Next Article in Journal
Use of Bioelectrical Impedance Analysis to Explore the Effectiveness of Stellate Ganglion Block in Patients with Post-Stroke Complex Regional Pain Syndrome: A Retrospective Pilot Study
Previous Article in Journal
Electrocauterization versus Ligation of Lymphatic Vessels to Prevent Lymphocele Development after Kidney Transplantation—A Meta-Analysis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Case Report

Genome Sequencing of Multiple Primary Lung Cancers Harbouring Mixed Histology and Spontaneously Regressing Small-Cell Lung Cancer

1
Genomic Oncology Research Group, Department of Physiology and Medical Physics, RCSI University of Medicine and Health Sciences, D02 YN77 Dublin, Ireland
2
Centre for Systems Medicine, RCSI University of Medicine and Health Sciences, D02 YN77 Dublin, Ireland
3
Our Lady of Lourdes Hospital, A92 VW28 Drogheda, Ireland
4
Medical Oncology Group, Department of Molecular Medicine, RCSI University of Medicine and Health Sciences, D02 YN77 Dublin, Ireland
5
St Luke’s Radiation Oncology Network, D08 E797 Dublin, Ireland
6
Department of Histopathology, St James’s Hospital, D08 E797 Dublin, Ireland
7
Department of Cardiothoracic Surgery, St James’s Hospital, D08 E797 Dublin, Ireland
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
These authors also contributed equally to this work.
J. Pers. Med. 2024, 14(3), 257; https://doi.org/10.3390/jpm14030257
Submission received: 13 December 2023 / Revised: 6 February 2024 / Accepted: 20 February 2024 / Published: 28 February 2024
(This article belongs to the Section Omics/Informatics)

Abstract

:
Up to 15% of lung cancer patients present two or more anatomically separate primary lung lesions, known as multiple primary lung cancers (MPLCs). While surgical resection or stereotactic body radiation therapy (SBRT) is the standard of care for most early-stage lung cancer cases, this may not be an option for patients with widespread tumours, highlighting the need for the improved targeted management of MPLC patients, which remains challenging. Moreover, the spontaneous regression (SR) of small-cell lung cancer (SCLC) is rare, with only four cases accounted for between 1988 and 2018. We report a rare MPLC case harbouring the mixed histology of non-small-cell lung cancer adenocarcinoma (NSCLCa) and SCLC and the SR of SCLC without treatment. The patient was diagnosed in 2015 with MPLCs, identified as NSCLCa and SCLC. In 2016, a restaging PET/CT scan prior to the start of treatment showed SCLC SR. In 2018, a further tumour was detected in the patient’s mandible, and a re-biopsy of the SCLC revealed histology consistent with NSCLCa. Whole-genome sequencing (WGS) analysis identified a high expression of programmed death ligand-1 (PDL-1) in the NSCLCa, which was treated with pembrolizumab. WGS revealed distinct genomic profiles and mutational mechanisms in MPLCs, suggesting the need for distinct targeted therapies to improve the management of MPLC patients and highlighting the importance of precision evaluation.

1. Introduction

Lung cancer is the leading cause of cancer death worldwide [1,2]. About 15% of lung lesions are small-cell lung cancers (SCLCs), while the remaining majority are non-small-cell lung cancers (NSCLCs), predominantly of the adenocarcinoma subtype (NSCLCa) [3,4]. These distinct clinical and histological subtypes display high molecular heterogeneity, which translates into different treatment responses: while chemotherapy shows beneficial results in the treatment of SCLCs, the standard of care for NSCLCs largely depends on the stage of the lesion, with early-stage tumours generally removed surgically or treated with SBRT, and more advanced ones treated with chemoradiation therapy [5,6]. Moreover, considerable survival benefits are provided by the administration of more specific and better-tolerated treatments in patients that exhibit specific, clinically relevant cancer biomarkers as targets for therapy [4]. Currently, several predictive biomarkers can be tested, such as the high expression of programmed death ligand-1 (PD-L1) and tumour mutational burden (TMB) for immune checkpoint inhibitors (i.e., pembrolizumab) [7,8,9,10], and EGFR mutations, with ALK and ROS1 gene fusions for tyrosine kinase inhibitors (i.e., gefitinib, erlotinib, crizotinib) [11,12,13,14,15]. Targeted therapies are especially relevant in the context of reduced tolerance to the standard of care in cases where the patient presents with widespread lesions, such as multiple primary lung cancers (MPLCs) and/or co-morbidities, and cannot endure surgery and chemoradiation therapy [16,17]. MPLCs are diagnosed in up to 15% of lung cancer patients [17]. MPLCs are physically distinct entities that occur in the same or different lung lobes. The development of MPLCs is an interesting phenomenon because it arises from an identical germline genetic background and environmental exposure and yet results in highly heterogeneous lesions. Previous studies on the molecular and genomic data of MPLCs have found that tumours developing in this context show clonally distinct molecular profiles, distinct oncogenic alterations, and can be of a different pathological type [18,19,20,21], suggesting variable responses of MPLCs to targeted therapies, and strong implications for the therapeutic management of MPLC patients. We describe a rare case of MPLCs, harbouring a mixed histology of SCLC and NSCLCa, which we analyse through whole genome sequencing. Moreover, the SCLC showed spontaneous regression (SR), making this case study a rare opportunity to gain insight into both MPLC development and the occurrence of SR.

2. Case Description

A 69-year-old female patient, a smoker known to have chronic obstructive pulmonary disease (COPD) and compensated alcoholic liver cirrhosis, presented in 2015 with the exacerbation of shortness of breath (SOB). A computed tomography scan (CT scan) and positron emission tomography/CT scan (PET/CT scan) revealed bilateral lung masses, one in the right lower lobe and one in the left upper lobe, with no suspicious lymph nodes or any other lesions. Endobronchial ultrasound (EBUS) biopsies were performed in September 2015 and identified an NSCLCa (sample NSC-1) in the right lung lesion and a SCLC (sample SC) in the left lung lesion. It was decided to surgically remove the NSCLCa first, followed by surgery on the SCLC. In October 2015, right lower lobe segmentectomy and mediastinal lymph node dissection were performed. Histopathology analysis confirmed the complete resection of pT1aN0 NSCLCa. However, the patient’s slow recovery made her unfit for further surgery. In April 2016, she was referred to the medical oncology service for the consideration of chemotherapy. In May 2016, a restaging PET/CT scan prior to starting treatment showed right lung post-surgical changes and partial left lung resolution. The left apical SCLC reduced in size from 28 mm to 8 mm, and its standardised uptake value (SUV) reduced from 16.5 to 2.1. A definite improvement and SR were, therefore, observed in the absence of any local or systemic treatment. Following a discussion at the lung cancer multidisciplinary meeting, it was decided to proceed with no treatment and start PET/CT scan surveillance. A stable disease, with no evidence of progression, was observed through three PET/CT scans from August 2016 to July 2017. In February 2018, the scan revealed a left apical mass increase to 22 mm and an SUV increase to 13.3, in addition to a 1 cm lesion in the right mandible. A biopsy from the mandibular lesion showed metastatic NSCLCa (sample NSC-3) with programmed death ligand-1 (PDL-1) = 60%. A re-biopsy of her left apical lung lesion showed NSCLCa (sample NSC-2), which prompted the start of patient treatment with pembrolizumab in late 2018. A restaging CT scan in January 2019 showed stable disease based on imRECIST. Histopathological analysis indicated that tumour NSC-1 was 16 mm with moderately differentiated invasive adenocarcinoma with a 40% acinar component, 30% lepidic, and 30% solid component. It was TTF-1-positive, CK 7-positive, and CK 20-negative. SC was 28 mm clinically on the PET/CT scan; the biopsy showed small-cell lung cancer with nuclear moulding and hyperchromasia with a high nuclear-to-cytoplasmic ratio. It was positive for TTF-1, AE/3, and synaptophysin with an MIB-1 index of 90%. For NSC-2, EBUS cellular aspirate showed cohesive clusters of malignant epithelial cells consistent with NSCLC. It was EGFR-negative, ALK-negative, and ROS-1-negative. NSC-3 was consistent with NSCLC with positive TTF-1, CK7, and Ber EP4. It was negative for Chromogranin, synaptophysin, CK5, 6, and CD56.
With the patient’s consent, it was decided to analyse the whole genome sequences (WGSs) of her four tumour samples (see Supplementary Methods). We identified the somatic single nucleotide variants (SNVs), small insertions and deletions (InDels), copy number alterations (CNAs), and structural variants (SVs) in the four tumour genomes to understand the different classifications of the left apical lung lesion (SCLC and NSCLCa), and to establish whether any genomic evidence existed for a potential transformation from SCLC to NSCLCa. Also, the results held potential benefits for the patient’s treatment if targetable driver mutations were identified.
Sadly, the patient subsequently developed autoimmune acute renal failure and passed away in 2019. A timeline of the patient’s clinical history is shown in Figure 1, and clinicopathologic data of the patient’s tumours are provided in Table 1.

2.1. Mutational Load, Mutational Overlap and Driver Analysis of SNVs, InDels and SVs

The NA WGS analysis of somatic variants revealed significant differences in mutational burdens across samples and between variant types. The SNV load was higher in NSC-3 (91,963) and SC (64,816) and much lower in NSC-1 (3303) and NSC-2 (6327). The InDel burden peaked in SC (2639), followed by NSC-3 (395), NSC-1 (56), and NSC-2 (13). The SV burden was highest in NSC-3 (153,274) and dropped in SC (5368), NSC-2 (4314), and NSC-1 (3167) (Figure 2A). The vast majority of somatic mutations were unique to each sample, with only 0.03% of SNVs common to all, suggesting that the primary lesions in the patient are genetically distinct and likely to have originated independently. The highest overlap of SNVs (1.95%) and InDels (0.16%) was seen between NSC-2 and NSC-3 (Figure 2B), which included a KRAS G12C mutation. TP53 was mutated in SC only (Figure 2C).

2.2. Mutational Signatures

Mutational signature analysis was performed to inform the exposures and biological history of each cancer and investigate the mutational processes that occurred during tumour development. Evidence of the smoking-related signature SBS4 was found in NSC-1, SC, and NSC-2 but not in NSC-3. NSC-1 and NSC-3 show evidence of the homologous recombination (HR)-related signature SBS3, though no mutations in HR-associated genes were found. SBS18, linked to damage by reactive oxygen species, was found in SC. The majority of the identified signatures, however, are of unknown aetiology (SBS12, SBS33, SBS37, SBS39, SBS40) or the result of possible sequencing artefacts (SBS46) (Figure 2D).

2.3. Copy Number Alterations and CNA Driver Analysis

Copy number alteration analysis revealed broadly stable genomes in the lung adenocarcinomas NSC-1 and NSC-2 (ploidy: 2.2 and 2.1, respectively) and higher chromosomal instability (CIN) in samples SC and NSC-3 (ploidy: 3.3 and 3.2, respectively) (Figure 3A). The copy number state of a set of known lung cancer genes is shown in Figure 3B and further highlights the heterogeneity of the samples.

3. Discussion

In this report, we describe an extremely rare case of SR in MPLC. This case presents the following several uncommon features: the development of histologically distinct MPLCs; a possible transition from SCLC to NSCLCa; and the SR of the SCLC.
We performed WGS on the four tumour samples diagnosed in the patient to establish the level of heterogeneity and the clonal relationship between the MPLCs and found evidence of clinically actionable alterations. We observed high genomic heterogeneity and distinct known cancer-associated events between all MPLC samples, suggesting independent origins in a carcinogen-damaged field. A KRAS G12C mutation, associated with response to KRAS inhibitors as well as resistance to tyrosine kinase inhibitors and poorer prognosis [22], was found in NSC-2 primary and NSC-3 mandible adenocarcinomas. This common driver might suggest that the mandibular tumour in the patient originated from the primary adenocarcinoma NSC-2; however, more evidence is needed to support this claim, especially as the KRAS G12C mutation is a frequent driver event that could have developed independently in each sample. EGFR amplification, associated with poor prognosis [8,11], was reported in all samples. MET amplifications were seen in NSC-1, SC, and NSC-3 and are associated with poor prognosis, though several MET inhibitors might show beneficial results in the NSCLC treatment [23,24]. The mandibular NSCLCa lesion carried the highest TMB, which, together with the high levels of PD-L1, makes it a good candidate for immunotherapy with pembrolizumab [7,10]. Overall, the scarcity of potential tumorigenic events found among known lung cancer driver genes in the patient might indeed be indicative of the occurrence of further drivers—genetic, epigenetic, or transcriptomic—that are yet to be identified.
A likely driving factor for developing bilateral synchronous lung malignancy, in particular SCLC, was the patient’s smoking history. While there is a known association between COPD and lung cancer, we were not able to directly correlate her stable COPD (or liver disease) to her primary cancer diagnosis. After primary surgery, the patient was on follow-up for her SCLC, maintained a healthy lifestyle, and had been a non-smoker for several years. We were not able to attribute the later recurrence and progression of her disease to her known medical and clinical history.
Due to the patient’s known exposure to tobacco smoke and as a result of the mutations induced by its associated carcinogens, we expected all lung samples to have high TMB. However, TMB varied greatly between the tumours, and the two lung NSCLCa samples displayed surprisingly low TMB. Evidence of the smoking-related signature (SBS4) was confirmed by mutational signature analysis in all primary lung tumours but not in the mandibular lesion. This is an interesting result because, although previous reports highlight the key role of tobacco smoke-related mutations in tumour initiation but not in tumour progression and the later stages of tumour development [25], we would still expect metastatic cancer cells to carry mutations that originate in their primary lesion [26].
While the histological transformation from adenocarcinoma to SCLC has been previously observed in the context of acquired resistance to EGFR-targeted therapies [27], the change from SCLC to NSCLCa is extremely unusual. The highly heterogeneous genomic profiles and the presence of the TP53 mutation in SC but not NSC_2 appear to rule out the hypothesis of a phenotypic transformation from SCLC to NSCLCa. Indeed, it is possible that what we initially considered as a phenotypic transformation might have been, as observed in a small subset of lung cancers [28], a tumour harbouring the combined histology of SCLC and adenocarcinoma, the co-existence of which was not detected by the two biopsies of the lesion. Nevertheless, this finding has strong implications in clinical practice, where the potential of histologic diversity should be accounted for and where repeat biopsies might be crucial to establish the most appropriate treatment strategy.
SR, defined by Everson and Cole as the partial or complete disappearance of a malignant tumour in the absence of all treatment or in the presence of therapy which is considered inadequate to exert a significant influence on neoplastic disease, is rarely observed in malignancy [29,30]. Kumar later developed a “modified Everson and Cole criterion”, which was defined as the partial or complete disappearance of the tumour in the absence of all systemic or local treatment of the primary or metastatic lesion [31]. Based on the modified criterion, between 1951 and 2008, only five cases of primary thoracic malignancies showed SR, none of which were SCLC [31]. Between 1988 and 2018, 14 cases showed true SR with lung primary origin, only 4 of which were SCLC [29].
Our case fulfils Everson and Cole’s original and modified criteria. Our patient experienced the SR of her primary SCLC without any local or systemic treatment. This makes her one of the very few cases reported in the last 70 years [29,31]. Factors that stimulate an immune response, such as infection, trauma, surgery, and blood transfusions, have been proposed as potential explanations for SR, though the underlying mechanisms of this phenomenon remain unclear [29,31,32,33,34,35].
This study also reveals the genomic heterogeneity between the mandibular NSC-3 tumour and the primary cancers in the patient in terms of mutational overlap, driver events, and mutational processes, thus questioning its metastatic nature [26]. The analysis of multi-region biopsies should be considered in the future, as this might provide key information on the evolutionary relationship between lesions. As reported in other cancer types [36], patients with multiple primary lesions present complex scenarios that need to be carefully assessed. Our study revealed the status of several known targetable biomarkers for each MPLC, providing essential information for the personalised treatment of the patient.

4. Conclusions

In conclusion, we presented an in-depth analysis of a rare case of MPLCs and SR. Our study of MPLCs within a patient highlights the heterogeneity of both the genomic landscape and the lung cancer biomarkers between multiple primary tumours, which could have strong implications for therapeutic management. The occurrences of MPLCs and SR are unusual and poorly understood phenomena, warranting further investigation of their underlying mechanisms.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/jpm14030257/s1, Supplementary Methods.

Author Contributions

Conceptualisation and design, A.R., S.J.F., S.T. and B.T.H.; sample acquisition, analysis, and data collection, A.R., S.N., C.F., J.H. and V.Y., bioinformatic analysis, V.T. and S.J.F., manuscript preparation and review, all authors. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by the North-East Cancer Research and Education Trust (NECRET). S.J.F. acknowledges support from the Royal College of Surgeons in Ireland StAR programme and the SFI-HRB-Wellcome Research Partnership (212529/Z/18/Z).

Informed Consent Statement

Informed consent was obtained from the subject involved in this study.

Data Availability Statement

The datasets supporting the conclusion of this article are available through the Genomic Oncology Research Group Data Access Committee. The names of the repository/repositories and accession number(s) can be found below: European Genome-Phenome Archive (https://ega-archive.org/—Project ID: EGAC00001001585; accessed on 10 December 2023).

Acknowledgments

We would like to thank the Irish Centre for High End Computing (https://www.ichec.ie/, accessed on 10 December 2023) for the use of HPC infrastructure.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef]
  2. Siegel, R.L.; Miller, K.D.; Jemal, A. Cancer statistics, 2020. CA Cancer J. Clin. 2020, 70, 7–30. [Google Scholar] [CrossRef]
  3. George, J.; Lim, J.S.; Jang, S.J.; Cun, Y.; Ozretić, L.; Kong, G.; Leenders, F.; Lu, X.; Fernández-Cuesta, L.; Bosco, G.; et al. Comprehensive genomic profiles of small cell lung cancer. Nature 2015, 524, 47–53. [Google Scholar] [CrossRef]
  4. Herbst, R.S.; Morgensztern, D.; Boshoff, C. The biology and management of non-small cell lung cancer. Nature 2018, 553, 446–454. [Google Scholar] [CrossRef]
  5. Zito Marino, F.; Bianco, R.; Accardo, M.; Ronchi, A.; Cozzolino, I.; Morgillo, F.; Rossi, G.; Franco, R. Molecular Heterogeneity in Lung Cancer: From Mechanisms of Origin to Clinical Implications. Int. J. Med. Sci. 2019, 16, 981–989. [Google Scholar] [CrossRef]
  6. van Meerbeeck, J.P.; Fennell, D.A.; De Ruysscher, D.K. Small-cell lung cancer. Lancet 2011, 378, 1741–1755. [Google Scholar] [CrossRef]
  7. Garon, E.B.; Rizvi, N.A.; Hui, R.; Leighl, N.; Balmanoukian, A.S.; Eder, J.P.; Patnaik, A.; Aggarwal, C.; Gubens, M.; Horn, L.; et al. Pembrolizumab for the Treatment of Non–Small-Cell Lung Cancer. N. Engl. J. Med. 2015, 372, 2018–2028. [Google Scholar] [CrossRef] [PubMed]
  8. Villalobos, P.W., II. Lung Cancer Biomarkers. Hematol. Oncol. Clin. N. Am. 2017, 31, 13–29. [Google Scholar] [CrossRef] [PubMed]
  9. Chan, T.; Yarchoan, M.; Jaffee, E.; Swanton, C.; Quezada, S.; Stenzinger, A.; Peters, S. Development of tumor mutation burden as an immunotherapy biomarker: Utility for the oncology clinic. Ann. Oncol. 2018, 30, 44–56. [Google Scholar] [CrossRef]
  10. Rizvi, N.A.; Hellmann, M.D.; Snyder, A.; Kvistborg, P.; Makarov, V.; Havel, J.J.; Lee, W.; Yuan, J.; Wong, P.; Ho, T.S.; et al. Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non–small cell lung cancer. Science 2015, 348, 124–128. [Google Scholar] [CrossRef] [PubMed]
  11. Sharma, S.V.; Bell, D.W.; Settleman, J.; Haber, D.A. Epidermal growth factor receptor mutations in lung cancer. Nat. Rev. Cancer 2007, 7, 169–181. [Google Scholar] [CrossRef] [PubMed]
  12. Lindeman, N.I.; Cagle, P.T.; Beasley, M.B.; Chitale, D.A.; Dacic, S.; Giaccone, G.; Jenkins, R.B.; Kwiatkowski, D.J.; Saldivar, J.S.; Squire, J.; 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 Pathology. J. Thorac. Oncol. 2013, 8, 823–859. [Google Scholar] [PubMed]
  13. Paez, J.G.; Jänne, P.A.; Lee, J.C.; Tracy, S.; Greulich, H.; Gabriel, S.; Herman, P.; Kaye, F.J.; Lindeman, N.; Boggon, T.J. EGFR mutations in lung cancer: Correlation with clinical response to gefitinib therapy. Science 2004, 304, 1497–1500. [Google Scholar] [CrossRef]
  14. Kwak, E.L.; Bang, Y.-J.; Camidge, D.R.; Shaw, A.T.; Solomon, B.; Maki, R.G.; Ou, S.-H.I.; Dezube, B.J.; Jänne, P.A.; Costa, D.B.; et al. Anaplastic Lymphoma Kinase Inhibition in Non–Small-Cell Lung Cancer. N. Engl. J. Med. 2010, 363, 1693–1703. [Google Scholar] [CrossRef] [PubMed]
  15. Bergethon, K.; Shaw, A.T.; Ou, S.-H.I.; Katayama, R.; Lovly, C.M.; McDonald, N.T.; Massion, P.P.; Siwak-Tapp, C.; Gonzalez, A.; Fang, R.; et al. ROS1 Rearrangements Define a Unique Molecular Class of Lung Cancers. J. Clin. Oncol. 2012, 30, 863–870. [Google Scholar] [CrossRef] [PubMed]
  16. Liu, Y.; Zhang, J.; Li, L.; Yin, G.; Zhang, J.; Zheng, S.; Cheung, H.; Wu, N.; Lu, N.; Mao, X.; et al. Genomic heterogeneity of multiple synchronous lung cancer. Nat. Commun. 2016, 7, 13200. [Google Scholar] [CrossRef]
  17. Zhao, L.; Liu, C.; Xie, G.; Wu, F.; Hu, C. Multiple Primary Lung Cancers: A New Challenge in the Era of Precision Medicine. Cancer Manag. Res. 2020, 12, 10361–10375. [Google Scholar] [CrossRef]
  18. Bai, Y.; Xu, Y.; Wang, X.; Liu, Y.; Sun, C.; Guo, Y.; Cai, Y.; Shao, G.; Yang, Z.; Qiu, S.; et al. Whole exome sequencing of lung adenocarcinoma and lung squamous cell carcinoma in one individual: A case report. Thorac. Cancer 2020, 11, 2361–2364. [Google Scholar] [CrossRef]
  19. Pei, G.; Li, M.; Min, X.; Liu, Q.; Li, D.; Yang, Y.; Wang, S.; Wang, X.; Wang, H.; Cheng, H.; et al. Molecular Identification and Genetic Characterization of Early-Stage Multiple Primary Lung Cancer by Large-Panel Next-Generation Sequencing Analysis. Front. Oncol. 2021, 11, 653988. [Google Scholar] [CrossRef]
  20. Song, Y.; Jia, Z.; Wu, P.; Wang, W.; Ou, Q.; Bao, H.; Yu, M.; Wu, X.; Liu, P.; Liang, N.; et al. Comprehensive genomic profiling aids in understanding the lesion origins of a patient with six synchronous invasive lung adenocarcinomas: A case study. BMC Pulm. Med. 2020, 20, 80. [Google Scholar] [CrossRef] [PubMed]
  21. Goodwin, D.; Rathi, V.; Conron, M.; Wright, G.M. Genomic and Clinical Significance of Multiple Primary Lung Cancers as Determined by Next-Generation Sequencing. J. Thorac. Oncol. 2021, 16, 1166–1175. [Google Scholar] [CrossRef]
  22. Wistuba, I.I.; Gazdar, A.F.; Minna, J.D. Molecular genetics of small cell lung carcinoma. Semin. Oncol. 2001, 28 (Suppl. S4), 3–13. [Google Scholar] [CrossRef]
  23. Menis, J.; Giaj Levra, M.; Novello, S. MET inhibition in lung cancer. Transl. Lung Cancer Res. 2013, 2, 23–39. [Google Scholar]
  24. De Mello, R.A.; Neves, N.M.; Amaral, G.A.; Lippo, E.G.; Castelo-Branco, P.; Pozza, D.H.; Tajima, C.C.; Antoniou, G. The Role of MET Inhibitor Therapies in the Treatment of Advanced Non-Small Cell Lung Cancer. J. Clin. Med. 2020, 9, 1918. [Google Scholar] [CrossRef]
  25. Shi, J.; Hua, X.; Zhu, B.; Ravichandran, S.; Wang, M.; Nguyen, C.; Brodie, S.A.; Palleschi, A.; Alloisio, M.; Pariscenti, G.; et al. Somatic Genomics and Clinical Features of Lung Adenocarcinoma: A Retrospective Study. PLoS Med. 2016, 13, e1002162. [Google Scholar] [CrossRef]
  26. Vignot, S.; Frampton, G.M.; Soria, J.C.; Yelensky, R.; Commo, F.; Brambilla, C.; Palmer, G.; Moro-Sibilot, D.; Ross, J.S.; Cronin, M.T.; et al. Next-Generation Sequencing Reveals High Concordance of Recurrent Somatic Alterations Between Primary Tumor and Metastases From Patients With Non–Small-Cell Lung Cancer. JCO 2013, 31, 2167–2172. [Google Scholar] [CrossRef]
  27. Oser, M.G.; Niederst, M.J.; Sequist, L.V.; Engelman, J.A. Transformation from non-small-cell lung cancer to small-cell lung cancer: Molecular drivers and cells of origin. Lancet Oncol. 2015, 16, e165–e172. [Google Scholar]
  28. Adelstein, D.J.; Tomashefski, J.F.; Snow, N.J.; Horrigan, T.P.; Hines, J.D. Mixed small cell and non-small cell lung cancer. Chest 1986, 89, 699–704. [Google Scholar] [CrossRef] [PubMed]
  29. Zhang, J.; Wang, H.; Li, C.; Qian, H. Chance to rein in a cancer--Spontaneous regression of lung carcinoma (1988–2018): A 30-year perspective. Int. J. Clin. Exp. Pathol. 2020, 13, 1190–1196. [Google Scholar] [PubMed]
  30. Everson, T.C.; Cole, W.H. Spontaneous Regression of Cancer: Preliminary Report. Ann. Surg. 1956, 144, 366–380. [Google Scholar] [CrossRef] [PubMed]
  31. Kumar, T.; Patel, N.; Talwar, A. Spontaneous regression of thoracic malignancies. Respir. Med. 2010, 104, 1543–1550. [Google Scholar] [CrossRef]
  32. Cafferata, M.A.; Chiaramondia, M.; Monetti, F.; Ardizzoni, A. Complete spontaneous remission of non-small-cell lung cancer: A case report. Lung Cancer 2004, 45, 263–266. [Google Scholar] [CrossRef]
  33. Hong, G.; Chung, C.; Park, D.; Lee, S.I.; Lee, J.E.; Kang, D.H. Spontaneous regression of recurrent pulmonary large cell neuroendocrine carcinoma with alteration of PD-L1 expression after surgical resection: A case report. Thorac. Cancer 2024, 15, 266–270. [Google Scholar] [CrossRef]
  34. Goto, M.; Fukumoto, K.; Ichikawa, Y.; Tsubouchi, H.; Uchiyama, M.; Mori, S. Pathologically confirmed spontaneous regression of small cell lung cancer after computed tomography-guided percutaneous transthoracic needle biopsy followed by surgery. Surg. Case Rep. 2023, 9, 187. [Google Scholar] [CrossRef] [PubMed]
  35. Otani, M.; Nishimori, M.; Iwasa, H.; Iwamura, M.; Izumi, T.; Nakaji, K.; Nitta, N.; Miyatake, K.; Yoshimatsu, R.; Yamanishi, T.; et al. Spontaneous regression of small cell lung cancer associated with Lambert-Eaton Myasthenic Syndrome: Case report. Radiol. Case Rep. 2023, 18, 4036–4041. [Google Scholar] [CrossRef] [PubMed]
  36. Thomas, V.; Cotter, M.B.; Tosetto, M.; Khaw, Y.L.; Geraghty, R.; Winter, D.C.; Ryan, E.J.; Sheahan, K.; Furney, S.J. Personalised mapping of tumour development in synchronous colorectal cancer patients. NPJ Genom. Med. 2020, 5, 27. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Timeline illustrating the clinical history of the patient diagnosed with multiple primary lung cancers and a potentially metastatic lesion in the mandible.
Figure 1. Timeline illustrating the clinical history of the patient diagnosed with multiple primary lung cancers and a potentially metastatic lesion in the mandible.
Jpm 14 00257 g001
Figure 2. Mutational burdens, mutational overlap, putative drivers, and mutational signatures of the tumours diagnosed in the patient. (A) Mutational burdens of SNVs, InDels, and SVs quantified by WGS; (B) Venn diagrams of SNVs (left), InDels (centre), and SVs (right) show the overlap of mutations between samples; (C) VAFs of putative driver mutations and clinically relevant alterations; (D) tumour mutational profiles of total SNVs, defined by the weighted contributions of each input reference signature from COSMIC and identified by WGS. Signatures found in the samples include a “Smoking” signature (SBS4), “HR-deficiency” signature (SBS3), “Damage by Reactive Oxygen Species” signature (SBS18), and “Other” signatures (SBS12, SBS33, SBS37, SBS39, SBS40, SBS46).
Figure 2. Mutational burdens, mutational overlap, putative drivers, and mutational signatures of the tumours diagnosed in the patient. (A) Mutational burdens of SNVs, InDels, and SVs quantified by WGS; (B) Venn diagrams of SNVs (left), InDels (centre), and SVs (right) show the overlap of mutations between samples; (C) VAFs of putative driver mutations and clinically relevant alterations; (D) tumour mutational profiles of total SNVs, defined by the weighted contributions of each input reference signature from COSMIC and identified by WGS. Signatures found in the samples include a “Smoking” signature (SBS4), “HR-deficiency” signature (SBS3), “Damage by Reactive Oxygen Species” signature (SBS18), and “Other” signatures (SBS12, SBS33, SBS37, SBS39, SBS40, SBS46).
Jpm 14 00257 g002
Figure 3. Copy number alteration analysis of the tumours diagnosed in the patient. (A) The genome landscapes show CNAs estimated by WGS. The x-axis coordinates represent positions along the genome, with vertical bars indicating the borders between chromosomes. The y-axis represents the chromosomal copy number log ratio in the tumour. Amplifications are depicted in red, and deletions in blue. Estimates of tumour ploidy are as follows: 2.2 for NSC-1, 3.3 for SC, 2.1 for NSC-2, and 3.2 for NSC-3. Estimates of tumour purity are as follows: 0.3 for NSC-1, NSC-2, and NSC-3, and 0.6 for SC; (B) Median log ratios of putative driver CNAs. Positive log ratios (gains) are represented in red, and negative log ratios (losses) are represented in blue.
Figure 3. Copy number alteration analysis of the tumours diagnosed in the patient. (A) The genome landscapes show CNAs estimated by WGS. The x-axis coordinates represent positions along the genome, with vertical bars indicating the borders between chromosomes. The y-axis represents the chromosomal copy number log ratio in the tumour. Amplifications are depicted in red, and deletions in blue. Estimates of tumour ploidy are as follows: 2.2 for NSC-1, 3.3 for SC, 2.1 for NSC-2, and 3.2 for NSC-3. Estimates of tumour purity are as follows: 0.3 for NSC-1, NSC-2, and NSC-3, and 0.6 for SC; (B) Median log ratios of putative driver CNAs. Positive log ratios (gains) are represented in red, and negative log ratios (losses) are represented in blue.
Jpm 14 00257 g003
Table 1. Clinicopathologic data of multiple primary tumours from a lung cancer patient.
Table 1. Clinicopathologic data of multiple primary tumours from a lung cancer patient.
SampleTumour
Subtype
Tumour
Location
Tumour StageDiagnosis and BiopsySequencing MethodTreatment
NSC-1NSCLCaRight lower lobepT1aN02015WGSSurgical removal
SCSCLCLeft upper lobecT1cN02015WGSNone
NSC-2NSCLCaLeft upper lobecT1cNx2018WGSPembrolizumab
NSC-3NSCLCaRight mandiblecT1cNxM1b2018WGSPembrolizumab
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

Thomas, V.; Rashed, A.; Faul, C.; Nicholson, S.; Young, V.; Hanson, J.; Hennessy, B.T.; Toomey, S.; Furney, S.J. Genome Sequencing of Multiple Primary Lung Cancers Harbouring Mixed Histology and Spontaneously Regressing Small-Cell Lung Cancer. J. Pers. Med. 2024, 14, 257. https://doi.org/10.3390/jpm14030257

AMA Style

Thomas V, Rashed A, Faul C, Nicholson S, Young V, Hanson J, Hennessy BT, Toomey S, Furney SJ. Genome Sequencing of Multiple Primary Lung Cancers Harbouring Mixed Histology and Spontaneously Regressing Small-Cell Lung Cancer. Journal of Personalized Medicine. 2024; 14(3):257. https://doi.org/10.3390/jpm14030257

Chicago/Turabian Style

Thomas, Valentina, Ahmed Rashed, Clare Faul, Siobhan Nicholson, Vincent Young, John Hanson, Bryan T. Hennessy, Sinead Toomey, and Simon J. Furney. 2024. "Genome Sequencing of Multiple Primary Lung Cancers Harbouring Mixed Histology and Spontaneously Regressing Small-Cell Lung Cancer" Journal of Personalized Medicine 14, no. 3: 257. https://doi.org/10.3390/jpm14030257

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