You are on page 1of 11

Send Orders for Reprints to reprints@benthamscience.

ae
1324 Current Medicinal Chemistry, 2015, 22, 1324-1334

A Simple and Reliable Approach for Assessing Anticancer Activity In Vitro

Miguel López-Lázaro*

Department of Pharmacology, Faculty of Pharmacy, University of Seville, Spain

Abstract: Cancer patients need better anticancer drugs, and medicinal chemistry can play a critical
role in the discovery of these drugs. For an efficient drug discovery process, chemists working on the
synthesis of potential anticancer agents need to use reliable screening methods. These methods should
not only detect the compounds with the highest therapeutic potential, but should also predict whether
such potential is high enough to deserve additional attention. Unfortunately, the current strategies for
assessing anticancer activity in vitro are unable to do this reliably. This review article analyzes these
strategies and describes an alternative screening approach. It is based on establishing suitable experi-
mental conditions to detect compounds that improve the selective cytotoxicity of the drugs used in
cancer therapy. This patient-oriented approach is easy to implement and may help medicinal chemists, and other research-
ers involved in cancer drug discovery, assess in vitro anticancer activity more reliably.
Keywords: Antitumor activity, cancer, cytotoxic activity, drug discovery, screening, selectivity.

1. INTRODUCTION Medicinal chemists involved in cancer drug discovery


typically follow a cytotoxic potency-based screening ap-
The chances of survival of people diagnosed with local-
proach: the lower the concentration required to kill cancer
ized tumors are relatively high. Unless the tumor is in a vital
cells the higher the potential for cancer therapy. If the most
organ or problematic location, surgery and radiation thera-
cytotoxic compound of the series improves the cytotoxic po-
pies can generally eliminate the tumor cells and cure the dis- tency of the anticancer drug used as control, the new com-
ease. These therapeutic modalities are not curative when
pound deserves additional attention [2-7]. Another common
cells from primary tumors have already metastasized to un-
screening strategy consists of assessing the ability of the
known locations. When this very common situation arises,
compounds to act on particular therapeutic targets. For exam-
oncologists need to administer drugs systemically to try to
ple, because DNA topoisomerases and tubulin are the targets
reach and kill the cancer cells. Unfortunately, the ability of
of well-established anticancer drugs, the most potent modula-
the existing anticancer drugs to eliminate the cancer cells in tors of these targets are considered to be the most promising
patients with metastasis is low, and these patients do not
compounds of their series [8-16]. Recently, compounds have
usually overcome the disease. For example, the five-year
been screened and selected based on their ability to target
relative survival rate for patients with distant metastasis is
molecular differences between cancer cells and healthy cells,
4% in lung cancer, 28% in prostate cancer, 24% in breast
e.g., particular protein kinases [17-22]. Several recent studies
cancer, 13% in colorectal cancer, 3% in liver cancer, 16% in
have also focused on the screening of small-molecule modu-
melanoma, 12% in renal cancer, 27% in ovarian cancer, 5% lators of microRNAs [23, 24]. Researchers usually combine
in bladder cancer, 4% in esophageal cancer, and 2% in pan-
cytotoxic potency-based approaches with target-based ap-
creatic cancer [1]. Many patients within these percentages
proaches to identify the best compounds of their series, and
eventually die of the disease despite surviving five years
use animal models to confirm the in vitro anticancer activity
after diagnosis.
of their selected compounds [8, 9, 11, 14, 16, 19, 20].
Because pharmacotherapy is the main form of treatment After analyzing the type of drugs that cancer patients
for patients with advanced metastatic cancers, it is vital to
need, this article shows that the current screening methods
discover better anticancer drugs. Medicinal chemistry plays
are unreliable to identify this type of drugs. Then, it de-
an important role in cancer drug discovery and development.
scribes a simple and patient-oriented screening approach,
For an efficient drug discovery process, chemists working on
which may help researchers assess the in vitro anticancer
the synthesis of potential anticancer agents need to use reli-
activity of their compounds more reliably.
able screening methods. These methods should not only de-
tect the compounds with the highest therapeutic potential,
2. CANCER PATIENTS NEED DRUGS THAT IM-
but should also predict whether such potential is high enough
PROVE THE ABILITY OF THE EXISTING DRUGS
to deserve additional attention.
TO KILL THEIR CANCER CELLS SELECTIVELY
*Address correspondence to this author at the Department of Pharmacology,
Faculty of Pharmacy, University of Seville, Spain; C/ Profesor Garcia Gon- When one treats cancer cells with standard anticancer
zalez 2, 41012, Sevilla, Spain; Tel: +34 954 55 63 48; drugs and examines the cells under the microscope, one ob-
Fax: + 34 954 55 60 74; E-mail: mlopezlazaro@us.es serves that specific concentrations and exposure times result

1875-533X/15 $58.00+.00 © 2015 Bentham Science Publishers


A Simple and Reliable Approach for Assessing Anticancer Activity Current Medicinal Chemistry, 2015, Vol. 22, No. 11 1325

in a 100% cancer cell death. Many of these drugs kill all the cells as possible without killing the patient, usually with the
cancer cells at micromolar concentrations, and others do it at final aim of extending patient survival (Fig. 1). The answer
nanomolar or millimolar concentrations. Researchers have to the second question is that oncologists often use drugs that
also found numerous compounds not used in cancer therapy kill cancer cells at high concentrations because the key fea-
despite their ability to kill cancer cells at very low concentra- ture of an effective anticancer drug is its ability to kill cancer
tions. These observations raise questions that chemists work- cells selectively and not its ability to kill cancer cells at low
ing on the synthesis of potential anticancer agents ask cancer concentrations. A drug that kills cancer cells at 1 mM with-
pharmacologists: If we already have drugs that are excellent out affecting healthy cells at 100 mM will probably kill more
at killing cancer cells, why do these drugs not save the lives cancer cells in patients than a drug that kills both cell types
of patients with metastatic cancers? Why do oncologists of- at 1 nM. The answer to the third question is that the new
ten use drugs that kill cancer cells at high concentrations compound should kill cancer cells at concentrations that do
instead of compounds that do it at lower concentrations? At not significantly affect nonmalignant cells in order to have
what concentration should a new compound kill cancer cells potential for cancer therapy [25].
to have potential for cancer therapy?
Selectivity is the most important feature of an effective
The current drugs do not usually save the lives of patients anticancer drug. Understanding how much selectivity a new
with metastatic cancers because they have a limited ability to drug should have to be clinically effective is essential. If
kill cancer cells at concentrations that do not significantly possible, the drug should kill all the cancer cells of the pa-
affect healthy cells. This narrow selectivity implies that on- tients without significantly affecting their healthy cells. It
cologists cannot use the doses of the drugs required to kill all should match the selectivity of antibiotics, which can kill the
the cancer cells of their patients. If they used such doses, bacterial cells of our bodies at concentrations that do not
they would also kill the cells from their normal tissues and significantly affect our cells. An anticancer drug with this
would cause the death of the patients. As a poor alternative, selectivity would probably save the lives of patients with
oncologists have to use the maximum doses tolerated by the metastatic cancers, like antibiotics commonly save the lives
patients, which are generally insufficient to reach the drug of patients with bacterial infections. If this is not possible,
concentrations required to eliminate all their cancer cells. the new drug should at least improve the survival rates of
The purpose of this strategy is to eliminate as many cancer cancer patients treated with the current pharmacological

Fig. (1). The anticancer drug sorafenib has a limited ability to kill cancer cells selectively. Sorafenib is the standard of care for patients
with unresectable or metastatic hepatocellular carcinoma (liver cancer). This oral multikinase inhibitor is known to induce direct and indirect
cytotoxic effects against cancer cells. Skin toxicity is one of the most common dose-limiting side effects of sorafenib. The recommended
maximum tolerated dose for patients receiving sorafenib is 400 mg twice daily, which results in mean plasma concentrations of approxi-
mately 10 µM. In the experiments shown in this Figure, human liver hepatocellular carcinoma cells (HepG2) and human skin nonmalignant
cells (VH10) were exposed to sorafenib 10 µM and 100 µM for 24 h. Sorafenib was then eliminated and the cells were exposed to drug-free
medium for an additional period of 48 h. At this point, representative photographs were taken, and the percentage of cell viability (shown at
the right bottom of the photographs) relative to the untreated cells was estimated with the MTT assay. At high concentrations (100 µM),
sorafenib killed all the liver cancer cells, but also killed the nonmalignant skin cells. At the maximum concentrations tolerated by patients (10
µM), sorafenib showed some selectivity for the cancer cell line, but could not kill all the liver cancer cells. This limited selectivity probably
explains why most of the patients with hepatocellular carcinoma receiving sorafenib live several months longer than untreated patients but do
not usually overcome the disease (the five-year survival rate in patients diagnosed with advanced metastatic liver disease is approximately
3%) [1, 65-67]. Figure taken from reference [25].
1326 Current Medicinal Chemistry, 2015, Vol. 22, No. 11 Miguel López-Lázaro

therapies. A drug that improved the ability of the standard The ability of a compound to modulate molecular targets
drugs to kill cancer cells selectively would probably increase involved in cancer cell proliferation and survival can neither
patient survival. We should therefore look for drugs that im- reliably predict its selectivity towards cancer cells. For ex-
prove the selectivity of the existing anticancer drugs [25]. ample, the enzymes DNA topoisomerases are necessary for
cancer cell proliferation, and several topoisomerase inhibi-
3. CURRENT SCREENING APPROACHES CANNOT tors are useful anticancer drugs. This does not mean, how-
RELIABLY IDENTIFY THE TYPE OF DRUGS THAT ever, that a good inhibitor of these enzymes is a promising
CANCER PATIENTS NEED anticancer compound. We have reported that curcumin is an
efficient inhibitor of DNA topoisomerases [33] with a very
Cancer patients need drugs that improve the ability of the
limited selectivity towards cancer cells [34, 35]. Wang et al.
standard drugs to kill their cancer cells without significantly
recently screened a chemical library of 359,484 compounds
affecting their healthy cells. The most common screening to identify potential inhibitors of the steroid receptor coacti-
strategies cannot detect this type of drugs reliably.
vators SRC-3 and SRC-1; these coactivators are involved in
Chemists involved in cancer drug discovery typically fol- cancer cell proliferation and survival. They identified the
low a cytotoxic potency-based screening approach: a com- cardiac glycoside bufalin as a potent inhibitor of SRC-3 and
pound is promising if it kills cancer cells at low concentra- SRC-1. This compound also inhibited breast cancer cell
tions and improves the potency of the standard drugs. For growth at nanomolar concentrations and reduced tumor
example, several compounds have recently been considered growth in mice xenotransplanted with breast cancer cells.
promising anticancer agents based on their ability to kill can- Based on these observations, the authors discussed that bu-
cer cells at lower concentrations than those of the standard falin could be useful for cancer therapy [36]. Clifford and
drugs etoposide [26], cytarabine (ara-C) [27], 5-fluorouracyl Kaplan observed, however, that human breast nonmalignant
[28], doxorubicin [29] and cisplatin [7, 30]. Despite its wi- cells were more sensitive than human breast cancer cells to
despread use, this approach is unsound and unreliable. the cytotoxic activity of bufalin [37]. This suggests that bu-
falin would probably be ineffective in breast cancer therapy
It is unsound because the aim of this approach is to find [38].
something that we do not need. Cancer patients do not need
drugs that kill their cancer cells at low concentrations. As Researchers do not routinely use healthy cells for assess-
explained previously, they need drugs that kill their cancer ing anticancer activity in vitro because they may not think it
cells at concentrations that do not significantly affect their is necessary. In the end, no matter what screening approach
healthy cells. If a drug kills cancer cells at low concentra- we follow, we will have to use animal models to assess pre-
tions and also kills normal cells at similar concentrations, the clinical anticancer activity properly. These models will con-
maximum doses tolerated by the patients will be insufficient firm the anticancer activity of the selected compounds, and
to reach the drug concentrations required to eliminate their will also reveal if they induce toxicity against healthy tis-
cancer cells. If a drug kills cancer cells without significantly sues.
affecting nonmalignant cells, it does not matter much at what Even if performed properly, in vivo animal experiments
concentrations it kills the cancer cells. cannot compensate for a deficient screening approach. First,
It is unreliable because the ability of a compound to kill although animal models can show if the chosen compounds
cancer cells at low concentrations is not related to its ability induce selective anticancer activity in vivo, they cannot re-
to kill cancer cells selectively. For example, after reporting veal the anticancer activity of the compounds left behind in
that several isoprenyl-thiourea and urea derivatives killed the screening process. Assessing selectivity after the screen-
colon cancer cells at low concentrations and improved the ing step is an inadequate approach to identify the most selec-
potency of the anticancer drug 5-fluorouracil, we found that tive compound of a chemical series. If we accept that selec-
these compounds were equally cytotoxic towards nonmalig- tivity is the key feature of an effective anticancer drug, we
nant cells [31]. More recently, we assessed the anticancer should assess selectivity as part of, and not after, the screen-
activity of a series of new aziridine derivatives, and found ing process so that we can detect the best anticancer com-
that the most cytotoxic compound against a cancer cell line pounds of our series. Second, rodent models can show the
did not show any selectivity versus a nonmalignant cell line selectivity of a compound in human cancer cells versus ro-
from the same tissue (it was actually 7 times more cytotoxic dent normal cells (xenograft models), or in rodent cancer
against the normal cell line). We selected for further studies cells versus rodent normal cells (allograft and spontaneous
a compound that was less cytotoxic in the cancer cell line, models). However, animal models cannot show if a com-
pound kills human cancer cells versus human normal cells
but more selective. When we tested the cytotoxicity of this
selectively. This distinction is important to avoid experimen-
compound against three pairs of human cancer cell lines and
tal artifacts caused by species differences in sensitivity. For
nonmalignant cell lines, we observed that the selectivity of
example, we have shown that rodent cells are extremely re-
this aziridine against breast cancer cells versus normal cells
sistant (over 1000-fold) to the cytotoxicity of some cardiac
from three different tissues was approximately 50-100 fold
glycosides in relation to human cancer and nonmalignant
[32]. These experimental data demonstrate that cytotoxic
cells [39]. This means that if we assess the anticancer activ-
potency and selective cytotoxicity are two unrelated parame- ity of these compounds in mice xenotransplanted with hu-
ters. Therefore, when we show that our compounds kill can- man cancer cells, we will find a marked in vivo anticancer
cer cells at low concentrations, we are not revealing whether activity [36, 40-43]. This activity, however, is probably
or not they have potential for cancer therapy. caused by the ability of cardiac glycosides to selectively kill
human cells versus rodent cells rather than by their ability to
A Simple and Reliable Approach for Assessing Anticancer Activity Current Medicinal Chemistry, 2015, Vol. 22, No. 11 1327

selectively kill human cancer cells versus human healthy of the many potential mechanisms of action of the active
cells [39, 44, 45]. This can explain why the cardiac glycoside compound would we select for our screening protocol? What
bufalin inhibited tumor growth in mice xenografted with if the anticancer drug that we are trying to identify acted on a
human breast cancer cells [36] despite its lack of selectivity molecular target not yet discovered? This example suggests
[38]. These observations suggest that animal models cannot that the most common screening approaches do not reliably
substitute a proper in vitro design. detect the best anticancer compound in a group of potential
anticancer agents. It also suggests that we need to develop
An inadequate in vitro design may result in the selection
novel strategies to assess in vitro anticancer activity more
of useless compounds and in the failure to detect promising
reliably [25].
anticancer agents. In the first case, we will waste resources
trying to prove in animal models, and too many times in can-
cer patients, the anticancer activity of ineffective com- 4. A SIMPLE APPROACH FOR ASSESSING ANTI-
CANCER ACTIVITY IN VITRO
pounds. In fact, most of the compounds selected for further
studies based on the current screening strategies do not show Cancer patients need drugs that improve the ability of the
relevant anticancer effects when tested in animal models of existing drugs to kill their cancer cells without significantly
metastasis and in clinical trials [46-48]. Drug attrition rates affecting their healthy cells. This patient-oriented approach
for cancer are actually much higher than in other therapeutic aims to establish the most adequate experimental conditions
areas, and only 5% of compounds that enter clinical trials are to answer the following question: Can any of my compounds
licensed after demonstrating sufficient efficacy in phase III improve the ability of the current drugs to kill cancer cells
testing [47]. But the most worrying consequence of an in- without affecting nonmalignant cells from suitable tissues?
adequate in vitro screening is that we are probably throwing
The general approach consists of 1) exposing cancer cells
away useful anticancer drugs. Thousands of novel com-
and nonmalignant cells to the experimental drugs and to the
pounds are routinely discarded for not being the most cyto-
toxic of their series or the best modulators of particular mo- standard anticancer drugs, 2) estimating cell viability with a
simple cytotoxicity test, 3) calculating one or several cyto-
lecular targets. Since these drug features are poor predictors
toxicity parameters (e.g., IC50 values) for each drug in each
of therapeutic potential, an unknown number of these com-
cell type, 4) calculating a selectivity index for each drug
pounds might be better than the existing anticancer drugs.
(e.g., dividing the IC50 value in the nonmalignant cells by
Finally, let us consider the following situation to examine that in the cancer cells) and 5) comparing the selectivity in-
the ability of the current screening strategies to detect effec- dices of the experimental drugs with those of the standard
tive anticancer drugs reliably. Let us imagine that one of us anticancer drugs [25, 49, 50]. Several aspects need to be con-
is diagnosed with advanced metastatic liver disease. Like sidered for implementing this approach.
most of the patients in this situation, we would only survive
several months after diagnosis despite being treated with 4.1. Selection of Cancer Cells, Nonmalignant Cells and
novel anticancer drugs such as sorafenib (see Fig. 1 legend). Standard Anticancer Drugs
Let us imagine that we have a collection of 1000 compounds,
and that one of them is an effective cure for patients with our The first important aspect to consider for implementing
cancer. Could we identify this effective anticancer com- this approach is the selection of the cancer cells and nonma-
pound using the current in vitro approaches? lignant cells. Distinguishing between cancer cells and nonma-
lignant cells and being familiar with several basic terms and
Some researchers would screen the collection against concepts is important for this purpose. Unlike nonmalignant
liver cancer cells and would select the most cytotoxic com- cells, cancer cells proliferate and disseminate in an uncon-
pound. Others would select the best modulator of a particular trolled manner and are able to induce tumors following inocu-
therapeutic target. For example, we could choose the best lation into susceptible animals. Primary cells are cells taken
inhibitor of a specific protein kinase of a particular signal directly from organisms, and can be regarded as such until
transduction pathway involved in liver cancer cell prolifera- they are successfully subcultured for the first time. Cell lines
tion and survival. We could also opt for a more classic thera- are cells derived from organisms after the first successful
peutic target and select, for instance, the best topoisomerase subculture. The term nonmalignant cells can be used for pri-
inhibitor or antitubulin compound. We could also combine mary nonmalignant cells, for cell lines derived from primary
several of these screening strategies. It is easy to predict that nonmalignant cells that may have been altered to facilitate
the most cytotoxic compound would not be the best topoi- cell growth in culture (e.g., cells that have been immortalized
somerase inhibitor, the best antitubulin compound, and the through transfection of the catalytic component of the enzyme
best inhibitor of all the molecular targets of the multiple sig- telomerase reverse transcriptase hTERT), or for fetal cells.
nal transduction pathways involved in liver cancer cell pro- The terms normal cells and healthy cells commonly refer to
liferation and survival. non-diseased cells and, therefore, to nonmalignant cells.
What would we think if a problem only has one solution Ideally, we should use primary cancer cells and primary
and each of the methods used to solve it leads to different nonmalignant cells for implementing this approach. Because
solutions? We would probably think that only one of the primary cells are freshly derived from living organisms, they
methods is adequate, or that all of them are inadequate. If we mimic the physiological state of cells in vivo more closely
think that only one is adequate, which one would it be? than cell lines, which may develop changes (e.g., unstable
Would it be the one based on assessing cytotoxic potency or karyotypes) as a result of subculturing. However, primary
the one based on assessing mechanisms of action? If we cells are less convenient to use than cell lines; one needs at
think that the second option is the most suitable one, which least one subject to obtain the primary cells required for each
1328 Current Medicinal Chemistry, 2015, Vol. 22, No. 11 Miguel López-Lázaro

independent experiment. Their value in the laboratory setting If the study is not focused on a particular cancer, we
is limited, especially when large quantities of cells are re- could use several pairs of cancer cell lines and nonmalignant
quired. Well-characterized cancer and nonmalignant cell cell lines, as well as standard drugs used to treat patients
lines are suitable for implementing this approach. with the selected cancers. It is important to note that specific
Both the cancer cells and the nonmalignant cells should anticancer drugs are used only in particular cancers. There-
be human to avoid species differences in sensitivity. For ex- fore, we should calculate selectivity indices for the experi-
ample, as discussed previously, rodent cells are extremely mental compounds in each cancer and compare them with
resistant to the cytotoxicity of cardiac glycosides. Therefore, the selectivity indices of the standard drugs used in those
if we use a human cancer cell line and a rodent nonmalignant cancers. For example, imagine that we decide to screen a
cell line to evaluate the selectivity of a cardiac glycoside, we series of compounds against cancer and healthy cells from
will mistakenly find a selective anticancer effect caused by the colon, lungs and liver, and that we use the standard anti-
species differences in sensitivity. The cancer cells and non- cancer drugs 5-fluorouracil, cisplatin and sorafenib. To show
malignant cells should also be from the same tissues to avoid that one of our compounds could be useful for treating pa-
tissue differences in sensitivity. Imagine a compound that tients with colon cancer, we should use the IC50 values calcu-
kills cancer and normal cells from the lungs at 1 µM and lated for our compound and for the standard drug 5-
cancer and normal cells from the skin at 100 µM. If we as- fluorouracil in the colon cancer cell line and in the three
sess the selectivity of this compound using lung cancer cells nonmalignant cell lines (i.e., colon, lung and liver). Then, we
versus skin normal cells, we will find a selective anticancer should calculate a selectivity index for our compound and for
effect caused by tissue differences in sensitivity. 5-fluorouracil dividing the mean IC50 value in the three
nonmalignant cell lines by the IC50 value in the colon cancer
Another important aspect to consider is the number of
cell line. Finally, we should see if the selectivity index of our
cell lines to use. Ideally, we should assemble a panel of well-
compound is higher than that of 5-fluorouracil.
characterized cancer cell lines representative of all cancer
types, and a variety of nonmalignant cells. The normal cells Depending on the aim of the study, we can use particular
should be from the same tissues than those of the cancer cells types of cancer cells. If the experimental treatment is being
and from tissues commonly affected by cancer pharma- developed for specific patients, primary cells from these pa-
cotherapy. With this panel of cell lines, we would reliably tients could be used to better predict the effectiveness of the
predict whether or not any of the compounds has potential treatment or to avoid the administration of inactive che-
for cancer therapy. If we use fewer cancer cell lines, useful motherapeutics to them [52, 53]. Treatment failure has been
compounds for specific types of cancer may be missed. If we associated with the ability of cancer cells to develop drug
use fewer nonmalignant cell lines, we may miss toxicity to- resistance. We, therefore, could also use resistant cancer cells
wards particular tissues that could limit the therapeutic po- for implementing this approach. Evidence also suggests that
tential of the compounds. Despite these limitations, we can resistance to treatment may be due to the fact that current
assess anticancer activity reliably using a much lower num- treatments preferentially target non-stem cancer cells. Cancer
ber of cell lines. stem cells have been associated with resistance to treatment,
worse prognosis and recurrence of the disease. Another way
If the study is focused on a particular cancer, the mini-
that could help to identify potential anticancer drugs would be
mum requirement for implementing this screening approach
to evaluate their effects on cancer stem cells [54-56].
is to use a human cancer cell line, a human nonmalignant
cell line from the same tissue, and an anticancer drug used to
4.2. Cell Treatment Conditions
treat patients with the selected cancer. This simple approach
is suitable to screen rapidly the in vitro anticancer activity of Cell density is an important parameter to consider to ob-
large series of compounds against a particular cancer. If none tain reliable results. The cytotoxicity of a drug in cells grown
of the compounds improves the selectivity index of the stan- at low density may be different from that in cells grown at
dard anticancer drugs, the compounds have a little potential high density. For example, the IC50 values for 5-fluorouracil
for the treatment of the selected cancer [51]. If one of the and a novel merosesquiterpene were approximately 6 times
compounds improves the selectivity index of the standard lower in MCF7 breast cancer cells seeded at 5 x103 cells/cm2
drugs, the compound is promising; however, we should use than in the same cells seeded at 5 x104 cells/cm2 [57]. Be-
more nonmalignant cell lines to assess anticancer activity cause nonmalignant cells generally grow slower than cancer
more robustly. Using more normal cell types is essential to cells, they are often seeded at higher densities than cancer
detect possible toxicity to normal tissues that could limit the cells, which may originate unreal selectivity values. The
potential use of the new compound. For example, a com- screening approach discussed in this manuscript minimizes
pound that kills lung cancer cells at 1 µM and lung nonma- these false positives. Provided that the density at which we
lignant cells at 100 µM will not be clinically effective if it seed each cell line is the same for the experimental drugs and
also kills normal cells from other tissues (e.g., skin cells or the standard drugs, we can seed different cell lines at differ-
blood cells) at 1 µM. If the study is focused on one cancer, in ent densities. What matters in this approach is whether or not
vitro anticancer activity can be assessed robustly with sev- the experimental compounds improve the selectivity of the
eral cancer cell lines representative of the most common current anticancer drugs and not the magnitude of their IC50
cancer subtypes and with several nonmalignant cell lines values or selectivity indices in particular cell types.
originated from a variety of normal tissues. This little panel
of cell lines could be used to confirm the activity of com- The compounds should be tested at concentrations that
pounds passing the initial screening step. allow the determination of the cytotoxic parameters required
to calculate the selectivity indices. For example, if we show
A Simple and Reliable Approach for Assessing Anticancer Activity Current Medicinal Chemistry, 2015, Vol. 22, No. 11 1329

that the IC50 value of a compound is e.g. 50 µM in cancer and a solubilization step, the quantity of the colored product
cells and >100 µM in nonmalignant cells, we are not truly is measured at 570 nm using a plate reading spectrophotome-
revealing its anticancer potential. We are simply showing ter. Like all assays that rely on an active cellular metabolism,
that its selectivity index is higher than 2, which could be 3 any change in culture conditions that alters the metabolism
(low therapeutic potential) or 10000 (high therapeutic poten- of the cells (e.g., high cell confluence, changes in pH, deple-
tial). When the solubility of the compounds is not a problem, tion of essential nutrients) will likely affect the rate of MTT
we should use wide concentration ranges to allow the calcu- reduction into the formazan product and will lead to a loss of
lation of the required cytotoxic parameters in all the cell linearity between absorbance and cell number. In addition,
lines. like all assays that rely on a reduction reaction, the MTT is
subject to interferences from compounds with inherent re-
Drug exposure times and drug-free recovery periods may
ductive capacity (e.g., ascorbic acid and sulfhydryl-
change the cytotoxic potency and selectivity of particular
containing compounds such as reduced glutathione). At high
compounds. Some drugs need more time to kill cells than
concentrations and exposure times, the compound MTT in-
other drugs. If we use short exposure times and recovery
duces cytotoxic effects. The solubilization step required for
periods, we may underestimate the cytotoxicity of drugs in-
the MTT assay can be eliminated using newer tetrazolium
ducing a slow cytotoxic effect. Other compounds may trigger reagents (e.g. XTT) that are reduced by viable cells into
selective cytotoxic effects at short exposure times and non- soluble formazan products. The assays based on these newer
selective cytotoxic effects at long exposure times. The selec- tetrazolium reagents also have their advantages and disad-
tivity of these compounds could be missed if cell viability is vantages [58-60].
assessed after a long drug incubation period (e.g., 72 h), but
could be detected after a shorter drug exposure time (e.g., 4 The resazurin assay is a redox-based colorimetric or
h) followed by a recovery period (e.g., 68 h). In addition, it fluorometric assay, which employs a methodology similar to
should be noted that many anticancer drugs used in patients that of the tetrazolium assays (MTT, XTT). This method
have a very short biological half life (time required to elimi- relies on the capacity of viable cells to reduce the blue com-
nate 50% of the drug from the plasma). Short drug exposure pound resazurin into the pink, fluorescent and soluble prod-
times followed by drug-free recovery periods may therefore uct resorufin. The quantity of resorufin produced is propor-
mimic the in vivo activity of these drugs more closely than tional to the number of viable cells. Fluorescence can be
long drug exposure times. Drug exposure times and drug- quantified using a fluorometer equipped with a 560 nm exci-
free recovery periods should be designed flexibly. However, tation / 590 nm emission filter set. Although resorufin can
if we test the experimental compound and the standard drug also be quantified by measuring a change in absorbance,
following several treatment schedules, we should not only spectrophotometric determination is not often used because
compare the selectivity indices obtained under the same of its much lower sensitivity. When used as a fluorometric
schedules, but also under the most selective schedules. assay, its sensitivity is higher than that of the tetrazolium
assays. However, the resazurin assay is also subject to
4.3. Methods for Assessing Cell Viability chemical interferences caused by reducing compounds. Re-
sazurin is also cytotoxic at high concentrations and exposure
Once the cells have been exposed to the experimental times. Fluorescent compounds may also interfere with the
compounds and the standard anticancer drugs, we need to resorufin product. An important advantage is that resazurin
estimate how many viable cells remain at the end of the ex- reagents probably generate the most cost-effective data on a
periments. This section does not seek to review all the avail- per well basis [58-60].
able assays used to estimate cell viability in culture. It briefly
The sulforhodamine B (SRB) assay is a cost-effective
discusses several standard assays that can be used to imple-
and widely used colorimetric method for screening, in which
ment the screening approach shown in this article. Each as-
cell density determination is based on the measurement of
say has its own set of advantages and disadvantages, and the
cellular protein content. After drug treatments, cells are fixed
selection of a particular method depends on many factors
with trichloroacetic acid and stained with the SRB com-
such as the number of samples to screen, its suitability for
pound. The excess dye is then removed by washing the
high-throughput screening (HTS), or the infrastructure and
plates with acetic acid, and the protein-bound dye is dis-
research funds of the laboratory. The assays discussed next
solved in Tris base solution for spectrophotometric determi-
can be used in multi-well formats, and have been reviewed
nation at 510 nm. Its sensitivity is comparable to that of
or described in detail elsewhere [58-63]. References [58]
some fluorometric methods. Another important advantage is
(freely available at http://www.ncbi.nlm.nih.gov/books/
that SRB staining is independent of cell metabolic activity;
NBK144065/) and [63] describe in detail several common
therefore, fewer steps are required to optimize assay condi-
protocols for assessing cell viability.
tions for cell lines with different metabolic activities. In ad-
The MTT assay is an inexpensive and widely used col- dition, the SRB assay is not subject to chemical interferences
orimetric assay. It was the first convenient 96-well method caused by reducing compounds. The SRB method, however,
developed for screening large numbers of samples. The MTT does not distinguish between viable and dead cells, although
assay is based on the ability of viable cells to convert the this does not seem to compromise its ability to detect cyto-
MTT compound (3-(4,5-dimethylthiazol-2-yl)-2,5- toxic effects of a drug. Several studies have shown that re-
diphenyltetrazolium bromide) into an insoluble and purple sults from the SRB assay correlate well with those of the
formazan product; dead cells are metabolically inactive and MTT assay, although the IC50 values of compounds tested
cannot reduce the MTT into the colored product. After an using the SRB method generally are slightly higher. The
incubation period (generally 1-4 h) of the cells with the MTT SRB assay is the screening method used by the National
1330 Current Medicinal Chemistry, 2015, Vol. 22, No. 11 Miguel López-Lázaro

Cancer Institute (NCI) in their anticancer drug discovery detected using two independent assays [59, 60]. Observing
program [62, 63]. the cells under the microscope is also a good practice to de-
tect these possible artifacts, particularly when the number of
The ATP assay is a fast and highly sensitive lumines-
test compounds is small. If we use a particular assay, and the
cence method widely used for estimating cell viability in
high-throughput screening. Cells contain regulated levels of results do not faithfully represent what we see under the mi-
croscope (cell number and morphology), we should consider
ATP, and drug-induced cytotoxicity results in a loss of abil-
repeating the experiments using another assay.
ity to synthesize new ATP along with a rapid depletion of
cellular ATP by endogenous ATPases. ATP is quantified by
4.4. Expression and Discussion of Results
measuring the light produced through its reaction with the
firefly enzyme luciferase using a luminometer. The amount After estimating cell viability with a cytotoxicity assay,
of light produced is directly proportional to the number of we need to calculate one or several cytotoxicity parameters
viable cells. Commercial kits contain a detergent to lyse the (e.g., IC50, IC90, LC50) for each drug in each cell type. Then,
cells, ATPase inhibitors to stabilize the ATP that is released we should use these cytotoxicity parameters to calculate one
from the lysed cells, luciferin as a substrate, and luciferase to or several selectivity indices for each drug (e.g., dividing the
catalyze the reaction. The main advantage of this assay is its IC50 value in the nonmalignant cells by that in the cancer
high sensitivity, which enables miniaturization into 1536 cells). Finally, we should compare the selectivity indices of
well formats for HTS protocols. In addition, luminescence the experimental drugs with those of the standard anticancer
signals are not altered by intrinsically fluorescent test com- drugs.
pounds. Another advantage of this assay is that, like the SRB
Selectivity indices based on only one cytotoxic parameter
assay and unlike the tetrazolium and resazurin assays, it does
(e.g., IC50) may be misleading when dose-response curves are
not require an incubation step of the substrate with a popula-
not analyzed carefully. Fig. (2) represents dose-response
tion of viable cells to yield the product that generates the
signal, which eliminates the step of returning the cells to the curves of three hypothetical compounds in a cancer cell line
and a nonmalignant cell line. Each panel also shows two se-
incubator. However, the assay would detect possible changes
lectivity indices based on IC50 values and IC90 values. In Fig.
in cellular ATP levels not related to changes in cell viability.
(2A), the selectivity index based on the IC50 values represents
In addition, the luciferase reaction is susceptible to tempera-
faithfully the selectivity of the compound observed in the
ture changes and may also be altered by luciferase inhibitors
dose-response curves. In Fig. (2B and 2C), however, the se-
[58-60].
lectivity index based on the IC50 values may lead to misinter-
The lactate dehydrogenase (LDH) assay has been widely pretation of results. Dose-response curves in Fig. (2B) show
used to detect in vitro cytotoxicity. LDH is a cytosolic en- that the selectivity of the compound is very limited; however,
zyme that is released into cell culture media when the plasma the selectivity index based on the IC50 values (171,3) suggests
membrane is damaged. The released LDH can be quantified that the compound has a high selectivity towards the cancer
by a coupled enzymatic reaction. First, LDH catalyzes the cells. In Fig. (2C), the selectivity index based on the IC50 val-
conversion of lactate to pyruvate via reduction of NAD+ to ues (3,0) suggests that the compound has a limited selectivity
NADH. Second, diaphorase uses NADH to reduce an appro- towards the cancer cell line. However, the dose-response
priate redox reagent such as a tetrazolium compound or resa- curves show that this compound kills all the cancer cells at
zurin into a colored formazan or fluorescent product. The 0.1 µM and all the healthy cells at 100 µM. This compound
levels of this product is directly proportional to the amount could be useful if the reduced cell viability (40%) observed in
of released LDH in the medium and, therefore, to the amount the normal cells in the 0.1-10 µM range is caused by reversi-
of dead cells. Although LDH activity can be measured spec- ble inhibition of cell proliferation. False positives (Fig. 2B)
trophotometrically, fluorescence detection is advisable to and false negatives (Fig. 2C) obtained when selectivity indi-
increase sensitivity. This assay is susceptible to background ces are calculated based on only one cytotoxic parameter
signal from serum sources of LDH found in supplemented (e.g., IC50) could be prevented by using additional selectivity
growth media, which elevate the background signal. This indices based on other cytotoxic parameters (e.g., IC90) or by
method is also subject to assay artifacts by compounds that showing the dose-response curves.
inhibit LDH activity and to fluorescence or color interfer-
In addition to comparing the selectivity indices of the ex-
ences. An important inconvenient is that activity-based sur-
perimental compounds with those of the standard anticancer
rogates of cell death such as LDH have a finite enzymatic
drugs, it is important to observe that none of the healthy cell
half-life. The loss of LDH activity over time can lead to un-
lines is highly sensitive to the cytotoxicity of the experimen-
derestimation of cytotoxicity in cells exposed for relatively
tal compounds. We should not forget that a high toxicity in
long incubation periods (48-72 h) to drugs (or drug concen-
trations) that induce a fast cytotoxic effect [59]. normal cells from just one particular tissue may cause that
the maximum doses tolerated by the patients are insufficient
Although these assays are based on different concepts, to reach the drug concentrations required to eliminate their
there is usually a good correlation between the results ob- cancer cells.
tained with different assays. For example, although the MTT
assay measures cellular activity and the SRB assay measures 4.5. Final Considerations
cellular content, comparisons of data from several hundred
compounds screened in parallel by MTT and SRB assays The screening approach discussed throughout this article
yielded quite similar results [63]. The possible experimental has the typical limitations of all in vitro methods. Because in
artifacts caused by a particular assay could be prevented and vitro conditions cannot represent in vivo conditions faith-
fully, compounds that work in vitro may not work in vivo
A Simple and Reliable Approach for Assessing Anticancer Activity Current Medicinal Chemistry, 2015, Vol. 22, No. 11 1331

A SI (IC50) = 141,3 B SI (IC50) = 171,3


SI (IC90) = 120,1 SI (IC90) = 1,2

100 healthy cells 100 healthy cells


90 cancer cells 90
cancer cells
80 80

% cell viability
% cell viability

70 70
60 60
50 IC50 50 IC50
40 40
30 30
20 20
10 IC90 10 IC90
0 0
0 0,01 0,1 1 10 100 0 0,01 0,1 1 10 100

concentration (µM) concentration (µM)

C SI (IC50) = 3,0
SI (IC90) = 764,1

100 healthy cells


90 cancer cells
80
% cell viability

70
60
50 IC50
40
30
20
10 IC90
0
0 0,01 0,1 1 10 100

concentration (µM)

Fig. (2). Selectivity indices based on IC50 values do not always represent selective anticancer activity faithfully. In panel A, the selectiv-
ity index based on IC50 values, SI (IC50), represents the selectivity of the hypothetical compound faithfully. In panels B and C, however, this
index does not truly represent what dose-response curves show. Graph B shows that the compound has a very limited selectivity towards the
cancer cells, and the SI (IC50) indicates a high selectivity (false positive). Graph C suggests that the compound can kill cancer cells selec-
tively, and the SI (IC50) indicates a very low selectivity towards cancer cells (false negative). Misinterpretation of results could be prevented
by showing the dose-response curves or by calculating an additional selectivity index, e.g., SI (IC90). IC50 values (concentration of compound
required to inhibit cell viability by 50%) and IC90 values (concentration of compound required to inhibit cell viability by 90%) for each com-
pound are not shown for simplicity. Selectivity indices (SI) are calculated by dividing the IC50 (or IC90) values in the healthy cells by those in
the cancer cells. See text for further details.

and vice versa. Another limitation is that we cannot exclude of tumor volumes), or do not test the experimental drugs
toxicity to all types of normal tissues by using a variety of and the standard drugs under comparable experimental
nonmalignant cells. Therefore, animal models are essential to conditions [25, 46].
assess preclinical anticancer activity properly. In addition
The screening strategy discussed in this article is based
to detecting pharmacokinetic limitations of the experimen-
on assessing selectivity. Evaluating selectivity is not a
tal compounds, animal models are important to confirm or
novel idea in anticancer drug discovery. Researchers from
detect possible in vivo activity and toxicity. To assess in
many laboratories use healthy cells to assess if active con-
vivo anticancer activity robustly, we should evaluate if the centrations of their selected compounds induce toxicity
experimental drug improves the survival rates of the stan-
against these cells. Although this strategy provides useful
dard drugs when tested under equivalent experimental con-
information about the selected compounds, it is inadequate
ditions in animal models representative of the patients who
to identify the most selective compound in a group of po-
would eventually receive the drugs (e.g., animal models of
tential anticancer agents. This strategy is also inadequate to
metastasis). Unfortunately, most in vivo studies do not use
show if the chosen compound improves the selectivity of
animal models of metastasis, do not assess survival as the the drugs used in cancer patients. Another negative aspect
endpoint of the experiments (they typically assess reduction
1332 Current Medicinal Chemistry, 2015, Vol. 22, No. 11 Miguel López-Lázaro

of assessing selectivity this way is that the magnitude of ACKNOWLEDGEMENTS


this parameter changes much depending on the type of can-
I thank my research group for helpful discussions.
cer cells and healthy cells that we choose; this makes inter-
pretation and extrapolation of results problematic. This
REFERENCES
problem is minimized in the screening approach described
in this article. What matters in the screening approach dis- [1] Siegel, R.; Ma, J.; Zou, Z.; Jemal, A. Cancer statistics, 2014. CA
cussed here is if any of the compounds improves the selec- Cancer J. Clin., 2014, 64, 9-29.
[2] Cheng, W.H.; Cao, B.; Shang, H.; Niu, C.; Zhang, L.M.; Zhang,
tivity of the drugs used to treat cancer patients, and not the Z.H.; Tian, D.L.; Zhang, S.; Chen, H.; Zou, Z.M. Synthesis and
magnitude of their cytotoxic effects in particular cancer evaluation of novel podophyllotoxin derivatives as potential anti-
cells and healthy cells. tumor agents. Eur. J. Med. Chem., 2014, 85, 498-507.
[3] Kim, J.H.; Yu, J.; Alexander, V.; Choi, J.H.; Song, J.; Lee, H.W.;
This approach can be used for high-throughput screening Kim, H.O.; Choi, J.; Lee, S.K.; Jeong, L.S. Structure-activity rela-
[50]. Current high-throughput screening approaches are typi- tionships of 2'-modified-4'-selenoarabinofuranosyl-pyrimidines as
cally based on testing library compounds at one concentra- anticancer agents. Eur. J. Med. Chem., 2014, 83, 208-225.
[4] Alanazi, A.M.; Abdel-Aziz, A.A.; Al Suwaidan, I.A.; Abdel-
tion against one specific target. The same chemical libraries Hamide, S.G.; Shawer, T.Z.; El Azab, A.S. Design, synthesis and
are continuously screened against different anticancer tar- biological evaluation of some novel substituted quinazolines as an-
gets. The approach discussed in this manuscript requires titumor agents. Eur. J. Med. Chem., 2014, 79, 446-454.
each compound to be tested at several concentrations against [5] Rodrigues, F.A.; Bomfim, I.S.; Cavalcanti, B.C.; Pessoa, C.O.;
Wardell, J.L.; Wardell, S.M.; Pinheiro, A.C.; Kaiser, C.R.; No-
several cell types. This would require changes in the imple-
gueira, T.C.; Low, J.N.; Gomes, L.R.; de Souza, M.V. Design, syn-
mentation and organization of the drug discovery process thesis and biological evaluation of (E)-2-(2-
and would increase costs. However, the cost of screening the arylhydrazinyl)quinoxalines, a promising and potent new class of
same libraries many times and of assessing the in vivo anti- anticancer agents. Bioorg. Med. Chem. Lett., 2014, 24, 934-939.
cancer activity of inefficient compounds is probably higher. [6] Liu, W.; Gust, R. Metal N-heterocyclic carbene complexes as po-
tential antitumor metallodrugs. Chem. Soc. Rev., 2013, 42, 755-
On top of this, the implementation and organization of the 773.
drug discovery process should not have priority over solid [7] Liu, Z.; Romero-Canelon, I.; Qamar, B.; Hearn, J.M.; Habte-
scientific foundations [64]. The only way forward to make mariam, A.; Barry, N.P.; Pizarro, A.M.; Clarkson, G.J.; Sadler, P.J.
an impact in the lives of cancer patients when their tumor The Potent Oxidant Anticancer Activity of Organoiridium Cata-
lysts. Angew. Chem. Int. Ed Engl., 2014, 53, 3941-3946.
cells have spread to unknown locations is to find drugs that [8] Shchekotikhin, A.E.; Glazunova, V.A.; Dezhenkova, L.G.; Luzi-
improve the ability of the current drugs to kill these cells kov, Y.N.; Buyanov, V.N.; Treshalina, H.M.; Lesnaya, N.A.; Ro-
selectively. High-throughput screening approaches should be manenko, V.I.; Kaluzhny, D.N.; Balzarini, J.; Agama, K.; Pom-
designed to detect this type of drugs [50]. mier, Y.; Shtil, A.A.; Preobrazhenskaya, M.N. Synthesis and
evaluation of new antitumor 3-aminomethyl-4,11-
dihydroxynaphtho[2,3-f]indole-5,10-diones. Eur. J. Med. Chem.,
CONCLUSION 2014, 86, 797-805.
Cancer patients need drugs that improve the ability of the [9] Chen, W.; Shen, Y.; Li, Z.; Zhang, M.; Lu, C.; Shen, Y. Design and
existing pharmacological treatments to kill their cancer cells synthesis of 2-phenylnaphthalenoids as inhibitors of DNA topoi-
someraseIIalpha and antitumor agents. Eur. J. Med. Chem., 2014,
without significantly affecting their normal cells. Instead of 86, 782-796.
looking for this type of drugs, the current screening strate- [10] Karki, R.; Park, C.; Jun, K.Y.; Jee, J.G.; Lee, J.H.; Thapa, P.; Ka-
gies typically look for drugs that kill cancer cells at low con- dayat, T.M.; Kwon, Y.; Lee, E.S. Synthesis, antitumor activity, and
centrations or that modulate particular targets involved in structure-activity relationship study of trihydroxylated 2,4,6-
cancer cell proliferation and survival. Because cytotoxic po- triphenyl pyridines as potent and selective topoisomerase II inhibi-
tors. Eur. J. Med. Chem., 2014, 84, 555-565.
tency and ability to modulate these targets do not reliably [11] Wang, M.J.; Liu, Y.Q.; Chang, L.C.; Wang, C.Y.; Zhao, Y.L.;
predict selectivity towards cancer cells, the current screening Zhao, X.B.; Qian, K.; Nan, X.; Yang, L.; Yang, X.M.; Hung, H.Y.;
approaches result in the selection of ineffective compounds Yang, J.S.; Kuo, D.H.; Goto, M.; Morris-Natschke, S.L.; Pan, S.L.;
and probably in the failure to detect promising anticancer Teng, C.M.; Kuo, S.C.; Wu, T.S.; Wu, Y.C.; Lee, K.H. Design,
synthesis, mechanisms of action, and toxicity of novel 20(s)-
drugs. In the first case, researchers waste resources trying to sulfonylamidine derivatives of camptothecin as potent antitumor
prove in animal models and in cancer patients the anticancer agents. J. Med. Chem., 2014, 57, 6008-6018.
activity of ineffective compounds. In the second case, re- [12] Hayashi, Y.; Takeno, H.; Chinen, T.; Muguruma, K.; Okuyama, K.;
searchers discard compounds that could change the lives of Taguchi, A.; Takayama, K.; Yakushiji, F.; Miura, M.; Usui, T.;
cancer patients. Hayashi, Y. Development of a new benzophenone-
diketopiperazine-type potent antimicrotubule agent possessing a 2-
This article describes an alternative screening approach. pyridine structure. ACS Med. Chem. Lett., 2014, 5, 1094-1098.
It is based on establishing the most adequate conditions to [13] Guan, Q.; Han, C.; Zuo, D.; Zhai, M.; Li, Z.; Zhang, Q.; Zhai, Y.;
Jiang, X.; Bao, K.; Wu, Y.; Zhang, W. Synthesis and evaluation of
answer the question: Can the experimental compounds im- benzimidazole carbamates bearing indole moieties for antiprolifera-
prove the ability of the existing anticancer drugs to kill can- tive and antitubulin activities. Eur. J. Med. Chem., 2014, 87C, 306-
cer cells without significantly affecting nonmalignant cells? 315.
This patient-oriented approach is easy to implement and can [14] Yang, Z.; Wu, W.; Wang, J.; Liu, L.; Li, L.; Yang, J.; Wang, G.;
Cao, D.; Zhang, R.; Tang, M.; Wen, J.; Zhu, J.; Xiang, W.; Wang,
help researchers assess in vitro anticancer activity more re- F.; Ma, L.; Xiang, M.; You, J.; Chen, L. Synthesis and biological
liably. evaluation of novel millepachine derivatives as a new class of tubu-
lin polymerization inhibitors. J. Med. Chem., 2014, 57, 7977-7989.
CONFLICT OF INTEREST [15] Kamal, A.; Srinivasa, R.T.; Polepalli, S.; Shalini, N.; Reddy, V.G.;
Subba Rao, A.V.; Jain, N.; Shankaraiah, N. Synthesis and biologi-
The authors confirm that this article content has no con- cal evaluation of podophyllotoxin congeners as tubulin polymeriza-
flict of interest. tion inhibitors. Bioorg. Med. Chem., 2014, 22, 5466-5475.
A Simple and Reliable Approach for Assessing Anticancer Activity Current Medicinal Chemistry, 2015, Vol. 22, No. 11 1333

[16] Zhu, C.; Zuo, Y.; Wang, R.; Liang, B.; Yue, X.; Wen, G.; Shang, tives as new farnesyl diphosphate analogues: synthesis and in vitro
N.; Huang, L.; Chen, Y.; Du, J.; Bu, X. Discovery of potent cyto- antimicrobial and cytotoxic activities. Eur. J. Med. Chem., 2012,
toxic ortho-aryl chalcones as new scaffold targeting tubulin and mi- 58, 591-612.
tosis with affinity-based fluorescence. J. Med. Chem., 2014, 57, [32] Vega-Perez, J.M.; Palo-Nieto, C.; Vega-Holm, M.; Gongora-
6364-6382. Vargas, P.; Calderon-Montano, J.M.; Burgos-Moron, E.; Lopez-
[17] Moreau, P.; Dezhenkova, L.G.; Anizon, F.; Nauton, L.; Thery, V.; Lazaro, M.; Iglesias-Guerra, F. Aziridines from alkenyl-beta-D-
Liang, S.; Kaluzhny, D.N.; Shtil, A.A. New Potent and Selective galactopyranoside derivatives: Stereoselective synthesis and in vi-
Inhibitor of Pim-1/3 Protein Kinases Sensitizes Human Colon Car- tro selective anticancer activity. Eur. J. Med. Chem., 2013, 70C,
cinoma Cells to Doxorubicin. Anticancer Agents Med. Chem., 380-392.
2014, 14, 1228-1236. [33] Lopez-Lazaro, M.; Willmore, E.; Jobson, A.; Gilroy, K.L.; Curtis,
[18] Sangani, C.B.; Makawana, J.A.; Duan, Y.T.; Yin, Y.; Teraiya, H.; Padget, K.; Austin, C.A. Curcumin induces high levels of
S.B.; Thumar, N.J.; Zhu, H.L. Design, synthesis and molecular topoisomerase I- and II-DNA complexes in K562 leukemia cells. J
modeling of biquinoline-pyridine hybrids as a new class of poten- Nat. Prod., 2007, 70, 1884-1888.
tial EGFR and HER-2 kinase inhibitors. Bioorg. Med. Chem. Lett., [34] Burgos-Moron, E.; Calderon-Montano, J.M.; Salvador, J.; Robles,
2014, 24, 4472-4476. A.; Lopez-Lazaro, M. The dark side of curcumin. Int. J. Cancer,
[19] Wu, J.; Chen, W.; Xia, G.; Zhang, J.; Shao, J.; Tan, B.; Zhang, C.; 2010, 126, 1771-1775.
Yu, W.; Weng, Q.; Liu, H.; Hu, M.; Deng, H.; Hao, Y.; Shen, J.; [35] Calderon-Montano, J.M.; Madrona, A.; Burgos-Moron, E.; Orta,
Yu, Y. Design, Synthesis, and Biological Evaluation of Novel Con- M.L.; Mateos, S.; Espartero, J.L.; Lopez-Lazaro, M. Selective Cy-
formationally Constrained Inhibitors Targeting EGFR. ACS Med. totoxic Activity of New Lipophilic Hydroxytyrosol Alkyl Ether
Chem. Lett., 2013, 4, 974-978. Derivatives. J. Agric. Food Chem., 2013, 61, 5046-5053.
[20] Sampson, P.B.; Liu, Y.; Patel, N.K.; Feher, M.; Forrest, B.; Li, [36] Wang, Y.; Lonard, D.M.; Yu, Y.; Chow, D.C.; Palzkill, T.G.;
S.W.; Edwards, L.; Laufer, R.; Lang, Y.; Ban, F.; Awrey, D.E.; Wang, J.; Qi, R.; Matzuk, A.J.; Song, X.; Madoux, F.; Hodder, P.;
Mao, G.; Plotnikova, O.; Leung, G.; Hodgson, R.; Mason, J.; Wei, Chase, P.; Griffin, P.R.; Zhou, S.; Liao, L.; Xu, J.; O'Malley, B.W.
X.; Kiarash, R.; Green, E.; Qiu, W.; Chirgadze, N.Y.; Mak, T.W.; Bufalin is a potent small-molecule inhibitor of the steroid receptor
Pan, G.; Pauls, H.W. The Discovery of Polo-Like Kinase 4 Inhibi- coactivators SRC-3 and SRC-1. Cancer Res., 2014, 74, 1506-1517.
tors: Design and Optimization of Spiro[cyclopropane- [37] Clifford, R.J.; Kaplan, J.H. Human breast tumor cells are more
1,3'[3H]indol]-2'(1'H)-ones as Orally Bioavailable Antitumor resistant to cardiac glycoside toxicity than non-tumorigenic breast
Agents. J. Med. Chem., 2014 [Epub ahead of print]. cells. PLoS. One., 2013, 8, e84306.
[21] Duan, Y.T.; Yao, Y.F.; Huang, W.; Makawana, J.A.; Teraiya, S.B.; [38] Calderon-Montano, J.M.; Burgos-Moron, E.; Orta, M.L.; Garcia-
Thumar, N.J.; Tang, D.J.; Tao, X.X.; Wang, Z.C.; Jiang, A.Q.; Zhu, Dominguez, I.; Maldonado-Navas, D.; Lopez-Lazaro, M. Bufalin is
H.L. Synthesis, biological evaluation, and molecular docking stud- a steroid receptor coactivator inhibitor—Letter. Cancer Res., 2015,
ies of novel 2-styryl-5-nitroimidazole derivatives containing 1,4- doi: 10.1158/0008-5472.CAN-14-0398 (in press).
benzodioxan moiety as FAK inhibitors with anticancer activity. [39] Calderon-Montano, J.M.; Burgos-Moron, E.; Lopez-Lazaro, M.
Bioorg. Med. Chem., 2014, 22, 2947-2954. The in vivo antitumor activity of cardiac glycosides in mice
[22] Dao, P.; Smith, N.; Tomkiewicz-Raulet, C.; Yen-Pon, E.; xenografted with human cancer cells is probably an experimental
Camacho-Artacho, M.; Lietha, D.; Herbeuval, J.P.; Coumoul, X.; artifact. Oncogene, 2014, 33, 2947-2948.
Garbay, C.; Chen, H. Design, Synthesis, and Evaluation of Novel [40] Zhang, H.; Qian, D.Z.; Tan, Y.S.; Lee, K.; Gao, P.; Ren, Y.R.; Rey,
Imidazo[1,2-a][1,3,5]triazines and Their Derivatives as Focal Ad- S.; Hammers, H.; Chang, D.; Pili, R.; Dang, C.V.; Liu, J.O.; Se-
hesion Kinase Inhibitors with Antitumor Activity. J. Med. Chem., menza, G.L. Digoxin and other cardiac glycosides inhibit HIF-
2015, 58, 237-251. 1alpha synthesis and block tumor growth. Proc. Natl. Acad. Sci. U.
[23] Young, D.D.; Connelly, C.M.; Grohmann, C.; Deiters, A. Small S. A., 2008, 105, 19579-19586.
molecule modifiers of microRNA miR-122 function for the treat- [41] Hiyoshi, H.; Abdelhady, S.; Segerstrom, L.; Sveinbjornsson, B.;
ment of hepatitis C virus infection and hepatocellular carcinoma. J. Nuriya, M.; Lundgren, T.K.; Desfrere, L.; Miyakawa, A.; Yasui,
Am. Chem. Soc., 2010, 132, 7976-7981. M.; Kogner, P.; Johnsen, J.I.; Andang, M.; Uhlen, P. Quiescence
[24] Xiao, Z.; Li, C.H.; Chan, S.L.; Xu, F.; Feng, L.; Wang, Y.; Jiang, and gammaH2AX in neuroblastoma are regulated by oua-
J.D.; Sung, J.J.; Cheng, C.H.; Chen, Y. A small-molecule modula- bain/Na,K-ATPase. Br. J. Cancer, 2012, 106, 1807-1815.
tor of the tumor-suppressor miR34a inhibits the growth of hepato- [42] Tailler, M.; Senovilla, L.; Lainey, E.; Thepot, S.; Metivier, D.;
cellular carcinoma. Cancer Res., 2014, 74, 6236-6247. Sebert, M.; Baud, V.; Billot, K.; Fenaux, P.; Galluzzi, L.; Boehrer,
[25] Lopez-Lazaro, M. (2014) Experimental Cancer Pharmacology for S.; Kroemer, G.; Kepp, O. Antineoplastic activity of ouabain and
Researchers: At What Concentration Should my Drug Kill Cancer pyrithione zinc in acute myeloid leukemia. Oncogene, 2012, 31,
Cells so that it has Potential for Cancer Therapy? Amazon Digital 3536-3546.
Services, Inc [43] Zhang, D.M.; Liu, J.S.; Deng, L.J.; Chen, M.F.; Yiu, A.; Cao, H.H.;
[26] Cheng, W.H.; Cao, B.; Shang, H.; Niu, C.; Zhang, L.M.; Zhang, Tian, H.Y.; Fung, K.P.; Kurihara, H.; Pan, J.X.; Ye, W.C. Areno-
Z.H.; Tian, D.L.; Zhang, S.; Chen, H.; Zou, Z.M. Synthesis and bufagin, a natural bufadienolide from toad venom, induces apopto-
evaluation of novel podophyllotoxin derivatives as potential anti- sis and autophagy in human hepatocellular carcinoma cells through
tumor agents. Eur. J. Med. Chem., 2014, 85, 498-507. inhibition of PI3K/Akt/mTOR pathway. Carcinogenesis, 2013, 34,
[27] Kim, J.H.; Yu, J.; Alexander, V.; Choi, J.H.; Song, J.; Lee, H.W.; 1331-1342.
Kim, H.O.; Choi, J.; Lee, S.K.; Jeong, L.S. Structure-activity rela- [44] Lopez-Lazaro, M. Digoxin, HIF-1, and cancer. Proc. Natl. Acad.
tionships of 2'-modified-4'-selenoarabinofuranosyl-pyrimidines as Sci. U. S. A., 2009, 106, E26.
anticancer agents. Eur. J. Med. Chem., 2014, 83, 208-225. [45] Calderon-Montano, J.M.; Burgos-Moron, E.; Lopez-Lazaro, M.
[28] Alanazi, A.M.; Abdel-Aziz, A.A.; Al Suwaidan, I.A.; Abdel- Comment on 'Quiescence and gammaH2AX in neuroblastoma are
Hamide, S.G.; Shawer, T.Z.; El Azab, A.S. Design, synthesis and regulated by Ouabain/Na,K-ATPase': ouabain and cancer. Br. J.
biological evaluation of some novel substituted quinazolines as an- Cancer, 2013, 108, 2189-2190.
titumor agents. Eur. J. Med. Chem., 2014, 79, 446-454. [46] Kerbel, R.S. Human tumor xenografts as predictive preclinical
[29] Rodrigues, F.A.; Bomfim, I.S.; Cavalcanti, B.C.; Pessoa, C.O.; models for anticancer drug activity in humans: better than com-
Wardell, J.L.; Wardell, S.M.; Pinheiro, A.C.; Kaiser, C.R.; No- monly perceived-but they can be improved. Cancer Biol. Ther.,
gueira, T.C.; Low, J.N.; Gomes, L.R.; de Souza, M.V. Design, syn- 2003, 2, S134-S139.
thesis and biological evaluation of (E)-2-(2- [47] Hutchinson, L.; Kirk, R. High drug attrition rates--where are we
arylhydrazinyl)quinoxalines, a promising and potent new class of going wrong? Nat. Rev. Clin. Oncol., 2011, 8, 189-190.
anticancer agents. Bioorg. Med. Chem. Lett., 2014, 24, 934-939. [48] Begley, C.G.; Ellis, L.M. Drug development: Raise standards for
[30] Liu, W.; Gust, R. Metal N-heterocyclic carbene complexes as po- preclinical cancer research. Nature, 2012, 483, 531-533.
tential antitumor metallodrugs. Chem. Soc. Rev., 2013, 42, 755- [49] Calderon-Montano, J.M.; Burgos-Moron, E.; Orta, M.L.; Mal-
773. donado-Navas, D.; Garcia-Dominguez, I.; Lopez-Lazaro, M.
[31] Vega-Perez, J.M.; Perinan, I.; Argandona, M.; Vega-Holm, M.; Evaluating the cancer therapeutic potential of cardiac glycosides.
Palo-Nieto, C.; Burgos-Moron, E.; Lopez-Lazaro, M.; Vargas, C.; Biomed. Res. Int., 2014, 2014:794930.
Nieto, J.J.; Iglesias-Guerra, F. Isoprenyl-thiourea and urea deriva-
1334 Current Medicinal Chemistry, 2015, Vol. 22, No. 11 Miguel López-Lázaro

[50] Lopez-Lazaro, M. How many times should we screen a chemical [60] Niles, A.L.; Riss, T.L. Multiplexed viability, cytotoxicity, and
library to discover an anticancer drug? 2014, Drug Discov. Today caspase activity assays. Methods Mol. Biol., 2015, 1219, 21-33.
doi: 10.1016/j.drudis.2014.12.006. [Epub ahead of print] [61] Hatzis, C.; Bedard, P.L.; Birkbak, N.J.; Beck, A.H.; Aerts, H.J.;
[51] Elhalem, E.; Recio, R.; Werner, S.; Lieder, F.; Calderon-Montano, Stem, D.F.; Shi, L.; Clarke, R.; Quackenbush, J.; Haibe-Kains, B.
J.M.; Lopez-Lazaro, M.; Fernandez, I.; Khiar, N. Sulforaphane Enhancing reproducibility in cancer drug screening: how do we
homologues: Enantiodivergent synthesis of both enantiomers, acti- move forward? Cancer Res., 2014, 74, 4016-4023.
vation of the Nrf2 transcription factor and selective cytotoxic activ- [62] Monks, A.; Scudiero, D.; Skehan, P.; Shoemaker, R.; Paull, K.;
ity. Eur. J. Med. Chem., 2014, 87C, 552-563. Vistica, D.; Hose, C.; Langley, J.; Cronise, P.; Vaigro-Wolff, A.; .
[52] Antonelli, A.; Bocci, G.; La Motta, C.; Ferrari, S.M.; Fallahi, P.; Feasibility of a high-flux anticancer drug screen using a diverse
Fioravanti, A.; Sartini, S.; Minuto, M.; Piaggi, S.; Corti, A.; Ali, panel of cultured human tumor cell lines. J. Natl. Cancer Inst.,
G.; Berti, P.; Fontanini, G.; Danesi, R.; Da Settimo, F.; Miccoli, P. 1991, 83, 757-766.
Novel pyrazolopyrimidine derivatives as tyrosine kinase inhibitors [63] Vichai, V.; Kirtikara, K. Sulforhodamine B colorimetric assay for
with antitumoral activity in vitro and in vivo in papillary dediffer- cytotoxicity screening. Nat. Protoc., 2006, 1, 1112-1116.
entiated thyroid cancer. J. Clin. Endocrinol. Metab., 2011, 96, [64] Sams-Dodd, F. Is poor research the cause of the declining produc-
E288-E296. tivity of the pharmaceutical industry? An industry in need of a
[53] Fallahi, P.; Ferrari, S.M.; Mazzi, V.; Vita, R.; Benvenga, S.; An- paradigm shift. Drug Discov. Today, 2013, 18, 211-217.
tonelli, A. Personalization of targeted therapy in advanced thyroid [65] Strumberg, D.; Richly, H.; Hilger, R.A.; Schleucher, N.; Korfee, S.;
cancer. Curr. Genomics., 2014, 15, 190-202. Tewes, M.; Faghih, M.; Brendel, E.; Voliotis, D.; Haase, C.G.;
[54] Carrasco, E.; Alvarez, P.J.; Prados, J.; Melguizo, C.; Rama, A.R.; Schwartz, B.; Awada, A.; Voigtmann, R.; Scheulen, M.E.; Seeber,
Aranega, A.; Rodriguez-Serrano, F. Cancer stem cells and their S. Phase I clinical and pharmacokinetic study of the Novel Raf
implication in breast cancer. Eur. J. Clin. Invest., 2014, 44, 678- kinase and vascular endothelial growth factor receptor inhibitor
687. BAY 43-9006 in patients with advanced refractory solid tumors. J.
[55] Dawood, S.; Austin, L.; Cristofanilli, M. Cancer Stem Cells: Impli- Clin. Oncol., 2005, 23, 965-972.
cations for Cancer Therapy. Oncology (Williston. Park). 2014, 28, [66] Cheng, A.L.; Kang, Y.K.; Chen, Z.; Tsao, C.J.; Qin, S.; Kim, J.S.;
202935. Luo, R.; Feng, J.; Ye, S.; Yang, T.S.; Xu, J.; Sun, Y.; Liang, H.;
[56] Sandoval, M.; Harris, L. Cancer stem cells. Oncology (Williston. Liu, J.; Wang, J.; Tak, W.Y.; Pan, H.; Burock, K.; Zou, J.; Voliotis,
Park). 2014, 28, 202936. D.; Guan, Z. Efficacy and safety of sorafenib in patients in the
[57] Carrasco, E.; Alvarez, P.J.; Melguizo, C.; Prados, J.; Alvarez- Asia-Pacific region with advanced hepatocellular carcinoma: a
Manzaneda, E.; Chahboun, R.; Messouri, I.; Vazquez-Vazquez, phase III randomised, double-blind, placebo-controlled trial. Lancet
M.I.; Aranega, A.; Rodriguez-Serrano, F. Novel merosesquiterpene Oncol., 2009, 10, 25-34.
exerts a potent antitumor activity against breast cancer cells in vitro [67] Llovet, J.M.; Ricci, S.; Mazzaferro, V.; Hilgard, P.; Gane, E.;
and in vivo. Eur. J. Med. Chem., 2014, 79, 1-12. Blanc, J.F.; de Oliveira, A.C.; Santoro, A.; Raoul, J.L.; Forner, A.;
[58] Riss, T.L.; Moravec, R.A.; Niles, A.L.; Benink, H.A.; Worzella, Schwartz, M.; Porta, C.; Zeuzem, S.; Bolondi, L.; Greten, T.F.;
T.J.; Minor, L.; Riss, T.L.; Moravec, R.A.; Niles, A.L. Cell Viabil- Galle, P.R.; Seitz, J.F.; Borbath, I.; Haussinger, D.; Giannaris, T.;
ity Assays. Cytotoxicity testing: measuring viable cells, dead cells, Shan, M.; Moscovici, M.; Voliotis, D.; Bruix, J. Sorafenib in ad-
and detecting mechanism of cell death. Methods Mol. Biol., 2011, vanced hepatocellular carcinoma. N. Engl. J. Med., 2008, 359, 378-
740, 103-114. 390.
[59] Niles, A.L.; Moravec, R.A.; Riss, T.L. In vitro viability and cyto-
toxicity testing and same-well multi-parametric combinations for
high throughput screening. Curr. Chem. Genomics, 2009, 3, 33-41.

Received: November 04, 2014 Revised: January 18, 2015 Accepted: January 25, 2015

You might also like