Genomic alterations in small cell lung cancer and their clinical relevance
Correspondence

Genomic alterations in small cell lung cancer and their clinical relevance

Gary Wildey, Afshin Dowlati

Department of Hematology & Oncology, University Hospitals Case Medical Center, Case Western Reserve University, Cleveland, Ohio, USA

Correspondence to: Afshin Dowlati, MD. Department of Hematology & Oncology, University Hospitals Case Medical Center, 11100 Euclid Avenue, Cleveland, Ohio 44016, USA. Email: axd44@case.edu.

Submitted Jun 27, 2016. Accepted for publication Jul 04, 2016.

doi: 10.21037/tlcr.2016.07.05


We thank Karachaliou et al. for their insightful editorial (1) regarding genomic studies on small cell lung cancer (SCLC), which included analysis of our own recent study (2). They raise several interesting points, such as the potential use of ARID1A mutations and alterations in the PIK3-AKT-mTOR pathway as biomarkers for response to PARP inhibition, that support the idea that real therapeutic progress against this cancer can be gained from genomic studies. The authors specifically pointed out two of our results, however, which they found inconsistent with previous genomic studies of SCLC. The first was our finding that only 58% of our patient cohort carried exome mutations in RB1. Our RB1 results are within the range, however, of several earlier exome studies, which ranged from 39–74% (3-7). This wide range of results is likely caused by the small cohort sizes studied, as well as the divergent demographic populations of the cohorts. The most recent analysis of our cohort (N=67) yielded a 76% mutation rate in RB1. As Karachaliou et al. correctly point out, a recent whole genome study of SCLC (8) indicates that the RB1 gene may be altered in >90% of all SCLC tumors, approaching the mutation rate of TP53 in this cancer. Whatever the true frequency is, we still believe that a small subset of SCLC tumors express RB1 protein, as evidenced by our results in Figure 3, and that this may affect clinical outcome. Even if ≤10% of all SCLC tumors harbored wild type RB1, this would still represent a significant population in SCLC because the vast majority of genes mutated in SCLC occur at a similar low frequency. We are currently investigating the question of whether or not the RB1 pathway is functional when RB1 protein is expressed in SCLC. Thus, we remain open to the idea that other genes, besides RB1, can participate with TP53 loss in a ‘two-hit’ paradigm for SCLC. While it is true that mouse models of SCLC can be produced by double knockout of TP53/RB1, we do not feel this eliminates other possible gene combinations. Interestingly, there is a long latency in tumor formation in this mouse model of SCLC (6–9 mo) (9,10), suggesting that additional oncogenic events may take place during tumorigenesis. Concerning MYC amplifications, our low rate of detection may simply imply that MYC amplification is more commonly seen in relapsed SCLC, after failure of platinum based therapy. Our patient population was mainly comprised of chemo-naïve patients. The challenge before us is to acquire much larger cohorts of SCLC patients for genomic studies so that we can improve our understanding of the clinical significance of gene mutations in this cancer.


Acknowledgements

None.


Footnote

Provenance: This is a Guest Correspondence commissioned by Section Editor Hongbing Liu, MD, PhD (Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China).

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

Response to: Karachaliou N, Sosa A, Rosell R. Unraveling the genomic complexity of small cell lung cancer. Transl Lung Cancer Res 2016;5:363-6.


References

  1. Karachaliou N, Sosa A, Rosell R. Unraveling the genomic complexity of small cell lung cancer. Transl Lung Cancer Res 2016;5:363-6.
  2. Dowlati A, Lipka MB, McColl K, et al. Clinical correlation of extensive-stage small-cell lung cancer genomics. Ann Oncol 2016;27:642-7. [Crossref] [PubMed]
  3. Rudin CM, Durinck S, Stawiski EW, et al. Comprehensive genomic analysis identifies SOX2 as a frequently amplified gene in small-cell lung cancer. Nat Genet 2012;44:1111-6. [Crossref] [PubMed]
  4. Umemura S, Mimaki S, Makinoshima H, et al. Therapeutic priority of the PI3K/AKT/mTOR pathway in small cell lung cancers as revealed by a comprehensive genomic analysis. J Thorac Oncol 2014;9:1324-31. [Crossref] [PubMed]
  5. Ross JS, Wang K, Elkadi OR, et al. Next-generation sequencing reveals frequent consistent genomic alterations in small cell undifferentiated lung cancer. J Clin Pathol 2014;67:772-6. [Crossref] [PubMed]
  6. Peifer M, Fernández-Cuesta L, Sos ML, et al. Integrative genome analyses identify key somatic driver mutations of small-cell lung cancer. Nat Genet 2012;44:1104-10. [Crossref] [PubMed]
  7. Iwakawa R, Kohno T, Totoki Y, et al. Expression and clinical significance of genes frequently mutated in small cell lung cancers defined by whole exome/RNA sequencing. Carcinogenesis 2015;36:616-21. [Crossref] [PubMed]
  8. George J, Lim JS, Jang SJ, et al. Comprehensive genomic profiles of small cell lung cancer. Nature 2015;524:47-53. [Crossref] [PubMed]
  9. Meuwissen R, Linn SC, Linnoila RI, et al. Induction of small cell lung cancer by somatic inactivation of both Trp53 and Rb1 in a conditional mouse model. Cancer Cell 2003;4:181-9. [Crossref] [PubMed]
  10. Schaffer BE, Park KS, Yiu G, et al. Loss of p130 accelerates tumor development in a mouse model for human small-cell lung carcinoma. Cancer Res 2010;70:3877-83. [Crossref] [PubMed]
Cite this article as: Wildey G, Dowlati A. Genomic alterations in small cell lung cancer and their clinical relevance. Transl Lung Cancer Res 2016;5(4):450-451. doi: 10.21037/tlcr.2016.07.05

Download Citation