Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2020 Mar 20;11(3):331.
doi: 10.3390/genes11030331.

Kinome Profiling of NF1-Related MPNSTs in Response to Kinase Inhibition and Doxorubicin Reveals Therapeutic Vulnerabilities

Affiliations

Kinome Profiling of NF1-Related MPNSTs in Response to Kinase Inhibition and Doxorubicin Reveals Therapeutic Vulnerabilities

Jamie L Grit et al. Genes (Basel). .

Abstract

Neurofibromatosis Type 1 (NF1)-related Malignant Peripheral Nerve Sheath Tumors (MPNST) are highly resistant sarcomas that account for significant mortality. The mechanisms of therapy resistance are not well-understood in MPNSTs, particularly with respect to kinase inhibition strategies. In this study, we aimed to quantify the impact of both the genomic context and targeted therapy on MPNST resistance using reverse phase phosphoproteome array (RPPA) analysis. We treated tumorgrafts from three genetically engineered mouse models using MET (capmatinib) and MEK (trametinib) inhibitors and doxorubicin, and assessed phosphosignaling at 4 h, 2 days, and 21 days. Baseline kinase signaling in our mouse models recapitulated an MET-addicted state (NF1-MET), P53 mutation (NF1-P53), and HGF overexpression (NF1). Following perturbation with the drug, we observed broad and redundant kinome adaptations that extended well beyond canonical RAS/ERK or PI3K/AKT/mTOR signaling. MET and MEK inhibition were both associated with an initial inflammatory response mediated by kinases in the JAK/STAT pathway and NFkB. Growth signaling predominated at the 2-day and 21-day time points as a result of broad RTK and intracellular kinase activation. Interestingly, AXL and NFkB were strongly activated at the 2-day and 21-day time points, and tightly correlated, regardless of the treatment type or genomic context. The degree of kinome adaptation observed in innately resistant tumors was significantly less than the surviving fractions of responsive tumors that exhibited a latency period before reinitiating growth. Lastly, doxorubicin resistance was associated with kinome adaptations that strongly favored growth and survival signaling. These observations confirm that MPNSTs are capable of profound signaling plasticity in the face of kinase inhibition or DNA damaging agent administration. It is possible that by targeting AXL or NFkB, therapy resistance can be mitigated.

Keywords: MEK; MET; MPNST; NF1; capmatinib; doxorubicin; kinase; kinome adaptation; kinome reprogramming; tram.

PubMed Disclaimer

Conflict of interest statement

The authors declare no conflicts of interest.

Figures

Figure 1
Figure 1
MET inhibition reveals differential innate and adaptive kinome reprogramming. Individual tumor growth curves for (A) Neurofibromatosis Type 1 (NF1)-MET, (B) NF1-P53, and (C) NF1 tumorgrafts plotted by treatment (colored lines) compared to the vehicle (black lines). The analysis of tumor growth data was previously reported [6]. The annotated tumors were analyzed by a reverse phase phosphoproteome array (RPPA) (DF). The fold change relative to the mean protein expression of control tumors (i.e., #1–3) was calculated for each tumor #4–6, with the first column of Panel A at 21 days corresponding to tumor #4, the second to tumor #5, and the third to tumor #6. Ranked balloon plots of the proteins with the highest and lowest fold change in expression after 4-h, 2-day, and 21-day treatment of the NF1-MET model with capmatinib. Each column represents a single animal. Balloon color indicates the fold change in expression relative to the vehicle mean (n = 3) for that time point. Balloon size indicates the absolute protein expression normalized to the total protein input and background.
Figure 2
Figure 2
MEK inhibition reveals differential innate and adaptive kinome reprogramming. Individual tumor growth curves for (A) NF1-MET, (B) NF1-P53, and (C) NF1 tumorgrafts plotted by treatment (colored lines) compared to the vehicle (black lines). The analysis of tumor growth data was previously reported [6]. The annotated tumors were analyzed by RPPA (DF). The fold change relative to the mean protein expression of control tumors (i.e., #1–3) was calculated for each tumor #4–6, with the first column of Panel A at 21 days corresponding to tumor #4, the second to tumor #5, and the third to tumor #6. Ranked balloon plots of the proteins with the highest and lowest fold change in expression after 4-h, 2-day, and 21-day treatment of the NF1-MET model with trametinib. Each column represents a single animal. Balloon color indicates fold change in expression relative to the vehicle mean (n = 3) for that time point. Balloon size indicates the absolute protein expression normalized to the total protein input and background.
Figure 3
Figure 3
Combination MEK and MET inhibition reveals differential innate and adaptive kinome reprogramming. Individual tumor growth curves for (A) NF1-MET, (B) NF1-P53, and (C) NF1 tumorgrafts plotted by treatment (colored lines) compared to the vehicle (black lines). The analysis of tumor growth data was previously reported [6]. The annotated tumors were analyzed by RPPA (DF). The fold change relative to the mean protein expression of control tumors (i.e., #1–3) was calculated for each tumor #4–6, with the first column of Panel A at 21 days corresponding to tumor #4, the second to tumor #5, and the third to tumor #6. Ranked balloon plots of the proteins with the highest and lowest fold change in expression after 4-h, 2-day, and 21-day treatment of the NF1-MET model with combination therapy. Each column represents a single animal. Balloon color indicates the fold change in expression relative to the vehicle mean (n = 3) for that time point. Balloon size indicates the absolute protein expression normalized to the total protein input and background.
Figure 4
Figure 4
Doxorubicin reveals differential innate and adaptive kinome reprogramming. Individual tumor growth curves for (A) NF1-MET, (B) NF1-P53, and (C) NF1 tumorgrafts plotted by treatment (colored lines) compared to the vehicle (black lines). The annotated tumors were analyzed by RPPA (DF). The fold change relative to the mean protein expression of control tumors (i.e., #1–3) was calculated for each tumor #4–6, with the first column of Panel A at 21 days corresponding to tumor #4, the second to tumor #5, and the third to tumor #6. Ranked balloon plots of the proteins with the highest and lowest fold change in expression after 4-h, 2-day, and 21-day treatment of the NF1-MET model with doxorubicin. Each column represents a single animal. Balloon color indicates the fold change in expression relative to the vehicle mean (n = 3) for that time point. Balloon size indicates the absolute protein expression normalized to the total protein input and background.
Figure 5
Figure 5
Combined doxorubicin, MET, and MEK inhibitor treatment reduces the response heterogeneity. Tumor growth of (A) NF-MET and (D) tumorgrafts are plotted as means with standard errors. 95% confidence intervals for the pairwise differences between the growth rates of the select treatments in the (B) NF1-MET and (E) NF1-P53 tumors, estimated and tested using linear mixed-effects models with random slopes and intercepts, and false discovery rate-adjusted contrasts. Statistically significant differences (p-value < 0.05) between compared therapies are highlighted in red. Individual tumor growth curves for (C) NF1-MET and (F) NF1-P53 tumorgrafts plotted by treatment (colored lines) compared to the vehicle (black lines). The analysis of tumor growth data and differences in treatment response were previously reported for single-agent treatment of capmatinib and trametinib, and combination treatment of capmatinib + trametinib [6].
Figure 5
Figure 5
Combined doxorubicin, MET, and MEK inhibitor treatment reduces the response heterogeneity. Tumor growth of (A) NF-MET and (D) tumorgrafts are plotted as means with standard errors. 95% confidence intervals for the pairwise differences between the growth rates of the select treatments in the (B) NF1-MET and (E) NF1-P53 tumors, estimated and tested using linear mixed-effects models with random slopes and intercepts, and false discovery rate-adjusted contrasts. Statistically significant differences (p-value < 0.05) between compared therapies are highlighted in red. Individual tumor growth curves for (C) NF1-MET and (F) NF1-P53 tumorgrafts plotted by treatment (colored lines) compared to the vehicle (black lines). The analysis of tumor growth data and differences in treatment response were previously reported for single-agent treatment of capmatinib and trametinib, and combination treatment of capmatinib + trametinib [6].
Figure 6
Figure 6
ERK reactivation or pathway indifference drive resistance to kinase inhibition. Phospho-ERK1/2 T202/Y204 expression in each genetic model after 21 days of (A) vehicle, (B) capmatinib, (C) trametinib, or (D) combination treatment. (E) phospho-ERK1/2 T202/Y204 expression values were measured by RPPA and calculated as the absolute protein expression normalized to the total protein input and background. Points represent the mean (n = 3) for each treatment-genotype-time group. Shaded bars represent +/− SEM.
Figure 6
Figure 6
ERK reactivation or pathway indifference drive resistance to kinase inhibition. Phospho-ERK1/2 T202/Y204 expression in each genetic model after 21 days of (A) vehicle, (B) capmatinib, (C) trametinib, or (D) combination treatment. (E) phospho-ERK1/2 T202/Y204 expression values were measured by RPPA and calculated as the absolute protein expression normalized to the total protein input and background. Points represent the mean (n = 3) for each treatment-genotype-time group. Shaded bars represent +/− SEM.
Figure 7
Figure 7
AXL Y702 and NFkB p65 S536 phosphorylation are highly correlated. Expression of phoshpo-AXL Y702 and phospho-NFkB p65 S536 plotted by (A) time and genotype group or (B) time and treatment group. Colors indicate treatment time and point shape indicates treatment or genotype groups. Lines indicate loess-predicted fit for each time point; shaded regions indicate 95% confidence intervals. Spearmen’s rank correlation rho = 0.832, 0.835, and 0.881 with p value = 3.72 × 10−9, 9.75 × 10−13, and 6.40 × 10−15 for the 4-h, 2-day, and-21 day groups, respectively.
Figure 7
Figure 7
AXL Y702 and NFkB p65 S536 phosphorylation are highly correlated. Expression of phoshpo-AXL Y702 and phospho-NFkB p65 S536 plotted by (A) time and genotype group or (B) time and treatment group. Colors indicate treatment time and point shape indicates treatment or genotype groups. Lines indicate loess-predicted fit for each time point; shaded regions indicate 95% confidence intervals. Spearmen’s rank correlation rho = 0.832, 0.835, and 0.881 with p value = 3.72 × 10−9, 9.75 × 10−13, and 6.40 × 10−15 for the 4-h, 2-day, and-21 day groups, respectively.

Similar articles

Cited by

References

    1. Konieczkowski D.J., Johannessen C.M., Garraway L.A. A Convergence-Based Framework for Cancer Drug Resistance. Cancer Cell. 2018;33:801–815. doi: 10.1016/j.ccell.2018.03.025. - DOI - PMC - PubMed
    1. Lammert M., Friedman J.M., Kluwe L., Mautner V.F. Prevalence of neurofibromatosis 1 in German children at elementary school enrollment. Arch. Dermatol. 2005;141:71–74. doi: 10.1001/archderm.141.1.71. - DOI - PubMed
    1. Evans D.G., Howard E., Giblin C., Clancy T., Spencer H., Huson S.M., Lalloo F. Birth incidence and prevalence of tumor-prone syndromes: Estimates from a UK family genetic register service. Am. J. Med. Genet. A. 2010;152:327–332. doi: 10.1002/ajmg.a.33139. - DOI - PubMed
    1. Upadhyaya M., Spurlock G., Majounie E., Griffiths S., Forrester N., Baser M., Huson S.M., Gareth Evans D., Ferner R. The heterogeneous nature of germline mutations in NF1 patients with malignant peripheral serve sheath tumours (MPNSTs) Hum. Mutat. 2006;27:716. doi: 10.1002/humu.9429. - DOI - PubMed
    1. Miller S.J., Rangwala F., Williams J., Ackerman P., Kong S., Jegga A.G., Kaiser S., Aronow B.J., Frahm S., Kluwe L., et al. Large-Scale Molecular Comparison of Human Schwann Cells to Malignant Peripheral Nerve Sheath Tumor Cell Lines and Tissues. Cancer Res. 2006;66:2584–2591. doi: 10.1158/0008-5472.CAN-05-3330. - DOI - PubMed

Publication types

MeSH terms