Cancer Letters

Cancer Letters

Volume 357, Issue 1, 1 February 2015, Pages 179-185
Cancer Letters

Original Articles
Gene mutations in primary tumors and corresponding patient-derived xenografts derived from non-small cell lung cancer

https://doi.org/10.1016/j.canlet.2014.11.024Get rights and content

Highlights

  • 23 PDXs from lung cancer and their primary tumors were sequenced by exome sequencing analysis on 202 cancer-related genes.

  • Numbers of primary tumors with mutations in TP53, KRAS, PI3KCA, ALK, STK11, and EGFR were 10, 5, 4, 4, 3, and 2, respectively.

  • Other genes with mutations in ≥3 tumors were MLL3, SETD2, ATM, ARID1A, CRIPAK, HGF, BAI3, EP300, KDR, PDGRRA and RUNX1.

  • 93% of mutations detected in the primary tumors were also detected in their corresponding PDXs.

  • Some mutations had higher allele frequencies in the PDXs than in the primary tumors, suggesting tumor cell enrichment in PDXs.

Abstract

Molecular annotated patient-derived xenograft (PDX) models are useful for the preclinical investigation of anticancer drugs and individualized anticancer therapy. We established 23 PDXs from 88 surgical specimens of lung cancer patients and determined gene mutations in these PDXs and their paired primary tumors by ultradeep exome sequencing on 202 cancer-related genes. The numbers of primary tumors with deleterious mutations in TP53, KRAS, PI3KCA, ALK, STK11, and EGFR were 43.5%, 21.7%, 17.4%, 17.4%, 13.0%, and 8.7%, respectively. Other genes with deleterious mutations in ≥3 (13.0%) primary tumors were MLL3, SETD2, ATM, ARID1A, CRIPAK, HGF, BAI3, EP300, KDR, PDGRRA and RUNX1. Of 315 mutations detected in the primary tumors, 293 (93%) were also detected in their corresponding PDXs, indicating that PDXs have the capacity to recapitulate the mutations in primary tumors. Nevertheless, a substantial number of mutations had higher allele frequencies in the PDXs than in the primary tumors, or were not detectable in the primary tumor, suggesting the possibility of tumor cell enrichment in PDXs or heterogeneity in the primary tumors. The molecularly annotated PDXs generated from this study could be useful for future translational studies.

Introduction

Lung cancer is the leading cause of cancer-related deaths both in the United States and worldwide, with an annual global incidence of about 1.6 million and mortality of 1.4–1.5 million [1], [2], [3]. Recent advances in genomic profiling have led to the identification of a number of frequently mutated genes in lung cancer [4], [5], [6], [7]. Lung cancers with the same histological diagnosis and clinical stages can be classified into molecular subgroups based on gene mutations. Substantial efforts have been made to develop genotype-specific anticancer therapeutics. The finding that lung cancer cells with mutations in the epidermal growth factor receptor gene (EGFR) are highly susceptible to the EGFR inhibitors gefitinib [8], [9], [10], erlotinib [8], [11] and afatinib has made these agents the first choice for treating EGFR mutant lung cancer. Both gefitinib and erlotinib have been reported to significantly prolong progression-free survival in patients with EGFR-mutant lung cancer [12], [13]. Similarly, small molecular inhibitors for anaplastic lymphoma kinase (ALK) and ROS1 have been proven to be highly effective for treatment of lung cancers with ALK and ROS1 gene translocations [14], [15], [16]. However, despite the excitement accompanying the targeted therapeutics, only a subset of patients with the aberration respond and responses are often unfortunately brief. Furthermore, our knowledge of genetic alterations, their functional consequences and combinatorial effects in lung cancer is still not comprehensive. For most potential driver mutations identified in lung cancer, there are no effective therapeutic agents available. The success of the EGFR inhibitors underscores the urgency of developing effective genotype-specific anticancer therapeutics.

Anticancer drug development is often impeded by a lack of preclinical tumor models that are highly predictive of therapeutic effects in humans. Previous studies have shown that in vitro cell line models and in vivo xenograft tumors derived from established human cancer cell lines have limited predictive value for antitumor activity of a drug in clinical trials [17], [18], [19]. Anticancer agents that showed promising in vivo antitumor activity in xenograft tumor models have often been ineffective for the same type of cancer in clinical trials [20]. In fact, only about 5% of anticancer agents evaluated in human studies between 1991 and 2000 were successfully registered [20]. The majority of failures in late-phase clinical trials result from a lack of clinical efficacy caused primarily by the lack of efficacy proof of concept in humans, lack of predictive biomarkers to identify patient responders, and safety issues [20], [21]. Thus, clinically relevant tumor models that accurately predict therapeutic efficacies would be highly valuable for anticancer drug development.

Evidence from recent studies has shown that patient-derived xenografts (PDXs) established directly from patients' primary tumors preserve the histomorphologic features, heterogeneity, gene expression pattern (including cytokine expression by tumor stromal cells), DNA copy number alterations, and gene mutations of the original tumors [22], [23], [24]. These features were preserved after a series of passages of the tumorgrafts in mice [22], [24]. When PDXs were treated with agents used in a parallel patient population, response rates similar to those reported in human studies were observed, suggesting that the PDX model is clinically relevant for evaluating the efficacy of anticancer drugs [22], [25], [26], [27], [28]. A remarkable correlation between drug activity in PDXs and clinical outcome was reported when patients with advanced cancer were treated with selected regimens based on the treatment responses of their PDX [29], [30], suggesting that PDXs could provide robust models for identifying effective treatment for cancer patients and for predicting clinical efficacy of drug candidates. Consequently, PDXs derived from various types of cancers have been reported recently, including those established from lung cancer [23], [26], [28], [31]. Those studies have demonstrated the feasibility of using PDXs for translational studies in drug development, for molecular characterization of cancer biology, and for strategic development of individualized therapy. Nevertheless, few molecularly-annotated lung cancer PDXs are reported in literature and are not readily available for preclinical studies.

Our purpose here was to develop molecularly annotated PDXs for evaluation of investigational anticancer agents and mechanistic characterization of lung cancers. We established PDXs from surgical specimens of lung cancer patients and characterized the gene mutations in those PDXs and the corresponding primary tumors. Our results show that some novel genes were frequently mutated in primary lung cancers and that the mutations in primary tumors can be recapitulated by their corresponding PDX.

Section snippets

Human lung tissue specimens

Fresh lung cancer samples were collected in 2012 and 2013 from surgically resected specimens under approved research protocols with informed consent from the patients. This study was approved by the Institutional Review Board at The University of Texas MD Anderson Cancer Center.

Generation of patient-derived xenografts in immune-defective mice

All animal experiments were carried out in accordance with Guidelines for the Care and Use of Laboratory Animals (NIH publication number 85-23) and the institutional guidelines of MD Anderson Cancer Center. Six- to

Establishing patient-derived xenografts from lung cancer specimens

We collected surgically resected tumor samples from 88 NSCLC patients and implanted each specimen into 2–3 NOD-SCID mice to develop PDXs. We obtained 23 PDXs (Table 1). The overall implantation rate for development of a PDX was 26%. Squamous cancer and neuroendocrinal carcinoma had relatively higher implantation rates than adenocarcinoma. Moderately and poorly differentiated tumors had relative high implantation rates than well differentiated tumors (Fig. 1A). Nevertheless, the difference among

Discussion

Our study resulted in 23 molecularly-annotated PDXs that will be useful for preclinical evaluation of investigational lung cancer-targeting agents and/or for molecular characterization of lung cancers. Although the number of cases where PDXs were established in this study is relatively small, our studies allowed us to detect a number of genes that were frequently mutated in lung cancer. Many of those genes were consistent with those reported previously by others. Some of those genes, such as

Funding

This work was supported in part by the National Institutes of Health through Specialized Program of Research Excellence (SPORE) Grant CA070907 and The University of Texas MD Anderson Cancer Center Core Support Grant CA016672 (Lung Program DNA Analysis and Bioinformatics Core Facilities). Further support came from MD Anderson Cancer Center endowed funds, including the Moon Shot Program, the Stading Lung Cancer Research Fund, and the M.W. Elkins Endowed Fund for Thoracic Surgical Oncology.

Conflict of interest

The authors declare no conflict of interest.

Acknowledgements

We thank Kathryn Hale of the Department of Scientific Publication at The University of Texas MD Anderson Cancer Center for editorial review of this manuscript.

References (58)

  • R. Siegel et al.

    Cancer statistics, 2014

    CA Cancer J. Clin

    (2014)
  • A. Jemal et al.

    Global cancer statistics

    CA Cancer J. Clin

    (2011)
  • L. Ding et al.

    Somatic mutations affect key pathways in lung adenocarcinoma

    Nature

    (2008)
  • P.S. Hammerman et al.

    Comprehensive genomic characterization of squamous cell lung cancers

    Nature

    (2012)
  • W. Pao et al.

    EGF receptor gene mutations are common in lung cancers from “never smokers” and are associated with sensitivity of tumors to gefitinib and erlotinib

    Proc. Natl. Acad. Sci. U.S.A.

    (2004)
  • T.J. Lynch et al.

    Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib

    N. Engl. J. Med

    (2004)
  • J.G. Paez et al.

    EGFR mutations in lung cancer: correlation with clinical response to gefitinib therapy

    Science

    (2004)
  • F.A. Shepherd et al.

    Erlotinib in previously treated non-small-cell lung cancer

    N. Engl. J. Med

    (2005)
  • M. Fukuoka et al.

    Biomarker analyses and final overall survival results from a phase III, randomized, open-label, first-line study of gefitinib versus carboplatin/paclitaxel in clinically selected patients with advanced non-small-cell lung cancer in Asia (IPASS)

    J. Clin. Oncol

    (2011)
  • A.T. Shaw et al.

    Ceritinib in ALK-rearranged non-small-cell lung cancer

    N. Engl. J. Med

    (2014)
  • M.A. Davare et al.

    Foretinib is a potent inhibitor of oncogenic ROS1 fusion proteins

    Proc. Natl. Acad. Sci. U.S.A.

    (2013)
  • K. Bergethon et al.

    ROS1 rearrangements define a unique molecular class of lung cancers

    J. Clin. Oncol

    (2012)
  • K. Garber

    From human to mouse and back: ‘tumorgraft’ models surge in popularity

    J. Natl. Cancer Inst

    (2009)
  • J.I. Johnson et al.

    Relationships between drug activity in NCI preclinical in vitro and in vivo models and early clinical trials

    Br. J. Cancer

    (2001)
  • T. Voskoglou-Nomikos et al.

    Clinical predictive value of the in vitro cell line, human xenograft, and mouse allograft preclinical cancer models

    Clin. Cancer Res

    (2003)
  • I. Kola

    The state of innovation in drug development

    Clin. Pharmacol. Ther

    (2008)
  • J. Arrowsmith

    Trial watch: phase II failures: 2008–2010

    Nat. Rev. Drug Discov

    (2011)
  • S. Sivanand et al.

    A validated tumorgraft model reveals activity of dovitinib against renal cell carcinoma

    Sci. Transl. Med

    (2012)
  • T. John et al.

    The ability to form primary tumor xenografts is predictive of increased risk of disease recurrence in early-stage non-small cell lung cancer

    Clin. Cancer Res

    (2011)
  • Cited by (79)

    • Cancer epigenetics: Past, present and future

      2022, Seminars in Cancer Biology
      Citation Excerpt :

      Interestingly, the latter involves methylation of HIF-1a protein rather than that of histone proteins 27]. In contrast to the tumor-promoting KMTs described above, SETD1 and SETD2 are often mutated in NSCLC, implying a tumor suppressive role [28,29]. In addition to KMTs, several lysine demethylases (KDMs) also contribute to lung cancer progression.

    • Unbiased peptoid cell screen identifies a peptoid targeting newly appeared cell surface vimentin on tumor transformed early lung cancer cells

      2022, Bioorganic and Medicinal Chemistry
      Citation Excerpt :

      Multiple genetic and epigenetic alterations have been identified in genome-wide analyses of lung tumors7–9. For example, p53 inactivation is an early mutation common in all NSCLC histological subtypes (adenocarcinoma, squamous cell and neuroendocrinal), while oncogenic KRAS mutations are found mainly in adenocarcinomas10–12. While targeting such key and common early mutation based markers would be ideal for cancer detection, for a variety of reasons p53 and KRAS mutation makers have not proved generally useful in detecting premalignant lesions.

    • Epstein Barr virus–positive B-cell lymphoma is highly vulnerable to MDM2 inhibitors in vivo

      2022, Blood Advances
      Citation Excerpt :

      XABCL cells were obtained during our study to establish patient-derived xenografts (PDXs) from clinical specimens from patients with non-small cell lung cancer (NSCLC). The origin of XABCL was verified by DNA fingerprint together with the patients' genomic DNA from peripheral blood mononuclear cells or tumor samples, as previously described.15,16 All clinical samples and data were collected with informed consent of the patients under a research protocol approved by the Institutional Review Board at The University of Texas MD Anderson Cancer Center (MDACC).

    • Kinomic profiling of tumour xenografts derived from patients with non–small cell lung cancer confirms their fidelity and reveals potentially actionable pathways

      2021, European Journal of Cancer
      Citation Excerpt :

      Indeed, PDTXs have emerged, like organoids, as a powerful platform to overcome typical limitations of conventional models (e.g. cell lines or GEMMs) [12,17,32]. Despite the relatively small number of genes sequenced, the genomic stability reported in this study is consistent with results previously described by other groups [18,20,33]. Indeed, defined ‘hotspot’ mutations are associated with higher rate of engraftment (i.e. RAS and PI3K versus EGFR or BRAF mutations), suggesting that certain defects, identified at higher stages, may sustain a more aggressive phenotype.

    View all citing articles on Scopus
    1

    These authors contributed equally.

    View full text