ArticlePDF Available

Chemotherapy and Dietary Phytochemical Agents

Authors:
  • NGO Praeventio Tartu Estonia

Abstract and Figures

Chemotherapy has been used for cancer treatment already for almost 70 years by targeting the proliferation potential and metastasising ability of tumour cells. Despite the progress made in the development of potent chemotherapy drugs, their toxicity to normal tissues and adverse side effects in multiple organ systems as well as drug resistance have remained the major obstacles for the successful clinical use. Cytotoxic agents decrease considerably the quality of life of cancer patients manifesting as acute complaints and impacting the life of survivors also for years after the treatment. Toxicity often limits the usefulness of anticancer agents being also the reason why many patients discontinue the treatment. The nutritional approach may be the means of helping to raise cancer therapy to a new level of success as supplementing or supporting the body with natural phytochemicals cannot only reduce adverse side effects but improve also the effectiveness of chemotherapeutics. Various plant-derived compounds improve the efficiency of cytotoxic agents, decrease their resistance, lower and alleviate toxic side effects, reduce the risk of tumour lysis syndrome, and detoxify the body of chemotherapeutics. The personalised approach using various phytochemicals provides thus a new dimension to the standard cancer therapy for improving its outcome in a complex and complementary way.
Content may be subject to copyright.
Hindawi Publishing Corporation
Chemotherapy Research and Practice
Volume 2012, Article ID 282570, 11 pages
doi:10.1155/2012/282570
Review Article
Chemotherapy and Dietary Phytochemical Agents
Katrin Sak
NGO Praeventio, N¨
aituse 22-3, 50407 Tartu, Estonia
Correspondence should be addressed to Katrin Sak, katrin.sak.001@mail.ee
Received 8 October 2012; Revised 23 November 2012; Accepted 29 November 2012
Academic Editor: G. J. Peters
Copyright © 2012 Katrin Sak. This is an open access article distributed under the Creative Commons Attribution License, which
permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
Chemotherapy has been used for cancer treatment already for almost 70 years by targeting the proliferation potential and
metastasising ability of tumour cells. Despite the progress made in the development of potent chemotherapy drugs, their toxicity
to normal tissues and adverse side eects in multiple organ systems as well as drug resistance have remained the major obstacles
for the successful clinical use. Cytotoxic agents decrease considerably the quality of life of cancer patients manifesting as acute
complaints and impacting the life of survivors also for years after the treatment. Toxicity often limits the usefulness of anticancer
agents being also the reason why many patients discontinue the treatment. The nutritional approach may be the means of helping
to raise cancer therapy to a new level of success as supplementing or supporting the body with natural phytochemicals cannot only
reduce adverse side eects but improve also the eectiveness of chemotherapeutics. Various plant-derived compounds improve
the eciency of cytotoxic agents, decrease their resistance, lower and alleviate toxic side eects, reduce the risk of tumour lysis
syndrome, and detoxify the body of chemotherapeutics. The personalised approach using various phytochemicals provides thus a
new dimension to the standard cancer therapy for improving its outcome in a complex and complementary way.
1. Introduction
Chemotherapy is one of the principal modes of the treatment
of cancer patients [1]. It was first used to treat advanced lym-
phoma in the late 1940s after it became known that the use
of mustard gas in the World War I caused leukopenia [2,
3]. Shortly after the World War II, it was also found that
folic acid stimulates the proliferation of acute lymphoblastic
leukaemia cells and antagonistic analogues to folic acid, first
aminopterin and then amethopterin (now known as metho-
trexate) induced the remission in children with acute lym-
phoblastic leukaemia [4]. Although the role of chemotherapy
in the treatment of common, epithelial malignancies was
limited to the treatment of symptomatic metastatic disease
for almost 30 years, nowadays cancer chemotherapy has three
main applications: it is curative for a small number of malig-
nancies including childhood leukaemia, Hodgkin’s and non-
Hodgkin’s lymphoma, and germ cell malignancies; it has a
palliative role for most metastatic epithelial malignancies;
and it has an adjuvant role in several types of resected
epithelial malignancies [3].
By definition, chemotherapy treatment should interfere
with the biochemical program that is involved or committed
to cellular replication and cause selective cell death. At that,
the host cell should be able to adapt and recover from
toxicity [2]. Many chemotherapeutic agents kill cancer cells
oxidatively via the production of reactive oxygen species and
the induction of either apoptosis or necrosis of tumorous
cells [2,5,6]; whereas others act on various components of
cellular metabolism influencing activities of dierent en-
zymes needful for cell division.
Cancer treatment is targeted at its proliferation potential
and its ability to metastasise; hence, the majority of chemo-
therapy drugs take advantage of the fact that cancer cells
divide rapidly [7]. Chemotherapy agents can be divided into
severalcategoriesbasedonthefactorssuchashowtheywork,
their chemical structure, and their relationship to another
drug. The most important categories of chemotherapeutics
include alkylating agents (e.g., cyclophosphamide, ifosfam-
ide, melphalan, busulfan), antimetabolites (e.g., 5-fluorou-
racil, capecitabine, methotrexate, gemcitabine), antitumour
antibiotics (e.g., daunorubicin, doxorubicin, epirubicin), to-
poisomerase inhibitors (e.g., topotecan, irinotecan, etopo-
side, teniposide), and mitotic inhibitors (e.g., paclitaxel,
docetaxel, vinblastine, vincristine) [8,9]. Most chemother-
apeutic drugs target the cell cycle machinery relying on the
2Chemotherapy Research and Practice
dierence in the frequency of cell division to dierentiate
between the cancer clones and normal cells. Within this
process slow-growing cancer clones will survive and evolve
into new fast growing strains. Chemotherapy is able to kill
omost of the susceptible tumorous cells succeeding to send
cancer into remission for weeks or months after which it
reemerges as a more aggressive organism [10,11]. In fact, the
more chemotherapy is given, the higher is the aggressiveness
of relapse. In these cases, chemotherapy may indirectly select
the most resistant mutant cell for clonal expansion [2].
Cancer is a highly heterogeneous disease, especially
in its advanced forms [2], and such heterogeneity gives
it an advantage to survive under selection pressure of
drugs [12]. Moreover, the unique characteristics of tumour
microenvironment (hypoxia, low extracellular pH, high
interstitial fluid pressure), developed at least in part as a
result of the malformed tumour vasculature, act as barriers
to chemotherapy impairing the transport and delivery of
circulating therapeutic molecules in tumour tissue [13
16]. Cancer hypoxia can thus reduce the eectiveness of
drugs [13,15,17]. Another major obstacle of conventional
chemotherapy proceeds from the drug resistance [1,12,18,
19]. Some tumours are intrinsically resistant to certain drugs,
whereas others can acquire resistance after treatment [1,18
20]. Cancer cells can often develop resistance not only to the
agent, which they have been exposed to, but also to other
drugs and chemicals that they have not encountered. There
are a number of mechanisms mediating such multidrug
resistance [2,20,21]. Upregulation of drug eux ATP-
binding cassette (ABC) transporters, such as P-glycoprotein
(P-gp), multidrug resistance protein 1 (MRP1), and breast
cancer resistance protein (BCRP), may be responsible for the
resistance to many chemotherapeutics aecting disposition
of these drugs in the tumour cells and modifying seriously
the clinical outcome [1,20,22,23]. Although the first
human ABC drug transporter P-gp was identified already for
more than 35 years ago, there are still no clinically applicable
inhibitors of ABC transporters available to date and this
failureislargelyduetotheunfavourabletoxicsideeects
of tested chemical compounds. Novel chemosensitisers as
inhibitors of dierent MDR-linked ABC transporters are
actively explored turning increasing attention also to various
naturalproducts[23,24].
2. Factors Affecting the
Effectiveness of Chemotherapy
Eectiveness of chemotherapy depends on various factors,
including properties of cancer cells (if tumour is hypoxic
or mitochondrial function is severely compromised, or the
number of mitochondria within the cancer cell is low,
chemotherapy will be of limited value, only increasing the
clonal selection of the most resistant and possibly also the
most aggressive cancer phenotype [2]); tumour size (the
microscopic form of tumour is much more successfully
treated than macroscopic cancer [2]); number of chemother-
apy cycles [25]; administrating polychemotherapy versus
monotherapy (polychemotherapy may be more active than
single agent, whereas the order of administration of drugs as
well as their time schedule is also important: combining
drugs with dierent modes of action may lead to enhanced or
even synergistic antitumour eects without injuring the host
[2528]); multitargeted approach (targeting both the cancer
cell and its microenvironment might increase the treatment
eciency [29]); and the phase of the circadian cycle [30].
Circadian periodicity in cell proliferation provides an oppor-
tunity to improve the tolerability or eciency (or both) of
anticancer treatment by targeting its timing at certain stages
of the circadian system of the host or the tumour [3037].
It has been shown in animal studies that the toxicity and
ecacy of more than 30 anticancer agents vary by more than
50% depending on the circadian time when the agent is given
[2,31,32,35,36]. The peak time of drug delivery to achieve
optimal tolerability is characteristic for each chemothera-
peutic agent and the circadian time of administration of
cancer chemotherapeutic agents when they are best tolerated
also usually shows the highest ecacy against the tumour
[31,35,37]. Such chronotherapy may have a significant
impact on the treatment success and can ultimately lead to a
better tumour management [33,35,37]. The patient’s gender
could also have an essential role in the determination of
optimal chronotherapeutic schedule [2,36].
Cancer is a systemic and not a local disease [2]. Metastatic
spread of tumour cells may take place already at an early
stage of the malignancy; however, little is known about
the tumour-biological parameters of such disseminated cells
[38]. Metastatic foci might vary with genotype, phenotype,
and drug response [21], whereas expression profiles and
signalling pathways between primary tumour and metastatic
tissue can be dierent [27,38]. The considerable number of
adjuvant therapy failures may be explained by such molec-
ular dierences between the surgically removed and histo-
pathologically examined primary tumour cells and the
remaining disseminated tumour cells [38]. Namely, the latest
would be the real targets for any systemic therapy to prevent
them forming a clinically relevant metastatic disease [21,38].
3. Typical Side Effects of Chemotherapy
Although the desired goal of chemotherapy is to eliminate
the tumour cells, diverse ranges of normal cell types are also
aected, leading to many adverse side eects in multiple
organ systems [5,3943]. Such debilitating eects are a
major clinical problem [44], whereas the toxicity often limits
the usefulness of anticancer agents [44,45].
Knowing how the chemotherapy agent works is impor-
tant in predicting its side eects. For instance, treatment with
alkylating agents and topoisomerase II inhibitors increases
the risk of secondary cancer (acute leukaemia); anthracy-
clines (like doxorubicin) induce cardiotoxicity; and mitotic
inhibitors have the potential to cause peripheral nerve
damage [6].
The most common acute complaints of cancer patients
undergoing cytotoxic therapy are fatigue, nausea, vomiting,
malaise, diarrhoea, mucositis, pain, rashes, infections, head-
aches, and other problems [2,42,44,46,47]. Normal
haematopoietic cells, intestinal epithelial cells, and hair
matrix keratinocytes are often susceptible to the toxic eects
Chemotherapy Research and Practice 3
of anticancer agents [44]. Over 75% of cancer patients suer
from therapy-associated fatigue; however, only about one-
third of treating physicians recognise this problem [42].
Fatigue is related to a reduced mitochondrial function of
tumorous cells and is often a significant reason why patients
discontinue the treatment [42,48]. Thus, cytotoxic agents
decrease considerably the quality of life of patients [43,
48,49]. Through the induction of nausea and vomiting,
diculties in swallowing, dry mouth, alterations in taste and
smell, depression, poor energy, and aversion to food cyto-
toxic drugs aect also the nutritional status of patients [2,47]
showing that chemotherapy is a sucient stressor in causing
malnutrition. It must be appreciated that malnutrition is the
reason why majority of the cancer patients die [2,50].
Most cytotoxic drugs have immune suppressive side
eects. Many chemotherapeutics kill dividing haematopoi-
etic cells manifesting as profound neutropenia and cytopenia
resulting in decreased immunity, increased susceptibility to
infections, and elevated risk of bleeding [2,3]. Coming from
the requirement of the bone marrow to repopulate white
cells and platelets in the blood, drugs are often administered
episodically followed by the drug-free intervals of 2-3 weeks.
Such scheme helps to minimise the chance of infection or
bleeding but allows also the tumour to recover [51,52].
Cancer patients frequently complain of neurological side
eects [53]. Such eects range from abnormalities in brain
volume and integrity detected by magnetic resonance imag-
ing on patients after chemotherapy to dierent clinical symp-
toms; manifesting acutely or as delayed neurotoxicities only
becoming apparent years after treatment [40,41,54]. Neu-
rological complications include memory loss and cogni-
tive dysfunction, seizures, vision loss, dementia, leukoen-
cephalopathy, cerebral infarctions, and other problems [41,
53,54]. These symptoms are commonly referred to as Chemo
Brain [53,55]aecting some 4–75% of cancer patients fol-
lowing with chemotherapy [55]. Nearly all frequently used
chemotherapeutic agents can cause adverse neurological
eects [40,41]. Chemotherapy-induced cognitive changes
might be associated with neurotoxic eects of inflammation
and cytokine deregulation. Treatment-induced changes in
the level of oestrogen and testosterone can be related to
cognitive decline. Also, corticosteroids, which are commonly
used as part of chemotherapy regimens or to manage side
eects such as nausea, alter neuroendocrine functioning and
cognition [40].
The most common long-term health problems of adju-
vant chemotherapy include poor memory and concen-
tration, visual deterioration, musculoskeletal complaints
including early onset osteoporosis, poor sleep patterns, skin
changes, sexual dysfunction, and chronic fatigue [3]. This
complex of problems is suggestive of accelerated aging lead-
ing potentially to early onset frailty [3,40]. Such long-term
toxicity can have an impact on the quality of life of cancer
survivals that could last for years [3].
Most chemotherapy drugs are genotoxic likely causing
epigenetic and genetic damage [2,44,53]. Many cancer
patients encounter thus the problem of developing second
malignancies as a result of treatment [44,48,53]. The com-
mon secondary tumours are a variety of acute leukaemias
and non-Hodgkin’s lymphomas, less common are carcino-
mas of the urinary bladder and other malignancies, which are
usually refractory to treatment. Secondary leukaemias con-
stitute approximately 10% of all leukaemias having the
average latent interval from the diagnosis and treatment of
the primary neoplasm to development of acute leukaemia for
four to six years. The International Agency for Research on
Cancer (IARC) has identified 20 single chemotherapeutic
agents or regimens which cause cancer in humans and about
50 others that are suspect [2]. Chemotherapy-associated
immunosuppression can result in an increased rate of
infection by oncogenic viruses which further increase the risk
of secondary cancers [2].
Current treatment protocols often apply multiagent
chemotherapy [54] and this may even increase the extent of
adverse side eects [50]. Several serious complications can
cause discontinuation of therapy, prolong the duration of
stay in hospitals, and may aect the overall prognosis and
outcome of the disease [50]. It is important to bear in mind
that, in general, older cancer patients are more susceptible
to treatment-related complications than younger individuals
[56]. One of the most critical and life-threatening adverse
conditions of chemotherapy needing immediate intervention
is tumour lysis syndrome. It results from the massive and
abrupt destruction and lysis of malignant cells and subse-
quent release of intracellular ions and metabolites into the
bloodstream leading to hyperuricemia, hyperphosphatemia,
hypocalcemia, and hyperkalemia [57,58]. This syndrome is
accompanied by renal failure and metabolic acidosis increas-
ing the risk of death [57]. Although the tumour lysis syn-
drome is most frequently observed in patients with haema-
tologic malignancies, it may also occur in solid tumours
[57,58]. These malignancies share the characteristics of a
high proliferative rate, large tumour burden, or high sensi-
tivity to cytotoxic therapy and in principle, any tumour that
is highly responsive to chemotherapeutic drugs, particularly
if the cancer cells die through the necrotic pathway, can
give rise to this severe metabolic syndrome [2,58]. Most of
the complications can be readily managed when they are
recognised early; however, delay in recognition and treat-
ment of metabolic abnormalities may become fatal [58].
4. Nutritional Approach in Chemotherapy
The ecacy of standard oncologic therapies (involving a
combination of surgery, multiple chemotherapeutic agents,
and ionising radiation) has reached a plateau for most of
solid tumours [48]. General protocol with these therapies is
just to follow a watch-and-wait strategy after the therapeutic
administration is concluded [9]. However, this is a period
when supplemental therapies are highly indicated to result in
a higher percentage of successful outcomes [9]. Cancer
patient must be stocked up with needful nutrients already
before receiving treatment in order to ensure the best health
possible before undergoing chemotherapy [59]. The adverse
side eects of anticancer agents exacerbate the nutritional
problems and the proper nutrition should be an integral part
of the treatment program of every cancer patients [47].
Dietary components may act as adaptogens protecting
4Chemotherapy Research and Practice
organism from the adverse eects of intervention or as
modifiers of biologic response [60] exerting additive or even
synergistic eects with pharmaceutical agents [61].
Thus, nutritional approach may be the means of helping
to raise cancer therapy to a new level of success, whereas sup-
plementing or supporting the body with natural compounds
may be the answer in not only reducing the side eects but
also improving the eectiveness of chemotherapy [2]. At
the same time, it is important to bear in mind that some
nutrients can aect the treatment outcome also adversely
by attenuating or inhibiting the therapeutic eect of certain
drugs. Optimal dietary regimen of cancer patients should
thus be prescribed by a nutritional oncologist considering
each individual case separately, whereas the consumption of
supplements on patient’s own initiative may be dangerous
and should be avoided.
5. Purposes of Nutritional Intervention
Intervention with dietary agents in chemotherapy has several
aims. It increases the ecacy of treatment and decreases its
side eects, improves cancer killing through apoptosis rather
than necrosis, reduces drug resistance or increases drug accu-
mulation within cancer cells, detoxifies body of chemothera-
peutics, decreases weight loss and malnutrition, improves the
quality of life, and reduces severity of comorbid conditions.
Dietary compounds may enhance the ecacy of cancer
therapeutics by modifying the activity of key cell prolifer-
ation and survival pathways [48,61,62]. Multiple antiox-
idants are eective in increasing the tumour response to
chemotherapy improving also the survival time of patients
[9,48,59]. Nutrients can be used also to mitigate the toxicity
of chemotherapy [46,48,62]. Reduction in adverse eects of
anticancer agents has been shown when given concurrently
with antioxidants [9,48,59]. Lowering the side eects may
permit to safely administer a higher and possibly more
eective dose of chemotherapeutic drugs [2]. Combination
of nutrients has generally a better eect getting the optimum
therapeutic response and reducing adverse side eects than
dierent nutrients separately. However, possible interactions
between the dietary components during chemotherapy need
to be carefully controlled as some supplements can abolish
the beneficial eects of others [2].
Oxidant stress shifts the mechanisms of cell killing away
from apoptosis to necrosis inducing inflammation and pro-
moting cancer progression. Necrosis will increase also the
risk of developing tumour lysis syndrome. Regulating the
oxidant state with specific antioxidants, supporting mito-
chondrial function, and increasing their numbers within
cancer cells can promote the apoptosis-induced death of
chemotherapy. This reduces the side eects associated with
chemotherapy and may also lower the resistance to antican-
cer agents and the progression of tumour [2,9].
Drug resistance is one of the primary obstacles of
successful chemotherapy. Inhibiting the function of ABC
transporters, such as P-gp and MRP-s, may lead to over-
coming drug resistance providing a possible mechanism
to improve chemotherapeutic eects [2,20]. Although co-
administration of transport inhibitors together with the
actual anticancer drug may enhance drug penetration into
the tumour [24], no real solution to multidrug resistance
has been found to date and no chemical P-gp inhibitors with
clinically satisfying results have been described mainly due
to their high toxicity [23,24]. Therefore, several novel com-
pounds including those isolated from natural sources (or the
so-called “fourth generation chemosensitisers”) are actively
explored and may hopefully provide some solution to
overcome these problems [20,23,24].
The body needs to be detoxified of chemotherapeutics
after the treatment is completed. Many of the anticancer
agents are carcinogenic themselves and the patients may suf-
fer secondary cancers following primary remission from the
initial tumour [2].
Nutritional status of cancer patients is associated with the
outcome of malignant disease while weight loss is related to
shortened survival and lowered response to chemotherapy
[2,47]. Malnutrition is the cause of death for the majority of
cancer patients [2]. Therefore, nutritional support enhances
the chances of complete remission of disease and improves
the quality of life [47].
Cancer patients may be antioxidant deficient and chem-
otherapy further generates free radicals that cause oxidative
damage in dierent organs. It is important to keep comorbid
conditions in mind considering the state of the entire patient
and not just to focus on the cancer. The antioxidant status of
the patient can remain depressed for some months after the
cancer treatment and nutritional support helps to ensure the
best possible general health state [59].
6. Plant-Derived Dietary Agents
in Chemotherapy
The optimal nutritional program proceeds from each indi-
vidual case and considers the needs of a certain patient.
Dietary phytochemical agents influence various aspects of
chemotherapy treatment and their involvement in the cureof
cancer patients is absolutely indicated and needful. Dierent
natural compounds can improve eciency of chemother-
apeutic agents, decrease the resistance of chemotherapeu-
tic drugs, lower and alleviate the adverse side eects of
chemotherapy, reduce the risk of tumour lysis syndrome, and
detoxify the body of chemotherapeutics. At the same time,
it is important to be aware that some phytochemical agents
can have also toxic eects and influence the treatment results
adversely.
Eects of plant-derived dietary agents on treatment out-
put have been intensively explored; however, most of the
studies about interactions between dietary phytochemical
agents and chemotherapy drugs are done using either in vitro
cell systems or in vivo animal experiments (see Ta ble 1 ).
Therefore and before more clinical data will be available,
some precaution must be taken in transferring these results
directly to the patients.
Various plant-derived agents like genistein, curcumin,
epigallocatechin gallate (EGCG), resveratrol, indole-3-car-
binol, and proanthocyanidin have been shown to be able to
aect the ecacy of traditional chemotherapeutic agents [48,
61]. Supplementation with bromelain can also increase the
Chemotherapy Research and Practice 5
Tab le 1: Examples of eects of dietary phytochemical agents on chemotherapy.
Compound Dietary source Chemotherapy drug Eect Biological system Reference
Influence on treatment ecacy
Ginsenosides Cisplatin Enhancement of drug-induced antiproliferative
eect Human breast carcinoma MCF-7 cells [63,64]
Ginseng
5-Fluorouracil Increase in antiproliferative eect Human colorectal cancer HCT-116 cells [63,65]
Curcumin Turmeric Vinorelbine Enhancement of chemotherapeutic ecacy Human squamous cell lung carcinoma H520 cells [66,67]
Catechins/theanine Green tea Doxorubicin Enhancement of antitumour activity Ehrlich ascites carcinoma and M5076 ovarian
sarcoma tumour-bearing mice [9,6870]
Quercetin
Increase in reduction of tumour growth Mice bearing human tumour xenografts [71]
Cisplatin Potentiation of cytotoxic eect Human ovarian and endometrial cancer cell lines [71]
Many foods such as
onions, apples,
berries, and tea Doxorubicin Potentiation of growth-inhibitory activity Doxorubicin-resistant human breast tumour
MCF-7 cells [68,71,72]
Busulfan Synergistic antiproliferative activity Human leukaemia K562 cells [71,73]
Genistein
Cisplatin Increased cytotoxic eect Cisplatin-sensitive and cisplatin-resistant human
2008 ovarian carcinoma cells [9,71]
Attenuation of inhibitory eect of tamoxifen on
tumour cell growth
Oestrogen-dependent human breast cancer
MCF-7 cells [9,74]
Soy foods
Tamo xi fen Attenuation of tamoxifen eect on reducing of
tumour burden
Female Sprague-Dawley rats with induced
mammary tumours [75]
Synergistic growth inhibition Oestrogen receptor-negative human breast
carcinoma MDA-MB-435 cells [9,76]
Daidzein Soy foods Tamoxifen Improvement of drug activity to reduce tumour
burden
Female Sprague-Dawley rats with induced
mammary tumours [9,75]
Tangeretin Tangerine and other
citrus peels Tamo xi fen Complete blocking of growth inhibitory eect of
tamoxifen
Female nude mice inoculated with human
MCF-7/6 mammary adenocarcinoma cells [9,68,77]
Influence on side eects of chemotherapy
Ginsenosides Cyclophosphamide
Protection against drug-induced genotoxicity and
apoptosis in bone marrow cells and peripheral
lymphocytes
Mouse peripheral lymphocytes and bone marrow
cells [78,79]
Ginseng
Cisplatin Attenuation of drug-induced nausea and vomiting Rat model [63,80]
Quercetin
Many foods such as
onions, apples,
berries, and tea
Cisplatin Protection of normal renal tubular cells from drug
toxicity Pig kidney tubular epithelial LLC-PK1 cells [71,81]
Influence on drug resistance
Ginsenosides Ginseng Paclitaxel Chemosensitisation Multidrug-resistant breast cancer cells [63,82]
Catechins/theanine
Doxorubicin Inhibition of drug eux from tumour cells Drug-resistant M5076 ovarian sarcoma
tumour-bearing mice [9,68,70]
Green tea Daunorubicin Increase in drug accumulation in tumour cells Multidrug-resistant P-gp overexpressing human
epidermal carcinoma KB-C2 cells [20,83]
Irinotecan, SN-38 Inhibiting drug transport into biliary elimination
and prolonging half-lives in plasma Male Sprague-Dawley rats [20,84]
6Chemotherapy Research and Practice
Tab le 1: Continued.
Compound Dietary source Chemotherapy drug Eect Biological system Reference
Quercetin
Vincristine Increase in drug uptake in tumour cells Doxorubicin-resistant human myelogenous
leukaemia K562 cells [20,85]
Tamo xi fen Enhancement of drug bioavailability decreasing the
eux by MDR transporters Female Sprague-Dawley rats [20,86]
Paclitaxel Enhancement of drug bioavailability Male Sprague-Dawley rats [20,87]
Many foods such as
onions, apples,
berries, and tea Doxorubicin Potentiation of antitumour eect reducing P-gp
expression
Multidrug-resistant human breast cancer MCF-7
cells [9,20,72]
Topotecan Chemosensitisation Mouse fibrosarcoma WEHI-S cells [71,88]
Gemcitabine Chemosensitisation Mouse fibrosarcoma WEHI-S cells [71,88]
Genistein Soy foods Paclitaxel Enhancement in systemic exposure of drug Male Sprague-Dawley rats [20,89]
Chemotherapy Research and Practice 7
cytotoxic activity of several drugs and reduce inflammatory
responses [2,42].
Saponins from Chinese ginseng enhance the therapeutic
eect and behave as adaptogens reducing haematopoietic
complications induced by systemic chemotherapy [60,78].
Laboratory experiments have shown that saponins are able to
protect against cyclophosphamide- (CY-) induced genotoxi-
city and apoptosis in bone marrow cells and peripheral lym-
phocytes [78], some ginsenosides are able to sensitise drug-
resistant breast cancer cells to paclitaxel [63], and American
ginseng can attenuate nausea and vomiting induced by cis-
platin while enhancing its antiproliferative eect on human
breast cancer cells [63]. Notoginseng extract can synergisti-
cally increase the antiproliferative eect of 5-fluorouracil (5-
FU) in human colorectal cancer cell line making it possible
to reduce the dose of 5-FU in combination with notoginseng
and thereby further decrease its dose-related toxicity [63].
Polyphenol quercetin is able to potentiate the cytotoxic
eect of cisplatin while protecting normal renal cells from
cisplatin toxicity. Quercetin can work synergistically with
busulfan against human leukaemia cells and with doxoru-
bicin in cultured multidrug-resistant human breast cancer
cells. It increases cytotoxic eect of CY and decreases resist-
ance to gemcitabine, topotecan, vincristine, tamoxifen, pacli-
taxel, and doxorubicin [9,20,68,71,90]. Quercetin stim-
ulates also the activity of macrophages thereby further
improving the chemotherapeutical eciency [2].
Catechins from green tea can increase the therapeutic
eect of doxorubicin in drug-resistant tumours in animal
studies [68]. Administration of green tea increases the con-
centration of this chemotherapeutic agent in tumour but not
in normal tissue enhancing its antitumour activity [9].
Curcumin can also enhance the tumoricidal ecacy of
cytotoxic chemotherapy [66] and behave as adaptogen [60].
However, administration of soy isoflavones has led to
contradictory results emphasising the necessity to maintain
caution in excessive consumption of dietary agents and sup-
plements during treatment with chemotherapy. While genis-
tein can potentiate the action of tamoxifen to inhibit the
tumour cell growth in oestrogen receptor-negative human
breast cancer cells, this isoflavone can attenuate the tumorici-
dal activity of tamoxifen in oestrogen-dependent breast car-
cinoma cells (see Tabl e 1). Also, as a combination of daidzein
with tamoxifen produces increased protection against mam-
mary carcinogenesis [9,75], supplementation of genistein
can negate the inhibitory eect of tamoxifen on tumour
burden [75]. The therapeutic eect of tamoxifen can be fully
blocked also by flavonoid tangeretin [9,68] and until the
interactions between flavonoids and tamoxifen will be more
clear, therapeutic doses of flavonoid compounds should be
avoided in nutritional supporting programs of breast cancer
patients treated with tamoxifen [9].
Several naturally occurring plant agents like flavonoids
can enhance the drug bioavailability by inhibiting ATP
transporters-mediated drug eux in vitro, suggesting that
such interactions could occur also in vivo [20]. It is rec-
ommended therefore that diet of cancer patients treated
with chemotherapy should be rich in herbal constituents
(including quercetin, kaempferol, naringenin, silymarin, cat-
echins), fruits and berries (e.g., grapefruit, orange, apricot,
strawberry), and spices (mint, rosemary, curcumin, garlic,
ginseng, piper nigrum, onion) [20].
To ensure the removal of toxic waste products, it is
important that phase II detoxification enzymes are stimu-
lated and restored. For this purpose, cancer patients should
increase the intake of isothiocyanates (found in various
cruciferous vegetables, particularly Brussels sprouts and red
cabbage), naringin (in grapefruit), and parsley and spice
their food with curcumin [2]. Detoxification program should
be undertaken to eliminate the potentially mutagenic chemo-
therapeutic agents from organism. Various dietary com-
pounds contribute to induce specific detoxification pathways
whereas such multifunctional inducers include many of the
flavonoid molecules found in various fruits and vegetables
[22]. Increase in activity of phase II enzymes supports the
better detoxification and it can be achieved by substances
abundantly found in red grapes, garlic oil, rosemary, soy,
cabbage, Brussels sprouts, and broccoli [2,22].
7. Role of Vitamins in Chemotherapy
It is becoming more and more clear that vitamins can reduce
the side eects of chemotherapy treatment. Thus, vitamin
Areducesadverseeects of cyclophosphamide in rats [48]
and its coadministration with methotrexate ameliorates
intestinal damage in studies of mice without any inhibition
of antitumour activity [9,68]. Also folic acid (or vitamin
B9) supplementation during chemotherapy may lead to
reduction of toxicity [2]; however, it should not be added
to methotrexate treatment, as this anticancer drug is a folic
acid inhibitor [9]. Oncologists should therefore advise their
patients who undergo chemotherapy with antifolate drugs
(such as methotrexate) not to take folate supplements or
consume folate-rich food in excess. A folate status of the
patient may influence the response to methotrexate and can
possibly support cancer growth [91].
Vitamin C reduces the adverse eects of some chem-
otherapeutic agents on normal cells, such as those from dox-
orubicin. This vitamin can lower tamoxifen-induced hyper-
lipidemia in women with breast cancer and has been shown
to reduce bleomycin-induced chromosomal breakage in
human lymphoid cells in vitro [48]. In experiments carried
out with mice and guinea pigs, vitamin C led to a reduction
in cardiotoxicity of doxorubicin without any reduce in anti-
tumour ecacy [9].
Animal studies carried out mainly with rats and rabbits
have shown the ability of vitamin E to reduce bleomycin-
induced lung fibrosis, doxorubicin-induced cardiac toxicity,
and doxorubicin-induced skin necrosis. It lowers also doxo-
rubicin-induced toxicity in liver, kidney, and intestinal
mucosa. Coadministration of vitamin E and vitamin C can
reduce the tamoxifen-induced hyperlipidemia in women
with breast cancer [48]. The most eective form of vitamin E
is α-tocopheryl succinate that can be used as an adjunct to
standard cancer therapies in order to improve their ecacy
8Chemotherapy Research and Practice
on tumour cells while protecting normal cells against some
of their toxicities [92,93].
Vitamin K3 can act synergistically when combined with
several conventional chemotherapeutic agents [94]. How-
ever, it is important to keep in mind that while this vitamin is
able to induce cell cycle arrest at the G2/M phase, it is useful
as an enhancer of G2 phase-dependent chemotherapeutic
drug etoposide, whereas reduces the cytotoxic activity of S-
phase-dependent agent irinotecan [95].
Last but not least, vitamin-like compound coenzyme
Q10 has also been shown to ameliorate the side eects
of chemotherapies [96]. It helped to prevent doxorubicin-
associated cardiotoxicity in a small human study whereas the
diarrhoea and stomatitis were also significantly reduced. This
compound can lower also daunorubicin-induced adverse
events in leukaemia patients without any reduce in the
therapeutic benefit [9,68].
These data demonstrate the potential of dietary agents to
reduce the symptoms of adverse side eects of chemotherapy
treatment and relieve the situation of patients, improving
their quality of life. Therefore, nutritional support pro-
gramme built upon the personal necessities and clinical pur-
poses is an absolutely important part of treatment scheme for
all cancer patients.
8. Potential Problems
Prescribing Phytochemical
Agents to Chemotherapy Patients
Despite a huge amount of studies published about the ben-
efits of nutrient supplementation in chemotherapy there are
still a lot of sceptics and opponents among both physicians as
well as patients. On the one hand, this can be due to the lack
of knowledge; on the other hand, spread of misconceptions
and false beliefs also play an important role.
It is estimate that roughly 50% of cancer patients use
some kind of dietary supplements [50,97]. Many standard
chemotherapeutic agents mediate their cytotoxic eects by
generating excessive amounts of free radicals [5,6,48,97]
and a long-standing concern with the use of phytochemical
antioxidants has been comprised of their potential theoret-
ical intervention to the eectiveness of chemotherapy [5,6,
9,68]. It might be expected that antioxidants, quenching
the reactive oxygen species, can protect tumour cells as
well as healthy cells from oxidative damage generated by
chemotherapeutic drugs [5,9799]. This could lead to
reduction in eectiveness of cytotoxic therapy and many
oncologists are of the opinion that taking antioxidants
concurrently with chemotherapeutic drugs might be harmful
[5,48,59,97,99,100]. However, studies involving thousands
of patients have demonstrated that antioxidants and other
herbal nutrients do not interfere with chemotherapy, but
instead provide a wide range of beneficial eects [5,101].
Antioxidants can protect normal tissues from chemotherapy-
induced damage without decreasing oncological ecacy [5,
9,98]. These nutrients reduce the treatment-related side
eects and lower the risk and severity of comorbidities
[5,6,9,59,98,101,102] and improve the quality of life
and overall survival of cancer patients [9,59,101]. Some of
antioxidants may enhance the eects of cytotoxic regimens
improving the response rate of tumour to chemotherapeutic
agents [9,59,98,101,102].
Currently, a number of scientists hold the position that
antioxidants do not interfere with chemotherapy and at
commonly used dosages they enhance the success of the
treatment. Except for some specific interactions (including
flavonoids with tamoxifen) [5,9], the weight of the literature
supports the use of antioxidants during the treatment and
also in maintenance phase [59]. It is better to consume com-
binations of food-based antioxidants rather than synthetic
single compounds (vitamins) [59], whereas the form of a
particular antioxidant as well as dosages and dose schedules
are also important [103]. Physicians need to remain aware
of the large body of evidence showing beneficial eects of
antioxidants on the outcomes of chemotherapy treatment [9]
and apply this knowledge in their everyday clinical practice.
9. Conclusions and Further Perspectives
Developing the rational strategies for fighting the cancer
requires first a clear understanding of the causes and path-
ogenesis of the disease [104]. This comprises a multidimen-
sional approach of the studies at genomic, proteomic, and
metabolomic level considering both the individual vari-
ances as well as interindividual dierences. Harnessing the
mutations that cancer cells need to promote their patho-
logical survival and expansion will be the basis of further
therapeutic strategies [19,44]. In the future, each patient
should have his own unique chemotherapy protocol, which
improves the therapeutic quality by selecting and prescribing
well-matched drugs and avoiding ineective ones [21].
Applying such individualised chemotherapeutics through a
personalised chronotherapy regime will further improve the
final outcome [2,36]. This will be accompanied by the iden-
tification and testing of novel more specific and selective
drugs either via synthetic routes or by purifying from herbal
sources [39].
Although the novel chemotherapeutic agents will be
more and more eective against the tumour cells, their tox-
icity to normal tissues as well as drug resistance remains the
major obstacles for clinical use. Personalised approach using
various phytochemical compounds provides a new dimen-
sion to the standard cancer therapy for improving its out-
come in a complex and complementary way.
References
[1] F. S. Liu, “Mechanisms of chemotherapeutic drug resistance
in cancer therapy—a quick review,Taiwanese Journal of
Obstetrics and Gynecology, vol. 48, no. 3, pp. 239–244, 2009.
[2] H. Osiecki, Cancer: A Nutritional, Biochemical Approach, Bio-
concepts Publishing, 2002.
[3] R. E. Maccormick, “Possible acceleration of aging by adjuvant
chemotherapy: a cause of early onset frailty?” Medical
Hypotheses, vol. 67, no. 2, pp. 212–215, 2006.
[4] J. R. Bertino, “Cancer research: from folate antagonism to
molecular targets,Best Practice & Research: Clinical Haema-
tology, vol. 22, no. 4, pp. 577–582, 2009.
Chemotherapy Research and Practice 9
[5] R. M. Howes, “Dangers of antioxidants in cancer patients: a
review,” Philica, Article ID 153, 2009.
[6] H. Ma, T. Das, S. Pereira et al., “Ecacy of dietary antiox-
idants combined with a chemotherapeutic agent on human
colon cancer progression in a fluorescent orthotopic mouse
model,Anticancer Research, vol. 29, no. 7, pp. 2421–2426,
2009.
[7] J. Sagar, B. Chaib, K. Sales, M. Winslet, and A. Seifalian,
“Role of stem cells in cancer therapy and cancer stem cells:
a review,Cancer Cell International, vol. 7, pp. 9–19, 2007.
[8] X. Z. Wu, “A new classification system of anticancer drugs—
based on cell biological mechanisms,Medical Hypotheses,
vol. 66, no. 5, pp. 883–887, 2006.
[9] D. W. Lamson and M. S. Brignall, “Antioxidants in cancer
therapy; their actions and interactions with oncologic thera-
pies,Alternative Medicine Review, vol. 4, no. 5, pp. 304–329,
1999.
[10] G. E. Parris, “Clinically significant cancer evolves from tran-
sient mutated and/or aneuploid neoplasia by cell fusion to
form unstable syncytia that give rise to ecologically viable
parasite species,Medical Hypotheses, vol. 65, no. 5, pp. 846–
850, 2005.
[11] W. Harless and Y. Qiu, “Cancer: a medical emergency,
Medical Hypotheses, vol. 67, no. 5, pp. 1054–1059, 2006.
[12] T. Dittmar, C. Nagler, S. Schwitalla, G. Reith, B. Niggemann,
andK.S.Z
¨
anker, “Recurrence cancer stem cells—made by
cell fusion?” Medical Hypotheses, vol. 73, no. 4, pp. 542–547,
2009.
[13] R. Cairns, I. Papandreou, and N. Denko, “Overcoming physi-
ologic barriers to cancer treatment by molecularly targeting
the tumor microenvironment,Molecular Cancer Research,
vol. 4, no. 2, pp. 61–70, 2006.
[14] J. E. Trosko and R. J. Ruch, “Gap junctions as targets for
cancer chemoprevention and chemotherapy,Current Drug
Ta r g e t s, vol. 3, no. 6, pp. 465–482, 2002.
[15] G. Myhr, “MR guided cancer treatment system for an ele-
vated therapeutic index—a macroscopic approach,Medical
Hypotheses, vol. 70, no. 3, pp. 665–670, 2008.
[16] K. Ghosh, C. K. Thodeti, A. C. Dudley, A. Mammoto, M.
Klagsbrun, and D. E. Ingber, “Tumor-derived endothelial
cells exhibit aberrant Rho-mediated mechanosensing and
abnormal angiogenesis in vitro,” Proceedings of the National
Academy of Sciences of the United States of America, vol. 105,
no. 32, pp. 11305–11310, 2008.
[17] P. Fishman, S. Bar-Yehuda, M. Synowitz et al., “Adenosine
receptors and cancer,Handbook of Experimental Pharmacol-
ogy, vol. 193, pp. 399–441, 2009.
[18] E. W. LaPensee, T. R. Tuttle, S. R. Fox, and N. Ben-Jonathan,
“Bisphenol A at low nanomolar doses confers chemoresis-
tance in estrogen receptor-α-positive and -negative breast
cancer cells,Environmental Health Perspectives, vol. 117, no.
2, pp. 175–180, 2009.
[19] S. W. Lowe, E. Cepero, and G. Evan, “Intrinsic tumour sup-
pression,Nature, vol. 432, no. 7015, pp. 307–315, 2004.
[20] T.Bansal,M.Jaggi,R.K.Khar,andS.Talegaonkar,“Emerg-
ing significance of flavonoids as P-glycoprotein inhibitors in
cancer chemotherapy,Journal of Pharmacy and Pharmaceu-
tical Sciences, vol. 12, no. 1, pp. 46–78, 2009.
[21] D. Y. Lu, X. L. Chen, and J. Ding, “Individualized cancer
chemotherapy integrating drug sensitivity tests, patholog-
ical profile analysis and computational coordination—an
eective strategy to improve clinical treatment,Medical
Hypotheses, vol. 66, no. 1, pp. 45–51, 2006.
[22] D. J. Liska, “The detoxification enzyme systems,Alternative
Medicine Review, vol. 3, no. 3, pp. 187–198, 1998.
[23] C.P.Wu,C.H.Hsieh,andY.S.Wu,“Theemergenceofdrug
transporter-mediated multidrug resistance to cancer chemo-
therapy,Molecular Pharmaceutics, vol. 8, pp. 1996–2011,
2011.
[24] T. Eichhorn and T. Eerth, “P-glycoprotein and its inhibition
in tumors by phytochemicals derived from Chinese herbs,
Journal of Ethnopharmacology, vol. 141, pp. 557–570, 2012.
[25] A. Berruti, M. P. Brizzi, D. Generali et al., “Presurgical sys-
temic treatment of nonmetastatic breast cancer: facts and
open questions,” Oncologist, vol. 13, no. 11, pp. 1137–1148,
2008.
[26] S. Bonnet, S. L. Archer, J. Allalunis-Turner et al., “A mito-
chondria-K+channel axis is suppressed in cancer and its nor-
malization promotes apoptosis and inhibits cancer growth,
Cancer Cell, vol. 11, no. 1, pp. 37–51, 2007.
[27] P. S. Steeg and D. Theodorescu, “Metastasis: a therapeutic tar-
get for cancer,Nature Clinical Practice Oncology, vol. 5, no.
4, pp. 206–219, 2008.
[28] W. Q. Ding, B. Liu, J. L. Vaught, R. D. Palmiter, and S. E. Lind,
“Clioquinol and docosahexaenoic acid act synergistically to
kill tumor cells,Molecular Cancer Therapeutics, vol. 5, no. 7,
pp. 1864–1872, 2006.
[29] A. Ahmad, S. Banerjee, Z. Wang, D. Kong, A. P. N. Majumdar,
and F. H. Sarkar, “Aging and inflammation: etiological cul-
prits of cancer,Current Aging Science, vol. 2, no. 3, pp. 174–
186, 2009.
[30] V. Mongrain and N. Cermakian, “Clock genes in health and
diseases,Journal of Applied Biomedicine, vol. 7, no. 1, pp. 15–
33, 2009.
[31] E. Haus, “Chronobiology in oncology,International Journal
of Radiation Oncology, Biology, Physics, vol. 73, no. 1, pp. 3–5,
2009.
[32] V. Y. Gorbacheva, R. V. Kondratov, R. Zhang et al., “Circadian
sensitivity to the chemotherapeutic agent cyclophosphamide
depends on the functional status of the CLOCK/BMAL1
transactivation complex,Proceedings of the National Acad-
emy of Sciences of the United States of America, vol. 102, no. 9,
pp. 3407–3412, 2005.
[33] J. Sajan, T. A. Cinu, A. J. Chacko, J. Litty, and T. Jaseeda,
“Chronotherapeutics and chronotherapeutic drug delivery
systems,TropicalJournalofPharmaceuticalResearch, vol. 8,
no. 5, pp. 467–475, 2009.
[34] T. C. Erren, “Could visible light contribute to the develop-
ment of leukaemia and other cancers in children?” Medical
Hypotheses, vol. 64, no. 4, pp. 864–871, 2005.
[35] S. Sahar and P. Sassone-Corsi, “Metabolism and cancer: the
circadian clock connection,Nature Reviews Cancer, vol. 9,
no. 12, pp. 886–896, 2009.
[36] F. L´
evi, E. Filipski, I. Iurisci, X. M. Li, and P. Innominato,
“Cross-talks between circadian timing system and cell divi-
sion cycle determine cancer biology and therapeutics,Cold
Spring Harbor Symposia on Quantitative Biology, vol. 72, pp.
465–475, 2007.
[37] M. P. Antoch, R. V. Kondratov, and J. S. Takahashi, “Circadian
clock genes as modulators of sensitivity to genotoxic stress,
Cell Cycle, vol. 4, no. 7, pp. 901–907, 2005.
[38] G. Gebauer, “On the way to specifically targeting minimal
residual disease?” Breast Cancer Research, vol. 10, no. 5, article
112, 2008.
[39] H.Zhou,P.Zou,Z.C.Chen,andY.You,“Anovelvicious
cycle cascade in tumor chemotherapy,Medical Hypotheses,
vol. 69, no. 6, pp. 1230–1233, 2007.
10 Chemotherapy Research and Practice
[40] T. A. Ahles and A. J. Saykin, “Candidate mechanisms for
chemotherapy-induced cognitive changes,Nature Reviews
Cancer, vol. 7, no. 3, pp. 192–201, 2007.
[41] R. Han, Y. M. Yang, J. Dietrich, A. Luebke, M. Mayer-
Pr¨
oschel, and M. Noble, “Systemic 5-fluorouracil treatment
causes a syndrome of delayed myelin destruction in the cen-
tral nervous system,Journal of Biology, vol. 7, no. 4, pp. 12–
33, 2008.
[42] G. L. Nicolson, “Lipid replacement/antioxidant therapy as an
adjunct supplement to reduce the adverse eects of cancer
therapy and restore mitochondrial function,Pathol ogy a nd
Oncology Research, vol. 11, no. 3, pp. 139–144, 2005.
[43] C. Constantinou, A. Papas, and A. I. Constantinou, “Vitamin
E and cancer: an insight into the anticancer activities of vit-
amin E isomers and analogs,International Journal of Cancer,
vol. 123, no. 4, pp. 739–752, 2008.
[44] R. W. Johnstone, A. A. Ruefli, and S. W. Lowe, “Apoptosis: a
link between cancer genetics and chemotherapy,Cell, vol.
108, no. 2, pp. 153–164, 2002.
[45] P. Kovacic, “Unifying mechanism for anticancer agents
involving electron transfer and oxidative stress: clinical
implications,Medical Hypotheses, vol. 69, no. 3, pp. 510–516,
2007.
[46] C. L. Loprinzi, D. L. Barton, A. Jatoi et al., “Symptom control
trials: a 20-year experience,Journal of Supportive Oncology,
vol. 5, no. 3, pp. 119–128, 2007.
[47] B. U. Philips, The Case for Cancer Nutritional Support,The
Cancer Nutrition Network of Texas, 1999.
[48] K.N.Prasad,W.C.Cole,B.Kumar,andK.C.Prasad,“Scien-
tific rationale for using high-dose multiple micronutrients as
an adjunct to standard and experimental cancer therapies,
Journal of the American College of Nutrition,vol.20,no.5,pp.
450S–463S, 2001.
[49] A. Montazeri, “Health-related quality of life in breast cancer
patients: a bibliographic review of the literature from 1974 to
2007,Journal of Experimental and Clinical Cancer Research,
vol. 27, no. 1, pp. 32–62, 2008.
[50] Y. Al-Tonbary, M. Al-Haggar, R. El-Ashry, S. El-Dakroory, H.
Azzam, and A. Fouda, “Vitamin E and N-acetylcysteine as
antioxidant adjuvant therapy in children with acute lym-
phoblastic leukemia,Advances in Hematology, vol. 2009,
Article ID 689639, 5 pages, 2009.
[51] J. J. Kim and I. F. Tannock, “Repopulation of cancer cells dur-
ing therapy: an important cause of treatment failure,” Nature
Reviews Cancer, vol. 5, no. 7, pp. 516–525, 2005.
[52] G. L. Semenza, “Defining the role of hypoxia-inducible factor
1 in cancer biology and therapeutics,Oncogene, vol. 29, no.
5, pp. 625–634, 2010.
[53] A. B. Csoka and M. Szyf, “Epigenetic side-eects of common
pharmaceuticals: a potential new field in medicine and
pharmacology,Medical Hypotheses, vol. 73, no. 5, pp. 770–
780, 2009.
[54]J.Dietrich,R.Han,Y.Yang,M.Mayer-Pr
¨
oschel, and M.
Noble, “CNS progenitor cells and oligodendrocytes are tar-
gets of chemotherapeutic agents in vitro and in vivo,” Journal
of Biology, vol. 5, pp. 22–44, 2006.
[55] G. E. Jackson, “Chemo brain—a psychotropic drug phenom-
enon?” Medical Hypotheses, vol. 70, no. 3, pp. 572–577, 2008.
[56] L. Balducci and W. B. Ershler, “Cancer and ageing: a nexus at
several levels,Nature Reviews Cancer, vol. 5, no. 8, pp. 655–
662, 2005.
[57] G. Feres, J. I. Salluh, C. Ferreira, and M. Soares, “Severe acute
tumor lysis syndrome in patients with germ-cell tumors,
Indian Journal of Urology, vol. 24, no. 4, pp. 555–557, 2008.
[58] J. Hochberg and M. S. Cairo, “Tumor lysis syndrome: current
perspective,Haematologica, vol. 93, no. 1, pp. 9–13, 2008.
[59] B. Brom, “The antioxidant controversy and cancer,South
African Family Practice, vol. 51, no. 2, p. 119, 2009.
[60] S. M. Sagar, D. Yance, and R. K. Wong, “Natural health prod-
ucts that inhibit angiogenesis: a potential source for investi-
gational new agents to treat cancer—part 2,Current Oncol-
ogy, vol. 13, no. 3, pp. 99–107, 2006.
[61] C. D. Davis, “Nutritional interactions: credentialing of mol-
ecular targets for cancer prevention,Experimental Biology
and Medicine, vol. 232, no. 2, pp. 176–183, 2007.
[62] B. Jung and N. Ahmad, “Melatonin in cancer management:
progress and promise,Cancer Research, vol. 66, no. 20, pp.
9789–9793, 2006.
[63] C. Z. Wang and C. S. Yuan, “Potential role of Ginseng in the
treatment of colorectal cancer,American Journal of Chinese
Medicine, vol. 36, no. 6, pp. 1019–1028, 2008.
[64] H. H. Aung, S. R. Mehendale, C. Z. Wang, J. T. Xie, E.
McEntee, and C. S. Yuan, “Cisplatin’s tumoricidal eect on
human breast carcinoma MCF-7 cells was not attenuated by
American ginseng,” Cancer Chemotherapy and Pharmacology,
vol. 59, no. 3, pp. 369–374, 2007.
[65] C. Z. Wang, X. Luo, B. Zhang et al., “Notoginseng enhances
anti-cancer eect of 5-fluorouracil on human colorectal can-
cer cells,” Cancer Chemotherapy and Pharmacology, vol. 60,
no. 1, pp. 69–79, 2007.
[66] S. M. Sagar, D. Yance, and R. K. Wong, “Natural health prod-
ucts that inhibit angiogenesis: a potential source for inves-
tigational new agents to treat cancer—part 1,Current
Oncology, vol. 13, no. 1, pp. 14–26, 2006.
[67] S. Sen, H. Sharma, and N. Singh, “Curcumin enhances Vino-
relbine mediated apoptosis in NSCLC cells by the mitochon-
drial pathway,Biochemical and Biophysical Research Com-
munications, vol. 331, no. 4, pp. 1245–1252, 2005.
[68] D. W. Lamson and M. S. Brignall, “Antioxidants and cancer
therapy II: quick reference guide,Alternative Medicine
Review, vol. 5, no. 2, pp. 152–163, 2000.
[69] Y. Sadzuka, T. Sugiyama, and S. Hirota, “Modulation of can-
cer chemotherapy by green tea,Clinical Cancer Research, vol.
4, no. 1, pp. 153–156, 1998.
[70] T. Sugiyama and Y. Sadzuka, “Enhancing eects of green tea
components on the antitumor activity of adriamycin against
M5076 ovarian sarcoma,Cancer Letters, vol. 133, no. 1, pp.
19–26, 1998.
[71] D. W. Lamson and M. S. Brignall, “Antioxidants and cancer
III: quercetin,Alternative Medicine Review,vol.5,no.3,pp.
196–208, 2000.
[72] G. Scambia, F. O. Ranelletti, P. B. Panici et al., “Quercetin
potentiates the eect of adriamycin in a multidrug-resistant
MCF-7 human breast-cancer cell line: P-glycoprotein as a
possible target,Cancer Chemotherapy and Pharmacology,
vol. 34, no. 6, pp. 459–464, 1994.
[73] R. Homan, L. Graham, and E. S. Newlands, “Enhanced anti-
proliferative action of busulphan by quercetin on the human
leukaemia cell line K562,British Journal of Cancer, vol. 59,
no. 3, pp. 347–348, 1989.
[74] Y.H.Ju,D.R.Doerge,K.F.Allred,C.D.Allred,andW.G.
Helferich, “Dietary genistein negates the inhibitory eect of
tamoxifen on growth of estrogen-dependent human breast
cancer (MCF-7) cells implanted in athymic mice,Cancer
Research, vol. 62, no. 9, pp. 2474–2477, 2002.
[75] A. I. Constantinou, B. E. P. White, D. Tonetti et al., “The soy
isoflavone daidzein improves the capacity of tamoxifen to
Chemotherapy Research and Practice 11
prevent mammary tumours,European Journal of Cancer,
vol. 41, no. 4, pp. 647–654, 2005.
[76] F. Shen, X. Xue, and G. Weber, “Tamoxifen and genistein
synergistically down-regulate signal transduction and pro-
liferation in estrogen receptor-negative human breast carci-
noma MDA-MB-435 cells,Anticance r Research, vol. 19, no.
3, pp. 1657–1662, 1999.
[77] M. E. Bracke, H. T. Depypere, T. Boterberg et al., “Influence
of tangeretin on tamoxifen’s therapeutic benefit in mammary
cancer,Journal of the National Cancer Institute, vol. 91, no.
4, pp. 354–359, 1999.
[78] H. Wang, D. Peng, and J. Xie, “Ginseng leaf-stem: bioactive
constituents and pharmacological functions,Chinese Medi-
cine, vol. 4, pp. 20–27, 2009.
[79] Q. H. Zhang, C. F. Wu, L. Duan, and J. Y. Yang, “Protective
eects of total saponins from stem and leaf of Panax ginseng
against cyclophosphamide-induced genotoxicity and apop-
tosis in mouse bone marrow cells and peripheral lymphocyte
cells,” Food and Chemical Toxicology, vol. 46, no. 1, pp. 293–
302, 2008.
[80] S. Mehendale, H. Aung, A. Wang et al., “American ginseng
berry extract and ginsenoside Re attenuate cisplatin-induced
kaolin intake in rats,Cancer Chemotherapy and Pharmacol-
ogy, vol. 56, no. 1, pp. 63–69, 2005.
[81] M. K. Kuhlmann, E. Horsch, G. Burkhardt, M. Wagner, and
H. K¨
ohler, “Reduction of cisplatin toxicity in cultured renal
tubular cells by the bioflavonoid quercetin,Archives of Toxi-
cology, vol. 72, no. 8, pp. 536–540, 1998.
[82] W. W. G. Jia, X. Bu, D. Philips et al., “Rh2, a compound
extracted from ginseng, hypersensitizes multidrug-resistant
tumor cells to chemotherapy,Canadian Journal of Physiology
and Pharmacology, vol. 82, no. 7, pp. 431–437, 2004.
[83] S. Kitagawa, T. Nabekura, and S. Kamiyama, “Inhibition of
P-glycoprotein function by tea catechins in KB-C2 cells,
Journal of Pharmacy and Pharmacology, vol. 56, no. 8, pp.
1001–1005, 2004.
[84] L. C. Lin, M. N. Wang, and T. H. Tsai, “Food-drug interaction
of (-)-epigallocatechin-3-gallate on the pharmacokinetics of
irinotecan and the metabolite SN-38,Chemico-Biological
Interactions, vol. 174, no. 3, pp. 177–182, 2008.
[85] T. Ikegawa, H. Ohtani, N. Koyabu et al., “Inhibition of P-
glycoprotein by flavonoid derivatives in adriamycin-resistant
human myelogenous leukemia (K562/ADM) cells,Cancer
Letters, vol. 177, no. 1, pp. 89–93, 2002.
[86] S. C. Shin, J. S. Choi, and X. Li, “Enhanced bioavailability
of tamoxifen after oral administration of tamoxifen with
quercetininrats,International Journal of Pharmaceutics, vol.
313, no. 1-2, pp. 144–149, 2006.
[87]J.S.Choi,B.W.Jo,andY.C.Kim,“Enhancedpaclitaxel
bioavailability after oral administration of paclitaxel or pro-
drug to rats pretreated with quercetin,European Journal of
Pharmaceutics and Biopharmaceutics, vol. 57, no. 2, pp. 313–
318, 2004.
[88] G. Sliutz, J. Karlseder, C. Tempfer, L. Orel, G. Holzer, and M.
M. Simon, “Drug resistance against gemcitabine and topote-
can mediated by constitutive hsp70 overexpression in vitro:
implication of quercetin as sensitiser in chemotherapy,Bri-
tish Journal of Cancer, vol. 74, no. 2, pp. 172–177, 1996.
[89] X. Li and J. S. Choi, “Eect of genistein on the pharmacoki-
netics of paclitaxel administered orally or intravenously in
rats,International Journal of Pharmaceutics, vol. 337, no. 1-2,
pp. 188–193, 2007.
[90] W. W¨
atjen, G. Michels, B. Stean et al., “Low concentrations
of flavonoids are protective in rat H4IIE cells whereas high
concentrations cause DNA damage and apoptosis,Journal
of Nutrition, vol. 135, no. 3, pp. 525–531, 2005.
[91] A. D. Smith, Y. I. Kim, and H. Refsum, “Is folic acid good for
everyone?” American Journal of Clinical Nutrition, vol. 87, no.
3, pp. 517–533, 2008.
[92] J. Neuzil, M. Tomasetti, Y. Zhao et al., “Vitamin E analogs, a
novel group of “mitocans,” as anticancer agents: the impor-
tance of being redox-silent,Molecular Pharmacology, vol. 71,
no. 5, pp. 1185–1199, 2007.
[93] K.N.Prasad,B.Kumar,X.D.Yan,A.J.Hanson,andW.C.
Cole, “α-tocopheryl succinate, the most eective form of vit-
amin E for adjuvant cancer treatment: a review,Journal of
the American College of Nutrition, vol. 22, no. 2, pp. 108–117,
2003.
[94] D. W. Lamson and S. M. Plaza, “The anticancer eects of vit-
amin K,Alternative Medicine Review, vol. 8, no. 3, pp. 303–
318, 2003.
[95] S. Matzno, Y. Yamaguchi, T. Akiyoshi, T. Nakabayashi, and
K. Matsuyama, “An attempt to evaluate the eectofvitamin
K3 using as an enhancer of anticancer agents, Biological and
Pharmaceutical Bulletin, vol. 31, no. 6, pp. 1270–1273, 2008.
[96] L. Pinto, A. Sloan, I. Persaud, and N. R. Narain, “Normaliza-
tion of BCL-2 family members in breast cancer by coenzyme
Q10,Ethnicity & Disease, vol. 19, pp. S3-17–S3-18, 2009.
[97] G. M. D’Andrea, “Use of antioxidants during chemotherapy
and radiotherapy should be avoided,CA: A Cancer Journal
for Clinicians, vol. 55, no. 5, pp. 319–321, 2005.
[98]B.D.Lawenda,K.M.Kelly,E.J.Ladas,S.M.Sagar,A.
Vickers, and J. B. Blumberg, “Should supplemental antiox-
idant administration be avoided during chemotherapy and
radiation therapy?” Journal of the National Cancer Institute,
vol. 100, no. 11, pp. 773–783, 2008.
[99] R. I. Salganik, “The benefits and hazards of antioxidants: con-
trolling apoptosis and other protective mechanisms in cancer
patients and the human population,JournaloftheAmerican
College of Nutrition, vol. 20, no. 5, pp. 464S–472S, 2001.
[100] J. R. DiPalma and R. McMichael, “The interaction of vita-
mins with cancer chemotherapy,CA:ACancerJournalfor
Clinicians, vol. 29, no. 5, pp. 280–286, 1979.
[101] C. B. Simone and C. B. Simone II, “Re: should supplemental
antioxidant administration be avoided during chemotherapy
and radiation therapy?” Journal of the National Cancer
Institute, vol. 100, no. 21, pp. 1558–1559, 2008.
[102] G. Manda, M. T. Nechifor, and T. M. Neagu, “Reactive oxygen
species, cancer and anti-cancer therapies, Current Chemical
Biology, vol. 3, no. 1, pp. 22–46, 2009.
[103] K. N. Prasad and W. C. Cole, “Antioxidants in cancer ther-
apy,Journal of Clinical Oncology, vol. 24, pp. e8–e9, 2006.
[104] L. G. Hersoug and J. Arnau, “A built-in co-carcinogenic eect
due to viruses involved in latent or persistent infections,
Medical Hypotheses, vol. 68, no. 5, pp. 1001–1008, 2007.
... Studies have shown that regular intake of phytochemicals such as flavonoids is inversely proportional to breast cancer risk in humans [9][10][11]. Furthermore, many studies suggest that anti-oxidant activity of dietary flavonoids plays a role in alleviating adverse side effects of chemotherapy, as well as in cancer chemo-prevention [12][13][14]. Flavonoid-induced cytotoxic and chemopreventive activities are linked to the ability of flavonoids to impact biological systems such as inhibition of growth-promoting protein kinases, cell cycle progression, metastasis, angiogenesis, multi-drug resistance, metabolism of carcinogens, and pro-oxidant enzymes [15]. Flavonoids possess a wide range of anti-cancer properties. ...
... Studies have shown that regular intake of phytochemicals such as flavonoids is inversely proportional to breast cancer risk in humans [9][10][11]. Furthermore, many studies suggest that anti-oxidant activity of dietary flavonoids plays a role in alleviating adverse side effects of chemotherapy, as well as in cancer chemo-prevention [12][13][14]. Flavonoid-induced cytotoxic and chemo-preventive activities are linked to the ability of flavonoids to impact biological systems such as inhibition of growth-promoting protein kinases, cell cycle progression, metastasis, angiogenesis, multi-drug resistance, metabolism of carcinogens, and pro-oxidant enzymes [15]. Flavonoids possess a wide range of anti-cancer properties. ...
Article
Full-text available
The eradication of cancer stem cells (CSCs) is vital to successful cancer treatment and overall disease-free survival. CSCs are a sub-population of cells within a tumor that are defined by their capacity for continuous self-renewal and recapitulation of new tumors, demonstrated in vitro through spheroid formation. Flavonoids are a group of phytochemicals with potent anti-oxidant and anti-cancer properties. This paper explores the impact of the flavonoid precursor phloridzin (PZ) linked to the ω-3 fatty acid docosahexaenoate (DHA) on the growth of MCF-7 and paclitaxel-resistant MDA-MB-231-TXL breast cancer cell lines. Spheroid formation assays, acid phosphatase assays, and Western blotting were performed using MCF-7 cells, and the cell viability assays, Annexin-V-488/propidium iodide (PI) staining, and 7-aminoactinomycin D (7-AAD) assays were performed using MDA-MB-231-TXL cells. PZ-DHA significantly reduced spheroid formation, as well as the metabolic activity of MCF-7 breast cancer cells in vitro. Treatment with PZ-DHA also suppressed the metabolic activity of MDA-MB-231-TXL cells and led to apoptosis. PZ-DHA did not have an observable effect on the expression of the drug efflux transporters ATP-binding cassette super-family G member 2 (ABCG2) and multidrug resistance-associated protein 1 (MRP1). PZ-DHA is a potential treatment avenue for chemo-resistant breast cancer and a possible novel CSC therapy. Future pre-clinical studies should explore PZ-DHA as a chemo-preventative agent.
... However, it can result in many problems, such as aching in the bones and muscles, dyspnea, chest pain, nausea, hypoxemia, anaphylaxis, syncope, and flushing (1,9). Andalso, the G-CSF injection is conditional only for patientswith a white blood cell (WBC) count of <1000/μL or anabsolute neutrophil count (ANC) of <500/μL (8). Therefore, using a subcutaneous injection of G-CSF to prevent ortreat CIL cannot benefit alloncologic patients (18). ...
... Various plant-derived compounds have shown to improve the efficiency of cytotoxic agents, decrease their resistance, lower and alleviate toxic side effects, reduce the risk of tumour lysis syndrome, and detoxify the body of chemotherapeutics. The personalised approach using various phytochemicals provides thus a new dimension to the standard cancer therapy for improving its outcome in a complex and complementary way (8). ...
Article
Full-text available
Objective: Leukopenia is the most commonly experienced side effect of cancer treatments especially chemotherapy. This systematic review assesses whether there are any dietary factors which can have plausible effect on reducing the CIL (Chemotherapy Induced Leukopenia). Methods: The search for immune enhancing dietary factors was carried out using the following databases; Google Scholar, Excerpta Medica Database (EMBASE), Public/Publisher MEDLINE (PubMed), Research gate and ELSEVIER. Also the ancient Indian literature particularly Ayurvedic scriptures such as Chikitsa Prabhakar, Ayurved for Cancer, Charak Samhita, Sushrut Samhita, Bhavaprakshetc and modern nutritional sciences’ books were also studied comprehensively for the collection of references to augment immunity via diet. The related researches were meticulously studied with the aim to discover the dietary factors which will supplement immunity to reduce or lessen the risk of Chemotherapy Induced Leukemia (CIL). Results: The review evidently explains the benefits of certain research assessed indigenous food components such as ginger, Yashti-madhu (licorice), betel leaf, Triphala, Neem, Aamla, Giloy, Punarnava and also certain phytochemicals like alkaloids, polysaccharides, phenolic compounds, glycosides, anthocyanins, saponins, sterols etc. towards credibly increasing WBC count by immuno-modulation and immuno-stimulation. Conclusion: Dietary phytochemicals from certain indigenous herbs and foods have prominently exhibited anti-metastatic properties as well as immune-modulating activities. Hence these dietary phytochemicals from indigenous herbs and foods have been studied in depth to aid in reducing the risk of above mentioned side effect. Most of them can be safely used in the prevention of CIL once chemotherapy is commenced.
... Plant-based diets, focusing on the consumption of not only vegetables and fruits, but also whole grains, are inversely associated with the incidence of malignancies, as they are rich in phytochemicals [10]. Over the past decades, the application of phytochemicals as adjuvant anti-neoplastic agents has been a rising trend [11], because NPs display the following main characteristics: (i) "metabolite-likeness", (ii) active transport metabolites, and (iii) high bioavailability [12][13][14]. Hence, studies on NPs skyrocketed in the past three decades [15]. ...
Article
Full-text available
The majority of blood malignancies is incurable and has unforeseeable remitting-relapsing paths in response to different treatments. Cynaropicrin, a natural sesquiterpene lactone from the edible parts of the artichoke plant, has gained increased attention as a chemotherapeutic agent. In this study, we investigated the effects of cynaropicrin against multiple myeloma (MM) cells in vitro and assessed its in vivo effectiveness in a xenograft tumor zebrafish model. We showed that cynaropicrin exerted potent cytotoxicity against a panel of nine MM cell lines and two leukemia cell lines with AMO1 being the most sensitive cell line (IC 50 = 1.8 ± 0.3 µM). Cynaropicrin (0.8, 1.9, 3.6 µM) dose-dependently reduced c-Myc expression and transcriptional activity in AMO1 cells that was associated with significant downregulation of STAT3, AKT, and ERK1/2. Cell cycle analysis showed that cynaropicrin treatment arrested AMO1 cells in the G 2 M phase along with an increase in the sub-G 0 G 1 phase after 24 h. With prolonged treatment times, cells accumulated more in the sub-G 0 G 1 phase, implying cell death. Using confocal microscopy, we revealed that cynaropicrin disrupted the microtubule network in U2OS cells stably expressing α-tubulin-GFP. Furthermore, we revealed that cynaropicrin promoted DNA damage in AMO1 cells leading to PAR polymer production by PARP1 hyperactivation, resulting in AIF translocation from the mitochondria to the nucleus and subsequently to a novel form of cell death, parthanatos. Finally, we demonstrated that cynaropicrin (5, 10 µM) significantly reduced tumor growth in a T-cell acute lymphoblastic leukemia (T-ALL) xenograft zebrafish model. Taken together, these results demonstrate that cynaropicrin causes potent inhibition of hematopoietic tumor cells in vitro and in vivo.
... While the synthetic anticancer drugs currently in use destroy cancer cells, they also kill normal cells in large amounts. In addition, if resistance develops against antineoplastic drugs, they will not affect cancer cells [10,11]. Therefore, the idea that natural anticancer drugs should be developed has emerged. ...
Article
Full-text available
Hypericum salsolifolium plant extracts prepared with different solvents. The total phenolic content, total flavonoid content, and antioxidant potential [(2,2-diphenyl-1-picrylhydrazyl, 2,2′-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid), ferric reducing antioxidant power, and cupric reducing antioxidant capacity assays] of Hypericum salsolifolium extracts obtained using solvents of different polarities (hexane, dichloromethane, methanol, and water) were measured using spectrophotometric methods. The contents of the extracts were analyzed using liquid chromatography–mass spectrometry and inductively coupled plasma—mass spectrometry methods. Anticancer detection was performed in human lung carcinoma cells using the 3-4,5-dimethylthiazol-2,5-diphenyltetrazolium bromide, annexin-V, and cell cycle assays, as well as fluorescence detection of acridine orange/ethidium bromide staining. The methanolic extract was determined to have higher activation values of total phenolics, total flavonoids, ferric reducing antioxidant power, and in the 2,2-diphenyl-1-picrylhydrazyl assay than the other extracts, and the aqueous extract had higher values in the 2,2′-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid) and cupric reducing antioxidant capacity assays. The methanolic extract showed a cytotoxic effect against human lung carcinoma cells (IC50: 141.96 µg/mL). It was found that Hypericum salsolifolium extract showed antioxidant and anticancer activities. It was concluded that this plant can be used as a nutritional supplement due to its glucose, phenolic compound, amino acid, and vitamin content.
... Apoptosis activation and cell cycle arrest are thought to be the main mechanisms behind propolis' anticancer effects [64]. The current study suggests that the occurrence of visible lesions following cisplatin therapy, such as inflammation, haemorrhagic RBCs and RBC extravasation in airway spaces, may be caused by the fact that cisplatin is not only harmful to cancer cells but also affects healthy cells [65]. ...
... Phytochemicals have emerged as prospective anticancer treatments, either alone or in combination with other chemotherapeutic drugs, due to their efficient tumour-targeting ability and low toxicity to normal tissues [10][11][12][13]. Phytochemicals act as chemopreventive and synergistic agents, increasing anticancer activity and decreasing chemotherapy-associated toxicity [14]. Pro-oxidative and antioxidative properties of phytochemicals positively regulate the homeostasis of reactive oxygen species (ROS), modifying apoptotic signals to prevent cancer [10,11]. ...
Article
Full-text available
Cancer is one of the most worldwide spread diseases and causes maximum death. Treatment of cancer depends on the host immune system and the type of drugs. The inefficiency of conventional cancer treatments as a result of drug resistance, nontargeted delivery, and chemotherapy-related negative side effects has caused bioactive phytochemicals to come into focus. As a result, recent years have seen an increase in research into screening and identifying natural compounds with anticancer properties. Recent studies on the isolation and use of polysaccharides derived from various marine algal species have revealed a variety of biological activities, including antioxidant and anticancer properties. Ulvan is a polysaccharide derived from various green seaweeds of the Ulva species in the family Ulvaceae. It has been demonstrated to have potent anticancer and anti-inflammatory properties through the modulation of antioxidants. It is vital to understand the mechanisms underlying the biotherapeutic activities of Ulvan in cancer and its role in immunomodulation. In this context, we reviewed the anticancer effects of ulvan based on its apoptotic effects and immunomodulatory activity. Additionally, we also focused on its pharmacokinetic studies in this review. Ulvan is the most conceivable candidate for use as a cancer therapeutic agent and could be used to boost immunity. Moreover, it may be established as an anticancer drug once its mechanisms of action are understood. Due to its high food and nutritive values, it can be used as a possible dietary supplement for cancer patients in the near future. This review may provide fresh perspectives on the potential novel role of ulvan, reveal a brand-new cancer-prevention strategy, and improve human health.
... Current drug treatments for cancer face many challenges that require careful consideration and improvement. One major issue is the toxicity of chemotherapeutic drugs, which can limit the effectiveness of treatment despite the various treatment options available [40,41]. Using nanotechnology to increase drug solubility may help overcome this challenge by opening new avenues for therapeutic development [42]. ...
Article
Full-text available
Non-thermal plasma (NTP) is thought to have a cytotoxic effect on tumor cells. Although its application in cancer therapy has shown considerable promise, the current understanding of its mechanism of action and cellular responses remains incomplete. Furthermore, the use of melatonin (MEL) as an adjuvant anticancer drug remains unexplored. In this study, we found that NTP assists MEL in promoting apoptosis, delaying cell cycle progression, and inhibiting cell invasion and migration in hepatocellular carcinoma (HCC) cells. This mechanism may be associated with the regulation of intracellular reactive oxygen species levels and ribonucleotide reductase regulatory subunit M2 expression. Our findings confirm the pharmacological role of MEL and the adjuvant value of NTP, emphasizing their potential in combination therapy for HCC. Our study may have important implications for the development of new approaches for HCC treatment.
Article
Full-text available
The present work aims to evaluate anticancer performance of Cassia alata methanolic leaf extracts (CMLE) in ethyl carbamate-stimulated lung adenocarcinoma (LAD) in differentiation to the function of Cisplatin (CIPL). Rats were divided into four groups: (1) control (CONT), (2) lung-adenocarcinoma (LAD) injected intra-peritoneally with 1g/kg ethyl carbamate once weekly for a month, (3) LAD+CMLE administered 500 mg/kg CMLE orally for the last two months of the experiment, and (4) LAD+CIPL treated group, injected 2.5 mg/kg Cisplatin intraperitoneally once weekly for the last two months of the experiment. Light and electron microscopic examinations revealed adenocarcinoma development in terminal bronchiole besides some histopathological changes in the LAD group such as atypical, exaggerated collagen fibers, increment of mucinous content, and increasing of PCNA positive immunoreactivity whereas electron microscopy investigation exposed that papillary adenocarcinoma originated from Clara cells in the LAD group. The LAD+CMLE treated group showed no tumor masses and nearly all with normal lung histology. It also recovered the normal ultrastructure of bronchiolar Clara cells. CMLE treatment offers a new alternative cure with less toxicity than Cisplatin for lung cancer therapy. Hence, CMLE would be employed as a novel supply of anti-cancer compounds combating lung cancer. Keywords: Cassia alata; Ethyl carbamate; lung adenocarcinoma; Cisplatin; clara cells; immunohistochemistry
Chapter
Considering the essentiality to find effectual biological agents as therapeutics, biosurfactants have shown remarkable significance in recent years. Biosurfactants are surface-active amphiphilic metabolites which are predominantly produced from biological sources, particularly from microorganisms. Glycolipids, lipopeptides, phospholipids, lipoproteins, polymeric biosurfactants, and particulate biosurfactants are the leading classes of biosurfactants. Disparate properties such as self-assembly, biodegradability, emulsification, low/no toxicity, and biocompatibility make biosurfactant a perfect biologically active substance with respect to the chemical surfactants. Over the past few years, biosurfactant is being used as capping, reducing, stabilizing, and coating agent in nanobiotechnology as eco-friendly alternative biomolecules to the physical and chemical substances with higher efficiency. Biosurfactants-mediated nanoparticles have exhibited multipurpose roles in biomedicine, particularly as antibacterial, antifungal, antibiofilm, anticancer, wound healing, anti-inflammatory, mosquitocidal, and dermal drug delivery agents, without showing toxicity to the normal cells. This chapter sheds light not only on the fundamental aspects (classification, structures, genetics, and properties) of biosurfactants but also on the recent updates emphasizing the foremost therapeutic attributes of biosurfactants-mediated nanoparticles as next-generation drug delivery system.KeywordsBiosurfactantsBiosynthesisDrug deliveryNanobiotechnologyNanoparticlesTherapeutics
Data
Full-text available
Elderly lung cancer patients and those with poor performance status/co-morbid conditions are deprived of chemotherapy because of high toxicity of multidrug regimens. Human squamous cell lung carcinoma H520 cells treated with Curcumin were sen-sitized to the cytotoxicity caused by chemotherapeutic agent, Vinorelbine. Both caused apoptosis by increasing the protein expres-sion of Bax and Bcl-xs while decreasing Bcl-2 and Bcl-X L , releasing apoptogenic cytochrome c, and augmenting the activity of caspase-9 and caspase-3. Expression of Cox-2, NF-jB, and AP-1 was also affected. 23.7% apoptosis was induced in the H520 cells by treatment with Curcumin while Vinorelbine caused 38% apoptosis. Pre-treatment with Curcumin enhanced the Vinorelbine induced apoptosis to 61.3%. The findings suggest that Curcumin has the potential to act as an adjuvant chemotherapeutic agent and enhance chemotherapeutic efficacy of Vinorelbine in H520 cells in vitro. Thus, Curcumin offers the prospect of being beneficial in the above-mentioned patient groups.
Article
Full-text available
Mammalian cells produce reactive oxygen species (ROS) which are carcinogens, key actors of the non-specific immune defense against pathogens and, in a more subtle way, of signal transduction, cellular metabolism and functions. Oxidative stress can induce severe damage to the host which in turn adapted to face oxidative injury. Disruption of redox balance leads to various pathological conditions, such as cancer. In this review we explore the network linking ROS, cancer cells, anti-tumor immunity and therapy. We emphasize recent findings regarding the oxidative tumor microenvironment and the correlation between ROS, proliferation and death of cancer cells. Further-on we highlight that granulocytes, as key inflammatory cells and ROS producers, are nowadays exploited for eradication of cancer cells. Finally, we focus on ROS-inducing anti-neoplastic therapies (radiotherapy and photodynamic therapy) and on controversial issues regarding the interference between chemotherapy, ROS and antioxidants. This review is directed mainly to researchers involved in anti-cancer drug development by pointing out that redox balance is a suitable therapeutic target, either alone or in combination with other pathways of cancer cells killing. We emphasize critical redox-controlled checkpoints that have to be taken into account in drug design for achieving good therapeutic efficiency and convenient side-effects.
Article
Cancer is second only to heart disease as the leading cause of death in the United States. Indeed, it is estimated that approximately 178,000 new breast cancer cases were diagnosed in 2007 and 40,000 women will succumb to the disease. The nature of the disease makes it very resistant to chemotherapeutic intervention and radiation. The balance of the Bcl-2 protein family has been implicated as the major contibuting factor to conferral of resistance to cancer therapy. Previous work from our research group has demonstrated that Coenzyme Q10 (Q10) is able to significantly decrease Bcl-2 and thereby induce apoptosis in melanoma and prostate cancer. Hence, we postulated that Q10 may have a pro-apoptotic effect in breast cancer. To investigate this hypothesis, we employed the Sk-Br3 and MCF-7 breast cancer lines which exhibit a mutation Her-2/neu and p53 respectively. We examined the effect of Coenzme Q10 on various members of the Bcl-2 family (bcl-2, bcl-xl, bid, bad, bak, mcl-1, bim, and bax), p53, and caspases 3, 6, 9. All cells were teated for 0-24 hours in the presence and absence of 50 μM and 100 μM Q10 under physiologic conditions after which total protein was isolated and subjected to Western blot analysis to measure the aforementioned protein products. The results of our study may provide a template for further investigation into the mechanism of action of mammary oncogenesis while providing support for the use of Coenzyme Q10 as an adjuvant breast cancer therapy. The result showed that there was an upregulation in protein expression of proapoptotic members and BH3 subfamily members such as bid, bad, bax, bim, and bak whereas the anti-apototic members bcl-xl, mcl-1, and bcl-2 significantly decreased in total protein expression between 4 and 12 hours. Commitment to apoptosis was confirmed by activation of caspase 3, 6 and 9. Conversely, administration of Coenzyme Q10 to mammary fibroblasts did not elicit a significant response on any of the aforementioned intracellular proteins involved in programmed cell death. The data herein suggest that Coenzyme Q10 is able to modulate the various subfamilies of the Bcl-2 family in a manner that restores the apoptotic potential in breast cancer without presenting any adverse effect to normal breast tissue. This provides a template for further investigation into the mechanism of action of mammary oncogenesis while providing support for the use of Coenzyme Q10 as an adjuvant in breast cancer therapy.
Article
Export Date: 18 October 2014
Article
Mutations that drive uncontrolled cell-cycle progression are requisite events in tumorigenesis. But evolution has installed in the proliferative programmes of mammalian cells a variety of innate tumour-suppressive mechanisms that trigger apoptosis or senescence, should proliferation become aberrant. These contingent processes rely on a series of sensors and transducers that act in a coordinated network to target the machinery responsible for apoptosis and cell-cycle arrest at different points. Although oncogenic mutations that disable such networks can have profound and varied effects on tumour evolution, they may leave intact latent tumour-suppressive potential that can be harnessed therapeutically.
Article
Many patients being treated for cancer use dietary supplements, particularly antioxidants, in the hope of reducing the toxicity of chemotherapy and radiotherapy. Some researchers have claimed, furthermore, that antioxidants also increase the effectiveness of cytotoxic therapy and have explicitly recommended their use. However, mechanistic considerations suggest that antioxidants might reduce the effects of conventional cytotoxic therapies. Preclinical data are currently inconclusive and a limited number of clinical studies have not found any benefit. Clinicians should advise their patients against the use of antioxidant dietary supplements during chemotherapy or radiotherapy. Such caution should be seen as the standard approach for any unproven agent that may be harmful.
Article
Quercetin (QC), a polyphenolic compound widely distributed in fruits and vegetables has recently gained interest due to its cisplatin (CP) sensitizing properties in cancer cells. It is currently unknown, whether quercetin also increases the susceptibility of the kidneys to cisplatin toxicity. We studied the effects of various bioflavonoids on CP toxicity in an in vitro model of cultured tubular epithelial cells (LLC-PK1). Viability of LLC-PK1 cells, as assessed by lactate dehydrogenase (LDH) release and MTT-test, was affected by CP (100–400 μM) in a time and dose dependent fashion. Pretreatment of cells with QC for 3 h significantly reduced the extent of cell damage. The protective activity of QC was concentration dependent, starting at 10–25 μM and reaching a plateau between 50 and 100 μM. Other bioflavonoids (catechin, silibinin, rutin) did not diminish cellular injury, even at higher concentrations (100–500 μM). Quercetin itself showed some intrinsic cytotoxicity at concentrations exceeding 75 μM. Our data indicate that quercetin reduces cisplatin toxicity in cultured tubular epithelial cells. The exact mechanism of protection is unclear, though scavenging of free oxygen radicals may play an important role.