Targeted NGS, array-CGH, and patient-derived tumor xenografts for precision medicine in advanced breast cancer: a single-center prospective study

Anthony Gonçalves, François Bertucci, Arnaud Guille, Severine Garnier, José Adelaide, Nadine Carbuccia, Oliver Cabaud, Pascal Finetti, Serge Brunelle, Gilles Piana, Jeanne Tomassin-Piana, Maria Paciencia, Eric Lambaudie, Cornel Popovici, Renaud Sabatier, Carole Tarpin, Magali Provansal, Jean-Marc Extra, François Eisinger, Hagay Sobol, Patrice Viens, Marc Lopez, Christophe Ginestier, Emmanuelle Charafe-Jauffret, Max Chaffanet, Daniel Birnbaum, Anthony Gonçalves, François Bertucci, Arnaud Guille, Severine Garnier, José Adelaide, Nadine Carbuccia, Oliver Cabaud, Pascal Finetti, Serge Brunelle, Gilles Piana, Jeanne Tomassin-Piana, Maria Paciencia, Eric Lambaudie, Cornel Popovici, Renaud Sabatier, Carole Tarpin, Magali Provansal, Jean-Marc Extra, François Eisinger, Hagay Sobol, Patrice Viens, Marc Lopez, Christophe Ginestier, Emmanuelle Charafe-Jauffret, Max Chaffanet, Daniel Birnbaum

Abstract

Background: Routine feasibility and clinical impact of genomics-based tumor profiling in advanced breast cancer (aBC) remains to be determined. We conducted a pilot study to evaluate whether precision medicine could be prospectively implemented for aBC patients in a single center and to examine whether patient-derived tumor xenografts (PDX) could be obtained in this population.

Results: Thirty-four aBC patients were included. Actionable targets were found in 28 patients (82%). A targeted therapy could be proposed to 22 patients (64%), either through a clinical trial (n=15) and/or using already registered drugs (n=21). Ten patients (29%) eventually received targeted treatment, 2 of them deriving clinical benefit. Of 22 patients subjected to mouse implantation, 10 had successful xenografting (45%), mostly in triple-negative aBC.

Methods: aBC patients accessible to tumor biopsy were prospectively enrolled at the Institut Paoli-Calmettes in the BC-BIO study (ClinicalTrials.gov, NCT01521676). Genomic profiling was established by whole-genome array comparative genomic hybridization (aCGH) and targeted next-generation sequencing (NGS) of 365 candidate cancer genes. For a subset of patients, a sample of fresh tumor was orthotopically implanted in humanized cleared fat pads of NSG mice for establishing PDX.

Conclusions: Precision medicine can be implemented in a single center in the context of clinical practice and may allow genomic-driven treatment in approximately 30% of aBC patients. PDX may be obtained in a significant fraction of cases.

Keywords: CGH; NGS; advanced breast cancer; patient-derived xenograft; precision medicine.

Conflict of interest statement

CONFLICTS OF INTEREST

Authors declare no potential conflicts of interest

Figures

Figure 1. Distribution of molecular alterations identified…
Figure 1. Distribution of molecular alterations identified by CGH arrays and NGS in all 32 samples
A. Upper, mid and lower panels indicate the percentage of altered genomes, the mutation rate and the depth of sequencing for each sample, respectively (dot lines indicate mean percentage of altered genome, median mutation rate and median depth of sequencing, respectively). Gain, amplification, loss and deletion as well as neutral, damaging, hot spot and indel mutations were defined as indicated in the material and methods section. B. The distribution of molecular alterations (blue, mutation; red, amplification; green, deletion) was shown by decreasing frequency across samples (unique and anonymized patient number). Only alterations present in more than 1 sample are shown. Sample subtypes (HR-positive/HER2-negative, blue; HER2-positive, pink; triple-negative, red) are indicated. Patients with PDX engraftment (“PDX”) and genomic-driven treatment (therapeutic) are also shown. Asterisks indicate patients without germline sequencing.
Figure 2. Impact of germline sequencing on…
Figure 2. Impact of germline sequencing on the number of retained mutations
Patients with available normal samples (N=27) were subjected to constitutional sequencing and somatic mutations were identified as indicated in the Material and methods section. Results are shown on a Venn diagram.
Figure 3. Correlations between molecular alterations and…
Figure 3. Correlations between molecular alterations and clinical features
The mutation rate A. and the fraction of altered genome B. were calculated as indicated in the material and method section and compared across molecular subtypes. C. The fraction of altered genome was compared according to the presence of visceral or non-visceral metastases. D. Overall survival was compared according to the fraction of altered genome. The median value of the overall population (0.2) was selected as threshold. * ANOVA ** student's t-test *** Log-rank test.
Figure 4. Actionable alterations retained by molecular…
Figure 4. Actionable alterations retained by molecular tumor board
Actionable alterations were selected according to rules defined in the material and methods section. Mutations, amplifications and deletions are shown across the samples analyzed (N=32) in red, yellow and blue, respectively.

References

    1. Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, Parkin DM, Forman D, Bray F. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer J Int Cancer. 2015;136:E359–386. doi: 10.1002/ijc.29210.
    1. Jemal A, Siegel R, Ward E, Hao Y, Xu J, Murray T. Cancer statistics, 2008. CA Cancer J Clin. 2008;58:71–96.
    1. Joy AA, Ghosh M, Fernandes R, Clemons MJ. Systemic treatment approaches in her2-negative advanced breast cancer—guidance on the guidelines. Curr Oncol. 201522:S29–S42. doi: 10.3747/co.22.2360.
    1. Perou C, Sorlie T, Eisen M, van de Rijn M, Jeffrey S, Rees C, Pollack J, Ross D, Johnsen H, Akslen L. Molecular portraits of human breast tumours. Nature. 2000;406:747–52.
    1. Network TCGA Comprehensive molecular portraits of human breast tumours. Nature. 2012;490:61–70.
    1. Nik-Zainal S, Davies H, Staaf J, Ramakrishna M, Glodzik D, Zou X, Martincorena I, Alexandrov LB, Martin S, Wedge DC, Van Loo P, Ju YS, Smid M, et al. Landscape of somatic mutations in 560 breast cancer whole-genome sequences. Nature. 2016;534:47–54. doi: 10.1038/nature17676.
    1. Garraway LA, Verweij J, Ballman KV. Precision oncology: an overview. J Clin Oncol. 2013;31:1803–5. doi: 10.1200/JCO.2013.49.4799.
    1. Early Breast Cancer Trialists' Collaborative Group (EBCTCG) Effects of chemotherapy and hormonal therapy for early breast cancer on recurrence and 15-year survival: an overview of the randomised trials. Lancet. 2005;365:1687–717. doi: 10.1016/S0140-6736(05)66544-0.
    1. Slamon DJ, Leyland-Jones B, Shak S, Fuchs H, Paton V, Bajamonde A, Fleming T, Eiermann W, Wolter J, Pegram M, Baselga J, Norton L. Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N Engl J Med. 2001;344:783–92.
    1. Romond EH, Perez EA, Bryant J, Suman VJ, Geyer CE, Davidson NE, Tan-Chiu E, Martino S, Paik S, Kaufman PA, Swain SM, Pisansky TM, Fehrenbacher L, et al. Trastuzumab plus adjuvant chemotherapy for operable HER2-positive breast cancer. N Engl J Med. 2005;353:1673–84.
    1. Piccart-Gebhart MJ, Procter M, Leyland-Jones B, Goldhirsch A, Untch M, Smith I, Gianni L, Baselga J, Bell R, Jackisch C, Cameron D, Dowsett M, Barrios CH, et al. Trastuzumab after adjuvant chemotherapy in HER2-positive breast cancer. N Engl J Med. 2005;353:1659–72.
    1. Stephens PJ, Tarpey PS, Davies H, Van Loo P, Greenman C, Wedge DC, Nik-Zainal S, Martin S, Varela I, Bignell GR, Yates LR, Papaemmanuil E, Beare D, et al. The landscape of cancer genes and mutational processes in breast cancer. Nature. 2012;486:400–4. doi: 10.1038/nature11017.
    1. Curtis C, Shah SP, Chin S-F, Turashvili G, Rueda OM, Dunning MJ, Speed D, Lynch AG, Samarajiwa S, Yuan Y, Gräf S, Ha G, Haffari G, et al. The genomic and transcriptomic architecture of 2,000 breast tumours reveals novel subgroups. Nature. 2012;486:346–52. doi: 10.1038/nature10983.
    1. André F, Bachelot T, Commo F, Campone M, Arnedos M, Dieras V, Lacroix-Triki M, Lacroix L, Cohen P, Gentien D, Adélaide J, Dalenc F, Gonçalves A, et al. Comparative genomic hybridisation array and DNA sequencing to direct treatment of metastatic breast cancer: a multicentre, prospective trial (SAFIR01/UNICANCER) Lancet Oncol. 2014;15:267–74. doi: 10.1016/S1470-2045(13)70611-9.
    1. DeRose YS, Wang G, Lin Y-C, Bernard PS, Buys SS, Ebbert MTW, Factor R, Matsen C, Milash BA, Nelson E, Neumayer L, Randall RL, Stijleman IJ, et al. Tumor grafts derived from women with breast cancer authentically reflect tumor pathology, growth, metastasis and disease outcomes. Nat Med. 2011;17:1514–20. doi: 10.1038/nm.2454.
    1. Zhang X, Claerhout S, Prat A, Dobrolecki LE, Petrovic I, Lai Q, Landis MD, Wiechmann L, Schiff R, Giuliano M, Wong H, Fuqua SW, Contreras A, et al. A renewable tissue resource of phenotypically stable, biologically and ethnically diverse, patient-derived human breast cancer xenograft models. Cancer Res. 2013;73:4885–97. doi: 10.1158/0008-5472.CAN-12-4081.
    1. Charafe-Jauffret E, Ginestier C, Bertucci F, Cabaud O, Wicinski J, Finetti P, Josselin E, Adelaide J, Nguyen T-T, Monville F, Jacquemier J, Thomassin-Piana J, Pinna G, et al. ALDH1-positive cancer stem cells predict engraftment of primary breast tumors and are governed by a common stem cell program. Cancer Res. 2013;73:7290–300. doi: 10.1158/0008-5472.CAN-12-4704.
    1. De Mattos-Arruda L, Caldas C. Cell-free circulating tumour DNA as a liquid biopsy in breast cancer. Mol Oncol. 2015 doi: 10.1016/j.molonc.2015.12.001..
    1. Jamal-Hanjani M, Quezada SA, Larkin J, Swanton C. Translational implications of tumor heterogeneity. Clin Cancer Res. 2015;21:1258–66. doi: 10.1158/1078-0432.CCR-14-1429.
    1. Le Tourneau C, Delord J-P, Gonçalves A, Gavoille C, Dubot C, Isambert N, Campone M, Trédan O, Massiani M-A, Mauborgne C, Armanet S, Servant N, Bièche I, et al. Molecularly targeted therapy based on tumour molecular profiling versus conventional therapy for advanced cancer (SHIVA): a multicentre, open-label, proof-of-concept, randomised, controlled phase 2 trial. Lancet Oncol. 2015;16:1324–34. doi: 10.1016/S1470-2045(15)00188-6.
    1. Le Tourneau C, Kamal M, Tsimberidou A-M, Bedard P, Pierron G, Callens C, Rouleau E, Vincent-Salomon A, Servant N, Alt M, Rouzier R, Paoletti X, Delattre O, et al. Treatment Algorithms Based on Tumor Molecular Profiling: The Essence of Precision Medicine Trials. J Natl Cancer Inst. 2016:108. doi: 10.1093/jnci/djv362.
    1. Swanton C, Soria J-C, Bardelli A, Biankin A, Caldas C, Chandarlapaty S, de Koning L, Dive C, Feunteun J, Leung S-Y, Marais R, Mardis ER, McGranahan N, et al. Consensus on precision medicine for metastatic cancers: a report from the MAP conference. Ann Oncol. 2016;27:1443–8. doi: 10.1093/annonc/mdw192.
    1. Bosserman L, Rogers K, Willis C, Davidson D, Whitworth P, Karimi M, Upadhyaya G, Rutledge J, Hallquist A, Perree M, Presant CA. Application of a drug-induced apoptosis assay to identify treatment strategies in recurrent or metastatic breast cancer. PloS One. 2015;10:e0122609. doi: 10.1371/journal.pone.0122609.
    1. Francies HE, Barthorpe A, McLaren-Douglas A, Barendt WJ, Garnett MJ. Drug Sensitivity Assays of Human Cancer Organoid Cultures. Methods Mol Biol. 2016 doi: 10.1007/7651_2016_10.
    1. Rooney MS, Shukla SA, Wu CJ, Getz G, Hacohen N. Molecular and genetic properties of tumors associated with local immune cytolytic activity. Cell. 2015;160:48–61. doi: 10.1016/j.cell.2014.12.033.
    1. Brown SD, Warren RL, Gibb EA, Martin SD, Spinelli JJ, Nelson BH, Holt RA. Neo-antigens predicted by tumor genome meta-analysis correlate with increased patient survival. Genome Res. 2014;24:743–50. doi: 10.1101/gr.165985.113.
    1. Howitt BE, Shukla SA, Sholl LM, Ritterhouse LL, Watkins JC, Rodig S, Stover E, Strickland KC, D'Andrea AD, Wu CJ, Matulonis UA, Konstantinopoulos PA. Association of Polymerase e-Mutated and Microsatellite-Instable Endometrial Cancers With Neoantigen Load, Number of Tumor-Infiltrating Lymphocytes, and Expression of PD-1 and PD-L1. JAMA Oncol. 2015;1:1319–23. doi: 10.1001/jamaoncol.2015.2151.
    1. Hammond MEH, Hayes DF, Wolff AC, Mangu PB, Temin S. American society of clinical oncology/college of american pathologists guideline recommendations for immunohistochemical testing of estrogen and progesterone receptors in breast cancer. J Oncol Pract Am Soc Clin Oncol. 2010;6:195–7. doi: 10.1200/JOP.777003.
    1. Wolff AC, Hammond MEH, Hicks DG, Dowsett M, McShane LM, Allison KH, Allred DC, Bartlett JMS, Bilous M, Fitzgibbons P, Hanna W, Jenkins RB, Mangu PB, et al. Recommendations for human epidermal growth factor receptor 2 testing in breast cancer: American Society of Clinical Oncology/College of American Pathologists clinical practice guideline update. J Clin Oncol. 2013;31:3997–4013. doi: 10.1200/JCO.2013.50.9984.
    1. Desmedt C, Di Leo A, de Azambuja E, Larsimont D, Haibe-Kains B, Selleslags J, Delaloge S, Duhem C, Kains J-P, Carly B, Maerevoet M, Vindevoghel A, Rouas G, et al. Multifactorial approach to predicting resistance to anthracyclines. J Clin Oncol. 2011;29:1578–86. doi: 10.1200/JCO.2010.31.2231.
    1. Adelaide J, Finetti P, Bekhouche I, Repellini L, Geneix J, Sircoulomb F, Charafe-Jauffret E, Cervera N, Desplans J, Parzy D, Schoenmakers E, Viens P, Jacquemier J, et al. Integrated profiling of basal and luminal breast cancers. Cancer Res. 2007;67:11565–75.
    1. Sircoulomb F, Bekhouche I, Finetti P, Adélaïde J, Ben Hamida A, Bonansea J, Raynaud S, Innocenti C, Charafe-Jauffret E, Tarpin C, Ben Ayed F, Viens P, Jacquemier J, et al. Genome profiling of ERBB2-amplified breast cancers. BMC Cancer. 2010;10:539. doi: 10.1186/1471-2407-10-539.
    1. Bekhouche I, Finetti P, Adelaïde J, Ferrari A, Tarpin C, Charafe-Jauffret E, Charpin C, Houvenaeghel G, Jacquemier J, Bidaut G, Birnbaum D, Viens P, Chaffanet M, et al. High-resolution comparative genomic hybridization of inflammatory breast cancer and identification of candidate genes. PloS One. 2011;6:e16950. doi: 10.1371/journal.pone.0016950.
    1. Bertucci F, Finetti P, Guille A, Adélaïde J, Garnier S, Carbuccia N, Monneur A, Charafe-Jauffret E, Gonçalves A, Viens P, Birnbaum D, Chaffanet M. Comparative genomic analysis of primary tumors and metastases in breast cancer. Oncotarget. 2016;7:27208–19. doi: 10.18632/oncotarget.8349.
    1. Collette Y, Prébet T, Goubard A, Adélaïde J, Castellano R, Carbuccia N, Garnier S, Guille A, Arnoulet C, Charbonier A, Mozziconacci MJ, Birnbaum D, Chaffanet M, et al. Drug response profiling can predict response to ponatinib in a patient with t(1;9)(q24;q34)-associated B-cell acute lymphoblastic leukemia. Blood Cancer J. 2015;5:e292. doi: 10.1038/bcj.2015.13.
    1. Li H, Durbin R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinforma Oxf Engl. 2009;25:1754–60. doi: 10.1093/bioinformatics/btp324.
    1. Garrison E, Marth G. ArXiv12073907 Q-Bio. 2012. Haplotype-based variant detection from short-read sequencing. [cited 2016 Feb 8]; Available from.
    1. DePristo MA, Banks E, Poplin R, Garimella KV, Maguire JR, Hartl C, Philippakis AA, del Angel G, Rivas MA, Hanna M, McKenna A, Fennell TJ, Kernytsky AM, et al. A framework for variation discovery and genotyping using next-generation DNA sequencing data. Nat Genet. 2011;43:491–8. doi: 10.1038/ng.806.
    1. Robinson JT, Thorvaldsdóttir H, Winckler W, Guttman M, Lander ES, Getz G, Mesirov JP. Integrative genomics viewer. Nat Biotechnol. 2011;29:24–6. doi: 10.1038/nbt.1754.
    1. Thorvaldsdóttir H, Robinson JT, Mesirov JP. Integrative Genomics Viewer (IGV): high-performance genomics data visualization and exploration. Brief Bioinform. 2013;14:178–92. doi: 10.1093/bib/bbs017.
    1. Cibulskis K, Lawrence MS, Carter SL, Sivachenko A, Jaffe D, Sougnez C, Gabriel S, Meyerson M, Lander ES, Getz G. Sensitive detection of somatic point mutations in impure and heterogeneous cancer samples. Nat Biotechnol. 2013;31:213–9. doi: 10.1038/nbt.2514.
    1. Dienstmann R, Jang IS, Bot B, Friend S, Guinney J. Database of genomic biomarkers for cancer drugs and clinical targetability in solid tumors. Cancer Discov. 2015;5:118–23. doi: 10.1158/-14-1118.

Source: PubMed

3
Sottoscrivi