Journal Information
Vol. 30. Issue 1.
Pages 4-7 (January - February 2024)
Share
Share
Download PDF
More article options
Vol. 30. Issue 1.
Pages 4-7 (January - February 2024)
Comment
Full text access
Improving non-small-cell lung cancer survival through molecular characterization: Perspective of a multidisciplinary expert panel
Visits
1546
M.G.O. Fernandesa,b,c,
Corresponding author
gfernandes@med.up.pt

Corresponding author at: Alameda Prof. Hernâni Monteiro, 4200–319 Porto, Portugal
, A.S. Vilariçad, B. Fernandese, C. Camachof, C. Saraivag, F. Estevinhoh, H. Novais e Bastosa,b,c, J.M. Lopesi, P. Fidalgoj, P. Garridok, S. Alvesl, S. Silvam, T. Sequeiran, F. Baratao
a Pulmonology Department, Centro Hospitalar e Universitário de São João, EPE, Porto, Portugal
b Faculdade de Medicina da Universidade do Porto, Porto, Portugal
c IBMC/i3S – Instituto de Biologia Molecular e Celular/Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal Pulmonology Department, Centro Hospitalar e Universitário de São João, EPE, Porto, Portugal
d Pulmonology Department, Centro Hospitalar e Universitário de Lisboa Norte, EPE – Hospital Pulido Valente, Lisboa, Portugal
e Pulmonology Department, Hospital de Braga, Braga, Portugal
f Oncology Department, Serviço de Saúde da Região Autónoma da Madeira, Funchal, Portugal
g Pulmonology Department, Centro Hospitalar e Universitário do Algarve, EPE – Hospital de Portimão, Portugal
h Oncology Department, Unidade Local de Saúde de Matosinhos, EPE - Hospital Pedro Hispano, Matosinhos, Portugal
i Pulmonology Department, Hospital Garcia de Orta, EPE, Almada, Portugal
j Oncology Department, Centro Hospitalar e Universitário do Porto, EPE – Hospital de Santo António, Porto, Portugal
k Pulmonology Department, Fundação Champalimaud, Lisboa, Portugal
l Oncology Department, Instituto Português de Oncologia do Porto Francisco Gentil, Porto, Portugal
m Pulmonology Department, Centro Hospital de Leiria, EPE – Hospital de Santo André, Leiria, Portugal
n Oncology Department, Centro Hospitalar e Universitário de Lisboa Central, EPE – Hospital Santo António dos Capuchos, Lisboa, Portugal
o Pulmonology Department; Centro Hospitalar e Universitário de Coimbra, EPE – Hospitais da Universidade de Coimbra, Coimbra, Portugal
Ver más
This item has received
Article information
Full Text
Bibliography
Download PDF
Statistics
Tables (1)
Table 1. Molecular alterations with approved therapies in non-small-cell lung cancer
Full Text
Perspective of an expert panel on current challenges in the molecular characterization of non-small-cell lung cancer: What can be improved towards better treatment outcomes?

Substantial progress has been made over the last years in understanding critical molecular and cellular mechanisms driving tumor initiation and progression, with more than 50% of lung adenocarcinomas the main subtype of non-small-cell lung cancer (NSCLC) harboring oncogenic drivers.1–3 These findings led to the development of several novel drugs and treatment strategies and shifted the treatment paradigm of advanced NSCLC from a morphology-based to a predictive biomarker-driven approach based on tumor molecular genotyping.

Targeted therapies are the first treatment option for patients with advanced or metastatic disease with tumors harboring oncogenic mutations. In the absence of targetable oncogenic drivers, immunotherapy, either in monotherapy or combination, is the treatment of choice. Molecular alterations with approved therapies in NSCLC are depicted in Table 1.

Table 1.

Molecular alterations with approved therapies in non-small-cell lung cancer

Gene/protein alteration  Approved therapy 
EGFR exon 19 deletion or exon 21 L858R mutations  Afatinib*, dacomitinib*, erlotinib*, gefitinib*, osimertinib* 
EGFR S678I, L861Q, G719X mutations  Afatinib* 
EGFR exon 20 insertions  Avimantanab⁎⁎, mobocertinib⁎⁎ 
BRAF V600E mutation  Dabrafenib*, trametinib* 
ALK rearrangement  Alectinib*, brigatinib*, ceritinib*, crizotinib*, lorlatinib* 
ROS1 rearrangement  Crizotinib*, entrectinib* 
RET rearrangement  Pralsetinib*, selpercatinib* 
NTRK rearrangements  Entrectinib*, larotrectinib* 
MET exon 14 skipping mutation  Capmatinib⁎⁎, crizotinib*, tepotinib⁎⁎ 
KRAS G12C mutation  Sotorasib⁎⁎ 

FDA- and EMA-approved

⁎⁎

only FDA-approved

ALK, anaplastic lymphoma kinase; BRAF, B-Raf proto-oncogene; EGFR, epidermal growth factor receptor; HER2, human epidermal growth factor 2; KRAS, kirsten rat sarcoma viral oncogene homologue; MET, mesenchymal-epithelial transition; NRG1, neuregulin-1; NTRK, neurotrophic tyrosine receptor kinase 1; RET, rearranged during transfection; ROS1, C-ros oncogene 1

Given the established efficacy of targeted therapies in tumors harboring oncogenic drivers, the European and American guidelines recommend molecular testing for all advanced NSCLCs of non-squamous histology, particularly those with probable or definite adenocarcinoma, as in patients with non-adenocarcinoma histology, with low tobacco exposure, young age, or small specimen biopsy regardless of the performance status, to retrieve the most complete information to define the first-line treatment.4–9

Despite the recognition of the relevance of molecular characterization in the management of NSCLC, several challenges still need to be addressed and overcome in the clinical practice to ensure a complete and fast molecular assessment. An expert panel of pulmonologists and oncologists dedicated to thoracic Oncology convened to debate the molecular characterization of NSCLC at diagnosis and progression and identify key aspects to improve patient outcomes, highlighting the current evidence on NSCLC molecular profiling and discussing its benefits and challenges.

The following aspects were identified as most relevant for standardizing and optimizing the molecular diagnostic process considering both the laboratory and clinical approach:

  • Reflex testing has the advantage of optimizing sample management. It reduces the time until treatment initiation and should be performed by the pathologist after histological assessment.5

  • Molecular analysis should include a comprehensive gene panel, ideally a targeted multiplex next-generation sequencing (NGS) panel including point mutations, deletions, and rearrangements. Given the increasing number of mutations with potential clinical impact, NGS allows to optimize sample processing and simultaneously screen for several genes, with high throughput and sensitivity and low cost per test.10–13 Although NGS is a costly technique for most centres, it will predictably become more accessible in the future, as demonstrated in studies exploring the cost-effectiveness of the method.10

  • Biomarkers assessed should target genomic drivers with approved therapies, including the epidermal growth factor receptor (EGFR), the anaplastic lymphoma kinase (ALK), the C-ros oncogene 1 (ROS1), the rearranged during transfection (RET), and the B-Raf proto-oncogene (BRAF). In addition, given the fast pace of therapeutic progress, molecular alterations with targeted therapies in advanced stages of development or likely to become therapeutic targets in the short term should also be assessed, specifically those in the human epidermal growth factor 2 (HER2), neurotrophic tyrosine receptor kinase (NTRK), mesenchymal-epithelial transition (MET), and Kirsten rat sarcoma viral oncogene homologue (KRAS). This approach is in accordance with the European Society for Medical Oncology (ESMO) recommendations for the use of NGS in the clinical practice,10 and allows the treatment in routine clinical practice as well as access to ongoing clinical trials and early access programs.5,6

  • In specific cases of very symptomatic patients with aggressive disease and urgent need for treatment, rapid tests can be considered to define the first-line treatment, namely polymerase chain reaction (PCR) to detect EGFR mutations and immunohistochemistry or FISH to detect ALK and ROS1 rearrangements, while maintaining NGS ongoing.

  • In patients without sufficient tumor tissue to undergo molecular testing who are ineligible for rebiopsy, liquid biopsy can be considered to identify therapeutic targets. Liquid biopsy has several advantages, like avoiding the potential complications of tissue biopsy and allowing serial monitoring. In addition, it can provide a complete and real-time molecular profile, with information about clonal evolution and dynamic modifications within the tumor.14 The clinical use of liquid biopsy in detecting EGFR mutations in plasma from advanced NSCLC patients has been validated6,13,15–21 and is currently being assessed for other oncogenic drivers, as ALK, BRAF, ROS1, MEK, and HER2.13,22–25

  • Despite the significant improvements in patient outcomes achieved with EGFR-tyrosine kinase inhibitors (TKI), most patients acquire resistance and develop progressive disease within 10–12 months of treatment, limiting its long-term efficacy. This is particularly true when considering first- and second-generation TKIs.26 The most commonly acquired resistance mutation to first- and second-generation EGFR-TKIs is the T790M mutation in EGFR exon 20, identified in around 50–60% of cases.27–29 Other acquired resistance mechanisms to these inhibitors include alternative pathway activation through c-Met amplification, HER2 activation, and PIK3CA and BRAF mutations and histological transformation.30 T790M confers resistance to gefitinib, erlotinib, and afatinib, and its detection allows the use of the third-generation EGFR-TKI osimertinib in second line.24 At disease progression, rebiopsy should be considered to look for targetable resistance mechanisms. In the setting of EGFR-mutated disease, liquid biopsy can be the first step, as it is more accessible and less invasive than other methods. In cases of progression to third-generation TKIs, NGS is preferred to single-detection testing.8 Tissue biopsy should be considered in cases of negative or inconclusive liquid biopsy, progression to third-generation TKIs, and rapidly progressive disease, to investigate histological transformation.8

  • The average time between histological diagnosis and getting the molecular test result is heterogeneous among institutions, but ideally should not exceed two weeks.6 Minimizing bureaucratic issues and adopting reflex testing can reduce this time. Irrespective of the test being performed in-house or in an external laboratory, timely retrieval of results should be ensured.

  • The molecular study report should be presented in a simplified and systematic way and include the molecular test results (gene panel used and genomic changes identified and respective allele frequencies), their clinical interpretation given the available evidence, therapeutic options, and ongoing clinical trials (which should be regularly updated).31

  • Molecular study results should always be discussed within a multidisciplinary context, to allow complete and thorough data analysis. Defining a molecular tumor board is a recommended best practice that all centers should adopt.8

The management of NSCLC remains challenging, and the integration of data from predictive biomarkers in routine clinical practice can contribute to an optimal, individualized patient approach, particularly given the rapid emergence of effective targeted therapies. When considering molecular biomarker testing, the choice of the biomarker panel, target population, testing approach, and turnaround time are key issues that, when properly addressed, can improve the survival outcomes of NSCLC patients.

Funding

Medical writing assistance, supported financially by Boehringer Ingelheim Portugal, was provided by Prime Focus, during the preparation of this article

References
[1]
T Kohno, T Nakaoku, K Tsuta, K Tsuchihara, S Matsumoto, K Yoh, et al.
Beyond ALK-RET, ROS1 and other oncogene fusions in lung cancer.
Transl lung cancer Res, 4 (2015), pp. 156-164
[2]
K Sehgal, R Patell, D Rangachari, DB. Costa.
Targeting ROS1 rearrangements in non-small cell lung cancer with crizotinib and other kinase inhibitors.
Transl Cancer Res, 7 (2018), pp. S779-S786
[3]
D Zheng, R Wang, T Ye, S Yu, H Hu, X Shen, et al.
MET exon 14 skipping defines a unique molecular class of non-small cell lung cancer.
Oncotarget, 7 (2016), pp. 41691-41702
[4]
NI Lindeman, PT Cagle, MB Beasley, DA Chitale, S Dacic, G Giaccone, et al.
Molecular testing guideline for selection of lung cancer patients for EGFR and ALK tyrosine kinase inhibitors: guideline from the college of American Pathologists, International Association for the Study of Lung Cancer, and Association for Molecular Patho.
J Thorac Oncol, 8 (2013), pp. 823-859
[5]
KM Kerr, L Bubendorf, MJ Edelman, A Marchetti, T Mok, S Novello, et al.
Second ESMO consensus conference on lung cancer: pathology and molecular biomarkers for non-small-cell lung cancer.
Ann Oncol, 25 (2014), pp. 1681-1690
[6]
NI Lindeman, PT Cagle, DL Aisner, ME Arcila, MB Beasley, EH Bernicker, et al.
Updated molecular testing guideline for the selection of lung cancer patients for treatment with targeted tyrosine kinase inhibitors: guideline from the college of American pathologists, the International Association for the Study of Lung Cancer, and the.
Arch Pathol Lab Med, 142 (2018), pp. 321-346
[7]
GP Kalemkerian, N Narula, EB Kennedy, WA Biermann, J Donington, NB Leighl, et al.
Molecular testing guideline for the selection of patients with lung cancer for treatment with targeted tyrosine kinase inhibitors: American Society of Clinical Oncology Endorsement of the College of American Pathologists/International Association for the.
J Clin Oncol, 36 (2018), pp. 911-919
[8]
D Planchard, S Popat, K Kerr, S Novello, EF Smit, C Faivre-Finn, et al.
Metastatic non-small cell lung cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up.
Ann Oncol, 29 (2018), pp. iv192-iv237
[9]
DS Ettinger, DE Wood, DL Aisner, W Akerley, JR Bauman, A Bharat, et al.
NCCN guidelines insights: non–small cell lung cancer, version 2.2021: featured updates to the NCCN Guidelines.
J Natl Compr Cancer Netw, 19 (2021), pp. 254-266
[10]
F Mosele, J Remon, J Mateo, CB Westphalen, F Barlesi, MP Lolkema, et al.
Recommendations for the use of next-generation sequencing (NGS) for patients with metastatic cancers: a report from the ESMO Precision Medicine Working Group.
Ann Oncol, 31 (2020), pp. 1491-1505
[11]
T Yu, C Morrison, E Gold, A Tradonsky, A. Layton.
MA 11.06 retrospective analysis of NSCLC testing in low tumor content samples: single-gene tests, NGS, & the OncomineTM Dx Target Test.
J Thorac Oncol, 12 (2017), pp. S1845
[12]
GM Blumenthal, R. Pazdur.
Approvals in 2017: gene therapies and site-agnostic indications.
Nat Rev Clin Oncol, 15 (2018), pp. 127-128
[13]
C Rolfo, PC Mack, GV Scagliotti, P Baas, F Barlesi, TG Bivona, et al.
Liquid biopsy for advanced non-small cell lung cancer (NSCLC): a statement paper from the IASLC.
J Thorac Oncol, 13 (2018), pp. 1248-1268
[14]
G Siravegna, S Marsoni, S Siena, A. Bardelli.
Integrating liquid biopsies into the management of cancer.
Nat Rev Clin Oncol, 14 (2017), pp. 531-548
[15]
DH Shin, HS Shim, TJ Kim, HS Park, Choi Y La, WS Kim, et al.
Provisional guideline recommendation for EGFR gene mutation testing in liquid samples of lung cancer patients: a proposal by the korean cardiopulmonary pathology study group.
J Pathol Transl Med, 53 (2019), pp. 153-158
[16]
S Jenkins, JC-H Yang, SS Ramalingam, K Yu, S Patel, S Weston, et al.
Plasma ctDNA analysis for detection of the EGFR T790M mutation in patients with advanced non–small cell lung cancer.
J Thorac Oncol, 12 (2017), pp. 1061-1070
[17]
KS Thress, R Brant, TH Carr, S Dearden, S Jenkins, H Brown, et al.
EGFR mutation detection in ctDNA from NSCLC patient plasma: A cross-platform comparison of leading technologies to support the clinical development of AZD9291.
Lung Cancer, 90 (2015), pp. 509-515
[18]
C Zhou, M Wang, Y Cheng, Y Chen, X Ye.
Detection of EGFR T790M in Asia-Pacific patients (pts) with EGFR mutation-positive advanced non-small cell lung cancer (NSCLC): circulating tumour (ct) DNA analysis across 3 platforms.
Ann Oncol, 28 (2017), pp. v460-v496
[19]
B Weber, P Meldgaard, H Hager, L Wu, W Wei, J Tsai, et al.
Detection of EGFR mutations in plasma and biopsies from non-small cell lung cancer patients by allele-specific PCR assays.
BMC Cancer, 14 (2014), pp. 294
[20]
GR Oxnard, KS Thress, RS Alden, R Lawrance, CP Paweletz, M Cantarini, et al.
Association between plasma genotyping and outcomes of treatment with osimertinib (AZD9291) in advanced non–small-cell lung cancer.
J Clin Oncol, 34 (2016), pp. 3375-3382
[21]
JD Merker, GR Oxnard, C Compton, M Diehn, P Hurley, AJ Lazar, et al.
Circulating tumor DNA analysis in patients with cancer: American society of clinical oncology and college of American pathologists joint review.
Arch Pathol Lab Med, 142 (2018), pp. 1242-1253
[22]
P Bordi, M Tiseo, E Rofi, I Petrini, G Restante, R Danesi, et al.
Detection of ALK and KRAS mutations in circulating tumor DNA of patients with advanced ALK-Positive NSCLC with disease progression during crizotinib treatment.
Clin Lung Cancer, 18 (2017), pp. 692-697
[23]
CE McCoach, CM Blakely, KC Banks, B Levy, BM Chue, VM Raymond, et al.
Clinical utility of cell-free DNA for the detection of ALK fusions and genomic mechanisms of ALK Inhibitor resistance in non–small cell lung cancer.
Clin Cancer Res, 24 (2018), pp. 2758-2770
[24]
Y Tong, Z Zhao, B Liu, A Bao, H Zheng, J Gu, et al.
5′/3′ imbalance strategy to detect ALK fusion genes in circulating tumor RNA from patients with non-small cell lung cancer.
J Exp Clin Cancer Res, 37 (2018), pp. 68
[25]
Y Yang, X Shen, R Li, J Shen, H Zhang, L Yu, et al.
The detection and significance of EGFR and BRAF in cell-free DNA of peripheral blood in NSCLC.
Oncotarget, 8 (2017), pp. 49773-49782
[26]
S Kobayashi, TJ Boggon, T Dayaram, PA Jänne, O Kocher, M Meyerson, et al.
EGFR mutation and resistance of non–small-cell lung cancer to gefitinib.
N Engl J Med, 352 (2005), pp. 786-792
[27]
HA Yu, ME Arcila, N Rekhtman, CS Sima, MF Zakowski, W Pao, et al.
Analysis of tumor specimens at the time of acquired resistance to EGFR-TKI therapy in 155 patients with EGFR-Mutant Lung Cancers.
Clin Cancer Res, 19 (2013), pp. 2240-2247
[28]
D Westover, J Zugazagoitia, BC Cho, CM Lovly, L. Paz-Ares.
Mechanisms of acquired resistance to first- and second-generation EGFR tyrosine kinase inhibitors.
Ann Oncol, 29 (2018), pp. i10-i19
[29]
LV Sequist, BA Waltman, D Dias-Santagata, S Digumarthy, AB Turke, P Fidias, et al.
Genotypic and histological evolution of lung cancers acquiring resistance to EGFR inhibitors.
Sci Transl Med, 3 (2011),
[30]
AB Cortot, PA. Janne.
Molecular mechanisms of resistance in epidermal growth factor receptor-mutant lung adenocarcinomas.
Eur Respir Rev, 23 (2014), pp. 356-366
[31]
MM Li, M Datto, EJ Duncavage, S Kulkarni, NI Lindeman, S Roy, et al.
Standards and guidelines for the interpretation and reporting of sequence variants in cancer.
J Mol Diagnostics, 19 (2017), pp. 4-23
Pulmonology
Article options
Tools

Are you a health professional able to prescribe or dispense drugs?