Skip to main content
Erschienen in: Cellular Oncology 2/2015

Open Access 01.04.2015 | Original Paper

Anti-tumor activity of the TGF-β receptor kinase inhibitor galunisertib (LY2157299 monohydrate) in patient-derived tumor xenografts

verfasst von: Armin Maier, Anne-Lise Peille, Vincent Vuaroqueaux, Michael Lahn

Erschienen in: Cellular Oncology | Ausgabe 2/2015

Abstract

Purpose

The transforming growth factor-beta (TGF-β) signaling pathway is known to play a critical role in promoting tumor growth. Consequently, blocking this pathway has been found to inhibit tumor growth. In order to achieve an optimal anti-tumor effect, however, it remains to be established whether blocking the TGF-β signaling pathway alone is sufficient, or whether the tumor microenvironment plays an additional, possibly synergistic, role.

Methods

To investigate the relevance of blocking TGF-β signaling in tumor cells within the context of their respective tissue microenvironments, we treated a panel of patient-derived xenografts (PDX) with the selective TGF-β receptor kinase inhibitor LY2157299 monohydrate (galunisertib) and assessed both the in vitro and in vivo effects.

Results

Galunisertib was found to inhibit the growth in an in vitro clonogenic assay in 6.3 % (5/79) of the examined PDX. Evaluation of the expression profiles of a number of genes, representing both canonical and non-canonical TGF-β signaling pathways, revealed that most PDX exhibited expression changes affecting TGF-β downstream signaling. Next, we subjected 13 of the PDX to an in vivo assessment and, by doing so, observed distinct response patterns. These results suggest that, next to intrinsic, also extrinsic or microenvironmental factors can affect galunisertib response. pSMAD2 protein expression and TGF-βRI mRNA expression levels were found to correlate with the in vivo galunisertib effects.

Conclusions

From our data we conclude that intrinsic, tumor-dependent TGF-β signaling does not fully explain the anti-tumor effect of galunisertib. Hence, in vivo xenograft models may be more appropriate than in vitro clonogenic assays to assess the anti-tumor activity of TGF-β inhibitors such as galunisertib.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1007/​s13402-014-0210-8) contains supplementary material, which is available to authorized users.

1 Introduction

The transforming growth factor beta (TGF-β) signaling pathway plays a pleiotropic role in both normal and tumor tissues, including tumor-stroma interactions [1, 2]. The canonical TGF-β signaling pathway becomes activated when 1 of the 3 ligands (TGF-β1, TGF-β2, TGF-β3) binds to the TGF-β receptor II (TGF-βRII), which subsequently heterodimerizes with the TGF-β receptor I (TGF-βRI or ALK5) and transphosphorylates the kinase domains of both receptors. This phosphorylation step leads to a recruitment and phosphorylation of SMAD2 and SMAD3 (pSMAD2 and pSMAD3). Next, this complex initiates the canonical or SMAD-dependent signaling cascade leading to nuclear translocation and downstream gene transcription [3]. In addition to the canonical signaling pathway, other activation pathways (non-canonical pathways) have been described, but these are less understood [4]. The non-canonical or non-SMAD-dependent activation of the TGF-β pathway includes signaling via jun N-terminal kinase (JNK), p38 MAPK, ERK or MEKK.
In the past, several small molecule inhibitors targeting the TGF-βRI serine/threonine kinase activity have been developed, including LY2157299 monohydrate (galunisertib) [5], which has been found to inhibit pSMAD2 expression in different tumor models [6, 7]. Galunisertib is now being investigated in a clinical trials and has very recently been shown to elicit anti-tumor effects in patients with glioblastoma or hepatocellular carcinoma [8, 9]. Since only a few TGF-β inhibitors are currently being studied in clinical trials, the development of appropriate preclinical models is considered imperative in order to reliably establish the mechanisms of action of TGF-β inhibitors and to specifically direct new drug screens.
In traditional models such as xenografts with established tumor cell lines or in vitro cell viability studies, galunisertib has shown moderate anti-tumor activity [10, 11]. Here, we used patient-derived xenografts (PDX) instead of established tumor-derived cell lines [12, 13]. In contrast to these cell lines, primary patient-derived cells generally retain their original phenotype [14, 15]. Initially, primary patient-derived cells were used to assess the effects of cytotoxic agents [16] but, more recently, these cells have also been found to be useful for characterizing anti-tumor activities of cytostatic or immunomodulatory agents [1719]. It has also been noted that the effects of anti-tumor drugs in PDX-based clonogenic assays correlated well with clinical responses observed in patients with various solid cancers [12, 20, 21]. Therefore, we hypothesized that PDX models might be useful for testing both the in vitro and in vivo effects of galunisertib in different primary tumor cell types and, as such, to delineate the roles of both intrinsic and extrinsic activities of TGF-β signaling in the respective responses in these models.

2 Materials and methods

2.1 Small molecule TGF-β receptor kinase inhibitor

The small molecule LY2157299 monohydrate (galunisertib), targeting TGF-βRI serine/threonine kinase activity, was provided by Eli Lilly and Company, Indianapolis, USA. Galunisertib was tested at concentrations ranging from 0.03 to 10.0 μM. Selected tumor xenografts were re-tested at higher concentrations ranging from 1.0 to 80.0 μM. Stock solutions of the compound were prepared in DMSO at 3.0 or 24.0 mM, respectively, and small aliquots were stored at −20 °C in the dark. Final dilutions were prepared in Iscove’s Modified Dulbecco’s Medium (IMDM, Life Technologies, Carlsbad, CA) immediately prior to use.

2.2 In vitro clonogenic assays on patient-derived xenograft samples

After obtaining the informed consent from patients and approvals from local ethics review boards, patient-derived xenograft (PDX) samples were derived from tumors subcutaneously growing as xenografts in NMRI nu/nu mice [13, 22] purchased from Elevage Janvier, France or Taconic Europe, Denmark. Details of the test procedure have been previously described [21]. Briefly, solid tumor xenografts were removed from mice under sterile conditions, mechanically disaggregated and subsequently incubated in an enzyme cocktail consisting of collagenase type IV (41 U/ml), DNase I (125 U/ml), hyaluronidase type III (100 U/ml) and dispase II (1.0 U/ml) in RPMI-1640 medium at 37 °C for 45–60 min. Single cells were passed through sieves of 200 and 50 μm mesh size and washed twice with sterile PBS. The percentage of viable cells was determined in a hemocytometer using trypan blue exclusion staining.
The tumor clonogenic assays were performed according to a modified soft agar assay introduced by Hamburger & Salmon [23]. Each test well contained three layers of equal volumes: two layers of semi-solid medium (bottom and top layer) and one layer of medium supernatent with or without test compound. The bottom layer consisted of IMDM, supplemented with 20 % (v/v) fetal calf serum (FCS, Sigma, St Louis, MO), 0.01 % (w/v) gentamicin (Life Technologies, Carlsbad, CA) and 0.75 % (w/v) agar (BD Biosciences, San Jose, CA). Cells were seeded at a final density of 7.5 × 104 to 2 × 105 cells/ml using the same culture medium, supplemented with 0.4 % (w/v) agar, and plated onto the bottom layer. The test compound was added by continuous exposure (drug overlay) in culture medium. Cultures were incubated at 37 °C and 7.5 % CO2 in a humidified atmosphere for 7–20 days and monitored closely for colony growth using an inverted microscope. Within this period, colonies were formed with a diameter > 50 μm. At the time of maximum colony formation, counts were performed using an automatic image analysis system (OMNICON 3600, Biosys GmbH, Germany) after staining vital colonies for 24 h prior to evaluation with a sterile aqueous solution of 2-(4-iodophenyl)-3-(4-nitrophenyl)-5-phenyltetrazolium chloride (1 mg/ml, 100 μl/well) [24]. Additionally, the viability of the colonies was determined using a CellTiter-Glo® viability assay (Promega, Madison, WI), as an equivalent to colony formation, and luminescence was measured using an EnVision® Xcite Multilabel Reader (Perkin Elmer, Waltham, MA) to quantify the amount of metabolically active and, thus, viable cells. All assays were performed in a standardized manner and the efficacy of galunisertib was assessed in relation to an untreated control only containing cells and the solvent (DMSO at 0.3 %). The efficacy of galunisertib was rated based on concentration-responses as: inhibition (T/C ≤ 75 %), no response (75 % < T/C < 125 %) or stimulation (T/C ≥ 125 %).

2.3 In vivo assays on patient-derived xenografts

Patient-derived xenografts (PDX) were established from primary patient material as described above. Cell line-derived xenografts were established from cells harvested from in vitro culture. Xenografts were subcutaneously grown in nude mice through serial passage and randomized after reaching tumor volumes of approximately 72–120 mm3.
Galunisertib was prepared as a suspension in 1 % NaCMC, 0.5 % SLS, 0.05 % Antifoam, 0.085 % PVP C-30, and administered twice daily for 14 days at a dose of 75 mg/kg orally by gavage (12 mice per group). The control group received an identical volume of the same mix without galunisertib. The tumor load was determined by caliper measurement twice weekly and the absolute tumor volume [mm3] was calculated according to the formula: a [mm] × b2 [mm2] × 0.5, where (a) is the largest diameter and (b) is the perpendicular diameter of the tumor representing an idealized ellipsoid. The relative volume of an individual tumor on day x (RTVx) was calculated by dividing the absolute volume [mm3] of the respective tumor on day x (Tx) by the absolute volume of the same tumor on the day of randomization, i.e., on day 0 (T0), multiplied by 100: RTVx [%] = Tx/T0 × 100. Group median RTVs were used for drawing tumor growth curves and for treatment evaluation. The tumor growth response was expressed quantitatively by the Area Between the Curves (ABC), comparing the Area Under the Curve (AUC) of tumor growth curves of the control group with the AUC of the group treated with galunisertib according to the formula below.
$$ \boldsymbol{A}\boldsymbol{B}\boldsymbol{C}\ \left(\%\right)=\frac{\boldsymbol{AU}{\boldsymbol{C}}_{\boldsymbol{control}}-\boldsymbol{AU}{\boldsymbol{C}}_{\boldsymbol{treatment}}}{\boldsymbol{AU}{\boldsymbol{C}}_{\boldsymbol{control}}}*100 $$
Using this formula, positive ABC values represent tumor growth curves below the control group and indicate growth inhibition. Tumor responses were classified according to ABC values of < −20 % = growth stimulation, −20 % < ABC < 20 % = no change, > 20 % = growth inhibition, compared to the control. All studies were performed in agreement with German animal welfare acts.

2.4 RNA isolation

For each untreated xenograft model, tissues were pooled from 4 different mice. Total RNA was extracted from frozen samples using the “mirVana miRNA Isolation kit” (Ambion, Carlsbad, CA) according to the manufacturer’s instructions. Genomic DNA was removed using the “RNase-free DNase Set” (Qiagen, Hilden, Germany). The quality of the RNA preparations was controlled using a Bioanalyzer (Agilent Technologies, Palo Alto, CA). Only RNA samples with an RNA integrity number (RIN) > 6.5 were used for gene expression profiling purposes (see below).

2.5 DNA isolation

For DNA isolation, snap frozen samples from untreated tumors were digested with proteinase K at 55 °C overnight and lysates were digested with RNase A (Qiagen). Next, DNAs were extracted using phenol:chloroform:isoamylalcohol and precipitated with ethanol. DNA pellets were washed and resuspended in TElow buffer (Tris 10 mM pH8, EDTA 0.1 mM pH8). The integrity of the DNA samples was checked after 1.3 % agarose gel electrophoresis, and the purity of the DNA samples was determined using a NanoDrop 2000 system (Thermo Scientific, Waltham, MA).

2.6 Gene expression profiling

Total RNAs were submitted to AROS Applied Biotechnologies (Aarhus, Denmark) or DNA vision for analysis on Affymetrix HGU133 plus 2.0 gene expression arrays. First- and second-strand synthesis, biotin labeling, fragmentation and hybridization were performed according to Affymetrix protocols. Evaluation and normalization of the Affymetrix GeneChip Data were performed in the “R” (version 2.15.3) statistical computing environment. The hybridizations were normalized using the gcRMA (gc robust multichip averaging) method from Bioconductor to obtain summary expression values for each probe set. One probeset for each gene was chosen according to Li et al. [25]. Gene expression levels were analyzed on a logarithmic scale and were expressed in arbitrary units (U). Affymetrix expression values < 6 U were considered as background.

2.7 Gene mutation analyses

The mutation status of key cancer genes was assessed in all samples using mass array sequencing panels from Sequenom, Inc. (OncoCarta panels I, II and III) and then confirmed by Sanger sequencing of individual exons or whole exome sequencing. Moreover, 64/79 PDX samples were profiled by whole exome sequencing. Exonic regions from Oncotest DNA samples were targeted using Agilent SureSelect Human All Exon kits 38 MB (60 samples) or 51 MB (4 samples). Enriched genomic DNA was sequenced using an Illumina HiSeq-2000 platform in 100 bp paired-end (PE) reads and an expected coverage of ~80×. To remove the mouse stroma content, PE reads that mapped better on the mouse (mm10) than on the human (hg19) genome were discarded from the human mapped read dataset (based on the Burrows-Wheeler Alignment mapping score) using PicardTools. Variants were detected by independently using 3 different variant callers: the GATK’s UnifiedGenotyper, the combination of Samtools mpileup and bcftools caller, and the Freebayes caller. Only variants identified with all 3 tools, showing a minimum number of variant-supporting reads of 3 and a minimum variant frequency of 5 %, were further analyzed.

2.8 SNP profiling

The Affymetrix Genome-Wide Human SNP Array 6.0 with 1.8 million genetic markers, including more than 906,600 single nucleotide polymorphisms (SNPs) and more than 946,000 probes for the detection of copy number variation (CNV), was employed using a standard protocol recommended by the manufacturer. According to the Affymetrix guidelines, contrast Quality Control (QC) and Median Absolute Pairwise Difference (MAPD) thresholds were set at values > 0.4 and 0.35, respectively.
CNVs were identified using the Affymetrix Genotyping Console v4.1 and the PICNIC software provided by the Cancer Genome Project from the Welcome Trust Sanger Institute [26].

2.9 Western blot analyses

Native tumor lysates were prepared for Western blotting as previously described [17]. Briefly, 4 parts of untreated PDX samples were homogenized using a Tissue Lyser (Qiagen) in cell lysis buffer supplemented with Tris pH7.4 20 mM (Roth, Karlsruhe, Germany), NaCl 100 mM (Merck, Darmstadt, Germany), EDTA 1 mM (Sigma-Aldrich), NP40 1 % (Sigma-Aldrich), deoxycholate 0.5 % (Sigma-Aldrich), Na4O7P2 10H2O 10 mM (Sigma-Aldrich), Na3VO4 2 mM (Sigma-Aldrich), NaF 20 mM (Sigma-Aldrich), PMSF 100 μM (Sigma-Aldrich), Benzonase 50 U/ml, and a protease inhibitor mix 1× (Roche, Mannheim, Germany). Lysates were cleared and protein concentrations measured (Protein Assay, Bio-Rad, Berkley, CA). After polyacrylamide gel electrophoresis and membrane transfer, the resulting membranes were incubated with primary antibodies (anti- TGF-β1 clone 56E4, Cat #3709, rabbit mAb, Cell Signaling, Beverly, CA; anti-Smad2: clone D43B4, Cat #5339, rabbit mAb, Cell Signaling; anti-pSmad2 (ser465/467): clone 138D4, Cat #3108, rabbit mAb, Cell Signaling; anti-GAPDH: clone 14C10, Cat #2118, rabbit mAb, Cell Signaling). The binding of primary antibodies was detected using a secondary HRP-coupled antibody (goat anti-rabbit IgG (H + L)-HRP conjugate, 170-6515-Biorad), followed by incubation with an ECL Western blotting detection reagent (GE Healthcare Cat # RPN2106, Little Chalfont, United Kingdom) and visualization using a GE Healthcare Image Quant LAS 4000 CCD camera system (GE Healthcare 28-9558-10). Quantification of protein expression was performed by subtracting the intensity of the signal of GAPDH from the intensity of the signal of the protein of interest, and was carried out using Image J software. The calculated quantities of the respective proteins are expressed in arbitrary units (AU). The Oncotest tumor lysate pool containing a mixture of 380 different PDX lysates was used as a normalization control to minimize plate-to-plate variability.

3 Results

3.1 TGF-β inhibitor galunisertib variably impairs clonogenic growth of tumor-derived xenografts

The TGF-β inhibitor galunisertib was evaluated in different PDX and cell line-derived xenografts (CDX) in different test series using a clonogenic assay. The test panel consisted of 11 different tumor types, including glioblastoma, melanoma, colon, gastric, liver, non-small cell lung (adenocarcinoma, squamous cell carcinoma and large cell lung carcinoma, NSCLC), small cell lung, mammary, ovary, pancreas, and renal cell cancers (Table 1). In addition, established cell lines derived from hematologic malignancies including leukemia, lymphoma and myeloma were tested. Initial screens with diverse numbers of tumor types have often been used to assess the range of anti-tumor efficacy for compounds in order to understand their differential activity [2729]. The majority of PDX in our current study were obtained from metastatic tumor lesions (42/79, 53 %) and were poorly differentiated. Most tumors were obtained from males (46/79; 58 %) and the mean age of the patients was 52 years (median age 55 years, range 11–82 years). The anti-tumor effects were recorded as inhibition of colony formation in relation to untreated controls (T/C values, see materials and methods). Although doses above 10 μM are expected to be associated with unspecific activity of galunisertib [6], we used higher concentrations to gain insight into anti-tumor effects beyond the pharmacologically targeted concentration.
Table 1
Main characteristics of 79 patient-derived xenografts (PDX) and cell line-derived xenografts (CDX) (solid and hematologic), which were tested against galunisertib in different test series
Histologic type
Origin
(PDX / CDX)
Tumor designation
Pathological stage at time of xenograft implantation
Histology classification
Origin of tumor lesion
Origin of organ
Differentiation
Patient age
Gender
Glioblastoma
PDX
CNXF 498
WHO Grade IV
glioblastoma
not known
Brain
poorly differentiated
62
male
Colon
PDX
CXF 1103
T3N1M1
adenocarcinoma
metastasis
Skin (peritoneum)
poorly differentiated
55
male
Colon
PDX
CXF 158
T2N0M0
adenocarcinoma
primary
Colon
moderately differentiated
68
female
Colon
PDX
CXF 1784
M1
adenocarcinoma
metastasis
Liver
moderately differentiated
65
male
Colon
PDX
CXF 260
TxN2Mx
adenocarcinoma
primary
Colon
poorly differentiated
68
female
Colon
PDX
CXF 269
TxN2Mx
colorectal adenocarcinoma
primary
Colon
poorly differentiated
56
male
Colon
PDX
CXF 280
M1
adenocarcinoma
metastasis
Skin
poorly differentiated
55
female
Colon
PDX
CXF 742
M1
adenocarcinoma
metastasis
Liver (mesenterium)
poorly differentiated
66
male
Colon
PDX
CXF 975
T3N1M0
adenocarcinoma
primary
Colon
moderately differentiated
44
female
Colon
CDX
CXF-HCT-116
not known
colon carcinoma
not known
Colon
poorly differentiated
not known
male
Gastric
PDX
GXF 1172
T4N3M1
adenocarcinoma
metastasis
Pleura
poorly differentiated
49
male
Gastric
PDX
GXF 251
T2N0M0
adenocarcinoma
primary
Stomach
poorly differentiated
60
male
Liver
PDX
LIXF 575
not known
hepatocellular carcinoma
primary
Liver
well differentiated
66
male
Liver
CDX
LIXF Hep-G2
not known
hepatocellular carcinoma
not known
not known
poorly differentiated
15
male
Lung, NSC
PDX
LXFA 1012
T2N0Mx
adenocarcinoma
primary
Lung
poorly differentiated
58
male
Lung, NSC
PDX
LXFA 1041
T4NxMx
adenocarcinoma
metastasis
Pleura
well differentiated
60
male
Lung, NSC
PDX
LXFA 1584
T4N1Mx
adenosquamous carcinoma
primary
Lung
poorly differentiated
53
male
Lung, NSC
PDX
LXFA 1647
M1
adenocarcinoma
metastasis
adrenal gland
poorly differentiated
53
male
Lung, NSC
PDX
LXFA 297
T2N2Mx
adenocarcinoma
metastasis
Lymph Node
poorly differentiated
46
male
Lung, NSC
PDX
LXFA 526
N3M1
adenocarcinoma
metastasis
Lymph Node
poorly differentiated
58
male
Lung, NSC
PDX
LXFA 677
T1N1Mx
adenocarcinoma
primary
Lung
poorly differentiated
62
male
Lung, NSC
PDX
LXFA 737
T3N2Mx
adenocarcinoma
primary
Lung
moderately differentiated
56
male
Lung, NSC
PDX
LXFA 749
T2N1-2 Mx
adenocarcinoma
primary
Lung
poorly differentiated
72
male
Lung, NSC
PDX
LXFA 983
T2N0M1
adenocarcinoma
metastasis
Brain
poorly differentiated
59
male
Lung, NSC
CDX
LXFA CALU-6
not known
adenocarcinoma
not known
not known
poorly differentiated
61
female
Lung, NSC
PDX
LXFE 1422
T2N3Mx
squamous cell carcinoma
primary
Lung
poorly differentiated
38
male
Lung, NSC
PDX
LXFE 211
M1 skin
squamous cell carcinoma
metastasis
Skin
poorly differentiated
58
male
Lung, NSC
PDX
LXFE 397
T1N0Mx
squamous cell carcinoma
primary
Lung
poorly differentiated
56
male
Lung, NSC
PDX
LXFL 1121
T2N1M0
large cell carcinoma
primary
Lung
poorly differentiated
56
female
Lung, NSC
PDX
LXFL 1176
TxN2Mx
large cell carcinoma
metastasis
Lymph Node
moderately differentiated
65
male
Lung, NSC
PDX
LXFL 529
T3N1M0
large cell carcinoma
primary
Lung
poorly differentiated
34
female
Lung, NSC
PDX
LXFL 625
T3N1M0
large cell carcinoma
primary
Lung
poorly differentiated
39
female
Lung, SC
PDX
LXFS 615
M1
small cell anaplastic carcinoma
metastasis
Bone Marrow
poorly differentiated
56
female
Lung, SC
PDX
LXFS 650
N1
small cell carcinoma
metastasis
Lymph Node
poorly differentiated
64
male
Breast
PDX
MAXF 1162
T4N0Mx
adenocarcinoma
primary
breast
poorly differentiated
55
female
Breast
PDX
MAXF 1322
M1
adenocarcinoma
metastasis
Brain
poorly differentiated
49
female
Breast
PDX
MAXF 1384
M1
adenocarcinoma
metastasis
Brain
poorly differentiated
32
female
Breast
PDX
MAXF 401
M1
adenocarcinoma
metastasis
Lung
well differentiated
51
female
Breast
PDX
MAXF 449
T3N1M0
invasive ductal carcinoma
primary
breast
poorly differentiated
53
female
Breast
PDX
MAXF 574
not known
invasive ductal carcinoma
not known
not known
poorly differentiated
not known
female
Breast
PDX
MAXF 583
M1
adenocarcinoma
metastasis
Lung
moderately differentiated
50
female
Breast
PDX
MAXF 857
T4NxM0
adenocarcinoma
primary
breast
poorly differentiated
60
female
Breast
PDX
MAXF MX1
not known
not known
not known
not known
poorly differentiated
29
female
Breast
CDX
MAXF-MDA-MB-231
not known
invasive ductal carcinoma
not known
not known
poorly differentiated
51
female
Melanoma
PDX
MEXF 1341
M1
melanoma
metastasis
Lung
not known
35
female
Melanoma
PDX
MEXF 1539
N1
melanoma
metastasis
Lymph Node
not known
36
male
Melanoma
PDX
MEXF 1732
N1
melanoma
metastasis
Lymph Node
not known
33
male
Melanoma
PDX
MEXF 1737
N1
melanoma
metastasis
Lymph Node
not known
62
female
Melanoma
PDX
MEXF 1765
N1
melanoma
metastasis
Lymph Node
not known
49
male
Melanoma
PDX
MEXF 1792
N1
melanoma
metastasis
Lymph Node
not known
60
male
Melanoma
PDX
MEXF 1829
M1
melanoma
metastasis
Lymph Node
not known
41
female
Melanoma
PDX
MEXF 1870
pM1 (SKI), R1
melanoma
metastasis
Skin
not known
63
female
Melanoma
PDX
MEXF 2090
not known
melanoma
primary
Skin
not known
not known
not known
Melanoma
PDX
MEXF 2095
not known
melanoma
primary
Skin
not known
not known
not known
Melanoma
PDX
MEXF 2106
not known
melanoma
primary
Skin
not known
not known
not known
Melanoma
PDX
MEXF 274
N1
melanoma
metastasis
Lymph Node
not known
51
male
Melanoma
PDX
MEXF 276
N1
melanoma
metastasis
Lymph Node
not known
22
male
Melanoma
PDX
MEXF 394
not known
melanoma
metastasis
Skin
not known
70
male
Melanoma
PDX
MEXF 462
M1
melanoma
metastasis
Skin
not known
63
male
Melanoma
PDX
MEXF 514
N1
melanoma
metastasis
Lymph Node
not known
40
female
Melanoma
PDX
MEXF 520
M1
melanoma
metastasis
Skin
not known
41
male
Melanoma
PDX
MEXF 535
N1
melanoma
metastasis
Lymph Node
not known
55
male
Melanoma
PDX
MEXF 622
M1
melanoma
metastasis
Lung
not known
68
male
Melanoma
PDX
MEXF 666
M1
melanoma
metastasis
Skin
not known
67
male
Melanoma
PDX
MEXF 672
N1
melanoma
metastasis
Lymph Node
not known
25
male
Melanoma
PDX
MEXF 989
M1
melanoma
metastasis
Lymph Node
not known
32
male
Ovary
PDX
OVXF 1353
N1
serous adenocarcinoma
metastasis
Lymph Node
poorly differentiated
41
female
Ovary
PDX
OVXF 550
not known
serous adenocarcinoma
not known
not known
poorly differentiated
not known
female
Ovary
PDX
OVXF 899
T3N0M0
serous adenocarcinoma
primary
ovary
well differentiated
76
female
Pancreas
PDX
PAXF 1657
M1
adenocarcinoma
metastasis
Lung
moderately differentiated
60
female
Pancreas
PDX
PAXF 736
M1
adenocarcinoma
recurrence
Pancreas
poorly differentiated
65
male
Kidney
PDX
RXF 1220
T3M1
hypernephroma
metastasis
Lung
not known
59
male
Kidney
PDX
RXF 1393
T4N0Mx G2
hypernephroma
metastasis
Lung
not known
56
male
Kidney
PDX
RXF 631
M1
hypernephroma
metastasis
Lung
not known
54
male
Hematologic tumors
Leukemia
CDX
LEXF-K-562
not known
chronic myelocytic leukemia
not known
not known
poorly differentiated
53
female
Lymphoma
CDX
LYXF-MYLA
not known
T cell cutaneous
not known
not known
well differentiated
82
male
Lymphoma
CDX
LYXF-RAJI
not known
B lymphocyte
not known
not known
poorly differentiated
11
male
Lymphoma
CDX
LYXF-U-937
not known
histiocystic lymphoma
not known
not known
poorly differentiated
37
male
Myeloma
CDX
MMXF-L-363
not known
multiple myeloma
not known
not known
poorly differentiated
36
female
The efficacy of galunisertib was assessed in a first series of experiments in a panel of 66 tumor models. The compound inhibited colony growth in 1/66 samples (1.5 %) in a concentration-dependent manner when tested up to 10 μM. Growth stimulation was observed in 15/66 (22.7 %) of the samples and no response was seen in 50/66 (75.8 %) of the samples. When tested up to 80 μM, galunisertib inhibited colony growth in 5/24 (20.8 %) selected samples. Stimulation of colony formation was observed in 10/24 (41.7 %) of these samples and no response was found in 9/24 (37.5 %) of the samples (Fig. 1a). The most sensitive xenograft samples were CXF 742 (colon cancer), LXFS 650 and LXFE 1422 (small cell lung cancer), and the hematologic xenograft samples LYXF MYLA (T-ALL) and LYXF RAJI (Burkitt’s lymphoma). No correlation between responses and histopathological characteristics of the different samples was observed (Fig. 1a).
We also assessed galunisertib in a second series of experiments in panel of melanoma PDX (Fig. 1b). This melanoma panel was chosen because of previous reports suggesting that TGF-β1 signaling is an autocrine activation pathway for tumor cell growth in melanoma [3032]. We found that galunisertib did not have any inhibitory effect in this panel of melanoma xenografts, i.e., when tested up to 10 μM galunisertib elicited no response in 12/17 (70.6 %) of the samples, whereas stimulation of colony formation was observed in 5/17 (29.4 %) of the samples. At a higher concentration (30 μM), no response was observed in 11/17 (64.7 %) of the samples, whereas stimulation of colony formation was observed in 6/17 (35.3 %) of the samples.

3.2 Molecular characteristics of tumor models investigated ex vivo

Because of the limited inhibitory effects observed in the clonogenic assays, we set out to investigate whether the canonical TGF-β signaling pathway is altered in these PDX. To this end, we assessed the expression level and mutation status of genes associated with the canonical (i.e., the TGF-β1, TGF-β2, SMAD2, SMAD3, SMAD4, SMAD7, TGF-βRI and TGF-βRII genes) as well as the non-canonical (i.e., the MAPK and AKT genes) TGF-β signaling pathways. The aims were (i) to characterize the molecular profiles of these pathways and (ii) to evaluate whether the respective genes predicted drug sensitivity. We also investigated the expression of two proteins that were previously reported to be associated with TGF-β -mediated drug resistance, i.e., TP53 and MED12 [33].
The copy numbers of the TGF-β1, TGF-β2, TGF-βR1 and TGF-βR2 genes were assessed in 70/79 PDX samples. No major rearrangements were observed. Two samples showed mutations in TGF-β1: CXF 260, A350V and MEXF 989, S138L. Two samples showed mutations in TGF-β2: CXF 269, S365R, and CXF 260, P387H. Three samples showed mutations in TGF-βR1: MAXF 401, E242D, CXF 260, A125V and LXFA 737, E111K. All these mutations were found in the non-responder group. TGF-βR2 was mutated in 4 samples, including one from the group that showed growth inhibition (LXFE 1422,153X [HGVS nomenclature for frameshift]), one from the group that showed growth stimulation (LXFA 1041, N384T) and two from the group that showed no response (LXFA 526, 153X and CXF 1103, 128 fs, C393F). No associations were found between the mutation status of the PDX samples and the responses to galunisertib (data not shown).
Next, we set out to investigate the mRNA expression levels of the TGF-β1, TGF-β2, TGF-βR1 and TGF-βR2 genes in 77/79 PDX samples (Fig. 2, Panel a). Heterogeneous mRNA expression levels were found for these 4 genes in the samples tested, and no significant associations were found between the mRNA expression levels and the responses to galunisertib. Despite this variability, however, some general trends were observed: (i) the expression levels of the TGF-β1 and TGF-β2 genes were generally low or undetectable, (ii) the TGF-βR1 gene was well expressed in most of the samples tested, whereas the TGF-βR2 gene was expressed at a low level or undetectable in at least some of the samples (Fig. 2, panel a).
We also assessed the status of the TGF-β1 downstream canonical activation pathway (SMAD-dependent activation). No major gene copy number alterations were found in the samples tested, except in LXFA 749 (deletion of SMAD3) and in LIXF 575 (deletion of SMAD4), both from the “stimulated” group. Sequence analysis revealed mutations in SMAD1 (CXF 260, A262V, not shown) and SMAD2 (LXFE 397, S287C). No mutations were found in SMAD3. SMAD4 was the most frequently mutated gene with 9 mutations in samples from the “no response” (7/9) and the “stimulated” (2/9) groups. SMAD6 was mutated in 3 samples of the “inhibited” and the “no response” groups (LXFS 650, L192P; CXF 260, D359G and LXFA 1012, P323L; not shown). SMAD7 was mutated in CXF 260 (A159V and D113G) and MAXF 449 (R131C). All SMAD genes (SMAD1, 2, 3, 4, 6 and 7) showed heterogeneous expression patterns, but these patterns were not found to be associated with a response to galunisertib (Fig. 2). Of note, SMAD7 expression was nearly absent in most samples, whereas SMAD2, SMAD3 and SMAD4 were expressed at similar levels, with similar minimum to maximum ranges (Fig. 2b), and variable patterns in most samples.
Subsequently, we subjected some non-canonical or SMAD-independent genes [4, 33] to genomic and transcriptomic analyses. No major genomic alterations in the AKT1 gene were discovered, and AKT1 mRNA expression was detected in most of the samples tested (range 5.5-11 Units [U], mean 7.9 U). There was no significant association with respons to galunisertib. The MAPK1 gene was found to be expressed in 30 % of the samples investigated, and no associations between its expression and galunisertib responses were observed. E-cadherin (CDH1) expression levels were observed in most of the samples tested (Fig. 3).
We next investigated the status of some genes presumed to be associated with drug resistance in relation to TGF-β signaling (Fig. 3). No overt MED12 gene copy number gains were detected. In 21 samples, however, loss of one MED12 gene copy was noted without any observable alteration in mRNA expression. MED12 gene mutations were detected in 8 samples, of which 6 exhibited mRNA expression levels that were below the detection threshold. None of the alterations observed were found to be associated with a galunisertib response. Of note, TP53 mutations were frequently encountered in most of the samples tested (not shown), but these mutations could not significantly be associated with responses to galunisertib (Kruskal-Wallis p-value = 0.13).
Based on the altered profiles observed in the TGF-β signaling cascade, we decided to further assess the expression of the pSMAD2 protein in order to substantiate the activated status of the TGF-β signaling pathway (Fig. 4) in the PDX samples before treatment. Most samples tested indeed showed pSMAD2 expression, and in some cases this expression was observed in spite of the fact that the TGF-βR1 protein levels were either low or undetectable.

3.3 Galunisertib elicits in vivo anti-tumor activity

From the 79 samples included in this study, 13 were selected for in vivo analyses. We used ABC calculations to assess the efficacy of galunisertib treatment. By doing so, we found that the ABC values ranked from −23.63 % (stimulation of tumor growth) to 28.32 % (inhibition of tumor growth) (Fig. 5a). The mRNA expression levels of genes involved in the TGF-β pathway were compared to the calculated ABC values (Fig. 5a and b). Tumor growth inhibition was seen in 2 of 13 (15.4 %) selected samples (i.e., lung cancer PDX LXFA 737 and prostate cancer PDX PRXF MRIH 1579). No changes were seen in 8/13 (61.5 %) samples. In 3/13 (23.1 %) samples, tumor growth stimulation was observed. Next, we assessed a possible association between the in vivo responses observed and the concomitant TGF-β1 and SMAD2 gene expression levels, or activation of the TGF-β signaling pathway by measuring pSMAD2 protein levels (Fig. 5a and c). Although the TGF-β1 and SMAD2 gene expression levels were not associated with the in vivo response, high TGF-βRI transcript levels and pSMAD2 protein levels were found to be correlated with the in vivo anti-tumor responses (Spearman correlation 0.67, p-value = 0.017 and Spearman correlation 0.68, p-value = 0.025, respectively).

4 Discussion

In the present study, we observed in vitro and in vivo differences between the anti-tumor activities of the TGF-β receptor kinase inhibitor galunisertib. We also observed a lack of broad activity across the tumor panel tested, which is in stark contrast to other mostly cytotoxic anti-cancer agents. These observations are unusual for anti-cancer agents and, given the activity of galunisertib observed in patients [8], we hypothesized that the tissue microenvironment might play a critical role in supporting TGF-β-dependent tumor growth. Our current observations can be explained in several ways. First, the activity of galunisertib in the tumor clonogenic assay may be dependent on its selectivity. Galunisertib is known to selectively target TGF-βRI (ALK5) at concentrations up to 1 μM [6, 34]. Concentrations above 10 μM are not considered to be given to humans because of the possible toxic implications of this drug [35, 36]. In our current study, however, we used concentrations up to 80 μM in order to assess its effects beyond the pharmacological range. By doing so, we found no overt differences between the higher and the lower concentrations. Thus, the selectivity of the drug appeared to have little or no effect in this study. Only 5 PDX showed small inhibitory effects, not related to a specific tumor type. None of the other tumors responded to galunisertib, including melanomas, which are presumed to grow using a TGF-β-dependent autocrine loop. We grouped the galunisertib responses into three categories: “inhibited”, “no response” or “stimulated”. Second, we set out to understand why the majority of the PDX either exhibited “no response” or a “stimulated” response in vivo. Therefore, we assessed whether either the canonical or the non-canonical TGF-β1signaling pathway was intact. Canonical, SMAD-dependent TGF-β signaling was found to be defective in 30 % of the PDX samples. From this result we conclude that galunisertib may have acted on kinases for which it was not designed, resulting in unspecific activity in the clonogenic assays. TGF-βR1 and TGF-βR2 transcripts were detected in most of the PDX samples, whereas TGF-β1 and TGF-β2 transcripts were only detected at a low level and in half of the PDX. In such PDX samples autocrine loops may be active, in spite of the fact that the TGF-β1 and TGF-β2 levels were low. The SMAD1, SMAD2 and SMAD4 genes were all expressed. Only the SMAD4 gene was found to be frequently mutated, while the others were rarely affected. Noteworthy, inhibitory effects of galunisertib were observed in those samples that expressed the TGF-βR1 and SMAD4 genes. Hence, the responses could not be traced back to a specific or intact signaling pathway, either canonical or non-canonical. Third, based on the clonogenic assay, the anti-tumor activity of galunisertib or other agents targeting the tumor microenvironment are underestimated because such agents will not show cell killing, but will arrest tumors by inhibition of cell migration and invasion [3739]. It is also possible that galunisertib targets signals that are not tumor cell-dependent, such as immune- or other microenvironment signals, as has been shown in a syngeneic mouse model in which a monoclonal antibody against TGF-βRII showed anti-tumor activity [40]. This anti-tumor effect appeared to be dependent on the presence of specific immune cells such as CD8-positive T-cells and NK cells. We also tested galunisertib in vivo in 13 of the PDX samples. Two of these samples (LXFA 737 and PRXF MRIH 1579) showed growth inhibition (Fig. 5, Fig. S1, S2, Table S1 and S2). Given the presence of residual NK cell functions in nude mice, we assume that in these two models blocking of TGF-β-dependent signaling has facilitated NK activity against tumor cells. In addition, we noted that the PDX samples investigated contained variable amounts of human tumor cells and murine stroma components, which may contain a fibroblast-rich microenvironment [41] or genomically instable tumor cells [42]. Although the amounts of stroma in the individual samples did not directly correlate with the response rates in vivo, we found that the presence of stroma not only affects the response rate in general, but also that the in vivo response patterns correlated better with the molecular characteristics of the PDX models (i.e., pSMAD protein and TGF-βR1 mRNA expression).
In conclusion, it appears that models that reflect the normal physiological condition, including all aspects of the microenvironment, are more useful to assess TGF-β1 inhibitors. In absence of such models, results from such experiments must be interpreted with caution.

Acknowledgments

The authors acknowledge the medical writing assistance of Durisala Desaiah, PhD., Eli Lilly and Company for the revisions and formatting of the manuscript.

Disclosure

Michael Lahn was a former employee of Eli Lilly and Company and holds stock. Eli Lilly and Company is currently developing galunisertib for indications in oncology.
Armin Maier, Anne-Lise Peille and Vincent Vuaroqueaux are employed by Oncotest. Oncotest was contracted to perform the present studies by Eli Lilly and Company.
Open Access This article is distributed under the terms of the Creative Commons Attribution License which permits any use, distribution, and reproduction in any medium, provided the original author(s) and the source are credited.

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.

Anhänge

Electronic supplementary material

Below is the link to the electronic supplementary material.
Literatur
1.
Zurück zum Zitat A.B.. Roberts, M.A. Anzano, L.C. Lamb, J.M. Smith, M.B. Sporn, New class of transforming growth factors potentiated by epidermal growth factor: isolation from non-neoplastic tissues. Proc. Natl. Acad. Sci. U. S. A. 78, 5339–5343 (1981) A.B.. Roberts, M.A. Anzano, L.C. Lamb, J.M. Smith, M.B. Sporn, New class of transforming growth factors potentiated by epidermal growth factor: isolation from non-neoplastic tissues. Proc. Natl. Acad. Sci. U. S. A. 78, 5339–5343 (1981)
3.
Zurück zum Zitat C.H. Heldin, A. Moustakas, Role of Smads in TGFβ signaling. Cell Tissue Res. 347, 21–36 (2012)CrossRefPubMed C.H. Heldin, A. Moustakas, Role of Smads in TGFβ signaling. Cell Tissue Res. 347, 21–36 (2012)CrossRefPubMed
4.
Zurück zum Zitat R. Derynck, Y.E. Zhang, Smad-dependent and Smad-independent pathways in TGF-beta family signalling. Nature 425, 577–584 (2003)CrossRefPubMed R. Derynck, Y.E. Zhang, Smad-dependent and Smad-independent pathways in TGF-beta family signalling. Nature 425, 577–584 (2003)CrossRefPubMed
5.
Zurück zum Zitat M. Lahn, S. Kloeker, B.S. Berry, TGF-beta inhibitors for the treatment of cancer. Expert Opin. Investig. Drugs 14, 629–643 (2005)CrossRefPubMed M. Lahn, S. Kloeker, B.S. Berry, TGF-beta inhibitors for the treatment of cancer. Expert Opin. Investig. Drugs 14, 629–643 (2005)CrossRefPubMed
6.
Zurück zum Zitat J.S. Sawyer, D.W. Beight, K.S. Britt, B.D. Anderson, R.M. Campbell, T. Goodson Jr., D.K. Herron, H.Y. Li, W.T. McMillen, N. Mort, S. Parsons, E.C. Smith, J.R. Wagner, L. Yan, F. Zhang, J.M. Yingling, Synthesis and activity of new aryl- and heteroaryl-substituted 5,6-dihydro-4H-pyrrolo[1,2-b]pyrazole inhibitors of the transforming growth factor-beta type I receptor kinase domain. Bioorg. Med. Chem. Lett. 14, 3581–3584 (2004)CrossRefPubMed J.S. Sawyer, D.W. Beight, K.S. Britt, B.D. Anderson, R.M. Campbell, T. Goodson Jr., D.K. Herron, H.Y. Li, W.T. McMillen, N. Mort, S. Parsons, E.C. Smith, J.R. Wagner, L. Yan, F. Zhang, J.M. Yingling, Synthesis and activity of new aryl- and heteroaryl-substituted 5,6-dihydro-4H-pyrrolo[1,2-b]pyrazole inhibitors of the transforming growth factor-beta type I receptor kinase domain. Bioorg. Med. Chem. Lett. 14, 3581–3584 (2004)CrossRefPubMed
7.
Zurück zum Zitat F. Dituri, A. Mazzocca, F.J. Peidro, P. Papappicco, I. Fabregat, F. De Santis, A. Paradiso, C. Sabba, G. Giannelli, Differential inhibition of the TGF-β signaling pathway in HCC cells using the small molecule inhibitor LY2157299 and the D10 monoclonal antibody against TGF-β receptor type II. PLoS ONE 8, e67109 (2013)CrossRefPubMedCentralPubMed F. Dituri, A. Mazzocca, F.J. Peidro, P. Papappicco, I. Fabregat, F. De Santis, A. Paradiso, C. Sabba, G. Giannelli, Differential inhibition of the TGF-β signaling pathway in HCC cells using the small molecule inhibitor LY2157299 and the D10 monoclonal antibody against TGF-β receptor type II. PLoS ONE 8, e67109 (2013)CrossRefPubMedCentralPubMed
8.
Zurück zum Zitat J. Rodon, M.A. Carducci, J.M. Sepúlveda, A. Azaro, E. Calvo, J. Seoane, I. Brana, E. Sicart, I. Gueorguieva, A. Cleverly, S. Pillay, D. Desaiah, M.M.F. Lahn, L. Paz-Ares, M. Holdhoff, J.O.N. Blakeley, J. Baselga, First-in-human dose study of the novel transforming growth factor-β receptor I kinase inhibitor LY2157299 monohydrate in patients with advanced cancer and glioma. Clin. Canc. Res. In Press (2014) J. Rodon, M.A. Carducci, J.M. Sepúlveda, A. Azaro, E. Calvo, J. Seoane, I. Brana, E. Sicart, I. Gueorguieva, A. Cleverly, S. Pillay, D. Desaiah, M.M.F. Lahn, L. Paz-Ares, M. Holdhoff, J.O.N. Blakeley, J. Baselga, First-in-human dose study of the novel transforming growth factor-β receptor I kinase inhibitor LY2157299 monohydrate in patients with advanced cancer and glioma. Clin. Canc. Res. In Press (2014)
9.
Zurück zum Zitat S.J. Faivre, A. Santoro, R.K. Kelley, P. Merle, E. Gane, J.-Y. Douillard, D. Waldschmidt, M.F. Mulcahy, C. Costentin, B. Minguez, P. Papappicco, I. Gueorguieva, A. Cleverly, D. Desaiah, M.M.F. Lahn, N. Murray, K.A. Benhadji, E. Raymond, G. Giannelli, Randomized dose comparison phase II study of the oral transforming growth factor-beta (TGF-ß) receptor I kinase inhibitor LY2157299 monohydrate (LY) in patients with advanced hepatocellular carcinoma (HCC). ASCO Gastrointestinal Cancers Symposium, (2014) S.J. Faivre, A. Santoro, R.K. Kelley, P. Merle, E. Gane, J.-Y. Douillard, D. Waldschmidt, M.F. Mulcahy, C. Costentin, B. Minguez, P. Papappicco, I. Gueorguieva, A. Cleverly, D. Desaiah, M.M.F. Lahn, N. Murray, K.A. Benhadji, E. Raymond, G. Giannelli, Randomized dose comparison phase II study of the oral transforming growth factor-beta (TGF-ß) receptor I kinase inhibitor LY2157299 monohydrate (LY) in patients with advanced hepatocellular carcinoma (HCC). ASCO Gastrointestinal Cancers Symposium, (2014)
10.
Zurück zum Zitat S. Parsons, S. Sawyer, L. Yan, R. Foreman, S. Weir, M.M.F. Lahn, J. Yingling and J.R. Graff, The combination of the small molecule TGFβR1 inhibitor LY2157299 monohydrate with CCNU substantially blocks SMAD phosphorylation and significantly suppresses human glioblastoma xenograft growth. Abstract C201. Poster Presentation. Molecular Cancer Therapeutics: November 2011; Volume 10, Issue 11, Supplement 1. doi: 10.1158/1535-7163.TARG-11-C201; San Francisco, CA. Abstracts: AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics--Nov 12–16, (2011) S. Parsons, S. Sawyer, L. Yan, R. Foreman, S. Weir, M.M.F. Lahn, J. Yingling and J.R. Graff, The combination of the small molecule TGFβR1 inhibitor LY2157299 monohydrate with CCNU substantially blocks SMAD phosphorylation and significantly suppresses human glioblastoma xenograft growth. Abstract C201. Poster Presentation. Molecular Cancer Therapeutics: November 2011; Volume 10, Issue 11, Supplement 1. doi: 10.​1158/​1535-7163.​TARG-11-C201; San Francisco, CA. Abstracts: AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics--Nov 12–16, (2011)
11.
Zurück zum Zitat E.C. Connolly, E.F. Saunier, D. Quigley, M.T. Luu, A. De Sapio, B. Hann, J.M. Yingling, R.J. Akhurst, Outgrowth of drug-resistant carcinomas expressing markers of tumor aggression after long-term TβRI/II kinase inhibition with LY2109761. Cancer Res. 71, 2339–2349 (2011)CrossRefPubMedCentralPubMed E.C. Connolly, E.F. Saunier, D. Quigley, M.T. Luu, A. De Sapio, B. Hann, J.M. Yingling, R.J. Akhurst, Outgrowth of drug-resistant carcinomas expressing markers of tumor aggression after long-term TβRI/II kinase inhibition with LY2109761. Cancer Res. 71, 2339–2349 (2011)CrossRefPubMedCentralPubMed
12.
Zurück zum Zitat D.P. Berger, H. Henss, B.R. Winterhalter, H.H. Fiebig, The clonogenic assay with human tumor xenografts: evaluation, predictive value and application for drug screening. Ann. Oncol. 1, 333–341 (1990)PubMed D.P. Berger, H. Henss, B.R. Winterhalter, H.H. Fiebig, The clonogenic assay with human tumor xenografts: evaluation, predictive value and application for drug screening. Ann. Oncol. 1, 333–341 (1990)PubMed
13.
Zurück zum Zitat H.H. Fiebig, in In Immunodeficient mice in oncology, ed. By D. P. Berger (S Karger, Basel, 1992), p. 321–351 H.H. Fiebig, in In Immunodeficient mice in oncology, ed. By D. P. Berger (S Karger, Basel, 1992), p. 321–351
14.
Zurück zum Zitat S. Julien, A. Merino-Trigo, L. Lacroix, M. Pocard, D. Goere, P. Mariani, S. Landron, L. Bigot, F. Nemati, P. Dartigues, L.B. Weiswald, D. Lantuas, L. Morgand, E. Pham, P. Gonin, V. Dangles-Marie, B. Job, P. Dessen, A. Bruno, A. Pierre, H. De The, H. Soliman, M. Nunes, G. Lardier, L. Calvet, B. Demers, G. Prevost, P. Vrignaud, S. Roman-Roman, O. Duchamp, C. Berthet, Characterization of a large panel of patient-derived tumor xenografts representing the clinical heterogeneity of human colorectal cancer. Clin. Cancer Res. 18, 5314–5328 (2012)CrossRefPubMed S. Julien, A. Merino-Trigo, L. Lacroix, M. Pocard, D. Goere, P. Mariani, S. Landron, L. Bigot, F. Nemati, P. Dartigues, L.B. Weiswald, D. Lantuas, L. Morgand, E. Pham, P. Gonin, V. Dangles-Marie, B. Job, P. Dessen, A. Bruno, A. Pierre, H. De The, H. Soliman, M. Nunes, G. Lardier, L. Calvet, B. Demers, G. Prevost, P. Vrignaud, S. Roman-Roman, O. Duchamp, C. Berthet, Characterization of a large panel of patient-derived tumor xenografts representing the clinical heterogeneity of human colorectal cancer. Clin. Cancer Res. 18, 5314–5328 (2012)CrossRefPubMed
15.
Zurück zum Zitat S. Kopetz, R. Lemos, G. Powis, The promise of patient-derived xenografts: the best laid plans of mice and men. Clin. Cancer Res. 18, 5160–5162 (2012)CrossRefPubMedCentralPubMed S. Kopetz, R. Lemos, G. Powis, The promise of patient-derived xenografts: the best laid plans of mice and men. Clin. Cancer Res. 18, 5160–5162 (2012)CrossRefPubMedCentralPubMed
16.
Zurück zum Zitat D.D. von Hoff, Human tumor cloning assays: applications in clinical oncology and new antineoplastic agent development. Cancer Metastasis Rev. 7, 357–371 (1988)CrossRef D.D. von Hoff, Human tumor cloning assays: applications in clinical oncology and new antineoplastic agent development. Cancer Metastasis Rev. 7, 357–371 (1988)CrossRef
17.
Zurück zum Zitat R. Krumbach, J. Schuler, M. Hofmann, T. Giesemann, H.H. Fiebig, T. Beckers, Primary resistance to cetuximab in a panel of patient-derived tumour xenograft models: activation of MET as one mechanism for drug resistance. Eur. J. Cancer 47, 1231–1243 (2011)CrossRefPubMed R. Krumbach, J. Schuler, M. Hofmann, T. Giesemann, H.H. Fiebig, T. Beckers, Primary resistance to cetuximab in a panel of patient-derived tumour xenograft models: activation of MET as one mechanism for drug resistance. Eur. J. Cancer 47, 1231–1243 (2011)CrossRefPubMed
18.
Zurück zum Zitat H.H. Fiebig, V. Vuaroqueaux, A. Korrat, F. Foucault, T. Beckers, Predictive gene signatures for bevacizumab and cetuximab as well as cytotoxic agents. Int. J. Clin. Pharmacol. Ther. 50, 70–71 (2012)CrossRefPubMed H.H. Fiebig, V. Vuaroqueaux, A. Korrat, F. Foucault, T. Beckers, Predictive gene signatures for bevacizumab and cetuximab as well as cytotoxic agents. Int. J. Clin. Pharmacol. Ther. 50, 70–71 (2012)CrossRefPubMed
19.
Zurück zum Zitat M. Singh, N. Ferrara, Modeling and predicting clinical efficacy for drugs targeting the tumor milieu. Nat. Biotechnol. 30, 648–657 (2012)CrossRefPubMed M. Singh, N. Ferrara, Modeling and predicting clinical efficacy for drugs targeting the tumor milieu. Nat. Biotechnol. 30, 648–657 (2012)CrossRefPubMed
20.
Zurück zum Zitat C.C. Scholz, D.P. Berger, B.R. Winterhalter, H. Henss, H.H. Fiebig, Correlation of drug response in patients and in the clonogenic assay with solid human tumour xenografts. Eur. J. Cancer 26, 901–905 (1990)CrossRefPubMed C.C. Scholz, D.P. Berger, B.R. Winterhalter, H. Henss, H.H. Fiebig, Correlation of drug response in patients and in the clonogenic assay with solid human tumour xenografts. Eur. J. Cancer 26, 901–905 (1990)CrossRefPubMed
21.
Zurück zum Zitat H.H. Fiebig, A. Maier, A.M. Burger, Clonogenic assay with established human tumour xenografts: correlation of in vitro to in vivo activity as a basis for anticancer drug discovery. Eur. J. Cancer 40, 802–820 (2004)CrossRefPubMed H.H. Fiebig, A. Maier, A.M. Burger, Clonogenic assay with established human tumour xenografts: correlation of in vitro to in vivo activity as a basis for anticancer drug discovery. Eur. J. Cancer 40, 802–820 (2004)CrossRefPubMed
22.
Zurück zum Zitat H.H. Fiebig, W.A. Dengler and T. Roth. Human tumor xenografts: predictivity, characterization and discovery of new anticancer agents. In Relevance of tumor models for anticancer drug development., ed. By H.H. Fiebig and A. M. Burger (S. Karger, Basel, 1999), p. 29–50 H.H. Fiebig, W.A. Dengler and T. Roth. Human tumor xenografts: predictivity, characterization and discovery of new anticancer agents. In Relevance of tumor models for anticancer drug development., ed. By H.H. Fiebig and A. M. Burger (S. Karger, Basel, 1999), p. 29–50
23.
Zurück zum Zitat A.W. Hamburger, S.E. Salmon, Primary bioassay of human tumor stem cells. Science 197, 461–463 (1977)CrossRefPubMed A.W. Hamburger, S.E. Salmon, Primary bioassay of human tumor stem cells. Science 197, 461–463 (1977)CrossRefPubMed
24.
Zurück zum Zitat M.C. Alley, C.B. Uhl, M.M. Lieber, Improved detection of drug cytotoxicity in the soft agar colony formation assay through use of a metabolizable tetrazolium salt. Life Sci. 31, 3071–3078 (1982)CrossRefPubMed M.C. Alley, C.B. Uhl, M.M. Lieber, Improved detection of drug cytotoxicity in the soft agar colony formation assay through use of a metabolizable tetrazolium salt. Life Sci. 31, 3071–3078 (1982)CrossRefPubMed
25.
Zurück zum Zitat Q. Li, N.J. Birkbak, B. Gyorffy, Z. Szallasi, A.C. Eklund, Jetset: selecting the optimal microarray probe set to represent a gene. BMC Bioinforma. 12, 474 (2011)CrossRef Q. Li, N.J. Birkbak, B. Gyorffy, Z. Szallasi, A.C. Eklund, Jetset: selecting the optimal microarray probe set to represent a gene. BMC Bioinforma. 12, 474 (2011)CrossRef
26.
Zurück zum Zitat C.D. Greenman, G. Bignell, A. Butler, S. Edkins, J. Hinton, D. Beare, S. Swamy, T. Santarius, L. Chen, S. Widaa, P.A. Futreal, M.R. Stratton, PICNIC: an algorithm to predict absolute allelic copy number variation with microarray cancer data. Biostatistics 11, 164–175 (2010)CrossRefPubMedCentralPubMed C.D. Greenman, G. Bignell, A. Butler, S. Edkins, J. Hinton, D. Beare, S. Swamy, T. Santarius, L. Chen, S. Widaa, P.A. Futreal, M.R. Stratton, PICNIC: an algorithm to predict absolute allelic copy number variation with microarray cancer data. Biostatistics 11, 164–175 (2010)CrossRefPubMedCentralPubMed
27.
Zurück zum Zitat A. Monks, D. Scudiero, P. Skehan, R. Shoemaker, K. Paull, D. Vistica, C. Hose, J. Langley, P. Cronise, A. Vaigro-Wolff, M. Gray-Goodrich, H. Campbell, J. Mayo, M. Boyd, Feasibility of a high-flux anticancer drug screen using a diverse panel of cultured human tumor cell lines. J. Natl. Cancer Inst. 83, 757 (1991)CrossRefPubMed A. Monks, D. Scudiero, P. Skehan, R. Shoemaker, K. Paull, D. Vistica, C. Hose, J. Langley, P. Cronise, A. Vaigro-Wolff, M. Gray-Goodrich, H. Campbell, J. Mayo, M. Boyd, Feasibility of a high-flux anticancer drug screen using a diverse panel of cultured human tumor cell lines. J. Natl. Cancer Inst. 83, 757 (1991)CrossRefPubMed
28.
Zurück zum Zitat R.H. Shoemaker, The NCI60 human tumour cell line anticancer drug screen. Nat. Rev. Cancer 6, 813 (2006)CrossRefPubMed R.H. Shoemaker, The NCI60 human tumour cell line anticancer drug screen. Nat. Rev. Cancer 6, 813 (2006)CrossRefPubMed
29.
Zurück zum Zitat Maier A, Metz T, Krumbach R, Fiebig HH, Beckers T, In vitro profiling of kinase inhibitors and targeted anticancer agents in panels of patient-derived xenografts using a clonogenic assay. Proc. Am. Assoc. Cancer Res. 52, abstract 3528 (2011) Maier A, Metz T, Krumbach R, Fiebig HH, Beckers T, In vitro profiling of kinase inhibitors and targeted anticancer agents in panels of patient-derived xenografts using a clonogenic assay. Proc. Am. Assoc. Cancer Res. 52, abstract 3528 (2011)
30.
Zurück zum Zitat L.A. Liotta, R. Mandler, G. Murano, D.A. Katz, R.K. Gordon, P.K. Chiang, E. Schiffmann, Tumor cell autocrine motility factor. Proc. Natl. Acad. Sci. U. S. A. 83, 3302–3306 (1986)CrossRefPubMedCentralPubMed L.A. Liotta, R. Mandler, G. Murano, D.A. Katz, R.K. Gordon, P.K. Chiang, E. Schiffmann, Tumor cell autocrine motility factor. Proc. Natl. Acad. Sci. U. S. A. 83, 3302–3306 (1986)CrossRefPubMedCentralPubMed
31.
Zurück zum Zitat U. Rodeck, T. Nishiyama, A. Mauviel, Independent regulation of growth and SMAD-mediated transcription by transforming growth factor beta in human melanoma cells. Cancer Res. 59, 547–550 (1999)PubMed U. Rodeck, T. Nishiyama, A. Mauviel, Independent regulation of growth and SMAD-mediated transcription by transforming growth factor beta in human melanoma cells. Cancer Res. 59, 547–550 (1999)PubMed
32.
Zurück zum Zitat K. Krasagakis, C. Garbe, P.I. Schrier, C.E. Orfanos, Paracrine and autocrine regulation of human melanocyte and melanoma cell growth by transforming growth factor beta in vitro. Anticancer Res. 14, 2565–2571 (1994)PubMed K. Krasagakis, C. Garbe, P.I. Schrier, C.E. Orfanos, Paracrine and autocrine regulation of human melanocyte and melanoma cell growth by transforming growth factor beta in vitro. Anticancer Res. 14, 2565–2571 (1994)PubMed
33.
Zurück zum Zitat S. Huang, M. Holzel, T. Knijnenburg, A. Schlicker, P. Roepman, U. McDermott, M. Garnett, W. Grernrum, C. Sun, A. Prahallad, F.H. Groenendijk, L. Mittempergher, W. Nijkamp, J. Neefjes, R. Salazar, P. Ten Dijke, H. Uramoto, F. Tanaka, R.L. Beijersbergen, L.F. Wessels, R. Bernards, MED12 controls the response to multiple cancer drugs through regulation of TGF-β receptor signaling. Cell 151, 937–950 (2012)CrossRefPubMedCentralPubMed S. Huang, M. Holzel, T. Knijnenburg, A. Schlicker, P. Roepman, U. McDermott, M. Garnett, W. Grernrum, C. Sun, A. Prahallad, F.H. Groenendijk, L. Mittempergher, W. Nijkamp, J. Neefjes, R. Salazar, P. Ten Dijke, H. Uramoto, F. Tanaka, R.L. Beijersbergen, L.F. Wessels, R. Bernards, MED12 controls the response to multiple cancer drugs through regulation of TGF-β receptor signaling. Cell 151, 937–950 (2012)CrossRefPubMedCentralPubMed
34.
Zurück zum Zitat H.Y. Li, Y. Wang, L. Yan, R.M. Campbell, B.D. Anderson, J.R. Wagner, J.M. Yingling, Novel and potent transforming growth factor beta type I receptor kinase domain inhibitor: 7-amino 4-(2-pyridin-2-yl-5,6-dihydro-4H-pyrrolo[1,2-b]pyrazol-3-yl)-quinolines. Bioorg. Med. Chem. Lett. 14, 3585–3588 (2004)CrossRefPubMed H.Y. Li, Y. Wang, L. Yan, R.M. Campbell, B.D. Anderson, J.R. Wagner, J.M. Yingling, Novel and potent transforming growth factor beta type I receptor kinase domain inhibitor: 7-amino 4-(2-pyridin-2-yl-5,6-dihydro-4H-pyrrolo[1,2-b]pyrazol-3-yl)-quinolines. Bioorg. Med. Chem. Lett. 14, 3585–3588 (2004)CrossRefPubMed
35.
Zurück zum Zitat A.J. Stauber, J.L. Zimmermann and B.R. Berridge, Pathobiology of a Valvulopathy in Fischer 344 Rats Given a Transforming Growth Factor-b RI Kinase Inhibitor. Abstract 290. 45th Annual Meeting and Tox Expo™, San Diego, California, Oxford University Press (2006), p 59, Abstract # 290 (2006) A.J. Stauber, J.L. Zimmermann and B.R. Berridge, Pathobiology of a Valvulopathy in Fischer 344 Rats Given a Transforming Growth Factor-b RI Kinase Inhibitor. Abstract 290. 45th Annual Meeting and Tox Expo, San Diego, California, Oxford University Press (2006), p 59, Abstract # 290 (2006)
36.
Zurück zum Zitat M.J. Anderton, H.R. Mellor, A. Bell, C. Sadler, M. Pass, S. Powell, S.J. Steele, R.R. Roberts, A. Heier, Induction of heart valve lesions by small-molecule ALK5 inhibitors. Toxicol. Pathol. 39, 916–924 (2011)CrossRefPubMed M.J. Anderton, H.R. Mellor, A. Bell, C. Sadler, M. Pass, S. Powell, S.J. Steele, R.R. Roberts, A. Heier, Induction of heart valve lesions by small-molecule ALK5 inhibitors. Toxicol. Pathol. 39, 916–924 (2011)CrossRefPubMed
37.
Zurück zum Zitat E. Fransvea, U. Angelotti, S. Antonaci, G. Giannelli, Blocking transforming growth factor-beta up-regulates E-cadherin and reduces migration and invasion of hepatocellular carcinoma cells. Hepatology 47, 1557–1566 (2008)CrossRefPubMed E. Fransvea, U. Angelotti, S. Antonaci, G. Giannelli, Blocking transforming growth factor-beta up-regulates E-cadherin and reduces migration and invasion of hepatocellular carcinoma cells. Hepatology 47, 1557–1566 (2008)CrossRefPubMed
38.
Zurück zum Zitat E. Fransvea, A. Mazzocca, S. Antonaci, G. Giannelli, Targeting transforming growth factor (TGF)-betaRI inhibits activation of beta1 integrin and blocks vascular invasion in hepatocellular carcinoma. Hepatology 49, 839–850 (2009)CrossRefPubMed E. Fransvea, A. Mazzocca, S. Antonaci, G. Giannelli, Targeting transforming growth factor (TGF)-betaRI inhibits activation of beta1 integrin and blocks vascular invasion in hepatocellular carcinoma. Hepatology 49, 839–850 (2009)CrossRefPubMed
39.
Zurück zum Zitat M. Serova, D. Garbay, M.E. Riveiro, I. Bieche, M. Bouattour, E. Raymond and S. Faivre. Targeting of TGF-beta signaling results in decreased motility and invasion in parental and mulitkinase inhibitor-insensitive hepatocacinoma cells. ILCA Annual Conference, Brussels, International Liver Cancer Association (ILCA) (2011) M. Serova, D. Garbay, M.E. Riveiro, I. Bieche, M. Bouattour, E. Raymond and S. Faivre. Targeting of TGF-beta signaling results in decreased motility and invasion in parental and mulitkinase inhibitor-insensitive hepatocacinoma cells. ILCA Annual Conference, Brussels, International Liver Cancer Association (ILCA) (2011)
40.
Zurück zum Zitat Z. Zhong, K.D. Carroll, D. Policarpio, C. Osborn, M. Gregory, R. Bassi, X. Jimenez, M. Prewett, G. Liebisch, K. Persaud, D. Burtrum, S. Wang, D. Surguladze, S. Ng, H. Griffith, P. Balderes, J. Doody, J.D. Schwartz, H. Youssoufian, E.K. Rowinsky, D.L. Ludwig, L. Witte, Z. Zhu, Y. Wu, Anti-transforming growth factor beta receptor II antibody has therapeutic efficacy against primary tumor growth and metastasis through multieffects on cancer, stroma, and immune cells Clin. Cancer Res. 16, 1191–1205 (2010) Z. Zhong, K.D. Carroll, D. Policarpio, C. Osborn, M. Gregory, R. Bassi, X. Jimenez, M. Prewett, G. Liebisch, K. Persaud, D. Burtrum, S. Wang, D. Surguladze, S. Ng, H. Griffith, P. Balderes, J. Doody, J.D. Schwartz, H. Youssoufian, E.K. Rowinsky, D.L. Ludwig, L. Witte, Z. Zhu, Y. Wu, Anti-transforming growth factor beta receptor II antibody has therapeutic efficacy against primary tumor growth and metastasis through multieffects on cancer, stroma, and immune cells Clin. Cancer Res. 16, 1191–1205 (2010)
41.
Zurück zum Zitat M.D. Hale, J.D. Hayden, H.I. Grabsch, Tumour-microenvironment interactions: role of tumour stroma and proteins produced by cancer-associated fibroblasts in chemotherapy response. Cell. Oncol. 36, 95–112 (2013) M.D. Hale, J.D. Hayden, H.I. Grabsch, Tumour-microenvironment interactions: role of tumour stroma and proteins produced by cancer-associated fibroblasts in chemotherapy response. Cell. Oncol. 36, 95–112 (2013)
42.
Zurück zum Zitat A. Geurts van Kessel, The cancer genome: from structure to function. Cell. Oncol. 37, 55–165 (2014) A. Geurts van Kessel, The cancer genome: from structure to function. Cell. Oncol. 37, 55–165 (2014)
Metadaten
Titel
Anti-tumor activity of the TGF-β receptor kinase inhibitor galunisertib (LY2157299 monohydrate) in patient-derived tumor xenografts
verfasst von
Armin Maier
Anne-Lise Peille
Vincent Vuaroqueaux
Michael Lahn
Publikationsdatum
01.04.2015
Verlag
Springer Netherlands
Erschienen in
Cellular Oncology / Ausgabe 2/2015
Print ISSN: 2211-3428
Elektronische ISSN: 2211-3436
DOI
https://doi.org/10.1007/s13402-014-0210-8

Weitere Artikel der Ausgabe 2/2015

Cellular Oncology 2/2015 Zur Ausgabe

Neu im Fachgebiet Pathologie

Molekularpathologische Untersuchungen im Wandel der Zeit

Open Access Biomarker Leitthema

Um auch an kleinen Gewebeproben zuverlässige und reproduzierbare Ergebnisse zu gewährleisten ist eine strenge Qualitätskontrolle in jedem Schritt des Arbeitsablaufs erforderlich. Eine nicht ordnungsgemäße Prüfung oder Behandlung des …

Vergleichende Pathologie in der onkologischen Forschung

Pathologie Leitthema

Die vergleichende experimentelle Pathologie („comparative experimental pathology“) ist ein Fachbereich an der Schnittstelle von Human- und Veterinärmedizin. Sie widmet sich der vergleichenden Erforschung von Gemeinsamkeiten und Unterschieden von …

Gastrointestinale Stromatumoren

Open Access GIST CME-Artikel

Gastrointestinale Stromatumoren (GIST) stellen seit über 20 Jahren ein Paradigma für die zielgerichtete Therapie mit Tyrosinkinaseinhibitoren dar. Eine elementare Voraussetzung für eine mögliche neoadjuvante oder adjuvante Behandlung bei …

Personalisierte Medizin in der Onkologie

Aufgrund des erheblichen technologischen Fortschritts in der molekularen und genetischen Diagnostik sowie zunehmender Erkenntnisse über die molekulare Pathogenese von Krankheiten hat in den letzten zwei Jahrzehnten ein grundlegender …