An approach to suppress the evolution of resistance in BRAFV600E-mutant cancer

Yaohua Xue, Luciano Martelotto, Timour Baslan, Alberto Vides, Martha Solomon, Trang Thi Mai, Neelam Chaudhary, Greg J Riely, Bob T Li, Kerry Scott, Fabiola Cechhi, Ulrika Stierner, Kalyani Chadalavada, Elisa de Stanchina, Sarit Schwartz, Todd Hembrough, Gouri Nanjangud, Michael F Berger, Jonas Nilsson, Scott W Lowe, Jorge S Reis-Filho, Neal Rosen, Piro Lito, Yaohua Xue, Luciano Martelotto, Timour Baslan, Alberto Vides, Martha Solomon, Trang Thi Mai, Neelam Chaudhary, Greg J Riely, Bob T Li, Kerry Scott, Fabiola Cechhi, Ulrika Stierner, Kalyani Chadalavada, Elisa de Stanchina, Sarit Schwartz, Todd Hembrough, Gouri Nanjangud, Michael F Berger, Jonas Nilsson, Scott W Lowe, Jorge S Reis-Filho, Neal Rosen, Piro Lito

Abstract

The principles that govern the evolution of tumors exposed to targeted therapy are poorly understood. Here we modeled the selection and propagation of an amplification in the BRAF oncogene (BRAFamp) in patient-derived tumor xenografts (PDXs) that were treated with a direct inhibitor of the kinase ERK, either alone or in combination with other ERK signaling inhibitors. Single-cell sequencing and multiplex fluorescence in situ hybridization analyses mapped the emergence of extra-chromosomal amplification in parallel evolutionary trajectories that arose in the same tumor shortly after treatment. The evolutionary selection of BRAFamp was determined by the fitness threshold, the barrier that subclonal populations need to overcome to regain fitness in the presence of therapy. This differed for inhibitors of ERK signaling, suggesting that sequential monotherapy is ineffective and selects for a progressively higher BRAF copy number. Concurrent targeting of the RAF, MEK and ERK kinases, however, imposed a sufficiently high fitness threshold to prevent the propagation of subclones with high-level BRAFamp. When administered on an intermittent schedule, this treatment inhibited tumor growth in 11/11 PDXs of lung cancer or melanoma without apparent toxicity in mice. Thus, gene amplification can be acquired and expanded through parallel evolution, enabling tumors to adapt while maintaining their intratumoral heterogeneity. Treatments that impose the highest fitness threshold will likely prevent the evolution of resistance-causing alterations and, thus, merit testing in patients.

Figures

Figure 1. ERK inhibitor-resistant populations with extrachromosomal…
Figure 1. ERK inhibitor-resistant populations with extrachromosomal BRAF amplification
(a) Patient-derived xenograft (PDX) models from patients with BRAFV600E-mutant lung cancer or melanoma were treated with ERK inhibitor (ERKi) SCH984 over time (n = 5 mice, mean ± s.e.m). (b) H&E stained sections of the PDX models before and after ERKi treatment. (c) Single nuclei extracted from PDX1D tumors were analyzed by FACS to determine the distribution of cells according to their DNA content. A human diploid cell line was used as a control. (d) Copy number (CN) profiles of 69 single cells derived from parental (Par) and ERK inhibitor-resistant (EiR) PDX-1D tumors. (e), Projection of single cells into the top three principal components. (f) Subclonal distribution of parental and resistant tumors. (g) Segment values spanning the BRAF locus in tumor and stromal cells. For stromal cells, sequenced reads were mapped to the mouse genome (see Supplementary Fig. 1e). (h) Representative images of fluorescence in situ hybridization (FISH) analysis with probes spanning BRAF or chr7 centromere in red or green, respectively (a representative of five different fields is shown). (i) Probes were quantified by manual counting (n = 100 cells, all data are shown). (j) Representative image of extra-chromosomal localization of the BRAF gene (arrows) in an 1D-EiR cell undergoing metaphase. (k) The expression of BRAFV600E protein in matched PDX1D and 1E tumor sets was determined by mass spectrometry (n = 3, mean ± s.e.m). Actin and tubulin were used as controls.
Figure 2. BRAF amp emerges in parallel…
Figure 2. BRAFamp emerges in parallel evolutionary tracts
(a) The clonal relationship of single cells, as inferred by Manhattan-Ward clustering of integer CN. The bar graph shows BRAF CN. (b, c) Single cells derived from parental (b) or resistant (c) tumors were subjected to hierarchical clustering and subclonal analysis independent of each other. Phylogenetic inference was established using a heuristic maximal parsimony approach. Subclones A and B were subdivided on the basis of additional CN alterations and their inferred phylogeny. (d) Single cells with high-level BRAFamp were found in three distinct resistant clones. (e) The CN state of select chromosomal regions with heterogeneous profiles. Note the emergence of BRAFamp cells in three distinct evolutionary trajectories depending on co-occurring losses in chr2p, 11q and/or 13 (arrows). (f, g) Multiplex FISH with probes targeting the indicated chromosomal regions in PDX1D-EiR. Manual quantification (f) and representative images (g) are shown.
Figure 3. BRAF amp is sufficient to…
Figure 3. BRAFamp is sufficient to confer a selective advantage in the presence of ERKi treatment
(a) Immunoblot analysis of cell lines derived from parental (1D) or ERK inhibitor-resistant (1D-EiR) PDX. (b) Immunoblot analysis of signaling intermediates in 1D and 1D-EiR cells treated for 1h with SCH984. (c) Cell viability at 72h after treatment. (d) Cell viability of 1D-EiR cells transfected with BRAF specific or control siRNAs followed by drug treatment as in c. (e & f) A375 cells, engineered to express BRAFV600E under a doxycycline (dox)-induced promoter, were treated as shown (dox, 2μg/mL; SCH984, 500 nM) to determine the effect on signaling by immunoblotting (e) or viability (f). Withdrawal of dox after a 6-week stimulation restored sensitivity to the ERKi. A representative of at least two independent experiments is shown for the immunoblots in this figure. In viability experiments, n = 3, mean ± s.e.m.
Figure 4. Fitness threshold model
Figure 4. Fitness threshold model
(a) The BRAF mRNA expression as a function of CN in 145 untreated BRAFV600E-mutant melanomas. The data were obtained from TCGA. The dotted area represents tumors at risk for selective propagation of BRAFamp during drug treatment. (b, c) A375 cells were stimulated with increasing concentrations of dox (24h), followed by treatment with ERK signaling inhibitors (RAFi: vemurafenib, 1 μM; MEKi: trametinib, 25 nM; or ERKi: SCH984, 500 nM) for 1h (b) or 72h (c), to determine the effect on signaling (b, quantification of representative immunoblots of 2 independent experiments) or relative fitness (c, n = 3, mean ± s.e.m.). The effect on signaling was adjusted for the effect of dox alone. Relative fitness is the change in log(IC50) with increasing concentrations of dox. (d) A schematic representation of the fitness threshold model. Sequential monotherapy is predicted to impose a selective gradient for the propagation of high level BRAFamp. In contrast, combination therapy is predicted to maximally elevate the fitness threshold, thus suppressing the selection and propagation of BRAFamp subclones. (e) Genomic DNA extracted from the patient biopsies before and after RAFi treatment (pre, post) or their derivative PDX models, before and after exposure to the ERKi, were sequenced to determine the BRAF CN. Diploid A375 cells were used as a control. (f) The duration of ERKi treatment response in patients who were either targeted therapy-naïve (pt. A and B) or pretreated with a RAF/MEKi combination (pt. C, D, E). See also Supplementary Table II. (g) The duration of treatment response in lung cancer patients treated first with a RAFi (left) followed by the addition of a MEKi (right). By comparison, the RAF/MEKi combination therapy has nearly a 60% response rate in treatment-native lung cancer patients. PR: partial response, SD: stable disease, PD: progressive disease.
Figure 5. Identification of a treatment to…
Figure 5. Identification of a treatment to suppress the evolution of BRAF amplified clones
(a) Immunoblot analysis (n = 2 independent experiments) of dox-induced A375 cells treated as shown (doses as in Fig. 4b) for 24h to determine the effect on ERK signaling intermediates. (b) As in a, but cells were treated for 72h to determine the effect on viability (n = 3, mean ± s.e.m). (c, d) Mice bearing PDX1D (c) or PDX1E (d) were treated with dabrafenib (RAFi), trametinib and/or SCH984 daily for 14 days followed by discontinuation of treatment to determine the effect on tumor growth (n = 5 mice, mean ± s.e.m). A vemurafenib analogue (PLX4720), alone or in combination, had a similar effect to dabrafenib (see below). Mice treated with the MEK/ERKi combination experienced significant toxicity leading to discontinuation of the experiment in (d). (e) Tumors that regrew after discontinuation of drug treatment from (c) were analyzed to determine BRAF CN by sequencing and BRAF protein expression by immunoblot analysis.
Figure 6. An intermittent combination treatment inhibits…
Figure 6. An intermittent combination treatment inhibits tumor growth in lung cancer and melanoma BRAFV600E PDX models
(a, b) A schematic representation (a) of several three-drug combination treatment schedules and their effect on the growth of PDX1D tumors in athymic mice (b, n = 5 mice, mean ± s.e.m, RAFi: vemurafenib analogue PLX4270, MEKi: trametinib, ERKi: SCH984). Treatment related toxicity was determined by monitoring animal weight or mortality. Mice treated on Schedule 5 remained free of tumor for up to 180 days after drug discontinuation. (c) Additional optimization of the off-drug interval in order to minimize toxicity, while retaining maximal tumor growth inhibition. (d) The expression of total BRAF in the PDX models was determined using mass spectrometry (n = 3, mean ± s.e.m). (e) The profile of genetic alterations in the BRAFV600E PDX models utilized in this study. (f) Effect of the intermittent regimen in a model with de-novo insensitivity to ERKi treatment. (g) The effect of the intermittent three drug combination treatment (administered on a 3/7-day schedule) in lung and melanoma PDX models (n = 5 mice, for each untreated or treated arm, mean ± range; ns: p>0.05; primary data are shown in Supplementary Fig. 6).

References

    1. Cancer Genome Atlas Research, N. Comprehensive molecular profiling of lung adenocarcinoma. Nature. 2014;511:543–550. doi: 10.1038/nature13385.
    1. Davies H, et al. Mutations of the BRAF gene in human cancer. Nature. 2002;417:949–954. doi: 10.1038/nature00766.
    1. Flaherty KT, et al. Inhibition of mutated, activated BRAF in metastatic melanoma. The New England journal of medicine. 2010;363:809–819. doi: 10.1056/NEJMoa1002011.
    1. Planchard D, et al. Dabrafenib in patients with BRAF(V600E)-positive advanced non-small-cell lung cancer: a single-arm, multicentre, open-label, phase 2 trial. The Lancet Oncology. 2016;17:642–650. doi: 10.1016/S1470-2045(16)00077-2.
    1. Flaherty KT, et al. Combined BRAF and MEK inhibition in melanoma with BRAF V600 mutations. The New England journal of medicine. 2012;367:1694–1703. doi: 10.1056/NEJMoa1210093.
    1. Larkin J, et al. Combined vemurafenib and cobimetinib in BRAF-mutated melanoma. The New England journal of medicine. 2014;371:1867–1876. doi: 10.1056/NEJMoa1408868.
    1. Long GV, et al. Combined BRAF and MEK inhibition versus BRAF inhibition alone in melanoma. The New England journal of medicine. 2014;371:1877–1888. doi: 10.1056/NEJMoa1406037.
    1. Planchard D, et al. Dabrafenib plus trametinib in patients with previously treated BRAF(V600E)-mutant metastatic non-small cell lung cancer: an open-label, multicentre phase 2 trial. The Lancet Oncology. 2016;17:984–993. doi: 10.1016/S1470-2045(16)30146-2.
    1. Poulikakos PI, et al. RAF inhibitor resistance is mediated by dimerization of aberrantly spliced BRAF(V600E) Nature. 2011;480:387–390. doi: 10.1038/nature10662.
    1. Shi H, et al. Melanoma whole-exome sequencing identifies (V600E)B-RAF amplification-mediated acquired B-RAF inhibitor resistance. Nature communications. 2012;3:724. doi: 10.1038/ncomms1727.
    1. Das Thakur M, et al. Modelling vemurafenib resistance in melanoma reveals a strategy to forestall drug resistance. Nature. 2013;494:251–255. doi: 10.1038/nature11814.
    1. Shi H, et al. Acquired resistance and clonal evolution in melanoma during BRAF inhibitor therapy. Cancer discovery. 2014;4:80–93. doi: 10.1158/-13-0642.
    1. Lito P, Rosen N, Solit DB. Tumor adaptation and resistance to RAF inhibitors. Nature medicine. 2013;19:1401–1409. doi: 10.1038/nm.3392.
    1. Bollag G, et al. Clinical efficacy of a RAF inhibitor needs broad target blockade in BRAF-mutant melanoma. Nature. 2010;467:596–599. doi: 10.1038/nature09454.
    1. Lito P, et al. Relief of profound feedback inhibition of mitogenic signaling by RAF inhibitors attenuates their activity in BRAFV600E melanomas. Cancer cell. 2012;22:668–682. doi: 10.1016/j.ccr.2012.10.009.
    1. Lipinski KA, et al. Cancer Evolution and the Limits of Predictability in Precision Cancer Medicine. Trends in cancer. 2016;2:49–63. doi: 10.1016/j.trecan.2015.11.003.
    1. McGranahan N, Swanton C. Biological and therapeutic impact of intratumor heterogeneity in cancer evolution. Cancer cell. 2015;27:15–26. doi: 10.1016/j.ccell.2014.12.001.
    1. Merlo LM, Pepper JW, Reid BJ, Maley CC. Cancer as an evolutionary and ecological process. Nature reviews Cancer. 2006;6:924–935. doi: 10.1038/nrc2013.
    1. Baslan T, et al. Genome-wide copy number analysis of single cells. Nature protocols. 2012;7:1024–1041. doi: 10.1038/nprot.2012.039.
    1. Garvin T, et al. Interactive analysis and assessment of single-cell copy-number variations. Nature methods. 2015;12:1058–1060. doi: 10.1038/nmeth.3578.
    1. Navin N, et al. Tumour evolution inferred by single-cell sequencing. Nature. 2011;472:90–94. doi: 10.1038/nature09807.
    1. Morris EJ, et al. Discovery of a novel ERK inhibitor with activity in models of acquired resistance to BRAF and MEK inhibitors. Cancer discovery. 2013;3:742–750. doi: 10.1158/-13-0070.
    1. Wong DJ, et al. Antitumor activity of the ERK inhibitor SCH772984 [corrected] against BRAF mutant, NRAS mutant and wild-type melanoma. Molecular cancer. 2014;13:194. doi: 10.1186/1476-4598-13-194.
    1. Baslan T, et al. Optimizing sparse sequencing of single cells for highly multiplex copy number profiling. Genome research. 2015;25:714–724. doi: 10.1101/gr.188060.114.
    1. Weir BA, et al. Characterizing the cancer genome in lung adenocarcinoma. Nature. 2007;450:893–898. doi: 10.1038/nature06358.
    1. Yao Z, et al. BRAF Mutants Evade ERK-Dependent Feedback by Different Mechanisms that Determine Their Sensitivity to Pharmacologic Inhibition. Cancer cell. 2015;28:370–383. doi: 10.1016/j.ccell.2015.08.001.
    1. Joseph EW, et al. The RAF inhibitor PLX4032 inhibits ERK signaling and tumor cell proliferation in a V600E BRAF-selective manner. Proceedings of the National Academy of Sciences of the United States of America. 2010;107:14903–14908. doi: 10.1073/pnas.1008990107.
    1. Kim KB, et al. Phase II study of the MEK1/MEK2 inhibitor Trametinib in patients with metastatic BRAF-mutant cutaneous melanoma previously treated with or without a BRAF inhibitor. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2013;31:482–489. doi: 10.1200/JCO.2012.43.5966.
    1. Corcoran RB, et al. EGFR-mediated re-activation of MAPK signaling contributes to insensitivity of BRAF mutant colorectal cancers to RAF inhibition with vemurafenib. Cancer discovery. 2012;2:227–235. doi: 10.1158/-11-0341.
    1. Prahallad A, et al. Unresponsiveness of colon cancer to BRAF(V600E) inhibition through feedback activation of EGFR. Nature. 2012;483:100–103. doi: 10.1038/nature10868.
    1. Shi H, et al. A novel AKT1 mutant amplifies an adaptive melanoma response to BRAF inhibition. Cancer discovery. 2014;4:69–79. doi: 10.1158/-13-0279.
    1. Sun C, et al. Reversible and adaptive resistance to BRAF(V600E) inhibition in melanoma. Nature. 2014;508:118–122. doi: 10.1038/nature13121.
    1. Maertens O, et al. Elucidating distinct roles for NF1 in melanomagenesis. Cancer discovery. 2013;3:338–349. doi: 10.1158/-12-0313.
    1. Johannessen CM, et al. A melanocyte lineage program confers resistance to MAP kinase pathway inhibition. Nature. 2013;504:138–142. doi: 10.1038/nature12688.
    1. Anderson K, et al. Genetic variegation of clonal architecture and propagating cells in leukaemia. Nature. 2011;469:356–361. doi:310.1038/nature09650. Epub 02010 Dec 09615.
    1. Melchor L, et al. Single-cell genetic analysis reveals the composition of initiating clones and phylogenetic patterns of branching and parallel evolution in myeloma. Leukemia. 2014;28:1705–1715. doi:1710.1038/leu.2014.1713. Epub 2014 Jan 1713.
    1. Campbell PJ, et al. The patterns and dynamics of genomic instability in metastatic pancreatic cancer. Nature. 2010;467:1109–1113. doi:1110.1038/nature09460.
    1. Nguyen A, et al. PKLR promotes colorectal cancer liver colonization through induction of glutathione synthesis. The Journal of clinical investigation. 2016;126:681–694. doi: 10.1172/JCI83587.
    1. Poirier JT, et al. DNA methylation in small cell lung cancer defines distinct disease subtypes and correlates with high expression of EZH2. Oncogene. 2015;34:5869–5878. doi: 10.1038/onc.2015.38.
    1. Einarsdottir BO, et al. Melanoma patient-derived xenografts accurately model the disease and develop fast enough to guide treatment decisions. Oncotarget. 2014;5:9609–9618. doi: 10.18632/oncotarget.2445.
    1. Cheng DT, et al. Memorial Sloan Kettering-Integrated Mutation Profiling of Actionable Cancer Targets (MSK-IMPACT): A Hybridization Capture-Based Next-Generation Sequencing Clinical Assay for Solid Tumor Molecular Oncology. The Journal of molecular diagnostics : JMD. 2015;17:251–264. doi: 10.1016/j.jmoldx.2014.12.006.
    1. Langmead B, Trapnell C, Pop M, Salzberg SL. Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome biology. 2009;10:R25. doi: 10.1186/gb-2009-10-3-r25.
    1. Li H, Durbin R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics. 2009;25:1754–1760. doi: 10.1093/bioinformatics/btp324.
    1. Gao R, et al. Punctuated copy number evolution and clonal stasis in triple-negative breast cancer. Nature genetics. 2016;48:1119–1130. doi: 10.1038/ng.3641.
    1. Wanjala J, et al. Identifying actionable targets through integrative analyses of GEM model and human prostate cancer genomic profiling. Molecular cancer therapeutics. 2015;14:278–288. doi: 10.1158/1535-7163.MCT-14-0542-T.
    1. Hembrough T, et al. Application of selected reaction monitoring for multiplex quantification of clinically validated biomarkers in formalin-fixed, paraffin-embedded tumor tissue. The Journal of molecular diagnostics : JMD. 2013;15:454–465. doi: 10.1016/j.jmoldx.2013.03.002.
    1. Catenacci DV, et al. Absolute quantitation of Met using mass spectrometry for clinical application: assay precision, stability, and correlation with MET gene amplification in FFPE tumor tissue. PloS one. 2014;9:e100586. doi: 10.1371/journal.pone.0100586.
    1. Lito P, Solomon M, Li LS, Hansen R, Rosen N. Allele-specific inhibitors inactivate mutant KRAS G12C by a trapping mechanism. Science. 2016;351:604–608. doi: 10.1126/science.aad6204.

Source: PubMed

3
購読する