ArticlePDF Available

A Standard Protocol for the Production and Bioevaluation of Ethical In Vivo Models of HPV-Negative Head and Neck Squamous Cell Carcinoma

Authors:

Abstract and Figures

Preclinical cancer research increasingly demands sophisticated models for the development and translation of efficient and safe cancer treatments to clinical practice. In this regard, tumor-grafted chorioallantoic membrane (CAM) models are biological platforms that account for the dynamic roles of the tumor microenvironment and cancer physiopathology, allowing straightforward investigations in agreement to the 3Rs concept (the concept of reduction, refinement, and replacement of animal models). CAM models are the next advanced model for tumor biological explorations as well as for reliable assessment regarding initial efficacy, toxicity, and systemic biokinetics of conventional and emerging neoplasm treatment modalities. Here we report a standardized and optimized protocol for the production and biocharacterization of human papillomavirus (HPV)-negative head and neck chick chorioallantoic membrane models from a commercial cell line (SCC-25). Oral malignancies continue to have severe morbidity with less than 50% long-term survival despite the advancement in the available therapies. Thus, there is a persisting demand for new management approaches to establish more efficient strategies toward their treatment. Remarkably, the inclusion of CAM models in the preclinical research workflow is crucial to ethically foster both the basic and translational oncological research on oral malignancies as well as for the advancement of efficient cancer treatment approaches.
Content may be subject to copyright.
A Standard Protocol for the Production and Bioevaluation of Ethical
In Vivo Models of HPV-Negative Head and Neck Squamous Cell
Carcinoma
Patrizia Sarogni, Ana Katrina Mapanao, Sabrina Marchetti, Claudia Kusmic, and Valerio Voliani*
Cite This: https://doi.org/10.1021/acsptsci.1c00083
Read Online
ACCESS Metrics & More Article Recommendations
ABSTRACT: Preclinical cancer research increasingly demands
sophisticated models for the development and translation of
ecient and safe cancer treatments to clinical practice. In this
regard, tumor-grafted chorioallantoic membrane (CAM) models
are biological platforms that account for the dynamic roles of the
tumor microenvironment and cancer physiopathology, allowing
straightforward investigations in agreement to the 3Rs concept
(the concept of reduction, renement, and replacement of animal
models). CAM models are the next advanced model for tumor
biological explorations as well as for reliable assessment regarding
initial ecacy, toxicity, and systemic biokinetics of conventional
and emerging neoplasm treatment modalities. Here we report a standardized and optimized protocol for the production and
biocharacterization of human papillomavirus (HPV)-negative head and neck chick chorioallantoic membrane models from a
commercial cell line (SCC-25). Oral malignancies continue to have severe morbidity with less than 50% long-term survival despite
the advancement in the available therapies. Thus, there is a persisting demand for new management approaches to establish more
ecient strategies toward their treatment. Remarkably, the inclusion of CAM models in the preclinical research workow is crucial
to ethically foster both the basic and translational oncological research on oral malignancies as well as for the advancement of
ecient cancer treatment approaches.
KEYWORDS: chick chorioallantoic membrane, head/neck, cancer, alternative models, 3Rs principle, ethics
Head and neck squamous cell carcinomas (HNSCCs)
represent a wide class of epithelial neoplasms localized in
the oral and nasal cavities, paranasal sinuses, salivary glands,
pharynx, and larynx and whose molecular mechanisms involved
in the progression of the disease are still to be completely
claried.
1,2
One of the leading causes of the development of
HNSCCs is the long-term consumption of tobacco and
alcohol.
3
Another associated risk factor for the onset of
HNSCCs is human papillomavirus (HPV) infection.
4
The
presence of HPV usually aects the prognosis of the disease,
with more favorable outcomes for the patients.
4,5
The general
workup for staging and diagnosis of squamous cell carcinoma
of the oral cavity, larynx, oropharynx, and hypopharynx
includes physical examinations, medical history, blood test,
MRI/CT imaging, and biopsy under local anesthesia.
4
These
features together with the evaluations on the genomic
alterations or gene expression prole are important for the
pathological staging and prognosis and to determine the type
of treatment. In general, the usual management strategies for
oral cavity, laryngeal, oropharyngeal, and hypopharyngeal
cancers are dierent between locally advanced tumors and
early stage tumors.
6
Indeed, according with the most recent
Clinical Practice Guidelines in Oncology (e.g., NCCN), the
standard treatments in early stage disease, consisting of
conservative surgery or radiotherapy, give similar locoregional
control. Standard options for locally advanced HNSCC are
either surgery plus adjuvant chemo/radiotherapy or primary
chemoradiotherapy alone, even if the best treatment regime is
chosen on a case-by-case analysis.
4,7
In the eld of prognostic
estimation, it appears that cases of HNSCC that survive
beyond 5 years after the initial diagnosis show decreased
overall survival when compared to noncancer subjects of the
same age.
8
In general, stage I cases had improved survival
compared to stage IIIV, where no particular dierence was
proved in long-term survival for cases alive 5 years after
diagnosis. However, site, stage, smoking, and cardiovascular
Received: March 17, 2021
Articlepubs.acs.org/ptsci
© XXXX The Authors. Published by
American Chemical Society A
https://doi.org/10.1021/acsptsci.1c00083
ACS Pharmacol. Transl. Sci. XXXX, XXX, XXXXXX
Downloaded via 178.171.64.25 on April 16, 2021 at 13:13:03 (UTC).
See https://pubs.acs.org/sharingguidelines for options on how to legitimately share published articles.
disease are signicant factors determinant of mortality.
8
Nevertheless, oral malignancies, including tongue cancer,
continue to have severe morbidity with less than 50% long-
term survival despite the advancement in the available and
emerging therapies.
4,5,9,10
In this regard, understanding the
biological processes at the basis of oral malignancies and the
development of new therapeutic strategies to improve the
survival rate of patients while preserving the structure and
function of the involved organs are crucial topics for their
management.
11
In this context, in vivo models are pivotal to
foster the advancements in oncology by bridging the gap
between preclinical investigations and human clinical trial on
conventional and emerging therapeutic approaches as well as
to understand tumor cell behaviors in a physiological
environment.
19
The most widely employed in vivo models
are murine, and they have been crucial to establish most of the
current models in pediatric oncology and to develop some
drugs, among which topotecan and irinotecan.
7
Moreover,
murine models are particularly relevant for absorption
distributionmetabolismexcretiontoxicity (ADMET) in-
vestigations.
1214
However, genetically immunocompromised
murine models have very high costs of maintenance, and the
tumor engraftment may require up to 4 months. Engraftment
failure can be high and cannot be usually determined before
some months post-implantation, and their employment is
increasingly discouraged by following the 3Rs concept (the
concept of reduction, renement, and replacement of animal
models) and the European Parliament Directive 2010/63/
EU.
15,16
Among other biological models, the chick chorioal-
lantoic membrane (CAM) is one of the most attractive and
ethical in vivo models that jointly combines reliability,
medium-/high-throughput screenings, and easy handling
during imaging/treatment evaluations.
1719
Notably, the
absence of a mature immune system of the embryo during
the early developmental stage reduces the risk of tumor
rejection after implantation.
19
Thus, CAM models can develop
visible solid tumors within 45 days after engraftment, in
comparison to the approximately 36 weeks of murine
models.
7
Besides the usually high rate of success for tumor
grafting, other major advantages of CAM models are the easy
daily inspection of the tumor site and their exibility of
employment. Moreover, CAM models are ethical models in
agreement with the 3Rs concept because the chick embryo
does not develop pain perception before the 17th day of
incubation.
19
Indeed, investigations in CAM models usually do
not require permissions or approval from ethics committees.
19
Despite some diculties in the application of CAM models to
long-term investigations, their highly vascularized membrane
together with the immature immune response allowed the low
rejection rate grafting of several tissues and the study of various
neoplasms, including osteosarcoma, glioblastoma, pancreatic
carcinoma, and colon carcinoma.
2023
Several variable tumor
engraftment methods for CAM model production have been
reported, yet these studies focus on the tumor biology or the
evaluation of a treatment avoiding reporting a standard
protocol for the production of the models.
24,25
In general, in
the literature there is a serious lack of work regarding the
standard production of CAM models. Thus, a detailed step-by-
step procedure for the reliable CAM tumor model fabrication
together with the standard biocharacterization is required to
serve as guide for researchers interested to advance the eld.
In order to address this demand, we report a standardized
protocol for the composition as well as the cascade assays for
the characterization of a commercial HPV-negative head and
neck cell line (SCC-25) grafted on chick chorioallantoic
membrane of fertilized Leghorn chicken eggs.
MATERIALS
Reagents and Consumables.
Fertilized red or white Leghorn chicken eggs
SCC-25 squamous cell carcinoma cell line (ATCC,
catalog number CRL-1628)
DMEM/Hams F12 1:1 medium (DMEM/F12 Gibco,
21041025)
Fetal bovine serum, qualied, heat-inactivated (FBS,
Thermo Fisher Scientic, 10500064)
L-Glutamine (Thermo Fisher Scientic, A2916801)
Hydrocortisone (Sigma-Aldrich, H0888)
Penicillinstreptomycin (Pen/Strep 100X, 5,000 U/
mL) (Thermo Fisher Scientic, 15070063)
Phosphate-buered saline without calcium and magne-
sium (PBS, Sigma-Aldrich, D8537)
Trypsin-EDTA (0.5%), phenol red (Thermo Fisher
Scientic, 25300054)
Serological pipette, pipette tips, microcentrifuge tubes,
conical tubes, and asks
TC Dish150, Standard (83.3903, SARSTEDT)
Matrigel Matrix (Corning, ref 354234)
Sterile water
Fixative solution, i.e., 4% paraformaldehyde (PFA) in
PBS
RNA extraction kit, Nucleospin RNA plus (740984.50
MACHEREY-NAGEL)
cDNA synthesis kit, iScript cDNA Synthesis (1708891
BIORAD)
iTaqUniversal SYBRGreen Supermix (1725121 BIO-
RAD)
RIPA buer (Pierce 89901)
Protease Inhibitors Cockatil Tablets (04693116001
Roche)
Bradford Reagent (B6916 Sigma-Aldrich)
Albumin Standard (23209 Thermo scientic)
Nitrocellulose membrane, TransBlot Turbo Midi-size
nitrocellulose (1620167 BIORAD)
anti-TFRC primary antibody (SAB4200398 Sigma-
Aldrich)
Goat anti-rabbit IgG (H+L)-HRP-conjugated secondary
antibody, (1706515 BIORAD)
Clarity Western ECL substrate (1705061 BIORAD)
Paran wax (melting point 56 °C)
Ethanol (70, 80, 95, and 100% alcohol)
Xylene
Mayers hematoxylin solution
Eosin Y aqueous solution 1%
Permount mounting medium
Equipment and Tools.
Cell counter (Invitrogen Countess cell counter)
Optical microscope
Egg incubator, 37.5 °C/99.5 °F, 60% humidity, FIEM
MG 140/200
Tilting egg racks
Sterile dissection scissors and tweezers
Soft tissue paper or cotton swab
Refrigerator or cold room at 4 °C
Analytical balance
ACS Pharmacology & Translational Science pubs.acs.org/ptsci Article
https://doi.org/10.1021/acsptsci.1c00083
ACS Pharmacol. Transl. Sci. XXXX, XXX, XXXXXX
B
Ruler
Adhesive tapes, preferably Scotch magic tape
Portable digital microscope, Dino-Lite AM7915MZT
(or any camera-associated microscope)
DinoCapture 2.0 Software
Rotary microtome
Forced ventilation histology oven
Paran-embedding station
Light microscope equipped with RGB video camera
METHODS
Experimental Outline. The establishment of the exper-
imental design is the rst important step to dene a complete
outline of the assay procedure. For the optimization of the
CAM assay employing SCC-25 cells, the schedules of egg
incubation and subsequent experimentations were based on
the proposed scheme by Kleibeuker et al. (Figure 1).
26
In
general, the start of the incubation corresponds to the
embryonic day of development 0 (EDD0). Starting from this
moment, under appropriate environmental conditions, the
fertilized eggs begin their embryonic growth. It is important to
take into account that the biological window available to
perform this assay must not exceed the 17 days of incubation
to prevent the hatching of the eggs and avoid ethical
restrictions.
19
Cleaning the Working Area and Eggs (Start of
Incubation). It is highly recommended to work under sterile
conditions and to adequately clean the working area with 70%
ethanol and/or bleach to avoid any type of microorganism
contamination. Carefully clean the shell of each egg with soft
tissue paper soaked in distilled water before tting them into
the tray.
27
Place the eggs horizontally next to each other and
insert a steel spring in the ends of the columns to secure the
eggs while the racks tilt (Figure 2A).
28
Make sure that the
temperature and humidity of the incubator reach the
appropriate parameters before starting the incubation (37.5
°C, 47% humidity).
29
Puncturing. Take the eggs out after 3 days of incubation
(EDD3) and put them in upright position so that the air sac
will translocate to the top of the eggs.
30
Gently scratch with
tweezers the tip of each egg until the shell becomes fragile and
breakable, nally making a small hole. Seal the hole with an
adhesive tape to prevent dehydration and infection, and put
the eggs back in the incubator in the stationary mode (no
tilting).
Tumor Grafting. Selection of Eggs. At EDD6, the
fertilized eggs must be accurately selected before proceeding
with the grafting of the tumor cell suspension on the CAM.
Remove the eggshell around the small hole to create a small
window of about 1 cm2.
31
Check the fertilization and discard
the unfertilized eggs (Figure 2C). Label the eggs with the
appropriate information (e.g., egg number, cell density, name
of the cell line, and treatment conditions) to avoid confusion.
Reseal the window, and put the eggs back in the incubator.
Preparation of Tumor Cell Suspension. Harvest the SCC-
25 cells, and collect them in one 50 mL tube.
32
Spin down the
cells, and discard the supernatant. Resuspend the pellet in fresh
cell culture medium. Count the cells, and adjust the
concentration in order to have at least 2 ×106cells per
egg.
33
Spin down the cells again, and discard the supernatant.
Resuspend the pellet in a mixture of Matrigel/medium without
FBS (in ratio 1:1) such that each egg will be dispensed with 25
μL of the cell suspension.
34
Grafting. Open the sealed window. Roll a soft tissue paper
and gently poke a small vein within the CAM region until it
starts to bleed (Figure 2D).
35
Pipette 25 μL of the cell
suspension on top of the bleeding blood vessel. Reseal the
window, and put the eggs in the incubator (Figure 2E). Always
make sure that the humidier and the tank are lled with water
and that the incubator is still maintaining the right conditions
of temperature and humidity.
Figure 1. Overview of the CAM schedule from EDD0 to EDD17. In general, chick embryo incubation is marked as embryonic day of development
0 (EDD0). The puncturing day (EDD3) allows the translocation of the natural air sac to the top of the egg. Grafting of 2 ×106SCC-25 cells
(EDD6) enables the generation of a solid visible tumor at 4 days post-grafting (EDD10). The topical treatment is applied on EDD10, and the
tumor mass is monitored until EDD17, the last day of incubation (harvesting).
ACS Pharmacology & Translational Science pubs.acs.org/ptsci Article
https://doi.org/10.1021/acsptsci.1c00083
ACS Pharmacol. Transl. Sci. XXXX, XXX, XXXXXX
C
Apply Treatment (EDD10). A reference concentration of
your drug/chemotherapeutic compound to be tested is
recommended.
36
The amount of the treatment solution to
be prepared will depend on the number of tumor-grafted eggs
to be treated, while the frequency of the treatment will depend
on your own schedule and experimental objectives. To
proceed, take the eggs out from the incubator, and mark
each egg for the corresponding treatment to avoid confusion.
For our experiments, each egg is topically administered with
chemotherapeutic drugs/test compounds suspended in 30 μL
of solution (serum-free medium).
37
Tumor Growth Monitoring (EDD10EDD17). Place a
ruler under the DinoLite camera and calibrate the software
before starting to acquire images of the tumors. Although
constantly noting the calibration by using the ruler can help in
obtaining consistent and accurate measurements, it is also
advised to take photographs at the same magnication to
better visualize the changes in tumor dimensions. The
complementary DinoLite software (DinoCapture 2.0) comes
with length measurement tools. Measure the tumor sizes, in
which the longer and shorter measurements are denoted as the
length (L) and width (W), respectively (Figure 2F). From
these measurements, the tumor volume is derived using a
modied ellipsoid formula: 0.5(L×(W2)).
38,22
Tumor Harvesting (EDD17). On EDD17, take the eggs
out of the incubator and place them in the cold room for at
least 2 h to restrict the movements of the chick embryo.
Thoroughly clean the dissection area, tweezers, and scissors
with 70% ethanol and bleach. Prepare wash containers with
ethanol and PBS for rinsing the tools to avoid cross-
contamination among dierent tumors. Open the sealed
window, and remove some eggshell around to make the
tumor more accessible for cutting. Carefully lift the membrane
with tweezers. Cut the tumor, and place it in a Petri dish with
PBS. Take a photograph of the tumor, and place it in an empty
microcentrifuge tube, and weigh it (Figure 2G).
39
Store the
harvested tumor samples directly in 80 °C or in formalin
xing solution for further analysis.
DOWNSTREAM ASSAYS
Quantitative Real Time-PCR. To extract the total RNA
from the SCC-25 cell line, we used Nucleospin RNA plus Kit
(740984.50 MACHEREY-NAGEL) following the manufac-
turers instruction. Briey, the harvested tissue is minced in
small pieces using a plastic pestle. The extracted RNA can be
used immediately or stored at 80 °C. The quality control of
RNA through agarose gel electrophoresis will avoid the
likelihood of nal problem solving, such as in cDNA reverse
transcription and amplication of the gene target. For cDNA
synthesis, 500 ng of RNA was reverse-transcribed with iScript
cDNA Synthesis Kit (1708891 BIORAD). Dilute 500 ng of the
total cDNA 1:10 in nuclease-free water in order to get 50 ng as
nal amount before using it for the PCR reaction. Quantitative
real-time PCR is carried out using iTaqUniversal SYBRGreen
Supermix (1725121 BIORAD). To prepare the PCR reaction
mix in a nal volume of 20 μL, use 12μL of the diluted
cDNA with around 510 ng of cDNA template. Use specic
primers for your gene of interest and primers for a
housekeeping gene. One of the commonly utilized house-
keeping genes, which we also use, is GAPDH. All the samples
are prepared in triplicate. The amplication curves are
visualized by SYBR Green Analysis on Applied Biosystem
Instrument (7300). The recommended thermal cycling for the
amplication is as follows: 95 °C for 10 min, 40 cycles at 95 °C
15 s, 64 °C for 30 s, and 72 °C for 30 s. The 2ΔΔCT method is
used to calculate the relative expression level.
40
Western Blotting. Cells pellets are resuspended in RIPA
buer (Pierce 89901) supplemented with protease inhibitors
and mechanically minced using a 200 μL pipet tip. The lysates
Figure 2. Images of the main steps of the CAM protocol. (A) Eggs are
horizontally placed in the tilting trays before the incubation starts. (B)
A light source placed on top/behind the egg allows identication of
the location of the air chamber and the existence of a vascular network
(Top, infertile egg; bottom, fertile egg). (C) An unfertilized egg is
associated with the absence of the embryo and of the vascular
network. (D) Distinction between artery (arrow) and vein (asterisk).
Arteries are thicker and darker compared to veins.
41
(E) Image
showing the grafting procedure of SCC-25 cancer cells onto the
bleeding blood vessel. (F) Tumor volume measurement determined
by means of width and length. The length is associated with the
longest diameter of the tumor mass. (G) Harvesting procedure of the
tumor at EDD17. The CAM membrane is gently lifted with tweezers
and the tumor is cut with scissors.
ACS Pharmacology & Translational Science pubs.acs.org/ptsci Article
https://doi.org/10.1021/acsptsci.1c00083
ACS Pharmacol. Transl. Sci. XXXX, XXX, XXXXXX
D
are then incubated for 30 min in ice, and the supernatants are
collected after centrifugation for 30 min at 14 000 rpm. The
protein concentration in the lysates is determined through the
Bradford assay, using a standard calibration curve method
prepared with bovine serum albumin (BSA) of known
concentrations (2000 μg/mL). The absorbance values at 595
nm of the samples and the standards are noted. The formula y
=mx +qis used to derive the protein concentration. Then,
3050 μg of the total protein was separated via SDS-PAGE.
Upon transferring the samples from the gel to a nitrocellulose
membrane, the proteins are treated with a blocking solution
(TBS 1×5% powdered milk) for 1 h at room temperature. For
the overnight primary antibody incubation at 4 °C, we used
anti-TFRC primary antibody (SAB4200398 Sigma-Aldrich).
The membrane was washed thrice with TBS 1×0.1%
Tween20, and the horseradish peroxidase (HRP)-conjugated
secondary antibody was then added and incubated for 1 h at
room temperature. Finally, after further washing in TBS 1×
0.1% Tween20, the bands are visualized through chemilumi-
nescence using an enhanced chemiluminescence (ECL) kit
(1705061 Biorad) and Image Quant LAS 4000 System.
Hematoxylin and Eosin (H&E) Staining. Tumor
samples, xed in 4% paraformaldehyde for 2448 h, were
rinsed in running tap water for 1015 min. Then, dehydrate
samples through increasing alcohol series, followed by three
changes of 100% alcohol applied for 5 min each. The tissues
were cleared in xylene for 12 min and then immersed in
paran wax for 10 min, followed by other two paran
changes, 5 min each, and nally embedded in paran blocks.
Serial sections of 56μm were cut with a microtome, placed
on slides, and heated overnight at 40 °C in forced ventilation
histology oven. The sections were cleared from paran by two
changes in xylene, 6 min each. The tissue was hydrated
through decreasing ethanol series and rinsed in distilled water
for at least 5 min.
Slices were stained with Mayers hematoxylin solution for 5
min, followed by a 10 min rinse in running tap water, and
nally stained in eosin Y aqueous solution for 2 min. Slides
were rinsed in distilled water, heated at 40 °C for 40 min,
dipped twice in xylene, and a coverslip was added placing a
drop of Permount mounting medium.
Histological images (40×, 100×, 200×, and 400×) are
acquired by light microscope (Olympus BX43, Japan) and
digitized using a RGB video camera (Olympus DP 20, Japan).
ANTICIPATED RESULTS
ThereliablegenerationofasolidtumoronCAMis
fundamental for both the comprehension of in vivo cancer
cell behaviors and the ecacy/toxicity evaluation of emerging
and conventional therapeutic strategies. Imaging of tumors
provides detailed information about the quality and the
presence/absence of blood vessel across the tumor itself; an
important parameter to monitor during the time frame of the
experiment and/or after the therapeutic treatments.
42
Imaging
also provides a practical method to identify the volume of the
tumor mass without interfering with its spatial organization
and allows for distinguishing the tumor from artifacts caused
by the aggregation of the Matrigel solution. The primary goals
in the production of CAM tumor models include the
achievement of a high embryo survival rate and visible tumor
grafting that allow the topical application of therapeutics. Our
optimized protocol allows the development of solid vascular-
ized SCC-25 tumor with high eciency (80%) and with a
volume of 520 mm3at EDD10 (Figure 3A). It is important
to notice that the assessment of tumor size following the
harvesting can be aected by the wrinkling of the membrane
Figure 3. Characterization of harvested SCC25 tumors. (A) Representative image of SCC-25 solid tumor grown onto the CAM. The arrows
indicate the blood vessels across the tumor mass. (B) Example of solid and vascularized tumor harvested at EDD17. (C) Western blotting analysis
depicting the expression of the TfR marker in SCC-25-tumor-derived cells. (D) H&E staining of SCC-25 tumor-derived cells showing the tissue
structure and cells distribution (TC = tumor cells; S = stroma; scale bar = 20 μm). (E) Real-time PCR measurement of VEGF-A mRNA expression
levels in SCC-25 cancer cells compared to HBEpc bronchial cell line.
ACS Pharmacology & Translational Science pubs.acs.org/ptsci Article
https://doi.org/10.1021/acsptsci.1c00083
ACS Pharmacol. Transl. Sci. XXXX, XXX, XXXXXX
E
after 2 h at 4 °C. However, the size of the excised tumors
usually signicantly correlates with their in vivo volume.
Measuring the weight of the excised tumors is a facile and
useful end-point evaluation that provides additional informa-
tion on the aggressiveness and ability of cancer cells to grow
and form solid tumors (Figure 3B). It is worth remembering
that the identication and introduction of new molecular
pathways involved in the neoplastic transition has rapidly
expanded, considerably advancing the diagnostic techniques.
43
In fact, the detection of specic biomolecular tumor markers
represents a signicant diagnostic screening approach that
allows dierentiation between cancer cells and the surrounding
cells. Among the number of molecular mechanisms already
identied in neoplasms, the expression of transferrin receptor
(TfR) appears to be compromised in HNSCCs, leading to an
overexpression of the receptor and therefore constituting a
promising tumor marker.
44,45
In this regard, to eectively
prove the human origin of the harvested tissue, Western blot
analysis has been carried out using an antihuman TfR
antibody. The results demonstrate the presence of the TfR
protein maker in the harvested tumor samples, conrming the
signicance of this approach to determinate the human origin
of the grafted tumor (Figure 3C).
H&E staining is another pivotal end-point assay that mainly
provides qualitative information and a general overview on the
structure of the tissues.
46
Remarkably, this approach enables
visualization of cellular morphology and the distribution
pattern of distinct cells, as well as their density and consistency.
The H&E staining of the collected SCC-25 tumor specimen
showed a well-organized, preserved, and homogeneous
structure, clearly identiable from the surrounding stroma
and the membrane border (Figure 3D).
Since genetic alterations are often events upstream of cell
metabolism dysfunction, advances in the assessment of gene
expression prole may be useful for the prognosis of various
type of cancers, hence oering a wide overview about the gene
interaction network in the development of the disease. In fact,
mutations that lead to the alteration of gene expression level of
cytokines, enzymes, and growth factors are the main
responsible of tumorigenesis.
47
The deregulation of the
vascular endothelial growth factor-A (VEGF-A) was observed
to be among the tumor promoting factors in SCC-25 cells.
Indeed, VEGF-A is relatively overexpressed in SCC-25 cells
compared to the level in normal cells (Figure 3E) and
constitute a promising molecular therapeutic target for head
and neck tumors. Overall, these ndings aim to emphasize the
use of the chick embryo as an in vivo model for the screening of
anticancer compounds.
SUMMARY
HNSCCs represent an aggressive class of neoplasms with a
worldwide high incidence due to two main risk factors:
tobacco/alcohol consumption and HPV infection.
48
The
translation of ecient therapeutic strategies in oncology
requires a deep understanding beyond the physiopathology
of neoplasms as well as ethical and reliable in vivo models that
provide an analogous environment for an accurate exploration
of the behavior of cancer tissues and their response to the
therapeutic approaches. Due to its highly vascularized
environment and immature immune responses, the CAM
model provides an optimal environment for the grafting of
cancer cell lines and patient-derived cancer cells.
49
Here, we have presented a standardized and optimized
protocol for the medium-/high-throughput production of solid
tumors using the commercial HPV-negative head and neck cell
line SCC-25. Each step of the protocol requires basic biological
practice and experience. In general, the high survival rate of the
embryos and tumor take rate are among the essential criteria
considered for the eciency of the protocol. Additionally, a
detailed outline and explanation of the steps and further
remarks have been included to provide a suitable guide for the
generation of tumor grafts and subsequent imaging and
biomolecular assays. The reported step-by-step method allows
the establishment of a feasible in vivo model that can provide
insights on the biological processes at the basis of oral
malignancies and the development of new therapeutic
strategies.
50
The CAM model may push oncological research
toward a more rapid evaluation and ecient screening/
selection of conventional and emerging antitumor treatments.
AUTHOR INFORMATION
Corresponding Author
Valerio Voliani Center for Nanotechnology Innovation@
NEST, Istituto Italiano di Tecnologia, Pisa 56126, Italy;
orcid.org/0000-0003-1311-3349;
Email: valerio.voliani@iit.it
Authors
Patrizia Sarogni Center for Nanotechnology Innovation@
NEST, Istituto Italiano di Tecnologia, Pisa 56126, Italy
Ana Katrina Mapanao Center for Nanotechnology
Innovation@NEST, Istituto Italiano di Tecnologia, Pisa
56126, Italy; NEST-Scuola Normale Superiore, Pisa 56126,
Italy
Sabrina Marchetti Institute of Clinical Physiology, CNR,
Pisa 56100, Italy
Claudia Kusmic Institute of Clinical Physiology, CNR, Pisa
56100, Italy
Complete contact information is available at:
https://pubs.acs.org/10.1021/acsptsci.1c00083
Notes
The authors declare no competing nancial interest.
ACKNOWLEDGMENTS
This work is supported by Associazione Italiana per la Ricerca
sul Cancro (AIRC) under MFAG 2017, ID 19852 project
(P.I.: V.V.). Figure 1 and the graphical abstract have been
created with BioRender.com.
REFERENCES
(1) Kim, N., Ryu, H., Kim, S., et al. (2019) CXCR7 promotes
migration and invasion in head and neck squamous cell carcinoma by
upregulating TGF-β1/Smad2/3 signaling. Sci. Rep. 9 (1), 111.
(2) Li, P., Bian, X.-y., Chen, Q., Yao, X.-f., Wang, X.-d., Zhang, W.-c.,
Tao, Y.-j., Jin, R., and Zhang, L. (2017) Blocking of stromal
interaction molecule 1 expression influence cell proliferation and
promote cell apoptosis in vitro and inhibit tumor growth in vivo in
head and neck squamous cell carcinoma. PLoS One 12 (5), e0177484.
(3) Argiris, A., Karamouzis, M. V, Raben, D., and Ferris, R. L (2008)
Head and neck cancer. Lancet 371, 16951709.
(4) Machiels, J. P., René Leemans, C., Golusinski, W., Grau, C.,
Licitra, L., and Gregoire, V. (2020) Squamous cell carcinoma of the
oral cavity, larynx, oropharynx and hypopharynx: EHNS-ESMO-
ESTRO Clinical Practice Guidelines for diagnosis, treatment and
follow-up.Ann. Oncol. 31 (11), 14621475.
ACS Pharmacology & Translational Science pubs.acs.org/ptsci Article
https://doi.org/10.1021/acsptsci.1c00083
ACS Pharmacol. Transl. Sci. XXXX, XXX, XXXXXX
F
(5) Mapanao, A. K., Santi, M., and Voliani, V. (2021) Combined
chemo-photothermal treatment of three-dimensional head and neck
squamous cell carcinomas by gold nano-architectures. J. Colloid
Interface Sci. 582, 10031011.
(6) Lango, M. N. (2009) Multimodal Treatment for Head and Neck
Cancer. Surg. Clin. North Am. 89 (1), 4352.
(7) Cognetti, D. M., Weber, R. S., and Lai, S. Y. (2008) Head and
neck Cancer an evolving treatment paradigm. Cancer 113 (7), 1911
1932.
(8) Du, E., Mazul, A. L., Farquhar, D., Brennan, P., Anantharaman,
D., Abedi-Ardekani, B., Weissler, M. C., Hayes, D. N., Olshan, A. F.,
and Zevallos, J. P. (2019) Long-term Survival in Head and Neck
Cancer: Impact of Site, Stage, Smoking, and Human Papillomavirus
Status. Laryngoscope 129, 25062513.
(9) Santi, M., Mapanao, A. K., Biancalana, L., Marchetti, F., and
Voliani, V. (2021) Ruthenium arene complexes in the treatment of
3D models of head and neck squamous cell carcinomas. Eur. J. Med.
Chem. 212, 113143.
(10) Santi, M., Mapanao, A. K., Cassano, D., Vlamidis, Y., Cappello,
V., and Voliani, V. (2020) Endogenously-activated ultrasmall-in-nano
therapeutics: Assessment on 3d head and neck squamous cell
carcinomas. Cancers 12 (5), 1063.
(11) Haddad, R. I., and Shin, D. M. (2008) Recent advances in head
and neck cancer reconstruction. N. Engl. J. Med. 359, 11431154.
(12) Mapanao, A. K., Giannone, G., Summa, M., Ermini, M. L.,
Zamborlin, A., Santi, M., Cassano, D., Bertorelli, R., and Voliani, V.
(2020) Biokinetics and clearance of inhaled gold ultrasmall-in-nano
architectures. Nanoscale Adv. 2, 38153820.
(13) Cassano, D., Mapanao, A.-K., Summa, M., Vlamidis, Y.,
Giannone, G., Santi, M., Guzzolino, E., Pitto, L., Poliseno, L.,
Bertorelli, R., and Voliani, V. (2019) Biosafety and biokinetics of
noble metals: the impact of their chemical nature. ACS Appl. Bio
Mater. 2 (10), 44644470.
(14) Cassano, D., Summa, M., Pocoví-Martínez, S., Mapanao, A.-K.,
Catelani, T., Bertorelli, R., and Voliani, V. (2019) Biodegradable
ultrasmall-in-nano gold architectures: mid-period in vivo biodistribu-
tion and excretion assessment. Part. Part. Syst. Charact. 36 (2),
1800464.
(15) European Parliament. Directive 2010/63/EU - On the
protection of animals used for scientic purposes. O. J. Eur.
Communities: Legis. 2010, 276,3379.
(16) Mapanao, A. K., and Voliani, V. (2020) Three-dimensional
tumor models: Promoting breakthroughs in nanotheranostics trans-
lational research. Appl. Mater. Today. 19, 100552.
(17) Ribatti, D. (2016) The chick embryo chorioallantoic membrane
(CAM). A multifaceted experimental model. Mech. Dev. 141,7077.
(18) Santi, M., Mapanao, A. K., Cappello, V., and Voliani, V. (2020)
Production of 3D tumor models of head and neck squamous cell
carcinomas for nanotheranostics assessment. ACS Biomater. Sci. Eng. 6
(9), 48624869.
(19) Mapanao, A. K., Che, P. P., Sarogni, P., Sminia, P., Giovannetti,
E., and Voliani, V. (2021) Tumor grafted - chick chorioallantoic
membrane as an alternative model for biological cancer research and
conventional/nanomaterial-based theranostics evaluation. Expert
Opin. Drug Metab. Toxicol. 00 (00), 122.
(20) Kunz, P., Schenker, A., Sähr, H., Lehner, B., and Fellenberg, J.
(2019) Optimization of the chicken chorioallantoic membrane assay
as reliable in vivo model for the analysis of osteosarcoma. PLoS One
14 (4), e0215312.
(21) Hagedorn, M., Javerzat, S., Gilges, D., et al. (2005) Accessing
key steps of human tumor progression in vivo by using an avian
embryo model. Proc. Natl. Acad. Sci. U. S. A. 102 (5), 16431648.
(22) Rovithi, M., Avan, A., Funel, N., Leon, L. G., Gomez, V. E.,
Wurdinger, T., Griffioen, A. W., Verheul, H. M. W., and Giovannetti,
E. (2017) Development of bioluminescent chick chorioallantoic
membrane (CAM) models for primary pancreatic cancer cells: A
platform for drug testing. Sci. Rep. 7, 44686.
(23) Cecilia Subauste, M., Kupriyanova, T. A., Conn, E. M., Ardi, V.
C., Quigley, J. P., and Deryugina, E. I. (2009) Evaluation of metastatic
and angiogenic potentials of human colon carcinoma cells in chick
embryo model systems. Clin. Exp. Metastasis 26 (8), 10331047.
(24) Crespo, P., and Casar, B. (2016) The Chick Embryo
Chorioallantoic Membrane as an in vivo Model to Study Metastasis.
Bio-Protocol. 6 (20), 111.
(25) Sys, G. M. L., Lapeire, L., Stevens, N., et al. (2013) The in ovo
CAM-assay as a xenograft model for sarcoma. J. Visualized Exp.
No. 77, 17.
(26) Kleibeuker, E. A., Schulkens, I. A. E., Castricum, K. C. M.,
Griffioen, A. W., and Thijssen, V. L. J. L. (2015) Examination of the
Role of Galectins During In Vivo Angiogenesis Using the Chick
Chorioallantoic Membrane Assay. Methods Mol. Biol. 1207, 305315.
(27) Kunz et al. demonstrated that the application of 70% ethanol or
other disinfectants on the shell considerably reduces the viability of
the embryo by approximately 30%, thereby aecting the overall
execution of the experiment.
20
(28) The tilting racks are usually provided upon the purchase of the
incubator. They have a metal pin that ts into a small hole in the back
of the incubator, hence allowing the automatic movement. If no tilting
racks are available, then the trays can be manually moved through
180°at least 2 or 3 times per day.
(29) Once all the eggs are inside and incubation starts, it is
recommend to avoid frequent opening of the incubator in order to
keep the temperature and humidity constantly on the set parameter
and not interfere with the growth of the embryo. The percentage of
the unfertilized eggs may interfere with the nal outcome of the
experiment, and for this reason, it is strongly recommended to start
with a large number of eggs to obtain consistent result, e.g., 10% more
than expected eggs for the experiment. It should also take in
consideration that the fertilization rate of the eggs can be seasonal
depending on the supplier.
(30) Placing a bright light source on top/behind the egg
(candling) allows you to see through the shell. The air chamber
should be located in the blunt end of the egg. The incubated eggs can
be also candled to check for fertilization. While the fertilization of
white eggs can be checked at the third day of incubation, the brown-
shelled eggs may require a few more days to check this parameter
(Figure 2B). The presence of vessels is an indication of fertilized egg.
(31) The small window should be carefully opened to avoid shell
debris falling onto the membrane. If this happens, then try to carefully
remove the debris with sterile tweezers, without damaging the
membrane, to prevent infections or microorganism contamination.
(32) SCC-25 cells are maintained in a complete growth medium
composed of a 1:1 mixture of Dulbeccos modied Eagles medium
and Hams F12 medium supplemented with 10% of fetal bovine
serum (FBS), 4 mM L-glutamine, 1mM sodium pyruvate, 100 U/mL
penicillin, and 100 mg/mL streptomycin. The medium is also
supplemented with 400 ng/mL hydrocortisone. It should be noted
that SCC-25 is a very adherent cell line. Thus, a 5 min incubation in
trypsin may not be sucient to completely detach the cells from the
culture dish. After the medium is removed, it is recommended to rst
wash the cells with PBS and, then, with a small amount of trypsin.
After discarding, add trypsin again and incubate the cells for 510
min to harvest a high and consistent number of cells. Knowing the
doubling time of your cells may be useful for setting up the
experimental design. For SCC-25, the doubling time was observed to
be approximately 45 hours. It is also suggested to use 150 mm plates
to promote an extensive cell growth and to lower the frequency of
splitting the cells.
(33) The nal cell number per egg was empirically determined. It
has been observed that 2 ×106cells for SCC-25 is the minimum
amount needed to obtain solid tumor formation by EDD 10 (i.e., 4
days post-grafting and assigned EDD in which treatments are initially
administered).
(34) Matrigel is a protein mixture that resembles the extracellular
environment required to have a conducive substrate for cell growth.
Moreover, it is a dense mixture and keeps the cells together once
deposited on the membrane. The Matrigel solution is stored at
20°C. It needs to be thawed overnight at 4°C before the grafting
ACS Pharmacology & Translational Science pubs.acs.org/ptsci Article
https://doi.org/10.1021/acsptsci.1c00083
ACS Pharmacol. Transl. Sci. XXXX, XXX, XXXXXX
G
experiment. It is recommended to prepare Matrigel aliquots to keep
the stocks in proper storage temperature and avoid repeated freeze
thaw cycles. While working with Matrigel, it should be kept constantly
on ice to prevent polymerization while preparing the working solution
(12.5μL Matrigel + 12.5μL medium without FBS per egg) or
dispensing the cells on the membrane. Importantly, the tumor grafting
eciency at EDD 10 is only 20% if the SCC-25 cells deposited on the
CAM are suspended in the medium alone.
(35) The veins and arteries of the CAM are easily distinguished from
each other by the dierent blood color and vessel motility.
41
The
arteries are darker, and their size is larger compared to the veins. Also,
the movement of the arteries is more active, and their walls are
thicker, making them dicult to rupture during cell grafting.
(36) It usually takes 4 days post-grafting for the SCC-25 cells to
grow and form a solid visible tumor on the CAM. This timeframe may
change for other cell lines. It is recommended to have at least 89
eggs per treatment condition in order to have consistent statistics as
well as to employ at least 3 eggs as a control group without any
treatment.
(37) The treatment solution must be freshly prepared just before its
application. It has been observed that 30 μL of solution is enough to
adequately cover the tumor mass. Do not touch the tumor with the
tip while applying the treatment in order to avoid tissue damage.
(38) The tumor measurements from EDD10 to EDD14 are taken
from a supercial angle. Indeed, their volume is calculated by
considering the width and length of the visible tumor estimated from
the top of the CAM. It should be considered that some neoplasms
may grow below or above the membrane. Image acquisition can be
time consuming, especially when handling a lot of samples. It is
recommended to always keep the same magnication for all the
collected images. Any magnication changes require a software
recalibration. The light and embryo movements may interfere with
the acquisition of good quality images. It is also important to adjust
the light and polarization of the microscope to remove as much light
reection as possible on the membrane in order to collect high-quality
images. Eggs should be taken out from the incubator in small batches
during visual analysis to prevent the frequent opening and closing of
the incubator, which can aect the settings.
(39) Considering that the appearance of the membranes and the
tumors can change after 2 hours at 4 °C due to the wrinkling of the
CAM, the visualization of the tumor can be dicult. In this regard,
the images from previous EDDs may be helpful to locate the position
of the tumor. While harvesting the tumor, cut as little of the
membrane as possible since it might interfere with the weight of the
tumor and other downstream assays such as RNA extraction and
analysis of gene expression prole, protein extraction and detection of
tumor marker, and qualitative information about the structure of the
tissue.
(40) Livak, K. J., and Schmittgen, T. D. (2001) Analysis of relative
gene expression data using real-time quantitative PCR and the 2-
ΔΔCT method. Methods 25 (4), 402408.
(41) Nam, K. H., Kim, J., Ra, G., Lee, C. H., and Paeng, D. G.
(2015) Feasibility study of Ex Ovo chick chorioallantoic artery model
for investigating pulsatile variation of arterial geometry. PLoS One 10
(12), e0145969.
(42) Deryugina, E. (2016) Chorioallantoic Membrane Microtumor
Model to Study the Mechanisms of Tumor Angiogenesis, Vascular
Permeability, and Tumor Cell Intravasation. Methods Mol. Biol. 1430,
283298.
(43) Handy, B. (2009) The clinical utility of tumor markers. Lab.
Med. 40 (2), 99103.
(44) Shan, L., Hao, Y., Wang, S., Korotcov, A., Zhang, R., Wang, T.,
Califano, J., Gu, X., Sridhar, R., Bhujwalla, Z. M., and Wang, P. C.
(2008) Visualizing head and neck tumors in vivo using near-infrared
fluorescent transferrin conjugate. Mol. Imaging 7 (1), 4249.
(45) Högemann-Savellano, D., Bos, E., Blondet, C., Sato, F., Abe, T.,
Josephson, L., Weissleder, R., Gaudet, J., Sgroi, D., Peters, P. J., and
Basilion, J. P. (2003) The Transferrin Receptor: A Potential
Molecular Imaging Marker for Human Cancer. Neoplasia 5 (6),
495506.
(46) Feldman, A. T., and Wolfe, W. (2014) Tissue Processing and
Hematoxylin and Eosin Staining. Methods Mol. Biol. 1180, 283291.
(47) Neufeld, G., and Kessler, O. (2006) Pro-angiogenic cytokines
and their role in tumor angiogenesis. Cancer Metastasis Rev. 25 (3),
373385.
(48) McDermott, J. D., and Bowles, D. W. (2019) Epidemiology of
Head and Neck Squamous Cell Carcinomas: Impact on Staging and
Prevention Strategies. Curr. Treat Options Oncol. 20 (5), 113.
(49) DeBord, L. C., Pathak, R. R., Villaneuva, M., Liu, H.-C.,
Harrington, D. A., Yu, W., Lewis, M. T., and Sikora, A. G. (2018) The
chick chorioallantoic membrane (CAM) as a versatile patient-derived
xenograft (PDX) platform for precision medicine and preclinical
research. Am. J. Cancer Res. 8 (8), 16421660.
(50) Vlamidis, Y., and Voliani, V. (2018) Bringing again noble metal
nanoparticles to the forefront of cancer therapy. Front. Bioeng.
Biotechnol. 6, 143.
ACS Pharmacology & Translational Science pubs.acs.org/ptsci Article
https://doi.org/10.1021/acsptsci.1c00083
ACS Pharmacol. Transl. Sci. XXXX, XXX, XXXXXX
H
... The highly vascularized CAM supports the fast composition of standardized alternative in vivo tumour models, as previously demonstrated with both HNSCCs and pancreatic ductal adenocarcinoma cells [27,29]. It should be noted that CAMs respect the 3Rs concept, in accordance with the European Parliament Directive 2010/63/EU [26]. Due to their intrinsic features, these biomodels perfectly suits for preclinical investigations in oncological research, high-throughput drug-screening, and metastasis evaluations [30]. ...
... At EDD 10, tumour-bearing embryos were randomized and divided into three groups of study: i) serum-free cell culture medium ± RT (control); ii) CisPt ± RT; iii) NAs ± RT ( Figure 1C). The chemo treatment (CT) was administered on EDD10 as already standardized elsewhere [26]. The applied dosage of cisplatin (6 μg) results from the clinically administered amount of the drug in human (100 mg/m 2 ) and the weight of chicken embryo at EDD10. ...
... SCC-25 cancer cell line (Human Squamous Cell Carcinoma, purchased from the American Type Culture Collection -ATCC) was cultured in Dulbecco's Modified Eagle Medium/Nutrient Mixture F12 (DMEM/F12; Gibco 11039-021) supplemented with 10% foetal bovine serum (FBS), 4 mM L-glutamine, 1 mM sodium pyruvate, 100 U/mL penicillin, 100 mg/mL streptomycin and 400 ng/mL of hydrocortisone. The cells were kept in a humidified incubator set at 37 °C and 5% CO2.CAM assay was performed following the step-by-step procedure reported in Sarogni et al.[26] Briefly, fertilized red Leghorn chicken eggs were incubated at 37 °C in a humidified incubator (FIEM). The start of incubation was assigned as ...
Article
Full-text available
Locally advanced head and neck squamous cell carcinoma (LA-HNSCC) is characterized by high rate of recurrence, resulting in a poor survival. Standard treatments are associated with significant toxicities that impact the patient’s quality of life, highlighting the urgent need for novel therapies to improve patient outcomes. On this regard, noble metal nanoparticles (NPs) are emerging as promising agents as both drug carriers and radiosensitizers. On the other hand, co-treatments based on NPs are still at the preclinical stage because of the associated metal-persistence. In this bioconvergence study, we introduce a novel strategy to exploit tumour chorioallantoic membrane models (CAMs) in radio-investigations within clinical equipment and evaluate the performance of non-persistent nanoarchitectures (NAs) in combination with radiotherapy with respect to the standard concurrent chemoradiotherapy for the treatment of HPV-negative HNSCCs. A comparable effect has been observed between the tested approaches, suggesting NAs as a potential platinum-free agent in concurrent chemoradiotherapy for HNSCCs. On a broader basis, our bioconvergence approach provides an advance for the translation of Pt-free radiosensitizer to the clinical practice, positively shifting the therapeutic vs. side effects equilibrium for the management of HNSCCs.
... The CAM is a highly vascularized extraembryonic membrane connected to the developing embryo through an easily accessible circulatory system (Harper et al., 2021;Nowak-Sliwinska et al., 2014). In comparison to other in vivo models, the CAM model presents several advantages: (a) reduced ethical issues, as the chick embryo does not perceive pain from the CAM tissue before the 17th day of incubation Sarogni et al., 2021), given that the nervous system in chick embryos does not develop until day 17 of ontogenesis (Mesas et al., 2024); (b) the small size of the egg embryo is relatively easy to handle; (c) the egg embryo is rich in nutrients; and (d) the egg does not possess a complete immune system at the used stages of the chick development, proving to be a cost-effective method when compared to immune-compromised animals. CAM models are also in agreement with the 3Rs concept: reduction, refinement, and Fig. 1. ...
... The optimized tumor grafting procedure with SCC25 cells on CAM model has been previously reported and discussed in Sarogni et al. 2 . Briefly, fertilized red Leghorn chicken eggs were immediately stored at 4 °C upon delivery. ...
Article
Full-text available
Oral malignancies continue to have severe morbidity with less than 50% long-term survival despite the advancement in the available therapies. There is a persisting demand for new approaches to establish more efficient strategies for their treatment. In this regard, the human topoisomerase II (topoII) enzyme is a validated chemotherapeutics target, as topoII regulates vital cellular processes such as DNA replication, transcription, recombination, and chromosome segregation in cells. TopoII inhibitors are currently used to treat some neoplasms such as breast and small cells lung carcinomas. Additionally, topoII inhibitors are under investigation for the treatment of other cancer types, including oral cancer. Here, we report the therapeutic effect of a tetrahydroquinazoline derivative (named ARN21934) that preferentially inhibits the alpha isoform of human topoII. The treatment efficacy of ARN21934 has been evaluated in 2D cell cultures, 3D in vitro systems, and in chick chorioallantoic membrane cancer models. Overall, this work paves the way for further preclinical developments of ARN21934 and possibly other topoII alpha inhibitors of this promising chemical class as a new chemotherapeutic approach for the treatment of oral neoplasms.
... All procedures were agreed with the 3Rs concept (the concept of reduction, refinement, and replacement of animal models). Since the chick embryo does not develop pain perception before the 17th day of incubation, no permission or approval from ethics committees is required [35]. ...
Article
Full-text available
Objectives Pre-vascularization of the collagen membranes with autologous platelet concentrates is a standard procedure in oral and maxillofacial surgery. This study analyzed the possible interaction of an acellular collagen membrane of porcine origin (NM) with platelet-rich fibrin (PRF) regarding its rehydration protocol with differences in pH values and effect on angiogenesis. Materials and methods NM was analyzed alone and combined with solid PRF by plotting or co-culturing with injectable PRF. Different media (venous blood, buffer solution with a fixed pH value of 7, saline solution, and injectable PRF) were used to analyze the influence on pH value during rehydration. Chorion allantois membrane assay (CAM) was applied to check pro-angiogenic effects after 24, 48, and 72 h, followed by immunohistochemical analysis. Results Rehydration in injectable PRF showed acidity over time (p < 0.05). A definite pro-angiogenic effect of NM alone was found regarding neo-vessel formation supported by the respective light microscopically analysis without significant differences to PRF alone (p > 0.005). This pro-angiogenic effect could not be exaggerated when NM was combined with liquid/solid PRF (each p > 0.005). Conclusions Rehydration with liquid PRF of the collagen membrane results in acidity compared to a saline solution or patient’s blood. The significant pro-angiogenic potential of the membrane alone resulted in enhanced neo-vessel formation that could not be optimized with the addition of PRF. Clinical relevance statement Using injectable PRF for rehydration protocol of the collagen membrane leads to acidosis that can ultimately optimize wound healing. Differences in the physio-mechanical interplay of collagen matrices and autologous platelet concentrates must result in clinical algorithms if pre-vascularization can maximize outcomes.
Article
Full-text available
Pancreatic cancer has a poor prognosis due to its aggressive nature and ability to metastasize at an early stage. Currently, its management is still a challenge because this neoplasm is resistant to conventional treatment approaches, among which is chemo‐radiotherapy (CRT), due to the abundant stromal compartment involved in the mechanism of hypoxia. Hyperthermia, among other effects, counteracts hypoxia by promoting blood perfusion and thereby can enhance the therapeutic effect of radiotherapy (RT). Therefore, the establishment of integrated treatments would be a promising strategy for the management of pancreatic carcinoma. Here, the effects of joint radiotherapy/hyperthermia (RT/HT) on optimized chick embryo chorioallantoic membrane (CAM) pancreatic tumor models are investigated. This model enables a thorough assessment of the tumor‐arresting effect of the combined approach as well as the quantitative evaluation of hypoxia and cell cycle‐associated mechanisms by both gene expression analysis and histology. The analysis of the lower CAM allows to investigate the variation of the metastatic behaviors of the cancer cells associated with the treatments. Overall, this study provides a potentially effective combined strategy for the non‐invasive management of pancreatic carcinoma.
Article
Full-text available
Head and neck squamous cell carcinomas (HNSCCs) are a complex group of malignancies that affect different body sites pertaining to the oral cavity, pharynx and larynx. Current chemotherapy relies on platinum complexes, the major exponent being cisplatin, which exert severe side effects that can negatively affect prognosis. For this reason, other metal complexes with less severe side effects are being investigated as alternatives or adjuvants to platinum complexes. In this context, exploiting (supra)additive effects by the concurrent administration of cisplatin and emerging metal complexes is a promising research strategy that may lead to effective cancer management with reduced adverse reactions. Here, the combined action of cisplatin and a ruthenium(II) η6-arene compound (RuCy), both as free molecules and loaded into hybrid nano-architectures (NAs), has been assessed on HPV-negative HNSCC models of increasing complexity: 2D cell cultures, 3D multicellular tumor spheroids, and chorioallantoic membranes (CAMs). Two new NAs have been established to explore all the delivery combinations and compare their ability to enhance the efficacy of cisplatin in the treatment of HNSCCs. A significant supra-additive effect has been observed in both 2D and 3D models by one combination of treatments, suggesting that cisplatin is particularly effective when loaded on NAs, whereas RuCy performs better when administered as a free compound. Overall, this work paves the way for the establishment of the next co-chemotherapeutic approaches for the management of HNSCCs.
Article
Angiogenesis and metastasis are two interdependent cancer hallmarks, the latter of which is the key cause of treatment failure. Thus, establishing effective antiangiogenesis/antimetastasis agents is the final frontier in cancer research. Gold nanoparticles (GNPs) may provide disruptive advancements in this regard due to their intrinsic physical and physiological features. Here, we comprehensively discuss recent potential therapeutical strategies to treat angiogenesis and metastasis and present a critical review on the state-of-the-art in vitro and in vivo evaluations of the antiangiogenic/antimetastatic activity of GNPs. Finally, we provide perspectives on the contribution of GNPs to the advancement of cancer management.
Article
Full-text available
The selective and localized delivery of active agents to neoplasms is crucial to enhance the chemotherapeutic efficacy while reducing the associated side effects. The encapsulation of chemotherapeutics in nanoparticles decorated with targeting agents is a strategy of special interest to improve drug delivery. However, serum protein adsorption often compromises the in vivo efficiency of targeting agents, leading to protein corona formation that interferes with the targeting process. Here, the enhanced efficacy of hybrid nano-architectures enclosing a platinum prodrug and decorated with a customized peptide (NAs-cisPt-Tf2) is demonstrated by employing alternative in vivo models of oral carcinoma. The peptide binds to transferrin and modulates the protein corona formation on NAs-cisPt-Tf2, supporting the identification of its receptor. Optimized chorioallantoic membrane cancer models (CAMs) enabled a thorough assessment of the tumor-suppressing effect of NAs-cisPt-Tf2 as well as the quantitative evaluation of angiogenesis and cell cycle associated mechanisms. The treatment strategy resulted in a significant tumor volume reduction coupled with anti-angiogenic and pro-apoptotic effects inferred from the downregulation of the vascular endothelial growth factor gene and increased expression of cleaved caspase-3. Overall, this study provides a potentially effective tumor-targeted approach for a non-invasive management of oral carcinoma.
Article
Full-text available
Introduction: Advancements in cancer management and treatment are associated with strong preclinical research data, in which reliable cancer models are demanded. Indeed, inconsistent preclinical findings and stringent regulations following the 3Rs principle of reduction, refinement, and replacement of conventional animal models currently pose challenges in the development and translation of efficient technologies. The chick embryo chorioallantoic membrane (CAM) is a system for the evaluation of treatment effects on the vasculature, therefore suitable for studies on angiogenesis. Apart from vascular effects, the model is now increasingly employed as a preclinical cancer model following tumor-grafting procedures. Areas covered: The broad application of CAM tumor model is highlighted along with the methods for analyzing the neoplasm and vascular system. The presented and cited investigations focus on cancer biology and treatment, encompassing both conventional and emerging nanomaterial-based modalities. Expert opinion: The CAM tumor model finds increased significance given the influences of angiogenesis and the tumor microenvironment in cancer behavior, then providing a qualified miniature system for oncological research. Ultimately, the establishment and increased employment of such a model may resolve some of the limitations present in the standard preclinical tumor models, thereby redefining the preclinical research workflow.
Article
Full-text available
Among the organism’s entry portals, the respiratory tract is one of the most promising routes for a non-invasive administration of therapeutics for local and systemic delivery. On the other hand, it is the subtlest to preserve from environment pollution and microbial occurrences. Here, the biokinetics, distribution, and clearance trends of gold ultrasmall-in-nano architectures administered through a single intranasal application have been quantitatively evaluated. Aside from reaching the lung parenchyma, the (bio)degradable nano-architectures are able to translocate as well to secondary organs and to be almost completely excreted within 10 days. These findings further promote the clinical relevance of plasmonic nanomaterials for oncology and infectious disease treatment and management. Noticeably, this investigation also provides crucial information regarding the associated risks consequential to the pulmonary delivery of nanoparticles.
Article
Full-text available
As a first approach, standard 2D cell culture techniques are usually employed for the screening of drugs and nanomaterials. Despite the easy -handling, findings achieved on 2D cultures are often not efficiently translatable to in vivo preclinical investigations. Furthermore, although animal models are pivotal in preclinical studies, more strict directives have been implemented to promote the use of alternative biological systems. In this context, the development and the integration into the preclinical research workflow of 3D neoplasms models is particularly appealing to promote the advancement and success of therapeutics in clinical trials while reducing the number of in vivo models. Indeed, 3D tumor models bridge several discrepancies between 2D cell culture and in vivo models, among which are morphology, polarity, drug penetration, osmolality, and gene expressions. Here, we comprehensively describe a robust and high-throughput hanging drop protocol for the production of 3D models of both Human Papillomavirus (HPV)-positive and HPV-negative head and neck squamous cell carcinomas (HNSCCs). We also report the standard cascade assays for their characterization and demonstrate their significance in investigations on these aggressive neoplasms. The employment of relevant 3D cancer models is pivotal to produce more reliable and robust findings in terms of biosafety, theranostic efficacy, and biokinetics as well as to promote further knowledge on HNSCCs pathophysiology.
Article
Full-text available
Negative or positive HPV-associated Head and Neck Squamous Cell Carcinomas (HNSCCs) are high recurrence neoplasms usually resulting in a poor prognosis, mainly due to metastasis formation. Despite the low overall patient survival rate and the severe side effects, the treatment of choice is still cisplatin-based chemotherapy. Here, we report a straightforward protocol for the production of high throughput 3D models of negative or positive HPV-associated HNSCCs, together with their employment in the therapeutic evaluation of gold ultrasmall-in-nano architectures comprising an endogenously-activatable cisplatin prodrug. Beyond enhancing the biosafety of cisplatin, our approach paves the way for the establishment of synergistic co-therapies for HNSCCs based on excretable noble metals.
Article
Full-text available
The chemokine receptor CXCR7 has been suggested to play important roles in the progression of several types of cancers. However, few studies have investigated the biological roles of CXCR7 in head and neck squamous cell carcinoma (HNSCC). CXCR7 expression and its clinical implications were examined in 103 HNSCC tissues using immunohistochemistry (IHC). The biological roles and mechanisms of CXCR7-mediated signaling pathways were investigated in HNSCC cells through CXCR7 overexpression in vitro and in vivo. High expression of CXCR7 was significantly associated with tumor size (P = 0.007), lymph node metastasis (P = 0.004), and stage (P = 0.020) in HNSCC. Overexpression of CXCR7 in HNSCC cells enhanced cell migration and invasion in vitro and promoted lymph node metastasis in vivo. CXCR7 also induced epithelial–mesenchymal transition through PI3K/AKT. CXCR7 increased secretion of transforming growth factor-β1 (TGF-β1) and promoted EMT through phosphorylated Smad2/3. Taken together, our results provide functional and mechanistic roles of CXCR7 as a master regulator of oncogenic TGF-β1/Smad2/3 signaling in HNSCC, suggesting that CXCR7 might be a therapeutic target for the treatment of HNSCC.
Article
Current chemotherapy for head and neck squamous cell carcinomas (HNSCCs) are based on cisplatin, which is usually associated to severe side effects. In general, the exploration for metal-based alternatives to cisplatin has resulted in the development of a series of ruthenium complexes that are able to produce a safe therapeutic action against some neoplasms, among which are lung and ovarian cancers. Here, we evaluate the efficacy of well defined, easily available and robust ruthenium(II) η⁶-arene compounds on 3D models of HNSCCs with or without human papillomavirus (HPV) infection and compare their effects to the state-of-the-art RAPTA-C, a promising ruthenium compound with known anti-cancer activity. One of the compounds induces a significant therapeutic action especially on HPV negative carcinoma. Besides viability and repopulation evaluations, we performed quantitative analysis of the internalized Ru compounds to further validate our findings and elucidate the possible mechanisms of action. These results show that Ru arene compounds represent a promising alternative for the treatment of HNSCCs and pave the way for the composition of innovative (co)therapies.
Article
Synergistic combined treatments are currently practiced in clinics for the management of several neoplasms. While surgery, radiotherapy, and chemotherapy remain as the standards of care for monomodal and co-treatments, emerging modalities like hyperthermia (HT) demonstrate promising features as (neo)adjuvant, particularly for recurrent cancers. However, the clinical relevance of HT is still debated due to a number of challenges, such as tumor specific temperature increase, uneven heating of the target, and the lack of agents that concurrently execute HT in combination with radio- and/or chemotherapy. Here, the application of non-persistent ultrasmall-in-nano gold architectures for synergistic chemo-photothermal treatment of head and neck squamous cell carcinomas (HNSCCs) is presented. The nano-architectures are composed of excretable narrow near-infrared (NIR)-absorbing gold ultrasmall nanoparticles and an endogenously double controlled cisplatin prodrug. The efficiency of the nano-architectures is evaluated on three-dimensional (3D) models of HNSCCs with positive or negative human papillomavirus (HPV) status. The combined treatment causes a more pronounced antitumor action on HPV-positive HNSCCs. Overall, the findings demonstrate the potential clinical relevance of translatable noble metal-based synergistic treatments in tumors management.
Article
Three-dimensional tumor models are becoming extensively employed in cancer research due to their accessibility and close representation of physiological tumor conditions. Hence, novel oncological diagnostic, therapeutic and combined theranostic platforms, including nanomaterial-based systems, are increasingly evaluated at preclinical stage by means of such models. Here, we provide an overview of the functional features and methods of preparation of some of the most interesting 3D tumor models, with a special attention to the classical and widely utilized multicellular tumor spheroids. Their applications in nanomaterial biosafety and efficacy assessment are highlighted, along with the suitable evaluating techniques. Overall, this review comprehensively illustrates the significant role of 3D tumor models towards the translation of nanomaterials to clinics. Their inclusion in biomedical and preclinical research workflow may address the issues related to both the conventional two-dimensional cell cultures and animal models, ultimately promoting breakthroughs in cancer nanotheranostics research.
Article
Effective excretion of nanostructured noble metals is still one of the most challenging bottle-neck for their employment in clinical practice. Beside the persistence issue, the clinical translation of inorganic nanomaterials is also affected by a bewildering lack of investigations regarding their quantitative biokinetics. Here, we have quantitatively correlated the chemical nature of the three most interesting noble metals for biomedical applications to their biosafety and biokinetics in, respectively, zebrafish and murine models. Gold, silver and platinum ultrasmall-in-nano architectures with comparable size elicit, after intravenous administration, different excretion pathways depending on their intrinsic metallic nature. Understanding the in vivo fate of noble metal nanoparticles is a significant breakthrough to unlock their clinical employment for the establishment of treatments for neoplasms, infectious diseases and neurological disorders.