Advertisement
Brief Report| Volume 15, ISSUE 7, P1232-1239, July 2020

High Prevalence of Somatic Oncogenic Driver Alterations in Patients With NSCLC and Li-Fraumeni Syndrome

Open ArchivePublished:March 13, 2020DOI:https://doi.org/10.1016/j.jtho.2020.03.005

      Abstract

      Introduction

      Actionable somatic molecular alterations are found in 15% to 20% of NSCLC in Europe. NSCLC is a tumor observed in patients with germline TP53 variants causing Li-Fraumeni syndrome (LFS), but its somatic molecular profile is unknown.

      Methods

      Retrospective study of clinical and molecular profiles of patients with NSCLC and germline TP53 variants.

      Results

      Among 22 patients with NSCLC and LFS (n = 23 lung tumors), 64% were women, median age was 51 years, 84% were nonsmokers, 73% had adenocarcinoma histological subtype, and 84% were diagnosed with advanced-stage disease. These patients harbored 16 distinct germline TP53 variants; the most common was p.R158H (5/22; three in the same family). Personal and family histories of cancer were reported in 71% and 90% of patients, respectively. In most cases (87%, 13/15), lung cancer was diagnosed with a late onset. Of the 21 tumors analyzed, somatic oncogenic driver mutations were found in 19 of 21 (90%), EGFR mutations in 18 (exon 19 deletion in 12 cases, L858R in three cases, and G719A, exon 20 insertion, and missing mutation subtype, each with one case), and ROS1 fusion in one case. A PI3KCA mutation was concurrently detected at diagnosis in three EGFR exon 19-deleted tumors (3/12). The median overall survival was 37.3 months in 14 patients treated with EGFR inhibitors; seven developed resistance, five (71%) acquired EGFR-T790M mutation, and one had SCLC transformation.

      Conclusions

      Driver oncogenic alterations were observed in 90% of the LFS tumors, mainly EGFR mutations; one ROS1 fusion was also observed. The germline TP53 variants and lung cancer carcinogenesis driven by oncogenic processes need further evaluation.

      Keywords

      Introduction

      Li-Fraumeni syndrome (LFS), resulting from germline pathogenic TP53 variants, is a rare autosomal dominant inheritance condition characterized by the early onset of different cancer types, including breast cancer, bone and soft tissue sarcomas, central nervous system tumors, and adrenocortical carcinoma.
      • Li F.P.
      • Fraumeni Jr., J.F.
      • Mulvihill J.J.
      • et al.
      A cancer family syndrome in 24 kindreds.
      • Bougeard G.
      • Renaux-Petel M.
      • Flaman J.-M.
      • et al.
      Revisiting Li-Fraumeni syndrome from TP53 mutation carriers.
      • McBride K.A.
      • Ballinger M.L.
      • Killick E.
      • et al.
      Li-Fraumeni syndrome: cancer risk assessment and clinical management.
      Patients with germline disease-causing TP53 variants have an increased risk of developing lung cancer,
      • McBride K.A.
      • Ballinger M.L.
      • Killick E.
      • et al.
      Li-Fraumeni syndrome: cancer risk assessment and clinical management.
      which is currently considered the third most frequent tumor in adults with LFS. In a cohort of 552 patients with LFS, 17 who were diagnosed with lung cancer had a median age of 45 years.
      • Bougeard G.
      • Renaux-Petel M.
      • Flaman J.-M.
      • et al.
      Revisiting Li-Fraumeni syndrome from TP53 mutation carriers.
      Caron et al. reported that lung adenocarcinoma was found in five of 107 germline TP53 variant carriers screened with whole-body magnetic resonance imaging.
      • Caron O.
      • Frebourg T.
      • Benusiglio P.R.
      • Foulon S.
      • Brugières L.
      Lung adenocarcinoma as part of the Li-Fraumeni syndrome spectrum: preliminary data of the LIFSCREEN randomized clinical trial.
      Although previous articles have reported individuals with LFS and lung cancer harboring EGFR-sensitizing mutations (Supplementary Table 1), the clinical phenotype has not been well characterized, and the molecular profile of LFS lung tumors remains unknown.
      The aim of this study was to describe the clinical presentation of 22 patients with LFS and lung cancer and the molecular phenotype of their tumors.

      Materials and Methods

      Patients with histologically confirmed NSCLC and pathogenic germline TP53 variants were included. Samples were collected from May 2002 to November 2017 in two academic centers (Gustave Roussy, France; Catalan Institute of Oncology, Spain). Clinical and molecular data were retrospectively extracted from electronic medical records.
      Germline TP53 screening, interpretation of TP53 variants, and the somatic molecular profile of NSCLC were determined according to standard procedures (Supplementary Material). The institutional review boards of both centers approved this study.

      Results

      Baseline characteristics of the 22 patients and germline variants are summarized in Table 1 and Table 2, respectively. Overall, the median age was 51 years, 84% were nonsmokers, 64% were women, 73% had adenocarcinoma histology, and 76% were diagnosed with an advanced stage.
      Table 1Baseline Characteristics of the Study Population
      Baseline CharacteristicsOverall Population N = 22
      One patient presented two synchronous lung adenocarcinoma (no. 10; no. 11) with different pathologic and molecular characteristics and stages at diagnosis.
      (%)
      Age (at lung cancer diagnosis)
       Median, range51 y, (21–77)
      Sex
       Female14 (64%)
       Male8 (36%)
      Smoking
       Nonsmoker16 (84%)
       Light smoker
      One case with one pack-year.
      1 (5%)
       Smoker (former)2 (11%)
       Missing3
      Histology
       Adenocarcinoma16 (73%)
       NSCLC, other6 (27%)
      Stage at diagnosis
       I–II5 (24%)
       IVA4 (19%)
       IVB12 (57%)
       Missing1
      Additional cancer personal history
       Yes15 (71%)
       No7 (33%)
       Missing1
      Family cancer history
       Yes18 (90%)
       No2 (10%)
       Missing2
      Previous radiotherapy
       Yes6 (27%)
       No16 (73%)
      Molecular assessment
       NGS12 (55%)
       RT-PCR2 (9%)
       Not specified6 (27%)
       Not performed2 (9%)
      NGS, next-generation sequencing; RT-PCR, reverse transcription polymerase chain reaction.
      a One patient presented two synchronous lung adenocarcinoma (no. 10; no. 11) with different pathologic and molecular characteristics and stages at diagnosis.
      b One case with one pack-year.
      Table 2Clinical, Pathologic, and Molecular Characterization of Patients With Lung Cancer and Germline TP53 Pathogenic Variant (n = 22 Tumors, 21 Patients)
      No. 1
      Three patients with NSCLC within the same family.
      No. 2
      Three patients with NSCLC within the same family.
      No. 3
      Three patients with NSCLC within the same family.
      No. 4No. 5No. 6No. 7No. 8No. 9No.10
      Two different lung tumors from the same patient.
      No.11
      Two different lung tumors from the same patient.
      No. 12No. 13No. 14No. 15No. 16No. 17No. 18No. 19No. 20No. 21No. 22No. 22
      TP53 germline mutation
      p.R158Hp.R158Hp.R158Hp.R158Hp.R158Hp.R337Hp.R248Wp.R248Wp.R175Hp.R196∗p.R196∗p.R267Gp.C275Sp.H214∗c.783-1G>A p.?p.R282Wp.C242Yp.R196∗p.H179Yp.R273Hp.L257Kc.559+1G>A p.?p.C277F
      Personal cancer history
      Additional personal cancer history.
       Breast cancer++unk+++++++
       Sarcomaunk+++
       Brain tumorunk++
       Thyroid carcinoma+unk
       H&N tumorunk++
       Nonmelanoma skinunk+++
       Melanoma skinunk+
       Esophageal cancerunk+
       Desmoid tumorunk+
       Fibromaunk+
      Family cancer
       Lung cancer+++++unk+unk
       Breast cancer++++++++unk++unk
       Sarcoma++unk++unk
       Leukemia+++unk+unk
       Lymphoma++unk+unk
       Adrenal carcinoma++unk++unk
       Brain tumor+unk+unk
       Thyroid carcinoma++unkunk
       Prostate cancer+++unk+unk
       Colorectal cancer+++unkunk
       Gastric cancer+unk+unk
       Hepatic cancer+unk+unk
       Ovarian cancer+unkunk
       Cervix carcinoma+++unkunk
       Thymoma+unkunk
       Germline tumor+unkunk
       Meduloblastomaunk+unk
       Neuroblastomaunk+unk
       Unknown primary cancer++unkunk
       Nonmelanoma skinunkunk+
      Lung cancer diagnosis
       SexFFFFFMFFMMMMFMMFFMMFFFF
       SmokingNonNonNonNonNonunkNonNonLightNonNonNonLightunkNonNonNonNonNonSmokerNonunkNon
       Age at diagnosis3854455353773763544848405465513729315557364343
       HistologyAdenoOtherAdenoAdenoAdenounkAdenoAdenoAdenoAdenoAdenoAdenoAdenoNSCLCAdenoAdenoAdenoAdenoAdenoAdenoNSCLCAdenoAdeno
       StageIVBIVBIVBIVBIVBIVBIAIVBIBI-IIIVAIBIVBunkIVAIVBIVAIVBIVBIVBI-IIIAIVA
      Molecular profile
       Driver oncogene alterationEGFR ex 19EGFR ex 21EGFR ex 19EGFR ex 19WTunkEGFR ex 19WTEGFR, ex unkEGFR, ex 20EGFR ex 21EGFR ex 19EGFR ex 19EGFR ex 19EGFR ex 19EGFR ex 18EGFR ex 19EGFR ex 19EGFR ex 19EGFR ex 21unkEGFR ex 19ROS1
       Other (NGS panel)unkunkunkNonNonunkNonunkunkNonNonPI3KCAPI3KCAunkPI3KCANonunkNonunkunkunkNonNon
      Plus symbol indicates that one specific patient has previous and/or family history of one specific tumor type.
      unk, unknown; ex, exon; WT, wildtype; NGS, next-generation sequencing; H&N, head and neck squamous cell carcinoma.
      a Three patients with NSCLC within the same family.
      b Two different lung tumors from the same patient.
      c Additional personal cancer history.
      The most common germline TP53 variant was p.R158H (5/22 patients), including three cases in the same family (Supplementary Fig. 1). The p.R248W variant was observed in two unrelated patients, and the p.R196∗ variant was found in one patient with two synchronous lung tumors.
      Previous personal and family cancer histories were reported in 71% and 90% of the patients, respectively (Table 1). Breast cancer was the most common cancer type, affecting 67% of women (14/22). Breast (41%), lung (27%), and adrenal (18%) were the most common tumors diagnosed in the families (Table 2) (Supplementary Table 2).
      Among patients who had previously developed another cancer, lung cancer was often the second or later event (13/15; 87%) (Fig. 1). The median time that elapsed between the first cancer and the subsequent lung cancer diagnosis was 7 years (range, −3 to 23 y). The most common first tumors identified were breast cancer (9/10) and mesenchymal tumors (3/5) in women and men, respectively. All four patients with breast cancer had HER2 amplification as per available analyses.
      Figure thumbnail gr1
      Figure 1Timeline of the personal cancer history of germline TP53-mutant cases with lung cancer and at least one additional cancer diagnosis. ∗Carcinoma in situ; Esophag, esophageal cancer; Sarc, sarcoma; Melan, melanoma; GBM, multiforme glioblastoma; H&N, head and neck squamous cell carcinoma.
      Molecular screening was performed for 21 tumors, and next-generation sequencing target panel for lung cancer was performed for 12 tumors (Supplementary Table 3). Somatic oncogenic driver alterations were detected in 90% of the tumors (19/21), with 18 cases of somatic EGFR mutation and one case of ROS1 fusion (Supplementary Fig. 2). Two tumors did not harbor any EGFR mutation or other molecular alterations (in one, only EGFR-sensitizing mutations were sequenced), but the fusions were not tested. PI3KCA mutations were concomitantly detected in three EGFR exon 19-deleted tumors (3/11). Three patients belonged to the same family, with two of them harboring an EGFR exon 19 deletion and one an EGFR exon 21 mutation (#appsec1).
      Among patients with EGFR-mutant tumors, 14 received an EGFR tyrosine kinase inhibitor (TKI) and 11 were evaluable for survival. Median progression-free survival was 25.9 months (95% confidence interval [CI]: 8.4‒not reached [NR]). With a median follow-up of 56.6 months (95% CI: 31.5–NR), seven patients had disease progression. The mechanisms of acquired resistance are displayed in Supplementary Figure 3. Five tumors developed EGFR-T790M resistance mutation (71%), and one case had SCLC transformation (14%). The emergence of the EGFR-T790M mutation on circulating tumor DNA in case no. 13 is represented in Supplementary Figure 4. The patient detected with the ROS1 fusion received ROS1 TKI (crizotinib) as a second-line therapy, after progression to platinum-based chemotherapy, with partial response as best response; at this moment, the patient is still under crizotinib after 18 months.
      The median overall survival was 46.3 months (95% CI: 35.0–NR) in the overall population, and 31.9 months (95% CI: 24.4–NR) in the metastatic EGFR-mutant population (Supplementary Fig. 5A and B).

      Discussion

      Here, we present the largest series of patients with LFS and lung cancer in which we identified a high rate of targetable oncogenic drivers, most of them EGFR mutations. This is consistent with previously published case reports (Supplementary Table 1). In addition, we report, for the first time, the occurrence of ROS1 fusion in a patient with NSCLC and LFS.
      Barbosa et al.
      • Barbosa M.V.R.
      • Cordeiro de Lima V.C.
      • Formiga M.N.
      • Andrade de Paula C.A.
      • Torrezan G.T.
      • Carraro D.M.
      High prevalence of EGFR mutations in lung adenocarcinomas from Brazilian patients harboring the TP53 p.R337H variant.
      have reported, very recently, a case series of nine patients with lung cancer harboring the known founder germline TP53 p.R337H variant (Supplementary Table 1). Of these nine cases, eight had an EGFR mutation and one had a KRAS mutation (EGFR wildtype, KRAS subtype not described); however, no next-generation sequencing was performed for testing other somatic mutations or fusions. In our cohort, we described one case harboring the germline TP53 p.R337H variant, but no tissue was available for molecular testing.
      In never smokers, who are more susceptible to lung cancer with somatic oncogene driver alterations, the reported distribution in Europe of the altered genes is 44% for EGFR, 14% for ALK, and 3% for ROS1.
      • Barlesi F.
      • Mazieres J.
      • Merlio J.-P.
      • et al.
      Routine molecular profiling of patients with advanced non-small-cell lung cancer: results of a 1-year nationwide programme of the French Cooperative Thoracic Intergroup (IFCT).
      ,
      • Gou L.Y.
      • Niu F.Y.
      • Wu Y.L.
      • Zhong W.Z.
      Differences in driver genes between smoking-related and non-smoking-related lung cancer in the Chinese population.
      In contrast, 90% were EGFR-mutant and 5% ROS1-positive in our LFS series. It has been described that breast tumors associated with LFS are enriched in HER2-positive subtype, which is in line with our data.
      • Gallardo-Alvarado L.N.
      • Tusié-Luna M.T.
      • Tussié-Luna M.I.
      • et al.
      Prevalence of germline mutations in the TP53 gene in patients with early-onset breast cancer in the Mexican population.
      These observations suggest that the development of tumors in TP53 variant carriers requires somatic driver alterations, with TP53 variants acting not as oncogenic but as permissive events. This oncogenic addiction might be specific of an EGFR and HER2 pathway belonging to the HER family. In this sense, enhanced chromosomal instability has been previously related to EGFR/HER2-driven carcinogenesis in the context of TP53 mutations.
      • Liu S.
      • Liu W.
      • Jakubczak J.L.
      • et al.
      Genetic instability favoring transversions associated with ErbB2-induced mammary tumorigenesis.
      ,
      • Ma Z.
      • Kim Y.M.
      • Howard E.W.
      • et al.
      DMBA promotes ErbB2–mediated carcinogenesis via ErbB2 and estrogen receptor pathway activation and genomic instability.
      Other epithelial tumors related to LFS should be investigated to check the activation of other HER family protein kinases. Some authors have presented the hypothesis that the oncogenic potential of certain genomic drivers may also be context-dependent, and thus potentially organ-dependent, with early compensatory mechanisms that could moderate the oncogenic potential of the alteration.
      • Kato S.
      • Lippman S.M.
      • Flaherty K.T.
      • Kurzrock R.
      The conundrum of genetic “drivers” in benign conditions.
      In NSCLC with germline TP53 variant and somatic EGFR mutation, a important benefit from EGFR TKI was observed, comparable to the benefit reported in the general population.
      • Soria J.C.
      • Ohe Y.
      • Vansteenkiste J.
      • et al.
      Osimertinib in untreated EGFR-mutated advanced non-small-cell lung cancer.
      This contrasts with the poorer prognosis reported when somatic TP53 inactivation is found concomitantly with EGFR mutations.
      • Canale M.
      • Petracci E.
      • Delmonte A.
      • et al.
      Impact of TP53 mutations on outcome in EGFR-mutated patients treated with first-line tyrosine kinase inhibitors.
      As somatic TP53 alterations can be either a clonal or subclonal event,
      • Jamal-Hanjani M.
      • Wilson G.A.
      • McGranahan N.
      • et al.
      Tracking the evolution of non–small-cell lung cancer.
      subclonal TP53 mutations may reflect higher tumor heterogeneity and increased tumor mutation burden at diagnosis reducing the prognosis.
      • Rizvi N.A.
      • Hellmann M.D.
      • Snyder A.
      • et al.
      Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer.
      In our NSCLC case with germline TP53 variant and somatic ROS1 fusion, we observed similar efficacy to crizotinib, as reported in the ROS1-positive population with unknown LFS status.
      • Shaw A.T.
      • Ou S.H.
      • Bang Y.J.
      • et al.
      Crizotinib in ROS1-rearranged non-small-cell lung cancer.
      More preclinical studies are needed to study the link between germline and somatic driver alterations in the carcinogenesis processes in lung cancer.
      To date, no lung cancer risk factors have been identified in patients with LFS. Nevertheless, patients with LFS have a higher risk of developing cancers related to radiation exposure, such as radon gas, considered by the WHO as the second cause of lung cancer and the first in never smokers. Therefore, a study of environmental factors combined with germline predisposition could help to better assess cancer risk and the molecular profile associated in these patients. Although data are limited, it is expected that smoking in patients with LFS might increase the risk of lung cancer even more when compared with those with NSCLC who do not harbor germline TP53 mutations.
      Given that metastatic lung cancer conveys high mortality rates, optimizing screening techniques is required to detect lung cancers at earlier stages. The potential predisposition to somatic driver mutations in individuals with LFS could lead to integration of molecular tools in the follow-up, such as circulating tumor DNA, to detect cancer early.
      Our study has several limitations including sample size, lack of a systematic comprehensive tumoral somatic molecular screening, particularly for fusion testing, and incomplete clinical-familial data from old cases. Despite these limitations, we observed a clinically relevant association between LFS and driver oncogene mutations in lung cancer, which warrants further studies.
      In conclusion, we report a high prevalence of somatic driver oncogenic alterations, mainly EGFR mutations but also the first case of ROS1 fusion associated with LFS, in patients with lung cancers and germline TP53 variants in the largest series of LFS lung cancer reported to date. Our data support the concept of oncogene addiction in TP53 variant carriers and should prompt extensive screening for oncogenic alterations in other LFS tumors.

      Acknowledgments

      Information Commissioner’s Office grant support: Supported by the Carlos III National Health Institute funded by the Federación Española de Enfermedades Raras (FEDER) funds—a way to build Europe—(PI19/00553; PI16/00563; PI16/01898; and Centros de Investigación Biomédica en Red Cáncer) and the Government of Catalonia (2017SGR448; 2017SGR1282; and 2017SGR496). Ernest Nadal received support from the SLT006/17/00127 grant, funded by the Department of Health of the Generalitat de Catalunya by the call “Acció instrumental d’intensificació de professionals de la salut.” This study has been funded by the Sociedad Española de Oncología Médica through the “Proyectos de Investigación para Grupo Emergente.” We thank the Centers de Recerca de Catalunya Program, Generalitat de Catalunya for their institutional support. Dr. Maria Jové is supported by a Rio Hortega contract (CM17/00008) from the Carlos III Institute. Gustave Roussy: Dr. Laura Mezquita received support from the 2018 International Association for the Study of Lung Cancer Research Fellowship Award, European Society for Medical Oncology Translational Research Fellowship 2019, and Sociedad Española de Oncología Médica Retorno de Investigadores 2019.

      Supplementary Data

      References

        • Li F.P.
        • Fraumeni Jr., J.F.
        • Mulvihill J.J.
        • et al.
        A cancer family syndrome in 24 kindreds.
        Cancer Res. 1988; 48: 5358-5362
        • Bougeard G.
        • Renaux-Petel M.
        • Flaman J.-M.
        • et al.
        Revisiting Li-Fraumeni syndrome from TP53 mutation carriers.
        J Clin Oncol. 2015; 33: 2345-2352
        • McBride K.A.
        • Ballinger M.L.
        • Killick E.
        • et al.
        Li-Fraumeni syndrome: cancer risk assessment and clinical management.
        Nat Rev Clin Oncol. 2014; 11: 260-271
        • Caron O.
        • Frebourg T.
        • Benusiglio P.R.
        • Foulon S.
        • Brugières L.
        Lung adenocarcinoma as part of the Li-Fraumeni syndrome spectrum: preliminary data of the LIFSCREEN randomized clinical trial.
        JAMA Oncol. 2017; 3: 1736-1737
        • Barbosa M.V.R.
        • Cordeiro de Lima V.C.
        • Formiga M.N.
        • Andrade de Paula C.A.
        • Torrezan G.T.
        • Carraro D.M.
        High prevalence of EGFR mutations in lung adenocarcinomas from Brazilian patients harboring the TP53 p.R337H variant.
        Clin Lung Cancer. 2020; 21: e37-e44
        • Barlesi F.
        • Mazieres J.
        • Merlio J.-P.
        • et al.
        Routine molecular profiling of patients with advanced non-small-cell lung cancer: results of a 1-year nationwide programme of the French Cooperative Thoracic Intergroup (IFCT).
        Lancet. 2016; 387: 1415-1426
        • Gou L.Y.
        • Niu F.Y.
        • Wu Y.L.
        • Zhong W.Z.
        Differences in driver genes between smoking-related and non-smoking-related lung cancer in the Chinese population.
        Cancer. 2015; 121: 3069-3079
        • Gallardo-Alvarado L.N.
        • Tusié-Luna M.T.
        • Tussié-Luna M.I.
        • et al.
        Prevalence of germline mutations in the TP53 gene in patients with early-onset breast cancer in the Mexican population.
        BMC Cancer. 2019; 19: 118
        • Liu S.
        • Liu W.
        • Jakubczak J.L.
        • et al.
        Genetic instability favoring transversions associated with ErbB2-induced mammary tumorigenesis.
        Proc Natl Acad Sci U S A. 2002; 99: 3770-3775
        • Ma Z.
        • Kim Y.M.
        • Howard E.W.
        • et al.
        DMBA promotes ErbB2–mediated carcinogenesis via ErbB2 and estrogen receptor pathway activation and genomic instability.
        Oncol Rep. 2018; 40: 1632-1640
        • Kato S.
        • Lippman S.M.
        • Flaherty K.T.
        • Kurzrock R.
        The conundrum of genetic “drivers” in benign conditions.
        J Natl Cancer Inst. 2016; 108: djw036
        • Soria J.C.
        • Ohe Y.
        • Vansteenkiste J.
        • et al.
        Osimertinib in untreated EGFR-mutated advanced non-small-cell lung cancer.
        N Engl J Med. 2018; 378: 113-125
        • Canale M.
        • Petracci E.
        • Delmonte A.
        • et al.
        Impact of TP53 mutations on outcome in EGFR-mutated patients treated with first-line tyrosine kinase inhibitors.
        Clin Cancer Res. 2017; 23: 2195-2202
        • Jamal-Hanjani M.
        • Wilson G.A.
        • McGranahan N.
        • et al.
        Tracking the evolution of non–small-cell lung cancer.
        N Eng J Med. 2017; 376: 2109-2121
        • Rizvi N.A.
        • Hellmann M.D.
        • Snyder A.
        • et al.
        Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer.
        Science. 2015; 348: 124-128
        • Shaw A.T.
        • Ou S.H.
        • Bang Y.J.
        • et al.
        Crizotinib in ROS1-rearranged non-small-cell lung cancer.
        N Engl J Med. 2014; 371: 1963-1971