Next Article in Journal
A Comparison Between First-, Second- and Third-Generation Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitors in Patients with Non-Small-Cell Lung Cancer and Brain Metastases
Previous Article in Journal
Persistent Ependymal Tumor Arising from an Immature Ovarian Teratoma: A Rare Case
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Case Report

Concomitant Rare KRAS and BRAF Mutations in Lung Adenocarcinoma: A Case Report

1
Department of Public Health, University of Naples Federico II, 80131 Naples, Italy
2
Department of Oncology, A.O.R.N. Sant’Anna e San Sebastiano, 81100 Caserta, Italy
*
Author to whom correspondence should be addressed.
J. Mol. Pathol. 2020, 1(1), 36-42; https://0-doi-org.brum.beds.ac.uk/10.3390/jmp1010006
Submission received: 8 October 2020 / Revised: 30 October 2020 / Accepted: 2 November 2020 / Published: 3 November 2020
(This article belongs to the Special Issue Molecular Pathology of Lung Cancer)

Abstract

:
In July 2020, an active smoker, 63-year old man was admitted to the oncology unit of A.O.R.N. Sant’Anna e San Sebastiano (Caserta, Italy). Chest radiology highlighted right pleural effusion. Total-body CT scanning revealed a solid lesion with lobulated contours in the apical segment of the upper right lobe. The patient’s oncologist requested a molecular assessment of EGFR, ALK, ROS1, BRAF, and KRAS, as well as an evaluation of PD-L1 expression level. To this end, we carried out NGS analysis, on DNA extracted from cytospins, by adopting a custom-designed NGS panel (SiRe®). Overall, no actionable mutations in the tested genes were identified. Conversely, concomitant BRAF exon 11 p.G469A and a KRAS exon 4 p.A146T mutations were detected. Owing to the limited data on the presence of KRAS exon 4 p.A146T point mutation in lung adenocarcinoma patients, a further molecular confirmatory analysis was carried out with a dedicated KRAS cartridge on a fully automated real time polymerase chain reaction. When DNA was extracted from the TTF-1 positive tumor cell slide, the same KRAS alteration was observed. Unfortunately, the patient died in August 2020 before having the chance to start any type of treatment.

1. Introduction

Non-small cell lung cancer (NSCLC) represents the leading cause of cancer-related deaths worldwide [1]. Since the disease is mostly diagnosed in its advanced stages, cytological samples may constitute the only source of tissue material for morph-molecular purposes [2]. However, because cytological samples are often scant, it becomes highly challenging to identify the myriad of disease-associated biomarkers for therapeutic purposes. Indeed, various institutions—namely, the College of American Pathologists (CAP), the International Association for the Study of Lung Cancer (IASLC), the Association for Molecular Pathology (AMP), the National Comprehensive Cancer Network (NCCN), and the American Society of Clinical Oncology (ASCO)—state that a minimum panel of genes, including Epidermal Growth Factor Receptor (EGFR), Anaplastic Lymphoma Kinase (ALK), ROS Proto-Oncogene 1 Receptor Tyrosine Kinase (ROS1), and V-Raf murine sarcoma viral oncogene homolog B (BRAF), must be tested before the administration of tyrosine kinase inhibitors (TKIs). In addition, it is required to evaluate the expression level of programmed death-ligand 1 (PD-L1) for treatment with immune-checkpoint inhibitors [3,4,5]. More recently, careful attention has also been paid to novel predictive biomarkers, such as Kirsten Rat Sarcoma Viral Oncogene Homolog (KRAS) exon 2 p.G12C, for the administration of specific TKIs (e.g., AMG510, Amgen, Thousand Oaks, CA) [6]. Hence, to optimize the limited amount of starting tissue material for the detection of a plethora of available biomarkers, laboratories have adopted next generation sequencing (NGS), a complex yet useful tool to assess several biomarkers for different patients, simultaneously [7].
Here, we report a rare case of NSCLC featuring an unusual association of BRAF and KRAS point mutations.

2. Case Report

In July 2020, an active smoker, 63-year old man was admitted to the oncology unit of A.O.R.N. Sant’Anna e San Sebastiano (Caserta, Italy). The patient reported severe dyspnoea and 30 days of persistent dry cough. Chest radiology highlighted right pleural effusion. Total-body CT scanning revealed a solid lesion with lobulated contours in the apical segment of the upper right lobe and great post-contrastographic enhancement with a large implantation base on the anterior mediastinal pleura indissociable from its bronco-vascular structures. The solid lesion measured 36 × 22 mm. The CT analysis also confirmed a concomitant apical–basal pleural effusion measuring up to 60 mm in diameter. Such lesion caused subtotal atelectasis of the middle and right lower lobe (Figure 1A,B). In addition, pathological pericentimetric para-tracheal, pre- and sub-carinal homolateral, aortic–pulmonary, and Barety window lymphadenopathies, measuring up to 2 cm in maximum diameter, were also observed. (Figure 1C,D). No other lesions were identified in the other examined body regions.
Evacuative palliative thoracentesis (about 1600 mL of bloody pleural fluid) and diagnostic bronchoscopy with biopsy of lymph node station number seven were performed. The cytological picture displayed atypical epithelial cells with severe nuclear abnormalities organized in glandular structures, as evidenced in H&E stained cytospins (Figure 2A,B). Immunocytochemical (ICC) analysis highlighted a strong positivity for thyroid transcription factor 1 (TTF-1) and negativity for calretinin and Wilms tumor protein (WT1); cytokeratin 7 (CK7) staining was not contributive (Figure 2C,D). Overall, on the basis of these clinical, radiological, morphological, and immunocytochemical features, a diagnosis of NSCLC favor adenocarcinoma was made.
Following the above mentioned guidelines, the patient’s oncologist requested a molecular assessment of EGFR, ALK, ROS1, BRAF, and KRAS, as well as an evaluation of PD-L1 expression level. To this end, we carried out NGS analysis, on DNA extracted from cytospins, by adopting a custom-designed NGS panel (SiRe®), developed and validated by the Predictive Molecular Pathology Laboratory of the University of Naples Federico II. This panel is able to detect 568 clinically relevant mutations in the following six genes: EGFR, KRAS, Neuroblastoma RAS Viral Oncogene Homolog (NRAS), BRAF, Platelet Derived Growth Factor Receptor Alpha [PDGFRA], and KIT Proto-Oncogene, Receptor Tyrosine Kinase (KIT) [8,9,10,11,12,13]. The analysis was performed on the Ion Torrent S5 (ThermoFisher Scientific, Waltham, MA) platform. Overall, no actionable mutations in the tested genes were identified. Conversely, concomitant BRAF exon 11 p.G469A (allelic frequency 42.9%) and a KRAS exon 4 p.A146T (allelic frequency 31.9%) mutations were detected (Figure 3A,B). Owing to the limited data on the presence of KRAS exon 4 p.A146T point mutation in lung adenocarcinoma patients, a further molecular confirmatory analysis was carried out with a dedicated KRAS cartridge on a fully automated real time polymerase chain reaction (RT-PCR) platform (IdyllaTM, Biocartis, Mechelen, Belgium) [14]. When DNA was extracted from the TTF-1 positive tumor cell slide, the same KRAS alteration was observed (Figure 3C). Unfortunately, the patient died in August 2020 before having the chance to start any type of treatment.

3. Ethical Statement

Written informed consent was obtained from the patient and documented in accordance with the general authorization to process personal data for scientific research purposes from ‘The Italian Data Protection Authority’ (http://www.garanteprivacy.it/web/guest/home/docweb/-/docwebdisplay/export/2485392). All information regarding human material was managed using anonymous numerical codes, and all samples were handled in compliance with the Declaration of Helsinki (http://www.wma.net/en/30publications/10policies/b3/).

4. Discussion

Here, we report a rare case of lung adenocarcinoma harboring concomitant KRAS exon 4 p.A146T (COSM19404) and BRAF exon 11 p.G469A (COSM460) point mutations. To the best of our knowledge, although KRAS and BRAF co-occurring mutations have already been described in the literature, no study has ever reported the co-occurrence of these two rare genomic events in NSCLC patients.
Typically, KRAS mutations are found in 25–30% of NSCLC patients, in particular in Western countries and in smokers [15]. Up to 90% of KRAS mutations are found in codon 12 (exon 2), and, in particular, in p.G12C and p.G12V, due to G:C to T:A transversion in smokers [16]. Preliminary studies from early clinical trials have indicated that although these mutations are associated with negative prognoses, KRAS exon 2 p.G12C mutations, which account for about 13% of NSCLC patients, have acquired a relevant positive predictive significance as actionable biomarker [17]. On the other hand, KRAS exon 4 p.A146T is a very rare event in NSCLC adenocarcinoma patients (0.08%) [18]. This mutation seems to be associated with elevated expression levels of both RAS-GTP and extracellular signal-regulated kinase (ERK) compared with wild-type RAS cells. Therefore, KRAS exon 4 p.A146T is considered an activating missense mutation [19,20]. Recently, Wang et al. further confirmed the very rare occurrence of KRAS exon 4 p.A146T (0.41%) in the metastatic lung adenocarcinoma setting [21]. Not surprisingly, KRAS exon 4 p.A146T point mutation plays a negative predictive role when EGFR TKIs are administered [22].
As opposed to KRAS mutations, BRAF mutations have a lower incidence, occurring in only 1.5–3.5% to 7–8% of NSCLC patients. They are typically associated with micropapillary adenocarcinoma histotype, and with current and former smokers [23]. Discordant results have however been reported on the prevalence of BRAF exon 15 p.V600E and non-p.V600E alterations [24,25,26]. Similar to KRAS exon 4 p.A146T point mutation, the occurrence of BRAF exon 11 p.G469A is reported in a low percentage of lung adenocarcinomas (0.42%) [18]. To date, some studies indicate that only BRAF exon 15 p.V600E point mutation may predict the response to the combination of dabrafenib (BRAF inhibitor) and trametinib (MEK inhibitor) [27,28]. By contrast, other studies have highlighted the sensitivity of BRAF non-p.V600 point mutations, including BRAF exon 11 p.G469A, to this drug combination [26].
As of today, the co-occurrence of KRAS and BRAF mutations in lung adenocarcinoma is not well established. In a large experience, the Lung Cancer Mutation Consortium evidenced the occurrence of BRAF mutations with other concomitant oncogenic drivers (including KRAS) in a limited number of patients (5%) [29]. Salimian et al. reported concomitant KRAS and BRAF alterations in only 11% of analyzed cases [25]. Interestingly, in our present case and in a previous experience [24], we found that all cases with a concomitant KRAS mutation harbored a BRAF non-p.V600E alteration [25]. Similarly, another study has reported activating KRAS mutations in 11 (18%) out of 63 BRAF mutated patients [30]. Of note, all cases featured a BRAF non-p.V600E point mutation. Furthermore, two harbored a KRAS exon 4 p.A146T with a BRAF exon 11 p.G466V or BRAF exon 15 p.K601N [30]. As far as treatment is concerned, Gautschi et al. highlighted that the co-existence of KRAS exon 2 p.G12V with a BRAF exon 15 p.V600K determined the resistance to BRAF TKI treatments [31]. However, novel therapeutic approaches are under investigation for these patients. Interestingly, promising results have been reported in both RAS and BRAF mutated cell lines and in phase I dose-escalation and expansion studies of two novel molecules, ulixertinib (ERK1/2 kinase inhibitor) and LY3009120 (pan-RAF inhibitor) [32,33].
In conclusion, this is the first report describing the association of two rare mutations in KRAS and BRAF (KRAS exon 4 p.A146T and BRAF exon 11 p.G469A) in a lung adenocarcinoma patient. In this experience, our custom-designed NGS panel enabled us to run a successful genetic testing despite having scant cytological material. Nonetheless, even though custom-designed NGS yield accurate data, we recommend that orthogonal methodologies should be implemented to guarantee the accuracy of very rare molecular events, such as the KRAS exon 4 p.A146T point mutation. Finally, one limitation of this case report is that, unfortunately, our patient died before he could benefit from a personalized treatment plan. Consequently, further studies will be needed to better define the role of these co-occurring mutations in the hope of designing personalized treatment strategies for advanced stage NSCLC patients.

Author Contributions

Conceptualization, A.I. and U.M.; Methodology, all authors; Software, all authors; Validation, all authors; Formal Analysis, all authors; Investigation, all authors; Resources, all authors; Data Curation, all authors; Writing—Original Draft Preparation, all authors; Writing—Review & Editing, all authors; Visualization, all authors; Supervision, U.M.; Project Administration, U.M. All authors have read and agreed to the published version of the manuscript.

Funding

The authors have not declared a specific grant for this research from any funding agency in the public, commercial, or not-for-profit sectors.

Acknowledgments

We thank Paola Merolla for editing the manuscript.

Conflicts of Interest

Umberto Malapelle reports personal fees (as speaker bureau or advisor) from Boehringer Ingelheim, AstraZeneca, Roche, MSD, Amgen and Merck, unrelated to the current work. The other authors declare that they have no conflict of interest.

References

  1. Siegel, R.L.; Miller, K.D.; Jemal, A. Cancer statistics, 2020. CA Cancer J. Clin. 2020, 70, 7–30. [Google Scholar] [CrossRef]
  2. Malapelle, U.; Bellevicine, C.; De Luca, C.; Salatiello, M.; De Stefano, A.; Rocco, D.; de Rosa, N.; Vitiello, F.; Russo, S.; Pepe, F.; et al. EGFR mutations detected on cytology samples by a centralized laboratory reliably predict response to gefitinib in non-small cell lung carcinoma patients. Cancer Cytopathol. 2013, 121, 552–560. [Google Scholar] [CrossRef] [PubMed]
  3. Lindeman, N.I.; Cagle, P.T.; Aisner, D.L.; Arcila, M.E.; Beasley, M.B.; Bernicker, E.H.; Colasacco, C.; Dacic, S.; Hirsch, F.R.; Kerr, K.; 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 Association for Molecular Pathology. Arch. Pathol. Lab. Med. 2018, 142, 321–346. [Google Scholar] [PubMed] [Green Version]
  4. Kalemkerian, G.P.; Narula, N.; Kennedy, E.B.; Biermann, W.A.; Donington, J.; Leighl, N.B.; Lew, M.; Pantelas, J.; Ramalingam, S.S.; Reck, M.; 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 Study of Lung Cancer/Association for Molecular Pathology Clinical Practice Guideline Update. J. Clin. Oncol. 2018, 36, 911–919. [Google Scholar] [PubMed]
  5. Ettinger, D.S.; Aisner, D.L.; Wood, D.E.; Akerley, W.; Bauman, J.; Chang, J.Y.; Chirieac, L.R.; D’Amico, T.A.; Dilling, T.J.; Dobelbower, M.; et al. NCCN Guidelines Insights: Non-Small Cell Lung Cancer, Version 5.2018. J. Natl. Compr. Cancer Netw. 2018, 16, 807–821. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. AMG 510 First to Inhibit “Undruggable” KRAS. Cancer Discov. 2019, 9, 988–989.
  7. Vigliar, E.; Malapelle, U.; de Luca, C.; Bellevicine, C.; Troncone, G. Challenges and opportunities of next-generation sequencing: A cytopathologist’s perspective. Cytopathology 2015, 26, 271–283. [Google Scholar] [CrossRef] [PubMed]
  8. Malapelle, U.; Mayo de-Las-Casas, C.; Rocco, D.; Garzon, M.; Pisapia, P.; Jordana-Ariza, N.; Russo, M.; Sgariglia, R.; De Luca, C.; Pepe, F.; et al. Development of a gene panel for next-generation sequencing of clinically relevant mutations in cell-free DNA from cancer patients. Br. J. Cancer. 2017, 116, 802–810. [Google Scholar] [CrossRef] [Green Version]
  9. Pepe, F.; De Luca, C.; Smeraglio, R.; Pisapia, P.; Sgariglia, R.; Nacchio, M.; Russo, M.; Serra, N.; Rocco, D.; Battiloro, C.; et al. Performance analysis of SiRe next-generation sequencing panel in diagnostic setting: Focus on NSCLC routine samples. J. Clin. Pathol. 2019, 72, 38–45. [Google Scholar] [CrossRef]
  10. Pisapia, P.; Pepe, F.; Smeraglio, R.; Russo, M.; Rocco, D.; Sgariglia, R.; Nacchio, M.; De Luca, C.; Vigliar, E.; Bellevicine, C.; et al. Cell free DNA analysis by SiRe(®) next generation sequencing panel in non small cell lung cancer patients: Focus on basal setting. J. Thorac. Dis. 2017, 9 (Suppl. 13), S1383–S1390. [Google Scholar] [CrossRef] [Green Version]
  11. Pisapia, P.; Rocco, D.; Pepe, F.; De Luca, C.; Battiloro, C.; Smeraglio, R.; Cieri, M.; Bellevicine, C.; Troncone, G.; Malapelle, U. EGFR exon 19 deletion switch and development of p.L792Q mutation as a new resistance mechanism to osimertinib: A case report and literature review. Transl. Cancer Res. 2019, 8 (Suppl. 1), S64–S69. [Google Scholar] [CrossRef]
  12. Clery, E.; Pisapia, P.; Migliatico, I.; Pepe, F.; De Luca, C.; Russo, M.; De Rosa, F.; Smeraglia, F.; Insabato, L.; Vigliar, E.; et al. Cytology meets next generation sequencing and liquid biopsy: A case of lung adenocarcinoma presenting as metastasis to the phalanx. Diagn. Cytopathol. 2020, 48, 759–764. [Google Scholar] [CrossRef] [PubMed]
  13. Malapelle, U.; Pepe, F.; Pisapia, P.; Sgariglia, R.; Nacchio, M.; De Luca, C.; Lacalamita, R.; Tommasi, S.; Pinto, R.; Palomba, G.; et al. Harmonization of Next-Generation Sequencing Procedure in Italian Laboratories: A Multi-Institutional Evaluation of the SiRe® Panel. Front Oncol. 2020, 10, 236. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Weyn, C.; Van Raemdonck, S.; Dendooven, R.; Maes, V.; Zwaenepoel, K.; Lambin, S.; Pauwels, P. Clinical performance evaluation of a sensitive, rapid low-throughput test for KRAS mutation analysis using formalin-fixed, paraffin-embedded tissue samples. BMC Cancer 2017, 17, 139. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Downward, J. Targeting RAS signalling pathways in cancer therapy. Nat. Rev. Cancer 2003, 3, 11–22. [Google Scholar] [CrossRef]
  16. Passiglia, F.; Malapelle, U.; Del Re, M.; Righi, L.; Pagni, F.; Furlan, D.; Danesi, R.; Troncone, G.; Novello, S. KRAS inhibition in non-small cell lung cancer: Past failures, new findings and upcoming challenges. Eur. J. Cancer 2020, 137, 57–68. [Google Scholar] [CrossRef]
  17. Dueling KRASG12C Inhibitors Achieve Responses. Cancer Discov. 2020, 10, 10. [CrossRef]
  18. AACR Project GENIE Consortium. AACR Project GENIE: Powering Precision Medicine through an International Consortium. Cancer Discov. 2017, 7, 818–831. [Google Scholar] [CrossRef] [Green Version]
  19. Janakiraman, M.; Vakiani, E.; Zeng, Z.; Pratilas, C.A.; Taylor, B.S.; Chitale, D.; Halilovic, E.; Wilson, M.; Huberman, K.; Ricarte Filho, J.C.; et al. Genomic and biological characterization of exon 4 KRAS mutations in human cancer. Cancer Res. 2010, 70, 5901–5911. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Poulin, E.J.; Bera, A.K.; Lu, J.; Lin, Y.J.; Strasser, S.D.; Paulo, J.A.; Huang, T.Q.; Morales, C.; Yan, W.; Cook, J.; et al. Tissue-Specific Oncogenic Activity of KRASA146T. Cancer Discov. 2019, 9, 738–755. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. Wang, H.; Lv, Q.; Xu, Y.; Cai, Z.; Zheng, J.; Cheng, X.; Dai, Y.; Jänne, P.A.; Ambrogio, C.; Köhler, J. An integrative pharmacogenomics analysis identifies therapeutic targets in KRAS-mutant lung cancer. EBioMedicine 2019, 49, 106–117. [Google Scholar] [CrossRef] [PubMed]
  22. Ruan, X.; Sun, Y.; Wang, W.; Ye, J.; Zhang, D.; Gong, Z.; Yang, M. Multiplexed molecular profiling of lung cancer with malignant pleural effusion using next generation sequencing in Chinese patients. Oncol. Lett. 2020, 19, 3495–3505. [Google Scholar] [CrossRef]
  23. Leonetti, A.; Facchinetti, F.; Rossi, G.; Minari, R.; Conti, A.; Friboulet, L.; Tiseo, M.; Planchard, D. BRAF in non-small cell lung cancer (NSCLC): Pickaxing another brick in the wall. Cancer Treat. Rev. 2018, 66, 82–94. [Google Scholar] [CrossRef]
  24. Pisapia, P.; Pepe, F.; Malapelle, U.; Troncone, G. BRAF Mutations in Lung Cancer. Acta Cytol. 2019, 63, 247–250. [Google Scholar] [CrossRef]
  25. Salimian, K.J.; Fazeli, R.; Zheng, G.; Ettinger, D.; Maleki, Z. V600E BRAF versus Non-V600E BRAF Mutated Lung Adenocarcinomas: Cytomorphology, Histology, Coexistence of Other Driver Mutations and Patient Characteristics. Acta Cytol. 2018, 62, 79–84. [Google Scholar] [CrossRef]
  26. Noeparast, A.; Teugels, E.; Giron, P.; Verschelden, G.; De Brakeleer, S.; Decoster, L.; De Grève, J. Non-V600 BRAF mutations recurrently found in lung cancer predict sensitivity to the combination of Trametinib and Dabrafenib. Oncotarget 2016, 8, 60094–60108. [Google Scholar] [CrossRef] [Green Version]
  27. Planchard, D.; Besse, B.; Groen, H.J.M.; Souquet, P.J.; Quoix, E.; Baik, C.S.; Barlesi, F.; Kim, T.M.; Mazieres, J.; Novello, S.; et al. Dabrafenib plus trametinib in patients with previously treated BRAF(V600E)-mutant metastatic non-small cell lung cancer: An open-label, multicentre phase 2 trial. Lancet Oncol. 2016, 17, 984–993. [Google Scholar] [CrossRef] [Green Version]
  28. Planchard, D.; Smit, E.F.; Groen, H.J.M.; Mazieres, J.; Besse, B.; Helland, Å.; Giannone, V.; D’Amelio AMJr Zhang, P.; Mookerjee, B.; Johnson, B.E. Dabrafenib plus trametinib in patients with previously untreated BRAFV600E-mutant metastatic non-small-cell lung cancer: An open-label, phase 2 trial. Lancet Oncol. 2017, 18, 1307–1316. [Google Scholar] [CrossRef]
  29. Villaruz, L.C.; Socinski, M.A.; Abberbock, S.; Berry, L.D.; Johnson, B.E.; Kwiatkowski, D.J.; Iafrate, A.J.; Varella-Garcia, M.; Franklin, W.A.; Camidge, D.R.; et al. Clinicopathologic features and outcomes of patients with lung adenocarcinomas harboring BRAF mutations in the Lung Cancer Mutation Consortium. Cancer 2015, 121, 448–456. [Google Scholar] [CrossRef] [Green Version]
  30. Illei, P.B.; Belchis, D.; Tseng, L.H.; Nguyen, D.; De Marchi, F.; Haley, L.; Riel, S.; Beierl, K.; Zheng, G.; Brahmer, J.R.; et al. Clinical mutational profiling of 1006 lung cancers by next generation sequencing. Oncotarget 2017, 8, 96684–96696. [Google Scholar] [CrossRef] [Green Version]
  31. Gautschi, O.; Milia, J.; Cabarrou, B.; Bluthgen, M.V.; Besse, B.; Smit, E.F.; Wolf, J.; Peters, S.; Früh, M.; Koeberle, D.; et al. Targeted Therapy for Patients with BRAF-Mutant Lung Cancer: Results from the European EURAF Cohort. J. Thorac. Oncol. 2015, 10, 1451–1457. [Google Scholar] [CrossRef] [Green Version]
  32. Sullivan, R.J.; Infante, J.R.; Janku, F.; Wong, D.J.L.; Sosman, J.A.; Keedy, V.; Patel, M.R.; Shapiro, G.I.; Mier, J.W.; Tolcher, A.W.; et al. First-in-Class ERK1/2 Inhibitor Ulixertinib (BVD-523) in Patients with MAPK Mutant Advanced Solid Tumors: Results of a Phase I Dose-Escalation and Expansion Study. Cancer Discov. 2018, 8, 184–195. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Sullivan, R.J.; Hollebecque, A.; Flaherty, K.T.; Shapiro, G.I.; Rodon Ahnert, J.; Millward, M.J.; Zhang, W.; Gao, L.; Sykes, A.; Willard, M.D.; et al. A Phase I Study of LY3009120, a Pan-RAF Inhibitor, in Patients with Advanced or Metastatic Cancer. Mol. Cancer Ther. 2020, 19, 460–467. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Radiological evaluation. (A,B) Total-body CT scanning revealed a solid lesion (36 × 22 mm) with lobulated contours in the apical segment of the upper right lobe and great post-contrastographic enhancement with a large implantation base on the anterior mediastinal pleura indissociable from its bronco-vascular structures. A concomitant apical–basal pleural effusion measuring up to 60 mm in diameter was also evaluated. As a consequence, subtotal atelectasis of the middle and right lower lobe was observed. (C,D) Pathological pericentimetric para-tracheal, pre- and sub-carinal homolateral, aortic–pulmonary and Barety window lymphadenopathies, measuring up to 2 cm in maximum diameter, were observed.
Figure 1. Radiological evaluation. (A,B) Total-body CT scanning revealed a solid lesion (36 × 22 mm) with lobulated contours in the apical segment of the upper right lobe and great post-contrastographic enhancement with a large implantation base on the anterior mediastinal pleura indissociable from its bronco-vascular structures. A concomitant apical–basal pleural effusion measuring up to 60 mm in diameter was also evaluated. As a consequence, subtotal atelectasis of the middle and right lower lobe was observed. (C,D) Pathological pericentimetric para-tracheal, pre- and sub-carinal homolateral, aortic–pulmonary and Barety window lymphadenopathies, measuring up to 2 cm in maximum diameter, were observed.
Jmp 01 00006 g001
Figure 2. Cytological evaluation. (A,B) The cytospins cytological picture displayed atypical epithelial cells with severe nuclear abnormalities organized in glandular structures. (C,D) Immunocytochemical analysis highlighted a strong positivity for thyroid transcription factor 1 (TTF-1). On the basis of these morphological and immunocytochemical features, a diagnosis of non-small cell lung cancer (NSCLC) favor adenocarcinoma was made.
Figure 2. Cytological evaluation. (A,B) The cytospins cytological picture displayed atypical epithelial cells with severe nuclear abnormalities organized in glandular structures. (C,D) Immunocytochemical analysis highlighted a strong positivity for thyroid transcription factor 1 (TTF-1). On the basis of these morphological and immunocytochemical features, a diagnosis of non-small cell lung cancer (NSCLC) favor adenocarcinoma was made.
Jmp 01 00006 g002
Figure 3. Molecular evaluation. (A,B) Next generation sequencing analysis revealed concomitant BRAF exon 11 p.G469A (allelic frequency 42.9%) and KRAS exon 4 p.A146T (allelic frequency 31.9%) mutations. (C) A further molecular analysis was carried out with a dedicated KRAS cartridge on a fully automated real time polymerase chain reaction platform (IdyllaTM, Biocartis, Mechelen, Belgium), confirming the same KRAS alteration.
Figure 3. Molecular evaluation. (A,B) Next generation sequencing analysis revealed concomitant BRAF exon 11 p.G469A (allelic frequency 42.9%) and KRAS exon 4 p.A146T (allelic frequency 31.9%) mutations. (C) A further molecular analysis was carried out with a dedicated KRAS cartridge on a fully automated real time polymerase chain reaction platform (IdyllaTM, Biocartis, Mechelen, Belgium), confirming the same KRAS alteration.
Jmp 01 00006 g003
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Iaccarino, A.; Pisapia, P.; De Felice, M.; Pepe, F.; Gragnano, G.; De Luca, C.; Ianniello, G.; Malapelle, U. Concomitant Rare KRAS and BRAF Mutations in Lung Adenocarcinoma: A Case Report. J. Mol. Pathol. 2020, 1, 36-42. https://0-doi-org.brum.beds.ac.uk/10.3390/jmp1010006

AMA Style

Iaccarino A, Pisapia P, De Felice M, Pepe F, Gragnano G, De Luca C, Ianniello G, Malapelle U. Concomitant Rare KRAS and BRAF Mutations in Lung Adenocarcinoma: A Case Report. Journal of Molecular Pathology. 2020; 1(1):36-42. https://0-doi-org.brum.beds.ac.uk/10.3390/jmp1010006

Chicago/Turabian Style

Iaccarino, Antonino, Pasquale Pisapia, Marco De Felice, Francesco Pepe, Gianluca Gragnano, Caterina De Luca, Giovanni Ianniello, and Umberto Malapelle. 2020. "Concomitant Rare KRAS and BRAF Mutations in Lung Adenocarcinoma: A Case Report" Journal of Molecular Pathology 1, no. 1: 36-42. https://0-doi-org.brum.beds.ac.uk/10.3390/jmp1010006

Article Metrics

Back to TopTop