Skip to main content
Erschienen in: Journal of Hematology & Oncology 1/2018

Open Access 01.12.2018 | Research

MET alterations detected in blood-derived circulating tumor DNA correlate with bone metastases and poor prognosis

verfasst von: Sadakatsu Ikeda, Maria Schwaederle, Mandakini Mohindra, Denis L. Fontes Jardim, Razelle Kurzrock

Erschienen in: Journal of Hematology & Oncology | Ausgabe 1/2018

Abstract

Background

We analyzed clinical associations of MET alterations in the plasma of patients with diverse malignancies.

Methods

Digital sequencing of circulating tumor DNA (ctDNA) (54–70 genes) was performed in 438 patients; 263 patients also had tissue sequencing (182–315 genes). The most represented tumor types were gastrointestinal (28.1%), brain (24.9%), and lung (23.2%). Most patients (71.2%) had recurrent/metastatic disease.

Results

MET alterations were observed in 31 patients (7.1%) and correlated with bone metastasis (P = 0.007), with TP53 (P = 0.001) and PTEN (P = 0.003) abnormalities, and with an increased number of alterations (median, 4 vs 1, P = 0.001) (all multivariable analyses). Patients with MET alterations demonstrated a significantly shorter median time to metastasis/recurrence (1.0 vs 10.4 months, P = 0.044, multivariable) and a poorer survival (30.6 vs 58.4 months, P = 0.013, univariate only). Of the 31 patients with MET alterations, 18 also had tissue testing; only two also had tissue MET alterations (11.1%); MET alterations were detected at a lower frequency in tissue (1.14%) compared to ctDNA (7.1%), with P = 0.0002.

Conclusions

In conclusion, the detection of MET alterations by liquid biopsy is feasible. MET ctDNA alterations were associated with a poorer prognosis, higher numbers of genomic abnormalities, and bone metastases. The correlation with bone metastases may explain the higher frequency of MET alterations in blood ctDNA than in tissue (since bones are rarely biopsied) and the previous observations of bone-predominant responses to MET inhibitors. The high number of co-altered genes suggests that MET inhibitors may need to be combined with other agents to induce/optimize responses.
Hinweise

Electronic supplementary material

The online version of this article (https://​doi.​org/​10.​1186/​s13045-018-0610-8) contains supplementary material, which is available to authorized users.
Abkürzungen
95% CI
95% confidence interval
CAP
College of American Pathologists
ctDNA
Circulating tumor DNA
FDA
Food and Drug Administration
FISH
Fluorescent in situ hybridization
GAB1
GRB2-associated binding protein 1
GRB2
Growth factor receptor-bound protein 2
HGF
Hepatocyte growth factor
HR
Hazard ratio
NCCN
National Comprehensive Cancer Network
NGS
Next-generation sequencing
NSCLC
Non-small cell lung cancer
OS
Overall survival
PREDICT
Profile Related Evidence Determining Individualized Cancer Therapy
VAF
Variant allele fraction

Background

MET, also called c-MET or hepatocyte growth factor (HGF) receptor, is a receptor tyrosine kinase discovered as an oncogene in the 1980s [1, 2]. Independent research found that HGF or scatter factor (SF) was a ligand for MET [3]. Upon binding of HGF to MET, the kinase domain phosphorylates growth factor receptor-bound protein 2 (GRB2) and GRB2-associated binding protein 1 (GAB1) and activates diverse downstream signaling pathways important in cancer, including the ERK/MAPK, PI3K-Akt/PKB, Crk-Rap, and Rac-Pak pathways [4]. These pathways form distinct branches that interact to regulate cell proliferation, invasion, migration, angiogenesis, development, organ regeneration, and tumorigenesis [4].
Activating MET mutations are found in diverse human cancer [5]. For instance, activating mutations in the kinase domain are a feature of both hereditary and some non-hereditary forms of papillary renal cell carcinoma [6]. MET amplification is seen in 5 to 20% of non-small cell lung cancer (NSCLC) and gastric cancer [7]. MET overexpression correlates with poor treatment outcome in some malignancies [8].
Drugs that target MET include inhibitors of the HGF/-MET pathway, MET antibodies, and MET kinase inhibitors [8]. Cabozantinib and crizotinib are both Food and Drug Administration (FDA)-approved multikinase inhibitors that are also potent suppressors of MET [9, 10]. Crizotinib induced responses in some patients with highly MET-amplified lung cancer [11]. Studies with the multi-target MET inhibitor cabozantinib have shown significant activity against a variety of solid tumors including melanoma, as well as renal and non-small cell lung, liver, medullary thyroid, breast, and ovarian cancer, but this activity is likely due to other targets of cabozantinib, such as VEGFR or RET [9]. In the phase I setting, patients with MET amplification did not respond to MET inhibitors (but the number of treated patients was small) [5].
Because of their non-invasive nature, liquid biopsies are increasingly used in the clinical setting. Indeed, numerous studies showed a relatively good correlation with tissue sequencing and the potential to detect actionable alterations [1216]. In this study, we analyzed MET alterations in the plasma-derived circulating tumor DNA (ctDNA) of 438 patients with diverse malignancies and explored the relationship between MET alterations, demographics, as well as other molecular alterations and clinical outcomes.

Methods

Patients

We reviewed the clinicopathology and clinical outcomes of 438 consecutive patients with cancer for whom ctDNA testing had been performed and who were seen at the UC San Diego Moores Cancer Center from June 2014 to July 2016. Pathology was reviewed at UCSD. Data was abstracted from the electronic medical record. This study was performed and consents were obtained in accordance with the UCSD Institutional Review Board guidelines [PREDICT-UCSD (Profile Related Evidence Determining Individualized Cancer Therapy); NCT02478931].

Next-generation sequencing

Digital sequencing of ctDNA (DNA) in all patients was performed by Guardant Health, Inc. (Guardant360, Redwood City, California, http://​www.​guardanthealth.​com/​guardant360/​), a Clinical Laboratory Improvement Amendment (CLIA)-certified and College of American Pathologists (CAP)-accredited clinical laboratory. The analytical and clinical validation of Guardant360 was conducted in accordance with evidentiary standards established by the Standards for Reporting of Diagnostic Accuracy (STARD), REporting of tumor MARKer Studies (REMARK), Evaluation of Genomic Applications in Practice and Prevention (EGAPP), and the recent Next-generation Sequencing: Standardization of Clinical Testing (Nex-StoCT) biomarker guidelines [17]. As described in Lanman et al. [17], 5–30 ng of ctDNA was isolated from plasma (two 10 ml Streck tubes drawn for each patient) and sequencing libraries were prepared with custom in-line barcode molecular tagging and complete sequencing at 15,000× read depth (~ 4000 unique double-stranded cfDNA fragments, each represented by 3–5 sequencing reads). The panels utilize hybrid capture followed by NGS of the critical exons in a panel of 54–70 genes (Additional file 1: Table S1 to S3) and report all four major types of genomic alterations (point mutations, indels, fusions, and copy number amplifications). Post-sequencing bioinformatics matches the complementary strands of each barcoded DNA fragment to remove false positive results [17]. The variant allele fraction (VAF) is computed as the number of mutated DNA molecules divided by the total number (mutated plus wild type) of DNA fragments at that allele; VAF is reported as a percentage. The majority of cell-free DNA is wild type (germline); thus, the median VAF of somatic alterations is < 0.5%. The analytic sensitivity reaches detection of one to two single-mutant fragments from a 10-ml blood sample (0.1% limit of detection), and the analytic specificity is greater than 99.9999% [17].
For 144 patients, a 54-gene panel was used, which identified potential tumor-related alterations in 54 cancer-related genes (Additional file 1: Table S1) including copy number amplifications in ERBB2, EGFR, and MET (indels and fusions were not detected as part of this panel). For 272 patients, a 68-gene version of the original panel (expanded to all four major alteration types) was used, and for 22 patients, the most recent 70-gene panel version (further expanded to amplifications in 18 genes and fusions in 6 genes) was applied (Additional file 1: Table S2 and S3). Only non-synonymous alterations were included in our analysis.
In addition, 263 patients (~ 60%) of the 438 patients with ctDNA test also had CLIA/CAP-accredited next-generation sequencing (NGS) performed on tumor tissue (FoundationOne™, Cambridge, Massachusetts, http://​www.​foundationone.​com.) (N = 182 to 315 gene panels).

Statistical and outcome analysis

Patient characteristics were summarized using descriptive statistics. Medians and respective 95% confidence intervals and range were calculated, whenever possible. Associations between categorical variables were tested using a binary logistic regression model. Linear variables were tested using the Mann-Whitney U test for univariable analyses and a multiple linear regression model for multivariable analyses.
Time to metastasis/recurrence was defined as the time interval between diagnosis and first metastasis/recurrence (whichever came first) or last follow-up date (patients who had not recurred/developed metastases at last follow-up were censored on that date). Overall survival (OS) was defined as the time from diagnosis to death or last follow-up date for patients who were alive (patients still alive at the last follow-up were censored on that date). Estimations for the time to first metastasis/recurrence and OS were done using Kaplan-Meier analyses and were compared among subgroups by the log-rank test for univariable analysis or Cox regression models for multivariable analysis. All statistical analyses were performed by author MS with SPSS version 24.0.

Results

Patient demographic characteristics

The median age of patients at diagnosis was 57.5 years (CI 95%, 54.5–59.1). Women comprised 52.1% (N = 228) of the population. The majority of patients were Caucasian (69.2%, N = 303). The most represented tumor types were gastrointestinal (28.1%, N = 123), brain (24.9%, N = 109), lung (23.2%, N = 102), and breast (11.6%, N = 51) cancers. The majority of the patients had recurrent or metastatic disease at the time of blood draw used for testing (71.2%, N = 312) (Table 1).
Table 1
Demographics comparison of 438 patients with or without MET alterations
Characteristics
Total patients, N = 438 (100%)
MET wild type, N = 407 (92.9%)
MET alteration(s), N = 31 (7.1%)
P values*
Age at diagnosis (years) (median, CI 95%)
57.5 (54.5–59.1)
57.7 (55.3–59.2)
53.8 (49.0–62.2)
0.791
Gender
   
0.288
 Women
228 (52.1%)
209 (91.7%)
19 (8.3%)
 
 Men
210 (47.9%)
198 (94.3%)
12 (5.7%)
 
Ethnicity
 Caucasian
303 (69.2%)
284 (93.7%)
19 (6.3%)
0.326
 Asian
52 (11.9%)
46 (88.5%)
6 (11.5%)
0.188
 Hispanic
30 (6.8%)
28 (93.3%)
2 (6.7%)
0.928
 African American
9 (2.1%)
8 (88.9%)
1 (11.1%)
0.637
 Middle Eastern
5 (1.1%)
5 (100%)
0 (0%)
0.998
 Unknown
39 (8.9%)
36 (92.3%)
3 (7.7%)
0.875
Type of cancer
 Gastrointestinal
123 (28.1%)
118 (95.9%)
5 (4.1%)
0.132
 Brain
109 (24.9%)
107 (98.2%)
2 (1.8%)
0.026**
 Lung
102 (23.2%)
90 (88.2%)
12 (11.8%)
0.039**
 Breast
51 (11.6%)
45 (88.2%)
6 (11.8%)
0.172
 Genitourinary
18 (4.1%)
15 (83.3%)
3 (16.7%)
0.120
 Head and neck
10 (2.3%)
9 (90.0%)
1 (10.0%)
0.717
 Gynecologic
7 (1.6%)
6 (85.7%)
1 (14.3%)
0.465
 Melanoma
5 (1.1%)
4 (80.0%)
1 (20.0%)
0.285
 Hematologic
3 (0.7%)
3 (100%)
0 (0%)
0.998
 Othera
10 (2.3%)
10 (100%)
0 (0%)
0.998
Presence of metastasis or recurrence at the time of blood draw
 Yes
312 (71.2%)
284 (91.0%)
28 (9.0%)
0.024**
 No
126 (28.8%)
123 (97.6%)
3 (2.4%)
 
The percentages on the first column are expressed over the total number of patients (N = 438); for the second and third columns, percentages are expressed over the total number of patients for each variable
aLymphoma (n = 2), sarcoma (n = 2), thymoma (n = 2), desmoid tumor, neurofibromatosis, peripheral nerve sheath tumor, and carcinoma of unknown primary (each n = 1)
*P values were computed using the independent sample Mann-Whitney U test for linear variables (age at diagnostic) and the logistic binary regression analysis for categorical variables, as appropriate
**MET alterations were negatively associated with brain tumors but positively associated with lung tumors and metastasis or recurrence at the time of blood draw

MET alterations and associations with patient characteristics

Overall, MET alterations were observed in 31 of the 438 patients whose ctDNA was tested (7.1%). Sixteen patients had a MET amplification only; 13 had a somatic mutation only; and two had both an amplification and a somatic MET mutation (Fig. 1a).

Univariate analyses

MET alterations were significantly (positively) associated with lung cancers (11.8%) and the presence of metastasis/recurrence at the time of blood draw and negatively associated with brain tumors 1.8% (all P < 0.05) (Table 1). We then examined the associations with other genomic alterations and found that MET alterations correlated in univariable analysis with alterations in TP53, EGFR, PIK3CA, BRAF, ARID1A, ALK, and PTEN genes (all P < 0.05, Additional file 1: Table S4). We also investigated the potential associations between MET alterations and the location of metastatic sites and showed a significant correlation with lymph nodes and bone metastasis (both P < 0.02, Additional file 1: Table S4). Of note, patients with MET alterations had a median of two metastatic sites versus a single site for patients without MET alterations (P = 0.001). The latter conclusions remained unchanged if the patients with brain tumors, lymphoma/leukemia, thymoma, and NF were excluded, with P values of < 0.05 for association between MET alterations and lymph node or bone metastases and increased number of metastases.

Multivariate analyses

In the multiple logistic regression model (that included any parameters that were significant (P ≤ 0.05) in univariate analysis), the only variables that remained statistically associated with MET alterations were aberrations in TP53 (P = 0.001) and PTEN (P = 0.003) genes, as well as an increased incidence of bone metastasis (P = 0.007) (Table 2). The univariate association between MET alterations and lung cancer, the negative association with brain tumors, and the correlation with metastases at the time of blood draw did not remain significant in multivariate analysis. Multivariable analysis also demonstrated that MET alterations correlated with an increased number of alterations (median of 4 alterations vs 1, P = 0.001) (Table 3). These conclusions remained valid if the patients with brain tumors, lymphoma/leukemia, thymoma, and NF were excluded, with P values of < 0.01 for association between MET alterations and TP53 or PTEN alterations, bone metastases, and increased number of metastases.
Table 2
Multivariable analysis of the variables associated with MET alterations*
 
MET wild type, N = 407
MET altered, N = 31
Univariable
Multivariable
Characteristics
  
Wald
Odds ratio (CI 95%)
P value
Wald
Odds ratio (CI 95%)
P value
Type of cancer
 Brain (n = 109)**
107 (26.3%)
2 (6.5%)
4.94
0.19 (0.05–0.83)
0.026**
 Lung (n = 102)
90 (22.1%)
12 (38.7%)
4.26
2.22 (1.04–4.76)
0.039
Presence of metastasis or recurrence at the time of blood draw (n = 312)
284 (69.8%)
28 (90.3%)
5.13
4.0 (1.2–14.3)
0.024
Genetic alteration type
TP53 (n = 149)
129 (31.7%)
20 (64.5%)
12.2
3.85 (1.82–8.33)
< 0.001
10.9
3.7 (1.7–8.3)
0.001
EGFR (n = 53)
43 (10.6%)
10 (32.4%)
11.2
4.0 (1.79–9.1)
0.001
PIK3CA (n = 42)
35 (8.6%)
7 (22.6%)
5.93
3.1 (1.25–7.7)
0.015
BRAF (n = 27)
22 (5.4%)
5 (16.1%)
5.14
3.3 (1.2–10.0)
0.023
ARID1A (n = 19)
15 (3.7%)
4 (12.9%)
5.14
3.85 (1.2–12.5)
0.023
ALK (n = 14)
11 (2.7%)
3 (9.7%)
3.94
3.85 (1.02–14.3)
0.047
PTEN (n = 9)
6 (1.5%)
3 (9.7%)
7.72
7.1 (1.7–33.3)
0.007
9.12
11.1 (2.3–50.0)
0.003
Metastatic sites
 Lymph node (n = 139)
123 (30.2%)
16 (51.6%)
5.77
2.4 (1.2–5.3)
0.016
 Bone (n = 102)
88 (21.6%)
14 (45.2%)
8.26
2.9 (1.4–6.3)
0.004
7.34
2.9 (1.35–6.25)
0.007
 No metastases (n = 147)**
145 (35.6%)
2 (6.5%)
7.96
0.13 (0.03–0.5)
0.005**
The Wald statistics test the unique contribution of each variable; the higher the Wald statistics, the higher the association/contribution in the model
*Only variables that were significant in the univariable models (logistic regression) were included in the multivariable analysis, with the final model containing only significant covariates in the multivariable analyses (forward stepwise selection model)
**“Brain tumors” and “no metastases” were negatively associated with MET alterations in univariate analysis. These variables were not significant in the final multivariate analysis model
Table 3
Association with the number of alterations*
Variables
Median N of alterations (95% CI) (range)
P value (univariable)
t statistic (multivariable)
P value (multivariable)
Overall
1 (1–1) (0–26)
   
Tumor type
 Gastrointestinal
2 (1–2) (0–26)
0.031
 Brain
0 (0–0) (0–5)
< 0.001
 Lung
2 (2–3) (0–21)
< 0.001
Genomic alterations
MET
4 (3–6) (1–21)
< 0.001
3.35
0.001
TP53
3 (3–3) (1–21)
< 0.001
6.86
< 0.001
EGFR
3 (3–4) (1–26)
< 0.001
PIK3CA
5 (4–6) (1–26)
< 0.001
7.31
< 0.001
BRAF
5 (4–6) (1–26)
< 0.001
6.54
< 0.001
KRAS
4 (3–4) (1–26)
< 0.001
6.01
< 0.001
MYC
5 (3–6) (2–7)
< 0.001
2.70
0.007
Metastatic/recurrence at the time of blood draw
2 (1–2) (0–26)
< 0.001
Metastatic sites
 Lymph node
2 (2–3) (0–21)
< 0.001
2.05
0.041
 Bone
2 (2–3) (0–26)
< 0.001
 Liver
2 (2–3) (0–26)
< 0.001
 Lung
2 (1–3) (0–19)
0.010
 Brain
2 (1–3) (0–21)
< 0.001
 Adrenal
2 (2–4) (0–20)
< 0.001
2.06
0.040
*Only variables with ≥ 20 patients in the overall population were tested. Only significant variables in the univariable analysis (non-parametric Mann-Whitney U test) were included in the multivariable model (multiple linear regression), with the final model containing only significant covariates (stepwise model selection). The t statistics test the unique contribution of each variable; the higher the t statistics, the higher the association/contribution in the model
The other variables that were significantly associated with a higher number of alterations in the multivariable analysis were alterations in TP53, PIK3CA, BRAF, KRAS, or MYC genes as well as the presence of metastasis in the lymph node or adrenal tissue (Table 3).

MET alterations and survival outcomes

Overall survival

Overall, 213 patients (213/438, 48.6%) had died at the time of our analysis (20/31 with MET alterations; 193/407 without MET alterations detected in their ctDNA). A log rank test demonstrated a poorer survival (time from diagnosis until death or last follow-up date) for patients bearing MET alterations (30.6 months vs 58.4, P = 0.013). In the multivariable analysis, only lung cancer as well as the presence of ARID1A, KRAS, ALK, and MYC alterations and liver metastasis remained significant predictors of a poorer survival (all P < 0.50, Fig. 1b) (Table 4).
Table 4
Overall survival analysis (N = 438 patients)*
 
Univariable
Multivariable
Characteristics
Median time (months) (std. error)
P value
Wald
Hazard ratio
(CI 95%)
P value
Type of cancer
 Gastrointestinal (n = 123) vs not
36.3 (4.5) vs 71.6 (6.5)
0.002
 Brain (n = 109) vs not**
95.0 (50.1) vs 51.5 (5.9)
0.003
 Lung (n = 102) vs not
31.8 (6.0) vs 61.0 (7.4)
0.002
9.59
1.7 (1.21–4.04)
0.002
 Breast (n = 51) vs not
104.8 (20.3) vs 50.0 (5.6)
0.027
   
Genetic alteration type
MET (n = 31) vs not
30.6 (6.2) vs 58.4 (6.3)
0.013
TP53 (n = 149) vs not
42.1 (6.1) vs 66.5 (10.3)
0.003
PIK3CA (n = 42) vs not
43.2 (10.6) vs (58.4 (6.3)
0.013
ARID1A (n = 19) vs not
24.8 (6.2) vs 58.4 (6.8)
0.002
7.98
2.7 (1.25–4.0)
0.005
KRAS (n = 50) vs not
18.3 (7.3) vs 59.7 (6.5)
< 0.001
15.68
2.1 (1.5–3.12)
< 0.001
ALK (n = 14) vs not
31.3 (9.0) vs 58.3 (6.7)
0.015
5.76
2.04 (1.14–3.7)
0.016
SMAD4 (n = 12) vs not
21.7 (8.2) vs 58.3 (6.2)
0.007
MYC (n = 20) vs not
22.2 (3.9) vs 59.1 (6.5)
< 0.001
7.59
2.17 (1.24–3.77)
0.006
Metastatic/recurrence at the time of blood draw vs not
53.6 (4.9) vs NR
0.011
Metastatic sites
 Liver (n = 111) vs not
49.3 (8.2) vs 59.1 (7.9)
0.023
4.85
1.4 (1.03–1.89)
0.028
 Lymph node (n = 139) vs not
39.9 (7.4) vs 74.1 (11.1)
< 0.001
Only variables that were significant in the univariable models (log rank test) were included in the multivariable analysis (Cox regression model), with the final model containing only significant covariates in the multivariable analyses (forward stepwise selection model). The Wald statistics test the unique contribution of each variable; the higher the Wald statistics, the higher the association/contribution in the model
NR not reached
*Overall survival was defined as the time from diagnosis to death or last follow-up date
**Brain tumors (N = 109) included n = 50 glioblastoma cases, while the rest of the tumors were lower grade astrocytomas or other lower grade brain tumors

Time to metastasis/recurrence

Patients with MET alterations had a significantly shorter median time to metastasis/recurrence, with a median of 1.0 months (95%CI could not be computed) versus 10.4 months (95%CI 6.9–14.0) (P = 0.003) (Table 5). MET alterations remained significantly associated with a shorter time to metastasis/recurrence in a multivariable analysis (P = 0.044), along with the presence of liver (P = 0.022) or lymph node metastases (P < 0.001). Inversely, breast cancer correlated with a longer time to metastasis/recurrence (P < 0.001) (Fig. 1b and Table 5).
Table 5
Time to metastasis/recurrence from diagnosis (N = 438 patients)*
 
Univariable
Multivariable
Characteristics
Median time (months) (95% CI)
P value
Wald
Hazard ratio
(CI 95%)
P value
Type of cancer
 Gastrointestinal (n = 123) vs not
0.67 (0–1.9) vs 14.1 (8.3–19.8)
< 0.001
 Brain (n = 109) vs not
42.8 (19.0–66.7) vs 2.5 (0.49–4.4)
< 0.001
 Lung (n = 102) vs not
0.6 (0–1.3) vs 14.0 (9.5–18.6)
< 0.001
 Breast (n = 51) vs not
30.4 (14.3–46.4) vs 7.0 (3.8–10.2)
0.001
40.03
0.33 (0.23–0.47)
< 0.001
 Genitourinary (n = 18) vs not
0 (−) vs 10.1 (6.8–13.4)
0.020
 Head and neck (n = 10) vs not
0 (−) vs 9.8 (6.2–13.4)
0.015
Genetic alteration type
MET (n = 31) vs not
1.0 (−) vs 10.4 (6.9–14.0)
0.003
4.05
1.47 (1.01–2.13)
0.044
TP53 (n = 149) vs not
4.1 (0.5–7.7) vs 12.7 (6.1–19.3)
0.001
EGFR (n = 53) vs not
3.6 (0–8.2) vs 11.4 (7.9–14.8)
0.003
ARID1A (n = 19) vs not
3.6 (0–10.1) vs 10.0 (6.3–13.7)
0.013
KRAS (n = 50) vs not
0 (−) vs 11.4 (8.3–13.7)
< 0.001
SMAD4 (n = 12) vs not
0 (−) vs 10.1 (6.7–13.4)
0.040
MYC (n = 20) vs not
0.2 (0–0.75) vs 10.4 (7.0–13.9)
< 0.001
Metastatic sites
 Adrenal (n = 29) vs not
0 (−--) vs 11.3 (8.0–14.6)
0.002
 Bone (n = 102) vs not
1.5 (0–6.7) vs 11.4 (8.2–14.5)
0.049
 Liver (n = 111) vs not
0.9 (0–2.6) vs 132.3 (9.7–16.9)
< 0.001
10.04
1.49 (1.16–1.92)
0.022
 Lymph node (n = 139) vs not
0.3 (−) vs 20.1 (14.3–25.9)
< 0.001
13.41
1.54 (1.22–1.92)
< 0.001
 Peritoneal (n = 49) vs not
0 (−) vs 11.3 (8.1–14.5)
< 0.001
 Brain (n = 63) vs not
3.6 (0–7.8) vs 10.4 (7.0–13.9)
0.045
 Lung (n = 90) vs not
1.2 (0–8.0) vs 11.3 (7.7–15.0)
0.003
The Wald statistics test the unique contribution of each variable; the higher the Wald statistics, the higher the association/contribution in the model
*Only variables that were significant in the univariable models (log rank test) were included in the multivariable analysis (Cox regression model), with the final model containing only significant covariates in the multivariable analyses (forward stepwise selection model). For some values, the 95% CI could not be computed (−)

Comparison with tissue testing

As noted, 438 patients had ctDNA testing; 263 of these patients also had tissue NGS performed.
Of the 31 patients with MET alterations in ctDNA, 18 also had tissue testing (Foundation Medicine see the “Methods” section). The median time interval between the blood draw and the tissue biopsy for these 18 patients was 6.1 months (95% CI (2–13.7); range (0.2–32.6)). In most of the patients with both types of testing, the ctDNA test was performed after the tissue testing (16/18 cases). Only two patients who had a MET alteration identified in their ctDNA also had a MET alteration found in their tissue testing (11.1%; MET amplification and MET Y501C, one patient each). In these two patients, the time interval between the ctDNA and tissue biopsies was 1.8 and 15.3 months, and both tissues used for the testing were from the primary tumor.
In only one patient, a MET amplification was detected in the tissue and not in ctDNA (1 of 263 total patients who had NGS tissue testing). The tissue test was performed on a pancreatic tumor that was surgically removed, and the ctDNA test was done more than 1 year later (recent scans showed appearance of new pulmonary and liver nodules 2 months prior the ctDNA testing).
Overall, MET alterations were detected at a significantly lower frequency in tissue (3/263 patients, 1.14%) compared to ctDNA (31/438, 7.1%), P = 0.0002. Further, of eight patients who harbored MET alterations in ctDNA, and had ctDNA and tissue testing within 2 months of each other, only one patient showed a similar MET alteration in the tissue. Six of the seven patients with only ctDNA positive for MET alterations had bone metastases.

Discussion

This is the largest study interrogating the feasibility and utility of MET alteration detection through blood-derived ctDNA. Liquid biopsy is a non-invasive method to find genomic aberrations and is increasingly utilized in the clinical setting as reflected by the non-small cell lung cancer National Comprehensive Cancer Network (NCCN) guidelines [18, 19]. Our study demonstrated that MET ctDNA alterations were detected in 7.1% of patients with solid tumors. This detection rate is higher compared to previous tissue studies [5, 20, 21]. For instance, a study of MET tissue amplification determined by fluorescent in situ hybridization (FISH) demonstrated that 2.6% of 1115 solid tumor specimens were positive [5]. Furthermore, the MSKCC-IMPACT study showed that 3% of patients had MET tissue alterations [20, 21]. The differences in rate of MET alterations between our study and the other studies could be due to following reasons: (i) our technology detected both single nucleotide substitutions and amplifications, while the previous investigation by Jardim et al. at MD Anderson Cancer Center [5] discerned only amplifications (though the MSKCC study would have discerned both single nucleotide substitutions and amplifications [21]); (ii) our study used blood-derived ctDNA, which could capture shed tumor DNA from multiple sites, while the previous reports used tissue-based testing, which would only detect aberrations in the piece of tissue biopsied. Consistent with the above observations, our tissue NGS testing also showed significantly lower rates of MET alterations than the ctDNA NGS: 3 of 263 patients (1.1%) (who also had ctDNA tests) were positive for MET alterations in tissue versus 31 of 438 ctDNA-tested patients (7%) being positive for MET alterations (P = 0.0002). Further, of 18 patients positive for MET ctDNA alterations who also had tissue NGS, only 2 (11.1%) were also positive for a MET alteration by tissue NGS. The biologic underpinnings of discordance between ctDNA and tissue NGS results have been previously documented and include spatial (intra-tumor and inter-tumor) and temporal heterogeneity in genomic anomalies in cancers along with the fact that ctDNA is comprised of DNA that has leaked into the circulation from diverse metastatic sites while tissue NGS reflects only the tissue specimen analyzed [14].
The question that arises is whether or not biological explanations can specifically account for the higher rate of MET alterations in ctDNA. Of interest in this regard, our study showed that bone metastases were independently correlated with MET alterations (Additional file 1: Table S2 and Table S4). Indeed, 14 of 31 patients (45%) positive for MET ctDNA alterations had bone metastases (Additional file 1: Table S4). Of possible relevance in this regard, MET inhibitors are known to show efficacy in bone lesions [22, 23]. In the COMET-1 trial, 682 patients with castrate-resistant prostate cancer who progressed after docetaxel and androgen modulators (abiraterone and/or enzalutamide) were randomly assigned to either cabozantinib (MET inhibitor) or prednisone. Although there was no difference in overall survival, 42% of cabozantinib-treated patients showed bone scan response compared to 3% of prednisone-treated patients (P < 0.001), albeit without prostate surface antigen (PSA) response [24]. In the METEOR trial, 658 patients with advanced renal cell carcinoma who progressed with at least one VEGFR small molecule inhibitor were randomized to the MET inhibitor cabozantinib or the mTOR inhibitor everolimus [25]. In a sub-group analysis, patients randomized to cabozantinib arm with bone metastasis were associated with better overall survival (OS) (hazard ratio (HR) 0.54, 95% confidence interval (CI) 0.34–0.84) compared with non-bone metastasis (HR 0.71, 95% CI 0.55–0.91) [25]. These data together with our results raise the possibility that cancer clones with MET alterations preferentially localize to bone and may therefore explain bone responses after MET inhibitor therapy. Tissue biopsies are rarely performed on bones (none of our 18 patients with MET alterations in ctDNA who also had tissue NGS had a bone biopsy). Of interest in this regard, MET is prominently expressed (as determined by immunohistochemistry) at the site of bone metastases in renal cell cancer [26]. It is therefore conceivable that the high rates of ctDNA positivity for MET alterations, which strongly and independently correlated with bone metastases in our study, reflect shed MET alteration-bearing DNA from bone lesions (Additional file 1: Table S2 and Table S4).
Our study also found that MET ctDNA alterations are associated with poor prognosis, including decreased survival and shorter time to recurrence/metastasis (Tables 4 and 5, Fig. 1b). These results are consistent with those found by correlating MET alterations found in tissue NGS with outcome in specific malignancies, such as astrocytomas [27], breast cancers [28], genitourinary malignancies [29], and ovarian [30] or gastric/esophageal cancers [31]. Specific genes that were co-altered with MET in multivariate analysis of our study participants include PTEN and TP53 (Table 2); previously, another study has also shown the association between MET and PTEN abnormalities [5]. Overall, MET alterations significantly correlated with a higher number of alterations, which may explain—at least in part—the relatively limited efficacy of MET inhibitors as single agents for the treatment of MET-altered advanced malignancies observed in prior studies [5, 2931]. Indeed, if patients with MET alterations generally have multiple genomic abnormalities, it is likely that combination therapy, rather than monotherapy with a MET inhibitor, may be necessary to achieve salutary effects.
Our study has several limitations. First, only 60% of our patients (N = 263) with ctDNA analyses also had tissue sequencing and the median time between the two tests in the patients with MET-altered ctDNA was about 6 months. Second, our patients did not have bone biopsies done, which would be of interest to determine if MET alterations are found in such biopsies, since ctDNA MET alterations correlate independently with the presence of bone metastases. Third, though our study with 438 patients is the largest to date, the rarity of MET alterations suggests that investigations of even greater numbers of patients may be worthwhile in order to best understand the biology and correlations of MET alterations.

Conclusions

In summary, our study demonstrated that assessment of MET genomic aberrations by liquid biopsy is feasible. We found that MET ctDNA anomalies were associated with bone metastases, multiple genomic alterations, and a poorer prognosis, including poorer overall survival and a shorter time to recurrence/metastases. Further studies are needed to better understand the biologic relationship between MET alterations and bone lesions, and next-generation trials with MET inhibitors may require combinations of drugs that address the genes such as PTEN that are frequently co-altered in these patients.

Acknowledgments

This work was supported in part by the Joan and Irwin Jacobs Fund philanthropic fund [no grant number is applicable—Razelle Kurzrock] and by the National Cancer Institute at the National Institutes of Health [grant P30 CA016672 (Razelle Kurzrock, rkurzrock@ucsd.edu)].

Availability of data and materials

All data generated or analyzed during this study are included in this published article [and its additional files].
Not applicable

Competing interests

Dr. Razelle Kurzrock has research funding from Incyte, Genentech, Merck Serono, Pfizer, Sequenom, Foundation Medicine, and Guardant as well as consultant fees from Sequenom, LOXO, and Actuate Therapeutics. Dr. Kurzrock is also a consultant for Genentech. She also has an ownership interest in CureMatch, Inc. The other authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Cooper CS, Park M, Blair DG, Tainsky MA, Huebner K, Croce CM, et al. Molecular cloning of a new transforming gene from a chemically transformed human cell line. Nature. 1984;311:b29–33.CrossRef Cooper CS, Park M, Blair DG, Tainsky MA, Huebner K, Croce CM, et al. Molecular cloning of a new transforming gene from a chemically transformed human cell line. Nature. 1984;311:b29–33.CrossRef
2.
Zurück zum Zitat Park M, Dean M, Kaul K, Braun MJ, Gonda MA, Vande WG. Sequence of MET protooncogene cDNA has features characteristic of the tyrosine kinase family of growth-factor receptors. Proc Natl Acad Sci U S A. 1987;84:6379–83.CrossRefPubMedPubMedCentral Park M, Dean M, Kaul K, Braun MJ, Gonda MA, Vande WG. Sequence of MET protooncogene cDNA has features characteristic of the tyrosine kinase family of growth-factor receptors. Proc Natl Acad Sci U S A. 1987;84:6379–83.CrossRefPubMedPubMedCentral
3.
Zurück zum Zitat Bottaro DP, Rubin JS, Faletto DL, Chan AM, Kmiecik TE, Vande Woude GF, et al. Identification of the hepatocyte growth factor receptor as the c-met proto-oncogene product. Science. 1991;251:802–4.CrossRefPubMed Bottaro DP, Rubin JS, Faletto DL, Chan AM, Kmiecik TE, Vande Woude GF, et al. Identification of the hepatocyte growth factor receptor as the c-met proto-oncogene product. Science. 1991;251:802–4.CrossRefPubMed
4.
Zurück zum Zitat Birchmeier C, Birchmeier W, Gherardi E, Vande Woude GF. Met, metastasis, motility and more. Nat Rev Mol Cell Biol. 2003;4:915–25.CrossRefPubMed Birchmeier C, Birchmeier W, Gherardi E, Vande Woude GF. Met, metastasis, motility and more. Nat Rev Mol Cell Biol. 2003;4:915–25.CrossRefPubMed
5.
Zurück zum Zitat Jardim DLF, Tang C, Gagliato DDM, Falchook GS, Hess K, Janku F, et al. Analysis of 1,115 patients tested for MET amplification and therapy response in the MD Anderson Phase I Clinic. Clin Cancer Res. 2014;20:6336–45.CrossRefPubMed Jardim DLF, Tang C, Gagliato DDM, Falchook GS, Hess K, Janku F, et al. Analysis of 1,115 patients tested for MET amplification and therapy response in the MD Anderson Phase I Clinic. Clin Cancer Res. 2014;20:6336–45.CrossRefPubMed
6.
Zurück zum Zitat Schmidt L, Duh FM, Chen F, Kishida T, Glenn G, Choyke P, et al. Germline and somatic mutations in the tyrosine kinase domain of the MET proto-oncogene in papillary renal carcinomas. Nat Genet. 1997;16:68–73.CrossRefPubMed Schmidt L, Duh FM, Chen F, Kishida T, Glenn G, Choyke P, et al. Germline and somatic mutations in the tyrosine kinase domain of the MET proto-oncogene in papillary renal carcinomas. Nat Genet. 1997;16:68–73.CrossRefPubMed
7.
Zurück zum Zitat Kawakami H, Okamoto I, Okamoto W, Tanizaki J, Nakagawa K, Nishio K. Targeting MET amplification as a new oncogenic driver. Cancers. 2014;6:1540–52.CrossRefPubMedPubMedCentral Kawakami H, Okamoto I, Okamoto W, Tanizaki J, Nakagawa K, Nishio K. Targeting MET amplification as a new oncogenic driver. Cancers. 2014;6:1540–52.CrossRefPubMedPubMedCentral
8.
Zurück zum Zitat Madoz-Gúrpide J, Zazo S, Chamizo C, Casado V, Caramés C, Gavín E, et al. Activation of MET pathway predicts poor outcome to cetuximab in patients with recurrent or metastatic head and neck cancer. J Transl Med. 2015;13:282.CrossRefPubMedPubMedCentral Madoz-Gúrpide J, Zazo S, Chamizo C, Casado V, Caramés C, Gavín E, et al. Activation of MET pathway predicts poor outcome to cetuximab in patients with recurrent or metastatic head and neck cancer. J Transl Med. 2015;13:282.CrossRefPubMedPubMedCentral
9.
Zurück zum Zitat Kurzrock R, Sherman SI, Ball DW, Forastiere AA, Cohen RB, Mehra R, et al. Activity of XL184 (cabozantinib), an oral tyrosine kinase inhibitor, in patients with medullary thyroid cancer. J Clin Oncol. 2011;29:2660–6.CrossRefPubMedPubMedCentral Kurzrock R, Sherman SI, Ball DW, Forastiere AA, Cohen RB, Mehra R, et al. Activity of XL184 (cabozantinib), an oral tyrosine kinase inhibitor, in patients with medullary thyroid cancer. J Clin Oncol. 2011;29:2660–6.CrossRefPubMedPubMedCentral
10.
Zurück zum Zitat Shaw AT, Kim DW, Nakagawa K, Seto T, Crinó L, Ahn M-J, et al. Crizotinib versus chemotherapy in advanced ALK-positive lung cancer. N Engl J Med. 2013;368:2385–94.CrossRefPubMed Shaw AT, Kim DW, Nakagawa K, Seto T, Crinó L, Ahn M-J, et al. Crizotinib versus chemotherapy in advanced ALK-positive lung cancer. N Engl J Med. 2013;368:2385–94.CrossRefPubMed
12.
Zurück zum Zitat Schwaederle M, Patel SP, Husain H, Ikeda M, Lanman R, Banks KC, et al. Utility of genomic assessment of blood-derived circulating tumor DNA (ctDNA) in patients with advanced lung adenocarcinoma. Clin Cancer Res. 2017;23:5101–11.CrossRefPubMedPubMedCentral Schwaederle M, Patel SP, Husain H, Ikeda M, Lanman R, Banks KC, et al. Utility of genomic assessment of blood-derived circulating tumor DNA (ctDNA) in patients with advanced lung adenocarcinoma. Clin Cancer Res. 2017;23:5101–11.CrossRefPubMedPubMedCentral
13.
Zurück zum Zitat Kato S, Krishnamurthy N, Banks KC, De P, Williams K, Williams C, et al. Utility of genomic analysis in circulating tumor DNA from patients with carcinoma of unknown primary. Cancer Res. 2017;77:4238–46.CrossRefPubMedPubMedCentral Kato S, Krishnamurthy N, Banks KC, De P, Williams K, Williams C, et al. Utility of genomic analysis in circulating tumor DNA from patients with carcinoma of unknown primary. Cancer Res. 2017;77:4238–46.CrossRefPubMedPubMedCentral
14.
Zurück zum Zitat Schwaederle M, Husain H, Fanta PT, Piccioni DE, Kesari S, Schwab RB, et al. Use of liquid biopsies in clinical oncology: pilot experience in patients. Clin Cancer Res. 2016;22:5497–505.CrossRefPubMed Schwaederle M, Husain H, Fanta PT, Piccioni DE, Kesari S, Schwab RB, et al. Use of liquid biopsies in clinical oncology: pilot experience in patients. Clin Cancer Res. 2016;22:5497–505.CrossRefPubMed
15.
Zurück zum Zitat Bidard F-C, Madic J, Mariani P, Piperno-Neumann S, Rampanou A, Servois V, et al. Detection rate and prognostic value of circulating tumor cells and circulating tumor DNA in metastatic uveal melanoma. Int J Cancer. 2014;134:1207–13.CrossRefPubMed Bidard F-C, Madic J, Mariani P, Piperno-Neumann S, Rampanou A, Servois V, et al. Detection rate and prognostic value of circulating tumor cells and circulating tumor DNA in metastatic uveal melanoma. Int J Cancer. 2014;134:1207–13.CrossRefPubMed
16.
Zurück zum Zitat Schwaederle M, Husain H, Fanta PT, Piccioni DE, Kesari S, Schwab RB, et al. Detection rate of actionable mutations in diverse cancers using a biopsy-free (blood) circulating tumor cell DNA assay. Oncotarget. 2016;7:9707–17.CrossRefPubMedPubMedCentral Schwaederle M, Husain H, Fanta PT, Piccioni DE, Kesari S, Schwab RB, et al. Detection rate of actionable mutations in diverse cancers using a biopsy-free (blood) circulating tumor cell DNA assay. Oncotarget. 2016;7:9707–17.CrossRefPubMedPubMedCentral
17.
Zurück zum Zitat Lanman RB, Mortimer SA, Zill OA, Sebisanovic D, Lopez R, Blau S, et al. Analytical and clinical validation of a digital sequencing panel for quantitative, highly accurate evaluation of cell-free circulating tumor DNA. PLoS One. 2015;10:e0140712.CrossRefPubMedPubMedCentral Lanman RB, Mortimer SA, Zill OA, Sebisanovic D, Lopez R, Blau S, et al. Analytical and clinical validation of a digital sequencing panel for quantitative, highly accurate evaluation of cell-free circulating tumor DNA. PLoS One. 2015;10:e0140712.CrossRefPubMedPubMedCentral
18.
Zurück zum Zitat Kwapisz D. The first liquid biopsy test approved. Is it a new era of mutation testing for non small cell lung cancer? Ann Transl. 2017;Med5:46.CrossRef Kwapisz D. The first liquid biopsy test approved. Is it a new era of mutation testing for non small cell lung cancer? Ann Transl. 2017;Med5:46.CrossRef
19.
Zurück zum Zitat Chabon JJ, Simmons AD, Lovejoy AF, Esfahani MS, Newman AM, Haringsma HJ, et al. Circulating tumour DNA profiling reveals heterogeneity of EGFR inhibitor resistance mechanisms in lung cancer patients. Nat Commun. 2016;7:11815.CrossRefPubMedPubMedCentral Chabon JJ, Simmons AD, Lovejoy AF, Esfahani MS, Newman AM, Haringsma HJ, et al. Circulating tumour DNA profiling reveals heterogeneity of EGFR inhibitor resistance mechanisms in lung cancer patients. Nat Commun. 2016;7:11815.CrossRefPubMedPubMedCentral
20.
Zurück zum Zitat Cheng DT, Mitchell TN, Zehir A, Shah RH, Benayed R, Syed A, 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. J Mol Diagn. 2015;17:251–64.CrossRefPubMedPubMedCentral Cheng DT, Mitchell TN, Zehir A, Shah RH, Benayed R, Syed A, 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. J Mol Diagn. 2015;17:251–64.CrossRefPubMedPubMedCentral
21.
Zurück zum Zitat Zehir A, Benayed R, Shah RH, Syed A, Middha S, Kim HR, et al. Mutational landscape of metastatic cancer revealed from prospective clinical sequencing of 10,000 patients. Nat Med. 2017;23:703–13.CrossRefPubMedPubMedCentral Zehir A, Benayed R, Shah RH, Syed A, Middha S, Kim HR, et al. Mutational landscape of metastatic cancer revealed from prospective clinical sequencing of 10,000 patients. Nat Med. 2017;23:703–13.CrossRefPubMedPubMedCentral
22.
Zurück zum Zitat Graham TJ, Box G, Tunariu N, Crespo M, Spinks TJ, Miranda S, et al. Preclinical evaluation of imaging biomarkers for prostate cancer bone metastasis and response to cabozantinib. J Natl Cancer Inst. 2014;106:dju033.CrossRefPubMed Graham TJ, Box G, Tunariu N, Crespo M, Spinks TJ, Miranda S, et al. Preclinical evaluation of imaging biomarkers for prostate cancer bone metastasis and response to cabozantinib. J Natl Cancer Inst. 2014;106:dju033.CrossRefPubMed
23.
Zurück zum Zitat McKay RR, Kroeger N, Xie W, Lee J-L, Knox JJ, Bjarnason GA, et al. Impact of bone and liver metastases on patients with renal cell carcinoma treated with targeted therapy. Eur Urol. 2014;65:577–84.CrossRefPubMed McKay RR, Kroeger N, Xie W, Lee J-L, Knox JJ, Bjarnason GA, et al. Impact of bone and liver metastases on patients with renal cell carcinoma treated with targeted therapy. Eur Urol. 2014;65:577–84.CrossRefPubMed
24.
Zurück zum Zitat Smith M, De Bono J, Sternberg C, Le Moulec S, Oudard S, De Giorgi U, et al. Phase III study of cabozantinib in previously treated metastatic castration-resistant prostate cancer: COMET 1. J Clin Oncol. 2016;34:3005–13.CrossRefPubMed Smith M, De Bono J, Sternberg C, Le Moulec S, Oudard S, De Giorgi U, et al. Phase III study of cabozantinib in previously treated metastatic castration-resistant prostate cancer: COMET 1. J Clin Oncol. 2016;34:3005–13.CrossRefPubMed
25.
Zurück zum Zitat Choueiri TK, Escudier B, Powles T, Tannir NM, Mainwaring PN, Rini BI, et al. Cabozantinib versus everolimus in advanced renal cell carcinoma (METEOR): final results from a randomised, open-label, phase 3 trial. Lancet Oncol. 2016;17:917–27.CrossRefPubMed Choueiri TK, Escudier B, Powles T, Tannir NM, Mainwaring PN, Rini BI, et al. Cabozantinib versus everolimus in advanced renal cell carcinoma (METEOR): final results from a randomised, open-label, phase 3 trial. Lancet Oncol. 2016;17:917–27.CrossRefPubMed
26.
Zurück zum Zitat Mukai S, Yorita K, Kawagoe Y, Katayama Y, Nakahara K, Kamibeppu T, et al. Matriptase and MET are prominently expressed at the site of bone metastasis in renal cell carcinoma: immunohistochemical analysis. Hum Cell. 2015;28:44–50.CrossRefPubMed Mukai S, Yorita K, Kawagoe Y, Katayama Y, Nakahara K, Kamibeppu T, et al. Matriptase and MET are prominently expressed at the site of bone metastasis in renal cell carcinoma: immunohistochemical analysis. Hum Cell. 2015;28:44–50.CrossRefPubMed
27.
Zurück zum Zitat Pierscianek D, Kim YH, Motomura K, Mittelbronn M, Paulus W, Brokinkel B, et al. MET gain in diffuse astrocytomas is associated with poorer outcome. Brain Pathol. 2013;23:13–8.CrossRefPubMed Pierscianek D, Kim YH, Motomura K, Mittelbronn M, Paulus W, Brokinkel B, et al. MET gain in diffuse astrocytomas is associated with poorer outcome. Brain Pathol. 2013;23:13–8.CrossRefPubMed
28.
Zurück zum Zitat de Melo Gagliato D, Fontes Jardim DL, Falchook G, Tang C, Zinner R, et al. Analysis of MET genetic aberrations in patients with breast cancer at MD Anderson Phase I Unit. Clin Breast Cancer. 2014;14:468–74.CrossRefPubMedPubMedCentral de Melo Gagliato D, Fontes Jardim DL, Falchook G, Tang C, Zinner R, et al. Analysis of MET genetic aberrations in patients with breast cancer at MD Anderson Phase I Unit. Clin Breast Cancer. 2014;14:468–74.CrossRefPubMedPubMedCentral
29.
Zurück zum Zitat Jardim DLF, de Melo Gagliato D, Falchook G, Zinner R, Wheler JJ, Janku F, et al. MET abnormalities in patients with genitourinary malignancies and outcomes with c-MET inhibitors. Clin Genitourin Cancer. 2015;13:e19–26.CrossRefPubMed Jardim DLF, de Melo Gagliato D, Falchook G, Zinner R, Wheler JJ, Janku F, et al. MET abnormalities in patients with genitourinary malignancies and outcomes with c-MET inhibitors. Clin Genitourin Cancer. 2015;13:e19–26.CrossRefPubMed
30.
Zurück zum Zitat Tang C, Fontes Jardim DL, Falchook GS, Hess K, Fu S, Wheler JJ, et al. MET nucleotide variations and amplification in advanced ovarian cancer: characteristics and outcomes with c-Met inhibitors. Oncoscience. 2013;1:5–13.CrossRefPubMedPubMedCentral Tang C, Fontes Jardim DL, Falchook GS, Hess K, Fu S, Wheler JJ, et al. MET nucleotide variations and amplification in advanced ovarian cancer: characteristics and outcomes with c-Met inhibitors. Oncoscience. 2013;1:5–13.CrossRefPubMedPubMedCentral
31.
Zurück zum Zitat Jardim DLF, Gagliato D de M, Falchook GS, Janku F, Zinner R, Wheler JJ, et al. MET aberrations and c-MET inhibitors in patients with gastric and esophageal cancers in a phase I unit. Oncotarget. 2014;5:1837–45.CrossRefPubMedPubMedCentral Jardim DLF, Gagliato D de M, Falchook GS, Janku F, Zinner R, Wheler JJ, et al. MET aberrations and c-MET inhibitors in patients with gastric and esophageal cancers in a phase I unit. Oncotarget. 2014;5:1837–45.CrossRefPubMedPubMedCentral
Metadaten
Titel
MET alterations detected in blood-derived circulating tumor DNA correlate with bone metastases and poor prognosis
verfasst von
Sadakatsu Ikeda
Maria Schwaederle
Mandakini Mohindra
Denis L. Fontes Jardim
Razelle Kurzrock
Publikationsdatum
01.12.2018
Verlag
BioMed Central
Erschienen in
Journal of Hematology & Oncology / Ausgabe 1/2018
Elektronische ISSN: 1756-8722
DOI
https://doi.org/10.1186/s13045-018-0610-8

Weitere Artikel der Ausgabe 1/2018

Journal of Hematology & Oncology 1/2018 Zur Ausgabe

Adjuvante Immuntherapie verlängert Leben bei RCC

25.04.2024 Nierenkarzinom Nachrichten

Nun gibt es auch Resultate zum Gesamtüberleben: Eine adjuvante Pembrolizumab-Therapie konnte in einer Phase-3-Studie das Leben von Menschen mit Nierenzellkarzinom deutlich verlängern. Die Sterberate war im Vergleich zu Placebo um 38% geringer.

Alectinib verbessert krankheitsfreies Überleben bei ALK-positivem NSCLC

25.04.2024 NSCLC Nachrichten

Das Risiko für Rezidiv oder Tod von Patienten und Patientinnen mit reseziertem ALK-positivem NSCLC ist unter einer adjuvanten Therapie mit dem Tyrosinkinase-Inhibitor Alectinib signifikant geringer als unter platinbasierter Chemotherapie.

Bei Senioren mit Prostatakarzinom auf Anämie achten!

24.04.2024 DGIM 2024 Nachrichten

Patienten, die zur Behandlung ihres Prostatakarzinoms eine Androgendeprivationstherapie erhalten, entwickeln nicht selten eine Anämie. Wer ältere Patienten internistisch mitbetreut, sollte auf diese Nebenwirkung achten.

ICI-Therapie in der Schwangerschaft wird gut toleriert

Müssen sich Schwangere einer Krebstherapie unterziehen, rufen Immuncheckpointinhibitoren offenbar nicht mehr unerwünschte Wirkungen hervor als andere Mittel gegen Krebs.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.