ArticlePDF Available

Regrowth Concentration Zero (RC0) as Complementary Endpoint Parameter to Evaluate Compound Candidates During Preclinical Drug Development for Cancer Treatment

Authors:
  • National University of San Martín

Abstract

The screening process for potential anticancer drugs involvesexpensive and time consuming preclinical and clinicaltrials (CT) before a drug is approved for clinical use(CU). At present, there is a “bottleneck” at the CT/CUtransition because many drugs that showed promising resultsduring preclinical research did not pass clinical trials.We speculated that the endpoint parameters (the inhibitoryconcentration 50 (IC50) or lethal concentration100 (CL100)) commonly used in proliferation assays forshort-term periods (24-72 h) are not useful to predict theantiproliferative effect in vivo, especially during clinicaltrials. We propose the use of a parameter, regrowth concentration0 (RC0), which will define the concentrationand time necessary to kill 100 % of the cells and preventregrowth when drug is removed. The RC0 might introducea new bottleneck at the preclinical stage, “preclinical bottleneck”,that will select for drugs with more chances to passclinical trials and improve the success rate of anticancerscreening programs. Our proposal is supported by experimentsdone with the DBTRG-05MG human glioma celllines exposed to short and long-term incubation with threedifferent DNA replication inhibitors (aphidicolin, hydroxyureaand etoposide) and retrospective analysis of clinicaltrials for these drugs.
O
M
I
C
S
P
u
b
l
i
s
h
i
n
g
G
r
o
u
p
ISSN:1948-5956
This is an open-access article distributed under the terms of the Creative Commons
Attribution License, which permits unrestricted use, distribution, and reproduction
in any medium, provided the original author and source are credited.
Journal of
Cancer Science & Therapy
The International Open Access
Journal of Cancer Science & Therapy
Executive Editors
Harvey I. Pass, MD
Department of Cardiothoracic Surgery
New York University Langone Medical Center (USA)
William E. Grizzle, MD, PhD
UAB Comprehensive Cancer Center
Birmingham, USA
Peter E. Barker, PhD
National Institute of Standards and Technology
Gaithersburg, MD, USA
Dan Crichton, MS
Jet Propulsion Laboratory (JPL)
JPL/Caltech, CA, USA
Sudhakar Akulapalli, PhD
Boys Town National Research Hospital, Omaha, USA
Available online at
OMICS Publishing Group
www.omicsonline.org
This article was originally published in a journal published by OMICS Publishing
Group, and the attached copy is provided by OMICS Publishing Group for the
author’s benefit and for the benefit of the authors institution, for
commercial/research/educational use including without limitation use in instruc-
tion at your institution, sending it to specific colleagues that you know, and
providing a copy to your institution’s administrator.
All other uses, reproduction and distribution, including without limitation
commercial reprints, selling or licensing copies or access, or posting on open
internet sites, your personal or institution’s website or repository, are requested
to cite properly.
Digital Object Identifier: http://dx.doi.org/10.4172/1948-5956.1000003
J Canc Sci Ther Volume 1(1) : 019-024 (2009) - 019
ISSN:1948-5 956 JCST, an open access journal
Research Article OPEN ACCESS Freely available online
doi:10.4172/1948-5956.1000003
*Corresponding author: Yakisich J. Sebastian, Department of Clinical
Neuroscience, Huddinge Division of Neurology, Karolinska Institute,
Huddinge University Hospital, Sweden Hospital, S-141 86, Huddinge,
Sweden, Tel: +46 8 585 89 533; Fax: +46 8 585 83810; E-mail:
Sebastian.Yakisich@ki.se
Received November 01, 2009; Accepted November 25, 2009; Pub-
lished November 25, 2009
Citation: Avramidis D, Cruz M, Sidén Å, Tasat DR, Yakisich JS (2009)
Regrowth Concentration Zero (RC0) as Complementary Endpoint Pa-
rameter to Evaluate Compound Candidates During Preclinical Drug De-
velopment for Cancer Treatment . J Canc Sci Ther 1: 019-024.
doi:10.4172/1948-5956.1000003
Copyright: © 2009 Avramidis D, et al. This is an open-access article
distributed under the terms of the Creative Commons Attribution License,
which permits unrestricted use, distribution, and reproduction in any
medium, provided the original author and source are credited.
Abstract
The screening process for potential anticancer drugs in-
volves expensive and time consuming preclinical and clini-
cal trials (CT) before a drug is approved for clinical use
(CU). At present, there is a “bottleneck” at the CT/CU
transition because many drugs that showed promising re-
sults during preclinical research did not pass clinical tri-
als. We speculated that the endpoint parameters (the in-
hibitory concentration 50 (IC
50
) or lethal concentration
100 (CL
100
)) commonly used in proliferation assays for
short-term periods (24-72 h) are not useful to predict the
antiproliferative effect in vivo, especially during clinical
trials. We propose the use of a parameter, regrowth con-
centration 0 (RC
0
), which will define the concentration
and time necessary to kill 100 % of the cells and prevent
regrowth when drug is removed. The RC
0
might introduce
a new bottleneck at the preclinical stage, “preclinical bottle-
neck”, that will select for drugs with more chances to pass
clinical trials and improve the success rate of anticancer
screening programs. Our proposal is supported by experi-
ments done with the DBTRG-05MG human glioma cell
lines exposed to short and long-term incubation with three
different DNA replication inhibitors (aphidicolin, hydrox-
yurea and etoposide) and retrospective analysis of clinical
trials for these drugs.
Regrowth Concentration Zero (RC
0
) as Complementary End-
point Parameter to Evaluate Compound Candidates During
Preclinical Drug Development for Cancer Treatment
Avramidis Dimitrios
1
, Cruz Mabel
1
, Sidén Åke
1
, Tasat Deborah Ruth
2
, Yakisich J. Sebastian
1
*
1
Department of Clinical Neuroscience, Huddinge Division of Neurology, Karolinska Institute, Huddinge University Hospital, Sweden
2
Universidad Nacional de San Martín Buenos Aires, Argentina
Keywords:
Drug screening; DNA replication; Glioma;
Aphidicolin; Hydroxyurea; Etoposide
Introduction
In general, the standard approach to evaluate novel compounds
for cancer treatment after drug synthesis or discovery is based
in preclinical testing and clinical trials (Figure 1 top). The pre-
clinical phase involves in vitro as well as in vivo research. The
entire process is very expensive and time consuming (DiMasi
et al., 2003; Emanuel et al., 2003). Commonly, in vitro experi-
ments for determining the antiproliferative effects of a poten-
tial antineoplastic drug are done either in cell free systems or in
cell lines by means of short term proliferation assays which
measure the incorporation of tritiated thymidine or BrDU into
DNA or mass cell by colorimetric methods (e.g. MTT assay).
In these assays, cell line(s) are tested against a broad range of
drug concentrations typically for 48-72 h and the results are
usually reported as a plot and the IC
50
and LC
100
(See Glossary)
are calculated by interpolation (Brown, 1997; Iljin et al., 2009).
For instance, in the NCI60 human tumor cell line anticancer
drug screen program, the GI50 (50% growth inhibition) and
LC50 (50% lethal concentration) are derived from concentration-
response curves by linear interpolation while the TGI (total
growth inhibition) is read as the x-axis intercept (Shoemaker,
2006). The main pitfall of these assays is that the short term
incubation is not enough to determine the minimum concentra-
tion of the drug that actually kill 100 % of the cells preventing
regrowth when the drug is removed from the culture. In prac-
tice, the continuous growth of the untreated cells (control) and
cells exposed to low concentration of the drug limits the assay
because of loss of linearity over time. Long term survival for
screening purposes has been assessed by the tumor colony-form-
ing assay on a moderate scale due to technical limitations (Shoe-
maker et al., 1985 ). When used, researchers often report the
IC
50
for the tested drug (Sasaki et al., 2008). After the optimal
drug concentration (e.g. IC
50
or LC
100
) is determined, the next
step involves testing the compound in animal models in vivo
where a tumour is induced by injecting cancer cells into spe-
cific organs. The drug is administered in control and experi-
mental groups and the antineoplastic effect is usually assessed
by tumor growth and/or survival rates (Kaplan-Meier plots).
Due to ethical considerations, animals are usually sacrificed after
few weeks and long term relapses are not evaluated after treat-
ment discontinuation. In case the compound shows promising
in vivo effect on tumor growth, acceptable side effects and tox-
icity, the drug is considered a good candidate to be tested in
clinical trials. These stages are associated with a significant
percentage of the total cost of the entire drug evaluation pro-
cess (DiMasi et al., 2003; Emanuel et al., 2003). There are several
examples of newly developed compounds that might be consid-
ered for clinical trials (Roth et al., 2009; Xu et al., 2009; Yakisich et
al., 2009 ) but at present it is difficult to estimate the chances that
they will be successful. Unfortunately, when promising drug
candidates are tested in clinical trials, the majority of them fail. In
most cases, the outcome has been disappointing and sometimes
Journal of Cancer Science & Therapy
- Open Access
www.omicsonline.org JCST/Vol.1 Issue 1
J Canc Sci Ther Volume 1(1) : 019-024 (2009) - 020
ISSN:1948-5 956 JCST, an open access journal
the trial terminated early due to evident failure (Robe et al., 2009).
This problem was recognized by Brown (Brown, 1997) who re-
ported that the popular NCI’s anticancer drug screening pro-
gram does not select for clinically active compounds, but no
potential solution or important changes have been implemented
to solve the problem.
Our hypothesis is that the high rate of failure when translating
preclinical drug screening into successful clinical trials is due to
the use of the IC
50
(and less common LC
100
) as guiding concen-
tration for mechanistic studies as well as goal for clinical trials.
At present, current drug development programs have a “bottle-
neck” at the “clinical trial”/”clinical use” transition, where the
success rate is low (Figure 1 Top). It is estimated that only 5% of
cancer drugs entering clinical trials reach marketing approval
(Collins and Workman, 2006). In this paper, success rate (SR) is
defined as the ratio between the number of drugs approved for
clinical use (n
CU
) and the number of drugs that enter the clinical
trial stage (n
CT
). Conceptually the SR (SR=n
CU
/n
CT
) is similar to
the “clinical approval success rate” defined as “the probability
that a compound that enters the clinical testing pipeline will
eventually be approved for marketing” (DiMasi et al., 2003). In
some cases (e.g. roscovitine as drug candidate for brain tumours),
the concentration that actually reach the target tissue is main-
tained at levels that are below the IC
50
reported in vitro and
might limit the clinical use (Yakisich et al., 2009 ). Moreover, even
if, in the target tissue, concentrations similar to the IC
50
are
reached, one can anticipate that a fraction of cells will survive
and resume cell proliferation when the drug is removed. Interest-
ingly, in a study measuring in vitro chemo sensitivity in patient-
derived cell lines by a short-term assay (24-72 h), the ID
50
(de-
fined as the dose of the drug that inhibited protein synthesis by
50 %), was used to classified between responder and non-re-
sponder patients. The relapse-free interval in patients that
showed in vitro chemo sensitivity (responders) was modestly
increased compared to patients designated as non-responder
(Thomas et al., 1985).
Increasing the number of clinical trials by simply screening
more drugs, without any additional rational guidance, will cre-
ate an unproductive “financial bottleneck” at the “preclinical/
clinical trial” transition due to the astronomical cost associated
with the clinical trials. Due to this “financial bottleneck” some
drugs that might be of clinical relevance, will probably never
be tested while other that enter the clinical trial stage will even-
tually fail at the same rate (keeping the 5 % rate success con-
stant).
In this paper we propose that the success rate and cost benefit
for drug development could be improved by introducing a
“bottleneck” during the preclinical stage (“preclinical bottle-
neck”) using the so called RC
0
(see glossary) as endpoint pa-
rameter instead of IC
50
or LC
100
(Figure 1 Bottom). The ratio-
nale of our proposal is supported by a series of experiments
using prolonged exposure to three classical DNA replication
inhibitors with different mechanism of action (aphidicolin, hy-
droxyurea and etoposide) in the human DBTRG-05MG glioma
cell line and a retrospective analysis of clinical trials with these
same drugs.
Materials and Methods
Cell lines
Stock cultures of human DBTRG-05MG glioma cell line were
obtained from the European Collection of Cell Culture
(ECACC). Cells were routinely cultured in RPMI-1640 medium
supplemented with 10 % Foetal Bovine Serum (FBS), 2 mM
glutamine, 1 % HT and 1 mM sodium pyruvate (complete me-
dia) at 37 ºC in a humidified atmosphere containing 5% CO
2
.
Drugs
Hydroxyurea, Aphidicolin and Etoposide (Sigma, Sweden)
were prepared as stock solutions (1 M in H
2
O, 2.5 mM in DMSO
and 25 mM in DMSO respectively) and stored at -20
°
C until use.
Fresh dilutions in culture media were prepared just before use.
Antiproliferative assay
DBTRG-05MG cells were plated in 96- well flat bottom plates at
5000 cells/well and were allowed to adhere overnight. Then, the
cultures were exposed to a concentration range of the three drugs
selected and control cultures where treated with the equivalent
concentration of the corresponding vehicle (DMSO or H
2
O).
After 72 h, cell growth was monitored using the CCK kit (Sigma,
Sweden) according to supplier instructions. For long-term
antiproliferative assays, drugs were maintained in culture for 2-
10 weeks. The media and drugs were changed twice a week and
were found to be sufficient to prevent cell growth (see result)
indicating that the drugs remain active during in vitro prolonged
cultures.
The presence of surviving cells during prolonged exposure
and the (re)growth after drug removal was evaluated using a
routine inverted microscope. Regrowth was defined as the abil-
Citation: Avramidis D, Cruz M, Sidén Å, Tasat DR, Yakisich JS (2009) Regrowth Concentration Zero (RC0) as Complementary
Endpoint Parameter to Evaluate Compound Candidates During Preclinical Drug Development for Cancer Treatment. J Canc Sci Ther
1: 019-024. doi:10.4172/1948-5956.1000003
Figure 1:
Top) Simplified diagram showing the common steps during drug
development from dru g discovery to clinical tri als. After discovery or
synthesis of a new potential antineoplastic drug, the compound undergoes
preclinical research where is tested in cell free systems as well as in viv o in
cell lines and animal models. Promising candidates that pass the preclinical
stage are approved for clinical trials and few reach the “clinical use” stage.
At the “clinical trial”/”drug use” transition the presence of a “bottleneck
limits the success rate. The success rate (defined as the number of drugs
that reach the stage of clinical use divided by t he number of drugs that
enter the number of drugs clinical trial; n
CU
/n
CT
) , value between 0-1, is an
indication of the cost/benefit of the program.
B) Proposed outcome of drug development by introducing a “preclinical
bottleneck” by using the RC
0
as endpoint parameter during preclinical
research. The presence of the “preclinical bottleneck” will reduce the
number of cells entering the “clinical trial stage” and might increase the
success rate improving the cost/benefit.
Preclinical
Cell Free Systems
Discovery
or
Synthesis
Discovery
or
Synthesis
Cell lines/Animal models
Clinical trials
(CT)
“Bottleneck”
“Bottleneck”
“Preclinical Bottleneck”
Clinical Use
(CU)
SR= n/n
CU CT
SR= n/n
CU CT
J Canc Sci Ther Volume 1(1) : 019-024 (2009) - 021
ISSN:1948-5 956 JCST, an open access journal
ity of surviving cells to form a monolayer after 1-2 weeks of
incubation with drug-free media.
Results
We evaluated the short term antiproliferative effect of hydrox-
yurea (HU), aphidicolin (Aph) and etoposide (Et) on DBTRG-
05MG cells in culture. Exponentially growing cells were incu-
bated with different concentrations of each drug and cell prolif-
eration was measured at 72 h using the CCK kit assay. All three
drugs tested showed a concentration-dependent inhibitory ef-
fect reaching the maximum effect at 2.5 µM, 10 mM and 50 µM for
Aph, HU and Et respectively (Figure 2). The IC
50
was estimated
by interpolation as 0.9 µM, 0.75 mM and 0.9 µM for Aph, HU and
Et respectively.
Long-term incubation (4 weeks) with concentrations > IC
50
(Aph, 2.5 µM; HU, 10 M; Et, 5 µM), showed a subpopulation of
cells resistant to these drug concentrations. Microscopic exami-
nation revealed that a small fraction of cells remained attached
to the surface. When the drugs were removed from the culture,
the surviving cells were able to resume cell division and, they
proliferate reaching a monolayer morphologically indistinguish-
able from the original culture. To prevent cell regrowth higher
concentrations of HU (50 mM) or Et (> 25 µM) for at least one
week were required.
Dis cussion
We used the human glioma cell line DBTRG-05MG as an ex-
perimental system to retrospectively analyze the failure of sev-
eral drugs that inhibit in vitro cell proliferation of cancer cell
lines during clinical trials. Gliomas are the most common primary
brain tumours and remain poorly responsive to multimodality
therapeutic interventions, including surgery, radiotherapy, and
chemotherapy. The highly proliferative activity of glioma cells
compared to normal brain makes DNA replication an interesting
target for therapeutic purposes. In this paper, we evaluated the
outcome of DBTRG-05MG glioma cells exposed to three differ-
ent DNA replication inhibitors for short and long-term expo-
sures (Figure 2, Figure 3).
Aphidicolin: The in vitro IC
50
for aphidicolin has been deter-
mined in cell free systems as 0.5, 0.9 and 5.8 µM for polymerase
α, δ, and ε respectively (Wright et al., 1994) without affecting
dNTP pools (Sheaff et al., 1991). Due to the poor solubility and
because pharmacologically active levels had not been achieved
for aphidicolin, it was decided early (after two phase I studies)
to stop further evaluation (Beijnen et al., 1995). Therefore,
aphidicolin represents a good example of a potential anticancer
drug that reached the stage of clinical trials and its use for can-
cer treatment was stopped early based in its pharmacokinetic
properties.
Hydroxyurea: In our study, the IC
50
for DBTRG-05MG cells
was 0.5 mM (Figure 2). Pharmacological studies showed that
the level of HU after a single dose of 1200 mg can reach only 0.04
mM and 0.26 mM in cerebrospinal fluid and plasma respectively
(Gwilt et al., 2003). DBTRG-05MG cells were able to resist pro-
longed treatments (> 4 weeks) with 10 mM while higher concen-
trations (50 mM) were required to prevent regrowth. Plasma con-
centration might be enough for treatment of some types of can-
cer, but definitely not for brain tumors. Hydroxyurea alone has
been proven at early stage to be of no use for glioma treatment
and represents an example of a drug that (based only in IC
50
values) could have been stopped at the “preclinical bottleneck”
before engaging in costly and time consuming clinical trials.
Etoposide: Several large numbers of clinical trials and pharma-
cokinetic data showed that a daily oral etoposide dose of 50 mg/
m
2
produces serum concentrations >1 mg/L (~ 1.7 µM) lasting
several hours each day (Hainsworth, 1999). Potentially cyto-
toxic concentration in cerebro spinal fluid (CSF) was achieved
with doses higher than 300 mg/m
2
i.v. ( levels of 0.175 µM; range,
0.066 to 2.12) in children with acute lymphoblastic leukemia
(Relling et al., 1996 ). In our study, the IC
50
for DBTRG-05MG
cells was < 1 µM but the RC
0
was > 25 µM (Figure 3). Even
though at present etoposide alone has been of limited use in
brain tumors, our data predict that clinical trials with etoposide
will continue to fail unless intracerebral levels of > 25 µM are
reached. The use of etoposide at very high doses (800 mg/m
2
) in
combination with autologous bone marrow transplantation in-
crease the CSF levels (similar to IC
50
but not to RC
0
levels) still,
in brain tumor tissue the outcome has been disappointing
(Giannone and Wolff, 1987; Leff et al., 1988 ). Etoposide is an
example of a drug that based in IC
50
values should have been
successful for glioma treatment, but clinical trials proved to be
of very limited use (Finn et al., 1985; Fulton et al., 1996; Tirelli et
Journal of Cancer Science & Therapy
- Open Access
www.omicsonline.org JCST/Vol.1 Issue 1
Figure 2:
Antiproliferative effect of short term incubation with Hydroxyurea, Aphidicolin or Etoposide. DBTRG-05MG glioma cells were incubated with
the indicated concentra tion of drugs during 72 h. Cell proliferation was measured by the CCK kit. Results are representative of three independent
experiments performed by quadruplicates.
Survival (%)
Aphidicolin (µM) Etoposide (µM)
Hydroxyurea (mM)
120
100
80
60
40
20
0
0 0 (D) 1 2,5 5 10 25 0 1 5 10 0 1 10 50 100 250
J Canc Sci Ther Volume 1(1) : 019-024 (2009) - 022
ISSN:1948-5 956 JCST, an open access journal
al., 1984). If the RC
0
have been used as guide, etoposide as single
agents would have never been attempted saving cost and time.
Instead, etoposide would have been used from the beginning in
combinational therapy with other drugs.
The three drugs we evaluated have RC
0
much higher than IC
50
and retrospectively demonstrate that clinical trials have poor
outcome when the drug concentration reach similar or just
slightly higher values than the IC
50
. In m-AMSA treated cultures
at LD
80
concentration, a fraction of stem cells survive prolonged
exposure (Sabisz and Skladanowski, 2009). m-AMSA is a
topoisomerase II inhibitor that also works as an alkylating agent,
suggesting that our study can be extrapolated to other drug
types. Thus, classical end-point proliferation parameters used
to evaluate drug effects (IC
50
, LC
100
) when used in short term
assays, are insufficient for preclinical testing since they do not
predict long term effects. This is due to the fact that a small
fraction of cells may survive prolonged exposure to similar or
slightly higher concentrations than the IC
50
. The surviving cells,
when the drug is removed from the culture will resume cell divi-
sion. At the clinical level, when the chemotherapy regime is
discontinuated, surviving cells will resume cell division leading
to relapse of the tumour. We propose the use of a new parameter,
RC
0
(Regrowth Concentration 0; defined as the lowest concen-
tration that produce 0% regrowth when the drug is removed
from the culture) as a complementary end-point proliferation
parameter. Other authors also proposed that assays that moni-
tor surviving cells should be included during drug screening
(Sabisz and Skladanowski, 2009). The main disadvantage of the
RC
0
is the length of the assay (2-4 weeks) that in turn limits the
number of drugs that can be screened. High throughput assays
such as the NCI60 (Shoemaker, 2006) or similar cell-based screen-
ing methods (Iljin et al., 2009) can in short time, select several
potential anticancer drugs (based in IC
50
values) from compound
Citation: Avramidis D, Cruz M, Sidén Å, Tasat DR, Yakisich JS (2009) Regrowth Concentration Zero (RC0) as Complementary
Endpoint Parameter to Evaluate Compound Candidates During Preclinical Drug Development for Cancer Treatment. J Canc Sci Ther
1: 019-024. doi:10.4172/1948-5956.1000003
libraries. After that, the RC
0
can further help to identify those
drugs with higher chances to succeed in clinical trials from the
initial selection.
The RC
0
provides two types of valuable information: 1) The
necessary concentration to kill 100 % of tumoral cells, 2) the
lapse of time the cells need to be exposed to a certain concentra-
tion. The time factor has been demonstrated to be important. For
instance extended-schedule oral etoposide shows more efficacy
in selected cancers (Hainsworth, 1999). Thus, preclinical in vitro
testing should include long term proliferation assays to deter-
mine the RC
0
. This can simply be done by incubating the cells
for a prolonged period of time (e.g. 2-4 weeks) after that, remove
the drugs and incubate the cells for another 1-2 weeks and deter-
mine the concentration of drug that killed 100% of the cells (No
regrowth after incubation in drug free media).
To be of clinical use, the CR
0
should fulfil two other essential
requirements: a) the drugs should be non toxic (or mild) to
normal cells for the period required to kill all tumoral cells and b)
It should be possible to reach this concentration in the target
tissue. In this context, the data obtained from phase 0 might
constitute and essential step before pursuing more advanced
research (e.g phase I). A rational use of CR
0
in combination with
pharmacokinetic and toxicological studies in healthy individu-
als (Phase 0) might lead to a more successful rate of antineoplas-
tic drugs with clinical relevance.
Since the RC
0
> LC
100
>LC
50
, higher toxicity to normal cells will
be expected, and less number of drugs will fulfil the criteria to be
approved for clinical trials. At a first glance, this will reduce the
number of clinical trials that in turn will reduce the number of
drugs that reach the market. However, by avoiding unnecessary
highly expensive clinical trials with drugs that have little or no
chances to be of clinical use, more drugs can be screened before
the “preclinical blottleneck”. Resources can be put into devel-
oping more specific drugs with RC
0
levels that can be achieved
in target tissue with tolerable toxicity. The RC
0
will eventually
move the drug development “bottleneck” from clinical trials to
preclinical stages. This displacement will eventually produce a
drop in drugs that reach clinical trials but it will select for those
with higher rates of success reducing the high expenses and
time of clinical trials.
Thus, the use of the RC
0
has the potential to increase the
successful rate and maybe, overcome the bottleneck when trans-
lating preclinical research into clinical trials. From the economi-
cal perspective, predicting which drug will successfully pass
clinical trials will have a tremendous impact in the drug industry
by lowering cost and time by stopping clinical trials of high
number of drugs. On the other hand the use of the RC
0
will help
at early stages to decide which drugs have little chance to be
useful for monotherapy but might have use in combinational
regimes.
Glossary
GI
50
: drug concentration that causes a 50% reduction in cell
number in test plates relative to control plates (equivalent to
IC
50
).
LC
100
: drug concentration that causes a 100% reduction in cell
number in test plates relative to control plates (equivalent to
Fig ure 3:
Top) Antiproliferative effect of long -term incubation with
Aphidicolin (Aph, 2.5 µM), Hydroxyurea (HU, 10 mM) or Etoposide (Et,
5 µM) on DBTRG-05MG glioma cells. Exponentially growing cells were
incubated in complete media for 2-3 days (A), exposed for 4 weeks to the
indicated drugs concentrations (B-C). During this period the media and the
drugs were changed twice a week. A clear decrease in the cell density was
observed by microscopic examination indicating extensive cell death (B)
followed by a stable low cell density (C). When the drug was removed, the
surviving cells resumed proliferation and formed a monolayer indicated as
regrowth in (D). Bottom) same as top but cells were incubated with higher
concentrations of HU (50 mM) or Et (25 µM).
A B C D
Complete Media
Proli feration Cell death
Complete Media
Proli feratio n Cell death
Survival
No Survival
Time
Cell # Cell #
Re
Growth
HU (10 mM) or Aph (2,5 µM), Et (5 µM)
HU (50 mM) or Et (> 25 µM)
Relapse
-----------------------------------------------------------------------------
-----------------------------------------------------------------------------
-----------------------------------------------------------------------------
J Canc Sci Ther Volume 1(1) : 019-024 (2009) - 023
ISSN:1948-5 956 JCST, an open access journal
IC
100
). It is usually determined by interpolation from concentra-
tion response curves.
RC
0
: drug concentration that kill 100% of cells preventing re-
growth when cells are incubated in drug free media. In contrast
to LC
100
, RC
0
is determined empirically, not by interpolation.
Disclosure of Potential Conflicts of Interest
None
Acknowledgement
This work was supported by grants from the Swedish Re-
search Council and the Karolinska Institute.
References
1. Beijnen JH, Flora KP, Halbert GW, Henrar RE, Slack JA (1995) CRC/
EORT C/NCI Joint Formulation Working Par ty: experiences in the
formulation of investigational cytotoxic drugs. Br J Cancer 72: 210-
218.
» PubMed » Go ogle Scholar
2. Brown JM (1997) NCI’s anticancer drug screening program may not
be selecting for clinically active compounds. Oncol Res 9: 213-215.
» Pub Med » Google Scholar
3. Collins I, Workman P (2006) New approaches to molecular cancer
therapeutics. Nat Chem Biol 2: 689-700.
» CrossR ef » PubMed » G oogle
Scholar
4. DiMasi JA, Hansen RW, Grabowski H (2003) The price of innovation:
new estimates of drug development costs. J Health Econ 22: 151-185.
» Cross Ref » P ubMed » Google Scholar
5. Emanuel EJ, Schnipper LE, Kamin DY, Levinson J, Lichter AS (2003)
The cost s of conducting clinical research. J Clin Oncol 21: 4145-
4150.
» CrossRef » Pub Med » Google Scholar
6. Finn GP, Bozek T, Souhami RL, Slevin ML, Thomas DG (1985) High-
dose etoposide in the treatment of relapsed prima ry brain tumors.
Cancer Treat Rep 69: 603-605.
» PubMed » Google Scholar
7. Fulton D, Urtasun R, Forsyth P (1996) Phase II study of prolonged
oral therapy with etoposide (VP16) for patients with recurrent malig-
nant glioma. J Neurooncol 27: 149-155 .
» CrossR ef » Pub Med » Google
Scholar
8. Giannone L, Wolff SN (1987) Phase II treatment of central nervous
system gliomas with high-dose etoposide and autologous bone marrow
transplantation. Cancer Treat Rep 71: 759-761.
» Pu bMed » Go ogle Sch olar
9. Gwilt PR, Manouilov KK, McNabb J, Swindells SS (2003) Pharmaco-
kinetics of hydroxyurea in plasma and cerebrospinal fluid of HIV-1-
infected patients. J Clin Pharmacol 43: 1003-1007.
» CrossRef » PubMed
» Go ogle Scho lar
10 . Ha insworth JD (1999) Extended-schedule oral etoposide in selected
neoplasms and overview of administration and scheduling issues. Drugs
58: 51-56.
» CrossRef » Pu bMed » Google Scholar
11 . Iljin K, Ketola K, Vainio P, Halonen P, Kohonen P, et al. (2009) High-
throughput cell-based screening of 4910 known drugs and drug-like
small molecules identifies disulfiram as an inhibitor of prostate cancer
cell growth. Clin Cancer Res 15: 6070-6078.
» Cross Ref » PubMed » Goo gle
Scholar
Journal of Cancer Science & Therapy
- Open Access
www.omicsonline.org JCST/Vol.1 Issue 1
12 . Leff RS, Thompson JM, Daly MB, Johnson DB, Harden EA, et al.
(1988) Acute neurologic dysfunction after high-dose etoposide therapy
for malignant glioma. Cancer 62: 32-35.
» CrossR ef » Pub Med » Go ogle
Scholar
13 . Relling MV, Mahmoud HH, Pui CH, Sandlund JT, Rivera GK, et al.
(1996) Etoposide achieves potentially cytotoxi c concentrations in
CSF of children with acute lymphoblastic leukemia. J Clin Oncol 14:
399-404.
» CrossRef » Pu bMed » Go ogle Scholar
14 . Robe PA, Martin DH, Nguyen-Khac MT, Artesi M, Deprez M, et al.
(2009) Early termination of ISRCTN45828668, a phase 1/2 prospec-
tive, randomized study of Sulfasalazine for the treatment of progress-
ing malignant gliomas in adults. BMC Cancer 9: 372.
» Cross Ref » PubMed
» Go ogle Scho lar
15 . Roth P, Kissel M, Herrmann C, Eisele G, Leban J, et al. (2009) SC68896,
a Novel Small Molecule Proteasome Inhibitor, Exerts Antiglioma
Activity In vitro and In vivo. Clin Cancer Res 15: 6609-18.
» CrossRef
» Pub Med » Google Scholar
16 . Sabisz M, Skladanowski A (2009) Cancer stem cells and escape from
drug-induced premature senescence in human lung tumor cells: impli-
cations for drug resista nce and in vitro drug screening model s. Cell
Cycle 8: 3208-3217.
» Cr ossRef » PubMed » Go ogle Sch olar
17 . Sasaki R, Suzuki Y, Yonezawa Y, Ota Y, Okamoto Y, et al. (200 8)
DNA polymerase gamma inhibition by vitamin K3 induces mitochon-
dria-mediated cytotoxicity in human cancer cells. Cancer Sci 99: 1040-
1048.
» CrossRef » Pub Med » Google Scholar
18 . Sheaff R, Ilsley D, Kuchta R (1991) Mechanism of DNA polymerase
alpha inhibition by aphidicolin. Biochemistry 30: 8590-8597.
» CrossRef
» Pub Med » Google Scholar
19 . Shoemaker RH (2006) The NCI60 human tumour cell line anticancer
drug screen. Nat Rev Cancer 6: 813-823.
» CrossR ef » P ubMed » Goo gle
Scholar
20 . Shoemaker RH , Wolpert-DeF ilippes MK, Kern DH, Lieber MM,
Mak uch RW, et al. (1985) Application of a huma n tumor colony-
forming assay to new drug screening. Cancer Res 45: 2145-2153.
» Cross Ref » P ubMed » Google Scholar
21 . Thomas DG, Darling JL, Paul EA, Mott TJ, Godlee JN, et al. (1985)
Assay of anti-cancer drugs in tissue culture: rela tionship of relapse
free interva l (RFI) and in vitro chemosensitivity in patients wit h
malignant cerebral glioma. Br J Cancer 51: 525-532.
» PubMed » Google
Scholar
22 . Tirelli U, D’Incalci M, Canetta R, Tumolo S, Franchin G, et al. (1984)
Etoposide (VP-16-213) in malignant brain tumors: a phase II study. J
Clin Oncol 2: 432-437.
» CrossRef » PubMed » Go ogle Sch olar
23 . Wright GE, Hübscher U, Khan NN, Focher F, Verri A (1994) Inhibitor
analysis of calf thymus DNA polymerases alpha, delta and epsilon.
FEBS Lett 341: 128-130.
» CrossRef » PubMed » Go ogle Sch olar
24 . Xu H, Wang Z, Donaldson JC, Yao H, Zhou S, et al. (2009) Antitumor
Effi cacy and Molecular Mechanism of TLK58747 , a Novel DNA-
Alkylating Prodrug. Anticancer Res 29: 3845-3855.
» CrossRef » PubMed
» Go ogle Scho lar
25 . Yakisich JS, Vita MF, Siden A, Tasat DR, Cruz M (2009) Strong inhi-
bition of repli cative DNA synthesis in the developing rat cerebral
cortex and glioma cells by roscovitine. Invest New Drugs.
» Cross Ref
» Pub Med » Google Scholar
J Canc Sci Ther Volume 1(1) : 019-024 (2009) - 024
ISSN:1948-5 956 JCST, an open access journal
Citation: Avramidis D, Cruz M, Sidén Å, Tasat DR, Yakisich JS (2009) Regrowth Concentration Zero (RC0) as Complementary
Endpoint Parameter to Evaluate Compound Candidates During Preclinical Drug Development for Cancer Treatment. J Canc Sci Ther
1: 019-024. doi:10.4172/1948-5956.1000003
OMICS Publishing Group
List of Journals
1.Journal of Proteomics & Bioinformatics (JPB) Open Access
2.Journal of Bioequivalence & Bioavailability (JBB) Open Access
3.Journal of Computer Science & Systems Biology (JCSB) Open Access
4.Journal of Antivirals & Antiretrovirals (JAA) Open Access
5.Journal of Data Mining in Genomics & Proteomics (JDMGP) Open Access
6.Journal of Glycomics & Lipidomics (JGL) Open Access
7.Journal of Pharmacogenomics & Pharmacoproteomics(JPP) Open Access
8.Journal of Postgenomics: Drug & Biomarker Development (JPDBD) Open Access
9.Journal of Biochips and Tissue Chips (JBTC) Open Access
10.Journal of Molecular Biomarkers & Diagnosis (JMBD) Open Access
11.Journal of Bioanalysis & Biomedicine (JBABM) Open Access
12.Journal of Nanomedicine & Biotherapeutic Discovery (JNBD) Open Access
13.Journal of Cancer Science & Therapy (JCST) Open Access
14.Journal of Biotechnology & Biomaterials (JBTBM) Open Access
15.Journal of Petroleum & Environmental Biotechnology (JPEB) Open Access
16.Journal of Microbial & Biochemical Technology (JMBT) Open Access
17.Journal of Tissue Sciences & Engineering (JTSE) Open Access
18.Briefings in Intellectual Property Rights (BIPR) Open Access
... In most studies, the fate of surviving cells after short term incubation with drugs is not evaluated. For instance, glioma cells can survive prolonged exposure of antineoplastic drugs at concentrations much higher than the IC 50 [14]. At the clinical level, these surviving cells explain the relapse of tumor when the treatment is discontinued and suggest that drugs that kill 100% of tumoral cells ( " pankillers " ) will be more effective anti-cancer drugs and may, cure cancer. ...
... Stock cultures of human DBTRG-05MG glioma cell line were obtained from the European Collection of Cell Culture (ECACC). Cells were routinely cultured as previously described [14]. Patients-derived glioblastoma cells hGCL1-hGCL8 were kindly provided by Dr Peter Siesjö (Lund University). ...
... The anti-proliferative effect of menadione has been studied in several glioma cell lines and showed IC 50 values ranging between 13.5 μM to ~25 μM [1, 24] . We recently suggested that drug concentrations much higher than the IC 50 might be required to eliminate 100% of tumor cells and that the IC 50 is not the best parameter to evaluate anti-cancer drugs because it does not select for useful clinical drugs [14]. In our study, the IC 50 for the DBTRG.05MG ...
Article
Full-text available
Menadione (Vitamin K3) has anti-tumoral effects against a wide range of cancer cells. Its potential toxicity to normal cells and narrow therapeutic range limit its use as single agent but in combination with radiation or other anti-neoplastic agents can be of therapeutic use. In this paper, we first evaluated the early (within 3 h) effect of menadione on ongoing DNA replication. In normal rat cerebral cortex mini-units menadione showed an age dependent anti-proliferative effect. In tissue mini-units prepared from newborn rats, menadione inhibited ongoing DNA replication with an IC (50) of approximately 10 μM but 50 μM had no effect on mini-units from prepared adult rat tissue. The effect of short (72 h) and prolonged exposure (1-2 weeks) to menadione alone in the DBTRG.05MG human glioma cells line and in combination with vitamin C was studied. After short period of exposure data show that menadione alone or in combination with vitamin C provided similar concentration-response curves (and IC(50) values). Prolonged exposure to these drugs was evaluated by their ability to kill 100% of glioma cells and prevent regrowth when cells are re-incubated in drug-free media. In this long-term assay, menadione:vitamin C at a ratio 1:100 showed higher anti-proliferative activity when compared to each drug alone and allowed to reduce each drug concentration between 2.5 to 5-fold. Similar anti-proliferative effect was demonstrated in 8 patient derived glioblastoma cell cultures. Our data should be able to encourage further advanced studies on animal models to evaluate the potential use of this combination therapy for glioma treatment.
... The prefix " pan " comes from the ancient Greek word πανpan-(all , every). To screen for pankiller drugs we introduced the " Regrowth Concentration zero " parameter (RC 0 ) that defines the minimum drug concentration and time exposure needed to kill 100% of cells [24] in order to prevent cancer relapse. On the other hand, the effect of anticancer drugs on cell proliferation is usually measured by short-term assays, such as the MTT assay [25] [26]. ...
... In order to efficiently monitor the ability of drugs to eliminate all cancer cells, according to our experience and despite the extended use of short-term assays, the screening of anticancer drugs may require other type, of proliferation assays such as long-term proliferation assays. For this purpose , we recently introduced a proliferation assay where cells are subjected to prolonged (between 7 and 14 days) drug exposure [22] [23] [24]. This assay has the potential to detect around 0.01% of resistant cells and is technically easy to perform compared to the clonogenic assay [22] that gives similar information. ...
Article
Full-text available
Exposure of cancer cells to anticancer agents in cultures induces detachment of cells that are usually considered dead. These drug-induced detached cells (D-IDCs) may represent a clinical problem for chemotherapy since they may survive anoikis, enter the circulation, invade other tissues and resume proliferation, creating a metastasis, especially in tissues where the bioavailability of anticancer agents is not enough to eliminate all cancer cells. In this study we evaluated the antiproliferative effect of menadione : sodium orthovanadate (M : SO) combination on A549 lung cancer cells as well as the ability of M : SO to induce cell detachment. In addition, we followed the fate and chemosensitivity of M : SO-induced detached cells. Using transwell chambers, we found that a fraction of the M : SO-induced detached cells were viable and, furthermore, were able to migrate, re-attach, and resume proliferation when re-incubated in drug-free media. The total elimination of A549 detachment-resistant cells and M : SO-induced detached cells were successfully eliminated by equivalent M : SO concentration (17.5 μ M : 17.5 μ M). Thus, M : SO prevented cell migration. Similar results were obtained on DBTRG.05MG human glioma cells. Our data guarantee further studies to evaluate the in vivo occurrence of D-IDCs, their implications for invasiveness and metastasis and their sensitivity to anticancer drugs.
... Recent studies in solid tumors indicate that the concept of cancer as a hierarchy initiated and maintained by a rare population of stem cells [35] may have larger implications beyond haematopoiesis field. Identification in the previous years of breast cancer [36,37] stem cells and characterization of central nervous system stem cells [38] responsible for the maintenance of some brain tumors have increased the evidence of the veracity of rare cancer stem cells that drive the formation of a number of different tumors types, raising the question of whether all cancers originate from and are maintained by cancer stem cells. ...
Article
Full-text available
Terrorism refers to the use or threat of force or violence against people or their property. The challenges of countering biological threats faced by the international community in the twenty-first century are broader and deeper in purpose the incidents connected with this series of events reveals certain important challenges and obstacles for the coordination and communication of public and private sector emergency responses to the possible terrorist bioweapon attacks. They do not only refer potential threat of States developing biological weapons but also to prevent the pathogens released into the environment and the accidental infection to the laboratory workers.
... Recent studies in solid tumors indicate that the concept of cancer as a hierarchy initiated and maintained by a rare population of stem cells [35] may have larger implications beyond haematopoiesis field. Identification in the previous years of breast cancer [36,37] stem cells and characterization of central nervous system stem cells [38] responsible for the maintenance of some brain tumors have increased the evidence of the veracity of rare cancer stem cells that drive the formation of a number of different tumors types, raising the question of whether all cancers originate from and are maintained by cancer stem cells. ...
Article
Full-text available
Stem cells likely play key roles in the repair of diverse injuries or disorders in body. Recent advances in biotechnology and regenerative medicine brought a revolutionary change in the practice of medicine. Advancements in stem cell biology, including embryonic and postnatal somatic stem cells, have made the prospect of tissue regeneration a potential clinical reality. Absolute concept for the use of stem cells in bone disorders has been centered on the strategies for reconstructing segmental regions of the skeleton, lost to trauma or surgery, and in muscle diseases, to provide normal population of cells in order to replace defective tissue.
... From the clinical aspect, brain tumor treatment will require the use of drugs or drugs combinations that should reach the brain tumoral tissue for extended periods of time at effective concentrations to eliminate all cancer cells. With this in mind, our lab developed several novel concepts that include (i) the regrowth concentration zero (RC 0 ): defined as the minimum drug concentration that kills 100% of cancer cells preventing regrowth when cells are re-incubated in drug free media [12], (ii) the term pankiller: defined as drugs or drugs combination that deplete cancer cells [13] [14], (iii) a novel two phases treatment (2PT) that in vitro was shown to be effective in eliminating all cancer cells. In this treatment regime, cells are exposed to drugs for few weeks (first phase) that eliminates most of the cancer cells leaving few surviving cells that are eliminated by treatment with a second drug (second phase). ...
Article
Full-text available
The anticancer drugs screening program is a long and expensive process. It is estimated that only 5% of drugs entering clinical trials are approved by the FDA. Moreover, many of the drugs that enter clinical trials are often of limited use in clinical practice, and most cancers remain untreatable. Brain tumors are particularly difficult to treat due to the presence of the blood brain barrier that limits the penetration of anticancer drugs. Additionally the isolation from most brain tumors of putative cancer stem cells and novel models of cancer stem cell biology suggest that anticancer drugs should be delivered for prolonged time and at higher concentrations to deplete any potential tumorigenic cell. In this paper, current concepts of cancer stem cell biology and novel concepts of anticancer drugs screening are integrated to develop a seven-steps algorithm as a guideline for the preclinical evaluation of active compounds for the treatment of brain tumors. The flexibility of the algorithm allows the inclusion of alternative studies to exhaustively investigate anticancer drugs and creates multiple opportunities where decisions to engage or not in early clinical trials can be made providing a useful tool for translational research in neurooncology.
Article
Nanotechnology is the study of tiny structures ranging in size from 0.1 to 100 nanometers. It includes biophysics, molecular biology, and bioengineering, as well as medical subspecialties such as cardiology, ophthalmology, endocrinology, oncology, and immunology. Pharmaceutical Nanotechnology combines the methods and ideas of nanoscience and nanomedicine with pharmacy to create novel medication delivery systems that transcend the limitations of traditional drug delivery systems. The purpose of this article is to provide an overview of nanotechnology and its uses in the pharmaceutical industry.
Article
Glioblastoma is the most common primary brain tumor and one of the most devastating cancers. High-grade glioma patients’ prognoses are very poor, usually with a median survival of less than one year. Maximal neurosurgical resection followed by external adjuvant radiation therapy and chemotherapy is the conventional treatment for newly diagnosed high-grade glioma. Resistance to cytotoxic agents remains the greatest barrier to the successful treatment of human cancer. The alkylating agents temozolomide (TMZ), carmustine (BCNU), and lomustine (CCNU), which readily cross the blood-brain barrier, are the major chemotherapeutic candidates in glioma treatment, but many patients do not benefit from these drugs due to inherent or acquired resistance. We recently developed a two-phase treatment (2PT) aimed at eliminating drug resistant glioma cells by a second-phase treatment with a low concentration of salinomycin. In this study, we evaluated 1) the effect of prolonged exposure of clinically useful TMZ, BCNU, and CCNU on glioma cells; and 2) the fate of surviving cells as well as their sensitivity to a low concentration of salinomycin. We found that prolonged treatment with TMZ, BCNU, and CCNU induces a senescent-like state in the resistant cells, and that they then can be partially eliminated with a second-phase treatment of a low concentration of salinomycin.
Article
The effectiveness of anticancer therapies relies on the ability of these substances to eliminate selectively the malignant cells with little or no toxicity to normal cells. The isolation in most human tumors of a rare subpopulation of cancer stem cells (CSCs) associated with chemo resistance lead to the "stem cell theory" (SCT). The SCT proposed that eliminating this fraction will eventually cure cancer, but experimental data supporting this classical view are controversial and are now being gradually replaced by other models. These novel models of cancer biology predict that to cure cancer only drugs or combination of drugs that eliminate all (CSCs and non-CSCs) cancer cells at once ("pankiller drugs") will be effective. The search for pankillers drugs will require tests to assess (i) the elimination of all cancer cells in in vitro systems and (ii) the ability to eradicate the tumors and prevent tumor relapse in in vivo systems. However, at present, most drugs are being tested in assays that can only provide a picture of the short-term activity of anticancer compounds. This in part explains why only a small fraction of the drugs that enter clinical trials are actually approved for clinical use. This article will provide a concise review of the systems, assays, and endpoint parameters routinely used to screen for potential anticancer drugs, and proposes, based in the current knowledge of cancer biology, a more rationale anticancer drug-screening program.
Article
Full-text available
Standard treatment for glioma includes surgery, radiotherapy and chemotherapy but the outcome of patients is very poor. Antineoplastic drugs are usually administered alone or in combination for variable times (continuously or in cycles) in a single phase schedule. In this study we explored in vitro the antiproliferative effect of a 2 phases treatment. In the first phase, glioma cells where treated for 3-4 weeks with hydroxyurea (HU) or aphidicolin and then for 4 weeks with salinomycin, a drug that preferentially inhibits the proliferation of cancer stem cells. We found that salinomycin, is able to slowly deplete the fraction of glioma cells that survive the exposure to HU or aphidicolin. Surviving cells were killed at salinomycin concentrations lower than those required to kill untreated cells. The fraction of surviving cell showed traits of senescence including increased activity of the senescence associated -β-galactosidase (SA-β-gal) marker. Our data suggest that drug-induced senescent cells may constitute a novel target for cancer treatment and can be exploited in a two phases therapeutic regimen.
Article
Gliomas are the most common primary brain tumor, and their treatment is still a challenge. Here, we evaluated the antiproliferative effect of a novel combination of two potent oxidative stress enhancers: menadione (M) and sodium orthovanadate (SO). We observed both short-term and prolonged growth inhibitory effects of M or SO alone as well as in combination (M:SO) on DBTRG.05MG human glioma cells. A stronger antiproliferative effect was observed in the short-term proliferation assay with the M:SO combination compared to either investigated agent alone. In the long-term proliferation assay, a 10-day exposure to M:SO at concentrations of 10 μM:17.5 μM or 17.5 μM:10 μM was enough to kill 100% of the cells; no cell regrowth was observed after re-incubation in drug-free media. When used in combination, the single concentration of M and SO could be decreased by 2.5- to 5-fold of those used for each experimental drug alone and still obtain a similar antiproliferative effect. The underlying molecular mechanism was investigated by co-incubating M:SO with dithiothreitol (DTT) and genistein. Both substances partially neutralized the effects of the M:SO combination, showing additive effects. This observation suggests a role of oxidative stress and tyrosine kinase stimulation in the M:SO cytotoxic effect. Our results indicate that M:SO combination is an attractive alternative for glioma treatment that encourages further study. The neutralizing effects of genistein and DTT reveal a possibility for their use in the minimization of potential M:SO systemic toxicity.
Article
Full-text available
Glioblastomas are among the most lethal neoplasms, with a median survival of <1 year. Modulation of the proteasome function has emerged as a novel approach to cancer pharmacotherapy. Here, we characterized the antitumor properties of SC68896, a novel small molecule proteasome inhibitor. Different tumor cell lines were tested by crystal violet staining for sensitivity to SC68896, given alone or in combination with death ligands. The molecular mechanisms mediating SC68896-induced cell death and changes in cell cycle progression were assessed by immunoblot and flow cytometry. An orthotopic human glioma xenograft model in nude mice was used to examine the in vivo activity of SC68896. SC68896 inhibits the proliferation of cell lines of different types of cancer, including malignant glioma. Exposure of LNT-229 glioma cells to SC68896 results in a concentration- and time-dependent inhibition of the proteasome, with a consequent accumulation of p21 and p27 proteins, cell cycle arrest, caspase cleavage, and induction of apoptosis. Using RNA interference, we show that the effect of SC68896 on glioma cells is facilitated by wild-type p53. SC68896 sensitizes glioma cells to tumor necrosis factor-related apoptosis-inducing ligand and CD95 ligand and up-regulates the cell surface expression of the tumor necrosis factor-related apoptosis-inducing ligand receptor cell death receptors 4 and 5, which may contribute to this sensitization. Intracerebral glioma-bearing nude mice treated either i.p. or intratumorally with SC68896 experience prolonged survival. SC68896 is the first proteasome inhibitor that exerts antiglioma activity in vivo. It may represent a novel prototype agent for the treatment of malignant gliomas and warrants clinical evaluation.
Article
Full-text available
One hundred and seventeen patients with cerebral glioma (Kernohan grades III and IV) were treated with adjuvant chemotherapy using procarbazine (PCB), CCNU and vincristine (VCR) following whole head irradiation. Cell cultures were prepared from 40 patients in this series and their sensitivity to each cytotoxic drug was assessed in a mictotitration assay with 35 S-methionine incorporation as the end point. Twenty-two of forty (55%) patients responded to PCB and/or CCNU in vitro, and sensitivity to these drugs was linked with increased RFI, whilst sensitivity to VCR was not. The RFI of patients who had responded to PCB or CCNU in vitro was significantly longer than the RFI of patients whose tumours failed to respond in vitro or patients who had not been tested. There was no difference in sex ratio, extent of operation, radiation dose and degree of steroid cover between responders, non-responders and untested groups. Grade III tumours tended to be more sensitive in vitro than grade IV tumours. The age of patients also influenced in vitro chemosensitivity. Patients with chemosensitive tumours in vitro tended to be younger than patients with insensitive tumours in vitro. Further statistical analysis, taking into account these prognostic factors, indicated an association between chemosensitivity in vitro and RFI.
Article
In this study, using an in vitro human tumor model, we show that non-small lung adenocarcinoma A549 cells after treatment with DNA damaging antitumor drugs become permanently growth-arrested as a result of so-called drug-induced premature senescence (pseudo-senescence). However, a small fraction of drug-treated cells escapes pseudo-senescence that leads to re-growth of tumor cell population after drug treatment. We show that this re-growth is associated with the presence of cancer stem cells (CSCs) in lung tumor cell population. We also document that re-growth of CSCs can be greatly delayed if lung tumor cells are treated with drug/caffeine combination that leads to the inhibition of the ATM/ATR pathway and decreased phosphorylation of PKB/Akt at Ser473. We show that in non-treated A549 cells caffeine by itself induces a reversible growth arrest that is associated with increased fraction of so-called side population cells, containing CSCs. These results point to the existence of an unknown, caffeine-sensitive mechanism that controls the number of CSCs in lung tumor cell population. Full characterization of this mechanism may lead to the development of innovative cancer therapies, which are based on small molecular weight inhibitors of CSC differentiation and self-renewal, which mimic caffeine action. Our results have also important implications for drug screening tumor models in vitro.
Article
Synthetic oligonucleotides of defined sequence were used to examine the mechanism of calf thymus DNA polymerase alpha inhibition by aphidicolin. Aphidicolin competes with each of the four dNTPs for binding to a pol alpha-DNA binary complex and thus should not be viewed as a dCTP analogue. Kinetic evidence shows that inhibition proceeds through the formation of a pol alpha.DNA.aphidicolin ternary complex, while DNase I protection experiments provide direct physical evidence. When deoxyguanosine is the next base to be replicated, Ki = 0.2 microM. In contrast, the Ki is 10-fold higher when the other dNMPs are at this position. Formation of a pol alpha.DNA.aphidicolin ternary complex did not inhibit the primase activity of the pol alpha.primase complex. Neither the rate of primer synthesis nor the size distribution of primers 2-10 nucleotides long was changed. Elongation of the primase-synthesized primers by pol alpha was inhibited both by ternary complex formation using exogenously added DNA and by aphidicolin alone.
Article
The applicability of a human tumor colony-forming assay to drug screening was investigated in terms of feasibility, validity, and potential for discovering new antitumor drugs. Feasibility was addressed in a pilot study during which basic methods, appropriate assay quality controls, and a standardized protocol for screening were developed. Considerable variability was noted in the availability and colony growth of different tumor types. The majority of the evaluable experiments utilized breast, colorectal, kidney, lung, melanoma, or ovarian tumors. For many tumor types, little evidence of growth was observed, or only rare specimens formed colonies. Colony-forming efficiencies ranged from 0.05 to 0.11% for the six most useful tumors listed above. A set of quality control measures was developed to address technical problems inherent in the assay. Testing of standard agents in the pilot study established that most of these agents could be detected as active. However, it also identified three assay limitations: compounds requiring systemic metabolic activation are inactive; medium constituents may block the activity of certain antimetabolites; and compounds without therapeutic efficacy may be positive in the assay. The assay categorized nontoxic clinically ineffective agents as true negatives with 97% accuracy. Of 79 compounds which were negative in the current National Cancer Institute prescreen (leukemia P388), 14 were active in the assay. Several demonstrated outstanding in vitro activity and are structurally unrelated to compounds already in development or in clinical trials. A subset of these active compounds were found to lack activity in a P388 in vitro colony-forming assay. This indication of differential cytotoxicity to human tumor cells makes this subset of compounds particularly interesting as antitumor drug leads. The demonstrated sensitivity to most standard agents, discrimination of nontoxic compounds, reproducibility of survival values within assays and between laboratories, and evidence of ability to identify active compounds which were negative in the in vivo prescreen suggest that the human tumor colony-forming assay may be a valuable tool for antitumor drug screening. However, because of technical limitations inherent in the current assay methodology, this must be confined to selected tumor types and limited to screening on a moderate scale.
Article
Twenty-two consecutive patients with recurrent malignant brain tumors after radiation therapy and systemic combination chemotherapy with BCNU and vincristine, four of whom were not evaluable due to early death, were treated with etoposide (VP-16-213) (50-100 mg/m2 for five days every three weeks). Response, defined as improvement in both clinical examination and computed tomography scan in absence of glucocorticoids dosage increase, was observed in three (17%) of 18 evaluable patients, lasting greater than 21, seven, and two months, respectively. Six additional patients had stable disease for greater than 10, seven, four, four, three, and two months: all of them had improvement of clinical symptoms but no variation in their scans. Overall median survival from the start of VP-16-213 was 4.5 months (range, 1-23 + months), whereas patients with response or stable disease had a median survival of eight months. Overall, treatment was well tolerated. In 10 patients concomitant plasma and cerebrospinal fluid samples were evaluated with a high-performance liquid chromatographic method for drug assay. The concentration of VP-16-213 in cerebrospinal fluid was less than 1% that found in plasma, even in the two patients with response. The activity of etoposide in patients with malignant, lomustine-vincristine-resistant brain tumors suggests an interesting potential use for this drug.
Article
Quantitative effects of inhibitors of the replicative DNA polymerases (pol) alpha, delta and epsilon from calf thymus are reported under similar assay conditions. Carbonyldiphosphonate was a competitive inhibitor of pols delta and epsilon, with 4- to 6-fold selectivity compared to pol alpha. Aphidicolin inhibited pols alpha and delta with 6- to 10-fold selectivity compared to pol epsilon. The 'butylphenyl' nucleotides, BuPdGTP and BuAdATP, inhibited pol alpha with at least 1000-fold selectivity compared to pols delta and epsilon. The use of these inhibitors under similar assay conditions permits the discrimination of the three enzymes.
Article
The US National Cancer Institute (NCI) 60 human tumour cell line anticancer drug screen (NCI60) was developed in the late 1980s as an in vitro drug-discovery tool intended to supplant the use of transplantable animal tumours in anticancer drug screening. This screening model was rapidly recognized as a rich source of information about the mechanisms of growth inhibition and tumour-cell kill. Recently, its role has changed to that of a service screen supporting the cancer research community. Here I review the development, use and productivity of the screen, highlighting several outcomes that have contributed to advances in cancer chemotherapy.
Article
Among the vitamin K (VK) compounds, VK3 exhibits distinct cytotoxic activity in cancer cells and is thought to affect redox cycling; however, the underlying mechanisms remain unclear. Here we demonstrate that VK3 selectively inhibits DNA polymerase (pol) gamma, the key enzyme responsible for mitochondrial DNA replication and repair. VK3 at 30 microM inhibited pol gamma by more than 80%, caused impairment of mitochondrial DNA replication and repair, and induced a significant increase in reactive oxygen species (ROS), leading to apoptosis. At a lower concentration (3 microM), VK3 did not cause a significant increase in ROS, but was able to effectively inhibit cell proliferation, which could be reversed by supplementing glycolytic substrates. The cytotoxic action of VK3 was independent of p53 tumor suppressor gene status. Interestingly, VK3 only inhibited pol gamma but did not affect other pol including human pol alpha, pol beta, pol delta, and pol epsilon. VK1 and VK2 exhibited no inhibitory effect on any of the pol tested. These data together suggest that the inhibition of pol gamma by VK3 is relatively specific, and that this compound seems to exert its anticancer activity by two possible mechanisms in a concentration-dependent manner: (1) induction of ROS-mediated cell death at high concentrations; and (2) inhibition of cell proliferation at lower concentrations likely through the suppression of mitochondrial respiratory function. These findings may explain various cytotoxic actions induced by VK3, and may pave the way for the further use of VK3.