Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

Review

Doxorubicin, mesenchymal stem cell toxicity and


antitumour activity: implications for clinical use
Mia Baxter-Hollanda and Crispin R. Dassa,b
a
School of Pharmacy and Biomedical Science, Curtin University, and bCurtin Health Innovation Research Institute, Perth, WA, Australia

Keywords Abstract
cancer; chemotherapy; doxorubicin; stem
cell; toxicity Objectives The use of doxorubicin, an antineoplastic medication used for the
treatment of cancers via mechanisms that prevent replication of cells or lead to
Correspondence their death, can result in damage to healthy cells as well as malignant. Among the
Crispin R. Dass, School of Pharmacy and
affected cells are mesenchymal stem cells (MSCs), which are involved in the
Biomedical Science, Curtin University, GPO
Box U1987, Perth, WA 6845, Australia.
maintenance and repair of tissues in the body. This review explores the mecha-
E-mail: Crispin.Dass@curtin.edu.au nisms of biological effects and damage attributed to doxorubicin on MSCs. The
PubMed database was used as a source of literature for this review.
Received July 19, 2017 Key findings Doxorubicin has the potential to lead to significant and irreversible
Accepted November 25, 2017 damage to the human bone marrow environment, including MSCs. The primary
known mechanism of these changes is through free radical damage and activation
doi: 10.1111/jphp.12869
of apoptotic pathways. The presence of MSCs in culture or in vivo appears to
either suppress or promote tumour growth. Interactions between doxorubicin
and MSCs have the potential to increase chemotherapy resistance.
Summary Doxorubicin-induced damage to MSCs is of concern clinically. How-
ever, MSCs also have been associated with resistance of tumour cells to drugs
including doxorubicin. Further studies, particularly in vivo, are needed to provide
consistent results of how the doxorubicin-induced changes to MSCs affect treat-
ment and patient health.

Introduction Class of drug, chemical structure and


mechanism of action
Doxorubicin is a medication employed in treatment regi-
mens of the ever-growing epidemic of cancer. Antineo- Doxorubicin (also known by trade names of Adriamycinâ,
plastic drugs used for chemotherapy are renowned for Doxilâ, Myocetâ) is classed as an anthracycline antibiotic anti-
their extensive side-effect profile, which ranges from neoplastic drug, derived from the Streptomyces peucetius var.
unpleasant to fatal, and doxorubicin is no exception.
Doxorubicin’s mechanism of action involves inhibition of
the synthesis of and damage to DNA, and formation of
reactive oxygen species (ROS) to create oxidative stress in
the cellular environment. These effects are not limited to
tumour cells; healthy cells are also affected, including
stem cells. As they are responsible for the growth and
repair of a wide range of tissues in the body, damage to
stem cells has the potential to cause lifelong health prob-
lems for a patient treated with doxorubicin. This review
discusses the potential ways doxorubicin can influence
cells, the current observed effects of chemotherapy on
stem cells, particularly mesenchymal stem cells (MSCs),
and what is already known about doxorubicin’s effects on
stem cells, but which has not been reviewed in this man-
ner to date.

320 © 2018 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 70 (2018), pp. 320–327
Mia Baxter-Holland and Crispin R. Dass Doxorubicin and stem cells

Caesius bacteria.[1] The anthracycline backbone is com-


Free radical damage to cells
prised of a tetracyclic aglycone ring with adjacent quinone-
hydroquinone groups and an attached daunosamine Doxorubicin is able to accept an electron at its quinone
sugar.[1,2] Doxorubicin is distinguished from other anthra- groups, forming a semiquinone in the presence of enzymes
cyclines by the addition of a methoxy group to the tetra- NADH dehydrogenase, xanthine oxidase and cytochrome
cyclic ring system, and methyl groups to the C-9 side chain P450 reductase. This semiquinone can damage DNA itself
and daunosamine sugar.[2] as well as return to its parent quinone by reducing oxygen,
Anthracyclines, including doxorubicin, exert their action forming ROS such as hydrogen peroxide and superox-
via multiple mechanisms. These biological effects do not ide.[3,9,10] Under normal conditions tissues can tolerate the
discriminate between cells of cancers and healthy cells and presence of ROS. However, when levels exceed the capacity
are discussed in turn below. of intrinsic antioxidant defences, cellular damage occurs.
This includes DNA strand scission and peroxidation of
unsaturated fatty acids leading to cellular membrane
Inhibition of DNA synthesis
damage.[3,10]
It has been shown that doxorubicin is able to intercalate Doxorubicin is given via intravenous injection or infu-
deoxyribonucleic acid (DNA) via the insertion of doxoru- sion and typically displays a triphasic plasma clearance and
bicin as a planar chromophore between the planar DNA a terminal half-life of 24–36 h.[11] Doxorubicin is used in
base pairs, binding to them via hydrogen bonds.[3] The multidrug regimens in the treatment of cancers including
drug’s hydroxyl and sugar amino groups are responsible for but not limited to solid tumours such as breast cancer,[1]
these covalent interactions.[4,5] Intercalation of DNA results ovarian cancer[12] and osteosarcoma[3] as well as
in double-stranded DNA breakage and gives rise to apopto- haematopoietic malignancies such as lymphomas and leu-
sis.[3,4] Doxorubicin has also been shown to lead to the kaemia.[13]
inhibition of DNA and RNA polymerase enzymes, though
it was found to have a higher preference for DNA poly-
Stem cells: an overview
merase.[6] Blockage of these enzymes prevents a cell from
undergoing normal DNA and RNA synthesis.[3,6] It must Stem cells can be subdivided into two main groups: adult/
be noted that outcomes of in-vivo studies on doxorubicin’s somatic and embryonic stem cells.[14] Embryonic stem cells
DNA and RNA polymerase inhibition are inconclusive – (ESCs) are derived from the inner cell mass of a human
some show that plasma concentrations above those used blastocyst, which forms 3 to 5 days after an egg is fertilised.
therapeutically (approximately 1–2 lM) are required for These cells are described as pluripotent because there is a
such effects, while others display enzyme inhibition at these possibility for them to develop into all three embryonic
levels. It has been proposed that enzyme inhibition is only a germ layers: endoderm (gut epithelium); ectoderm (neutral
cytostatic (rather than cytotoxic) and transient component epithelium, embryonic ganglia and stratified squamous
of drug action.[3] epithelium); and mesoderm (cartilage, bone, smooth
muscle and striated muscle).[15] ESCs are shown to express
high levels of telomerase, a ribonucleoprotein that is
Interference of topoisomerase II activity
involved in maintaining telomere length. The expression of
Topoisomerase II is an enzyme that regulates DNA topol- this protein is correlated with immortality in cell lines,
ogy (the pattern by which two complementary strands are allowing ESCs to have a long replicative lifespan and main-
intertwined) via creating temporary cuts in both strands tain the capacity to differentiate into the three germ layers
of DNA during replication. This relaxes the DNA and pre- in vitro.[15]
vents both negative and positive superhelical twists, knots Somatic stem cells are present in almost every tissue in
and nucleic acid tangles.[7,8] Doxorubicin takes advantage an adult body and have a narrower differentiation potential
of topoisomerase II by stabilising the enzyme-DNA inter- than ESC, only being able to differentiate into several or a
mediate complex that is responsible for the cleavage, single type of cell. They are used for tissue maintenance
resulting in higher concentrations of the complex within and repair.[14] The bone marrow is a large source of
the cell. Thus, more than the standard number of double- somatic stem cells, including both subcategories:
stranded DNA cuts are formed and rather than being tran- haematopoietic and mesenchymal.[14] MSCs are able to dif-
sient, much of the damage becomes permanent.[8] Cell ferentiate into osteoblasts (bone), adipocytes (fat), chon-
death then occurs as fragmented DNA cannot be tran- drocytes (cartilage) and myoblasts (muscle);[14]
scribed, and the cell is unable to undergo normal mitotic haematopoietic stem cells can form every type of blood cell,
processes. such as erythrocytes and platelets.[16]

© 2018 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 70 (2018), pp. 320–327 321
Doxorubicin and stem cells Mia Baxter-Holland and Crispin R. Dass

Mesenchymal stem cells Effects of chemotherapeutics on


stem cells
When isolated, MSCs are normally derived from bone mar-
row, however are also present in umbilical cord blood, sali- Medication-induced elevation of ROS is one method of tar-
vary glands, adipose tissue and other organs including the geting cancer cells via causing damage to proteins, DNA
gut.[17,18] The International Society for Cellular Therapy and lipids.[29] Stem cells have mechanisms for DNA repair
states that in order for a cell to be classed as MSC, it must and oxidative stress defence. Compared to somatic cells,
(1) possess certain surface markers such as CD105, CD70 ESCs have been shown to have higher antioxidant enzyme
and CD90 and lack expression of others including CD45, levels, including superoxide dismutase and glutathione/
CD34, CD14 or CD11b, (2) must be plastic-adherent when thioredoxin systems, and reduced accumulation of ROS,
maintained in standard culture conditions and (3) must resulting in a better defence mechanism against oxidative
differentiate to osteoblasts, adipocytes and chondroblasts stress.[30] However, these features were lost upon differenti-
in vitro.[19] The differentiation of MSCs firstly involves ation. In-vitro studies[29,31] found that higher levels of ROS
commitment to a lineage-specific progenitor and then mat- were associated with suppression of MSC osteogenesis
uration from progenitors to a specific cell type.[20] Lineage while favouring adipogenesis. Free radical generation adja-
commitment is under the control of several critical sig- cent to rodent bones in vivo is associated with formation of
nalling pathways, including wingless-type MMTV integra- new osteoclasts and higher levels of bone resorption, not
tion site (Wnt), Hedgehog, Notch, transforming growth unlike the effects of signalling molecules such as parathy-
factorb (TGFb) and bone morphogenetic protein (BMP), roid hormone (PTH) and interleukin-1 (IL-1).[32] PTH
and fibroblast growth factors. It has also been proposed and IL-1 induce osteoclastic breakdown of bone to release
that the nature of the cell’s microenvironment has some minerals with the intention of raising the levels of free cal-
control over its fate, with the phenotype formed being cium, phosphate, magnesium and other minerals in the
determined by the level of surrounding tissue elasticity. For blood: tipping the balance in favour of resorption over ossi-
example, MSCs in rigid matrices that resemble bone give fication. Indeed, higher levels of oxidative stress have been
rise to osteogenic progenitors; softer matrices mimicking associated with reduced bone mineral density and higher
brain give neurogenic cells.[21] More recently, differentia- risks of osteoporosis.[33]
tion has been found to be under the influence of micro- Nicolay et al. studied the effects of bleomycin, an anti-
RNAs, molecules that control post-translational gene neoplastic drug which forms free radicals and induces
expression.[20,22] By targeting transcription factors, micro- double-stranded DNA breaks, on MSCs.[34] It was found
RNAs can up- or downregulate the differentiation of differ- that following the administration of the drug, MCSs
ent cell lineages.[22] underwent an increased rate of apoptosis and while their
adipogenic differentiation potential was reduced, chon-
drogenic differentiation was not affected. The cells were
Patterns of differentiation
more sensitive to the drug than the lung fibroblasts they
In-vitro studies have proposed a hierarchal model of differ- were compared with. However, there were no changes to
entiation, with the early MSCs undergoing a sequential loss cell morphology, surface marker expression and the abil-
of lineage potential as they differentiate down the proposed ity to migrate and adhere. It was also found that the
levels; first adipogenic is lost, then chondrogenic. Following MSCs could repair the DNA breaks to some extent; thus,
this, the residual cells were found to be primarily osteo- the main proposed mechanism of cell death was via free
genic, suggesting that the osteogenic pathway is the ‘de- radical damage.[34]
fault’ outcome remaining if a cell is not committed to Prevention of mitosis is a mechanism of action expressed
another phenotype.[23,24] Formation of mature osteocytes is by drugs such as paclitaxel, etoposide, anthracyclines and
a multistep process. Firstly, the cells differentiate into pre- cytarabine by means of inhibiting or enhancing the activity
osteoblasts under the direction of multiple signalling path- of certain enzymes such as topoisomerase II or arresting
ways and transcription factors such as RUNX2.[25] Sec- the mitotic cycle. In 1997, Carlo-Stella et al. studied the
ondly, TGFb induces the migration of pre-osteoblasts to effects of acute myeloid leukaemia treatment protocols
sites of bone resorption,[26] the microenvironment of which including such drugs (idarubicin, daunorubicin and etopo-
is stiff and elastic (thus favouring osteogenesis) and where side) on the entire bone marrow environment and found
mechanical factors promote a more flattened cell that they left the marrow defective and unable to com-
shape.[21,27] Then, local soluble factors including BMP and pletely support haematopoietic progenitors and stromal
insulin-like growth factors (IGFs) activate multiple sig- cells.[35] This study did not include MSCs nor the mecha-
nalling pathways which promote pre-osteoblast maturation nism of the damage. However, a later study (Li et al.[36])
into mineralised tissue.[28] measured the effects of etoposide, cytarabine and paclitaxel

322 © 2018 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 70 (2018), pp. 320–327
Mia Baxter-Holland and Crispin R. Dass Doxorubicin and stem cells

on MSCs, and the results were consistent: reduction in pro- proliferation and lineage potential cannot be confidently
liferation, apoptosis and lack of recovery after 9 days (re- predicted from studies on similar drugs. To make in-vivo
tention of lineage potential was not studied). These results data harder to obtain, doxorubicin is often used in combi-
are contradictory to Mueller et al.,[37] who found that nation therapy with drugs such as cyclophosphamide,
MSCs were indeed resistant to etoposide, retaining prolifer- methotrexate and corticosteroids, making it difficult to
ation and lineage potential. Although these cells were trea- measure the effects of doxorubicin alone.
ted for shorter amounts of time, the authors were able to Negative effects of doxorubicin were reported on MSCs
isolate viable MSCs from the bone marrow of humans trea- both in vitro and in vivo (murine) by Oliveira et al.[42]
ted with chemotherapy.[37] Mueller et al. also compared Responses in the cells included lower proliferation rates,
the characteristics of MSCs isolated from the bone marrow alkaline phosphatase production deficiency, reduced poten-
of patients who had been exposed to standard- or high- tial for differentiation into cardiomyocytes and cytotoxicity
dose chemotherapy with those from unexposed bone mar- via apoptosis; however, positive and negative cell surface
row; no obvious differences in proliferation capacity and markers remained the same as the control. The doxoru-
differentiation potential were found.[37] bicin-exposed cells also did not express cardiac troponin T
Interestingly, Li et al.[36] also found that MSCs suffered following cardiomyogenic differentiation induction, unlike
minimal damage and almost complete recovery after expo- the control group. This hints that the drug’s influence on
sure to the alkylating agents cyclophosphamide and busul- MSCs may be partially responsible for its cardiotoxic
fan. The mechanism of action of these drugs involves adverse effects. Similar cytotoxic responses to doxorubicin
irreversibly attaching an alkyl group to the DNA helix were obtained by Yang et al.,[43] namely cell shrinkage,
which causes strand breakage and replication inhibition. A apoptosis and enhanced ROS production leading to mito-
case report of a human patient receiving cyclophosphamide chondrial membrane damage. It was also found that dox-
treatment obtained similar outcomes.[38] MSCs have also orubicin increased expression of mitogen-activated protein
shown resistance in vitro to treatment with drugs such as kinases (MAPK) signalling pathway precisely p38 and jun
cisplatin, camptothecin and vincristine (alkylating agent, N-terminal kinase (JNK), which plays an important role in
topoisomerase I inhibitor and mitotic inhibitor, respec- induction of inflammatory and apoptotic responses to
tively); full recovery and maintenance of lineage potential oxidative stress, among others.[43,44] When MAPK inhibi-
were observed.[36,37,39] Inconsistency in results suggests that tors were introduced into culture, lower levels of apoptosis
resistance may be influenced by external factors, such as the were observed. Elevated levels of these factors have consis-
composition of the cell culture medium or duration of tently been reported in other studies of doxorubicin-treated
treatment; further in-vivo studies are needed to form con- cells.[45,46] Yang et al.[43] also reported that doxorubicin
clusions that can be extrapolated to humans. increased the expression of p53 and cleaved-caspase-3 pro-
The above findings suggest that the extent of damage to teins, which signalled downstream genes including B-cell
the MSCs and the bone marrow environment depends on the lymphoma 2 (Bcl-2) and Bcl-2-like protein 4 (Bax) to cause
mechanism of action of the chemotherapy drug in question, cellular apoptosis. Thus, one hypothesised reason for apop-
whether it be DNA damage, mitosis inhibition, enzyme inhi- tosis was via ROS activation of phosphorylated p38, p53
bition or free radical generation. There is a lack of data and JNK.[43,47] In the same study, the secretion of vascular
involving the long-term effects of chemotherapy on in-vivo endothelial growth factor and insulin-like growth factor 1
MSCs and the bone marrow environment. Additionally, the (IGF-1) was decreased by doxorubicin. These factors are
precise mechanisms of resistance to chemotherapeutics have involved in the proliferation of and anabolic responses of
not been pinpointed and require further investigation; it has types of cells[48] and angiogenesis.[49] While this is a desir-
been suggested that the cells possess or have the ability to able effect in reducing tumour size, it could decrease the
develop mechanisms for repairing DNA cuts and damage, ability of MSCs to heal and maintain adult tissues.[43,47,48]
that the in-vivo response to acute injury may involve cell As MSCs have such a high regenerative potential, their
migration, induction of differentiation and proliferation, and presence and influence have been associated with promo-
that the apoptotic threshold may be elevated.[34,37,40,41] It has tion of tumour growth, such as apoptosis control, vascular
also been found that antioxidants were able to reduce the support, suppression of immune response and modulation
levels of apoptosis in MSCs.[36] of response to cytotoxic therapy.[50,51] MSCs appear to
migrate to injured tissue sites which includes both solid
tumours and the bone marrow in the case of haematologi-
Doxorubicin and stem cells
cal malignancies as such tissues often resemble those in
The complete range of influence doxorubicin has on MSCs need of repair.[52] MSCs can potentially promote tumour
has not yet been clearly elucidated. Due to its multiple and growth via stimulating angiogenesis in ischaemic tissue,[53]
unique mechanisms of action, changes to MSC health, paracrine secretion of multiple anti-apoptotic factors and

© 2018 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 70 (2018), pp. 320–327 323
Doxorubicin and stem cells Mia Baxter-Holland and Crispin R. Dass

Figure 1 Mesenchymal stem cells promote malignancy survival. TGFb, transforming growth factor b; Th1, helper T cell 1; BCRP, breast cancer resis-
tance protein; STAT3, signal transducer and activator of transcription 3; CCL5, C-C motif chemokine ligand 5; IL-6, interleukin 6; IL-8, interleukin 8.
[Colour figure can be viewed at wileyonlinelibrary.com]

growth factors, and differentiating into carcinoma-asso- that only the isolated secretome was used; this suggests the
ciated fibroblasts which promote metastasis and drug resis- secretome alone may not be responsible for resistance/sen-
tance.[54,55] Results are conflicting in experimental models. sitivity induction, or may be unable to produce such out-
Chen and colleagues[56] reported triple negative breast can- comes without presence of MSCs themselves. Ryu et al.[63]
cer showing increased resistance to doxorubicin upon and Kucerova et al.[64] observed suppression of human
interaction with adipose-derived MSCs (ASCs). The mech- breast cancer cell growth and increased chemosensitivity to
anism of such results involved decreased intracellular dox- doxorubicin, respectively, when in contact with ASCs.
orubicin accumulation within the breast cancer cells due to Causes of increased sensitivity were unclear. Similar growth
increased breast cancer resistance protein (BCRP) expres- inhibitory responses were expressed when human MSCs
sion, and increased secretion of IL-8 cytokines. MSCs also were intravenously injected into mice suffering Kaposi’s
appear able to aid breast cancer cells to evade immune sys- Sarcoma; they migrated to tumour sites, and tumours
tem attack via increasing helper T cell (Th) Th1 and TGF- decreased in size in a dose-dependent manner.[65] Bergfeld
ß1 production.[57] Building upon these findings, a study by et al.[66] found that circulating bone marrow MSCs were
Karnoub et al.[58] showed that the combination of MCSs incorporated into tumour cells and localised at the invasive
with weakly metastatic human breast carcinoma cells led to front of tumours, enhancing their expansion and protecting
a significant increase in the cells’ metastatic potential when them from drug-induced apoptosis. No definitive answer
the cells formed a xenograft. Increased secretion of the che- on the effects of MSCs on tumour growth in the presence
mokine CCL5 from MSCs led to paracrine responses in the of doxorubicin can yet be gleaned from the literature. It
breast cancer cells, stimulating their motility and metasta- appears that the influence on drug resistance depends on a
sis. A study by Tu et al.[59] showed osteosarcoma cells of multitude of factors including the context of MSC presence
the subtypes Saos-2 and U2-OS also displaying improved and treatment, and the specific cell type, for example ASCs.
survival rates from both doxorubicin and cisplatin treat- The nature of the tumour also plays a part, whether the cells
ment when grown in a medium containing MSCs com- are actively dividing or dormant, the type of malignancy
pared to control. Such cells displayed significantly lower and surrounding stromal cells, and the cellular interactions
levels of caspase 3/7 activity, thus preventing apoptosis, and within the tumour microenvironment.[64,67] This raises the
higher activity of IL-6 and STAT3 (signal transducer and question of whether the cytotoxic effect of doxorubicin is
activator of transcription 3), mediators in osteosarcoma indeed a negative thing; if MSCs aid tumour survival under
proliferation. Conclusive results were also obtained in vivo exposure to treatment with drugs such as doxorubicin,
from mouse models.[59] Multiple studies have shown toxic effects may not be so undesirable (Figure 1).
STAT3 inhibition being to be able to reverse drug (includ- Doxorubicin has a wide range of adverse effects includ-
ing doxorubicin) resistance, suggesting that STAT3 is ing immunosuppression, dyspigmentation, gastrointestinal
important in resistance development.[59–61] disturbances leading to severe nausea and vomiting, and
Other experimental models present different outcomes. life-threatening cardiotoxicity. The latter typically presents
No significant changes in doxorubicin sensitivity were dis- as acute or insidious cardiomyopathy and heart failure and
played by A459 lung cancer cells that had been treated in is exponentially dose-dependent.[68,69] Anthracycline expo-
culture with Wharton’s jelly-derived MSC secretome, the sure to the cardiac progenitor cells or MSCs has been pro-
name given to the immunomodulatory cytokines, growth posed as a cause for such toxicity, reducing the body’s
factors and chemokines produced by MSCs.[62] It is noted ability to heal and repair the heart.[43,48] Current treatments

324 © 2018 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 70 (2018), pp. 320–327
Mia Baxter-Holland and Crispin R. Dass Doxorubicin and stem cells

Table 1 Key papers on doxorubicin and mesenchymal stem cells

References Findings

[42] Lower proliferation rates, alkaline phosphatase production deficiency, reduced potential for differentiation into cardiomyocytes
and cytotoxicity via apoptosis
Dox dose = 5 mg/kg per week for 4 weeks
[43] Cell shrinkage, apoptosis and enhanced ROS production lead to mitochondrial membrane damage
Dox dose = 1, 3 and 10 lM
[56] MSCs increased drug resistance in triple negative breast cancer cells
Dox dose = 200 nM, 2 lM
[59] MSC co-culture with osteosarcoma cells displayed improved survival rates against doxorubicin
Dox dose = 10 mg/kg per week for 4 weeks
[64] MSCs cause suppression of human breast cancer cell growth and sensitivity to doxorubicin, respectively
Dox dose = 6.25–100 ng/ml
Administration of MSCs after or during doxorubicin treatment improves cardiac function in animal studies
[72] Dox dose = 3 mg/kg weekly for 6 weeks
[73] Dox dose = 2.5 mg/kg thrice weekly for 2 weeks
ROS, reactive oxygen species; MSCs, mesenchymal stem cells.

include angiotensin-converting enzyme inhibitors, beta- importance in the maintenance and repair of human tis-
blockers or heart transplantation.[70] The use of MSCs for sues. Certainly, the literature shows that drugs such as
cardio-protection or cardio-regenerative therapy in dox- doxorubicin have the possibility to cause severe and pos-
orubicin patients has been considered, however such ther- sibly irreversible damage to the bone marrow environ-
apy is undertaken with an air of caution due to the ment and MSCs, via mechanisms such as free radical
observed tumour-proliferation effects of MSCs in response damage and induction of apoptotic pathways (sum-
to doxorubicin.[71] Several studies of such therapy have marised in Table 1). Whether this damage is clinically sig-
been successful and promising. Administration of bone nificant for the remainder of the patient’s lifetime is not
marrow-derived MSCs (BMMSCs) 2 weeks after doxoru- yet known. MSCs have been shown to lead to tumour
bicin treatment generated significant improvement in left resistance when under treatment with doxorubicin, this
ventricular function in rabbits.[72] Another study in rats could affect the success of cancer treatment and the use
showed that daily dosing of BMMSCs for 10 days 10 weeks of MSCs to repair tissues damaged by chemotherapy. Fur-
after doxorubicin treatment improved cardiac contractility ther investigation into the mechanisms of resistance to
and reduced myocardium fibrosis and left ventricular chemotherapy treatment could shed light on ways to
diameter.[73] Finally, contradictory to fears that MSCs may overcome them.
enhance tumour proliferation in the presence of doxoru-
bicin, Oliveira et al. observed a partial cardioprotective
Declarations
effect in rats when treated with MSCs during doxorubicin
therapy.[74] However, the duration of the beneficial effect is
Conflict of interest
not known.
The Author(s) declare(s) that they have no conflicts of
interest to disclose.
Conclusions
Due to the lack of long-term clinical studies on the conse-
Acknowledgements
quences on stem cells of chemotherapy, projections of
how doxorubicin may influence a patient’s health post- The authors acknowledge support via an Academic50 grant
treatment are not very clear. Stem cells are of vital to CRD.

References 2. Krohn K. Anthracycline Chemistry and anthracycline antibiotics adriamycin


Biology II. Berlin: Springer, Berlin and daunorubicin. Biochem Pharmacol
1. Agrawal K. Doxorubicin. xPharm: The Heidelberg, 2008. 1999; 57: 727–741.
Comprehensive Pharmacology Refer- 3. Gewirtz DA. A critical evaluation of 4. Chen NT et al. Probing the dynamics
ence. New Orleans, LA: Elseiver Pub- the mechanisms of action proposed of doxorubicin-DNA intercalation
lishing, 2007. for the antitumor effects of the during the initial activation of

© 2018 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 70 (2018), pp. 320–327 325
Doxorubicin and stem cells Mia Baxter-Holland and Crispin R. Dass

apoptosis by fluorescence lifetime improving regenerative medicine. 30. Saretzki G et al. Stress defense in
imaging microscopy (FLIM). PLoS Stem Cells Transl Med 2016; 5. murine embryonic stem cells is supe-
ONE 2012; 7: e44947. 17. Okolicsanyi RK et al. Mesenchymal rior to that of various differentiated
5. Agudelo D et al. Intercalation of anti- stem cells, neural lineage potential, murine cells. Stem Cells 2004; 22:
tumor drug doxorubicin and its ana- sulfate proteoglycans and the matrix. 962–971.
logue by DNA duplex: structural Dev Biol 2014; 388: 1–10. 31. Turker I et al. Oxidative stress as a
features and biological implications. 18. da Silva Meirelles L et al. Mesenchy- regulator of adipogenesis. FASEB J
Int J Biol Macromol 2014; 66: 144–150. mal stem cells reside in virtually all 2007; 21: A1053.
6. Zunino F et al. The inhibition in vitro post-natal organs and tissues. J Cell 32. Garret IR et al. Oxygen-derived free
of DNA polymerase and RNA poly- Sci 2006; 119: 2204–2213. radicals stimulate osteoclastic bone
merases by daunomycin and adri- 19. Dominici M et al. Minimal criteria for resorption in rodent bone in vitro and
amycin. Biochem Pharmacol 1975; 24: definiting multipotent mesenchymal in vivo. J Clin Investig 1990; 85: 632–
309–311. stromal cells. The International Society 639.
7. Bush N et al. DNA topoisomerases. for Cellular Therapy position state- 33. Basu S et al. Association between
EcoSal Plus 2009; 6. ment. Cytotherapy 2006; 8: 315–317. oxidative stress and bone mineral den-
8. Corbett AH, Osheroff N. When good 20. Chen Q et al. Fate decisions of mes- sity. Biochem Biophys Res Commun
enzymes go bad: conversion of topoi- enchymal stem cells: adipocytes of 2001; 288: 275–279.
somerase II to a cellular toxin by anti- osteoblasts? Cell Death Differ 2016; 23: 34. Nicolay NH et al. Mesenchymal stem
neoplastic drugs. Chem Res Toxicol 1128–1139. cells are sensitive to bleomycin treat-
1993; 6: 585–597. 21. Engler AJ et al. Matrix elasticity ment. Sci Rep 2016; 6: 26645.
9. Mizutani H et al. Mechanism of directs stem cell lineage specification. 35. Carlo-Stella C et al. Effects of
apoptosis induced by doxorubicin Cell 2006; 126: 677–689. chemotherapy for acute myelogenous
through the generation of hydrogen 22. Huang C et al. MicroRNAs in regula- leukaemia on haematopoietic and
peroxide. Life Sci 2005; 76: 1439– tion of osteogenic differentiation of fibroblast marrow progenitors. Bone
1453. mesenchymal stem cells. Cell Tissue Marrow Transplant 1997; 20: 465–471.
10. Goodman J, Hochstein P. Generation Res 2017; 368: 229–238. 36. Li J et al. Differential damage and
of free radicals and lipid peroxidation 23. Muralgia A et al. Clonal mesenchymal recovery of human mesenchymal stem
by redox cycling of adriamycin and progenitors from human bone mar- cells after exposure to chemotherapeu-
daunomycin. Biochem Biophys Res row differentiate in vitro according to tic agents. Br J Haematol 2004; 127:
Comm 1977; 77: 797–803. a hierarchical model. J Cell Sci 2000; 326–334.
11. Tacar O et al. Doxorubicin: an 113: 1161–1166. 37. Mueller LP et al. Presence of mes-
update on anticancer molecular 24. Pittenger M et al. Multilineage poten- enchymal stem cells in human bone
action, toxicity and novel drug deliv- tial of adult human mesenchymal marrow after exposure to chemother-
ery systems. J Pharm Pharmacol 2013; stem cells. Science 1999; 284: 143–147. apy: evidence of resistance to apopto-
65: 157–170. 25. Franceschi RT et al. Transcriptional sis induction. Stem Cells 2006; 24:
12. Brucker J et al. Non-pegylated liposo- regulation of osteoblasts. Cells Tissues 2753–2765.
mal doxorubicin for patients with Organs 2008; 189: 144–152. 38. Baran DT et al. Recovery from aplas-
recurrent ovarian cancer: a multicen- 26. Tang Y et al. TGF-ß1-induced migra- tic anaemia after treatment with
tric phase II trial. Oncol Lett 2016; 12: tion of bone mesenchymal stem cells cyclophosphamide. N Engl J Med
1211–1215. couples bone resorption and forma- 1976; 295: 1522–1523.
13. Keizer HG et al. Doxorubicin (adri- tion. Nat Med 2009; 15: 757–765. 39. Liang W et al. Human adipose tissue
amycin) – a critical review of free rad- 27. Wang YK, Chen CS. Cell adhesion derived mesenchymal stem cells are
ical dependent mechanisms of and mechanical stimulation in the resistant to several chemotherapeutic
cytotoxicity. Pharmacol Ther 1990; 47: regulation of mesenchymal stem cell agents. Cytotechnology 2011; 63: 523–
219–231. differentiation. J Cell Mol Med 2013; 530.
14. Davie LA, Zur Neiden NI. Mesoder- 17: 823–832. 40. Almohamad K et al. Marrow stromal
mal fate decisions of a stem cell: the 28. Li J et al. The role of bone marrow cell recovery after radiation induced
wnt switch. Cell Mol Life Sci 2008; 65: microenvironment in governing the aplasia in mice. Int J Radiat Biol 2003;
2658. balance between osteoblastogenesis and 79: 259–267.
15. Thomson JA et al. Embryonic stem adipogenesis. Aging Dis 2016; 7: 514– 41. Gimble JM et al. Peroxisome prolifer-
cell lines derived from human blasto- 525. ator-activated receptor-gamma activa-
cysts. Science 1998; 282: 1145–1147. 29. Bai XC et al. Oxidative stress inhibits tion by thiazolidinediones induces
16. De La Garza A et al. Concise review: osteoblastic differentiation of bone adipogenesis in bone marrow stromal
hematopoietic stem cell origins: les- cells by ERK and NF-jB. Biochem Bio- cells. Mol Pharmacol 1996; 50: 1087–
sons from embryogenesis for phys Res Commun 2004; 314: 197–207. 1094.

326 © 2018 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 70 (2018), pp. 320–327
Mia Baxter-Holland and Crispin R. Dass Doxorubicin and stem cells

42. Oliveira MS et al. Doxorubicin has murine skin. Microvasc Res 2012; 83: 64. Kucerova L et al. Altered features and
in vivo toxicological effects on ex vivo 267–275. increased chemosensitivity of human
cultured mesenchymal stem cells. Tox- 54. Hung SC et al. Angiogenic effects of breast cancer cells mediated by
icol Lett 2014; 224: 380–386. human multipotent stromal cell adipose-tissue derived mesenchymal
43. Yang F et al. Doxorubicin caused conditioned medium activate the stromal cells. BMC Cancer 2013; 13:
apoptosis of mesenchymal stem cells PI3K-Akt pathway in hypoxic 535.
via p38, JNK and p53 pathway. Cell endothelial cells to inhibit apoptosis, 65. Khakoo AY et al. Human mesenchy-
Physiol Biochem 2013; 32: 1072–1082. increase survival, and stimulate mal stem cells exert potent antitu-
44. Sun Y et al. Signaling pathway of angiogenesis. Stem Cells 2007; 25: morigenic effects in a model of
MAPK/ERK in cell proliferation, dif- 2363–2370. Kaposi’s sarcoma. J Exp Med 2006;
ferentiation, migration, senescence 55. Mishra PJ et al. Carcinoma-associated 203: 1235–1247.
and apoptosis. J Recept Signal Trans- fibroblast-like differentiation of 66. Bergfeld SA et al. Bone marrow-
duct Res 2015; 35: 600–604. human mesenchymal stem cells. Can- derived mesenchymal stromal cells
45. Ghosh J et al. The protective role of cer Res 2008; 68: 4331–4339. promote survival and drug resistance
arjunolic acid against doxorubicin 56. Chen DR et al. Mesenchymal stem in tumor cells. Mol Cancer Ther 2014;
induced intracellular ROS dependent cell-induced doxorubicin resistance in 13: 962–975.
JNK-p38 and p-53 mediated cardias triple negative breast cancer. Biomed 67. Zimmerlin L et al. Mesenchymal stem
apoptosis. Biomaterials 2011; 32: Res Int 2014; 2014: 532161. cell secretome and regenerative ther-
4857–4866. 57. Patel SA et al. Mesenchymal stem cells apy after cancer. Biochimie 2013; 95:
46. Das J et al. Taurine suppresses dox- protect breast cancer cells through 2235–2245.
orubicin-triggered oxidative stress and regulatory T cells: role of mesenchy- 68. Gianni L et al. Anthracycline car-
cardiac apoptosis in rat via up-regula- mal stem cell-derived TGF-beta. J diotoxicity: from bench to bedside. J
tion of PI3-K/Akt and inhibition of Immunol 2010; 184: 5885–5894. Clin Oncol 2008; 26: 3777–3784.
p53, p38-JNK. Biochem Pharmacol 58. Karnoub AE et al. Mesenchymal stem 69. Volkova M, Russel R 3rd. Anthracy-
2011; 81: 891–909. cells within tumour stroma promote cline cardiotoxicity: prevalence, patho-
47. Johnson GL, Lapadat R. Mitogen-acti- breast cancer metastasis. Nature 2007; genesis and treatment. Curr Cardiol
vated protein kinase pathways medi- 449: 557–565. Rev 2011; 7: 214–220.
ated by ERK, JNK and p38 protein 59. Tu B et al. Mesenchymal stem cells 70. Georgakopoulos P et al. Cardioprotec-
kinases. Science 2002; 298: 1911–1912. promote osteosarcoma cell survival tive effect of metoprolol and enalapril
48. Ren J et al. Insulin-like growth factor and drug resistance through activation in doxorubicin-treated lymphoma
I as a cardiac hormone: physiological of STAT3. Oncotarget 2016; 7: 48296– patients: a prospective, parallel-group,
and pathophysiological implications in 48308. randomized, controlled study with 36-
heart disease. J Mol Cell Cardiol 1999; 60. Huang S et al. Inhibition of activated month follow-up. Am J Hematol 2010;
31: 2049–2061. STAT3 reverses drug resistance to 85: 894–896.
49. Ferrara N. Vascular endothelial chemotherapeutic agents in gastric 71. Ezquer F et al. Mesenchymal stem cell
growth factor. Arterioscler Thromb cancer cells. Cancer Lett 2012; 315: therapy for doxorubicin cardiomyopa-
Vasc Biol 2009; 29: 789–791. 198–205. thy: hopes and fears. Stem Cell Res
50. Klopp AH et al. Concise review: dis- 61. Gariboldi MB et al. Inhibition of Stat3 Ther 2015; 6: 116.
secting a discrepancy in the literature: increases doxorubicin sensitivity in a 72. Garbade J et al. Bone marrow-derived
do mesenchymal stem cells support or human metastatic breast cancer cell stem cells attenuate impaired contrac-
suppress tumour growth? Stem Cells line. Cancer Lett 2007; 258: 181–188. tility and enhance capillary density in
2011; 29: 11–19. 62. Hendijani F et al. Effect of human a rabbit model of Doxorubicin-
51. Houthuijzen JM et al. The role of Wharton’s jelly mesenchymal stem cell induced failing hearts. J Card Surg
mesenchymal stem cells in anti-cancer secretome on proliferation, apoptosis 2009; 24: 591–599.
drug resistance and tumour progres- and drug resistance of lung cancer 73. Mohammadi Gorji S et al. Effects of
sion. Br J Cancer 2012; 106: 1901– cells. Res Pharm Sci 2015; 10: 134– mesenchymal stem cells on doxoru-
1906. 142. bicin-induced fibrosis. Cell J 2012; 14:
52. Stagg J. Mesenchymal stem cells in 63. Ryu H et al. Adipose tissue-derived 142–151.
cancer. Stem Cell Rev 2008; 4: 119– mesenchymal stem cells cultured at 74. Oliveira MS et al. Doxorubicin car-
124. high density express IFN-b and sup- diotoxicity and cardiac function
53. Schlosser S et al. Paracrine effects of press the growth of MCF-7 human improvement after stem cell therapy
mesenchymal stem cells enhance vas- breast cancer cells. Cancer Lett 2014; diagnosed by strain echocardiography.
cular regeneration in ischaemic 352: 220–227. J Cancer Sci Ther 2013; 5: 52–57.

© 2018 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 70 (2018), pp. 320–327 327

You might also like