ArticlePDF Available

Ex-Vivo Treatment of Tumor Tissue Slices as a Predictive Preclinical Method to Evaluate Targeted Therapies for Patients with Renal Carcinoma

Authors:

Abstract and Figures

Clear cell renal cell carcinoma (ccRCC) is the third type of urologic cancer. At time of diagnosis, 30% of cases are metastatic with no effect of chemotherapy or radiotherapy. Current targeted therapies lead to a high rate of relapse and resistance after a short-term response. Thus, a major hurdle in the development and use of new treatments for ccRCC is the lack of good pre-clinical models that can accurately predict the efficacy of new drugs and allow the stratification of patients into the correct treatment regime. Here, we describe different 3D cultures models of ccRCC, emphasizing the feasibility and the advantage of ex-vivo treatment of fresh, surgically resected human tumor slice cultures of ccRCC as a robust preclinical model for identifying patient response to specific therapeutics. Moreover, this model based on precision-cut tissue slices enables histopathology measurements as tumor architecture is retained, including the spatial relationship between the tumor and tumor-infiltrating lymphocytes and the stromal components. Our data suggest that acute treatment of tumor tissue slices could represent a benchmark of further exploration as a companion diagnostic tool in ccRCC treatment and a model to develop new therapeutic drugs.
Treatment of slice cultures from 786-O tumor xenografts. 786-O cells were injected under the renal capsula of Balb/c nude mice. One month later, mice were euthanized, tumors were harvested and processed for tissue slice cultures. (A) Tissue slice cultures were treated with 10 µM of either GDC-0941 + saracatinib (10 µM each, GDC/SRC), pazopanib (PAZO), sunitinib (SUN), temsirolimus (TEM), or vehicle (DMSO 0.2%) for 48 h. Nuclei were stained with Hoechst 33342 and dead cells were visualized by Ethidium homodimer staining. Images were taken with an Apotome-equipped Zeiss microscope. Bar scale 50 µm. (B) The intensity of Ethidium homodimer positive cells was measured in each nucleus on five independent areas of the tumor slices as described in Material and Methods. The y-axis represents the ratio of the percentage of dead cells in the different groups divided by the corresponding value in the DMSO-treated-slices. Significant differences in cell death were observed between DMSO versus the GDC/SRC combination (*** p ≤ 0.001) or each drug alone (** p ≤ 0.05) using a Mann-Whitney test. (C) Tumor slices were treated as described in A, then fixed and embedded in paraffin. Fixed tissue slices were stained with Hematoxylin-Eosin (HE). Representative pictures of treated slices are shown at two magnifications (lower magnifications, upper images and higher magnifications, middle images). Tumor slices were also stained with the anti-PCNA antibody to visualize cell proliferation (lower panel). Negative controls (no primary antibody) are shown in the insets. Scale bars 20 µm.
… 
Content may be subject to copyright.
cancers
Article
Ex-Vivo Treatment of Tumor Tissue Slices as a
Predictive Preclinical Method to Evaluate Targeted
Therapies for Patients with Renal Carcinoma
Caroline Roelants 1,2, Catherine Pillet 3, Quentin Franquet 1,4, Clément Sarrazin 1,4,
Nicolas Peilleron 1,4, Sofia Giacosa 1, Laurent Guyon 1, Amina Fontanell 4, Gaëlle Fiard 4,
Jean-Alexandre Long 4, Jean-Luc Descotes 4, Claude Cochet 1and Odile Filhol 1, *
1UniversitéGrenoble Alpes, Inserm, CEA, IRIG-Biology of Cancer and Infection, UMR_S 1036,
F-38000 Grenoble, France; caroline.roelants@inovarion.com (C.R.); qfranquet@chu-grenoble.fr (Q.F.);
csarrazin1@chu-grenoble.fr (C.S.); nicolas.peilleron@gmail.com (N.P.); sofiagiacosa@gmail.com (S.G.);
laurent.guyon@cea.fr (L.G.); claude.cochet@cea.fr (C.C.)
2Inovarion, 75005 Paris, France
3UniversitéGrenoble Alpes, Inserm, CEA, IRIG-Biologie àGrande Echelle, UMR 1038,
F-38000 Grenoble, France; catherine.pillet@cea.fr
4Centre hospitalier universitaire Grenoble Alpes, CS 10217, 38043 Grenoble CEDEX 9, France;
lafontanell@chu-grenoble.fr (A.F.); g.fiard@ucl.ac.uk (G.F.); JALong@chu-grenoble.fr (J.-A.L.);
jldescotes@chu-grenoble.fr (J.-L.D.)
*Correspondence: odile.filhol-cochet@cea.fr; Tel.: +33-(0)4-38785645; Fax: +33-(0)4-38785058
Received: 30 November 2019; Accepted: 15 January 2020; Published: 17 January 2020


Abstract:
Clear cell renal cell carcinoma (ccRCC) is the third type of urologic cancer. At time of
diagnosis, 30% of cases are metastatic with no eect of chemotherapy or radiotherapy. Current targeted
therapies lead to a high rate of relapse and resistance after a short-term response. Thus, a major hurdle
in the development and use of new treatments for ccRCC is the lack of good pre-clinical models
that can accurately predict the ecacy of new drugs and allow the stratification of patients into the
correct treatment regime. Here, we describe dierent 3D cultures models of ccRCC, emphasizing
the feasibility and the advantage of ex-vivo treatment of fresh, surgically resected human tumor
slice cultures of ccRCC as a robust preclinical model for identifying patient response to specific
therapeutics. Moreover, this model based on precision-cut tissue slices enables histopathology
measurements as tumor architecture is retained, including the spatial relationship between the
tumor and tumor-infiltrating lymphocytes and the stromal components. Our data suggest that acute
treatment of tumor tissue slices could represent a benchmark of further exploration as a companion
diagnostic tool in ccRCC treatment and a model to develop new therapeutic drugs.
Keywords:
drug sensitivity; immune infiltration; renal cancer; targeted therapy; tumor slice culture
1. Introduction
Clear cell renal cell carcinoma (ccRCC) is the most frequent subtype of kidney cancer representing
above 3% of all cancers. At the time of diagnosis, 30% of cases are metastatic and are associated
with a poor prognosis and without long-lasting eects of traditional oncologic treatment such
as chemotherapy or radiotherapy [
1
]. With the advance of targeted therapies for RCC, several
agents targeting angiogenesis and signal transduction pathways such as sunitinib, temsirolimus,
and pazopanib have appeared and showed improved clinical benefit and survival in randomized
prospective clinical trials. Yet, improvements are still required, as many of these current therapies are
limited by acquired resistance mostly through activation of alternative pathways [
2
]. The tumor immune
Cancers 2020,12, 232; doi:10.3390/cancers12010232 www.mdpi.com/journal/cancers
Cancers 2020,12, 232 2 of 18
microenvironment of ccRCC is known to be highly immunosuppressive and immune infiltration of
tumors is closely associated with clinical outcome. Recently, immune checkpoint inhibitors have
demonstrated significant anti-tumor activity in the first-line treatment of intermediate to poor risk
RCC patients, but these therapies are only eective for a small fraction of patients, and are associated
with problems, such as side eects and high costs [
3
6
]. Thus, new treatment strategies are needed to
improve ecacy in a broader patient population. In the last decade, eorts have primarily focused
on establishing a framework for predictions of anticancer drug responses using
in vitro
tumor cell
line models [
7
14
]. These techniques are limited by the cell dissociation that selects the more robust
cells and the ones that can attach to the cell culture substratum [
15
17
]. Moreover, in these conditions,
inadequate representation of the tumor heterogeneity and microenvironment interactions during a
preclinical screen can result in inaccurate predictions of drug candidate eects.
Organoids derived from patient tumors have recently gained much interest as promising tools
for several translational applications, such as high-throughput drug screens and personalized
medicine [1820]
. Tumor organoids grown with undefined natural (e.g., Matrigel
®
) or synthetic
extracellular matrix gels show improved resemblance to the original tumor compared to 2D cultured
cancer cell lines. However, they do not model tumor–stromal interactions (cancer cells, immune,
and endothelial cells) and the growth selection pressures applied during their generation have the
potential to introduce bias [
13
,
21
]. Consequently, the prediction of treatment outcome extrapolated
from organoids may not recapitulate each cancer patient tumor. Moreover, it is not clear whether the
timescales are quick enough to aect patient care [22].
Another approach often considered more representative is the use of patient-derived xenograft
(PDX) systems. However, the generation of PDX models exhibits a low engraftment rate, and the
timescales and costs involved in this process are very significant [
23
]. Furthermore, the PDX deviates
from the original tumor over time [
24
], and dierence in pathophysiology between animal models and
humans contributes to high failure rates of current small-molecule inhibitors in preclinical trials [
25
27
].
Thus, predicting successful anticancer therapy remains extremely challenging, largely due to extensive
inter- and intratumor heterogeneity [
28
] and there remains a need for alternative, innovative models
that allow the precise balance between manipulability and biological complexity.
To address these challenges, ex-vivo culture of intact tumor slices is potentially an extremely
attractive system that has been already validated in various types of cancers [2938].
This method has several advantages: (1) it can be rapidly established using only small samples
of fresh tissue with a limited cost, (2) it preserves the tumor architecture and the spatial interaction
between tumor and stroma, (3) testing of drug susceptibility can be combined with gene sequencing
and immunohistochemistry analysis. To the best of our knowledge, tumor slice culture has never been
validated in renal carcinoma.
In this study, we developed dierent biological cell-based systems like 3D tumor spheroids,
mice orthotopic tumor xenografts, and patient-derived tumor slice cultures (PDTSC) for ex-vivo
assessment of drug eects in renal carcinoma. As we recently showed, a combination of two
inhibitors targeting both the PI3K and Src kinases impedes cell viability of renal carcinoma cells [
39
],
we compared the ecacy of this combination to standard-of-care-drugs for RCC like sunitinib,
pazopanib, and temsirolimus using 3D tumor spheroids and PDTSC methods. We show that PDTSC
has the potential to be exploited for cancer cell sensitivity assessment to novel molecularly targeted
therapies among patients with ccRCC, and to identify suitable candidates for drug combinations in a
cost-eective and patient-friendly manner. We also demonstrate that PDTSC faithfully preserves the
molecular landscape of the original renal carcinoma, retaining histopathology, including the stromal
components and the immune cells that innately infiltrate the patient’s malignant epithelial cells,
features that can be potentially useful to evaluate predictive biomarkers of treatment response and for
patient stratification in prospective trials with immune checkpoint inhibitors.
Cancers 2020,12, 232 3 of 18
2. Results
2.1. Evaluation of Drug Sensitivity on 786-0 Cell-Derived Spheroids
We first compared the induction of cell mortality in 786-O spheroids after their treatment with
either a combination of GDC-0941 and saracatinib (GDC/SRC), two small-molecule inhibitors that target
the PI3K and Src kinases respectively [
39
], or the currently clinically used inhibitorssunitinib, pazopanib,
or temsirolimus at the indicated concentrations. Treated spheroids were recorded for 48 h using an
Essen IncuCyte Zoom live-cell microscopy instrument (Figure 1A). Cell death induced by the different
treatments at 6, 12, 24, and 48 h, was quantified through propidium iodide (PI) incorporation normalized
by the surface of the spheroid. The results show that the drug effects on 786-O spheroids could be easily
quantified using the Incucyte microscopy instrument (Figure 1B). Moreover, monitoring the size of the
spheroids after 36 h of treatment showed that the GDC/SRC combination induces a significant reduction
of the spheroid size (35%) while the effects of the other drugs were weaker compared to DMSO for
which spheroid area declined by 15%, probably due to the maturation of the organoids that were under
culture condition for five days (Figure 1C). During the last 12 h of treatment, cells in the spheroid
center that was hypoxic, might have begun to die. Next, immunohistochemistry was performed on
paraffin-embedded spheroids to visualize both the cellular architecture and the cell proliferation inside
the 3D-spheroids. As shown in Figure 1D–F, both the integrity of the spheroids and the cell proliferation
detected by PCNA labeling were affected by the GDC/SRC combination or Temsirolimus treatments
confirming their effects on spheroids viability. Moreover, spheroid area measurements (Figure 1E) were
consistent with the analysis of PI incorporation determined with the Incucyte microscope (Figure 1A).
However, although promising, these data obtained with a 3D cancer cell line model suffered from
inherent limitations due to inadequate representation of the heterogeneous architecture of human tumor
and tumor–stromal interactions, which renders the interpretation on efficacy testing challenging. This is
attested by the observation that among all the new molecules discovered for their action on cancer cell
line models, only very few reached the FDA agreement. Therefore, implementation of physiologically
relevant in- vitro models closer to patient-derived tumors is required.
Figure 1. Cont.
Cancers 2020,12, 232 4 of 18
Figure 1.
Treatment of 786-O spheroids. 786-O-WT (VHL
-
) cells were grown as spheroids and treated
with 10
µ
M of either GDC-0941 +saracatinib (10
µ
M each, GDC/SRC,
N
), pazopanib (PAZO,
#
), sunitinib
(SUN,
), temsirolimus (TEM,
), or vehicle (DMSO,
) in the presence of propidium iodide. Cell death
was monitored on spheroids using either an Essen IncuCyte Zoom live-cell microscopy incubator or by
immunohistochemistry. (
A
) Bright field and fluorescent overlaid images show 786-O-treated spheroids
at indicated times (0, 6, 12, 24, and 48 h). Bar scale 300
µ
m. (
B
) Images taken automatically every
6 h over 48 h of culture were analyzed for PI fluorescent area quantification. Cell death values (PI
labeling area) was divided by the corresponding spheroid area and multiplied by 100. This percentage
of cell death was divided by the one at T
0
, for all the others time points and was expressed as mean
±
SEM. The statistical analysis of dead cells was performed with 2 way ANOVA test for each time point
compared to DMSO treatment. (
C
) The same images were analyzed for spheroid area quantification.
Significant dierence was observed between GDC/SRC (**** p
0.001), SUN (**** p
0.01), TEM (****
p
0.01) versus DMSO after 36 h of treatment using a Kruskal-Wallis test. (
D
) PCNA staining to
visualize proliferation of fixed paran-embedded (scale bar, 20
µ
m). (
E
) Spheroid area quantification
by surface calculation of (
D
), (n
8). Significant dierence was observed between GDC/SRC (*** p
0.001), TEM (** p
0.01) versus DMSO in a Kruskal-Wallis test. (
F
) The number of PCNA positive cells
was quantified in each spheroid and divided by the corresponding spheroid surface. Histogram plot
represents mean of PCNA-stained cells pooled from 4 to 6 spheroids (biological replicated/condition)
with error bar (±SEM).
2.2. Tissue Slice Cultures of Renal Tumors
We set out to determine whether an ex-vivo treatment protocol could be used as a means of
determining ccRCC sensitivity to various cytotoxic agents. The PDTSC methodology has been
previously used to evaluate the drug sensitivity of normal and tumor tissues [
29
38
]. Therefore, we set
up an adaptation of this method outlined in Figure 2A, as an ex-vivo protocol to examine responses of
ccRCC to dierent therapeutic agents. Cultures of slices, obtained either from 786-O-derived tumors
generated in mouse xenografts or from human ccRCC surgical resection specimens, were prepared as
detailed in the Methods section, and then subjected to a variety of tests. First, we noticed that over
96 h of culture, luminescence measurement of 786-O-luc cells in the tumor slice remained constant,
attesting their viability during this time schedule (Figure 2B).
Cancers 2020,12, 232 5 of 18
Figure 2.
The procedure flowchart for renal tumor slice culture. (
A
) 786-O-derived tumors generated
in mouse xenografts or human ccRCC surgical resection specimens are cut into 300
µ
m slices in buer
solution using a Vibratome
®
. The slices are transferred to culture medium and then carefully placed
on membrane insert in 6-well plates to create an air-liquid interface. After 48 h of drug treatments,
slices are analyzed for cell viability and biomarker immuno-detection. Correlation between drug
sensitivity and biomarker expression is visualized with the graphical display of a correlation matrix
(Corrplot, R package). (
B
) Tumor slices maintain cell survival over four days of culture. Slices from
786-O-luc-derived tumors were cultured for up to four days, with fresh media changes performed
every two days. Each day, luminescence was recorded from slices after luciferin addition using IVIS
imaging (upper panel). Plotted normalized photon quantification showed minimal changes over the
culture periods.
2.3. The Cytotoxic Eects of Drug Treatments Can Be Evaluated in Tissue-Slice Cultures
In order to evaluate the PDTSC approach, we first used the renal carcinoma mouse xenograft
model. The tumors were extracted from the mice, directly processed into 300
µ
m slices, and treated for
48 h as described in Materials and Methods and indicated in Figure 2A. Cell viability evaluated by
ethidium homodimer staining of treated tumor-slice cultures are illustrated in Figure 3A. Mortality
quantified on five to seven images using ImageJ, was reported as “Cell death/DMSO” that represents
the percentage of dead cells in the dierent groups divided by the percentage of dead cells in the
DMSO-treated slices. The mortality rate showed a significant dierence between DMSO and drugs
alone or the GDC/SRC combination (p<0.05) (Figure 3A,B). Immuno-histochemistry (IHC) analysis
was used to determine whether a dierential proliferative (PCNA) response to drug treatment could be
detected. For this, paran-embedded sections were stained with a PCNA antibody and counter-colored
with hematoxylin. We found that the GDC/SRC combination caused a significant decrease in PCNA
staining, while temsirolimus, sunitinib, and pazopanib were less ecient (Figure 3C). Taken together,
these results demonstrate that PDTSC allows for the rapid investigation of ccCRCC sensitivity to
targeted therapies.
Cancers 2020,12, 232 6 of 18
Figure 3.
Treatment of slice cultures from 786-O tumor xenografts. 786-O cells were injected under the
renal capsula of Balb/c nude mice. One month later, mice were euthanized, tumors were harvested
and processed for tissue slice cultures. (
A
) Tissue slice cultures were treated with 10
µ
M of either
GDC-0941 +saracatinib (10
µ
M each, GDC/SRC), pazopanib (PAZO), sunitinib (SUN), temsirolimus
(TEM), or vehicle (DMSO 0.2%) for 48 h. Nuclei were stained with Hoechst 33342 and dead cells were
visualized by Ethidium homodimer staining. Images were taken with an Apotome-equipped Zeiss
microscope. Bar scale 50
µ
m. (
B
) The intensity of Ethidium homodimer positive cells was measured
in each nucleus on five independent areas of the tumor slices as described in Material and Methods.
The y-axis represents the ratio of the percentage of dead cells in the dierent groups divided by the
corresponding value in the DMSO-treated-slices. Significant dierences in cell death were observed
between DMSO versus the GDC/SRC combination (*** p
0.001) or each drug alone (** p
0.05) using
a Mann–Whitney test. (
C
) Tumor slices were treated as described in A, then fixed and embedded
in paran. Fixed tissue slices were stained with Hematoxylin-Eosin (HE). Representative pictures
of treated slices are shown at two magnifications (lower magnifications, upper images and higher
magnifications, middle images). Tumor slices were also stained with the anti-PCNA antibody to
visualize cell proliferation (lower panel). Negative controls (no primary antibody) are shown in the
insets. Scale bars 20 µm.
Cancers 2020,12, 232 7 of 18
To further evaluate the potential of this approach, slices from surgical resections of human ccRCC
tumors were analyzed using the same optimized protocol. In this study, we focused on patient tumors
that were later on characterized as renal clear cell carcinoma by a board certified histo-pathologist at the
Urology Department—University Hospital Center of Grenoble-Alpes. We note that our protocol did
not interfere with the pathologist’s analysis. Warm ischemia was reduced to 15 min including tumor
dissection and extraction during the surgery. Cold ischemia between extraction and the beginning of
the culture was less than 2 h (including tumor sample dissection, transport and slicing). Two small
pieces from two distinct regions (A and B) of each tumor were taken and processed in slices using a
Vibratome
®
. Then, each tumor slice was cultured in the presence of the vehicle (0.2% DMSO) or the
indicated therapeutic agents and assayed for cell viability after 48 h of drug treatment. Figure 4A shows
that sample A disclosed high sensitivity to pazopanib or the GDC/SRC combination whereas sunitinib
and temsirolimus were almost without eect. In contrast, sunitinib significantly compromised cell
viability in sample B. Samples B-treated slices were further analyzed for their proliferation status after
fixation and paran inclusion to assess functional response and cell viability. PCNA staining was
detected in DMSO-treated slices (11.9%), whereas very few cells were stained in sunitinib-treated slices
(0.2%). Moreover, a strong staining of cleaved-caspase-3 that reflects apoptotic cell death was observed
in sunitinib-treated tumor slices (42.1%) but almost undetectable in DMSO-treated samples (4.2%)
(Figure 4B).
Figure 4. Cont.
Cancers 2020,12, 232 8 of 18
Figure 4.
Treatment of slice cultures from human renal tumors. Tissue slice cultures from human
renal tumors were treated for 48 h with a panel of drugs (10
µ
M each) and cell viability assayed as
in Figure 3A. (
A
) Intra-tumor heterogeneity. Two fragments A and B of the same tumor (NM014)
were analyzed for their sensitivity to indicated drug treatments. Mean DMSO was normalized to 1 to
compare the two fragments of NM014. The y-axis represents the ratio of the percentage of dead cells
in the dierent groups divided by the corresponding value in the DMSO-treated-PDTSC. Cell death
measurement in fragment A (black bars) from NM014 shows significant dierences between DMSO
versus the combination (GDC/SRC, * p<0.5), pazopanib (PAZO, ** p<0.01) and temsirolimus (TEM, *
p<0.5) but not versus sunitinib (SUN). The same analysis of fragment B (white bars) from the same
NM014 tumor, shows similar profile except for sunitinib that in this case induced significant cell death
(SUN, **** p<0.0005). (
B
) Apoptosis and proliferation assays. Representative pictures of tumor slices
from fragment B of NM014 treated for 48 h with DMSO (
upper panels
) or 10
µ
M sunitinib (SUN,
lower
panels
) and stained with Cleaved-Caspase-3 (
left panel
) or with anti-PCNA antibody (
right panel
).
The PCNA stain identifies cells that are proliferating while the Cleaved-Caspase-3 stain shows cells
undergoing apoptosis. The percentages of PCNA and Cleaved-Caspase 3 positive cells were plotted
below each set of pictures. Scale bars, 20
µ
m. Negative controls (no primary antibody) are shown in
insets. (
C
) Inter-tumor heterogeneity. Four dierent tumors were treated and analyzed as in Figure 3A
showing distinct drug sensitivity profiles. Each color represents one patient tumor (Yellow, NB029;
Blue, YL024; Green, NM014; Purple, MD034). (
D
) VHL and HIF expressions. Representative pictures
of two untreated tumor slices GD022 and NM014 stained with anti-VHL, anti-HIF1
α
or anti-HIF2
α
antibodies. Scale bars, 50
µ
m. For each staining, images taken from five independent areas of a tumor
slice were quantified with ImageJ and plotted as percentage of specific staining relative to tumor area
(respective right panels).
These results highlight the intra-tumor heterogeneity of ccRCC, a property that has been well
documented by extensive multi-regional whole-genome and -exome sequencing [
40
]. Collectively,
these data demonstrated that PDTSC can be used to assess functional response and cell survival of
human renal carcinoma specimens to drug treatments, reinforcing its value as a companion diagnostic
tool in ccRCC treatment.
Cancers 2020,12, 232 9 of 18
2.4. Acute Ex-Vivo Drug Treatments Identify Renal Tumor Subsets with Distinct Therapeutic Profiles
We compared the cell death rate of four dierent patient tumors upon the same panel of drug
treatments (Figure 4C). Interestingly, this approach allowed for the identification of dierential patient
responses revealing sensitive and resistant tumors. For example, pazopanib was completely inactive
on NB029, YL024 and MD034, whereas it was the most ecient on NM014. Temsirolimus was without
eect on YL024.
Inactivation of the Von Hippel–Lindau (VHL) tumor suppressor gene has been shown to play an
important role in the process of angiogenesis in RCC. As a component of an E3 ubiquitin ligase complex,
the VHL protein targets the hypoxia-inducible transcription factors (HIF1
α
and HIF2
α
) for degradation.
Loss of VHL function in ccRCC leads to the constitutive stabilization of these transcription factors,
leading to a highly angiogenic environment [
41
] and HIF2
α
has recently emerged as a therapeutic
target in ccRCC [
42
]. In line with this, we determined the protein expression level of VHL, HIF1
α
and
HIF2
α
in human renal tumor slices (Figure 4D, left panels). Immuno-staining quantification shows
that in slice NM014 where VHL expression was undetectable, HIF1
α
and mainly HIF2
α
were more
abundant than in slice GD022 where VHL was present (Figure 4D, right panels).
2.5. Predictive Biomarkers in Renal Tumor Slice Cultures
The tracking of immune cells in human cancers aects their immunobiology but also could
have a major prognostic and predictive impact on the ecacy of the patient treatment. Indeed,
renal cell carcinoma is an immunogenic tumor that characteristically harbors abundant infiltrating
lymphocytes [
43
] and it has been shown that across renal tumors, there is a wide range of immune
infiltrates [
44
]. Therefore, we tracked immune cells and their interaction with cancer cells within
fixed slice cultures of dierent patient tumor samples (Figure 5). As an example, Figure 5A shows
representative images of two non-treated tumor slices ML025 and DP027 in which the microvessel
density labeled by CD34 staining was similar (Figure 5, right panel). Tumor slice DP027 was infiltrated
with fewer cytotoxic CD8
+
T cells than the tumor slice ML025. Interestingly, it has been suggested that
in highly infiltrated ccRCC tumors, T-cell activation state is a key determinant of ccRCC prognosis and
likely of immunotherapy response. Given the variety of mechanisms triggered by molecularly targeted
agents in cancers and their late-stage clinical trials, the validation of drug sensitivity predictive models
may be critical to identify the right drug for the right patients and help to understand determinants of
responsiveness, wherein alternative treatments could potentially overcome resistance [
45
]. There is a
recently growing body of literature describing PDTSC from dierent normal and tumor tissues [
34
38
].
However, to our knowledge, the present study is the first to demonstrate the potential use of this
approach to evaluate renal cancer response to novel therapies while modeling the tumor immune
microenvironment. An important benefit of the PDTSC strategy is that it provides a rapid and easy
readout of the functional eects and drug responses that result from a complex array of molecular
alterations among patients with ccRCC. PDTSC delivers a much faster timeline than PDX animal
models, which require at least 6 to 7 weeks to become established versus 48 h for the PDTSC method.
There is an important limitation inherent to PDTSC: the frequent intra-tumor heterogeneity may not be
represented in individual slices from specific regions of a surgical resection specimen. However, this can
be taken into account by a careful geographical collection of replicated tumor slices. In agreement with
the key role played by the immune infiltrate in ccRCC, a phase 3 clinical trial (CheckMate214) showed
benefits in terms of overall survival and objective response rate using an immunotherapy combination
(ipilinumab plus nivolumab) versus sunitinib for intermediate and poor-risk patients with previously
untreated advanced renal cell carcinoma [46].
Cancers 2020,12, 232 10 of 18
Figure 5.
Predictive biomarkers in renal tumor slice cultures. (
A
) Vascular, immune and stem cell
type characterization. Representative pictures of untreated tumor slices ML025 and DP027 stained
with the following antibodies: anti-CD34, anti-CD8, anti-CD45, anti-PDL1, anti-LIM1. Scale bars,
50
µ
m. For each staining, images taken from five independent areas of a tumor slice were quantified
with ImageJ and plotted (
right panels
). (
B
) Correlation plot between the percentage of positive cells
following various IHC staining and the normalized proportion of dying cells following application of
drug treatments. The Spearman rank correlation was used. The diagonal indicates the biomarker used
for the IHC staining (
left part
) or the drug treatment (
right part
). Below the diagonal is the pairwise
correlation value, and above the diagonal is the corresponding representation, with the color legend
that is the bar on the right side of the plot. Blue (resp. red) colors correspond to positive (resp. negative)
correlations. Boxes correspond to cases described in the text. For example, the two blue boxes on
the top-left side highlight a correlation of 0.5 of the percentage of positive cells between HIF2 and
PDL1 staining.
Cancers 2020,12, 232 11 of 18
In both tumor slices, we also detected a dierential intra-tumor positive staining for the protein
tyrosine phosphatase receptor CD45, one of the key players in the initiation of T cell receptor
signaling [
47
]. CD45
+
cells were abundant in DP027 tumor and localized in close proximity with
microvessels and red blood cells. These cells were more intricately distributed in the ML025 slice than
in DP027 reflecting a potential immune infiltration.
The tumor microenvironment deploys various immune escape mechanisms that neutralize CD8
T cell-mediated tumor rejection. One mechanism implies the aberrant expression of programmed
death-ligand 1 (PD-L1) that targets the neutralization of activated CD8 T cells. PD-L1 has been reported
in several human cancers including RCC [
48
]. This ligand is aberrantly expressed on the surface
of both primary and metastatic RCC tumor cells [
49
] and several studies have described a positive
correlation between PD-L1 expression, metastasis, and poor outcomes in ccRCC [
50
]. Consistent with
this, we found that PD-L1 was strongly expressed in the ML025 tumor slice while undetectable in the
DP027 tumor slice. Of note, ML025 slices were both positive for PD-L1 and cytotoxic CD8
+
T cells.
Interestingly, it has been suggested that metastatic melanoma that are both expressing PD-L1 and CD8
+
T cells will likely respond to immunotherapy [51].
The LIM1 transcription factor which is essential for the development of human kidney is
reactivated in nephroblastomas [
52
] and implicated in the metastatic spread of ccRCC [
53
]. While being
undetectable in the DP027 slice, a strong intratumor LIM1 expression was observed in the ML025 slice.
Altogether, these data support the contention that the PDTSC method allows for precise, short-term
modeling of the stromal/immune microenvironment of renal tumors.
2.6. Prediction of Potential Correlations between Drug Sensitivity Responses and Tumor Immune Infiltration
To investigate whether there are links between drug-sensitivity and specific biomarkers previously
analyzed in Figure 5A, we performed correlation analysis (Figure 5B). Eighteen human renal tumors
(26 tumor specimens) that have been challenged for both drug-sensitivity and IHC- specific labeling
were compared. Pairwise correlated variables were plotted in a graph of correlation matrix according
to correlation coecients indicated either by colored circles or numbers. This analysis highlights
three subgroups/clusters of correlations between biomarkers only, drugs only and both drugs and
biomarkers. In the first cluster, we visualized a strong positive correlation between CD8 and CD45
expressions (correlation coecient, 0.54) and between HIF2
α
and PD-L1 (correlation coecient, 0.5).
These results are consistent with the literature as CD8 cytotoxic T cells are a subpopulation of CD45
positive leucocytes [
54
] and HIF2
α
as a transcription factor, binds to the PD-L1 promotor to induce its
expression [
55
]. Inside the “drug” cluster, correlation are high (correlation coecients >0.35 except
for the temsirolimus compared to the sunitinib situations). This result can be explained because the
four treatments tested have similar action mechanisms (all are kinase inhibitors), however, they are
not equal and more samples could help to find dierences. Finally, the most informative cluster that
compares biomarkers and drug treatments highlights two positive correlations between CD45 and
temsirolimus (TEM) (correlation coecient, 0.49) and PD-L1 and SUN (correlation coecient, 0.22)
and a negative correlation between GDC/SRC and HIF1
α
(correlation coecient,
0.28). TEM has
been demonstrated to have immune-modulating activity [
56
]. Obviously, the degree of correlation
should be established by increasing the amount of tumor tissue samples for IHC analysis but even
with this small cohort of tumor samples, potential valuable correlations dawned in this analysis and
warrants further investigations.
3. Discussion
Given the variety of mechanisms triggered by molecularly targeted agents in cancers and their
late-stage clinical trials, the validation of drug sensitivity predictive models may be critical to identify
the right drug for the right patients and help to understand determinants of responsiveness, wherein
alternative treatments could potentially overcome resistance [
45
]. There is a recently growing body
of literature describing PDTSC from dierent normal and tumor tissues [
29
38
]. However, to our
Cancers 2020,12, 232 12 of 18
knowledge, the present study is the first to demonstrate the potential use of this approach to evaluate
renal cancer response to novel therapies while modeling the tumor immune microenvironment.
An important benefit of the PDTSC strategy is that it provides a rapid and facile readout of the
functional eects and drug responses that result from a complex array of molecular alterations among
patients with ccRCC. PDTSC delivers a much faster timeline than PDX animal models, which require
at least 6 to 7 weeks to become established versus 48 h for the PDTSC method. There are several
limitations inherent to PDTSC: (1) the frequent intra-tumor heterogeneity may not be represented in
individual slices from specific regions of a surgical resection specimen. However, this can be taken into
account by a careful geographical collection of replicated tumor slices; (2) fresh primary tissue may
not be available when needed (in case of recurrent disease) and radical nephrectomy is not always
performed on metastasized patients, whereas screening of these patients would be highly beneficial
in the context of a predictive assay. The proof that the PDTSC can identify the best treatment need
further investigations. In particular, as 30% of ccRCC becomes metastatic, one third of the patients will
probably need specific treatments. In this context, the therapeutic profiling generated from the PDTSC
may be informative after a retrospective clinical follow-up from patients who will develop metastasis;
(3) PDTSC may be not relevant for some active drugs that are metabolites (e.g., for sunitinib).
4. Materials and Methods
4.1. Reagents, Drugs and Antibodies
Saracatinib (SRC) and GDC-0941(GDC) were obtained from LC Laboratories (Woburn, MA,
USA). temsirolimus (TEM), pazopanib (PAZO), and sunitinib (SUN) were purchased from Selleck
Chemicals (Houston, TX, USA), propidium iodide and Hoechst 33342 from Sigma-Aldrich (St Louis,
MO, USA), and Live & Dead kit from Life Technologies (Carlsbad, CA, USA). The antibodies against
the following targets were used: PCNA, CD8, CD34, PD-L1 (Ab29, Ab101500, Ab81289, Ab205921,
Abcam, Cambridge, UK), Cleaved-Caspase-3, CD45 (#9664, #13917, Cell Signaling, Danvers, MA, USA);
HIF1α, HIF2α(NB100-479, NB100-122, Novus Biologicals, Centennial, CO, USA), VHL (MA-1-12638,
Thermo Scientific, Waltham, MA, USA).
4.2. 3D-Spheroid Culture and Live Cell Tracking
786-O cells (ATCC-CRL-1932) are derived from a human primary clear cell adenocarcinoma.
This highly metastatic cell line is negative for VHL and is cultured in RPMI-1640 Medium supplemented
with 10% SVF and penicillin [100 U/mL], streptomycin [100 µg/mL].
Spheroids were prepared in 96-wells U-bottom with low evaporation lid (MicrotestTM,
Becton Dickinson Labware, San Jose, CA, USA) coated with 20 mg/mL poly-HEMA (Sigma-Aldrich). A
786-O cell suspension (1
×
10
3
cells) was seeded in each well and cells were allowed to form spheroids
within three days. Then, they were treated with indicated inhibitors for 48 h in the presence of
Propidium iodide (0.5
µ
g/mL) to visualize dead cells and video recorded every hour using an Incucyte
microscope, an automated live cell imager with high-throughput capabilities and built-in data analysis
(Essen Biosciences, Welwyn Garden City, UK). Experiments were conducted at 37
C and 5% CO
2
.
Quantification of cell death was measured after 48 h as a percentage of confluence in the red channel
(PI%) using the software incorporated into the IncuCyte Zoom. To normalize the data, all values for
each time point was divided by the value at T
0
. Experimental data are shown as mean
±
standard
error mean (SEM) except for Figure 1E for which whole the points are shown overlaid on boxplots and
whiskers. Classically, the box corresponds to the first and third quartiles, and the horizontal bar is the
median, whereas the whiskers demarcate here the extreme values.
4.3. Mice Orthotopic Tumor Xenograft Models
All animal studies were approved by the institutional guidelines and those formulated by the
European Community for the Use of Experimental Animals. Six week-old BALB/c female nude
Cancers 2020,12, 232 13 of 18
mice (Charles River Laboratories, Wilmington, MA, USA) with a mean body weight of 18–20 g were
used to establish orthotopic xenograft tumor models. The mice were housed and fed under specific
pathogen-free conditions. To produce tumors, renal cancer cells 786-O-luc (Roelants et al.) were
harvested from sub-confluent cultures by a brief exposure to 0.25% trypsin-EDTA. Trypsinization was
stopped with medium containing 10% FBS, and the cells were washed once in serum-free medium
and resuspended in 500
µ
L PBS. Renal orthotopic implantation was carried out by injection of 3
×
10
6
786-O luc cells into the left kidney of athymic nude mice. Mice were weighed once a week to monitor
their health and tumor growth was measured by imaging luminescence of 786-O-luc cells (IVIS).
4.4. Patients and Clinical Samples
All human renal carcinoma samples were obtained from patients, with their informed consent and
all procedures were approved by the ethic committee (Patient protection committee No 2017 A0070251).
All patients had serology to detect blood transmissible diseases before surgery and all samples were
anonymized. Fresh renal tumor tissues were obtained from patient undergoing a partial or a total
nephrectomy for cancer at the Urology Department—University Hospital Center of Grenoble-Alpes
(CHUGA). The minimal size of tumor samples for inclusion was 2 cm. After resection, tissue samples
were directly transported to the pathology department of the CHUGA in a cold saline solution (Sterile
0.9% NaCl). A macroscopic dissection was performed by a pathologist and as far as possible two
distinct tumor samples (A and B) were placed in a sterile conical tube containing a conservation
medium (ice-cold sterile balanced salt HBSS solution containing [100 U/mL] penicillin and [100
µ
g/mL]
streptomycin) on wet ice during transport from the pathology department to the INSERM research
laboratory (CEA).
4.5. Preparation of Tissue Slices and Organotypic Culture
Upon arrival, resections were manually minced using a sterile scalpel and samples were soaked
in ice-cold sterile HBSS, orientated, mounted in low-melting agarose (5%), and immobilized using
cyanoacrylate glue. Thick tissue slices (300
µ
m) were prepared from fresh tissue under sterile conditions
using a Vibratome VT1200 (Leica Microsystems, Wetzlar, Germany). Slicing speed was optimized
according to tissue density and type; in general, slower slicing speed was used on the softer tissues
and vice versa (0.2–0.7 mm/s). Vibration amplitude was set between 1.85 to 2.45 mm.
Tissue slices were then carefully placed on 0.4
µ
m pore size Teflon membrane culture inserts
(Millipore Corporation, Burlington, MA, USA) containing one slice per insert and cultured for up to 96
h. at 37
C in a 5% CO
2
humidified incubator using 2 mL of DMEM media supplemented with 20%
inactivated FBS (GIBCO), 100 U/mL penicillin (Invitrogen, Carlsbad, CA, USA). Inserts were placed in
a rotor agitator to allow gas and fluid exchanges with the medium. For each tumor, two slice samples
(A and B) were treated with the inhibitors at the indicated concentrations for 48 h.
4.6. Slice Viability Assay
At the end of treatment, lived slices were stained with the Live & Dead kit (Life Technologies)
as recommended and nuclei were labeled with Hoechst 33342. Images were taken with an
Apotome-equipped Zeiss Axio-Imager microscope with a 20
×
PlanApochromat objective (Numerical
Aperture 0.8). A minimum of three regions of interest (ROI) were taken, at three positions in z
with 7
µ
m intervals to avoid counting the same nuclei twice. Dead cells in the tissue slices were
quantified with scripts of ImageJ and further processed with R version 3.4.3 [
57
]. The histogram of red
fluorescence intensity shows a peak of low intensity corresponding to live cells, and high and widely
spread intensity values, corresponding to dying cells., called M
raw
. A minimum of 1200 total cells
was analyzed for each group. Experimental data are shown as mean
±
standard error mean (SEM).
As the percentage of dead cells varies significantly between dierent untreated tumor specimen due
to variation inherent to surgery, the percentage of dead cells in the dierent groups was divided by
Cancers 2020,12, 232 14 of 18
the corresponding value in the DMSO-treated-PDTSC. Thus, the y-axis untitled “Cell death/DMSO”
represents arbitrary units corresponding to this ratio.
For Figure 5B, mortality in a given condition is obtained by taking the median value among the
dierent z acquisition and ROI. Only conditions associated with a mortality below 55% were kept
for further analysis (four slices removed). As there is a correlation between cell mortality on a given
slide treated with the DMSO and the other drugs, we normalized cell mortality values using the
following formulae:
Mnorm =(Mraw MDMSO)/f(MDMSO )
where M
norm
is the normalized mortality for a given drug, typically between 0 and 1, M
raw
is the raw
mortality of the given drug, between 0 and 100, M
DMSO
is the raw mortality of the DMSO for the
nearby slice, between 0 and 100, f(M
DMSO)
is the normalization function, depending on the DMSO,
which evaluates the maximum amplitude of the drug eect on a given slice. It is given by:
f(MDMSO)=aMDMSO +(100 a)(1exp(MDMSO /b)),
where a=15 and b=10, chosen to fit with the maximal mortality.
The correlation analysis was performed using R version 3.4.3 [
57
], pairwise. The Spearman’s
rank correlation coecient was used to perform a robust analysis. The figure was generated using the
corrplot package version 0.84.
4.7. Immunohistochemistry Analysis
Sections (5
µ
m thick) of formalin-fixed, paran embedded tumor tissue samples were dewaxed,
rehydrated through graded ethanol and subjected to heat-mediated antigen retrieval in citrate buer
(Antigen Unmasking Solution, Vector Laboratories, Burlingame, CA, USA). Slides were incubated for
10 min in hydrogen peroxide H
2
O
2
to block endogenous peroxidases and then 30 min in saturation
solution (Histostain, Invitrogen) to block nonspecific antibody binding. This was followed by overnight
incubation with indicated primary antibodies at 4
C. After washing, sections were incubated with
a suitable biotinylated secondary antibody (Histostain, Invitrogen) for 10 min. Antigen-antibody
complexes were visualized by applying a streptavidin-biotin complex (Histostain, Invitrogen) for
10 min followed by NovaRED substrate (Vector Laboratories). Sections were counterstained with
hematoxylin to visualize nucleus. Control sections were incubated with pool secondary antibodies
without primary antibody.
4.8. Statistical and Correlation Analyses
Experimental data are shown as mean
±
standard error mean (SEM). Statistical analyses were
performed using one-way analysis of variance (ANOVA) with multiple comparisons test (GraphPad
Prism 6). A p-value of less than 0.05 was considered statistically significant.
5. Conclusions
Because the PDTSC strategy maintains the landscape of the original tumor sample, including the
stromal and the immune tumor compartment, this approach is a relevant model for individualized
testing of drug susceptibility to improve clinical success rates [
22
,
58
]. In agreement with the key role
played by the immune infiltrate in ccRCC, a phase 3 clinical trial (CheckMate214) showed benefits in
term of overall survival and objective response rate using an immunotherapy combination (ipilinumab
plus nivolumab) versus sunitinib for intermediate and poor-risk patients with previously untreated
advanced renal cell carcinoma [
46
]. Therefore, PDTSC also warrants further investigations to confirm
potential correlations between drug sensitivity responses and the level of tumor vascularization and
tumor-infiltrating immune cell populations. Finally, a recent study [
59
] suggests that further work will
eventually make this technique useful for personalized clinical immunotherapy.
Cancers 2020,12, 232 15 of 18
Author Contributions:
Conceptualization, C.C. and O.F.; investigation and methodology, C.R., C.P., S.G., Q.F.,
N.P., C.S., and O.F.; validation, C.P., Q.F., C.S., L.G., C.C., and O.F.; formal analysis, C.R., Q.F., L.G., C.S., and O.F.;
software, L.G. and C.S.; resources, C.R., C.P., S.G., Q.F., N.P., A.F., C.S., O.F., G.F., J.-A.L., and J.-L.D.; data curation,
C.R., L.G., Q.F., C.S., and O.F.; visualization, C.R., C.S., L.G., and O.F.; writing—review and editing, Q.F., C.R., C.S.,
L.G., O.F., and C.C.; supervision, O.F., C.C., and J.-L.D.; project administration and funding acquisition, C.C.; O.F.,
and J.-L.D. All authors have read and agreed to the published version of the manuscript.
Funding:
This work was supported by recurrent institutional funding from INSERM, CEA, Ligue Nationale
contre le Cancer (accredited team 2010–2012) and Ligue Comit
é
de l’Is
è
re, University Grenoble Alpes, Centre
Hospitalier Universitaire de Grenoble-Alpes (CHUGA), Groupement des Entreprises Françaises dans la LUtte
contre le Cancer (GEFLUC), Grenoble Alliance for Integrated Structural & Cell Biology (GRAL) and Association
Française d’Urologie (AFU).
Acknowledgments:
We thank the animal unit sta(Jeannin I., Bama S., Magallon C., Chaumontel N. and Pointu
H.) at Interdiciplinary Research Institute of Grenoble (IRIG) for animal husbandry.
Conflicts of Interest: The authors declare no financial or commercial conflict of interest.
References
1.
Negrier, S.; Escudier, B.; Lasset, C.; Douillard, J.Y.; Savary, J.; Chevreau, C.; Ravaud, A.; Mercatello, A.;
Peny, J.; Mousseau, M.; et al. Recombinant human interleukin-2, recombinant human interferon alfa-2a, or
both in metastatic renal-cell carcinoma. Groupe Francais d’Immunotherapie. N. Engl. J. Med.
1998
,338,
1272–1278. [CrossRef]
2.
Figlin, R.; Sternberg, C.; Wood, C.G. Novel agents and approaches for advanced renal cell carcinoma. J. Urol.
2012,188, 707–715. [CrossRef]
3.
Atkins, M.B.; Clark, J.I.; Quinn, D.I. Immune checkpoint inhibitors in advanced renal cell carcinoma:
Experience to date and future directions. Ann. Oncol. O. J. Eur. Soc. Med Oncol.
2017
,28, 1484–1494.
[CrossRef]
4.
Motzer, R.J.; Penkov, K.; Haanen, J.; Rini, B.; Albiges, L.; Campbell, M.T.; Venugopal, B.; Kollmannsberger, C.;
Negrier, S.; Uemura, M.; et al. Avelumab plus Axitinib versus Sunitinib for Advanced Renal-Cell Carcinoma.
N. Engl. J. Med. 2019,380, 1103–1115. [CrossRef] [PubMed]
5.
Rini, B.I.; Plimack, E.R.; Stus, V.; Gafanov, R.; Hawkins, R.; Nosov, D.; Pouliot, F.; Alekseev, B.; Soulieres, D.;
Melichar, B.; et al. Pembrolizumab plus Axitinib versus Sunitinib for Advanced Renal-Cell Carcinoma.
N. Engl. J. Med. 2019,380, 1116–1127. [CrossRef] [PubMed]
6.
Cella, D.; Grunwald, V.; Escudier, B.; Hammers, H.J.; George, S.; Nathan, P.; Grimm, M.O.; Rini, B.I.; Doan, J.;
Ivanescu, C.; et al. Patient-reported outcomes of patients with advanced renal cell carcinoma treated with
nivolumab plus ipilimumab versus sunitinib (CheckMate 214): A randomised, phase 3 trial. Lancet Oncol.
2019,20, 297–310. [CrossRef]
7.
Garnett, M.J.; Edelman, E.J.; Heidorn, S.J.; Greenman, C.D.; Dastur, A.; Lau, K.W.; Greninger, P.;
Thompson, I.R.; Luo, X.; Soares, J.; et al. Systematic identification of genomic markers of drug sensitivity in
cancer cells. Nature 2012,483, 570–575. [CrossRef] [PubMed]
8.
Barretina, J.; Caponigro, G.; Stransky, N.; Venkatesan, K.; Margolin, A.A.; Kim, S.; Wilson, C.J.; Lehar, J.;
Kryukov, G.V.; Sonkin, D.; et al. The Cancer Cell Line Encyclopedia enables predictive modelling of anticancer
drug sensitivity. Nature 2012,483, 603–607. [CrossRef] [PubMed]
9.
Shoemaker, R.H. The NCI60 human tumour cell line anticancer drug screen. Nat. Rev. Cancer
2006
,6,
813–823. [CrossRef]
10.
Basu, A.; Bodycombe, N.E.; Cheah, J.H.; Price, E.V.; Liu, K.; Schaefer, G.I.; Ebright, R.Y.; Stewart, M.L.; Ito, D.;
Wang, S.; et al. An interactive resource to identify cancer genetic and lineage dependencies targeted by small
molecules. Cell 2013,154, 1151–1161. [CrossRef]
11.
Holbeck, S.L.; Collins, J.M.; Doroshow, J.H. Analysis of Food and Drug Administration-approved anticancer
agents in the NCI60 panel of human tumor cell lines. Mol. Cancer Ther.
2010
,9, 1451–1460. [CrossRef]
[PubMed]
12.
Garnett, M.J.; McDermott, U. The evolving role of cancer cell line-based screens to define the impact of cancer
genomes on drug response. Curr. Opin. Genet. Dev. 2014,24, 114–119. [CrossRef] [PubMed]
Cancers 2020,12, 232 16 of 18
13.
Van de Wetering, M.; Francies, H.E.; Francis, J.M.; Bounova, G.; Iorio, F.; Pronk, A.; van Houdt, W.; van Gorp, J.;
Taylor-Weiner, A.; Kester, L.; et al. Prospective derivation of a living organoid biobank of colorectal cancer
patients. Cell 2015,161, 933–945. [CrossRef] [PubMed]
14.
Iorio, F.; Knijnenburg, T.A.; Vis, D.J.; Bignell, G.R.; Menden, M.P.; Schubert, M.; Aben, N.; Goncalves, E.;
Barthorpe, S.; Lightfoot, H.; et al. A Landscape of Pharmacogenomic Interactions in Cancer. Cell
2016
,166,
740–754. [CrossRef] [PubMed]
15.
Koerfer, J.; Kallendrusch, S.; Merz, F.; Wittekind, C.; Kubick, C.; Kassahun, W.T.; Schumacher, G.; Moebius, C.;
Gassler, N.; Schopow, N.; et al. Organotypic slice cultures of human gastric and esophagogastric junction
cancer. Cancer Med. 2016,5, 1444–1453. [CrossRef]
16.
Merz, F.; Gaunitz, F.; Dehghani, F.; Renner, C.; Meixensberger, J.; Gutenberg, A.; Giese, A.; Schopow, K.;
Hellwig, C.; Schafer, M.; et al. Organotypic slice cultures of human glioblastoma reveal dierent
susceptibilities to treatments. Neuro Oncol. 2013,15, 670–681. [CrossRef]
17.
Senkowski, W.; Zhang, X.; Olofsson, M.H.; Isacson, R.; Hoglund, U.; Gustafsson, M.; Nygren, P.; Linder, S.;
Larsson, R.; Fryknas, M. Three-Dimensional Cell Culture-Based Screening Identifies the Anthelmintic Drug
Nitazoxanide as a Candidate for Treatment of Colorectal Cancer. Mol. Cancer Ther.
2015
,14, 1504–1516.
[CrossRef]
18.
Sachs, N.; Clevers, H. Organoid cultures for the analysis of cancer phenotypes. Curr. Opin. Genet. Dev.
2014
,
24, 68–73. [CrossRef]
19.
Weeber, F.; Ooft, S.N.; Dijkstra, K.K.; Voest, E.E. Tumor Organoids as a Pre-clinical Cancer Model for Drug
Discovery. Cell Chem. Biol. 2017,24, 1092–1100. [CrossRef]
20.
Bleijs, M.; van de Wetering, M.; Clevers, H.; Drost, J. Xenograft and organoid model systems in cancer
research. EMBO J. 2019,38, e101654. [CrossRef]
21.
Lancaster,M.A.; Renner, M.; Martin, C.A.; Wenzel, D.; Bicknell, L.S.; Hurles,M.E.; Homfray, T.; Penninger, J.M.;
Jackson, A.P.; Knoblich, J.A. Cerebral organoids model human brain development and microcephaly. Nature
2013,501, 373–379. [CrossRef] [PubMed]
22.
Pauli, C.; Hopkins, B.D.; Prandi, D.; Shaw, R.; Fedrizzi, T.; Sboner, A.; Sailer, V.; Augello, M.; Puca, L.;
Rosati, R.; et al. Personalized In Vitro and In Vivo Cancer Models to Guide Precision Medicine. Cancer Discov.
2017,7, 462–477. [CrossRef] [PubMed]
23.
Lang, H.; Beraud, C.; Bethry, A.; Danilin, S.; Lindner, V.; Coquard, C.; Rothhut, S.; Massfelder, T. Establishment
of a large panel of patient-derived preclinical models of human renal cell carcinoma. Oncotarget
2016
,7,
59336–59359. [CrossRef] [PubMed]
24.
Morgan, K.M.; Riedlinger, G.M.; Rosenfeld, J.; Ganesan, S.; Pine, S.R. Patient-Derived Xenograft Models of
Non-Small Cell Lung Cancer and Their Potential Utility in Personalized Medicine. Front. Oncol.
2017
,7, 2.
[CrossRef]
25.
Maeda, H.; Khatami, M. Analyses of repeated failures in cancer therapy for solid tumors: Poor tumor-selective
drug delivery, low therapeutic ecacy and unsustainable costs. Clin. Transl. Med. 2018,7, 11. [CrossRef]
26.
Wong, C.C.; Cheng, K.W.; Rigas, B. Preclinical predictors of anticancer drug ecacy: Critical assessment with
emphasis on whether nanomolar potency should be required of candidate agents. J. Pharmacol. Exp. Ther.
2012,341, 572–578. [CrossRef]
27.
Ward, C.; Meehan, J.; Gray, M.; Kunkler, I.H.; Langdon, S.P.; Murray, A.; Argyle, D. Preclinical Organotypic
Models for the Assessment of Novel Cancer Therapeutics and Treatment. Curr. Top. Microbiol. Immunol.
2019
.
[CrossRef]
28.
Altman, R.B. Predicting cancer drug response: Advancing the DREAM. Cancer Discov.
2015
,5, 237–238.
[CrossRef]
29.
Guyot, C.; Combe, C.; Clouzeau-Girard, H.; Moronvalle-Halley, V.; Desmouliere, A. Specific activation of the
dierent fibrogenic cells in rat cultured liver slices mimicking
in vivo
situations. Virchows Arch.
2007
,450,
503–512. [CrossRef]
30.
Schmeichel, K.L.; Bissell, M.J. Modeling tissue-specific signaling and organ function in three dimensions.
J. Cell. Sci. 2003,116, 2377–2388. [CrossRef]
31.
Vaira, V.; Fedele, G.; Pyne, S.; Fasoli, E.; Zadra, G.; Bailey, D.; Snyder, E.; Faversani, A.; Coggi, G.; Flavin, R.;
et al. Preclinical model of organotypic culture for pharmacodynamic profiling of human tumors. Proc. Natl.
Acad. Sci. USA 2010,107, 8352–8356. [CrossRef] [PubMed]
Cancers 2020,12, 232 17 of 18
32.
De Hoogt, R.; Estrada, M.F.; Vidic, S.; Davies, E.J.; Osswald, A.; Barbier, M.; Santo, V.E.; Gjerde, K.; van
Zoggel, H.; Blom, S.; et al. Protocols and characterization data for 2D, 3D, and slice-based tumor models
from the PREDECT project. Sci. Data 2017,4, 170170. [CrossRef] [PubMed]
33.
Misra, S.; Moro, C.F.; Del Chiaro, M.; Pouso, S.; Sebestyen, A.; Lohr, M.; Bjornstedt, M.; Verbeke, C.S. Ex vivo
organotypic culture system of precision-cut slices of human pancreatic ductal adenocarcinoma. Sci. Rep.
2019,9, 2133. [CrossRef] [PubMed]
34.
Gerlach, M.M.; Merz, F.; Wichmann, G.; Kubick, C.; Wittekind, C.; Lordick, F.; Dietz, A.; Bechmann, I. Slice
cultures from head and neck squamous cell carcinoma: A novel test system for drug susceptibility and
mechanisms of resistance. Br. J. Cancer 2014,110, 479–488. [CrossRef] [PubMed]
35.
Marciniak, A.; Cohrs, C.M.; Tsata, V.; Chouinard, J.A.; Selck, C.; Stertmann, J.; Reichelt, S.; Rose, T.; Ehehalt, F.;
Weitz, J.; et al. Using pancreas tissue slices for in situ studies of islet of Langerhans and acinar cell biology.
Nat. Protoc. 2014,9, 2809–2822. [CrossRef]
36.
Rebours, V.; Albuquerque, M.; Sauvanet, A.; Ruszniewski, P.; Levy, P.; Paradis, V.; Bedossa, P.; Couvelard, A.
Hypoxia pathways and cellular stress activate pancreatic stellate cells: Development of an organotypic
culture model of thick slices of normal human pancreas. PLoS ONE 2013,8, e76229. [CrossRef]
37.
Kang, C.; Qiao, Y.; Li, G.; Baechle, K.; Camelliti, P.; Rentschler, S.; Efimov, I.R. Human Organotypic Cultured
Cardiac Slices: New Platform For High Throughput Preclinical Human Trials. Sci. Rep.
2016
,6, 28798.
[CrossRef]
38.
Jiang, T.; Zhou, C.; Ren, S. Role of IL-2 in cancer immunotherapy. Oncoimmunology
2016
,5, e1163462.
[CrossRef]
39.
Roelants, C.; Giacosa, S.; Pillet, C.; Bussat, R.; Champelovier, P.; Bastien, O.; Guyon, L.; Arnoux, V.; Cochet, C.;
Filhol, O. Combined inhibition of PI3K and Src kinases demonstrates synergistic therapeutic ecacy in
clear-cell renal carcinoma. Oncotarget 2018,9, 30066–30078. [CrossRef]
40.
Ricketts, C.J.; Linehan, W.M. Multi-regional Sequencing Elucidates the Evolution of Clear Cell Renal Cell
Carcinoma. Cell 2018,173, 540–542. [CrossRef]
41.
Kaelin, W.G., Jr. The von Hippel-Lindau tumour suppressor protein: O
2
sensing and cancer. Nat. Rev. Cancer
2008,8, 865–873. [CrossRef] [PubMed]
42.
Ricketts, C.J.; Crooks, D.R.; Linehan, W.M. Targeting HIF2alpha in Clear-Cell Renal Cell Carcinoma.
Cancer Cell 2016,30, 515–517. [CrossRef] [PubMed]
43.
Webster, W.S.; Lohse, C.M.; Thompson, R.H.; Dong, H.; Frigola, X.; Dicks, D.L.; Sengupta, S.; Frank, I.;
Leibovich, B.C.; Blute, M.L.; et al. Mononuclear cell infiltration in clear-cell renal cell carcinoma independently
predicts patient survival. Cancer 2006,107, 46–53. [CrossRef] [PubMed]
44.
Vuong, L.; Kotecha, R.R.; Voss, M.H.; Hakimi, A.A. Tumor Microenvironment Dynamics in Clear-Cell Renal
Cell Carcinoma. Cancer Discov. 2019,9, 1349–1357. [CrossRef]
45.
Kraus, V.B. Biomarkers as drug development tools: Discovery, validation, qualification and use. Nat. Rev.
Rheumatol. 2018,14, 354–362. [CrossRef]
46.
Motzer, R.J.; Tannir, N.M.; McDermott, D.F.; Aren Frontera, O.; Melichar, B.; Choueiri, T.K.; Plimack, E.R.;
Barthelemy, P.; Porta, C.; George, S.; et al. Nivolumab plus Ipilimumab versus Sunitinib in Advanced
Renal-Cell Carcinoma. N. Engl. J. Med. 2018,378, 1277–1290. [CrossRef]
47.
Rheinlander, A.; Schraven, B.; Bommhardt, U. CD45 in human physiology and clinical medicine. Immunol. Lett.
2018,196, 22–32. [CrossRef]
48.
Wu, P.; Wu, D.; Li, L.; Chai, Y.; Huang, J. PD-L1 and Survival in Solid Tumors: A Meta-Analysis. PLoS ONE
2015,10, e0131403. [CrossRef]
49.
Thompson, R.H.; Gillett, M.D.; Cheville, J.C.; Lohse, C.M.; Dong, H.; Webster, W.S.; Chen, L.; Zincke, H.;
Blute, M.L.; Leibovich, B.C.; et al. Costimulatory molecule B7-H1 in primary and metastatic clear cell renal
cell carcinoma. Cancer 2005,104, 2084–2091. [CrossRef]
50.
Thompson, R.H.; Dong, H.; Lohse, C.M.; Leibovich, B.C.; Blute, M.L.; Cheville, J.C.; Kwon, E.D. PD-1 is
expressed by tumor-infiltrating immune cells and is associated with poor outcome for patients with renal
cell carcinoma. Clin. Cancer Res. O. J. Am. Assoc. Cancer Res. 2007,13, 1757–1761. [CrossRef]
51.
Tumeh, P.C.; Harview, C.L.; Yearley, J.H.; Shintaku, I.P.; Taylor, E.J.; Robert, L.; Chmielowski, B.; Spasic, M.;
Henry, G.; Ciobanu, V.; et al. PD-1 blockade induces responses by inhibiting adaptive immune resistance.
Nature 2014,515, 568–571. [CrossRef] [PubMed]
Cancers 2020,12, 232 18 of 18
52.
Guertl, B.; Senanayake, U.; Nusshold, E.; Leuschner, I.; Mannweiler, S.; Ebner, B.; Hoefler, G. Lim1, an
embryonal transcription factor, is absent in multicystic renal dysplasia, but reactivated in nephroblastomas.
Pathobiology 2011,78, 210–219. [CrossRef] [PubMed]
53.
Hamaidi, I.; Coquard, C.; Danilin, S.; Dormoy, V.; Beraud, C.; Rothhut, S.; Barthelmebs, M.;
Benkirane-Jessel, N.; Lindner, V.; Lang, H.; et al. The Lim1 oncogene as a new therapeutic target for
metastatic human renal cell carcinoma. Oncogene 2019,38, 60–72. [CrossRef] [PubMed]
54.
Schnizlein-Bick, C.T.; Mandy, F.F.; O’Gorman, M.R.; Paxton, H.; Nicholson, J.K.; Hultin, L.E.; Gelman, R.S.;
Wilkening, C.L.; Livnat, D. Use of CD45 gating in three and four-color flow cytometric immunophenotyping:
Guideline from the National Institute of Allergy and Infectious Diseases, Division of AIDS. Cytometry
2002
,
50, 46–52. [CrossRef]
55.
Zerdes, I.; Matikas, A.; Bergh, J.; Rassidakis, G.Z.; Foukakis, T. Genetic, transcriptional and post-translational
regulation of the programmed death protein ligand 1 in cancer: Biology and clinical correlations. Oncogene
2018,37, 4639–4661. [CrossRef]
56.
Wang, Y.; Wang, X.Y.; Subjeck, J.R.; Shrikant, P.A.; Kim, H.L. Temsirolimus, an mTOR inhibitor, enhances
anti-tumour eects of heat shock protein cancer vaccines. Br. J. Cancer 2011,104, 643–652. [CrossRef]
57.
R Core Team. R: A Language and Environment for Statistical Computing; R Foundation for Statistical Computing:
Vienna, Austria; Available online: https://www.R-project.org/(accessed on 17 February 2018).
58.
Dienstmann, R.; Tabernero, J. Cancer: A precision approach to tumour treatment. Nature
2017
,548, 40–41.
[CrossRef]
59.
Jiang, X.; Seo, Y.D.; Chang, J.H.; Coveler, A.; Nigjeh, E.N.; Pan, S.; Jalikis, F.; Yeung, R.S.; Crispe, I.N.;
Pillarisetty, V.G. Long-lived pancreatic ductal adenocarcinoma slice cultures enable precise study of the
immune microenvironment. Oncoimmunology 2017,6, e1333210. [CrossRef]
©
2020 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access
article distributed under the terms and conditions of the Creative Commons Attribution
(CC BY) license (http://creativecommons.org/licenses/by/4.0/).
... Those subclones are often responsible of metastasis and are not many to be detected by bulk sequencing [6][7][8][9]. This leads to mRCC patients with a therapeutic response that differs significantly independently of their trunk mutation status [10,11]. ...
... A Vibratome VT1200 (Leica Microsystems) was used to cut thin (300 µm) slices from fresh tumor tissue as previously described [10,11]. Briefly, samples were soaked in ice-cold sterile-balanced salt solution (HBSS), orientated, mounted and immobilized using cyanoacrylate glue. ...
... Tissue slices were cultured as previously described [10,11]. Briefly, they were cultured on organotypic inserts for 48 h (one slice per insert; Millipore) at 37°C in a 5% CO 2 humidified incubator under agitation, using 2 mL of DMEM media supplemented with 10% inactivated fetal bovine serum (FBS) (GIBCO), 100 U/mL penicillin + streptomycin (Invitrogen). ...
Article
Full-text available
Clear cell Renal Cell Carcinoma (ccRCC) is one of the most prevalent kidney cancers, which is often asymptomatic and thus discovered at a metastatic state (mRCC). mRCC are highly heterogeneous tumors composed of subclonal populations that lead to poor treatment response rate. Several recent works explored the potential of ccRCC tumoroids culture derived from patients. However, these models were produced following a scaffold-based method using collagen I or Matrigel that exhibit lot variability and whose complexity could induce treatment response modifications and phenotypic alterations. Following the observation that ccRCC tumoroids can create their own niche by secreting extracellular matrix components, we developed the first scaffold-free tumoroid model of ccRCC tumors. Tumoroids from mice as well as from human tumors were generated with high success rate (≥90%) using a magnetic suspension method and standard culture media. Immunofluorescence analysis revealed their self-organization capacities to maintain multiple tumor-resident cell types, including endothelial progenitor cells. Transcriptomic analysis showed the reproducibility of the method highlighting that the majority of gene expression patterns was conserved in tumoroids compared to their matching tumor tissue. Moreover, this model enables to evaluate drug effects and invasiveness of renal cancer cells in a 3D context, providing a robust preclinical tool for drug screening and biomarker assessment in line with alternative ex vivo methods like tumor tissue slice culture or in vivo xenograft models.
... Those subclones are often responsible of metastasis and are not many to be detected by bulk sequencing [6,7,8,9]. This leads to mRCC patients with a therapeutic response that differs signi cantly independently of their trunk mutation status [10,11]. ...
... A Vibratome VT1200 (Leica Microsystems) was used to cut thin (300 µm) slices from fresh tumor tissue as previously described [10,11]. Brie y, samples were soaked in ice-cold sterile-balanced salt solution (HBSS), orientated, mounted and immobilized using cyanoacrylate glue. ...
... Tissue slices were cultured as previously described [10], [11]. Brie y, they were cultured on organotypic inserts for 48 h (one slice per insert; Millipore) at 37°C in a 5 % CO 2 humdi ed incubator under agitation, using 2 mL of DMEM media supplemented with 10% inactivated fetal bovine serum (FBS) (GIBCO), 100 U/mL penicillin + streptomycin (Invitrogen). ...
Preprint
Full-text available
Clear cell Renal Cell Carcinoma (ccRCC) is one of the most prevalent kidney cancers, which is often asymptomatic and thus discovered at a metastatic state (mRCC). mRCC are highly heterogeneous tumors composed of subclonal populations that lead to poor treatment response rate. Several recent works explored the potential of ccRCC tumoroids culture derived from patients. However, these models were produced following a scaffold-based method using collagen I or Matrigel that exhibit lot variability and whose complexity could induce treatment response modifications and phenotypic alterations. Following the observation that ccRCC tumoroids can create their own niche by secreting extracellular matrix components, we developed the first scaffold-free tumoroid model of ccRCC tumors. Tumoroids from mice as well as from human tumors were generated with high success rate (≥90 %) using a magnetic suspension method and standard culture media. Immunofluorescence analysis revealed their self-organization capacities to maintain multiple tumor-resident cell types, including endothelial progenitor cells. Transcriptomic analysis showed the reproducibility of the method highlighting that the majority of gene expression patternswas conserved in tumoroids compared to their matching tumor tissue. Moreover, this model enables to evaluate drug effects and invasiveness of renal cancer cells in a 3D context, providing a robust preclinical tool for drug screening and biomarker assessment in line with alternative ex vivo methods like tumor tissue slice culture or in vivo xenograft models.
... Biological hallmarks that contribute to the diagnosis, treatment and monitoring of RCC are urgently required [7,8]. Experimental research on RCC triggers the emergence of drug-target therapy [9,10]. Identification of specifically expressed genes during the progression of RCC may guide novel treatment in clinical practice [11]. ...
... In addition, TNM staging, incidental carcinoma, radical resection and postoperative immune therapy largely influence the prognosis in RCC [5][6][7][8]. Molecular mechanisms of RCC have been highlighted recently [9][10][11]. Recently discovered biomolecules, circRNAs, offer a promising application in gene therapy for tumors [16][17][18]. ...
Article
Purpose: To investigate the biological function and mechanisms of circPDSS1 in triggering malignant progression of renal cell carcinoma (RCC). Methods: Quantitative real-time polymerase chain reaction (qRT-PCR) was conducted to determine circPDSS1 levels in 50 pairs of RCC and para-cancerous tissues. The relationship between circPDSS1 level and pathological indices in RCC patients was analyzed, while the in vitro effect of circPDSS1 in regulating RCC proliferation was assessed using cell counting kit-8 (CCK-8), colony formation and 5- ethynyl-2’- deoxyuridine (EdU) assay. The sponge effect of circPDSS1 on miR-182-5p was examined by bioinformatics analysis and dual- luciferase reporter assay, while their involvement in mediating malignant progression of RCC was analyzed using rescue experiments. In vivo, the influence of circPDSS1 on RCC growth was determined by establishing a xenograft model in nude mice. Thereafter, RCC tissues were harvested from mice to assess relative levels of miR-182-5p and Ki-67. Results: CircPDSS1 was highly expressed in RCC tissues (p < 0.05). A high level of circPDSS1 correlated with advanced tumor staging and low overall survival. Knockdown of circPDSS1 inhibited RCC cell proliferation, and CircPDSS1 sponged and negatively regulated miR-182-5p (p < 0.05). MiR182-5p was able to abolish regulatory effect of circPDSS1 on malignant proliferative potential in RCC cells. In nude mice bearing RCC, in vivo knockdown of circPDSS1 slowed down tumor growth and decreased positive expression of Ki-67 in tumor tissues (p < 0.05). Conclusion: CircPDSS1 predicts tumor stage and prognosis in RCC patients. It triggers malignant progression of RCC through sponging of miR-182-5p.
... To this day, this is the closest human cancer model system to strengthen preclinical drug discovery and treatment decision in oncology [34][35][36][37][38]. In the past, tumor tissue slices have been used to study culture conditions and drug response assays with antineoplastic agents [8,35,39,40], analyze metastatic processes [41], and investigate new therapeutic or diagnostic strategies [42][43][44]. Other applications include studies on the regulation of tumor markers [45], the immune microenvironment [46], gene therapy [47,48], the antineoplastic potential of various agents [24,49,50], and drug sensitivity using electrochemical sensors [51], among others. ...
... As previously proven by Gerlinger et al. [25] and Kenerson et al. [76], which demonstrated different intratumoral zones that were separated only by a few millimeters and had different metabolic activities and gene expression profiles, which reflect real tumor behavior due in part to this heterogeneity. Ex vivo 3D models like the one we describe in this work allows the inclusion of different inter-and intratumor regions that constitute the microenvironment of each tumor, which in turn emphasizes the influences in the selection of optimal treatment for each patient [39,66,76,77] and provides a closer view of what happens in the niche of cancer cells in vivo. ...
Article
Full-text available
Breast cancer is one of the main causes of death worldwide. Lately, there is great interest in developing methods that assess individual sensitivity and/or resistance of tumors to antineoplastics to provide personalized therapy for patients. In this study we used organotypic culture of human breast tumor slices to predict the experimental effect of antineoplastics on the viability of tumoral tissue. Samples of breast tumor were taken from 27 patients with clinically advanced breast cancer; slices were obtained and incubated separately for 48 h with paclitaxel, docetaxel, epirubicin, 5-fluorouracil, cyclophosphamide, and cell culture media (control). We determined an experimental tumor sensitivity/resistance (S/R) profile by evaluating tissue viability using the Alamar Blue® metabolic test, and by structural viability (histopathological analyses, necrosis, and inflammation). These parameters were related to immunohistochemical expression of the estrogen receptor, progesterone receptor, and human epidermal growth factor receptor 2. The predominant histological type found was infiltrating ductal carcinoma (85.2%), followed by lobular carcinoma (7.4%) and mixed carcinoma (7.4%). Experimental drug resistance was related to positive hormone receptor status in 83% of samples treated with cyclophosphamide (p = 0.027). Results suggest that the tumor S/R profile can help to predict personalized therapy or optimize chemotherapeutic treatments in breast cancer.
... To further investigate NFE2L1 and NQO1 expression in podocytes, we used an ex vivo kidney injury model [26]. Slices of normal kidney parenchyma were cultured in DMSO, as a vehicle carrier control, and 20 µg/mL of PAN. ...
Article
Full-text available
Podocyte cellular injury and detachment from glomerular capillaries constitute a critical factor contributing to kidney disease. Notably, transcription factors are instrumental in maintaining podocyte differentiation and homeostasis. This study explores the hitherto uninvestigated expression of Nuclear Factor Erythroid 2-related Factor 1 (NFE2L1) in podocytes. We evaluated the podocyte expression of NFE2L1, Nuclear Factor Erythroid 2-related Factor 2 (NFE2L2), and NAD(P)H:quinone Oxidoreductase (NQO1) in 127 human glomerular disease biopsies using multiplexed immunofluorescence and image analysis. We found that both NFE2L1 and NQO1 expressions were significantly diminished across all observed renal diseases. Furthermore, we exposed human immortalized podocytes and ex vivo kidney slices to Puromycin Aminonucleoside (PAN) and characterized the NFE2L1 protein isoform expression. PAN treatment led to a reduction in the nuclear expression of NFE2L1 in ex vivo kidney slices and podocytes.
... For advanced ccRCC cases (stages III-IV), surgery remains the primary treatment (when the tumor is accessible), although adjuvant therapy is recommended [5]. Novel therapeutical options, such as targeted therapies and immunotherapies, often come with side effects and high costs and are often prescribed to patients with intermediate to poor prognoses [5,9]. Nevertheless, there is an urgent need for biomarker-driven studies with the overarching goal of incorporating newly proven biomarkers into innovative trial designs and giving patients a more accurate and individualized prognosis. ...
Article
Full-text available
Simple Summary Clear cell renal cell carcinoma (ccRCC) is the most common subtype of kidney cancer. Although it ranks as the seventh most diagnosed type of cancer, ccRCC is increasingly prevalent in the Western world. While most cases of ccRCC are diagnosed in the early stages, the cancer can often progress, spread to distant organs, and recur after treatment. Assessing the survival of ccRCC patients, and thus the aggressiveness of the cancer, can help clinicians make more informed decisions on patient management. We employed a systematic approach to find new biomarkers that can improve patient survival prediction. Our research revealed that increased expression of the COL7A1 gene is correlated with an aggressive form of the cancer and improve patient survival assessment, complementing the clinical characteristics that are already in use for survival prediction. Furthermore, our findings indicate that COL7A1 is a robust biomarker, that can be applied to patients of different origins and who undergo different treatment procedures. Interestingly, preliminary experiments led to the hypothesis that COL7A1 may play a role in cancer progression. Abstract Clear-cell renal cell carcinoma (ccRCC) accounts for 75% of kidney cancers. Due to the high recurrence rate and treatment options that come with high costs and potential side effects, a correct prognosis of patient survival is essential for the successful and effective treatment of patients. Novel biomarkers could play an important role in the assessment of the overall survival of patients. COL7A1 encodes for collagen type VII, a constituent of the basal membrane. COL7A1 is associated with survival in many cancers; however, the prognostic value of COL7A1 expression as a standalone biomarker in ccRCC has not been investigated. With five publicly available independent cohorts, we used Kaplan–Meier curves and the Cox proportional hazards model to investigate the prognostic value of COL7A1, as well as gene set enrichment analysis to investigate genes co-expressed with COL7A1. COL7A1 expression stratifies patients in terms of aggressiveness, where the 5-year survival probability of each of the four groups was 72.4%, 59.1%, 34.15%, and 8.6% in order of increasing expression. Additionally, COL7A1 expression was successfully used to further divide patients of each stage and histological grade into groups of high and low risk. Similar results were obtained in independent cohorts. In vitro knockdown of COL7A1 expression significantly affected ccRCC cells’ ability to migrate, leading to the hypothesis that COL7A1 may have a role in cancer aggressiveness. To conclude, we identified COL7A1 as a new prognosis marker that can stratify ccRCC patients.
... Still, reoccurrence occurs in around 30% of patients that undergo surgery, and this includes 10-25% of patients with localized disease [6]. Novel therapeutical options such as targeted therapies and immuno-therapies often come with side effects and high costs and are often prescribed to patients with intermediate to poor prognoses [7]. Nevertheless, there is an urgent need for biomarker-driven studies with an overarching goal to incorporate newly proven biomarkers for innovative trial designs and to give a more accurate and individualized prognosis to patients. ...
Preprint
Full-text available
Clear cell renal cell carcinoma (ccRCC) accounts for 75% of kidney cancers. Due to the high recurrence rate, and treatment options that come with high costs and potential side effects correct prognosis of patient survival is essential for the successful and effective treatment of patients. Novel biomarkers could play an important role in the assessment of the overall survival of patients. COL7A1 encodes for collagen type VII, a constituent of the basal membrane. COL7A1 is associated with survival in many cancers; however, the prognostic value of COL7A1 expression as a standalone biomarker in ccRCC has not been investigated. We used Kaplan-Meier curves and Cox proportional hazards model to investigate the prognostic value of COL7A1, as well as Gene Set Enrichment Analysis to investigate genes that are co-expressed with COL7A1. COL7A1 expression was used to stratify patients into four groups of expression, where the 5-year survival probability of each group was 72.4%, 59.1%, 34.15%, and 8.6% in order of increasing expression. Additionally, COL7A1 expression was successfully used to further divide patients of each stage and histological grade into groups of high and low risk. Similar results were obtained in independent cohorts. In-vitro knockdown of COL7A1 expression significantly impacted ccRCC cells' ability to migrate and proliferate. To conclude, we identified COL7A1 as a new prognosis marker that can stratify ccRCC patients.
... Kenerson et al. confirmed that PDTSCs reflect the metabolic activities, tumor cell density, proliferation rate, heterogeneity and clinical response to chemotherapies of the tumor [83]. In this context, PDTSC is a suitable alternative model for evaluating effectiveness of anticancer compounds in a complex tumor microenvironment [85,90]. ...
Article
Full-text available
Cancer is a heterogeneous disease. Each individual tumor is unique and characterized by structural, cellular, genetic and molecular features. Therefore, patient-derived cancer models are indispensable tools in cancer research and have been actively introduced into the healthcare system. For instance, patient-derived models provide a good reproducibility of susceptibility and resistance of cancer cells against drugs, allowing personalized therapy for patients. In this article, we review the advantages and disadvantages of the following patient-derived models of cancer: (1) PDC—patient-derived cell culture, (2) PDS—patient-derived spheroids and PDO—patient-derived organoids, (3) PDTSC—patient-derived tissue slice cultures, (4) PDX—patient-derived xenografts, humanized PDX, as well as PDXC—PDX-derived cell cultures and PDXO—PDX-derived organoids. We also provide an overview of current clinical investigations and new developments in the area of patient-derived cancer models. Moreover, attention is paid to databases of patient-derived cancer models, which are collected in specialized repositories. We believe that the widespread use of patient-derived cancer models will improve our knowledge in cancer cell biology and contribute to the development of more effective personalized cancer treatment strategies.
Article
In the era of personalized oncology, there have been accelerated efforts to develop clinically relevant platforms to test drug sensitivities of individual cancers. An ideal assay will serve as a diagnostic companion to inform the oncologist of the various treatments that are sensitive and insensitive, thus improving outcome while minimizing unnecessary toxicities and costs. To date, no such platform exists for clinical use, but promising approaches are on the horizon that take advantage of improved techniques in creating human cancer models that encompass the entire tumor microenvironment, alongside technologies for assessing and analyzing tumor response. This review summarizes a number of current strategies that make use of intact human cancer tissues as organotypic cultures in drug sensitivity testing.
Article
Full-text available
Patient-derived tumour xenografts and tumour organoids have become important preclinical model systems for cancer research. Both models maintain key features from their parental tumours, such as genetic and phenotypic heterogeneity, which allows them to be used for a wide spectrum of applications. In contrast to patient-derived xenografts, organoids can be established and expanded with high efficiency from primary patient material. On the other hand, xenografts retain tumour-stroma interactions, which are known to contribute to tumorigenesis. In this review, we discuss recent advances in patient-derived tumour xenograft and tumour organoid model systems and compare their promises and challenges as preclinical models in cancer research.
Article
Full-text available
Pancreatic ductal adenocarcinoma (PDAC) has a poor prognosis, which is mainly due to late diagnosis and profound resistance to treatment. The latter is to a large extent attributed to the tumor stroma that is exceedingly prominent in PDAC and engages in complex interactions with the cancer cells. Hence, relevant preclinical models of PDAC should also include the tumor stroma. We herein describe the establishment and functional validation of an ex vivo organotypic culture of human PDAC that is based on precision-cut tissue slices from surgical specimens and reproducibly recapitulates the complex cellular and acellular composition of PDAC, including its microenvironment. The cancer cells, tumor microenvironment and interspersed remnants of nonneoplastic pancreas contained in these 350 µm thick slices maintained their structural integrity, phenotypic characteristics and functional activity when in culture for at least 4 days. In particular, tumor cell proliferation persisted and the grade of differentiation and morphological phenotype remained unaltered. Cultured tissue slices were metabolically active and responsive to rapamycin, an mTOR inhibitor. This culture system is to date the closest surrogate to the parent carcinoma and harbors great potential as a drug sensitivity testing system for the personalized treatment of PDAC.
Article
Full-text available
Metastatic clear cell renal cell carcinoma (CCC) remains incurable despite advances in the development of anti-angiogenic targeted therapies and the emergence of immune checkpoint inhibitors. We have previously shown that the sonic hedgehog-Gli signaling pathway is oncogenic in CCC allowing us to identify the developmental Lim1 transcription factor as a Gli target and as a new oncogene in CCC regulating cell proliferation and apoptosis, and promoting tumor growth. In this previous study, preliminary in vitro results also suggested that Lim1 may be implicated in metastatic spread. Here we investigated the potential pro-metastatic role of Lim1 in advanced CCC (1) in vitro using a panel of CCC cell lines expressing or not the von Hippel-Lindau (VHL) tumor suppressor gene either naturally or by gene transfer and (2) ex vivo in 30 CCC metastatic tissues, including lymph nodes, lung, skin, bone, and adrenal metastases, and (3) in vivo, using a metastatic model by intravenous injection of siRNA-transfected cells into Balb/c nude. Our in vitro results reveal that Lim1 knockdown time-dependently decreased CCC cell motility, migration, invasion, and clonogenicity by up to 50% regardless of their VHL status. Investigating the molecular machinery involved in these processes, we identified a large panel of Lim1 targets known to be involved in cell adhesion (paxillin and fibronectin), epithelial-mesenchymal transition (Twist1/2 and snail), invasion (MMP1/2/3/8/9), and metastatic progression (CXCR4, SDF-1, and ANG-1). Importantly, Lim1 was found constitutively expressed in all metastatic tissues. The H-score in metastatic tissues being significantly superior to the score in the corresponding primary tumor tissues (P value = 0.009). Furthermore, we showed that Lim1 silencing decreases pulmonary metastasis development in terms of number and size in the in vivo metastatic model of human CCC. Taken together, these experiments strengthen the potential therapeutic value of Lim1 targeting as a promising novel approach for treating metastatic human CCC.
Article
Full-text available
Potent inhibitors of PI3K (GDC-0941) and Src (Saracatinib) exhibit as individual agents, excellent oral anticancer activity in preclinical models and have entered phase II clinical trials in various cancers. We found that PI3K and Src kinases are dysregulated in clear cell renal carcinomas (ccRCCs), an aggressive disease without effective targeted therapies. In this study we addressed this challenge by testing GDC-0941 and Saracatinib as either single agents or in combination in ccRCC cell lines, as well as in mouse and PDX models. Our findings demonstrate that combined inhibition of PI3K and Src impedes cell growth and invasion and induces cell death of renal carcinoma cells providing preclinical evidence for a pairwise combination of these anticancer drugs as a rational strategy to improve renal cancer treatment.
Article
Renal cell carcinoma stands out as one of the most immune-infiltrated tumors in pan-cancer comparisons. Features of the tumor microenvironment heavily affect disease biology and may affect responses to systemic therapy. With evolving frontline options in the metastatic setting, several immune checkpoint blockade regimens have emerged as efficacious, and there is growing interest in characterizing features of tumor biology that can reproducibly prognosticate patients and/or predict the likelihood of their deriving therapeutic benefit. Herein, we review pertinent characteristics of the tumor microenvironment with dedicated attention to candidate prognostic and predictive signatures as well as possible targets for future drug development. Significance: Tumor microenvironment features broadly characterizing angiogenesis and inflammatory signatures have shown striking differences in response to immune checkpoint blockade and antiangiogenic agents. Integration of stromal and immune biomarkers may hence produce predictive and prognostic signatures to guide management with existing regimens as well as future drug development.
Chapter
The immense costs in both financial terms and preclinical research effort that occur in the development of anticancer drugs are unfortunately not matched by a substantial increase in improved clinical therapies due to the high rate of failure during clinical trials. This may be due to issues with toxicity or lack of clinical effectiveness when the drug is evaluated in patients. Currently, much cancer research is driven by the need to develop therapies that can exploit cancer cell adaptations to conditions in the tumor microenvironment such as acidosis and hypoxia, the requirement for more-specific, targeted treatments, or the exploitation of ‘precision medicine’ that can target known genomic changes in patient DNA. The high attrition rate for novel anticancer therapies suggests that the preclinical methods used in screening anticancer drugs need improvement. This chapter considers the advantages and disadvantages of 3D organotypic models in both cancer research and cancer drug screening, particularly in the areas of targeted drugs and the exploitation of genomic changes that can be used for therapeutic advantage in precision medicine.
Article
Background In a single-group, phase 1b trial, avelumab plus axitinib resulted in objective responses in patients with advanced renal-cell carcinoma. This phase 3 trial involving previously untreated patients with advanced renal-cell carcinoma compared avelumab plus axitinib with the standard-of-care sunitinib. Methods We randomly assigned patients in a 1:1 ratio to receive avelumab (10 mg per kilogram of body weight) intravenously every 2 weeks plus axitinib (5 mg) orally twice daily or sunitinib (50 mg) orally once daily for 4 weeks (6-week cycle). The two independent primary end points were progression-free survival and overall survival among patients with programmed death ligand 1 (PD-L1)–positive tumors. A key secondary end point was progression-free survival in the overall population; other end points included objective response and safety. Results A total of 886 patients were assigned to receive avelumab plus axitinib (442 patients) or sunitinib (444 patients). Among the 560 patients with PD-L1–positive tumors (63.2%), the median progression-free survival was 13.8 months with avelumab plus axitinib, as compared with 7.2 months with sunitinib (hazard ratio for disease progression or death, 0.61; 95% confidence interval [CI], 0.47 to 0.79; P<0.001); in the overall population, the median progression-free survival was 13.8 months, as compared with 8.4 months (hazard ratio, 0.69; 95% CI, 0.56 to 0.84; P<0.001). Among the patients with PD-L1–positive tumors, the objective response rate was 55.2% with avelumab plus axitinib and 25.5% with sunitinib; at a median follow-up for overall survival of 11.6 months and 10.7 months in the two groups, 37 patients and 44 patients had died, respectively. Adverse events during treatment occurred in 99.5% of patients in the avelumab-plus-axitinib group and in 99.3% of patients in the sunitinib group; these events were grade 3 or higher in 71.2% and 71.5% of the patients in the respective groups. Conclusions Progression-free survival was significantly longer with avelumab plus axitinib than with sunitinib among patients who received these agents as first-line treatment for advanced renal-cell carcinoma. (Funded by Pfizer and Merck [Darmstadt, Germany]; JAVELIN Renal 101 ClinicalTrials.gov number, NCT02684006.)
Article
Background The combination of pembrolizumab and axitinib showed antitumor activity in a phase 1b trial involving patients with previously untreated advanced renal-cell carcinoma. Whether pembrolizumab plus axitinib would result in better outcomes than sunitinib in such patients was unclear. Methods In an open-label, phase 3 trial, we randomly assigned 861 patients with previously untreated advanced clear-cell renal-cell carcinoma to receive pembrolizumab (200 mg) intravenously once every 3 weeks plus axitinib (5 mg) orally twice daily (432 patients) or sunitinib (50 mg) orally once daily for the first 4 weeks of each 6-week cycle (429 patients). The primary end points were overall survival and progression-free survival in the intention-to-treat population. The key secondary end point was the objective response rate. All reported results are from the protocol-specified first interim analysis. Results After a median follow-up of 12.8 months, the estimated percentage of patients who were alive at 12 months was 89.9% in the pembrolizumab–axitinib group and 78.3% in the sunitinib group (hazard ratio for death, 0.53; 95% confidence interval [CI], 0.38 to 0.74; P<0.0001). Median progression-free survival was 15.1 months in the pembrolizumab–axitinib group and 11.1 months in the sunitinib group (hazard ratio for disease progression or death, 0.69; 95% CI, 0.57 to 0.84; P<0.001). The objective response rate was 59.3% (95% CI, 54.5 to 63.9) in the pembrolizumab–axitinib group and 35.7% (95% CI, 31.1 to 40.4) in the sunitinib group (P<0.001). The benefit of pembrolizumab plus axitinib was observed across the International Metastatic Renal Cell Carcinoma Database Consortium risk groups (i.e., favorable, intermediate, and poor risk) and regardless of programmed death ligand 1 expression. Grade 3 or higher adverse events of any cause occurred in 75.8% of patients in the pembrolizumab–axitinib group and in 70.6% in the sunitinib group. Conclusions Among patients with previously untreated advanced renal-cell carcinoma, treatment with pembrolizumab plus axitinib resulted in significantly longer overall survival and progression-free survival, as well as a higher objective response rate, than treatment with sunitinib. (Funded by Merck Sharp & Dohme; KEYNOTE-426 ClinicalTrials.gov number, NCT02853331.)
Article
Background: In the ongoing phase 3, CheckMate 214 trial, nivolumab plus ipilimumab improved overall survival compared with sunitinib in patients with intermediate or poor risk, previously untreated, advanced renal cell carcinoma. We aimed to assess whether health-related quality of life (HRQoL) could be used to further describe the benefit-risk profile of nivolumab plus ipilimumab versus sunitinib. Methods: In the phase 3, randomised, controlled, CheckMate 214 trial, patients aged 18 years and older with previously untreated, advanced or metastatic renal cell carcinoma with a clear-cell component were recruited from 175 hospitals and cancer centres in 28 countries. Patients were categorised by risk status into favourable, intermediate, and poor risk subgroups and randomly assigned (1:1) to open-label nivolumab 3 mg/kg plus ipilimumab 1 mg/kg every 3 weeks for four doses followed by nivolumab 3 mg/kg every 2 weeks, or sunitinib 50 mg/day for 4 weeks of each 6-week cycle. Randomisation was done with a block size of four and stratified by risk status and geographical region. Patient-reported outcomes (PROs) were assessed using the Functional Assessment of Cancer Therapy Kidney Symptom Index-19 (FKSI-19), Functional Assessment of Cancer Therapy-General (FACT-G), and EuroQol five dimensional three level (EQ-5D-3L) instruments. The coprimary endpoints of the trial, reported previously, were overall survival, progression-free survival, and the proportion of patients who had an objective response in those categorised as at intermediate or poor risk. PROs in all randomised participants were assessed as an exploratory endpoint; here we report this exploratory endpoint. This study is registered with ClinicalTrials.gov, number NCT02231749, and is ongoing but is now closed to recruitment. Findings: Between Oct 16, 2014, and Feb 23, 2016, of 1390 patients screened, 1096 (79%) were randomly assigned to treatment, of whom 847 (77%) were at intermediate or poor risk and randomly assigned to nivolumab plus ipilimumab (n=425) or sunitinib (n=422). Median follow-up was 25·2 months (IQR 23·0-27·4). PROs were more favourable with nivolumab plus ipilimumab than sunitinib throughout the first 103 weeks after baseline, with mean change from baseline at week 103 for FKSI-19 total score being 4·00 (95% CI 1·91 to 6·09) for nivolumab plus ipilimumab versus -3·14 (-6·03 to -0·25) for sunitinib (p<0·0001), and for FACT-G total score being 4·77 (1·73 to 7·82) for nivolumab plus ipilimumab versus -4·32 (-8·54 to -0·11) for sunitinib (p=0·0005). Significant differences were also seen for four of five FKSI-19 domains (disease-related symptoms, physical disease-related symptoms, treatment side-effects, and functional wellbeing) and FACT-G physical and functional wellbeing domains. However, there was no significant difference between the treatment groups at week 103 in EQ-5D-3L visual analogue rating scale (VAS) scores, with mean change from baseline to week 103 of 10·07 (95% CI 4·35 to 15·80) for nivolumab plus ipilimumab and 6·40 (-1·36 to 14·16) for sunitinib (p=0·45). Compared with sunitinib, nivolumab plus ipilimumab reduced risk of deterioration in FKSI-19 total score (hazard ratio [HR] 0·54; 95% CI 0·46-0·63), FACT-G total score (0·63, 0·52-0·75), and EQ-5D-3L VAS score (HR 0·75, 95% CI 0·63-0·89) and UK utility scores (0·67, 0·57-0·80). Interpretation: Nivolumab plus ipilimumab leads to fewer symptoms and better HRQoL than sunitinib in patients at intermediate or poor risk with advanced renal cell carcinoma. These results suggest that the superior efficacy of nivolumab plus ipilimumab over sunitinib comes with the additional benefit of improved HRQoL. Funding: Bristol-Myers Squibb and ONO Pharmaceutical.
Article
The systematic translation of cancer genomic data into knowledge of tumour biology and therapeutic possibilities remains challenging. Such efforts should be greatly aided by robust preclinical model systems that reflect the genomic diversity of human cancers and for which detailed genetic and pharmacological annotation is available¹. Here we describe the Cancer Cell Line Encyclopedia (CCLE): a compilation of gene expression, chromosomal copy number and massively parallel sequencing data from 947 human cancer cell lines. When coupled with pharmacological profiles for 24 anticancer drugs across 479 of the cell lines, this collection allowed identification of genetic, lineage, and gene-expression-based predictors of drug sensitivity. In addition to known predictors, we found that plasma cell lineage correlated with sensitivity to IGF1 receptor inhibitors; AHR expression was associated with MEK inhibitor efficacy in NRAS-mutant lines; and SLFN11 expression predicted sensitivity to topoisomerase inhibitors. Together, our results indicate that large, annotated cell-line collections may help to enable preclinical stratification schemata for anticancer agents. The generation of genetic predictions of drug response in the preclinical setting and their incorporation into cancer clinical trial design could speed the emergence of ‘personalized’ therapeutic regimens².