47
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Setting up a wide panel of patient-derived tumor xenografts of non–small cell lung cancer by improving the preanalytical steps

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          With the ongoing need to improve therapy for non–small cell lung cancer (NSCLC) there has been increasing interest in developing reliable preclinical models to test novel therapeutics. Patient-derived tumor xenografts (PDX) are considered to be interesting candidates. However, the establishment of such model systems requires highly specialized research facilities and introduces logistic challenges. We aimed to establish an extensive well-characterized panel of NSCLC xenograft models in the context of a long-distance research network after careful control of the preanalytical steps. One hundred fresh surgically resected NSCLC specimens were shipped in survival medium at room temperature from a hospital-integrated biobank to animal facilities. Within 24 h post-surgery, tumor fragments were subcutaneously xenografted into immunodeficient mice. PDX characterization was performed by histopathological, immunohistochemical, aCGH and next-generation sequencing approaches. For this model system, the tumor take rate was 35%, with higher rates for squamous carcinoma (60%) than for adenocarcinoma (13%). Patients for whom PDX tumors were obtained had a significantly shorter disease-free survival (DFS) compared to patients for whom no PDX tumors ( =  0.039) were obtained. We established a large panel of PDX NSCLC models with a high frequency of mutations (29%) in EGFR, KRAS, NRAS, MEK1, BRAF, PTEN, and PI3KCA genes and with gene amplification (20%) of c-MET and FGFR1. This new patient-derived NSCLC xenograft collection, established regardless of the considerable time required and the distance between the clinic and the animal facilities, recapitulated the histopathology and molecular diversity of NSCLC and provides stable and reliable preclinical models for human lung cancer research.

          Related collections

          Most cited references20

          • Record: found
          • Abstract: found
          • Article: not found

          Genome Remodeling in a Basal-like Breast Cancer Metastasis and Xenograft

          Massively parallel DNA sequencing technologies provide an unprecedented ability to screen entire genomes for genetic changes associated with tumor progression. Here we describe the genomic analyses of four DNA samples from an African-American patient with basal-like breast cancer: peripheral blood, the primary tumor, a brain metastasis, and a xenograft derived from the primary tumor. The metastasis contained two de novo mutations and a large deletion not present in the primary tumor, and was significantly enriched for 20 shared mutations. The xenograft retained all primary tumor mutations, and displayed a mutation enrichment pattern that paralleled the metastasis (16 of 20 genes). Two overlapping large deletions, encompassing CTNNA1, were present in all three tumor samples. The differential mutation frequencies and structural variation patterns in metastasis and xenograft compared to the primary tumor suggest that secondary tumors may arise from a minority of cells within the primary.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Highly tumorigenic lung cancer CD133+ cells display stem-like features and are spared by cisplatin treatment.

            The identification of lung tumor-initiating cells and associated markers may be useful for optimization of therapeutic approaches and for predictive and prognostic information in lung cancer patients. CD133, a surface glycoprotein linked to organ-specific stem cells, was described as a marker of cancer-initiating cells in different tumor types. Here, we report that a CD133+, epithelial-specific antigen-positive (CD133+ESA+) population is increased in primary nonsmall cell lung cancer (NSCLC) compared with normal lung tissue and has higher tumorigenic potential in SCID mice and expression of genes involved in stemness, adhesion, motility, and drug efflux than the CD133(-) counterpart. Cisplatin treatment of lung cancer cells in vitro resulted in enrichment of CD133+ fraction both after acute cytotoxic exposure and in cells with stable cisplatin-resistant phenotype. Subpopulations of CD133+ABCG2+ and CD133+CXCR4+ cells were spared by in vivo cisplatin treatment of lung tumor xenografts established from primary tumors. A tendency toward shorter progression-free survival was observed in CD133+ NSCLC patients treated with platinum-containing regimens. Our results indicate that chemoresistant populations with highly tumorigenic and stem-like features are present in lung tumors. The molecular features of these cells may provide the rationale for more specific therapeutic targeting and the definition of predictive factors in clinical management of this lethal disease.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              A renewable tissue resource of phenotypically stable, biologically and ethnically diverse, patient-derived human breast cancer xenograft models.

              Breast cancer research is hampered by difficulties in obtaining and studying primary human breast tissue, and by the lack of in vivo preclinical models that reflect patient tumor biology accurately. To overcome these limitations, we propagated a cohort of human breast tumors grown in the epithelium-free mammary fat pad of severe combined immunodeficient (SCID)/Beige and nonobese diabetic (NOD)/SCID/IL-2γ-receptor null (NSG) mice under a series of transplant conditions. Both models yielded stably transplantable xenografts at comparably high rates (∼21% and ∼19%, respectively). Of the conditions tested, xenograft take rate was highest in the presence of a low-dose estradiol pellet. Overall, 32 stably transplantable xenograft lines were established, representing 25 unique patients. Most tumors yielding xenografts were "triple-negative" [estrogen receptor (ER)-progesterone receptor (PR)-HER2+; n = 19]. However, we established lines from 3 ER-PR-HER2+ tumors, one ER+PR-HER2-, one ER+PR+HER2-, and one "triple-positive" (ER+PR+HER2+) tumor. Serially passaged xenografts show biologic consistency with the tumor of origin, are phenotypically stable across multiple transplant generations at the histologic, transcriptomic, proteomic, and genomic levels, and show comparable treatment responses as those observed clinically. Xenografts representing 12 patients, including 2 ER+ lines, showed metastasis to the mouse lung. These models thus serve as a renewable, quality-controlled tissue resource for preclinical studies investigating treatment response and metastasis. ©2013 AACR.
                Bookmark

                Author and article information

                Journal
                Cancer Med
                Cancer Med
                cam4
                Cancer Medicine
                BlackWell Publishing Ltd (Oxford, UK )
                2045-7634
                2045-7634
                February 2015
                03 December 2014
                : 4
                : 2
                : 201-211
                Affiliations
                [1 ]Laboratory of Clinical and Experimental Pathology, Louis Pasteur Hospital Nice, France
                [2 ]IRCAN Team 3, Inserm U1081/UMR CNRS 7284, Faculty of Medicine of Nice, University of Nice Sophia Antipolis Nice, France
                [3 ]Faculty of Medicine, University of Nice Sophia-Antipolis Nice, France
                [4 ]Hospital Related Biobank BB-0033-00025, Louis Pasteur Hospital Nice, France
                [5 ]Oncology Department, Sanofi Laboratories Vitry-sur-Seine, France
                [6 ]SCP Biologics Department, Sanofi Laboratories Vitry-sur-Seine, France
                [7 ]Department of Thoracic Surgery, Louis Pasteur Hospital Nice, France
                Author notes
                Correspondence Paul Hofman, Laboratory of Clinical and Experimental Pathology, Pasteur Hospital, 30 Voie Romaine, BP69, 06001 Nice Cedex, France. Tel: +33 4 92 03 88 55; Fax: +33 4 92 88 50; E-mail: hofman.p@ 123456chu-nice.fr

                Funding Information This work was supported by the French Government (National Research Agency, ANR) through the « Investments for the Future » LABEX SIGNALIFE: program reference # ANR-11-LABX-0028-01.

                [a]

                These authors contributed equally to this work.

                Article
                10.1002/cam4.357
                4329004
                25470237
                390666b6-86c1-41ca-84a6-09a70cb5cb3a
                © 2014 The Authors. Cancer Medicine published by John Wiley & Sons Ltd.

                This is an open access article under the terms of the Creative Commons Attribution License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited.

                History
                : 12 June 2014
                : 26 August 2014
                : 31 August 2014
                Categories
                Cancer Research
                Original Research

                Oncology & Radiotherapy
                molecular pathology,nsclc,pdx,preanalytical
                Oncology & Radiotherapy
                molecular pathology, nsclc, pdx, preanalytical

                Comments

                Comment on this article