ArticlePDF Available

A Myristoyl Amide Derivative of Doxycycline Potently Targets Cancer Stem Cells (CSCs) and Prevents Spontaneous Metastasis, Without Retaining Antibiotic Activity

Frontiers
Frontiers in Oncology
Authors:
  • Lunellabiotech Inc.

Abstract and Figures

Here, we describe the chemical synthesis and biological activity of a new Doxycycline derivative, designed specifically to more effectively target cancer stem cells (CSCs). In this analog, a myristic acid (14 carbon) moiety is covalently attached to the free amino group of 9-amino-Doxycycline. First, we determined the IC50 of Doxy-Myr using the 3D-mammosphere assay, to assess its ability to inhibit the anchorage-independent growth of breast CSCs, using MCF7 cells as a model system. Our results indicate that Doxy-Myr is >5-fold more potent than Doxycycline, as it appears to be better retained in cells, within a peri-nuclear membranous compartment. Moreover, Doxy-Myr did not affect the viability of the total MCF7 cancer cell population or normal fibroblasts grown as 2D-monolayers, showing remarkable selectivity for CSCs. Using both gram-negative and gram-positive bacterial strains, we also demonstrated that Doxy-Myr did not show antibiotic activity, against Escherichia coli and Staphylococcus aureus. Interestingly, other complementary Doxycycline amide derivatives, with longer (16 carbon; palmitic acid) or shorter (12 carbon; lauric acid) fatty acid chain lengths, were both less potent than Doxy-Myr for the targeting of CSCs. Finally, using MDA-MB-231 cells, we also demonstrate that Doxy-Myr has no appreciable effect on tumor growth, but potently inhibits tumor cell metastasis in vivo, with little or no toxicity. In summary, by using 9-amino-Doxycycline as a scaffold, here we have designed new chemical entities for their further development as anti-cancer agents. These compounds selectively target CSCs, e.g., Doxy-Myr, while effectively minimizing the risk of driving antibiotic resistance. Taken together, our current studies provide proof-of-principle, that existing FDA-approved drugs can be further modified and optimized, to successfully target the anchorage-independent growth of CSCs and to prevent the process of spontaneous tumor cell metastasis.
This content is subject to copyright.
ORIGINAL RESEARCH
published: 15 September 2020
doi: 10.3389/fonc.2020.01528
Frontiers in Oncology | www.frontiersin.org 1September 2020 | Volume 10 | Article 1528
Edited by:
Anna Sebestyén,
Semmelweis University, Hungary
Reviewed by:
Anna Maria Tokes,
Semmelweis University, Hungary
Chris Albanese,
Georgetown University, United States
*Correspondence:
Federica Sotgia
fsotgia@gmail.com
Michael P. Lisanti
michaelp.lisanti@gmail.com
Specialty section:
This article was submitted to
Cancer Metabolism,
a section of the journal
Frontiers in Oncology
Received: 23 May 2020
Accepted: 16 July 2020
Published: 15 September 2020
Citation:
Ózsvári B, Magalhães LG, Latimer J,
Kangasmetsa J, Sotgia F and
Lisanti MP (2020) A Myristoyl Amide
Derivative of Doxycycline Potently
Targets Cancer Stem Cells (CSCs)
and Prevents Spontaneous
Metastasis, Without Retaining
Antibiotic Activity.
Front. Oncol. 10:1528.
doi: 10.3389/fonc.2020.01528
A Myristoyl Amide Derivative of
Doxycycline Potently Targets Cancer
Stem Cells (CSCs) and Prevents
Spontaneous Metastasis, Without
Retaining Antibiotic Activity
Béla Ózsvári1, Luma G. Magalhães1, Joe Latimer2, Jussi Kangasmetsa3,
Federica Sotgia1,4*and Michael P. Lisanti1,4*
1Translational Medicine, School of Science, Engineering and Environment (SEE), University of Salford, Manchester,
United Kingdom, 2Salford Antibiotic Research Network, School of Science, Engineering and Environment (SEE), University of
Salford, Manchester, United Kingdom, 3Eurofins Integrated Discovery UK Ltd., Essex, United Kingdom, 4Lunella Biotech,
Inc., Ottawa, ON, Canada
Here, we describe the chemical synthesis and biological activity of a new Doxycycline
derivative, designed specifically to more effectively target cancer stem cells (CSCs).
In this analog, a myristic acid (14 carbon) moiety is covalently attached to the free
amino group of 9-amino-Doxycycline. First, we determined the IC50 of Doxy-Myr using
the 3D-mammosphere assay, to assess its ability to inhibit the anchorage-independent
growth of breast CSCs, using MCF7 cells as a model system. Our results indicate that
Doxy-Myr is >5-fold more potent than Doxycycline, as it appears to be better retained
in cells, within a peri-nuclear membranous compartment. Moreover, Doxy-Myr did not
affect the viability of the total MCF7 cancer cell population or normal fibroblasts grown
as 2D-monolayers, showing remarkable selectivity for CSCs. Using both gram-negative
and gram-positive bacterial strains, we also demonstrated that Doxy-Myr did not show
antibiotic activity, against Escherichia coli and Staphylococcus aureus. Interestingly,
other complementary Doxycycline amide derivatives, with longer (16 carbon; palmitic
acid) or shorter (12 carbon; lauric acid) fatty acid chain lengths, were both less potent
than Doxy-Myr for the targeting of CSCs. Finally, using MDA-MB-231 cells, we also
demonstrate that Doxy-Myr has no appreciable effect on tumor growth, but potently
inhibits tumor cell metastasis in vivo, with little or no toxicity. In summary, by using
9-amino-Doxycycline as a scaffold, here we have designed new chemical entities
for their further development as anti-cancer agents. These compounds selectively
target CSCs, e.g., Doxy-Myr, while effectively minimizing the risk of driving antibiotic
resistance. Taken together, our current studies provide proof-of-principle, that existing
FDA-approved drugs can be further modified and optimized, to successfully target the
anchorage-independent growth of CSCs and to prevent the process of spontaneous
tumor cell metastasis.
Keywords: cancer stem-like cells (CSCs), Doxycycline, myristic acid, fatty acylation, cancer cell metastasis,
prophylaxis of metastasis, 9-amino-Doxycycline, antimitoscins
Ózsvári et al. Doxycycline Derivatives for Metastasis Prevention
INTRODUCTION
Cancer stem cells (CSCs) are thought to be the root cause of
recurrence, metastasis, and drug-resistance, in a host of cancer
types (15). As a consequence, there is an unmet need to
develop new therapeutics to target and selectively kill CSCs,
while avoiding side effects, especially severe chemo-toxicity.
Metastasis is believed to be driven by this unique sub-population
of cancer-initiating cells (13). CSCs have the ability to generate
de novo tumors in immuno-deficient host organisms. Moreover,
they have the capacity to engage in anchorage-independent
growth, which facilitates their invasive spread throughout the
various tissues and organ systems, resulting in local, and distant
disseminated lesions (4,5). Remarkably, these metastatic lesions
are resistant to both chemo-therapy and radiation treatments.
Unfortunately, the Achilles’ heel of these pro-metastatic CSCs
remains largely unknown. As a consequence, currently there are
no anti-cancer drugs that are FDA-approved for the prevention
of metastasis.
Over the last 5 years, we identified that mitochondria in CSCs
may be a novel tractable therapeutic target, for inhibiting their
anchorage-independent growth (5). More specifically, increased
mitochondrial biogenesis may facilitate efficient high energy
production, resulting in the rapid propagation of CSCs (610).
In addition, within metastatic lymph nodes isolated from breast
cancer patients, disseminated cancer cells show elevated levels
of mitochondrial activity, especially Complex IV activation, as
revealed by functional activity assays (11).
Interestingly, mitochondrial biogenesis is critically linked
to the activity of mitochondrial ribosomal proteins, that
functionally translate key mitochondrial proteins, which are
genetically encoded by mitochondrial DNA (mt-DNA); this
includes 13 proteins that are necessary to functionally maintain
OXPHOS and mitochondrial ATP synthesis (613).
The reason that mitochondria have their own DNA and
specific machinery for protein translation is that they originally
evolved from engulfed aerobic bacteria, over the last 1.4
billion years, after invading eukaryotic cells. This evolutionary
symbiotic relationship has certain functional consequences
that could be safely exploited to achieve anti-mitochondrial
therapy, specifically targeting CSCs. For example, because of
the similarities between bacteria and mitochondria, a sub-
set of bacteriostatic antibiotics block mitochondrial protein
translation, as a manageable side effect (6,10,13). In this
context, Doxycycline inhibits the activity of the small mito-
ribosome, while Azithromycin blocks the large mito-ribosome,
both as off-target side effects (6,10,13). Similarly, Doxycycline
and Azithromycin both inhibit the propagation of CSCs, in an
anchorage-independent fashion, in numerous breast cancer cell
lines, as well as in cell lines derived from many other solid tumor
types (6,10,13). As such, we have suggested that these side-effects
could be re-purposed to clinically target and therapeutically
eradicate CSCs.
In direct support of this notion, a Phase II clinical trial
has documented that brief treatment with Doxycycline, in
early breast cancer patients, is indeed sufficient to significantly
decrease the content of CSCs in the tumor mass, by employing
CD44-staining as an established marker of CSCs in ER(+)
patients (14). Overall, the response rate approached 90%,
resulting in reductions of up to nearly 67% in CSC tumor burden
(14). Quantitatively similar results were obtained in HER2(+)
patients, using ALDH1 as a CSC marker. As such, blocking
mitochondrial protein synthesis may be a viable approach for
targeting and removing CSCs in vivo, prior to surgical excision,
possibly preventing the development of metastases.
Consistent with the above observations, other groups have
shown that Doxycycline treatment effectively reduces the
expression of a panel of CSC markers, in breast cancer cell
lines, such as CD44, ALDH, Oct4, Sox2, and Nanog (9,12).
Moreover, Doxycycline treatment functionally inhibits multiple
CSC signaling pathways, including Wnt, Notch, Hedgehog, and
STAT1/3-signaling (10).
Here, we describe a medicinal chemistry approach to design
novel therapeutics to more selectively target CSCs. More
specifically, we show that several new potent Doxycycline analogs
can be generated by attaching a fatty acid onto 9-amino-
Doxycycline, which is a relatively straightforward chemical
modification. Importantly, these analogs, such as Doxy-Myr, lack
antibiotic activity, but more potently target CSCs and effectively
prevent metastasis, in an in vivo pre-clinical model. Therefore,
Doxy-Myr could be used to eradicate CSCs, without exerting the
selective pressures required for the development of antimicrobial
resistance and without significant toxicity.
To better describe this general class of novel compounds
which are lipid-modified FDA-approved antibiotics, we propose
the term Antimitoscins, to specifically reflect their intrinsic anti-
mitochondrial activity.
MATERIALS AND METHODS
Materials
MCF7 and MDA-MB-231 cells were obtained from the American
Type Culture Collection (ATCC). hTERT-BJ1 fibroblasts were
as we previously described (13). Cells were cultured in DMEM,
supplemented with 10% fetal calf serum (FCS), Glutamine
and Pen/Strep.
Chemical Synthesis
Custom-chemical syntheses were performed by Eurofins
Integrated Discovery UK Ltd., (Essex, UK). Conventional
peptide synthesis methods were used to covalently attach each
free fatty acid to 9-amino-Doxycycline. The desired reaction
products were identified, chromatographically purified and the
chemical structures were validated, by using a combination
of NMR and mass spectrometry. The IUPAC names for the
chemical compounds are as follows.
Doxycycline
(4S,5S,6R,12aS)-4-(dimethylamino)-3,5,10,12,12a-
pentahydroxy-6-methyl-1,11-dioxo-4a,5,5a,6-tetrahydro-
4H-tetracene-2-carboxamide.
Doxycycline-Myr
(4S,5S,6R,12aS)-4-(dimethylamino)-3,5,10,12,12a-
pentahydroxy-6-methyl-1,11-dioxo-9-(tetradecanoylamino)-
4a,5,5a,6-tetrahydro-4H-tetracene-2-carboxamide.
Frontiers in Oncology | www.frontiersin.org 2September 2020 | Volume 10 | Article 1528
Ózsvári et al. Doxycycline Derivatives for Metastasis Prevention
Doxycycline-Laur
(4S,5S,6R,12aS)-4-(dimethylamino)-9-(dodecanoylamino)-
3,5,10,12,12a-pentahydroxy-6-methyl-1,11-dioxo-4a,5,5a,6-
tetrahydro-4H-tetracene-2-carboxamide.
Doxycycline-Pal
(4S,5S,6R,12aS)-4-(dimethylamino)-9-(hexadecanoylamino)-
3,5,10,12,12a-pentahydroxy-6-methyl-1,11-dioxo-4a,5,5a,6-
tetrahydro-4H-tetracene-2-carboxamide.
Doxycycline-TPP
[6-[[(5R,6S,7S,10aS)-9-carbamoyl-7-(dimethylamino)-
1,6,8,10a,11-pentahydroxy-5-methyl-10,12-dioxo-5a,6,6a,7-
tetrahydro-5H-tetracen-2-yl]amino]-6-oxo-hexyl]-triphenyl-
phosphonium oxalate.
9-Amino-Doxycycline
(4S,5S,6R,12aS)-9-amino-4-(dimethylamino)-3,5,10,12,12a-
pentahydroxy-6-methyl-1,11-dioxo-4a,5,5a,6-tetrahydro-4H-
tetracene-2-carboxamide.
9-Amino-Doxycycline was synthesized, essentially as
previously described (15).
See Supplemental Materials and Methods, and
Supplemental Figure 1, for further details on the chemical
synthesis of these Doxycycline derivatives.
3D-Mammosphere Assay
The mammosphere assay is considered as the gold-standard
for functionally measuring “stemness” and CSC propagation in
breast cancer cells. Single cells are first plated at low density
on low-attachment plates and >90% of “bulk” cancer cells die
under these conditions, in a process of programmed cell death,
termed anoikis. Only stem-like cells survive and propagate in
suspension. Each 3D mammosphere is formed from a single
CSC. Test compounds are added at the moment of single
cell plating and then the number of 3D mammospheres are
counted 5 days after plating. More specifically, a single cell
suspension of MCF7 cells was prepared using enzymatic (1x
Trypsin-EDTA, Sigma Aldrich) and manual disaggregation (25-
gauge needle) (1619). Cells were then plated at a density of
500 cells/cm2in mammosphere medium (DMEM-F12/B27/ EGF
(20-ng/ml)/PenStrep) in non-adherent conditions, in culture
dishes coated with (2-hydroxyethylmethacrylate) (poly-HEMA,
Sigma). Cells were then grown for 5 days and maintained in a
humidified incubator at 37C at an atmospheric pressure in 5%
(v/v) carbon dioxide/air. After 5 days in culture, spheres >50 µm
were counted using an eye-piece graticule, and the percentage of
cells plated which formed spheres was calculated and is referred
to as percent mammosphere formation, normalized to vehicle-
alone treated controls. Mammosphere assays were performed in
triplicate and repeated three times independently.
Fluorescence Imaging
Fluorescent images were taken after 72 h of incubation of MCF7
cells treated with either Doxycycline or Doxy-Myr (both at
10 µM), or vehicle control. Cell cultures were imaged with the
EVOS Cell Imaging System (Thermo Fisher Scientific, Inc.),
using the GFP channel. No fluorescent dye was used before
imaging, therefore, any changes in signal were exclusively due to
the auto-fluorescent nature of the Doxycycline compounds.
Cell Viability Assay
The Sulphorhodamine (SRB) assay is based on the measurement
of cellular protein content (1619). After treatment for 72 h
in 96-well plates (8,000 cells/well), cells were fixed with 10%
trichloroacetic acid (TCA) for 1 h in the cold room, and were
dried overnight at room temperature. Then, cells were incubated
with SRB for 15 min, washed twice with 1% acetic acid, and air
dried for at least 1 h. Finally, the protein-bound dye was dissolved
in a 10 mM Tris, pH 8.8, solution and read using the plate reader
at 540-nm.
Cell Proliferation
Briefly, MCF7 cells were seeded in each well (10,000 cells/well)
and employed to assess the efficacy of Doxycycline and Doxy-
Myr, using RTCA (real-time cell analysis), via the measurement
of cell-induced electrical impedance plate (Acea Biosciences Inc.)
(18). This approach allows the quantification of the onset and
kinetics of the cellular response. Experiments were repeated
several times independently, using quadruplicate samples for
each condition.
Cell Cycle Analysis
We performed cell-cycle analysis on MCF7 cells treated
with Doxycycline, Doxy-Myr, or vehicle-alone. Briefly, after
trypsinization, the re-suspended cells were incubated with
10 ng/ml of Hoechst solution (Thermo Fisher Scientific)
for 40 min at 37C under dark conditions. Following a
40 min period, the cells were washed and re-suspended
in PBS Ca/Mg for acquisition on the Attune NxT flow
cytometer (Thermo Scientific). We analyzed 10,000 events
per condition. Gated cells were manually-categorized into
cell-cycle stages (19).
Bacterial Growth Assays
Briefly, antibiotic activity was assessed using standard assay
systems (2022). The antibiotic activity of Doxycycline analogs
was determined experimentally, using Resazurin (R7017; Sigma-
Aldrich, Inc.) as an indicator of bacterial metabolism/vitality, in
a 96-well plate format, using Escherichia coli and Staphylococcus
aureus. The minimum inhibitory concentration (MIC) for
the studied compounds was determined using the broth
microdilution method, the reference susceptibility test for rapidly
growing aerobic or facultative microorganism(s). The assays
were performed according to the Clinical and Laboratory
Standards Institute (CLSI) guidelines. The test compounds
and positive control (doxycycline, Sigma Aldrich #D1822)
stock solutions were prepared at 25 mM in DMSO and
serially diluted (2-fold dilution from 200 to 1.56 µM) in
cation adjusted Mueller Hinton Broth (MHB, Sigma Aldrich
#90922) in 96-well transparent plates (VWR #734-2781) into
a final volume of 50 µL/well. Staphylococcus aureus (ATCC
29213) and E. coli (ATCC 25922) cultures were maintained
on Mueller Hinton Agar (MHA, Sigma Aldrich #70191). A
single colony of each strain was then grown overnight at 37
Frontiers in Oncology | www.frontiersin.org 3September 2020 | Volume 10 | Article 1528
Ózsvári et al. Doxycycline Derivatives for Metastasis Prevention
C in MHB until OD600 0.6–0.8 and diluted into MHB to
a concentration of 106 colony forming units (CFU)/mL, which
was equivalent to an OD600 0.01. Diluted inocula (50 µL)
were transferred to the wells of the previously prepared 96-
well plates containing the test compounds, negative control
(1% DMSO in MHB) and positive control (Doxycycline).
Final wells volume was 100 µL, final concentrations for
the testing compounds were between 100 and 0.78 µM and
final microorganism concentration was 5 ×105CFU/mL.
Subsequently, 10 µL of one negative control well was plated
in a petri dish containing MHA to check CFU and the
purity of the cultures. The plates were incubated at 37C for
24 h after which 20 µL of resazurin solution (0.2 mg/mL)
was added to the wells followed by 1 h 30 min incubation at
37C. The OD570 and OD600 were measured in a microplate
reader (BMG FLUOstar Omega). The ratio between OD570
and OD600 was determined and the MIC represents the lowest
concentration of compound that inhibited bacterial growth
(OD570/OD600 ratio inferior to the average ratio determined for
negative control wells). MIC values were determined by three
independent experiments.
Assays for Tumor Growth, Metastasis, and
Embryo Toxicity
These xenograft assays were carried out, essentially as previously
described, without any major modifications (2327).
Preparation of Chicken Embryos
Fertilized White Leghorn eggs were incubated at 37.5C with
50% relative humidity for 9 days. At that moment (E9), the
chorioallantoic membrane (CAM) was dropped down by drilling
a small hole through the eggshell into the air sac, and a 1 cm2
window was cut in the eggshell above the CAM.
Amplification and Grafting of Tumor Cells
The MDA-MB-231 tumor cell line was cultivated in
DMEM medium supplemented with 10% FBS and 1%
penicillin/streptomycin. On day E9, cells were detached
with trypsin, washed with complete medium and suspended in
graft medium. An inoculum of 1 ×106cells was added onto
the CAM of each egg (E9) and then eggs were randomized
into groups.
Tumor Growth Assays
At day 18 (E18), the upper portion of the CAM was removed
from each egg, washed in PBS and then directly transferred to
paraformaldehyde (fixation for 48 h) and weighed. For tumor
growth assays, at least 10 tumor samples were collected and
analyzed per group (n10).
Metastasis Assays
On day E18, a 1 cm2portion of the lower CAM was collected to
evaluate the number of metastatic cells in 8 samples per group (n
=8). Genomic DNA was extracted from the CAM (commercial
kit) and analyzed by qPCR with specific primers for Human
Alu sequences. Calculation of Cq for each sample, mean Cq,
and relative amounts of metastases for each group are directly
managed by the Bio-Rad R
CFX Maestro software. A one-way
ANOVA analysis with post-tests was performed on all the data.
Embryo Tolerability Assay
Before each administration, the treatment tolerability was
evaluated by scoring the number of dead embryos.
Statistical Analysis
Statistical significance was determined using the Student’s t-test,
values of <0.05 were considered significant. Data are shown as
the mean ±SEM, unless stated otherwise. Also, ANOVA was
conducted, where appropriate.
RESULTS
Generating New Analogs of Doxycycline
for Targeting CSCs: Doxy-Myr and
Doxy-TPP
Doxycycline is known to function as an inhibitor of the
propagation of CSCs, through its ability to inhibit the small
mitochondrial ribosome, which is an off-target side-effect
(6,10,13). Normally, Doxycycline is used as a broad-
spectrum bacteriostatic antibiotic to treat a wide range
of infections caused by gram-negative and gram-positive
bacteria. Therefore, we sought to optimize the ability of
Doxycycline for the targeting of CSCs, while minimizing its
antibiotic activity, to derive a new chemical entity to selectively
target CSCs.
As a first step, we synthesized 9-amino-Doxycycline, to which
we covalently attached either a 14 carbon fatty acid moiety
(myristic acid) or a six carbon spacer arm containing tri-phenyl-
phosphonium (TPP). The chemical structures of these two
Doxycycline analogs, as well as the parent compound, are all
shown in Figure 1. We speculated that the TPP-moiety would
better target Doxy-TPP to mitochondria in CSCs. In contrast,
the addition of myristic acid could act as a membrane targeting
signal, possibly leading to the increased retention of Doxy-
Myr within membranous compartments, such as the plasma
membrane, the endoplasmic reticulum (ER), the Golgi apparatus,
and/or mitochondria.
To determine the functional activity of these Doxycycline
analogs, we used the 3D-mammosphere assay, to assess their
ability to inhibit the 3D anchorage-independent propagation of
MCF7 CSCs. Interestingly, Doxy-TPP was not more potent that
Doxycycline itself, so further assays with Doxy-TPP were not
carried out (data not shown).
Figure 2 shows a direct comparison of Doxycycline with
Doxy-Myr. Remarkably, Doxy-Myr was >5-fold more potent
than Doxycycline, with an IC50 of 3.46 µM. In contrast,
Doxycycline had an IC50 of 18.1 µM. Therefore, Doxy-Myr
is more potent for targeting the 3D anchorage-independent
propagation of CSCs.
To experimentally test the hypothesis that Doxy-Myr was
better retained within cells, we took advantage of the observation
that Doxycycline is fluorescent (Ex. 390–425 nm/Em. 520–
560 nm). Doxy-Myr was more easily detected and retained in
monolayer MCF7 cells, when compared to Doxycycline or cells
Frontiers in Oncology | www.frontiersin.org 4September 2020 | Volume 10 | Article 1528
Ózsvári et al. Doxycycline Derivatives for Metastasis Prevention
FIGURE 1 | Chemical structures of two new Doxycycline derivatives: Doxy-Myr and Doxy-TPP. Note that Doxy-Myr contains a 14-carbon fatty acid (myristate)
covalently attached to 9-amino-Doxycycline and Doxy-TPP contains a TPP-moiety attached via a 6-carbon spacer to 9-amino-Doxycycline.
FIGURE 2 | Doxy-Myr more potently inhibits the anchorage-independent propagation of CSCs. (A) MCF7 cells were plated under anchorage-independent growth
conditions and the number of 3D-mammospheres were counted after 5 days. Note that Doxycycline and Doxy-Myr both inhibited 3D-mammosphere formation, all
relative to vehicle-alone controls. However, Doxy-Myr was >5-fold more potent than Doxycycline (IC-50 of 3.46 vs. 18.1 µM). (B) Representative phase images of
3D-mammospheres are shown. Bar =200 µm.
Frontiers in Oncology | www.frontiersin.org 5September 2020 | Volume 10 | Article 1528
Ózsvári et al. Doxycycline Derivatives for Metastasis Prevention
FIGURE 3 | Doxy-Myr is better retained within cells and reveals a peri-nuclear staining pattern. MCF7 cells were cultured for 72 h as 2D-monolayers, in the presence
of Doxycycline or Doxy-Myr, at a concentration of 10 µM. Vehicle-alone controls were processed in parallel. Then, MCF7 cells were washed twice with PBS and
subjected to live cell imaging to capture the auto-fluorescent signal retained within cells. Note that Doxy-Myr showed the strongest intracellular retention, and was
concentrated within peri-nuclear intracellular compartments. No nuclear staining was observed. Quantitation of mean pixel intensity, using Image J software, revealed
that relative to Doxycycline, Doxy-Myr showed a near 3-fold increase in intracellular fluorescence.
treated with vehicle alone (Figure 3). Doxy-Myr fluorescence
showed a peri-nuclear staining pattern, consistent with its
partitioning and retention within intracellular membranous
compartments. This observation could mechanistically explain
its increased potency. No nuclear staining for Doxy-Myr was
observed, indicating that it was predominantly excluded from
the nucleus.
Doxy-Myr Is Non-toxic in 2D-Monolayers of
MCF7 Cells and Normal Human Fibroblasts
To further assess the effects of Doxy-Myr on 2D-cell growth, we
next used MCF7 cells and normal human fibroblasts (hTERT-
BJ1) treated over a period of 3 days. Figure 4 shows that
both Doxycycline and Doxy-Myr had no appreciable effects
on cell viability, as determined over the concentration range
of 5–20 µM.
Potential 2D-effects on cell proliferation and the cell cycle
were also determined using MCF7 cell monolayers. Clearly,
Doxycycline and Doxy-Myr did not inhibit the proliferation
of MCF7 cells, as assessed using the xCELLigence (Figure 5).
Similarly, relative to the parent compound Doxycycline,
Doxy-Myr did not have any significant effects on reducing cell
cycle progression in 2D-monolayers of MCF7 cells (Figure 6).
Therefore, overall Doxy-Myr did not significantly reduce
the viability, proliferation or cell cycle progression of 2D-
monolayers of MCF7 cells, indicating that its effects were
specific for cell propagation under 3D anchorage-independent
growth conditions.
Doxy-Laur and Doxy-Pal Are Less Potent
Than Doxy-Myr in Targeting CSCs
We also synthesized two other new Doxycycline analogs to
study the influence of the fatty acid chain length on their
functional activity. These two analogs included Doxy-Laur
(harboring a 12 carbon fatty acid) and Doxy-Pal (harboring
a 16 carbon fatty acid) (Figure 7). Therefore, we directly
compared the functional inhibitory activity of Doxy-Myr, Doxy-
Laur, and Doxy-Pal in the 3D-mammosphere assay, using
MCF7 cells. Interestingly, Figure 8 demonstrates that the rank
order potency is: Doxy-Myr >Doxy-Laur >Doxy-Pal >
Doxycycline, with no direct correlation observed between chain
length and activity. As such, the addition of myristic acid
Frontiers in Oncology | www.frontiersin.org 6September 2020 | Volume 10 | Article 1528
Ózsvári et al. Doxycycline Derivatives for Metastasis Prevention
FIGURE 4 | Doxy-Myr does not affect the viability of MCF7 cells or normal fibroblasts, when grown as 2D-monolayers. To determine the effects of Doxy-Myr on cell
viability, we next treated MCF7 cells and normal human fibroblasts (hTERT-BJ1) over a period of 3 days. Note that Doxycycline and Doxy-Myr had no appreciable
effects on cell viability, in the concentration range of 5–20 µM. (A) MCF7 cells, (B) hTERT-BJ1 cells.
FIGURE 5 | Doxy-Myr does not inhibit the proliferation of MCF7 cell 2D-monolayers. Potential effects of Doxycycline and Doxy-Myr on cell proliferation were
determined using MCF7 cell monolayers. Note that Doxycycline and Doxy-Myr did not inhibit the proliferation of MCF7 cells, as assessed using the xCELLigence, in
the concentration range of 5–20 µM. *p<0.05.
Frontiers in Oncology | www.frontiersin.org 7September 2020 | Volume 10 | Article 1528
Ózsvári et al. Doxycycline Derivatives for Metastasis Prevention
FIGURE 6 | Doxy-Myr does not induce cell cycle arrest, in MCF7 2D-monolayers. MCF7 cells were cultured for 72 h as 2D-monolayers, in the presence of
Doxycycline or Doxy-Myr, at a concentration of 10 µM. Vehicle-alone controls were processed in parallel. Note that relative to the parent compound Doxycycline,
Doxy-Myr did not have any significant effects on reducing cell cycle progression in 2D-monolayers of MCF7 cells. Representative FACS cell cycle profiles are shown.
FIGURE 7 | Chemical structures of two other Doxycycline derivatives: Doxy-Laur and Doxy-Pal. We created two other new Doxycycline analogs by varying the chain
length of the fatty acid attached to 9-amino-Doxycycline. These two analogs included Doxy-Laur (harboring a 12 carbon fatty acid) and Doxy-Pal (harboring a 16
carbon fatty acid). Their chemical structures are as shown.
(a 14 carbon fatty acid) appears to be the optimal chain
length modification.
Doxy-Myr, Doxy-Laur, and Doxy-Pal Lack
Antibiotic Activity Against Common
Gram-Negative and Gram-Positive
Bacteria
Doxycycline is a well-established broad-spectrum antibiotic,
that is routinely used for therapeutically targeting both
gram-negative and gram-positive bacterial infections. As
a consequence, we also assessed the antibiotic activity
of the Doxycycline analogs, as compared to the parent
compound Doxycycline.
Figure 9 reveals that, as expected, Doxycycline potently and
effectively inhibits the growth of both gram-negative (E. coli) and
gram-positive (S. aureus) micro-organisms. Minimum inhibitory
concentrations were 3.125 µM (1.3 mg/L) and 12.5 µM (5.5
mg/L), respectively. However, in striking contrast, Doxy-Myr,
Frontiers in Oncology | www.frontiersin.org 8September 2020 | Volume 10 | Article 1528
Ózsvári et al. Doxycycline Derivatives for Metastasis Prevention
FIGURE 8 | Rank order potency of the new Doxycycline derivatives. Note that Doxy-Myr is the most potent Doxycycline derivative for targeting CSC propagation, as
assayed using the 3D mammosphere assay to quantitatively measure anchorage-independent growth. The rank order potency is Doxy-Myr >Doxy-Laur >Doxy-Pal
>Doxycycline.
FIGURE 9 | Doxy-Myr, Doxy-Laur and Doxy-Pal do not show any residual antibiotic activity. The novel Doxycycline analogs (Doxy-Myr, Doxy-Laur, and Doxy-Pal) were
screened against a gram-positive (S. aureus; ATCC 29213) and a gram-negative (E. coli; ATCC 25922) strain of bacteria to verify the maintenance of the antibiotic
activity, when compared to the parent compound. While Doxycycline presented MIC values of 3.125 µM against E. coli and 12.5 µM against S. aureus, none of the
Doxycycline analogs inhibited bacterial growth up to the concentration of 100 µM.
Doxy-Laur, and Doxy-Pal did not show any obvious antibiotic
activity, in the same concentration range. Minimum inhibitory
concentrations of the Doxycycline analogs were >100 µM (>65
mg/L). The clinical breakpoints for tetracyclines against E. coli
and S. aureus are between 0.5–2.0 mg/L and 2.0–6.0 mg/L,
respectively (28).
Frontiers in Oncology | www.frontiersin.org 9September 2020 | Volume 10 | Article 1528
Ózsvári et al. Doxycycline Derivatives for Metastasis Prevention
Therefore, these simple chemical modifications of
Doxycycline have successfully removed its ability to act as
a functional antibiotic, while simultaneously increasing its
specificity for targeting CSCs.
FIGURE 10 | Doxycycline and Doxy-Myr have no effect on tumor growth.
MDA-MB-231 cells and the well-established chorio-allantoic membrane (CAM)
assay in chicken eggs were used to quantitatively measure tumor growth. An
inoculum of 1 ×106MDA-MB-231 cells was added onto the Upper CAM of
each egg (on Day E9) and then eggs were then randomized into groups. On
day E10, tumors were detectable and they were then treated daily for 8 days
with vehicle alone (1% DMSO in PBS), Doxycycline or Doxy-Myr. After 8 days
of drug administration, on day E18 all tumors were weighed. Note that
Doxycycline and Doxy-Myr did not have any significant effects on tumor
growth. Averages are shown ±SEM. NS, not significant.
Doxy-Myr Potently Inhibits Cancer Cell
Metastasis in vivo, Without Significant
Toxicity
To experimentally evaluate the functional effects of Doxycycline
and Doxy-Myr in vivo, we next used MDA-MB-231 cells and
the well-established chorio-allantoic membrane (CAM) assay
in chicken eggs, to quantitatively measure tumor growth and
metastasis (1619). MDA-MB-231 breast cancer cells were used
for our in vivo studies, as they are estrogen-independent,
intrinsically more aggressive, form larger tumors and are
significantly more migratory, invasive and metastatic. As such,
they are a better in vivo model, for simultaneously evaluating
both tumor growth and spontaneous metastasis. Moreover, we
have previously demonstrated that Doxycycline also effectively
inhibits the 3D anchorage-independent growth of MDA-MB-231
cells (13).
Briefly, an inoculum of 1 ×106MDA-MB-231 cells was added
onto the Upper CAM of each egg (day E9) and then eggs were
randomized into groups. On day E10, tumors were detectable
and they were then treated daily for 8 days with vehicle alone
(1% DMSO in PBS), Doxycycline or Doxy-Myr. After 8 days
of drug administration, on day E18 all tumors were weighed,
and the Lower CAM was collected to evaluate the number of
metastatic cells, as analyzed by qPCR with specific primers for
Human Alu sequences.
Morphologically, the CAM is constructed of two opposing
sheets of epithelial cells, which are separated by a middle stromal
layer, containing blood vessels and lymphatics. One epithelial
layer is of ectodermal origin, while the other epithelial layer
is of mesodermal/endodermal origin. Importantly, movement
FIGURE 11 | Doxycycline and Doxy-Myr selectively target and prevent cancer metastasis. MDA-MB-231 cells and the well-established chorio-allantoic membrane
(CAM) assay in chicken eggs were used to quantitatively measure spontaneous tumor metastasis. An inoculum of 1 ×106MDA-MB-231 cells was added onto the
Upper CAM of each egg (on day E9) and then eggs were then randomized into groups. On day E10, tumors were detectable and they were then treated daily for 8
days with vehicle alone (1% DMSO in PBS), Doxycycline or Doxy-Myr. After 8 days of drug administration, the Lower CAM was collected to evaluate the number of
metastatic cells, as analyzed by qPCR with specific primers for Human Alu sequences. Note that Doxycycline and Doxy-Myr both showed significant effects on
MDA-MB-231 metastasis. However, Doxy-Myr was clearly more effective than Doxycycline in inhibiting metastasis. Averages are shown ±SEM. ****p<0.0001.
Frontiers in Oncology | www.frontiersin.org 10 September 2020 | Volume 10 | Article 1528
Ózsvári et al. Doxycycline Derivatives for Metastasis Prevention
TABLE 1 | Chick embryo toxicity of Doxycycline and Doxy-Myr.
Group. # Group description Total Alive Dead % Alive % Dead
1 Neg. Ctrl. 18 15 3 83.33 16.67
2 Doxy, 0.125 mM 13 11 2 84.62 15.38
3 Doxy, 0.250 mM 13 10 3 76.92 23.08
4 Doxy-Myr, 0.125 mM 14 11 3 78.57 21.43
5 Doxy-Myr, 0.250 mM 13 12 1 92.31 7.69
of metastatic MDA-MB-231 cells from the Upper CAM to the
Lower CAM involves their migration away from the primary
tumor, cellular invasion, intravasation, extravasation, and the
formation of a new distant lesion, all of the normal steps
that are key features of spontaneous tumor cell metastasis (see
Supplemental Figure 2).
Figure 10 shows the effects of Doxycycline and Doxy-Myr on
MDA-MB-231 tumor growth. Note that that they both did not
show any significant effects on tumor growth, as a result of the
8-day period of drug administration.
However, both Doxycycline and Doxy-Myr showed significant
effects on MDA-MB-231 cancer cell metastasis. Figure 11
illustrates that Doxycycline inhibited metastasis (by 44–57.5%).
In contrast, Doxy-Myr inhibited metastasis (by 85–87%), at the
same concentrations tested. Interestingly, the effects of Doxy-
Myr on metastasis were significantly more pronounced.
Surprisingly, little or no embryo toxicity was observed for
Doxycycline and Doxy-Myr (Table 1). Therefore, we conclude
that Doxy-Myr can be further developed as an anti-metastatic
agent, selectively inhibiting tumor metastasis, without showing
significant toxicity or antibiotic activity.
DISCUSSION
Here, we report the chemical synthesis and biological activity
of several new Doxycycline analogs, modified to increase their
effectiveness in the targeting of CSCs. The most promising
compound was Doxy-Myr, a Doxycycline analog in which a
myristic acid (14 carbon) moiety is covalently attached to
the free amino group of 9-amino-Doxycycline. We analyzed
the potency of Doxy-Myr, using the 3D-mammosphere assay,
to assess its potential inhibitory effects on the anchorage-
independent propagation of breast CSCs. Overall, we observed
that Doxy-Myr is >5-fold more potent than Doxycycline,
the parent compound. Moreover, Doxy-Myr showed better
intracellular retention, and was specifically localized within a
peri-nuclear membranous compartment. In striking contrast,
when MCF7 breast cancer cells or normal fibroblasts were
grown as 2D-monolayers, Doxy-Myr did not reveal any
effects on cell viability or proliferation, highlighting its unique
selectivity for targeting the 3D-propagation of CSCs. In addition,
we evaluated other Doxycycline analogs, with longer (16
carbon; palmitic acid) or shorter (12 carbon; lauric acid)
chain lengths (Figure 12). However, these two analogs were
less effective than Doxy-Myr for targeting of CSCs. Finally,
FIGURE 12 | Schematic diagram highlighting our systematic approach to
generating Doxycycline derivatives, to target CSCs and prevent metastasis.
Lipid moieties were covalently conjugated to 9-amino-Doxycycline.
Importantly, these three Doxycycline derivatives lack anti-microbial activity.
using MDA-MB-231 cells, we demonstrated that Doxy-Myr
has no appreciable effects on tumor growth, but potently
inhibits tumor cell metastasis in vivo, with little or no chick
embryo toxicity.
Our results also showed that the lipophilic amide substituents
in Doxycycline on the C9 of the Tetracycline (TC) skeleton
led to the loss of its antibacterial activity. Previously published
structure-activity relationship (SAR) studies have shown that
chemical modification of the TC skeleton on C9 can be tolerated,
leading to diverse antibacterial activity, as is exemplified by
the antibiotic Tigecycline. The lipophilicity of the TCs seems
to play a key role in the biological potency of this family
of drugs. There is a trend of decreased antibiotic activity
with the increase of lipophilicity, specifically against gram-
negative species, with eventual loss of activity when high
lipophilicity is achieved, which could partially explain the loss
of activity, we observed after the addition of fatty acids to the
Doxycycline scaffold.
Importantly, our improvement in the biological properties of
these Doxycycline analogs for targeting CSCs and the associated
loss of the anti-microbial activity, make these new analogs
extremely promising, because tetracycline resistance among
gram-negative and gram-positive pathogens requires exposure to
inhibitory concentrations as a selective pressure (2830). MICs
exceeding 65 mg/L also suggest that these analogs might be non-
inhibitory to members of the human microbiome, the complex
community of microorganisms which exerts wide-ranging effects
on human immunity and disease (29,30).
Interestingly, a previous study successfully used the parent
compound, Doxycycline, to prevent bone metastasis in a mouse
model, by employing MDA-MD-231 cells (31). However, these
Frontiers in Oncology | www.frontiersin.org 11 September 2020 | Volume 10 | Article 1528
Ózsvári et al. Doxycycline Derivatives for Metastasis Prevention
FIGURE 13 | In vitro inhibition of 3D-growth predicts in vivo inhibition of metastasis. Here, we used two complementary breast cancer cell lines for our in vitro
screening (MCF7) and in vivo (MDA-MB-231) validation assays. More specifically, Doxy-Myr had no effect on 2D-growth in vitro and no effect on tumor growth in vivo.
Conversely, we showed that Doxy-Myr potently inhibited 3D-growth in vitro, which directly correlated with inhibition of metastasis in vivo. In support of this
observation, 3D anchorage-independent growth is thought to be a required step for metastasis in vivo. N.E., no effect.
FIGURE 14 | Summary: Properties of Doxy-Myr. Briefly, Doxy-Myr is a lipid modified Doxycycline derivative. Our results show that Doxy-Myr potently targets CSCs
and selectively prevents metastasis, without affecting tumor growth. Moreover, Doxy-Myr was non-toxic in the chick embryo assay and did not affect the viability of
normal cells, or MCF7 cells, grown as a 2D-monolayer. Importantly, Doxy-Myr lacked antibiotic activity, and did not affect the growth of gram-positive (E. coli) or
gram-negative (S. aureus) organisms.
Frontiers in Oncology | www.frontiersin.org 12 September 2020 | Volume 10 | Article 1528
Ózsvári et al. Doxycycline Derivatives for Metastasis Prevention
authors did not examine the effects of Doxycycline on tumor
growth, but only focused on bone metastasis. They attributed the
efficacy of Doxycycline to its tropism for bone and to its ability
to act as a protease inhibitor for lysosomal cysteine proteinases,
the cathepsins, and MMPs, because Doxycycline behaves as a
zinc chelator.
In contrast, herein, we have demonstrated that Doxycycline
and Doxy-Myr both act as inhibitors of metastasis, by targeting
the 3D anchorage-independent growth of CSCs, which is a
completely different molecular mechanism (Figures 13,14).
However, as predicted, Doxy-Myr was significantly more effective
than Doxycycline, at the same concentrations examined. As such,
based on these functional observations, we propose that this
overall lipid modification strategy may be generally applicable,
to facilitate the development and discovery of other drugs,
for effectively preventing tumor progression, recurrence, and
distant metastasis.
Moreover, our current results are consistent with recent
studies showing that prophylaxis with other classes of
mitochondrial inhibitors is indeed sufficient to prevent
metastasis, using the same pre-clinical xenograft model, with
little or no effect on tumor growth and minimal toxicity (32).
DATA AVAILABILITY STATEMENT
All datasets generated for this study are included in the
article/Supplementary Material.
AUTHOR CONTRIBUTIONS
ML and FS conceived and initiated this project, they selected the
clinically-approved drug Doxycycline for chemical modification
and optimization by medicinal chemistry. JK performed the
custom-chemical syntheses. The phenotypic drug screening and
the majority of other wet-lab experiments described in this paper
were performed by BÓ. LM determined the antibiotic activity of
Doxycycline and its derivatives. BÓ, JK, and LM generated the
final figures for the paper. ML and FS wrote the first draft of the
manuscript, which was then further edited and approved by BÓ,
LM, JL, JK, FS, and ML. ML generated the schematic summary
diagrams. All authors contributed to the article and approved the
submitted version.
FUNDING
This work was supported by research grant funding, provided by
Lunella Biotech, Inc. The funder, Lunella Biotech, Inc., provided
the necessary monetary resources to carry out the current study.
ACKNOWLEDGMENTS
We are grateful to Rumana Rafiq, for her kind and dedicated
assistance, in keeping the Translational Medicine Laboratory
at Salford running smoothly. We would like to thank the
Foxpoint Foundation (Canada) and the Healthy Life Foundation
(UK) for their philanthropic donations toward new equipment
and infrastructure, in the Translational Medicine Laboratory at
the University of Salford. We are thankful to Inovotion, Inc.
(Grenoble, France), for independently performing the tumor
growth and metastasis studies, using the CAM assay, as well as
evaluating chicken embryo toxicity, through a research contract
with Lunella Biotech, Inc. (Ottawa, Canada).
SUPPLEMENTARY MATERIAL
The Supplementary Material for this article can be found
online at: https://www.frontiersin.org/articles/10.3389/fonc.
2020.01528/full#supplementary-material
Supplemental Figure 1 | Chemical synthesis of Doxycycline derivatives.
Supplemental Figure 2 | CAM model for measuring tumor growth, metastasis,
and embryo toxicity. On day E9, an inoculum of 1 million MDA-MB-231 breast
tumor cells was layered on top of the Upper CAM and was allowed to form a
primary tumor. Potential therapeutics were applied for a period of 8-days. Then,
on day E18, the primary tumor was harvested from the upper CAM and the
magnitude of distant metastases was quantitated in the Lower CAM, by
performing qPCR with specific primers for recognizing Human Alu sequences. In
order for the cells to metastasize, from the Upper CAM to the Lower CAM, it has
been established that they need to undergo migration, invasion, intravasation,
extravasation, and secondary lesion formation. Toxicity was measured by scoring
embryo viability on day E18. See Materials and Methods for further details.
Reproduced and modified, under a creative commons license, from the following
source (33).
REFERENCES
1. De Francesco EM, Sotgia F, Lisanti MP. Cancer stem cells (CSCs): metabolic
strategies for their identification and eradication. Biochem J. (2018) 475:1611–
34. doi: 10.1042/BCJ20170164
2. Martinez-Outschoorn UE, Peiris-Pagés M, Pestell RG, Sotgia F, Lisanti MP.
Cancer metabolism: a therapeutic perspective. Nat Rev Clin Oncol. (2017)
14:11–31. doi: 10.1038/nrclinonc.2016.60
3. Peiris-Pagès M, Martinez-Outschoorn UE, Pestell RG, Sotgia F,
Lisanti MP. Cancer stem cell metabolism. Breast Cancer Res. (2016)
18:55. doi: 10.1186/s13058-016-0712-6
4. Yu Z, Pestell TG, Lisanti MP, Pestell RG. Cancer stem cells. Int
J Biochem Cell Biol. (2012) 44:2144–51. doi: 10.1016/j.biocel.2012.
08.022
5. Sotgia F, Ozsvari B, Fiorillo M, De Francesco EM, Bonuccelli G,
Lisanti MP. A mitochondrial based oncology platform for targeting
cancer stem cells (CSCs): MITO-ONC-RX. Cell Cycle. (2018) 17:2091–
100. doi: 10.1080/15384101.2018.1515551
6. De Luca A, Fiorillo M, Peiris-Pagès M, Ozsvari B, Smith DL, Sanchez-
Alvarez R, et al. Mitochondrial biogenesis is required for the anchorage-
independent survival and propagation of stem-like cancer cells. Oncotarget.
(2015) 6:14777–95. doi: 10.18632/oncotarget.4401
7. Farnie G, Sotgia F, Lisanti MP. High mitochondrial mass identifies a sub-
population of stem-like cancer cells that are chemo-resistant. Oncotarget.
(2015) 6:30472–86. doi: 10.18632/oncotarget.5401
8. Lamb R, Harrison H, Hulit J, Smith DL, Lisanti MP, Sotgia F. Mitochondria
as new therapeutic targets for eradicating cancer stem cells: quantitative
proteomics and functional validation via MCT1/2 inhibition. Oncotarget.
(2014) 5:11029–37. doi: 10.18632/oncotarget.2789
9. Zhang L, Xu L, Zhang F, Vlashi E. Doxycycline inhibits the cancer stem cell
phenotype and epithelial-to-mesenchymal transition in breast cancer. Cell
Cycle. (2017) 16:737–45. doi: 10.1080/15384101.2016.1241929
Frontiers in Oncology | www.frontiersin.org 13 September 2020 | Volume 10 | Article 1528
Ózsvári et al. Doxycycline Derivatives for Metastasis Prevention
10. Lamb R, Fiorillo M, Chadwick A, Ozsvari B, Reeves KJ, Smith L, et al.
Doxycycline down-regulates DNA-PK and radiosensitizes tumor initiating
cells: implications for more effective radiation therapy. Oncotarget. (2015)
6:14005–25. doi: 10.18632/oncotarget.4159
11. Sotgia F, Whitaker-Menezes D, Martinez-Outschoorn UE, Flomenberg
N, Birbe RC, Witkiewicz K, et al. Mitochondrial metabolism in cancer
metastasis: visualizing tumor cell mitochondria and the “reverse Warburg
effect” in positive lymph node tissue. Cell Cycle. (2012) 11:1445–
54. doi: 10.4161/cc.19841
12. Lin CC, Lo MC, Moody RR, Stevers NO, Tinsley SL, Sun D. Doxycycline
targets aldehyde dehydrogenase-positive breast cancer stem cells. Oncol Rep.
(2018) 39:3041–7. doi: 10.3892/or.2018.6337
13. Lamb R, Ozsvari B, Lisanti CL, Tanowitz HB, Howell A, Martinez-Outschoorn
E, et al. Antibiotics that target mitochondria effectively eradicate cancer stem
cells, across multiple tumor types: treating cancer like an infectious disease.
Oncotarget. (2015) 6:4569–84. doi: 10.18632/oncotarget.3174
14. Scatena C, Roncella M, Di Paolo A, Aretini P, Menicagli M, Fanelli G, et al.
Doxycycline, an inhibitor of mitochondrial biogenesis, effectively reduces
Cancer Stem Cells (CSCs) in early breast cancer patients: a clinical pilot study.
Front Oncol. (2018) 8:452. doi: 10.3389/fonc.2018.00452
15. Barden CT, Buckwalter LB, Testa TR, Petersen JP, Lee VJ.
“Glycylcyclines”. 3. 9-aminodoxycyclinecarboxamides. J Med Chem. (1994)
37:3205–11. doi: 10.1021/jm00046a003
16. Ozsvari B, Fiorillo M, Bonuccelli G, Cappello AR, Frattaruolo L, Sotgia F,
et al. Mitoriboscins: mitochondrial-based therapeutics targeting cancer stem
cells (CSCs), bacteria and pathogenic yeast. Oncotarget. (2017) 8:67457–
72. doi: 10.18632/oncotarget.19084
17. Ozsvari B, Sotgia F, Lisanti MP. Exploiting mitochondrial targeting signal(s),
TPP and bis-TPP, for eradicating cancer stem cells (CSCs). Aging. (2018)
10:229–40. doi: 10.18632/aging.101384
18. Ozsvari B, Nuttall JR, Sotgia F, Lisanti MP. Azithromycin and roxithromycin
define a new family of “senolytic” drugs that target senescent human
fibroblasts. Aging. (2018) 10:3294–307. doi: 10.18632/aging.101633
19. Fiorillo M, Sotgia F, Lisanti MP. “Energetic” cancer stem cells
(e-CSCs): a new hyper-metabolic and proliferative tumor cell
phenotype, driven by mitochondrial energy. Front Oncol. (2019)
8:677. doi: 10.3389/fonc.2018.00677
20. The European Committee on Antimicrobial Susceptibility Testing. Routine
Extended Internal Quality Control for MIC Determination Disk Diffusion as
Recommended by EUCAST,Version 9.0 (2019). Available online at: http://
www.eucast.org (accessed August 12, 2020).
21. M02-A12. Performance Standards for Antimicrobial Disk Susceptibility Tests;
Approved Standard—Twelfth Edition. Wayne, PA: Clinical and Laboratory
Standards Institute. (2015).
22. Blackwood RK, English AR. Structure-activity relationships in the tetracycline
series. In: Perlman D, editor. Advances in Applied Microbiology. London:
Academic Press, Inc. (1970). p. 237–65. doi: 10.1128/AAC.01536-15
23. Alsamri H, El Hasasna H, Al Dhaheri Y, Eid AH, Attoub S, Iratni R. Carnosol,
a natural polyphenol, inhibits migration, metastasis, and tumor growth of
breast cancer via a ROS-dependent proteasome degradation of STAT3. Front
Oncol. (2019) 9:743. doi: 10.3389/fonc.2019.00743
24. Nascimento BFO, Laranjo M, Pereira NAM, Dias-Ferreira J, Piñeiro M,
Botelho MF, et al. Ring-fused diphenylchlorins as potent photosensitizers
for photodynamic therapy applications: in vitro tumor cell biology and in
vivo chick embryo chorioallantoic membrane studies. ACS Omega. (2019)
4:17244–50. doi: 10.1021/acsomega.9b01865
25. Gilson P, Couvet M, Vanwonterghem L, Henry M, Vollaire J, Baulin
V, et al. The pyrrolopyrimidine colchicine-binding site agent PP-13
reduces the metastatic dissemination of invasive cancer cells in vitro
and in vivo.Biochem Pharmacol. (2019) 160:1–13. doi: 10.1016/j.bcp.2018.
12.004
26. El Hasasna H, Saleh A, Al Samri H, Athamneh K, Attoub S, Arafat K, et al.
Rhus coriaria suppresses angiogenesis, metastasis and tumor growth of breast
cancer through inhibition of STAT3, NFκB and nitric oxide pathways. Sci Rep.
(2016) 6:21144. doi: 10.1038/srep21144
27. Al Dhaheri Y, Attoub S, Arafat K, Abuqamar S, Viallet J, Saleh A, et al.
Anti-metastatic and anti-tumor growth effects of Origanum majorana
on highly metastatic human breast cancer cells: inhibition of NFκB
signaling and reduction of nitric oxide production. PLoS ONE. (2013)
8:e68808. doi: 10.1371/journal.pone.0068808
28. The European Committee on Antimicrobial Susceptibility Testing. Breakpoint
Tables for Interpretation of MICs and Zone Diameters, Version 10.0. (2020).
Available online at: https://eucast.org/ (accessed August 12, 2020).
29. Belkaid Y, Segre JA.Dialogue between skin microbiota and immunity. Science.
(2014) 346:954–9. doi: 10.1126/science.1260144
30. Lazar V, Ditu LM, Pircalabioru GG, Gheorghe I, Curutiu C, Holban
AM, et al. Aspects of gut microbiota and immune system interactions in
infectious diseases, immunopathology, and cancer. Front Immunol. (2018)
9:1830. doi: 10.3389/fimmu.2018.01830
31. Duivenvoorden WC, Popovi´
c SV, Lhoták S, Seidlitz E, Hirte HW, Tozer RG,
et al. Doxycycline decreases tumor burden in a bone metastasis model of
human breast cancer. Cancer Res. (2002) 62, 1588–91.
32. Ózsvári B, Sotgia F, Lisanti MP. First-in-class candidate therapeutics
that target mitochondria and effectively prevent cancer cell
metastasis: mitoriboscins and TPP compounds. Aging. (2020)
12:10162–79. doi: 10.18632/aging.103336
33. Dünker N, Jendrossek V. Implementation of the Chick Chorioallantoic
Membrane (CAM) model in radiation biology and experimental radiation
oncology research. Cancers. (2019) 11:E1499. doi: 10.3390/cancers111
01499
Conflict of Interest: JK is employed by the company Eurofins Integrated
Discovery UK Ltd. FS holds a part-time affiliation with Lunella Biotech, Inc. ML
holds a part-time affiliation with Lunella Biotech, Inc.
The remaining authors declare that the research was conducted in the absence of
any commercial or financial relationships that could be construed as a potential
conflict of interest.
Copyright © 2020 Ózsvári, Magalhães, Latimer, Kangasmetsa, Sotgia and Lisanti.
This is an open-access article distributed under the terms of the Creative Commons
Attribution License (CC BY). The use, distribution or reproduction in other forums
is permitted, provided the original author(s) and the copyright owner(s) are credited
and that the original publication in this journal is cited, in accordance with accepted
academic practice. No use, distribution or reproduction is permitted which does not
comply with these terms.
Frontiers in Oncology | www.frontiersin.org 14 September 2020 | Volume 10 | Article 1528
... Xenograft assays were carried out by INOVOTION (Société: 811310127), La Tronche-France, essentially as we previously described, without any major modifications [23,24]. Fertilized White Leghorn eggs were incubated at 37.5°C with 50% relative humidity for 9 days. ...
... Comparative slope analysis of the three cell subpopulations. At each time point (24,48,72,96, and 120 h) the slope of the ATP-high cell population, was significantly higher compared to the other two subpopulations. Data represent mean ± SD, n = 3. Two-way ANOVA, Tukey corrected, *p < 0.01. ...
... Recently, we presented evidence that treatment with a panel of distinct anti-mitochondrial therapeutics, that target the mitochondrial ribosome, is sufficient to potently inhibit cancer cell metastasis, using an in vivo xenograft model [23,24]. These results functionally imply that high ATP levels are critical for the process of CSC metastasis. ...
... Xenograft assays were carried out by INOVOTION (Société: 811310127), La Tronche-France, essentially as we previously described, without any major modifications [23,24]. Fertilized White Leghorn eggs were incubated at 37.5°C with 50% relative humidity for 9 days. ...
... Comparative slope analysis of the three cell subpopulations. At each time point (24,48,72,96, and 120 h) the slope of the ATP-high cell population, was significantly higher compared to the other two subpopulations. Data represent mean ± SD, n = 3. Two-way ANOVA, Tukey corrected, *p < 0.01. ...
... Recently, we presented evidence that treatment with a panel of distinct anti-mitochondrial therapeutics, that target the mitochondrial ribosome, is sufficient to potently inhibit cancer cell metastasis, using an in vivo xenograft model [23,24]. These results functionally imply that high ATP levels are critical for the process of CSC metastasis. ...
Article
Full-text available
Here, we provide evidence that high ATP production by the mitochondrial ATP-synthase is a new therapeutic target for anticancer therapy, especially for preventing tumor progression. More specifically, we isolated a subpopulation of ATP-high cancer cells which are phenotypically aggressive and demonstrate increases in proliferation, stemness, anchorage-independence, cell migration, invasion and multi-drug resistance, as well as high antioxidant capacity. Clinically, these findings have important implications for understanding treatment failure and cancer cell dormancy. Using bioinformatic analysis of patient samples, we defined a mitochondrial-related gene signature for metastasis, which features the gamma-subunit of the mitochondrial ATP-synthase (ATP5F1C). The relationship between ATP5F1C protein expression and metastasis was indeed confirmed by immunohistochemistry. Next, we used MDA-MB-231 cells as a model system to functionally validate these findings. Importantly, ATP-high MDA-MB-231 cells showed a nearly fivefold increase in metastatic capacity in vivo. Consistent with these observations, ATP-high cells overexpressed (i) components of mitochondrial complexes I–V, including ATP5F1C, and (ii) markers associated with circulating tumor cells (CTCs) and metastasis, such as EpCAM and VCAM1. Knockdown of ATP5F1C expression significantly reduced ATP-production, anchorage-independent growth, and cell migration, as predicted. Similarly, therapeutic administration of the FDA-approved drug, Bedaquiline, downregulated ATP5F1C expression in vitro and prevented spontaneous metastasis in vivo. In contrast, Bedaquiline had no effect on the growth of non-tumorigenic mammary epithelial cells (MCF10A) or primary tumors in vivo. Taken together, our results suggest that mitochondrial ATP depletion is a new therapeutic strategy for metastasis prophylaxis, to avoid treatment failure. In summary, we conclude that mitochondrial ATP5F1C is a promising new biomarker and molecular target for future drug development, for the prevention of metastatic disease progression.
... This is clearly demonstrated by numerous examples: QPS-derivatives of chlorambucil [22], paclitaxel [23], doxorubicin [24], and metformin [25]. Incorporation of QPS into antibiotic molecules such as ciprofloxacin [26,27], azithromycin [28,29], doxycycline [28,29] significantly improves the antibacterial activity of doxycycline against Gram-positive bacteria. Azithromycin and doxycycline can also be considered as anti-tumor agents. ...
... This is clearly demonstrated by numerous examples: QPS-derivatives of chlorambucil [22], paclitaxel [23], doxorubicin [24], and metformin [25]. Incorporation of QPS into antibiotic molecules such as ciprofloxacin [26,27], azithromycin [28,29], doxycycline [28,29] significantly improves the antibacterial activity of doxycycline against Gram-positive bacteria. Azithromycin and doxycycline can also be considered as anti-tumor agents. ...
Article
Full-text available
It has been shown for a wide range of epoxy compounds that their interaction with triphenylphosphonium triflate occurs with a high chemoselectivity and leads to the formation of (2-hydroxypropyl)triphenylphosphonium triflates 3 substituted in the 3-position with an alkoxy, alkylcarboxyl group, or halogen, which were isolated in a high yield. Using the methodology for the disclosure of epichlorohydrin with alcohols in the presence of boron trifluoride etherate, followed by the substitution of iodine for chlorine and treatment with triphenylphosphine, 2-hydroxypropyltriphenylphosphonium iodides 4 were also obtained. The molecular and supramolecular structure of the obtained phosphonium salts was established, and their high antitumor activity was revealed in relation to duodenal adenocarcinoma. The formation of liposomal systems based on phosphonium salt 3 and L-α-phosphatidylcholine (PC) was employed for improving the bioavailability and reducing the toxicity. They were produced by the thin film rehydration method and exhibited cytotoxic properties. This rational design of phosphonium salts 3 and 4 has promising potential of new vectors for targeted delivery into mitochondria of tumor cells.
... Recently, we also demonstrated that treatment with a panel of mitochondrially-targeted therapeutics, which potently inhibit mitochondrial protein translation or OXPHOS, could block tumor cell metastasis, using an in vivo pre-clinical model (54)(55)(56). These results indicated that ATP levels are functionally critical for the processes fueling aggressive tumor cell behaviors and spontaneous metastasis. ...
... In further support of these clinical observations, MT-CO2 is over-expressed by >20fold in an hTERT-enriched sub-population of breast cancer stem cells (86). Pharmacologically, MT-CO2 is effectively targeted by the FDA-approved antibiotic Doxycycline (87), which behaves as an inhibitor of mitochondrial protein translation and prevents ATP production (87), ultimately blocking metastasis in preclinical models (54). Therefore, Doxycycline may also provide a therapeutic solution for inhibiting MT-CO2 in breast cancer patients, to help prevent disease progression. ...
Article
Full-text available
Recently, we presented evidence that high mitochondrial ATP production is a new therapeutic target for cancer treatment. Using ATP as a biomarker, we isolated the “metabolically fittest” cancer cells from the total cell population. Importantly, ATP-high cancer cells were phenotypically the most aggressive, with enhanced stem-like properties, showing multi-drug resistance and an increased capacity for cell migration, invasion and spontaneous metastasis. In support of these observations, ATP-high cells demonstrated the up-regulation of both mitochondrial proteins and other protein biomarkers, specifically associated with stemness and metastasis. Therefore, we propose that the “energetically fittest” cancer cells would be better able to resist the selection pressure provided by i) a hostile micro-environment and/or ii) conventional chemotherapy, allowing them to be naturally-selected for survival, based on their high ATP content, ultimately driving tumor recurrence and distant metastasis. In accordance with this energetic hypothesis, ATP-high MDA-MB-231 breast cancer cells showed a dramatic increase in their ability to metastasize in a pre-clinical model in vivo. Conversely, metastasis was largely prevented by treatment with an FDA-approved drug (Bedaquiline), which binds to and inhibits the mitochondrial ATP-synthase, leading to ATP depletion. Clinically, these new therapeutic approaches could have important implications for preventing treatment failure and avoiding cancer cell dormancy, by employing ATP-depletion therapy, to target even the fittest cancer cells.
... In this regard, antibiotics such as doxycycline that target the mitochondrial ribosome and inhibit translation prevent mitochondrial biogenesis which is required for the clonal expansion and survival of cancer stem cells (CSCs) [209]. Doxycycline has also been used in combination with other anticancer therapies [210][211][212][213] and their effectiveness lies in their ability to prevent metastasis by inhibiting the growth of CSCs. Interestingly, a Phase II clinical pilot study has demonstrated that doxycycline reduces the expression of stemness markers limiting the CSC burden in early breast cancer patients [214]. ...
Article
Full-text available
Simple Summary Cancer is a global health problem with high personal and economic burden worldwide. The reprogramming of metabolism experienced by carcinomas offers a large set of enzymes that could be exploited to prevent cancer growth and metastasis. This review emphasizes the interplay between mitochondrial ATP synthase and its physiological inhibitor, the ATPase Inhibitory Factor 1 (IF1), in the metabolic reprogramming of OXPHOS in cancer cells to an enhanced glycolytic phenotype. We highlight the cell-type specificity by which the ATP synthase/IF1 axis exerts its activity as a tumor promotor or signals an anti-metastatic phenotype. Moreover, the implication of the ATP synthase/IF1 axis in cell death, and as a promising target for cancer therapy, is also stressed. We think that investigations aimed at characterizing the posttranscriptional mechanisms that regulate the activity of ATP synthase/IF1 axis will provide additional promising biomarkers for effective treatment of the disease. Abstract Cancer poses a significant global health problem with profound personal and economic implications on National Health Care Systems. The reprograming of metabolism is a major trait of the cancer phenotype with a clear potential for developing effective therapeutic strategies to combat the disease. Herein, we summarize the relevant role that the mitochondrial ATP synthase and its physiological inhibitor, ATPase Inhibitory Factor 1 (IF1), play in metabolic reprogramming to an enhanced glycolytic phenotype. We stress that the interplay in the ATP synthase/IF1 axis has additional functional roles in signaling mitohormetic programs, pro-oncogenic or anti-metastatic phenotypes depending on the cell type. Moreover, the same axis also participates in cell death resistance of cancer cells by restrained mitochondrial permeability transition pore opening. We emphasize the relevance of the different post-transcriptional mechanisms that regulate the specific expression and activity of ATP synthase/IF1, to stimulate further investigations in the field because of their potential as future targets to treat cancer. In addition, we review recent findings stressing that mitochondria metabolism is the primary altered target in lung adenocarcinomas and that the ATP synthase/IF1 axis of OXPHOS is included in the most significant signature of metastatic disease. Finally, we stress that targeting mitochondrial OXPHOS in pre-clinical mouse models affords a most effective therapeutic strategy in cancer treatment.
... AGING Next steps would include: 1) their evaluation in preclinical animal models; and 2) clinical trials, as well. For example, using Doxycycline, we have previously shown that it eradicates CSCs and prevents metastasis in a preclinical animal model [95]. Moreover, a phase II clinical trial (window study) showed that Doxycycline eradicates CSCs in vivo, using CD44 and ALDH1 as CSC-markers [96]. ...
Article
Full-text available
Here, we report the identification of key compounds that effectively inhibit the anchorage-independent growth and propagation of cancer stem cells (CSCs), as determined via screening using MCF7 cells, a human breast adenocarcinoma cell line. More specifically, we employed the mammosphere assay as an experimental format, which involves the generation of 3D spheroid cultures, using low-attachment plates. These positive hit compounds can be divided into 5 categories: 1) dietary supplements (quercetin and glucosamine); 2) FDA-approved drugs (carvedilol and ciprofloxacin); 3) natural products (aloe emodin, aloin, tannic acid, chlorophyllin copper salt, azelaic acid and adipic acid); 4) flavours (citral and limonene); and 5) vitamins (nicotinamide and nicotinic acid). In addition, for the compounds quercetin, glucosamine and carvedilol, we further assessed their metabolic action, using the Seahorse to conduct metabolic flux analysis. Our results indicate that these treatments can affect glycolytic flux and suppress oxidative mitochondrial metabolism (OXPHOS). Therefore, quercetin, glucosamine and carvedilol can reprogram the metabolic phenotype of breast cancer cells. Despite having diverse chemical structures, these compounds all interfere with mitochondrial metabolism. As these compounds halt CSCs propagation, ultimately, they may have therapeutic potential.
Article
Full-text available
Breast cancer is the most frequently diagnosed malignancy worldwide and the leading cause of cancer mortality in women. Despite the recent development of new therapeutics including targeted therapies and immunotherapy, triple-negative breast cancer remains an aggressive form of breast cancer, and thus improved treatments are needed. In recent decades, it has become increasingly clear that breast cancers harbor metabolic plasticity that is controlled by mitochondria. A myriad of studies provide evidence that mitochondria are essential to breast cancer progression. Mitochondria in breast cancers are widely reprogrammed to enhance energy production and biosynthesis of macromolecules required for tumor growth. In this review, we will discuss the current understanding of mitochondrial roles in breast cancers and elucidate why mitochondria are a rational therapeutic target. We will then outline the status of the use of mitochondria-targeting drugs in breast cancers, and highlight ClpP agonists as emerging mitochondria-targeting drugs with a unique mechanism of action. We also illustrate possible drug combination strategies and challenges in the future breast cancer clinic.
Article
Full-text available
Transcription factors of the SOX family were first discovered in mammals in 1990. The sex-determining region Y box 9 belongs to the SOX transcription factor family. It plays an important role in inducing tissue and cell morphogenesis, survival, and many developmental processes. Furthermore, it has been shown to be an oncogene in many tumors. Gynecological malignancies are tumors that occur in the female reproductive system and seriously threaten the lives of patients. Common gynecological malignancies include ovarian cancer, cervical cancer, and endometrial cancer. So far, the molecular mechanisms related to the incidence and development of gynecological malignancies remain unclear. This makes it particularly important to discover their common causative molecule and thus provide an effective therapeutic target. In recent years, studies have found that multiple mechanisms are involved in regulating the expression of the sex-determining region Y box 9, leading to the occurrence and development of gynecological malignancies. In this review, we discuss the prognostic value of SOX9 expression and the potential of targeting SOX9 for gynecological malignancy treatment. We also discuss progress regarding the role of SOX9 in gynecological malignancy pathogenesis through its mediation of important mechanisms, including tumor initiation and proliferation, apoptosis, migration, invasion, chemoresistance, and stem cell maintenance.
Article
Mitochondria are the cytoplasmic organelles mostly known as the “electric engine” of the cells; however, they also play pivotal roles in different biological processes, such as cell growth/apoptosis, Ca²⁺ and redox homeostasis, and cell stemness. In cancer cells, mitochondria undergo peculiar functional and structural dynamics involved in the survival/death fate of the cell. Cancer cells use glycolysis to support macromolecular biosynthesis and energy production (“Warburg effect”); however, mitochondrial OXPHOS has been shown to be still active during carcinogenesis and even exacerbated in drug-resistant and stem cancer cells. This metabolic rewiring is associated with mutations in genes encoding mitochondrial metabolic enzymes (“oncometabolites”), alterations of ROS production and redox biology, and a fine-tuned balance between anti-/proapoptotic proteins. In cancer cells, mitochondria also experience dynamic alterations from the structural point of view undergoing coordinated cycles of biogenesis, fusion/fission and mitophagy, and physically communicating with the endoplasmic reticulum (ER), through the Ca²⁺ flux, at the MAM (mitochondria-associated membranes) levels. This review addresses the peculiar mitochondrial metabolic and structural dynamics occurring in cancer cells and their role in coordinating the balance between cell survival and death. The role of mitochondrial dynamics as effective biomarkers of tumor progression and promising targets for anticancer strategies is also discussed.
Article
Background The proposed central role of cancer stem cells (CSCs) in tumor development has been extended to explain the diverse oncologic phenomena such as multidrug resistance, metastasis and tumor recurrence in clinics. Due to the enhanced expression of ATP-binding cassette transporters and anti-apoptotic factors, stagnation on G0 phase and the strong ability of self-renewal, the CSCs were highly resistant to clinical anticancer drugs. Therefore, the discovery of new drug candidates that could effectively eradicate cancer stem cells afforded promising outcomes in cancer therapy. Introduction Natural products and their synthetic analogues are a rich source of biologically active compounds and several of them have already been recognized as potent CSCs killers. We aim to provide a collection of recently identified natural products that suppressed the survival of the small invasive CSC populations and combated the drug resistance of these cells in chemotherapy. Results and Conclusion These anti-CSCs natural products included flavonoids, stilbenes, quinones, terpenoids, polyketide antibiotics, steroids and alkaloids. In the present review, we highlighted the therapeutic potential of natural products and their derivatives against the proliferation and drug resistance of CSCs, their working mechanisms and related structure-activity relationships. Meanwhile, in this survey, several natural products with diverse cellular targets such as the naphthoquinone shikonin and the stilbene resveratrol were characterized as promising lead compounds for future development.
Article
Full-text available
Cancer stem cells (CSCs) have been proposed to be responsible for tumor recurrence, distant metastasis and drug-resistance, in the vast majority of cancer patients. Therefore, there is an urgent need to identify new drugs that can target and eradicate CSCs. To identify new molecular targets that are unique to CSCs, we previously compared MCF7 2D-monolayers with 3D-mammospheres, which are enriched in CSCs. We observed that 25 mitochondrial-related proteins were >100-fold over-expressed in 3D-mammospheres. Here, we used these 25 proteins to derive short gene signatures to predict distant metastasis (in N=1,395 patients) and tumor recurrence (in N=3,082 patients), by employing a large collection of transcriptional profiling data from ER(+) breast cancer patients. This analysis resulted in a 4-gene signature for predicting distant metastasis, with a hazard ratio of 1.91-fold (P=2.2e-08). This provides clinical evidence to support a role for CSC mitochondria in metastatic dissemination. Next, we employed a panel of mitochondrial inhibitors, previously shown to target mitochondria and selectively inhibit 3D-mammosphere formation in MCF7 cells and cell migration in MDA-MB-231 cells. Remarkably, these five mitochondrial inhibitors had only minor effects or no effect on MDA-MB-231 tumor formation, but preferentially and selectively inhibited tumor cell metastasis, without causing significant toxicity. Mechanistically, all five mitochondrial inhibitors have been previously shown to induce ATP-depletion in cancer cells. Since 3 of these 5 inhibitors were designed to target the large mitochondrial ribosome, we next interrogated whether genes encoding the large mitochondrial ribosomal proteins (MRPL) also show prognostic value in the prediction of distant metastasis in both ER(+) and ER(-) breast cancer patients. Interestingly, gene signatures composed of 6 to 9 MRPL mRNA-transcripts were indeed sufficient to predict distant metastasis, tumor recurrence and Tamoxifen resistance. These gene signatures could be useful as companion diagnostics to assess which patients may benefit most from anti-mito-ribosome therapy. Overall, our studies provide the necessary proof-of-concept, and in vivo functional evidence, that mitochondrial inhibitors can successfully and selectively target the biological process of cancer cell metastasis. Ultimately, we envision that mitochondrial inhibitors could be employed to develop new treatment protocols, for clinically providing metastasis prophylaxis, to help prevent poor clinical outcomes in cancer patients.
Article
Full-text available
Ring-fused diphenylchlorins as potent low-dose photosensitizers for photodynamic therapy of bladder carcinoma and esophageal adenocarcinoma are described. All studied molecules were very active against HT1376 urinary bladder carcinoma and OE19 esophageal adenocarcinoma cell lines, showing IC50 values below 50 nM. The in vivo evaluation of the more promising photosensitizer, using an OE19 tumor/chick embryo chorioallantoic membrane model, showed a tumor weight regression of 33% with a single photodynamic therapy treatment with the photosensitizer dose as low as 37 ng/embryo.
Article
Full-text available
Radiotherapy (RT) is part of standard cancer treatment. Innovations in treatment planning and increased precision in dose delivery have significantly improved the therapeutic gain of radiotherapy but are reaching their limits due to biologic constraints. Thus, a better understanding of the complex local and systemic responses to RT and of the biological mechanisms causing treatment success or failure is required if we aim to define novel targets for biological therapy optimization. Moreover, optimal treatment schedules and prognostic biomarkers have to be defined for assigning patients to the best treatment option. The complexity of the tumor environment and of the radiation response requires extensive in vivo experiments for the validation of such treatments. So far in vivo investigations have mostly been performed in time-and cost-intensive murine models. Here we propose the implementation of the chick chorioallantoic membrane (CAM) model as a fast, cost-efficient model for semi high-throughput preclinical in vivo screening of the modulation of the radiation effects by molecularly targeted drugs. This review provides a comprehensive overview on the application spectrum, advantages and limitations of the CAM assay and summarizes current knowledge of its applicability for cancer research with special focus on research in radiation biology and experimental radiation oncology.
Article
Full-text available
We have previously demonstrated that carnosol, a naturally occurring diterpene, inhibited in vitro cell viability and colony growth, as well as induced cell cycle arrest, autophagy and apoptosis in human triple negative breast cancer (TNBC) cells. In the present study, we evaluated the ability of carnosol to inhibit tumor growth and metastasis in vivo. We found that non-cytotoxic concentrations of carnosol inhibited the migration and invasion of MDA-MB-231 cells in wound healing and matrigel invasion assays. Furthermore, gelatin zymography, ELISA, and RT-PCR assays revealed that carnosol inhibited the activity and downregulation the expression of MMP-9. Mechanistically, we demonstrated that carnosol suppressed the activation of STAT3 signaling pathway through a ROS-dependent targeting of STAT3 to proteasome-degradation in breast cancer cells (MDA-MB-231, Hs578T, MCF-7, and T47D). We show that blockade of proteasome activity, by MG-132 and bortezomib, or ROS accumulation, by N-acetylcysteine (NAC), restored the level of STAT3 protein. In addition, using chick embryo tumor growth assay, we showed that carnosol significantly and markedly suppressed tumor growth and metastasis of breast cancer xenografts. To the best of our knowledge, this is the first report which shows that carnosol specifically targets signal transducer and activator of transcription 3 (STAT3) for proteasome degradation in breast cancer. Our study further provide evidence that carnosol may represent a promising therapeutic candidate that canmodulate breast cancer growth and metastasis.
Article
Full-text available
Here, we provide the necessary evidence that mitochondrial metabolism drives the anchorage-independent proliferation of CSCs. Two human breast cancer cell lines, MCF7 [ER(+)] and MDA-MB-468 (triple-negative), were used as model systems. To directly address the issue of metabolic heterogeneity in cancer, we purified a new distinct sub-population of CSCs, based solely on their energetic profile. We propose the term “energetic” cancer stem cells (e-CSCs), to better describe this novel cellular phenotype. In a single step, we first isolated an auto-fluorescent cell sub-population, based on their high flavin-content, using flow-cytometry. Then, these cells were further subjected to a detailed phenotypic characterization. More specifically, e-CSCs were more glycolytic, with higher mitochondrial mass and showed significantly elevated oxidative metabolism. e-CSCs also demonstrated an increased capacity to undergo cell cycle progression, as well as enhanced anchorage-independent growth and ALDH-positivity. Most importantly, these e-CSCs could be effectively targeted by treatments with either (i) OXPHOS inhibitors (DPI) or (ii) a CDK4/6 inhibitor (Ribociclib). Finally, we were able to distinguish two distinct phenotypic sub-types of e-CSCs, depending on whether they were grown as 2D-monolayers or as 3D-spheroids. Remarkably, under 3D anchorage-independent growth conditions, e-CSCs were strictly dependent on oxidative mitochondrial metabolism. Unbiased proteomics analysis demonstrated the up-regulation of gene products specifically related to the anti-oxidant response, mitochondrial energy production, and mitochondrial biogenesis. Therefore, mitochondrial inhibitors should be further developed as promising anti-cancer agents, to directly target and eliminate the “fittest” e-CSCs. Our results have important implications for using e-CSCs, especially those derived from 3D-spheroids, (i) in tumor tissue bio-banking and (ii) as a new cellular platform for drug development.
Article
Full-text available
Here, we employed a "senolytic" assay system as a screening tool, with the goal of identifying and repurposing FDA-approved antibiotics, for the targeting of the senescent cell population. Briefly, we used two established human fibroblast cell lines (MRC-5 and/or BJ) as model systems to induce senescence, via chronic treatment with a DNA-damaging agent, namely BrdU (at a concentration of 100 μM for 8 days). Cell viability was then monitored by using the SRB assay, to measure protein content. As a consequence of this streamlined screening strategy, we identified Azithromycin and Roxithromycin as two novel clinically-approved senolytic drugs. However, Erythromycin - the very closely-related parent compound - did not show any senolytic activity, highlighting the dramatic specificity of these interactions. Interestingly, we also show that Azithromycin treatment of human fibroblasts was indeed sufficient to strongly induce both aerobic glycolysis and autophagy. However, the effects of Azithromycin on mitochondrial oxygen consumption rates (OCR) were bi-phasic, showing inhibitory activity at 50 μM and stimulatory activity at 100 μM. These autophagic/metabolic changes induced by Azithromycin could mechanistically explain its senolytic activity. We also independently validated our findings using the xCELLigence real-time assay system, which measures electrical impedance. Using this approach, we see that Azithromycin preferentially targets senescent cells, removing approximately 97% of them with great efficiency. This represents a near 25-fold reduction in senescent cells. Finally, we also discuss our current results in the context of previous clinical findings that specifically document the anti-inflammatory activity of Azithromycin in patients with cystic fibrosis - a genetic lung disorder that results in protein mis-folding mutations that cause protein aggregation.
Article
Full-text available
Background and objectives: Cancer stem cells (CSCs) have been implicated in tumor initiation, recurrence, metastatic spread and poor survival in multiple tumor types, breast cancers included. CSCs selectively overexpress key mitochondrial-related proteins and inhibition of mitochondrial function may represent a new potential approach for the eradication of CSCs. Because mitochondria evolved from bacteria, many classes of FDA-approved antibiotics, including doxycycline, actually target mitochondria. Our clinical pilot study aimed to determine whether short-term pre-operative treatment with oral doxycycline results in reduction of CSCs in early breast cancer patients. Methods: Doxycycline was administered orally for 14 days before surgery for a daily dose of 200 mg. Immuno-histochemical analysis of formalin-fixed paraffin-embedded (FFPE) samples from 15 patients, of which 9 were treated with doxycycline and 6 were controls (no treatment), was performed with known biomarkers of “stemness” (CD44, ALDH1), mitochondria (TOMM20), cell proliferation (Ki67, p27), apoptosis (cleaved caspase-3), and neo-angiogenesis (CD31). For each patient, the analysis was performed both on pre-operative specimens (core-biopsies) and surgical specimens. Changes from baseline to post-treatment were assessed with MedCalc 12 (unpaired t-test) and ANOVA. Results: Post-doxycycline tumor samples demonstrated a statistically significant decrease in the stemness marker CD44 (p-value < 0.005), when compared to pre-doxycycline tumor samples. More specifically, CD44 levels were reduced between 17.65 and 66.67%, in 8 out of 9 patients treated with doxycycline. In contrast, only one patient showed a rise in CD44, by 15%. Overall, this represents a positive response rate of nearly 90%. Similar results were also obtained with ALDH1, another marker of stemness. In contrast, markers of mitochondria, proliferation, apoptosis, and neo-angiogenesis, were all similar between the two groups. Conclusions: Quantitative decreases in CD44 and ALDH1 expression are consistent with pre-clinical experiments and suggest that doxycycline can selectively eradicate CSCs in breast cancer patients in vivo. Future studies (with larger numbers of patients) will be conducted to validate these promising pilot studies.
Article
Full-text available
Here, we wish to propose a new systematic approach to cancer therapy, based on the targeting of mitochondrial metabolism, especially in cancer stem cells (CSCs). In the future, we envision that anti-mitochondrial therapy would ultimately be practiced as an add-on to more conventional therapy, largely for the prevention of tumor recurrence and cancer metastasis. This mitochondrial based oncology platform would require a panel of FDA-approved therapeutics (e.g. Doxycycline) that can safely be used to inhibit mitochondrial OXPHOS and/or biogenesis in CSCs. In addition, new therapeutics that target mitochondria could also be developed, to optimize their ability to eradicate CSCs. Finally, in this context, mitochondrial-based biomarkers (i.e. “Mito-signatures”) could be utilized as companion diagnostics, to identify high-risk cancer patients at diagnosis, facilitating the early detection of tumor recurrence and the prevention of treatment failure. In summary, we suggest that new clinical trials are warranted to test and possibly implement this emerging treatment strategy, in a variety of human cancer types. This general approach, using FDA-approved antibiotics to target mitochondria, was effective in killing CSCs originating from many different cancer types, including DCIS, breast (ER(+) and ER(-)), prostate, ovarian, lung and pancreatic cancers, as well as melanoma and glioblastoma, among others. Thus, we propose the term MITO-ONC-RX, to describe this anti-mitochondrial platform for targeting CSCs. The use of re-purposed FDA-approved drugs will undoubtedly help to accelerate the clinical evaluation of this approach, as these drugs can move directly into Phase II clinical trials, saving considerable amounts of time (10–15 y) and billions in financial resources.
Article
Full-text available
The microbiota consists of a dynamic multispecies community of bacteria, fungi, archaea, and protozoans, bringing to the host organism a dowry of cells and genes more numerous than its own. Among the different non-sterile cavities, the human gut harbors the most complex microbiota, with a strong impact on host homeostasis and immunostasis, being thus essential for maintaining the health condition. In this review, we outline the roles of gut microbiota in immunity, starting with the background information supporting the further presentation of the implications of gut microbiota dysbiosis in host susceptibility to infections, hypersensitivity reactions, autoimmunity, chronic inflammation, and cancer. The role of diet and antibiotics in the occurrence of dysbiosis and its pathological consequences, as well as the potential of probiotics to restore eubiosis is also discussed.
Article
Standard chemotherapies that interfere with microtubule dynamics are a chemotherapeutic option used for the patients with advanced malignancies that invariably relapse after targeted therapies. However, major efforts are needed to reduce their toxicity, optimize their efficacy, and reduce cancer chemoresistance to these agents. We previously identified a pyrrolo[2,3d]pyrimidine-based microtubule-depolymerizing agent (PP-13) that binds to the colchicine site of β-tubulin and exhibits anticancer properties in solid human cancer cells, including chemoresistant subtypes. Here, we investigated the therapeutic potential of PP-13 in vitro and in vivo. PP-13 induced a mitotic blockade and apoptosis in several cancer cells cultured in two-dimensions or three-dimensions spheroids, in conjunction with reduced cell proliferation. Capillary-like tube formation assays using HUVECs showed that PP-13 displayed antiangiogenic properties. It also inhibited cancer cell motility and invasion, in in vitro wound-healing and transwell migration assays. Low concentration PP-13 (130 nmol.L-1) treatment significantly reduced the metastatic invasiveness of human cancer cells engrafts on chicken chorioallantoic membrane. In nude mice, 0.5 or 1 mg.kg-1 PP-13 intraperitoneally administered three-times a week reduced the sizes of paclitaxel-refractory orthotopic breast tumors, delayed the progression of metastasis, and decreased the global metastatic load compared to 0.5 mg.kg-1 paclitaxel or vehicle alone. PP-13 did not show any apparent early adverse effect in vivo. These data suggest that PP-13 is a promising alternative to standard chemotherapy in antimitotic drug-refractory tumors, especially through its impact on metastasis.