crossref] [PubMed]
  4. Marusyk A, Almendro V, Polyak K. Intra-tumour heterogeneity: a looking glass for cancer? Nat Rev Cancer 2012;12:323-34. [Crossref] [PubMed]
  5. Lo YM, Corbetta N, Chamberlain PF, et al. Presence of fetal DNA in maternal plasma and serum. Lancet 1997;350:485-7. [Crossref] [PubMed]
  6. Leon SA, Shapiro B, Sklaroff DM, et al. Free DNA in the serum of cancer patients and the effect of therapy. Cancer Res 1977;37:646-50. [PubMed]
  7. Toro PV, Erlanger B, Beaver JA, et al. Comparison of cell stabilizing blood collection tubes for circulating plasma tumor DNA. Clin Biochem 2015;48:993-8. [Crossref] [PubMed]
  8. Diehl F, Schmidt K, Choti MA, et al. Circulating mutant DNA to assess tumor dynamics. Nat Med 2008;14:985-90. [Crossref] [PubMed]
  9. Underhill HR, Kitzman JO, Hellwig S, et al. Fragment Length of Circulating Tumor DNA. PLoS Genet 2016;12:e1006162. [Crossref] [PubMed]
  10. Lowes LE, Bratman SV, Dittamore R, et al. Circulating Tumor Cells (CTC) and Cell-Free DNA (cfDNA) Workshop 2016: Scientific Opportunities and Logistics for Cancer Clinical Trial Incorporation. Int J Mol Sci 2016.17. [PubMed]
  11. Giacona MB, Ruben GC, Iczkowski KA, et al. Cell-free DNA in human blood plasma: length measurements in patients with pancreatic cancer and healthy controls. Pancreas 1998;17:89-97. [Crossref] [PubMed]
  12. Dong L, Meng Y, Sui Z, et al. Comparison of four digital PCR platforms for accurate quantification of DNA copy number of a certified plasmid DNA reference material. Sci Rep 2015;5:13174. [Crossref] [PubMed]
  13. Li M, Diehl F, Dressman D, et al. BEAMing up for detection and quantification of rare sequence variants. Nat Methods 2006;3:95-7. [Crossref] [PubMed]
  14. Diehl F, Li M, He Y, et al. BEAMing: single-molecule PCR on microparticles in water-in-oil emulsions. Nat Methods 2006;3:551-9. [Crossref] [PubMed]
  15. Dressman D, Yan H, Traverso G, et al. Transforming single DNA molecules into fluorescent magnetic particles for detection and enumeration of genetic variations. Proc Natl Acad Sci U S A 2003;100:8817-22. [Crossref] [PubMed]
  16. Kinde I, Wu J, Papadopoulos N, et al. Detection and quantification of rare mutations with massively parallel sequencing. Proc Natl Acad Sci U S A 2011;108:9530-5. [Crossref] [PubMed]
  17. Forshew T, Murtaza M, Parkinson C, et al. Noninvasive identification and monitoring of cancer mutations by targeted deep sequencing of plasma DNA. Sci Transl Med 2012;4:136ra68. [Crossref] [PubMed]
  18. Dagogo-Jack I, Bernicker E, Li T, et al. Genomic profiling of circulating tumor DNA (ctDNA) from patients (pts) with advanced non-small cell lung cancer (NSCLC). J Clin Oncol 2017;35:9025. [Crossref]
  19. Bigot F, Pecuchet N, Blanchet B, et al. Early TKI-pharmokinetics and circulating tumor DNA (ctDNA) to predict outcome in patients with EGFR-mutated non-small cell lung cancer (NSCLC). J Clin Oncol 2017;35:11544.
  20. Garcia-Murillas I, Schiavon G, Weigelt B, et al. Mutation tracking in circulating tumor DNA predicts relapse in early breast cancer. Sci Transl Med 2015;7:302ra133. [Crossref] [PubMed]
  21. Ahn SM, Chan JY, Zhang Z, et al. Saliva and plasma quantitative polymerase chain reaction-based detection and surveillance of human papillomavirus-related head and neck cancer. JAMA Otolaryngol Head Neck Surg 2014;140:846-54. [Crossref] [PubMed]
  22. Lin JC, Wang WY, Chen KY, et al. Quantification of plasma Epstein-Barr virus DNA in patients with advanced nasopharyngeal carcinoma. N Engl J Med 2004;350:2461-70. [Crossref] [PubMed]
  23. Tie J, Cohen J, Wang Y, et al. The potential of circulating tumor DNA (ctDNA) to guide adjuvant chemotherapy decision making in locally advanced rectal cancer (LARC). J Clin Oncol 2017;35:3521.
  24. Crowley E, Di Nicolantonio F, Loupakis F, et al. Liquid biopsy: monitoring cancer-genetics in the blood. Nat Rev Clin Oncol 2013;10:472-84. [Crossref] [PubMed]
  25. Yung RC. Tissue diagnosis of suspected lung cancer: selecting between bronchoscopy, transthoracic needle aspiration, and resectional biopsy. Respir Care Clin N Am 2003;9:51-76. [Crossref] [PubMed]
  26. Schwaederlé MC, Patel SP, Husain H, et al. Utility of genomic assessment of blood-derived circulating tumor DNA (ctDNA) in patients with advanced lung adenocarcinoma. Clin Cancer Res 2017;23:5101-11. [Crossref] [PubMed]
  27. Qiu M, Wang J, Xu Y, et al. Circulating tumor DNA is effective for the detection of EGFR mutation in non-small cell lung cancer: a meta-analysis. Cancer Epidemiol Biomarkers Prev 2015;24:206-12. [Crossref] [PubMed]
  28. Russo M, Siravegna G, Blaszkowsky LS, et al. Tumor Heterogeneity and Lesion-Specific Response to Targeted Therapy in Colorectal Cancer. Cancer Discov 2016;6:147-53. [Crossref] [PubMed]
  29. Piotrowska Z, Niederst MJ, Karlovich CA, et al. Heterogeneity Underlies the Emergence of EGFRT790 Wild-Type Clones Following Treatment of T790M-Positive Cancers with a Third-Generation EGFR Inhibitor. Cancer Discov 2015;5:713-22. [Crossref] [PubMed]
  30. Barnes TA, O’Kane GM, Vincent MD, et al. Third-Generation Tyrosine Kinase Inhibitors Targeting Epidermal Growth Factor Receptor Mutations in Non-Small Cell Lung Cancer. Front Oncol 2017;7:113. [Crossref] [PubMed]
  31. Doebele RC, Pilling AB, Aisner DL, et al. Mechanisms of resistance to crizotinib in patients with ALK gene rearranged non-small cell lung cancer. Clin Cancer Res 2012;18:1472-82. [Crossref] [PubMed]
  32. Shaw AT, Friboulet L, Leshchiner I, et al. Resensitization to Crizotinib by the Lorlatinib ALK Resistance Mutation L1198F. N Engl J Med 2016;374:54-61. [Crossref] [PubMed]
  33. Katayama R, Friboulet L, Koike S, et al. Two novel ALK mutations mediate acquired resistance to the next-generation ALK inhibitor alectinib. Clin Cancer Res 2014;20:5686-96. [Crossref] [PubMed]
  34. Ignatius Ou SH, Azada M, Hsiang DJ, et al. Next-generation sequencing reveals a Novel NSCLC ALK F1174V mutation and confirms ALK G1202R mutation confers high-level resistance to alectinib (CH5424802/RO5424802) in ALK-rearranged NSCLC patients who progressed on crizotinib. J Thorac Oncol 2014;9:549-53. [Crossref] [PubMed]
  35. Lin JJ, Riely GJ, Shaw AT. Targeting ALK: Precision Medicine Takes on Drug Resistance. Cancer Discov 2017;7:137-55. [Crossref] [PubMed]
  36. Yamaguchi N, Lucena-Araujo AR, Nakayama S, et al. Dual ALK and EGFR inhibition targets a mechanism of acquired resistance to the tyrosine kinase inhibitor crizotinib in ALK rearranged lung cancer. Lung Cancer 2014;83:37-43. [Crossref] [PubMed]
  37. Sasaki T, Koivunen J, Ogino A, et al. A novel ALK secondary mutation and EGFR signaling cause resistance to ALK kinase inhibitors. Cancer Res 2011;71:6051-60. [Crossref] [PubMed]
  38. Morgillo F, Della Corte CM, Fasano M, et al. Mechanisms of resistance to EGFR-targeted drugs: lung cancer. ESMO Open 2016;1:e000060. [Crossref] [PubMed]
  39. Fisher R, Pusztai L, Swanton C. Cancer heterogeneity: implications for targeted therapeutics. Br J Cancer 2013;108:479-85. [Crossref] [PubMed]
  40. Oxnard GR, Paweletz CP, Kuang Y, et al. Noninvasive detection of response and resistance in EGFR-mutant lung cancer using quantitative next-generation genotyping of cell-free plasma DNA. Clin Cancer Res 2014;20:1698-705. [Crossref] [PubMed]
  41. Jänne PA, Yang JC, Kim DW, et al. AZD9291 in EGFR inhibitor-resistant non-small-cell lung cancer. N Engl J Med 2015;372:1689-99. [Crossref] [PubMed]
  42. Kris MG, Johnson BE, Berry LD, et al. Using multiplexed assays of oncogenic drivers in lung cancers to select targeted drugs. JAMA 2014;311:1998-2006. [Crossref] [PubMed]
  43. Thress KS, Paweletz CP, Felip E, et al. Acquired EGFR C797S mutation mediates resistance to AZD9291 in non-small cell lung cancer harboring EGFR T790M. Nat Med 2015;21:560-2. [Crossref] [PubMed]
  44. Guo W, Giancotti FG. Integrin signalling during tumour progression. Nat Rev Mol Cell Biol 2004;5:816-26. [Crossref] [PubMed]
  45. Douma S, Van Laar T, Zevenhoven J, et al. Suppression of anoikis and induction of metastasis by the neurotrophic receptor TrkB. Nature 2004;430:1034-9. [Crossref] [PubMed]
  46. Valastyan S, Weinberg RA. Tumor metastasis: molecular insights and evolving paradigms. Cell 2011;147:275-92. [Crossref] [PubMed]
  47. Nagrath S, Sequist LV, Maheswaran S, et al. Isolation of rare circulating tumour cells in cancer patients by microchip technology. Nature 2007;450:1235-9. [Crossref] [PubMed]
  48. Pantel K, Brakenhoff RH, Brandt B. Detection, clinical relevance and specific biological properties of disseminating tumour cells. Nat Rev Cancer 2008;8:329-40. [Crossref] [PubMed]
  49. Yap TA, Lorente D, Omlin A, et al. Circulating tumor cells: a multifunctional biomarker. Clin Cancer Res 2014;20:2553-68. [Crossref] [PubMed]
  50. de Wit S, van Dalum G, Lenferink AT, et al. The detection of EpCAM(+) and EpCAM(-) circulating tumor cells. Sci Rep 2015;5:12270. [Crossref] [PubMed]
  51. de Bono JS, Scher HI, Montgomery RB, et al. Circulating tumor cells predict survival benefit from treatment in metastatic castration-resistant prostate cancer. Clin Cancer Res 2008;14:6302-9. [Crossref] [PubMed]
  52. Cohen SJ, Punt CJ, Iannotti N, et al. Relationship of circulating tumor cells to tumor response, progression-free survival, and overall survival in patients with metastatic colorectal cancer. J Clin Oncol 2008;26:3213-21. [Crossref] [PubMed]
  53. Cristofanilli M, Hayes DF, Budd GT, et al. Circulating tumor cells: a novel prognostic factor for newly diagnosed metastatic breast cancer. J Clin Oncol 2005;23:1420-30. [Crossref] [PubMed]
  54. Hofman V, Ilie MI, Long E, et al. Detection of circulating tumor cells as a prognostic factor in patients undergoing radical surgery for non-small-cell lung carcinoma: comparison of the efficacy of the CellSearch Assay™ and the isolation by size of epithelial tumor cell method. Int J Cancer 2011;129:1651-60. [Crossref] [PubMed]
  55. Wu S, Liu S, Liu Z, et al. Classification of circulating tumor cells by epithelial-mesenchymal transition markers. PLoS One 2015;10:e0123976. [Crossref] [PubMed]
  56. Krebs MG, Sloane R, Priest L, et al. Evaluation and prognostic significance of circulating tumor cells in patients with non-small-cell lung cancer. J Clin Oncol 2011;29:1556-63. [Crossref] [PubMed]
  57. Farace F, Massard C, Vimond N, et al. A direct comparison of CellSearch and ISET for circulating tumour-cell detection in patients with metastatic carcinomas. Br J Cancer 2011;105:847-53. [Crossref] [PubMed]
  58. Kolostova K, Spicka J, Matkowski R, et al. Isolation, primary culture, morphological and molecular characterization of circulating tumor cells in gynecological cancers. Am J Transl Res 2015;7:1203-13. [PubMed]
  59. Baccelli I, Schneeweiss A, Riethdorf S, et al. Identification of a population of blood circulating tumor cells from breast cancer patients that initiates metastasis in a xenograft assay. Nat Biotechnol 2013;31:539-44. [Crossref] [PubMed]
  60. Zhang Z, Shiratsuchi H, Lin J, et al. Expansion of CTCs from early stage lung cancer patients using a microfluidic co-culture model. Oncotarget 2014;5:12383-97. [Crossref] [PubMed]
  61. Schehr JL, Schultz ZD, Warrick JW, et al. High Specificity in Circulating Tumor Cell Identification Is Required for Accurate Evaluation of Programmed Death-Ligand 1. PLoS One 2016;11:e0159397. [Crossref] [PubMed]
  62. Anantharaman A, Friedlander T, Lu D, et al. Programmed death-ligand 1 (PD-L1) characterization of circulating tumor cells (CTCs) in muscle invasive and metastatic bladder cancer patients. BMC Cancer 2016;16:744. [Crossref] [PubMed]
  63. Sienel W, Seen-Hibler R, Mutschler W, et al. Tumour cells in the tumour draining vein of patients with non-small cell lung cancer: detection rate and clinical significance. Eur J Cardiothorac Surg 2003;23:451-6. [Crossref] [PubMed]
  64. Hofman V, Bonnetaud C, Ilie MI, et al. Preoperative circulating tumor cell detection using the isolation by size of epithelial tumor cell method for patients with lung cancer is a new prognostic biomarker. Clin Cancer Res 2011;17:827-35. [Crossref] [PubMed]
  65. Hashimoto M, Tanaka F, Yoneda K, et al. Significant increase in circulating tumour cells in pulmonary venous blood during surgical manipulation in patients with primary lung cancer. Interact Cardiovasc Thorac Surg 2014;18:775-83. [Crossref] [PubMed]
  66. Matikas A, Syrigos KN, Agelaki S. Circulating Biomarkers in Non-Small-Cell Lung Cancer: Current Status and Future Challenges. Clin Lung Cancer 2016;17:507-16. [Crossref] [PubMed]
  67. Juan O, Vidal J, Gisbert R, et al. Prognostic significance of circulating tumor cells in advanced non-small cell lung cancer patients treated with docetaxel and gemcitabine. Clin Transl Oncol 2014;16:637-43. [Crossref] [PubMed]
  68. Wang J, Huang J, Wang K, et al. Prognostic significance of circulating tumor cells in non-small-cell lung cancer patients: a meta-analysis. PLoS One 2013;8:e78070. [Crossref] [PubMed]
  69. Marchetti A, Del Grammastro M, Felicioni L, et al. Assessment of EGFR mutations in circulating tumor cell preparations from NSCLC patients by next generation sequencing: toward a real-time liquid biopsy for treatment. PLoS One 2014;9:e103883. [Crossref] [PubMed]
  70. Punnoose EA, Atwal S, Liu W, et al. Evaluation of circulating tumor cells and circulating tumor DNA in non-small cell lung cancer: association with clinical endpoints in a phase II clinical trial of pertuzumab and erlotinib. Clin Cancer Res 2012;18:2391-401. [Crossref] [PubMed]
  71. Bronkhorst AJ, Wentzel JF, Aucamp J, et al. Characterization of the cell-free DNA released by cultured cancer cells. Biochim Biophys Acta 2016;1863:157-65.
  72. Alix-Panabières C, Pantel K. Clinical Applications of Circulating Tumor Cells and Circulating Tumor DNA as Liquid Biopsy. Cancer Discov 2016;6:479-91. [Crossref] [PubMed]
Cite this article as: Velcheti V, Pennell NA. Non-invasive diagnostic platforms in management of non-small cell lung cancer: opportunities and challenges. Ann Transl Med 2017;5(18):378. doi: 10.21037/atm.2017.08.24

Download Citation