0007B PDF

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 22

Send Orders for Reprints to reprints@benthamscience.

net
Current Pharmaceutical Design, 2014, 20, 201-222 201

Cancer Therapy: Targeting Mitochondria and other Sub-cellular Organelles

Obinna C. Ubah1 and Heather M. Wallace2*

1
Department of Molecular and Cell Biology, College of Life Sciences and Medicine, University of Aberdeen, AB25 2ZP, UK;
2
Division of Applied Medicine and Therapeutics, School of Medicine and Dentistry, University of Aberdeen, Aberdeen AB25 2ZD, UK

Abstract: Tumour cell death is required for the clearance of malignant cells and is a vital part of the mechanism of natural tumour sup-
pression. Cancer cells, having acquired multiple deregulated pathways involving several cellular oragenelles, are capable of disrupting
these normally finely tuned processes thereby evading both physiological and therapeutic intervention. Although current available data
indicate the dependence of successful tumour cell clearance on classical apoptotic pathways (intrinsic and/or extrinsic pathways), there is
now evidence suggesting that alternative apoptotic and non-apoptotic pathways may effectively contribute to tumour cell death. The mi-
tochondria, proteasomes, endoplasmic reticulum, Golgi apparatus, lysosomes and lysosome-related organelles of tumour cells exhibit a
number of deregulations which have been identified as potential druggable targets for successful rational drug design and therapy. In this
review, we summarise the roles of these cellular organelles in tumour initiation and establishment as well as current trends in develop-
ment of agents that target deregulations in these organelles.

Keywords: Cancer therapy, polyamines, apoptosis, Endoplasmic reticulum, Golgi apparatus, Lysosome, Proteasome, , ROS, MOMP,
Warburg effect.

INTRODUCTION membrane-bound vesicles that encapsulate proteases and other


There are six established alterations in cell physiology that dic- hydrolytic enzymes and these are the cellular organelles responsible
tate malignant transformation: growth signalling autonomy, insensi- for degrading extracellular and transmembrane proteins, whereas
tivity to anti-proliferating signals, limitless proliferative potential, proteasomes degrade intracellular proteins [7]. It is the compart-
sustained angiogenesis, adjacent tissue invasion and metastasis, and mentalisation of these proteolytic enzymes that prevent them from
avoidance of cell death. This intrinsic lack of cell death remains a producing uncontrolled protein degradation [5]. For tumourigenesis
major cause of therapeutic failure and may itself contribute to car- to be successful, an effective suppression of both the classical apop-
cinogenesis and tumour progression [1, 2]. Therefore, activation of tosis and lysosomal death pathways must occur, thus supporting the
the cell death machinery via drugs designed rationally may signify crucial involvement of these organelles in tumorigenesis [8].
a more effective cancer treatment option. The cell death machinery
MITOCHONDRIA
is composed of catabolic hydrolases which are modulated by spe-
cific inhibitors or via sequestration of their activators. Numerous i. Structure and Roles
cell death cascades (apoptotic and necrotic) converge on mitochon-
dria to cause deregulation of function and permeabilisation with the The rationale for targeting the mitochondria for therapeutic
activators of the cell death machinery being liberated following the benefit is based on their critical involvement in bioenergetics, redox
permeability change in the mitochondrial outer membrane [3]. To balancing and vitally their active regulation of several cell sur-
remove abnormal malignant cells, their death is an important event vival/death pathways. Other important roles include thermogenesis,
which also provides a critical mechanism of innate tumour suppres- Ca2+ homeostasisand essential anabolic cascades. Mitochondrial
sion. Aberrations blocking these tightly regulated processes tilt the dysfunction such as impaired oxidative phosphorylation and/or
balance in favour of cancer cells successfully evading cell death increased oxidative stress coupled with deregulation of apoptosis
from either therapeutic means or physiological control systems. has been described recently as a vital pathophysiological mecha-
Although the clearance of malignant cells often relies on manipulat- nism not only in human mitochondrial diseases but also in the
ing the classical apoptotic pathways (mitochondrial and/or death pathogenesis of other congenital and acquired pathologies such as
receptor cascades), evidence is emerging that other apoptotic and cancer [9], neurodegenerative disorders [10], diabetes [11], and
non-apoptotic pathways may play vital roles in tumour cell death. cardiovascular diseases [12].
Experimental evidence has suggested that the endoplasmic reticu- Mitochondria were regarded exclusively as the power house of
lum (ER) and Golgi apparatus can activate both pro-survival cas- the cell throughout the second-half of the 20th century. It was not
cades as well as cell suicide programmes if the stress-signalling until around 1995 that it became clear that they are not only a site
threshold is exceeded [4]. The proteasome is an abundant multi- for energy production via oxidative phosphorylation, but also have
enzyme complex providing the focal pathway for the degradation of crucial role in cell death regulation. This latter discovery which
intracellular proteins in eukaryotes, thus it is the modulator of the revitalised the field of mitochondrial and cell death research ap-
protein pool and is important for cell-cycle progression and apop- peared counterintuitive because experts doubted whether the meta-
totic cell death in both healthy and malignant cells [5, 6]. Cells use bolic functionality of the cell which converge oin the mitochondria
an exceptional assortment of mechanisms to regulate intracellular could be manipulated to cause cell death. The other argument was
cytosolic protein stability and degradation, lysosomes and protea- that the mitochondria during apoptosis do not suffer significant
somes remain the two main routes amongst other mechanisms re- ultra-structural changes thus may not be controlling the fate of the
sponsible for protein integrity. Lysosomes are cytoplasmic cell. Since 2001, over 13,000 articles have been published linking
apoptosis and mitochondria, thus the initial opposition to the con-
cept of mitochondrial cell death control has been overcome [13].
*Address correspondence to this author at the Division of Applied Medicine
and Therapeutics, School of Medicine and Dentistry, University of Aber- In healthy cells, the inner mitochondrial membrane (IM) is
deen, Aberdeen AB25 2ZD, UK; Tel/Fax: +44 1224 437956; almost impermeable to all ions and including protons (H+). This
E-mail: h.m.wallace@abdn.ac.uk allows respiratory chain complexes I - IV to accumulate proton

1873-4286/14 $58.00+.00 © 2014 Bentham Science Publishers


202 Current Pharmaceutical Design, 2014, Vol. 20, No. 2 Ubah and Wallace

gradient across the IM which is obligatory for oxidative phosphory- off the Bcl2-homology domain 3 (BH3) into the active form tBid
lation [14, 15]. Ions and small molecules crossing the inner mem- (truncated BID), the latter providing a cross-talk between the ex-
brane do so via specific transporters. The inner mitochondrial trinsic and intrinsic death pathways. tBid then translocates to the
transmembrane potential (m) is generated from the charge imbal- mitochondria where it promotes the mitochondrial permeability
ance resulting from the creation of an electrochemical gradient transition (elevated OM permeability) causing cytochrome c (cyt c)
across the inner membrane. Finally, ATP synthesis is driven by the release. The mitochondrial pathway is initiated by the release of
complex V (mitochondrial ATP synthase) of the respiratory chain various apoptogenic factors such as cyt c, apoptosis inducing factor
via the exploitation of the proton gradient. Therefore the mainte- (AIF), second mitochondria-derived activator of caspases
nance of the proton gradient is crucial for cellular bioenergetics [14 (Smac)/direct inhibitor of apoptosis proteins (IAP) binding protein
- 16]. All components of the mitochondrial matrix and those me- with Low PI (DIABLO), Omi/high temperature requirement protein
tabolites crossing the IM are strictly regulated with the aid of highly A (HtrA2) and Endonuclease G (Endo-G) from the IMS into the
selective transport proteins. A permanent dissipation of m is cytosol [13, 29 - 31]. The apoptogenic proteins can be released into
associated with cell death [17, 18], however cases of transient loss the cytosol using one of the four models hypothesised for the open-
of m secondary to functional instability in one or more IM pores ing of the permeability transition pore; [1] Oligomerised Bax/Bak
may occur in physiological conditions [19, 20]. The most vital generates channels sufficient for the liberation of the factors [32].
function of the mitochondria is the synthesis of ATP. Dysfunction This hypothesis is sustained by the findings that the structure of
of this process is associated with large number of mitochondrial Bcl-2 family proteins is similar to that of the pore-forming helices
pathologies. of bacterial toxins, and that Bak can form channels in artificial
In 1972-1973, Boveris and Chance described mitochondria, membranes and liberate cyt c from liposomes with large complexes
isolated from rat liver and pigeon heart, as a source of hydrogen containing Bax and Bak, as seen by electron microscopy [33]. [2]
peroxide (H2O2) that diffuses to the cytosol as a major supplier of The proapoptotic proteins may form complexes with mitochondrial
superoxide anions [21, 22]. Although the cytochrome oxidase membrane proteins such as the voltage-dependent anion channel
(complex IV) mediates the four-electron reduction of molecular (VDAC) or adenine nucleotide translocator (ANT) to assemble
oxygen to form H2O which is the end product of the respiratory pores [34]. This is supported by findings that microinjection of
chain, a minor fraction of molecular oxygen takes part in one- VDAC antibodies into the entrance of the channel inhibited the
electron reduction processes generating reactive oxygen species release of cyt c and prevented apoptosis [35, 36]. [3] Permeability
(ROS) and they include; O2-, H2O2, and the highly reactive hydroxyl transition pores are regulated by misfolded proteins and chaperones
radical (HO-). such as Hsp25. These permeability transition pores are formed by
aggregates of misfolded membrane proteins [37, 38]. [4] Membrane
The intramitochondrial metabolites O2-, H2O2, NO and HO - are damage resulting from the matrix swelling may induce cristae struc-
pro-oxidants with potential to cause oxidative stress and tissue ture distortion and rupture of the outer membrane thus releasing cyt
damage. These are the chemical species that are linkedto the mo- c [39, 40].
lecular mechanism of tissue dysfunction in aging, ischemia-
reperfusion, neurological diseases, and inflammation [16, 23]. Also the mitochondrial pathways can be triggered by various
Normally, the native defense systems consisting primarily of mito- intracellular and extracellular stress signals which ultimately lead to
chondrial (manganese-containing) and cytosolic (copper-zinc- activation of pro-apoptotic proteins such as Bcl2 antagonist/killer
containing) superoxide dismutases, glutathione peroxidase, and (Bak), Bcl2-associated X protein (Bax), or inactivation of anti-
phospholipid hydroperoxide neutralise these ROS and their peroxi- apoptotic Bcl2 family members such as Bcl2 or Bcl-xL [41, 42]. The
dation products respectively [23 - 25]. Antioxidant supplements and release of cyt c from the mitochondria causes activation of down-
drugs have been used as mitochondria-generated free radical scav- stream effector pro-caspases such as caspases-3 and 7 via formation
engers. These drugs inhibit mitochondria in tumour cells, thereby of a large cytosolic complex, the cyt c/Apaf-1/caspases-9 complex
killing these cancer cells; they also protect cells from oxidative [2, 13, 42]. Endo-G and apoptosis inducing factor (AIF) are capable
destruction, and aid in the repair of defects. of inducing caspase-independent cell death and produce DNA
fragmentation [31, 37]. Although the mitochondrial membrane
In healthy cells and during cell death, the outer mitochondrial permeabilisation (MMP) is vital for caspase-dependent and inde-
membrane (OM) permeability is also altered. Due to the presence of pendent apoptotic cell death pathways, the cell death cascade can
the voltage dependent anion channel diffusion of metabolites and also be induced by triggers found in other organelles such as cal-
solutes of ~5kDa across the OM occurs. Although the assumption cium (Ca2+) in the endoplasmic reticulum (ER) and cathepsins in
that the OM is freely permeable by such solutes has been chal- lysosomes in response to ROS or ceramide [37, 43]. The apoptosis
lenged recently because real-time quantification of mitochondrial pathways are strictly controlled by a variety of pro- and anti- apop-
Ca2+ concentrations revealed the existence of Ca2+ microdomains in totic regulators under physiological conditions, since any form of
which the voltage dependent anion channel and a variety of addi- uncontrolled stimulation of the apoptotic machinery would poten-
tional OM proteins regulate and limit Ca2+ diffusion across the OM tially translate to a detrimental effects on cell survival. In human
[26, 27]. The OM permeability often increases during cell death cancers, the anti-apoptotic systems are often abnormally upregu-
allowing for the liberation of soluble proteins into the intermem- lated thus enabling the cancer cells to evade apoptotic cell death
brane space (IMS), these proteins are otherwise situated and re- [44].
tained within the mitochondria in healthy cell conditions. More so,
the death associated outer membrane permeabilisation can be an iii. Mitochondria in Cancer Cells
accidental process as well as a finely controlled process with major
Metabolic imbalances and improved resistance to mitochondrial
implications for health and disease [13, 23].
cell death are considered as characteristic features of cancer cells.
ii. Mitochondria and Cell Death Tumours relying on glycolysis to meet their metabolic burdens have
been acknowledged since the twentieth century [45, 46]. Cancer
In general, two semi-interdependent apoptotic signalling path- cells are inclined to synthesise ATP mainly through aerobic glyco-
ways have been delineated; ligand binding to the death receptors at lysis (the so-called Warburg effect), a metabolic state that is associ-
the cell surface (extrinsic) pathway, and the mitochondrial (intrin- ated with elevated glucose uptake and local acidification owing to
sic) pathway [28]. Activation of either signalling pathway will lead lactate production. The Warburg effect is actually the principle
to the activation of caspases, a family of cysteine proteases that behind the development of the positron emission tomography in
function as death effector molecules. However, active caspase-8 which a glucose analogue tracer (2-18fluoro-2-deoxy-D-glucose) is
can cleave the protein BID (BH3-interacting-domain death agonist) utilised to differentiate between normal and malignant tissue. How-
Cancer Therapy Current Pharmaceutical Design, 2014, Vol. 20, No. 2 203

ever, a comprehensive clarification of the so-called Warburg effect  Putatively, in a one-step process cancer cells might repress the
has not been achieved. Some of the non-exclusive hypotheses mitochondrial apoptotic programme via mechanisms that in
which have been put forward to explain this phenotype are de- concert stimulate the Warburg phenomenon. The observation
scribed below: that mitochondrial DNA mutations may induce, through un-
 Cancer cells are deficient in mitochondrial respiration and known mechanisms, a decrease of anti-cancer-induced or
ATP generation as a result of accumulation of defects in the spontaneous apoptosis in cancer cells is in favour of this hy-
mitochondrial genome. Also, mitochondrial germline muta- pothesis [48]. Moreover, hexokinase II which increases in gly-
tions have been implicated as a predisposing factor in cancer colysis and inhibits MOMP when it is in association with
development. In most cases, such mitochondrial mutations are VDAC in the OM provides a possible link between MOMP
acquired during and after tumourigenesis. Brandon et al suppression and aerobic glycolysis. This interaction between
grouped these acquired mitochondrial DNA mutations in two hexokinase and VDAC is favoured by activation of the Akt
different classes; the first class involves severe mutations that pathway [53] or by the cancer-specific overexpression of mi-
impede oxidative phosphorylation, thus causing an increased tochondrion-binding hexokinase isoenzymes [48, 62].
and enhanced ROS production and tumour cells proliferation  During multistep carcinogenesis, metabolic alteration and
while the second category is of milder mutations permitting MOMP inhibition could be autonomously acquired by cancer
tumour cells adaptation to their new microenvironment, espe- cells. Increased ROS and respiratory dysfunction resulting
cially during progression and metastases [47 - 49]. from mitochondrial mutations could increase the probability of
 Mitochondrial enzymes can in specific cases act as tumour cancer cells committing to apoptotic MOMP, and MOMP in-
suppressors and mutations of these nuclear encoded mitochon- hibition would be necessary to maintain carcinogenesis. Oth-
drial proteins have been linked to cancer and will indirectly erwise, suppression of the apoptotic pathway might be permis-
encourage aerobic glycolysis. For instance, the inactivating sive for genomic instability [63], and the Darwinian selection
mutation of two enzymes involved in the TCA cycle; mito- of genetically variant cells would then favour the accumulation
chondria succinate dehydrogenase (SDH subunits B, C or D) of cells expressing the Warburg phenotype.
or fumarate dehydrogenase is considered an oncogenic event In the last ten years, there has been substantial interest in the
causing phaeochromocytoma and leiomyoma, leiomyosacr- possibility that mtDNA mutations might predispose or at least play
coma or renal carcinoma respectively. Also accumulation of a role in human malignancies and other common diseases. Al-
succinate and fumarate in the cytosol as a result of loss of though the mechanism involved in the initiation and progression of
function of their respective dehydrogenases favours the activa- mtDNA mutations, and their involvement in tumourigenesis and
tion of the hypoxia-inducible transcription factor (HIF) and other pathogenesis awaits comprehensive understanding. Interest-
generate a pseudohypoxic state together with HIF-dependent ingly, a recent report showed that the high heterogeneity of human
reprogramming of the metabolism towards aerobic glycolysis mtDNA was significantly amplified in tumours [64]. Polyak et al.
[50]. A mutation in the third TCA cycle enzyme, isocitrate de- demonstrated a consistent presence of somatic mtDNA mutations in
hydrogenase has recently been linked to the majority of grade human tumours. The authors provided evidence of the presence of
II and III gliomas and secondary glioblastomas [51, 52]. homoplasmic mtDNA mutations in 7 out of 10 cell lines from pa-
 As the pre-malignant lesion grows progressively further away tients with colorectal carcinomas which were neither found in nor-
from the blood supply, cancer cells (if not all solid tumours) mal colon nor in other tissues from the same patients [65]. Recent
possess the characteristic ability to adapt to decreased oxygen papers report somatic mutations in the mitochondrial genome in
tension (hypoxia). One way in which these cells adapt to de- nearly one out of every four gastric cancer specimens and stress the
creased oxygen tension is to shut down mitochondrial respira- potential role of those mutations in the evolution of the disease
tion and simultaneously switch on glycolysis [50, 53]. [66]. Kulawiec et al. [67] demonstrated that mtDNA mutations
were not associated with ROS generation in some samples of breast
 Cancer cells often upregulate the enzymes and rate-limiting cancer cells, but constitutively activated the PI3K/AKT pathway
processes of glycolysis, including glucose transporters, for in- contributing to enhanced metastasis. More over, the PI3K/AKT
stance as a result of the expression of oncogenic factors in- pathway is strictly linked and activated in association with the ser-
cluding Ras, Src, or Bcl-Abl or as a result of the constitutive ine/threonine kinase target of rapamycin (mTOR) that regulates key
signalling through the Akt pathway [48, 54]. cellular processes such as cell survival, growth and proliferation.
Cancer cells have been shown to be resistant to the induction of The mTOR pathway is regularly hyperactivated in many human
mitochondrial outer membrane permeabilisation (MOMP), a proc- tumours and this is consistent with its role in cell proliferation [68].
ess which mediates the intrinsic apoptotic pathway [55]. MOMP In cancer, oxidative stress has been linked to DNA instability,
being a complex phenomenon is tightly regulated by Bcl-2 family hypermethylation and mutation in the DNA repair machinery, point
of proteins [41, 42, 56], proteins resident in the permeability transi- mutations and loss of heterozygosity in DNA microsatellites. In
tion pore complex [57], proteins that influence mitochondrial fusion addition, cell cycle deregulation coupled with a shortened cycle
and fission dynamics [58] and even transcription factors with the resulting in inadequate opportunity to repair acquired DNA muta-
capacity to translocate from the nucleus to mitochondria to encour- tions prior to cells entering into S phase from G1 phase [69 - 71].
age MOMP. The tumour-suppressor protein p53 is an example of
such transcription factors [59]. Inhibition of MOMP disabling the Reports to date indicate that cancer cells exhibit huge varieties
apoptotic system is vital for the development of solid tumours, and of metabolic change which are associated with alterations in the
also haematological cancers as evident in the switch from pre- mitochondrial structures, dynamics and function, and with tumour
neoplasia (e.g., low-grade myelodysplastic syndrome) in which growth and survival. Mitochondria regulate tumour growth through
cells spontaneously commit to apoptotic MOMP, to overt neoplasia modulation of the TCA cycle and oxidative phosphorylation, and
(such as acute myeloid leukaemia developing from a myelodysplas- also they are vital in regulating redox homeostasis in the cells, in-
tic syndrome) in which MOMP is hindered [48, 60, 61]. ducing them to either commit to or evade apoptotic cell death sig-
nals. The mitochondrion has been identified as key organelle to
The mechanistic link between the Warburg effect (aerobic gly- understanding the molecular basis of tumour proliferation and also
colysis) and MOMP resistance in cancer cells has not been fully rational design of chemotherapeutic agents [52].
elucidated; however the following hypothetical explanations have
been put forward to explain the concurrent occurrence of aerobic
glycolysis and MOMP inhibition in cancer:
204 Current Pharmaceutical Design, 2014, Vol. 20, No. 2 Ubah and Wallace

MITOCHONDRIA AS CANCER THERAPEUTIC TARGETS damine with the anthracycline, epirubicin, increased by 9% the
The underlying principle for targeting the mitochondria for response rate of patients with solid tumours in comparison with
therapeutic benefits lies in the knowledge that this organelle plays a epirubicin alone [89, 90]. In another phase II study designed to
vital role in the regulation of bioenergetics, ROS production and examine the efficacy of cisplatin, paclitaxel and lonidamine co-
apoptotic cell death. Preferentially targeting the energy metabolism administration in patients with advanced ovarian cancer, an 80 %
of tumour cells and not the healthy cells which also rely upon the overall response rate was reported, including 40% complete re-
essential pathway for ATP supply poses a major challenge. How- sponses and 40% partial responses [91].
ever, the cancer cell mitochondria are structurally and functionally Betulinic acid (BetA) is a plant-derived pentacyclic triterpe-
different from their healthy counterparts [72, 73]. Cancer cells dis- noid that exerts potent anti-cancer effects in vitro and in vivo with
play an extensive metabolic reprogramming that makes them more remarkable selectivity for tumour cells over non-transformed cells.
susceptible to mitochondrial perturbations than healthy cells [74]. Apoptosis induced by BetA has been shown to involve the activa-
In a broader perspective, cancer-specific mitochondrial alteration tion of caspases, PARP cleavage and DNA fragmentation. In addi-
and energy metabolism can be exploited for the development of two tion, the production of ROS and decrease in the mitochondrial
novel classes of anti-tumour agents. These novel agents will target membrane potential has been described, thus suggesting a reliance
glycolysis and/or reverse the Warburg phenomenon or aim at induc- on the mitochondrial cell death pathway [92 - 94]. BetA triggers
ing apoptosis via targeting mitochondrial proteins and membranes MOMP directly in association with mitochondrial membrane poten-
[48]. As stated earlier, mitochondrially-targeted agents emerged as tial dissipation and cyt c release when added to isolated mitochon-
a means of selectively targeting tumour cells as a result of these dria in a cell-free system [95]. This cytotoxicity induced by BetA
differences that exist between them and non-immortalised cells. We could not be blocked by the pan-caspase inhibitor zVAD.fmk [96].
discuss below a number of mitochondrial features which has been This would imply that the cytotoxic effects of BetA are mediated by
exploited or may be considered as therapeutic target in human can- the MPT which may be induced by ROS overproduction [3]. BetA
cer treatment. modulates the expression levels of Bcl-2 family proteins which
includes the upregulation of the pro-apoptotic mewmbers such as
i. Targeting Mitochondrial Permeability Transition and Ade- the BAX and BCL-X5 (also known as the BCL2L1). Moreover,
nine Nucleotide Translocase (ANT) BetA mediated apoptosis has been shown not to be associated with
The permeability transition pore complex (PTPC) is a highly p53 accumulation and occurred in a p53-indepndent fashion [97].
dynamic supra-molecular complex with a poorly understood struc- GSAO (4-[N-[S-glutathionylacetyl]amino] phenylarsenoxide),
tural identity. This is probably as a result of the existence of multi- a glutathione-coupled trivalent arsenical compound which is a hy-
ple isoforms of its constituents and also a number of distinct but drophilic derivative of the protein tyrosine phosphates inhibitor
functionally related solute carrier proteins that can substitute for phenylarsine oxide (PAO). This small synthetic mitochondrial poi-
each other within the PTPC. Prototypical PTPC is composed of the son targets angiogenic endothelial cells. The trivalent arsenical of
VDAC in the outer membrane, the adenine nucleotide translocase GSAO interacts with and disrupts ANT of the inner mitochondrial
(ANT) in the mitochondrial inner membrane and cyclophilin D membrane of endothelial cells thus causing growth arrest [98, 99].
(CYPD) in the mitochondrial matrix [13], however studies showing It has been shown to cross-link critical cysteine residues of ANT
that the mitochondrial permeability transition (MPT) occurred in (Cys160 and Cys257) resulting in the inhibition of the ATP/ADP
mouse liver mitochondria with ANT1 and ANT2 knock-out mouse antiporter activity, cytosolic ATP depletion, ROS overproduction,
suggests that both VDAC and ANT (but not CYPD) may not be mitochondrial depolarisation and apoptosis [100]. GSAO would
essential for the lethal functions of the PTPC [75 - 77]. Other stud- preferentially target proliferating cells due to their high mitochon-
ies have also demonstrated that CYPD was also not essential for the drial Ca2+ levels and elevated respiration which renders them more
permeability transition [78, 79], thus leading to the assessment of susceptible to PTPC opening than normal cells [100]. PENAO (4-
the MPT (and ANT) as being relevant for necrotic rather than apop- [N-[S-penicillaminylacetyl]amino] phenylarsonous acid) a second
totic cell death [80]. Despite these findings, the suggestion that the generation ANT inhibitor was designed to evade the pro-drug proc-
ANT isoforms may play a vital role in apoptosis is credible; ANT1 essing and metabolism of GSAO [101]. It is a cysteine mimetic of
and ANT3 are pro-apoptotic, whereas ANT2 (which is often over active metabolite of GSAO, CAO (4-[N-[S-cysteinylacetyl] amino]
expressed in proliferating cells) is anti-apoptotic [81 - 84]. Compo- phenylarsenoxide). PENAO accumulates in cells 85-fold faster than
nents of the PTPC can be targeted by several compounds to induce GSAO, resulting in a 44-fold enhanced anti-proliferative activity
MPT and apoptosis. By depleting the endogenous inhibitors of and a ~20-fold increased anti-tumour efficacy in mice. PENAO
PTPC opening such as glucose, phosphocreatine and glutathione, an targets both proliferating endothelial and tumour cells, unlike
indirect permeabilisation effect can be achieved. Similarly, agents GSAO [102]. A phase 1/IIa dose escalation study of PENAO in
that cause increase in cytosolic Ca2+ or stimulate ROS production patients with solid tumours resistant to standard chemotherapy is
can trigger MPT [85]. Although several ANT ligands have been currently recruiting.
shown to induce intrinsic cell death, they lack ANT isoform-
Retinoid-related compounds such as CD437 (6-[3-(-1-
specific targeting.
adamantyl)-4-hydroxyphenyl]-2-naphthalene carboxylic acid) and
Lonidamine, 1-(2,4-dichlorobenzyl)-1-H-indazole-3- all trans-retinoic acid are well-known for their ability to induce the
carboxylic acid is a putative ANT ligand that triggers the mito- expression of retinoic acid receptor-responsive genes resulting in
chondrial apoptotic pathway [86]. It is a glycolysis-targeting com- complete clinical remission in a high proportion of patients with
pound which has exited clinical trials and gained approval in acute promyelocytic leukaemia [103]. Interestingly, Belzacq et al.,
Europe for the treatment of solid tumours. Lonidamine acts as a and Notario et al reported that these retinoids (CD437 and the all-
hexokinase inhibitor with selectivity for the mitochondria-bound trans-retinoic acid, 9-cis-retinoic acid and 13-cis-retinoic acids)
form of hexokinase which seems to play a key role in enhancing also trigger ANT-dependent MPT and apoptosis independent from
glycolytic flux in both proliferating normal and transformed cells nuclear receptor binding [88, 104].
[87]. Lonidamine in combination with the peripheral benzodi-
azepine receptor (PBR) ligand diazepam showed a cytostatic effect ii. Agents that Induce ROS Overproduction and Targeting Mi-
on tumour growth in patients with recurrent glioblastoma multi- tochondrial Peroxiredoxin III
forme in a phase II clinical study. 50 % of patients exhibited disease Motexafin gadolinium (gadolinium texaphyrin) is an aromatic
stabilisation following treatment including one case in which pro- macrocylic compound that exhibits an elevated oxidising potential,
gression occurred after 12 months [86, 88]. The inclusion of loni- thus triggering excess production of ROS and interfering with the
Cancer Therapy Current Pharmaceutical Design, 2014, Vol. 20, No. 2 205

antioxidant machinery. Motexafin gadolinium has been shown to apoptotic and autophagic death is initiated by production of ROS
preferentially accumulate in malignant cells, probably due to their resulting from inhibiton of oxidative phosphorylation[118,119].
metabolic perturbations, and to improve in vivo sensitisation to PEITC in combination with metformin demonstrated a significant
radiation and chemotherapy of xenotransplanted tumours [105]. In a anti-proliferative and cytotoxic effect in ovarian cancer cells in
phase III study based on motexafin gadolinium and brain radiother- vitro. This novel combination was effective in cisplatin resistant
apy for the treatment of patients with lung cancer and brain metas- cell lines [120].
tasis reported a motexafin gadolinium-mediated delayed neurologi- 2-methoxyoestradiol is selectively cytotoxic to human leu-
cal progression. In addition, paediatric glioblastoma patients toler- kaemia cells but not normal lymphocytes. It inhibits angiogenesis
ated the same regimen [106], suggesting a further investigation of by reducing endothelial cell proliferation and causing apoptotic cell
this combination therapy is warranted. death. There is accumulation of superoxide via its ability to inhibit
Menadione (2-methyl-1,4-naphthoquinone) and thiol cross- SOD [121, 122]. 2-methoxyoestradiol has shown disease stabilisa-
linking agents such as diamide (diazenediacarboxylic acid bis tion and it is also well tolerated according to reports from several
5N,N- dimethylamide), bismaleimido-hexane (BMH) and dithio- phase I/II studies in patients with solid tumours or multiple mye-
dipyridine (DTDP) cause oxidation of a critical cysteine residue loma [123, 124]. ANT-224, an orally bioavailable second-
(Cys 56) of ANT and can bypass Bcl-2 mediated cytoprotection. generation copper-chelating tetrathiomolybdate analogue showed
Recombinant Bcl-2 fails to prevent thiol modification of ANT. similar effects as to the oestradiol analogue with potential antiangi-
Moreover, these thiol cross-linkers induce mitochondrial membrane ogenic and antineoplastic activities [125].
permeabilization and cell death irrespective of the expression level The synthesis of ATP requires large amounts of oxygen, which
of Bcl-2 [107]. This suggests that thiol crosslinking agents cause a usually leads to the generation of ROS. Cellular damage ensues if
covalent modification of ANT which supercedes the control by Bcl- these ROS are not detoxified by the antioxidant systems. Elevated
2 thus commits the cells to apoptosis. pools of mitochondrial ROS and the disturbance of peroxiredoxin
-lapachone (ARQ 501) which undergoes futile redox cycles (Prx) generation in malignant cells may cause oxidative stress and
catalysed by NAD(P)H:quinone oxidoreducatse 1, thereby inducing the induction of apoptosis. Peroxiredoxins are family of enzymes
the overproduction of ROS, poly(ADP-ribose) polymerase 1 that catalyze the reduction of H2O2 and hydroperoxides to water
(PARP1) hyperactivation and apoptosis. -lapachone is in clinical and alcohol respectively and there are six isoforms of the enzyme
trial as a monotherapy and in combination therapy with gemcitabine (Prx I - VI). The Prx system is a cellular defence system against
in the management of patients with cancer of the pancreas and also oxidative stress, and Prx III and V have been reported to be ele-
head and neck malignancy [108]. vated in the mitochondria of cancer cells [126, 127]. Also elevated
Accumulation of ROS can be achieved by inhibiting the anti- expression of Prx I has been reported in several human cancers
oxidant systems, thus suggesting an alternative approach. including non-small cell lung, oral, breast and liver cancer. Over-
Buthionine sulphoximine causes an accumulation of ROS by in- expression of Prx II has also been reported in breast cancer, meso-
hibiting the synthesis of reduced gluthatione (GSH) whereas thelioma, and head and neck cancers [128]. Elevated Prx II is asso-
imexon, an aziridine-containing small pro-oxidant molecule, de- ciated with the chemotherapeutic resistance of leukemia and stom-
pletes the GSH pool due to its thiol-binding activity [109, 110]. The ach cancers. Downregulation of Prx II sensitised head-and neck
combination therapy of buthionine sulphoximine and mephalan, an cancers to radiation and gastric carcinoma to cisplatin [128, 129].
alkylating agent, is being investigated in a phase I clinical study As mentioned above, recent studies have suggested that the overex-
involving patients with neuroblastoma and melanoma. Also, pression of Prx III and its electron donors might provide a primary
imexon alone and in combination with docetaxel demonstrated line of defence against hydrogen peroxide produced by the mito-
some efficacy in another phase I clinical study in patients with chondrial respiratory chain, thus agents targeting Prx III and the
breast, prostate and lung cancer [110, 111]. mitochondrion-specific electron suppliers Trx2, Trx reductase
(TrxR)2 and sulfiredoxin (Srx) may potentially be administered in
Elesclomol sodium (STA-4783) induces oxidative stress with a combination with various chemotherapeutic agents. Srx plays a
consequent increase in ROS in both transformed and healthy cells. vital role by reducing hyperoxidised Prx III via translocation into
Since tumour cells have elevated ROS levels compared to healthy mitochondria. Overexpression of the mitochondrion-targeted Srx
cells, STA-4783 will induce oxidative stress beyond baseline levels efficiently enhances the restoration of Prx III and leads to cellular
with a resultant elevated ROS levels overwhelming the tumour cell resistance to apoptotic cell death achieved by an enhanced elimina-
antioxidant systems. This may result in the induction of the apop- tion of mitochondrial H2O2 and decreased rates of m collapse
totic cell death pathway [112, 113]. STA-4783 alone and in combi- [130].
nation with paclitaxel showed promising data in phase I/II clinical
studies in patients with refractory solid tumours, however a recent iii. MOMP as a Target
phase III study in patients with melanoma has been discontinued on
As discussed earlier, MOMP can cause MPT to proceed, how-
safety grounds [114, 115].
ever MPT can also result from events originating from the outer
Mangafodipir is a superoxide dismutase (SOD) mimetic pos- membrane. Pro-apoptotic proteins such as BAX and BAK can me-
sessing both catalase and gluthatione reductase activities. In non- diate MOMP via their pore-forming activity [33, 41]. BAX is cyto-
transformed cells, Mangafodipir acts as an antioxidant whereas in solic in healthy cells whereas BAK is an integral OM protein. BAX
malignant cells it has been shown to elevate H2O2 pool and also translocates to the OM in response to pro-apoptotic triggers and
potentiate the anticancer activity of paclitaxel against xenotrans- BAK undergoes conformational modifications locally. These two
plants of colon cancer in mice [116]. Mangafodipir is in phase II processes consequently result in the assembly of homo-oligomers
trial in subjects with colon cancer. and/or hetero-oligomers that form protein-permeable conduits
Isothiocyanates such as -phenylethyl isothiocyanate through which toxic intermembrane space proteins are liberated
(PEITC), a consitutuent of edible cruciferous vegetables such as into the cytosol [33, 131]. Bcl-2 and Bcl-XL can cause the seques-
watercress, is a thiol modifier that interacts with redox regulatory tration of BAX and BAK thus displaying their strong cytoprotective
proteins [117]. PEITC does not only provide significant protection ability. Moreover, BAX can be directly activated or liberated from
against chemically induced oncogenesis in experimental rodents but inhibitory interactions with anti-apoptotic Bcl-2 family proteins by
also inhibits proliferation of human cancer cells via induction of cytoplasmic p53 [59]. The binding groove of BAX that mediates its
apoptosis and autophagy. Although the underlying mechanism is interaction with anti-apoptotic proteins has been known for many
not fully understood, studies demonstrate that PEITC-induced years, but the site for direct activation by pro-apoptotic BH3-only
206 Current Pharmaceutical Design, 2014, Vol. 20, No. 2 Ubah and Wallace

proteins of the Bcl-2 family has only recently been identified with clinical activity and it is being evaluated currently as a monother-
the help of nuclear magnetic resonance analysis. This represents a apy or incombination with temozolomide or topotecan in the
novel druggable target for therapeutic modulation of apoptosis treatement of patients with SCLC, leukaemia, chronic lymphoma or
[132]. TOM22 which is a core component of the OM protein trans- advanced B-cell malignancies [148]. Apogossypol (NSC736630) is
location pore has been reported as the mitochondrial receptor for a semi-synthetic derivative of gossypol with reported superior anti-
BAX [133], although a recent study outcome has suggested that tumour activity and reduced toxicity compared to the parent com-
BAX and BAK oligomerisation occur independent of TOM22 pound. Daily oral dosing studies showed that mice tolerated doses
[134]. Endophilin B1 is another BAX- interacting protein which has of the derivative 2 - to - 4 times higher than gossypol. Hepatotoxic-
been reported to mediate BAX-dependent MOMP [135]. ity and gastrointestinal toxicity represent the major adverse effects
MOMP can also be initiated via mitochondrial lipids destabili- of gossypol, with the derivative apogossypol showing far less toxic-
sation by pro-apoptotic stimuli, with a consequent formation of ity. Efficacy tested both in vitro and in vivo, apogossypol displayed
transient gaps in the outer membrane that permits leakage of inter- superior activity [149].
membrane space proteins. Inner membrane proteases such as prese- A-385358 is a small molecule with some selectivity for binding
nilin-associated rhomboid-like (PARL) also regulate the release of to Bcl-X L versus Bcl-2 [150]. It enters the cells efficiently and co-
cyt c from the mitochondria by cleaving the dynamin-related pro- localises with the mitochondrial membrane. As a monotherapy, A-
tein optic atrophy 1 (OPA1), in this way directing the remodelling 385358 shows a modest cytotoxicity towards tumour cell lines, and
of cristae independent of mitochondrial fusion [136, 137]. PARL- enhances the in vitro cytotoxic effects of other chemotherapeutic
deficient cells have been shown to display low levels of OPA1 and agents like paclitaxel, etoposide, cisplatin and doxorubicin in sev-
were more susceptible to mitochondrial apoptotic triggers [136]. eral tumour types. In A549, non-small-cell lung cancer, A-385358
Therefore inhibition of PARL seems a promising strategy to induce enhanced the activity of paclitaxel by about 25-fold, and this poten-
MOMP and commit transformed cells to apoptosis. tiation of paclitaxel by A-385358 is reproducible in vivo. Combina-
The susceptibility of cancer cells to undergo apoptotic cell tion of A-385358 with the maximally or half maximally tolerated
death has repeatedly been demonstrated to be determined by the dose of paclitaxel showed a significant growth inhibition in A-549
ratio of pro-apoptotic versus anti-apoptotic Bcl-2 proteins. Shifting xenograft model.more so, significant increase in mitotic arrest and
the balance of the “so-called” Bcl-2 rheostat in favour of the pro- consequent apoptosis have been reported for the combination ther-
apoptotic protein members, such as the BH3-only proteins gener- apy compared to paclitaxel monotherapy [150].
ates a powerful means to initiate MOMP-dependent apoptosis [3]. Obatoclax (GX15-070) is a small-molecule indole bipyrrole
Several inhibitors of Bcl-2 anti-apoptotic proteins have been identi- compound which is predicted to occupy a hydrophobic pocket
fied recently as being capable of inducing apoptosis in a variety of within the BH3 binding groove of Bcl-2 anti-apoptotic proteins thus
tumour cells, thus indicating their potential in cancer therapy. How- antagonising their actions with a consequent induction of apoptosis
ever, following the investigation of some of the putative Bcl-2 in- dependent on BAX and BAK [151, 152]. Unlike ABT-737, obato-
hibitors (obatoclax, gossypol, apogossypol, EM20-25, chelerythrine clax efficiently disrupts the direct interaction between BAK and
and ABT-737), it appears that only ABT-737 specifically targeted MCL-1 in intact mitochondrial outer membrane and also inhibits
Bcl-2 proteins and induced apoptosis via caspase-9 activation. the association between MCL-1 and BAK in intact cells thereby
These BH3 mimetics currently under preclinical and clinical testing overcoming the MCL-1 dependent resistance to ABT-737 and
have shown that they also interact with cellular targets that are un- bortezomib, a proteasome inhibitor and the gold standard treatment
related to Bcl-2 with the exception of ABT-737 and ABT-263, and for patients with multiple myeloma [151]. Obatoclax has demon-
this “unwanted” interaction might mitigate further development of strated a modest activity as a monotherapy in a phase I clinical
this class of drugs due to potential toxicity concerns [138, 139]. study involving patients with advanced chronic lymphocytic leu-
ABT-737, a small-molecule inhibitor of the anti-apoptotic pro- kaemia [153], however it is currently undergoing clinical evaluation
teins Bcl-2, Bcl-X L and Bcl-W, with affinity two to three orders of as a monotherapy and in combination regimen in phase I/II studies
magnitude more potent than the previously mentioned counterparts. for the treatment of haematological malignancies and solid tumours
Data obtained from mechanistic studies suggests that ABT-737 [154]. In another phase II clinical study using obatoclax for the
does not directly initiate the apoptotic cell death process; rather it treatment of myelofibrosis (MF) the molecule had no significant
enhances the effects of death signals [140]. It has shown a synergis- clinical activity in the MF patients at the dose and schedule evalu-
tic cytotoxicity profile with conventional chemotherapeutics and ated [155].
radiotherapy against haematological malignancies and solid tu- Oblimersen (G3139) is an antisense oligonucleotide compound
mours [141, 142]. ABT-737 exhibits single-agent-mechanism-based rationally designed to specifically bind to the first 6 codons of the
killing of small-cell lung cancer and lymphoma cell lines, as well as human bcl-2 mRNA sequence resulting in the degradation of the
in primary tumours and animal models. ABT-737 improves sur- bcl-2 mRNA and subsequent decrease in Bcl-2 protein translation
vival, regresses established tumours and is associated with high [156]. Oblimersen in combination with chemotherapeutic agents
cure rates of tumours in mice [140]. Furthermore, ABT-737 re- such as doxorubicin, docetaxel, cyclophosphamide and fludarabine
versed the chemoresistance of cancer cells against conventional has been clinically evaluated in various cancer types (157, 158]. In
chemotherapeutic agents, and ABT-737 resistance could be over- a phase III study involving patients with relapsed or refractory
come by using any of the classical cytotoxic agents [143, 144]. chronic lymphocytic leukaemia, there was a significant increase in
ABT-263, is an orally bioavailable derivative of ABT-737 with response rate and its duration with the addition of oblimersen to
improved clinical potential. It mimics the mode of action of ABT- fludarabine plus cyclophosphamide therapy [158]. In a phase II
737 and has shown promising preclinical outcome. It is currently in multicentre study, oblimersen in combination with rituximab
Phase I/II clinical trials for chronic lymphocytic leukaemia, lym- showed clinical benefit and safety in relapsed/refractory B-cell non-
phoma and small cell lung cancer, as monotherapy or in combina- Hodgkin lymphoma patients [159]. However, another phase III
tion with conventional chemotherapeutic agents, or with mono- clinical study in patients with multiple myeloma showed no signifi-
clonal antibodies [145]. cant differences in response rate between both groups treated with
either dexamethasone plus oblimersen or dexamethasone alone
Gossypol (AT-101) is a natural phenol derived from the cotton [160]. Despite extensive data supporting a critical role for Bcl-2 in
plant (genus Gossypium) [146] that simultaneously inhibits the chemoresistance in small cell lung cancer, addition of oblimersen to
anti-apoptotic Bcl-2 proteins, BCL-2, Bcl-XL, Bcl-W and Mcl1 a standard regimen for the tumour (carboplatin and etoposide) did
[138, 147]. In a phase I study against prostate cancer, it showed not enhance any clinical outcome measure [161]. It has been sug-
Cancer Therapy Current Pharmaceutical Design, 2014, Vol. 20, No. 2 207

gested that the reported lack of efficacy may be due to insufficient therapeutic strategies that preferentially target glycolysis and con-
suppression of Bcl-2 in vivo, however further evaluation of sequently kill tumour cells (see Fig. 1).
oblimersen in small cell lung cancer is not necessary. 3-bromopyruvate (3-BrPA), a lactate/pyruvate analogue is an
HA14-1 (2-amino-6-bromo-4-[1-cyano-2-ethoxy-2-oxoethyl)- alkylating agent and a potent inhibitor of glycolysis. Its anticancer
4H-chromene-3-carboxylate), a small organic molecule (molecular effect is mediated by its capacity to inhibit hexokinase thus causing
weight = 409) and nonpeptidic ligand of a bcl-2 surface pocket. In a depletion of ATP in cancer cells. This effect is most severe in
vitro binding studies demonstrated the interaction of HA14-1 with cells with mitochondrial DNA deletion and respiratory defects,
this bcl-2 surface pocket that is vital for bcl-2 biological function leading to massive apoptotic cell death [176, 177]. Inhibition of
[162]. HA14-1 enhances the sensitivity of cultured glioblastoma hexokinase also results in a rapid dephosphorylation of Bcl-2 asso-
cells to chemotherapy or radiotherapy [163]. It effectively and se- ciated death promoter homologue (BAD), a molecule known to be
lectively induces apoptosis in human acute myeloid leukaemia (HL- vital in both apoptosis and glycolysis. Dephosphorylation of BAD
60) cells overexpressing Bcl-2 anti-apoptotic proteins and its activ- at Ser-112 is associated with re-localisation of BAX to mitochon-
ity is not altered by the expression of multidrug-resistance pheno- dria, cyt c release and apoptotic cell death [177]. Ganapathy-
type [162, 164]. Kanniappan et al. (2010) have described that the primary mecha-
Polyamine analogues and conjugates, the naturally occurring nism of 3-BrPA is via preferential alkylation of GAPDH and that 3-
polyamines spermidine and spermine and their diamine precursor BrPA mediated cell death is linked to free radicals generation. Fur-
putrescine are vital for eukaryotic cell growth, differentiation and thermore, research from their laboratory also revealed that 3-BrPA
death [165]. Although the targeted inhibition of specific biosyn- induced endoplasmic reticulum stress, inhibited global protein syn-
thetic enzymes in the polyamine metabolic pathway has yet to show thesis which contributed to cancer cell death [178]. 3-BrPA has
significant clinical success in the treatment of cancer, there is a been shown to have antitumour effects when injected intraarterially
large body of evidence in favour of the rational for targeting the in the hepatic artery of rabbits with VX-2 tumours [178]. The glu-
polyamine function and metabolism in cancer [165 - 167]. Poly- cose transporter isoform 1 (GLUT1) gene encodes a key factor for
amine analogues and conjugates have been developed as a strategy glucose transport into cancerous tissue [179, 180]. Although the
to exploit the self-regulatory nature of polyamine metabolism. The expression and functional significance of GLUT1 in neuroblastoma
macrocyclic polyamine analogues, symmetrically substituted have not been fully described, 3-BrPA significantly suppressed the
bis(alkyl) and asymmetrically substituted polyamine analogues, and proliferation of neuroblastoma cells with high GLUT1 gene expres-
the novel oligoamine analogues have been associated with mito- sion compared to those with low expression [181], suggesting that
chondrial DNA as their target for cytotoxicity activity [168 - 170]. glycolysis inhibitors are a potential therapeutic option for treatment
Cyt c release from the mitochondria of treated cells also suggested of tumours expressing GLUT1. Combination of 3-BrPA with an
some involvement of the mitochondria in unleashing the apoptotic mTOR inhibitor demonstrated synergistic effects on leukemia and
cell death response to these compounds. Xie et al showed that an- lymphoma cells [182]. 3-BrPA may interact with other cellular
tharcenylmethyl homospermidine (ANTMHspd), a polyamine con- components owing to its alkylating properties suggesting that ex-
jugate induced mitochondrial membrane potential loss followed by tensive toxicity study needs to be carried out. The interaction be-
the release of cytochrome c and apoptotic cell death in promyelo- tween HK and VDAC offers another fascinating target to selec-
cytic leukemia (HL-60) cells [171]. This group further demon- tively commit cancer cells to apoptotic cell death. Disrupting this
strated that the combination therapy of ANTMHspd with alpha- interaction has been shown to preferentially kill tumour cells both
difluoromethylornithine (DFMO), an irreversible inhibitor of or- in vitro and in vivo via the promotion of PTPC and MPT [183, 184].
nithine decarboxylase enzyme (ODC) was synergistic in inducing 3-BrPA and Methyl Jasmonate, a plant hormone has been shown
the classical apoptosis in HL-60 cells via mitochondria/caspase- to disrupt this HK-VDAC interaction and selectively trigger cell
9/caspase-3 dependent pathway. Ant 4, a putrescine-anthracene death in cancer cells. It binds to HK thereby disrupting its associa-
conjugate, also demonstrated similar cytotoxicity profile in HL-60 tion with the mitochondria and apoptotic cell death is initiated
cell line. Ant 4 also induced oxidative stress in the cells; the latter [184]. Methyl jasmonate disrupts the HK-mitochondrial interaction
discovery provides a possible mechanism for the triggering of apop- at a concentration as high as 1 mM, thus it may serve as a lead
tosis via the mitochondrial pathway [172]. compound in the development of more potent and specific agents.
2-deoxy-D-glucose (2-DG) is a glucose analogue and has long
iv. Targeting the Warburg Effect been known to act as a competitive inhibitor of glucose metabolism
Elevated glycolytic rate and improved glucose uptake can pro- [185]. 2-DG is transported into cells and is phosphorylated by
vide several benefits to a proliferating tumour cell. Although oxida- hexokinase to 2-DG-P. However, unlike its analogue metabolite
tive phosphorylation generates more ATP per molecule of glucose, glucose-6-phosphate (G-6-P), 2-DG-P cannot be metabolised by
glycolysis can provide ATP at a higher rate provided glucose sup- phosphohexose isomerase which usually converts G-6-P to fruc-
ply is unlimited [173]. While Warburg believed that the preferential tose-6-phosphate. This leads to the accumulation of 2-DG-P in the
use of glycolysis even in the presence of oxygen by tumour cells cells with a consequent inhibition of glycolysis at the step of
was as a result of defects in their mitochondrial respiratory path- hexokinase mediated phosphorylation of glucose. Cellular ATP
ways , recent studies have suggested that tumour cells do contain depletion leading to cell cycle arrest and cell death occurs following
functional mitochondria and that the enhanced glycolytic flux con- the inhibition of the rate-limiting step involving hexokinase medi-
fer a growth advantage [45, 46]. ated glucose phosphorylation [54]. Nonetheless, the efficacy of 2-
In tumour cells much of the carbon entering the TCA cycle is DG is significantly affected by the presence of glucose and appears
extruded as citrate resulting in a truncated TCA cycle that is used to only partially reduce glucose availability for glycolysis. 2-DG
for lipid and fatty acids synthesis [174]. Glucose plays a key role in causes endoplasmic reticulum (ER) stress because of its ability to
maintaining mitochondrial integrity by promoting the association of affect protein glycosylation causing aberrant GlcNAcylation of
hexokinase II with the mitochondria, thus preventing the liberation proteins and induces accumulation of misfolded proteins in the ER
of cyt c [62], as well as regulating various cell death effectors such [186]. In vitro studies demonstrated that 2-DG exhibits cytotxic
as the pro-survival Mcl-1, pro-apoptotic BAD and Bax [139]. Lac- effect in cancer cells especially in those harbouring mitochondrial
tate, the by-product of glycolysis is believed to promote tumour defects or cells in hypoxic environment [187]. Inhibition of glyco-
invasion and metastasis via degradation of extracellular matrices lysis by 2-DG significantly increased the cytotoxicity of cisplatin in
[175]. The understanding of the role of glucose in sustaining tu- human head and neck cancers via enhancing oxidative stress [188].
mour cell proliferation formed the basis for the development of There has been concern that 2-DG might compromise the glycolytic
208 Current Pharmaceutical Design, 2014, Vol. 20, No. 2 Ubah and Wallace

metabolism of the brain and heart, however discussions centre on of a 28-KDa  and -subunits, and also a third sequence homolo-
whether the therapeutic window of the 2-DG will be broad enough gous -subunit [197]. The -subunit has different enzymatic fea-
to justify its clinical development and also to develop specific in- tures and seems not to interact with the - and -subunits to form
hibitors of some glucose transporter isoforms that are frequently the 11S regulatory complex [198].
upregulated in cancer. Data from a clinical study suggests that 2- Proteasomes are situated in the both the nucleus and cytoplasm
DG at doses up to 250 mg/kg seems safe for use in combination of eukaryotic cells, however their distribution varies with regards to
with radiotherapy in glioblastoma multiforme patients [189]. 2-DG cell or tissue type. It is now known that they undergo cell-cycle-
has been shown to stimulate Akt phosphorylation in vitro and an- specific redistribution between the nucleus and cytoplasm. In the
tagonise the anti-tumour action of a radio-immunotherapeutic inter- cytoplasm, proteasomes localise near centrosomes, on the outer
vention in vivo. These effects undermines the prospective therapeu- surface of the endoplasmic reticulum and in cytoskeletal network
tic value of 2-DG [190]. The pursuit of further clinical development [199].
of this compound has recently been halted according to Threshold
Pharmaceuticals [61]. Proteasomes regulate the levels of proteins such as cyclins, Bcl-
2, caspases, nuclear factor B (NF-B) that are crucial in cell-cycle
Dichloroacetate (DCA) is a small molecule inhibitor of pyru- progression and cell death [5, 6], and many of these proteasome
vate dehydrogenase kinase (PDK) which has the capacity to induce substrates are known mediators of deregulated systems in cancer.
a metabolic switch from aerobic glycolysis to glucose oxidation, There is evidence that proteasome affects cell-cycle progression via
thus decreasing mitochondrial hyperpolarisation and rendering its regulatory action on the cyclins. Also, it can cause an elevation
tumour cells more sensitive to apoptotic signals [191]. Pyruvate or attenuation in apoptotic events through its action on the caspases,
dehydrogenase is a mitochondrial enzyme which converts pyruvate Bcl-2, and NF-B [6]. Suffice to say that specific defects in protea-
to acetyl CoA, and its activity is negatively regulated by PDK thus some function or structure are yet to be linked to neoplasia although
DCA indirectly stimulates the pyruvate to acetyl CoA conversion. abnormalities of the aforementioned regulatory pathways are linked
DCA also upregulates the expression of the K+ channel Kv1.5, to tumourigenesis.
which is often underexpressed in tumour cells [191]. DCA has pre-
viously been utilised in the clinic for the management of lactic aci- ii. Targeting the Proteasome for Cancer Therapy (Table 1)
dosis and showed a promising safety profile even when used The dilemma associated with targeting the proteasome as a
chronically [192]. A phase I study for DCA in currently under way means of chemotherapeutic intervention is that this organelle plays
in Canada and is accepting patients with recurrent or metastatic a crucial role in the execution of many cellular functions, thus mak-
solid tumours [3, 61]. ing it difficult to target selectively and at the same time maintain a
ATP citrate lyase (ACL) is a key enzyme that associates glu- tolerable therapeutic window. However, studies have shown that
cose metabolism with lipid biosynthesis via catalysing the conver- many types of actively proliferating cancer cells display a higher
sion of citric acid to cytosolic acetyl-CoA has become a promising sensitivity to proteasome inhibition than the non-transformed cells
potential drug target in cancer treatment. Frequently malignant cells [6].
redirect pyruvate towards lipid synthesis which is a key component The proteasome inhibitors N-carbobenzoxy-L-leucyl-L-leucyl-
required to support the elevated demand for membrane generation norvalinal (LLnV) and N-carbobenzoxy-L-isoleucyl-L--t-butyl-
in highly proliferating cells [193]. Tumour growth was shown to be L-glutamyl-L-alanyl-L-leucinal (PSI) demonstrated a selective
suppressed in vitro following the inhibition of ACL by RNAi or p53-dependent apoptotic cell death induction in the proliferating
using a pharmacological inhibitor, SB-204990 [193]. It has been human leukemic HL60 cells compared with the quiescent HL-60
demonstrated that ACL is essential for the provision of adequate cells [200]. Also compared with actively proliferating primary en-
acetyl CoA required for histone acetylation and hence gene expres- dothelial cells, a 340-fold higher concentration of PSI was required
sion [194]. The implication regarding this recent discovery is that to induce apoptosis in contact inhibited quiescent cells [6]. Another
ACL inhibition might not only normalise tumour metabolism at the proteasome inhibitor (PI), Lactacystin, which is an irreversible
lipid synthesis level but it will also play a part in the epigenetic inhibitor of the catalytic -subunit of the proteasome induces apop-
reprogramming of malignant cells. totic cell death in human chronic lymphocytic leukaemia sensitive
PROTEASOMES or insensitive to radiation at doses that do not affect normal lym-
phocytes [201]. Apoptotic cell death was also observed in oral
i. Structure, Localisation and Function squamous-cell carcinoma cells, but not in normal oral epithelial
The functionally active 26S proteasome is a very large 2.4 MDa cells or gingival fibroblasts following treatment with LLnV and
ATP-dependent proteolytic complex that is situated in the cyto- Lactacystin [201]. It is fascinating that treatment with LLnV or
plasm and nucleus of eukaryotic cells. The 26S proteasome consists lactacystin was associated with an accumulation of KIP1 and re-
of a 20S core catalytic cylindrical complex capped at both ends by duced expression of the anti-apoptotic protein, Bcl-2 thus leading to
19S regulatory subunits [195]. Although ubiquitinylation is not a cell-cycle arrest and consequent apoptosis [6]. Several studies have
prerequisite for the degradation of every protein, the 26S protea- provided proof of principle that the proteasome is a valid target for
some identifies proteins and degrades proteins that have been anticancer therapy; however the available inhibitors lack specificity
marked for degradation for their ubiquitin ‘tag’ [6]. The 20S protea- [202]. This led to the design and development of boronic acid-
some is a complex of 28 protein subunits arranged into 4 stacked derived compounds that inhibited the proteasome pathway in a
rings to create a cylindrical structure. The -subunits made up of highly specific fashion. Most of these compounds developed by
seven polypeptides form the top and bottom rings of the 20S cylin- Adam et al showed activity across the 60 panel of cell lines from
drical structure. The enzyme active site of the proteasome complex the National Cancer Institute. Bortezomib was identified from these
is in the two inner rings which consist of seven -subunits that form compounds as the best candidate on the basis of its potency and
the central chamber [196]. Proteasome enzymatic function is per- cytotoxicity [203].
formed by three (1, 2 and 5) of the seven -subunits, while the Bortezomib (N-pyrazinecarbonyl-L-phenylalanine-L-
function of the other four subunits is unclear [6]. For the 20S pro- leucineboronic acid; previously known as PS-341 or MLN-341) a
teasome to function in vivo, it will require the association of a regu- boronic acid dipeptide was the first PI to enter clinical trials and
latory unit that partially determine the specificity in function. 19S was granted accelerated approval by the FDA in 2003 after demon-
regulatory complex is an example of such regulatory unit [6]. The strating remarkable single-agent responses in patients with relapsed
mammalian cells contain another regulatory complex that associates and refractory myeloma. It has since been approved for the treat-
with the 20S proteasome: the 11S regulator or PA28. It is made up ment of newly diagnosed multiple myeloma and also mantle cell
Cancer Therapy Current Pharmaceutical Design, 2014, Vol. 20, No. 2 209

Table 1. Proteins affected by the Ubiquitin-Proteasome Pathway inhibition

lymphoma [202]. It reversibly and rapidly inhibits the proteasome zomib when cells are exposed for short duration mimicking in vivo
pathway by directly binding to the 20S proteasome complex and single-dose exposure; however both compounds have superior ac-
blocking its enzymatic activity [203]. The underlying mechanism tivity when cells are exposed for prolonged periods of time [208,
leading to cell-cycle arrest and apoptotic cell death include NF-B 209]. To achieve a prolonged inhibition in vivo, PR-171 was admin-
inhibition, upregulation of various apoptotic pathways and effects istered daily and well tolerated at doses that inhibited over 80 % of
on tumour micro-environment. It is unlikely that proteasome inhibi- proteasome chymotrypsin-like-5 subunit (CT-L) activity in blood
tion with bortezomib will be observed in tumours resident in the and tissues. This observation contrasts that of bortezomib in which
brain or the testes unless the blood-brain or blood-testes barrier has a twice-weekly clinical-dosing schedule that allows full recovery of
been disrupted by the tumour. This was observed in tumour tissues proteasome function between doses was preferred because of ex-
isolated from brain and testes showing no proteasome inhibition cessive toxicity associated with more frequent dosing schedule
[203]. Preclinical studies demonstrated that the cytotoxic and anti- [210, 211]. Carfilzomib has also been investigated in combination
proliferating actions of bortezomib are correlated with the inhibi- with lenalidomide and low-dose dexamethasone in patients with
tion of proteasome in a p53 independent manner and also non- relapsed/refractory multiple myeloma [212]. The FOCUS trial
overlapping with other chemotherapeutics [203]. Tumour resistance (N=302) is comparing carfilzomibmonotherapy with best suppor-
to conventional anticancer agents can be overcome by the addition tive care in the relapsed/refractory stage. Moreover, other early-
of bortezomib as shown in two phase II multiple myeloma studies phase studies are investigating carfilzomib in frontline multiple
[204, 205]. There is strong evidence suggesting that the activity of myeloma in combination with either thalidomide or lenalidomide
bortezomib in solid tumours is significantly enhanced when used in and dexamethasone [213].
combination with chemotherapeutic agents such as doxorubicin, Marizomib (NPI-0052; salinosporamide A) is a structurally
gemcitabine, docetaxel, paclitaxel, and irinotecan [202, 206, 207]. and pharmacologically unique -lactone--lactam proteasome in-
Phase II clinical trials of bortezomib with gemcitabine, docetaxel hibitor. Its potent and sustained inhibition of all three proteolytic
and other conventional cytotoxic agents are on-going. activities of the proteasome has generated a renewed motivation
inspiring the extensive preclinical evaluation in multiple myeloma,
SECOND-GENERATION PROTEASOME INHIBITORS
mantle cell lymphoma, acute and chronic lymphocytic leukemia,
(PIs)
Waldenstrom’s macroglobulinemia, glioma, colorectal and pancre-
Following an improvement in our understanding of the ubiq- atic cancer models. It has shown efficacy as monotherapy, and in
uitin-proteasome pathway, the development of new PIs with distinct combination with chemotherapeutics, biologics and targeted thera-
efficacy and safety profiles was embarked upon. More so the sec- peutics [214]. Clinical trials in patients with multiple myeloma,
ond generation PIs provide potential options for patients with borte- lymphomas, leukemias and solid tumours, and also patients with
zomib resistant tumours. bortezomib treatment failure, as well as in patients with diagnoses
Carfilzomib (PR-171) has in vitro anti-proliferative effect where other PIs have been shown to be significantly efficacious is
across a range of tumour cell types including multiple myeloma underway [214 - 216].
cells resistant to bortezomib. PR-171 is more active than borte-
210 Current Pharmaceutical Design, 2014, Vol. 20, No. 2 Ubah and Wallace

MLN9708/MLN2238 is a selective, orally bioavailable second- ER. Nonetheless, the UPR will induce cell death programmes to
generation PI that is currently in a phase I clinical development eliminate stressed cells in situations where attempts to recover the
[217]. It has a shorter proteasome dissociation half-life with an cells from ER stress fails [226]. During carcinogenesis, there is a
enhanced PK-PD, and anticancer activity compared with borte- high demand for an increased activity of ER protein folding, as-
zomib. Treatment of multiple myeloma cells with MLN9708 pre- sembly, and transport to support the elevated proliferation rate, and
dominantly blocked CT-L activity of the proteasome and induced this condition can induce physiological ER stress. Moreover, there
accumulation of ubiquitinated proteins. Multiple myeloma cells is an increased nutrient starvation and hypoxia as tumour cells
resistant to bortezomib and other conventional therapies were grow, and these are strong inducers of the accumulation of unfolded
shown to respond to anti-proliferative and apoptotic effects of or misfolded proteins in the ER and the subsequent activation of the
MLN9708 and these activities are selective for tumour cells rather UPR pathways [227]. There is valid and accumulating evidence
than non-transformed cells [218]. Mechanistic studies showed that showing that the UPR is a key mechanism required for cancer cells
MLN9708-mediated apoptosis is linked with the activation of to maintain malignancy and resistance to therapy [226].
caspases-3, 8, and 9; elevated p53, p21, NOXA, PUMA, and E2F; Experimental evidence suggests that ER and Golgi apparatus
induction of endoplasmic reticulum stress response proteins Bip, can activate both pro-survival and apoptotic mechanisms if the
phosphor-eIF2-, and CHOP; inhibition of NF-B [218]. MLN9708 stress-signalling threshold is exceeded [4]. The Golgi apparatus is
in combination with lenalidomide, dexamethasone or histone deace- known to dissemble during apoptosis in a fashion resembling that
tylase inhibitor suberoylanilidehydroxamic acid triggers synergistic observed during mitosis [225]. It is plausible that the subtle balance
activity in the treatment of multiple myeloma [210, 218]. of protein trafficking between various subcellular compartments
ONX-0912, is an orally bioactive tripeptideepoxyketone with afford an extremely sensitive damage sensor. It is worth mentioning
anti-proliferative effects and also induces apoptosis multiple mye- the interaction between the mitochondrion with the Bcl-2 family
loma cells resistant to conventional and bortezomib therapies. Its proteins regulating various cell death signals and the ER. Thus,
anti-myeloma activity is associated with activation of caspases-3, 8, overexpression of Bax has recently been shown to induce Ca2+ mo-
and 9, and poly(ADP) ribose polymerase, also the suppression of bilisation from ER stores [228, 229]. There is substantial evidence
multiple myeloma cells migration [219]. In animal tumour models, supporting the Bcl-2 co-localisation with the ER and protects cells
ONX-0912 significantly reduced tumour progression and improved from thapsigargin-mediated apoptosis [225]. More so, evidence is
survival, and it improved the anti-tumour activity of bortezomib, mounting that signals originating from the ER-Golgi network may
lenalidomide, histone deacetylase inhibitors or dexamethasone in bypass the mitochondrion and efficiently execute apoptotic cell
multiple myeloma [210, 219]. A phase I study with ONX-0912 in death [4, 225].
patients with solid tumours is on-going, and an interim results from
the study showed that at doses that were tolerated, more that 80% Targeting the UPR/ER Stress Components
proteasome inhibition in blood could be achieved on a once-daily The role of UPR in tumour maintenance has stimulated great
dosing regimen for 5 consecutive days [220]. interest in exploring therapeutic potential by targeting the UPR
components. The understanding that healthy cells are not subjected
ENDOPLASMIC RETICULUM (ER) - GOLGI NETWORK to stress and the UPR pathway remains in an inactive state while
Interestingly a growing number of reports recognise proteins in that of transformed cells rely on activated UPR for survival creates
control of ER and Golgi homoeostasis as potential therapeutic tar- a discrepancy between tumour cells and their healthy counterpart
gets in the treatment of malignancies. Eukaryotic cells consist of a and thus offer an advantage for the agents targeting these compo-
set of organelles with a tightly and dynamically regulated abun- nents to achieve specificity (Table 2).
dance with regards to cellular demand. These organelles are
equipped with specific cellular functions. The ER is an organelle in PROTEASOME INHIBITORS
which secretory and membrane proteins are synthesised, and cor- The degradation of the majority of cellular proteins is mediated
rectly folded proteins by the ER chaperones are transported to the by the 26S proteasome. The role of proteasome in regulating cell
Golgi apparatus [221]. The ER retains the unfolded or misfolded growth, survival and metastasis of malignant cells is well reported
proteins and they are subsequently degraded by the ER-associated leading to the development of PIs (e.g., Bortezomib; see previous
degradation (ERAD) [222, 223]. In cases of accumulation of un- section for discussion on PIs). It is believed that PIs cause a build-
folded proteins in the ER, an upregulated expression of the ER up of misfolded proteins which overwhelms the ERAD pathway
chaperone and components of ER-associated degradation machin- resulting in ER stress [230].
ery occurs in an effort to enhance the protein folding capacity and Multiple myeloma cells constitutively express ER stress sur-
degradation of unfolded or misfolded proteins. This process occurs vival factors in order to function as secretory cells, and it is also
through the cytoprotective mechanism referred to as the ER stress assumed that the UPR components are required for B-cell differen-
response or unfolded protein response [224]. Large amounts of tiation into antibody-secreting cells [231]. Thus agents that interfere
secretory proteins are transported to the Golgi apparatus when the with the UPR and ER-stress induced apoptosis will show greater
ER stress response enhances the capacity of ER function, thus caus- effect in myeloma cells and other secretory cells as a result of the
ing inadequacy of Golgi function. This is referred to as Golgi stress, reliance of these cells on the UPR pathway. As discussed in the
which initiates the Golgi stress response to cope with the situation previous section, multiple myeloma cells are very sensitive to the PI
[221]. To ensure the fidelity of protein folding and to sustain the bortezomib which rapidly induces components of the pro-apoptotic
accumulation of unfolded or misfolded proteins, the ER has devel- /terminal UPR, including PERK ultimately resulting in increased
oped a group of signal transduction pathways, the unfolded protein cell death [230]. Since multiple myeloma cells are dependent on an
response (UPR) signalling pathways to alter transcriptional and activated UPR for survival suggesting that these cells have a lower
translational programs [225]. The basic mammalian cell UPR path- threshold for ER stress. Thus, any additional stress can slant the
ways is composed of three major signalling pathways initiated by balance in favour of cell death.
three primary ER-localised protein stress sensors; IRE1 (inositol-
requiring enzyme 1 alpha), PERK (double-strand RNA-activated HSP90 INHIBITORS
protein kinase-like ER kinase), and ATF6 (activating transcription
Heat shock protein 90 (HSP90) is involved in regulating the
factor 6). The primary function of the UPR is to prevent the cell
proper folding, stability and function of a wide variety of signal-
from ER stress by limiting the amount of proteins translocated into
transducing proteins that regulate cell proliferation and differentia-
the ER lumen, increasing retrotranslocation and degradation of ER-
tion. These proteins include steroid hormone receptors and protein
localised proteins, and enhancing the protein-folding capacity of the
Cancer Therapy Current Pharmaceutical Design, 2014, Vol. 20, No. 2 211

Table 2. A representative list of ER stress-aggravating agents

kinases and most notably are Akt, Bcr-Abl, Apaf-1, surviving and
INHIBITORS OF PRO-SURVIVAL MODULE, GPR78
cyclin dependent kinases with established roles in tumorigenesis
[230]. A number of studies have presented strategies to incapacitate
GPR78 function in vitro and in vivo, although few have made it to
17-Allylamino-17-demethoxygeldanamycin (17-AAG) and 17
clinical testing. There are several naturally occurring compounds
(Dimethylaminoethylamino)-17-demethoxygeldanamycin (17-
that interfere with GPR78 expression or activity at pharmacological
DMAG) are analogues of geldanamycin, they are currently in clini-
concentrations, for example salicylic acid from plants [236]; the
cal trials. 17-DMAG was found to activate all three components of
polyphenolic green tea component a (-) -epigallocatechin-3-gallate
the UPR in myeloma plasma cells leading to the suggestion that
(EGCG) [237]; genistein, an active ingredient of soy [238]; the
HSP90 inhibitors induce apoptotic cell death at least in part via the
microbial metabolites verispelostatin [239], prunustatin A [240],
ER stress and the UPR death pathway [230, 232].
verrucosidin [241], piericidin A [242], deoxyverrucosidin [243];
HIV PROTEASE INHIBITORS synthetic members of the biguanide class of compounds (met-
formin, phenformin and buformin) [244]; the cyanine dye pyrv-
Nelfinavir, is an HIV protease inhibitor which has recently inium [245]; and the plant product arctigenin [246]. Since GPR78
been shown to induce ER stress, autophagy and apoptotic cell death has been located outside the ER lumen; it has been identified in the
in vitro and in vivo in non-small cell lung cancer [233]. Ata- cytosol, mitochondria, nucleus and tumour cells surface distinctly
zanavir, another HIV protease inhibitor alongside Nelfinavir suggesting that this protein exerts functions beyond its ER trans-
showed in a separate report to trigger ER stress and cause cell death membrane signalling function [244, 247]. This discovery has lim-
in various malignant glioma cell lines in vitro due to their ability to ited the progress of some of the GPR78 inhibitors since they seem
induce UPR [234]. As outlined earlier, PIs appear to be more effec- to have a non-specific GPR78 inhibition.
tive in secretory cells however solid tumours frequently display
reduced sensitivity to bortezomib. Kraus et al considered utilising a Versipelostatin (VST) is the first compound reported to spe-
dual therapeutic approach to achieve a more effective tumour kill- cifically inhibit GPR78 expression with a unique selective cytotox-
ing using agents that will enhance ER protein load and simultane- icity in glucose-deprived tumor cells by preventing the UPR [239].
ously blocking the proteasome degradation pathway using borte- Under stress conditions, it causes a robust cell death by inhibiting
zomib [235]. In this setting, another HIV protease inhibitor Rito- GPR78 and GPR94 induction and repression of ATF4 and XBP1,
navir was used to interfere with the ERAD machinery with a con- which are the UPR transcription activators [230, 239]. In glucose-
sequent decrease in the degradation of newly synthesised proteins deprivation conditions, VST exerts actions similar to those of ra-
thus inducing the UPR in sarcoma cells previously resistant to pamycin, thus suggesting that it may have dual activity as antican-
bortezomib. This combination of ritonavir and bortezomib resulted cer agent, inhibiting the UPR and modulating the mTOR signalling
in the activation of PERK, IRE1, and ATF. This dual approach pathway. The translational control during UPR is complex, thus
showed synergistic actions in the induction of JNK, caspases - 4 further studies on the mechanism of action of VST is required to
and 9, and apoptotic cell death greater than 90 % [230, 235]. elucidate this translational events.
212 Current Pharmaceutical Design, 2014, Vol. 20, No. 2 Ubah and Wallace

Other Therapeutic Agents that Trigger ER Stress of ER stress markers in vitro and also in vivo xenograft tumour
Brefeldin A (BFA), a fungal macrolytic lactone with a wide models [257]. DMC is yet to be tested in the clinic. OSU-03012 is a
range of antibiotic activity show anti-tumor activity by disrupting product of an extensive structural adaptation of the celecoxib mole-
the vesicular coating process involved in intracellular protein trans- cule which also shows no inhibitory activity on COX-2, and has
port thereby inhibiting intracellular transport of proteins from the demonstrated promising preclinical antitumour outcomes as mono-
ER to the Golgi thus causing disintegration of the Golgi complex. therapy and in combination with other conventional chemothera-
BFA has been shown to induce p53-independent apoptosis in di- peutics such as imatinib, tamoxifen, trastuzumab and many others
verse human cancer cell lines [230, 248, 249]. BFA acts by inhibit- [259 - 261]. A phase 1 clinical study of OSU-03012 (as AR-12) in
ing directly the ADP-ribosylation factor (ARF) activity which is patients with advanced or recurrent solid tumours or lymphoma is
essential for coatamer assembly onto Golgi membranes and results in progress.
in the activation of the UPR [230]. The critical role of ER stress in Chloroquine, the classical antimalarial agent is now causing
mediating BFA cytotoxicity was demonstrated using the flavones excitement in cancer research field because of its capacity to block
baicalein which prevented BFA-induced ER stress with a resultant autophagy. Since autophagy blockade is associated with aggrava-
significantly reduced BFA cytotoxicity [250]. Breflate, a water- tion of ER stress, it was not completely unexpected that chloroquine
soluble pro-drug analogue of BFA was developed as a parenterally may induce ERSA activity [262]. In a clinical trial in Mexico,
administered agent; unfortunately this agent did not progress to chloroquine when used as an adjuvant sensitised glioblastoma mul-
clinical trials [244]. tiforme to temozolamide [263]. Temozolomide is the standard che-
Thapsigargin, is a sesquiterpene lactone of natural origin that motherapeutic care for patients with glioblastoma multiforme, how-
potently inhibits sarcoplasmic/endoplasmic reticulum calcium AT- ever it was shown to activate autophagy as a defense mechanism of
Pase (SERCA). The inhibition of SERCA resulted in significant the tumour cells [264]. The greatest benefit from chloroquine in
calcium leakage from the ER store into the cytosol causing a pow- cancer therapy has been linked to its ability to chemosensitise and
erful ER stress induction. The clinical use of this agent was never radiosensitise tumor cells [265, 266].Clinical trials are underway in
achieved because of poor tolerance in experimental animals. Be- the U.S. to evaluate the anticancer potentials of chloroquine in
sides its potent anti-tumour activity, it also caused histamine release breast (NCT01023477) and lung (NCT00969306) cancer patients
and stimulates arachidonic acid metabolism [244]. Overall, these [244].
features were perceived as shortcomings in the context of systemic
LYSOSOMES AND LYSOSOME-RELATED ORGANELLES
therapy.
Lysosomes were first described in 1955 by De Duve et al as
To circumvent systemic toxicity, a pro-drug of thapsigargin
organelles enriched with acidic hydrolases and potentially harmful
with tumour-targeting potential was developed. It is a chemically
to cells [267]. They were viewed as ‘waste-disposal’ compartment
modified thapsigargin coupled to a peptide carrier that is a substrate
of the cell, however this view have been challenged following the
for prostate-specific antigen (PSA) protease. The protease acts on
discovery of other related organelles and the finding that lysosomes
the pro-drug to liberate the active agent within metastatic prostate
are dynamic organelles that move along the cytoskeleton and asso-
cancer sites [251]. G202, a related pro-drug construct whose anti-
ciate with other cellular compartments. There are well documented
tumour activity is activated by carboxypeptidase prostate-specific
evidence showing that the acid vacuolar compartment in all eukary-
membrane antigen (PSMA) showed a significant anti-tumour activ-
otic cells participate in a wide range of cellular processes (Fig. See
ity against a panel of human cancer xenografts in vivo with minimal
Castino et al., 2003, Destination “Lysosomes”) such as partial or
host toxicity [252]. A phase 1 dose escalation clinical study with
extensive degradation of various substrates, trafficking and recy-
G202 has commenced in patients with advanced tumours [244,
cling of molecules among internal organelles to and from the exte-
253].
rior of the cell, post-translational maturation of secretory products
Celecoxib, is a selective cyclooxygenase 2 (COX-2) inhibitor and storage of undigested material [268]. For the purpose of this
and a non-steroidal anti-inflammatory drug (NSAID) developed and review, we will focus on the roles of lysosomes that are more rele-
originally approved for management of acute pain and inflamma- vant in tumorigenesis, although the lysosomes or lysosome-related
tion, and later as an adjunct for the management of familial adeno- organelles have other functions such as the enlargement of the
matous polyposis (FAP) [254]. In the last decade, novel pharmacol- membrane surface and resealing of plasma membrane following
ogical activity and targets most likely independent of its COX-2 calcium-dependent exocytosis [269, 270]. Lysosomes participate in
inhibition emerged. They include ER stress aggravation and car- various events associated with neoplastic phenotype, namely the
bonic anhydrases inhibition [255, 256]. Its ability to induce ER loss of cell mass homeostasis, loss of function of ‘programmed cell
stress is associated with its inhibition of SERCA function causing death’, acquisition of chemo-resistance and evading immune sur-
calcium efflux from the ER [257]. Generally, the clinical therapeu- veillance system, and the acquisition of invasive and metastatic
tic outcome for celecoxib in advanced cancer management has been potentials.
unimpressive. It has been suggested that the poor outcome observed
Lysosomes being the organelles with the highest concentration
may be due to the fact that celecoxib is optimised towards COX-2
of proteases and other hydrolytic enzymes inside the cell are largely
inhibition rather than to its subsequently recognised ESRA activity,
involved in the maintenance of cellular homeostasis by realizing the
thus celecoxib might not accomplish adequately high levels of ER
degradation of autologous material. In malignancy, lysosome-
stress in tumour cells [244]. Although this current assumption re-
mediated protein degradation is compromised [268, 271]. Both
mains unresolved, efforts are geared towards developing celecoxib
heterophagic and autophagic cargos find their final destiny in
analogues with optimised ERSA activity and minimised COX-2
lysosomes, where they are acted upon by many hydrolytic enzymes.
inhibitory activity.
Amongst the most studied lysosomal hydrolases are the proteases of
OSU-03012 and 2, 5 - dimethyl - celecoxib (DMC) are ana- the cathepsin family. Cathepsins are optimally active in acidic pH
logues of celecoxib with no COX-2 inhibitory activity and en- of lysosomes (pH 4-5), although they can function with reduced
hanced ERSA potency [258]. DMC contains a 2,5-dimethyl sub- efficacy in neutral pH outside the lysosomes [272]. Besides their
stituent at its terminal phenyl ring compared with celecoxib with a function in protein turnover, certain cathepsins participate in the
4-methyl moiety. This chemical modification resulted in the loss of control of cell cycle, antigen presentation, hair follicle morpho-
COX-2 inhibitory function with a simultaneous anti-tumour activity genesis and epidermal homeostasis [273]. Emerging evidence sug-
enhancement. DMC is a stronger ER stress inducer than its parent gests that cathepsins degrade the extracellular matrix when secreted
compound. It inhibits SERCA activity with a consequent induction to the extracellular space and also execute apoptosis when released
Cancer Therapy Current Pharmaceutical Design, 2014, Vol. 20, No. 2 213

to the cytosol [272]. The most implicated cathepsins in cancer are small molecule drugs and macromolecules requires transporters,
the cysteine cathepsins B and L, and the aspartate cathepsin D, and thus shifting the burden of selectively targeting the mitochondria of
their elevated expression is a reliable diagnostic marker of poor cancer cells to the transporters. However the major obstacle facing
disease outcome [274]. The altered lysosomal function found in this approach is the rapid elimination of liposomes and nanoparti-
murine fibroblasts and breast epithelial cells transformed with ras cles from systemic circulation due to the elevated clearance rate by
oncogene was an influence from the oncogene suggesting that its the reticuloendothelial system (RES) and other organs. This limits
activation may play a role in the alteration of lysosomal functions the use of intravenously administered liposomes and nanoparticles
[275]. Together with matrix metalloproteases and the plasminogens in cancer therapy [287], although conjugation to polyethyleneglycol
activator system, cathepsins secreted into the cytosol have been (PEG) chain enhances half-life and affords longer-lasting particles
suggested to participate in the degradation of extracellular matrix, in the systemic circulation [288, 289]. Carrier-size reduction to size
thus empowering cellular motility, invasion and angiogenesis [276]. below 200 nm allows for efficient accumulation in tumour tissue
This hypothesis has been recently supported by reports from two in due to permeability and retention effect enhancement [286]. We
vivo mouse model of pancreatic cancer and glioblastoma where discuss below some of the cancer therapies based on liposome and
pharmacological inhibition of cysteine cathepsin activity impaired nanoparticle delivery to mitochondria of malignant cells. Mito-
angiogenic switching in progenitor lesions and repressed tumour chondriotropic liposomes made of positively charged amphiphilic
growth, vascularity and invasiveness [277, 278]. molecules have been produced. One member which have been ex-
Parallel to tumour progression, cathepsins B, L, and D have tensively studied is the DQAsomes formed by dequalinium, a
been implicated as mediators of the lysosomal cell death pathway, symmetrical molecule in which a relatively long aliphatic chain
and death stimuli such as death-receptor activation, p53 activation, seperates two charged moieties. DQAsomes was investigated for
oxidative stress, growth factor deprivation and microtubule- mitochondrial delivery of mitochondrial leader sequence-DNA
stabilising agents can as induce partial lysosomal membrane per- conjugates and paclitaxel, and it showed positive outcome in initial
meabilisation and the release of cathepsins into the cytosol [8]. experiments studying the growth of a human colon tumour in nude
From the cancer therapy point of view, it was exciting to note that mice [290, 291]. However systemic toxicity associated with de-
cathepsins are able to mediate caspase- and mitochondrion- qualinium poses a major challenge. Triphenylphosphonium head
independent apoptotic cell death, allowing cell death to ensue even carried on fatty acid chains have been used to bequeath liposomes
in malignant cells with multiple defects in their apoptosis pathway. with mitochondriotropic feature, and enhance internalisation by
It is believed that they activate the classical apoptosis pathway by cultured cells. Rh123 and ceramide have been successfully deliv-
cleaving the proapoptotic Bcl-2 family proteins [8, 279]. ered into the mitochondria using this system, and also the latter
showed growth inhibitory efficacy in an in vivo murine model of
Targeting Lysosomes as Cancer Therapy mammary carcinoma [292, 293].
For tumourigenesis to be successful, an effective suppression of The novel Mito-Porter; a nano-carrier system for delivery of
both the classical apoptosis and lysosomal death pathways must macromolecules into mitochondria via membrane fusion was de-
occur [8]. Many malignant cells display characteristics of increased veloped by the Harashima group. The “Mito-Porter”, a DOPE (1,2-
activity of phosphatidylinositol-3'-kinase (PI3K) and they contrib- dioleoylsn-glycero-3-phosphatidyl ethanolamine; found to be opti-
ute to the stability of tumour cell lysosomes [280]. PI3K inhibition mal for membrane fusion) liposome carrying a high density of fu-
in certain settings tends to shift the TNF-induced death pathway sogenic R8 (octaArg) or similar surface peptide was developed as a
from caspase dependent to cathepsin dependent [281]. The translo- variation of the “multifunctional envelope-type nanodevices”
cation of heat shock protein 70 (HSP 70) into the lysosomal mem- (MEND) [294, 295]. Oligo-Arg peptides are known to act in TAT-
brane confers protection on tumour cells against lysosomal leakage. like fashion enhancing membrane fusion as well as exhibiting mito-
It has been shown that cells with HSP 70 on their lysosomal mem- chondrotropic functions. In cultured cells, the Mito-porter appeared
branes contain larger and more stable lysosomes and the depletion to be internalised via macropinocytosis and the resulting vesicles
of these HSP 70 triggers a tumour cell-specific lysosomal death deliver their cargo to the mitochondrial inter-membrane space
programme [282]. Thus, agents that inhibit the PI3K pathway or the [296].
lysosomal localisation of HSP 70 have great potential in cancer
therapy as tumour sensitisers to conventional chemotherapeutics by SUMMARY
facilitating lysosomal membrane permeabilisation. In addition, The successful activation of the cell death machinery of cancer
antiblastic agents such as the C1311, a topoisomerase II-inhibitor cells cannot be overemphasised. Cancer cells exhibit a significant
[268]; the synthetic retinoid CD437 [283]; and the -tocopheryl number of metabolic aberrations associated with the mitochondria
succinate [284] have been shown to induce apoptotic cell death via and other sub-cellular organelles. These aberrations promote tu-
the induction of primary lysosomal leakage. One ultimate goal in mour growth and survival, and at the same time exhibit unique
cancer medicine research is geared towards identifying a carrier that properties which enhances their vulnerability to certain anticancer
would allow site-specific and delivery of cytotoxic cargo into tu- agents. By these unique features cancer cells have been successfully
mour tissue. Over-expression of cell surface molecules in cancer targeted selectively. All the organelles discussed display some sort
cells is a regular occurrence and these over-expressed molecules of deregulation in cancer cells as compared with their counterparts
can be exploited as Trojan Horses to internalise anti-cancer agents. in healthy cells, this being one of the reasons for the recorded suc-
For instance, the peroxidised low-density lipoproteins (LDL) can cess in drug development and cancer therapy (Tables 1-3).
be internalised via LDL receptors and consequently accumulate Although significant progress has been made towards elucidat-
within the lysosomes, where they induce lysosomal destabilisation, ing the mechanism and role of these alterations in oncogenesis and
acid hydrolases leakage into the cytosol with an attendant cell death resistance to chemotherapy, a better understanding of the key
pathway induction [268, 285]. pathophysiological differences between organelles in normal and
cancer cells will in no doubt complement effort towards improving
Liposomes and Nanocarriers
selectivity of targeted anticancer agents. For instance, a conclusive
Using mitochondrially targeted carriers such as liposomes or elucidation of the association involving the Warburg effect and
nanoparticles, active molecules can be selectively delivered to the MOMP resistance will be clinically important in reversing related
mitochondria instead of directing the molecules themselves to mi- chemoresistance.
tochondria. This research area of cancer-selective nanoparticles and
Most of the agents discussed in this review, e.g. ABT-737 and
carriers is currently highly active [286]. Cellular internalisation of
its orally bioavailable derivative, ABT-263 possess single-agent
214 Current Pharmaceutical Design, 2014, Vol. 20, No. 2 Ubah and Wallace

Table 3. Mechanism of action and targets of some compounds with anticancer activity

mechanism based cytotoxicity as well as capapcity to reverse An efficient drug delivery system is as crucial as the drug
chemoresistance in cancer cells. Interestingly, their cytotoxicity is molecule itself. A balanced effort between research in anticancer
enhanced in combination with other chemotherapeutic agents or drug discovery and drug delivery systems such as the liposomes,
monoclonal antibodies. Other compounds with enhanced cytotoxic- nanoparticles and conjugates is likely to be the most successful.
ity resulting from combination therapy were lonidamine (with PBR,
anthracycline, epirubicin);  - lapachone (with gemcitabine); Borte- CONFLICT OF INTEREST
zomib (with doxorubicin, paclitaxel, gemcitabine). The use of “in- The authors confirm that this article content has no conflicts of
hibitory” analogues and their conjugates such as the ANTMHspd interest.
and Ant-4 in the form of “Trojan horses” has shown some promise
as monotherapies and in combination with DFMO. ACKNOWLEDGEMENTS
Declared none.
Cancer Therapy Current Pharmaceutical Design, 2014, Vol. 20, No. 2 215

Appendix

Fig. (1). Metabolic alterations and Warburg phenotype in tumour cells and related therapeutic targets. Cancer cells rely mostly on glycolysis through
enhanced expression and activities of glucose transporters and HK. In healthy cells, glucose is converted to pyruvate through series of reactions in the cytosol.
In the mitochondria, Pyruvate is converted to Acetyl-CoA which is a key component of the TCA cycle, and drives the synthesis of ATP via oxidative phos-
phorylation. During malignancy, pyruvate is preferentially converted to lactate which is transported out of the cell. Thus, pyruvate which enters the mitochon-
dria drives the truncated TCA cycle in which citrate is expelled from the mitochondria to stimulate lipid biosynthesis. Lonidamine, 3-BrPA, 2-DG, methyl
jasmonate, DCA, Arsenic, SB-204990 and  - cyano-4-hydroxycinnamate are listed as therapeutic agents while GAPDH, HK, PDK, ACL, and monocarboxy-
late transporters are listed as therapeutic targets.

GPR78 = G-protein coupled receptor 78


ABBREVIATIONS
HIF = Hypoxia inducible transcription factor
ACL = ATP citrate lyase
HIV = Human Immunodeficiency virus
AIF = Apoptotic inducing factor
HK = Hexokinase
ANT = Adenine nucleotide translocator
HSP = Heat shock protein
Ant 4 = Putrescine-anthracene conjugate
I - KB = Nuclear factor kappa B inhibitor
ANTMHspd = anthracenylmethyl homospermidine
IAP = Inhibitor of apoptosis protein
ARF = ADP-ribosylation factor
IM = Inner Mitochondrial membrane
ATF6 = Activating transcription factor 6
IMS = Intermembrane space
BAD = Bcl-2 associated death promoter homologue
IRE1 = Inositol – requiring enzyme 1 alpha
BAK = Bcl-2 antagonist/killer
JNK = c-Jun N-terminal kinase
BAX = Bcl-2 associated X protein
KIP1 = Kinase interacting protein 1
BH3 = Bcl-2 homology domain 3
LLnV = N-carbobenzoxy-L-leucyl-L-leucyl-
BID = BH3 – interacting domain death agonist
norvalinal
CYPD = Cyclophilin D
MEND = Multifunctional envelope-type nanodevices
Cyt c = Cytochrome C
MM = Multiple myeloma
DFMO = Difluoromethyl ornithine
MOMP = Mitochondrial outer membrane permeabilisa-
ER = Endoplasmic reticulum tion
ERSA = Endoplasmic reticulum stress aggravator MPT = Mitochondrial permeability transition
FAP = Familial adenomatous polyposis mtDNA = mitochondrial DNA
GAPDH = glyceraldehydes -3 – phosphate dehydro- mTOR = serine/threonine kinase target of rapamycin
genase
NF - KB = Nuclear factor kappa B
GLUT1 = Glucose transporter isoform 1
216 Current Pharmaceutical Design, 2014, Vol. 20, No. 2 Ubah and Wallace

ODC = Ornithine decarboxylase [20] Zoratti M, Szabo I. The mitochondrial permeability transition.
Biochim Biophys Acta 1995; 1241: 139-76.
OM = Outer mitochondrial membrane [21] Boveris A, Chance B. The mitochondrial generation of hydrogen
PARP = Poly ADP ribose polymerase peroxide. General properties and effect of hyperbaric oxygen. Bio-
PBR = Peripheral benzodiazepine receptor chem J 1973; 156: 435-44.
[22] Boveris A, Oshino N, Chance B. The cellular production of hydro-
PDK = Pyruvate dehydrogenase kinase gen peroxide. Biochem J 1972; 128: 617-30.
PERK = Protein Kinase RNA-like endoplasmic reticu- [23] Yamada Y, Harashima H. Mitochondrial drug-delivery systems for
lum kinase macromolecule and their therapeutic application to mitochondrial
diseases. Adv Drug Deliv Rev 2008; 60: 1439-62.
PSI = N-carbobenzoxy-L-isoleucyl-L--t-butyl-L- [24] Augustin W, Wiswedel I, Noack H, Reinheckel T, Reichelt O. Role
glutamyl-L-alanyl-L-leucinal of endogenous and exogenous antioxidants in the defense against
PTPC = Permeability transition pore complex functional damage and lipid peroxidation in rat liver mitochondria.
Mol Cell Biochem1997; 174: 199-205.
ROS = Reactive oxygen species [25] Fridovich I. Superoxide dismutases. Adv Enzymol Relat Areas Mol
SCLC = Small cell lung cancer Biol 1974; 41: 35-97.
[26] Rizzuto R, Pozzan T. Microdomains of intracellular Ca2+: molecu-
SERCA = Sarcoplasmic /endoplasmic reticulum cal- lar determinants and functional consequences. Physiol Rev 2006;
cium ATPase 86: 369-408.
SOD = Superoxide dismutases [27] Rapizzi E, Pinton P, Szabadkai G, et al. Recombinant expression of
the voltage-dependent anion channel enhances the transfer of Ca2+
tBid = truncated Bid microdomains to mitochondria. J Cell Biol 2002; 159: 613-624.
UPR = Unfolded protein response [28] Hengartner MO. The biochemistry of apoptosis. Nature 2000; 407:
770-76.
VDAC = Voltage dependent anion channel [29] Adams JM, Cory S. The Bcl-2 apoptotic switch in cancer develop-
ment and therapy. Oncogene 2007; 26: 1324-37.
REFERENCES [30] Barczyk K, Kreuter M, Pryjma J. Serum cytochrome c indicates in
[1] Hanahan D, Weinberg RA. Hallmarks of cancer: the next genera- vivo-apoptosis and it can serve as a prognostic marker during can-
tion. Cell 2011; 144: 646-74. cer therapy. Int J Cancer 2005; 114: 167-73.
[2] Fulda S. Tumor resistance to apoptosis. Int J Cancer 2009; 124: [31] Li LY, Luo X, Wang X. Endonuclease G is an apoptotic DNase
511-5. when released from mitochondria. Nature 2001; 412: 95-99.
[3] Fulda S, Galluzzi L, Kroemer G. Targeting mitochondria for cancer [32] Antonsson B, Martinou, JC. The Bcl-2 protein family. Exp Cell
therapy. Nat Rev Drug Discovery 2010; 9: 447-64. Res 2000; 256: 50-7.
[4] Maag RS, Hicks SW, Machamer CE. Death from within: apoptosis [33] Pavlov EV, Priault M, Pietkiewicz D. A novel, high conductance
and secretory pathway. Curr Opin Cell Biol 2003; 15: 456-61. channel of mitochondria linked to apoptosis in mammalian cells
[5] Waxman L, Fagan JM, Goldberg AL. Demonstration of two dis- and Bax expression in yeast. J Cell Biol 2001; 155: 725-32.
tinct high molecular weight proteases in rabbit reticulocytes, one of [34] Loeffler M, Kroemer G. The mitochondrion in cell death control:
which degrades ubiquitin conjugates. J Biol Chem 1987; 262: certainties and incognita. Exp Cell Res 2000; 256: 19-26.
2451-7. [35] Shimizu S, Matsuoka Y, Shinohara Y, Yoneda Y, Tsujimoto Y.
[6] Adams J. The proteasome: a suitable antineoplastic target. Nature Essential role of voltage-dependent anion channel in various forms
Reviews 2004; 4: 349-60. of apoptosis in mammalian cells. J Cell Biol 2001; 152: 237-50.
[7] Ciechanover A. The ubiquitin-proteasome proteolytic pathway. [36] Verrier F, Deniaud A, Lebras M. Dynamic evolution of adenine
Cell 1994; 79: 13-21. nucleotide translocase interactome during chemotherapy-induced
[8] Jäättelä M. Multiple cell death pathways as regulators of tumour apoptosis. Oncogene 2004; 23: 8049-64.
initiation and progression. Oncogene 2004; 23: 2746-56. [37] Kim R, Emi M, Tanabe M. Role of mitochondria as the gardens of
[9] Ferrin F, Linares CI, Muntane J. Mitochondrial drug targets in cell cell death. Cancer Chemother Pharmacol 2006; 57: 545-53.
death and cancer. Curr Pharm Des 2011; 17: 2002-16. [38] He L, Lemaster JJ. Regulated and unregulated mitochondrial per-
[10] Serviddio G, Ronmano AD, Cassano T, Bellanti F, Altomare E, meability transition pores: a new paradigm of pore structure and
Vendemial G. Principles and therapeutic relevance for targeting function? FEBS Lett 2002; 512: 1-7.
mitochondria in aging and neurodegenerative diseases. Curr Pharm [39] Martinou JC, Desagher S, Antonsson B. Cytochrome c release from
Des 2011; 17: 2036-55. mitochondria: all or nothing. Nat Cell Biol 2000; 2: E41-3.
[11] Victor VM, Rocha M, Banuls C, Bellod L, Hernandez-Mijares A. [40] Lemasters JJ, Qian T, He L. Role of mitochondrial inner membrane
Mitochondrial dysfunction and targeted drugs: a focus on diabetes. permeabilisation in necrotic cell death, apoptosis, and autophagy.
Curr Pharm Des 2011; 17: 1986-2001. Antioxid Redox Signal 2002; 4: 769-81.
[12] Carreira RS, Lee P, Gottlieb RA. Mitochondrial therapeutics for [41] Finkel E. The mitochondrian: is it central to apoptosis? Science
cardioprotection. Curr Pharm Des 2011; 17: 2017-35. 2001; 292: 624-6.
[13] Kroemer G, Galluzi L, Brenner C. Mitochondrial Membrane Per- [42] Maddika S, Ande SR, Panigrahi S. Cell survival, cell death and cell
meabilization in Cell Death. Physiol Rev 2007; 87: 99-163. cycle pathways are interconnected: Implications for cancer therapy.
[14] Mitchell P, Moyle J. Evidence discriminating between the chemical Drug Resistance Updates 2007; 10: 13-29.
and the chemiosmotic mechanisms of electron transport phosphory- [43] Jacotot E, Costantini P, Laboureau E, Zamzami N, Susin SA, Kro-
lation. Nature 1965; 208: 1205-6. emer G. Mitochondrial membrane permeabilisation during the
[15] Mitchell P, Moyle J. Stoichiometry of proton translocation through apoptotic process. Ann N Y Acad Sci 1999; 887: 18-30.
the respiratory chain and adenosine triphosphatase systems of rat [44] Fulda S, Debatin KM. Apoptosis signalling pathways in anticancer
mitochondria. Nature 1965; 208: 147-51. therapy. Curr Cancer Ther Rev 2008; 4: 14-20.
[16] Navarro A, Boveris A. The mitochondrial energy transduction [45] Warburg O, Dickens F. The metabolism of tumors. Am J Med Sci
system and aging process. Am J Physiol Cell Physiol 2007; 292: 1931; 182: 123
C670-86. [46] Warburg O, Wind F, Negelein E. The metabolism of tumor in the
[17] Marchetti P, Castedo M, Susin SA et al. Mitochondrial permeabil- body. J Gen Physiol 1927; 8:519-30.
ity transition is central coordinating event of apoptosis. J Exp Med [47] Brandon M, Baldi PA, Wallace DC. Mitochondrial mutations in
1996; 184: 1155-60. cancer. Oncogene 2006; 25: 4647-62.
[18] Zamzami N, Larochette N, Kroemer G. Mitochondrial permeability [48] Kroemer G. Mitochondria in cancer. Oncogene 2006; 25: 4630-2.
transition in apoptosis and necrosis. Cell Death Differ 2005; 12: [49] Singh KK, Ayyasamy V, Owens KM, Koul MS, Vujcic M. Muta-
1478-80. tions in mitochondrial DNA polymerase-gamma promote breast
[19] Kroemer G, Dallaporta B, Resche-Rigon M. The mitochondrial tumorigenesis. J Hum Genet 2009; 54: 516-24.
death/life regulator in apoptosis and necrosis. Annu Rev Physiol
1998; 60: 619-42.
Cancer Therapy Current Pharmaceutical Design, 2014, Vol. 20, No. 2 217

[50] King A, Selak MA, Gottlieb E. Succinate dehydrogenase and fu- [78] Baines CP, Kaiser RA, Purcell NH, et al. Loss of cyclophilin D
marate hydratase: linking mitochondrial dysfunction and cancer. reveals a critical role for mitochondrial permeability transition in
Oncogene 2006; 25: 4675-82. cell death. Nature 2005; 434: 658-62.
[51] Dang L, White DW, Gross S, et al. Cancer associated IDH1 muta- [79] De Marchi U, Basso E, Szabo I, Zoratti M. Electrophysiological
tions produce 2-hydroxyglutarate. Nature 2009; 462: 739-44. characterisation of the Cyclophilin D-deleted mitochondrial perme-
[52] Solani G, Sgarbi G, Baracca A. Oxidative phosphorylation in can- ability transition pore. Mol Membr Biol 2006; 23: 521-30.
cer cells. Biochim Biophys Acta 2011; 1807: 534-42. [80] Tsujimoto Y, Shimizu S. Role of the mitochondrial membrane
[53] Robey RB, Hay N. Mitochondrial hexokinases, novel mediators of permeability transition in cell death. Apoptosis 2007; 12: 835-40.
the antiapoptotic effects of growth factors and Akt. Oncogene [81] Hu Z, Guo X, Yu Q, et al. Down-regulation of adenine nucleotide
2006; 25: 4683-96. translocase 3 and its role in camptothecin-induced apoptosis in hu-
[54] Pelicano H, Martin DS, Xu RH, Huang P. Glycolysis inhibition for man hepatoma QGY7703 cells. FBS Lett 2009; 583: 383-8.
anticancer treatment. Oncogene 2006; 25: 4633-46. [82] Lee J, Schriner SE, Wallace DC. Adenine nucleotide translocator 1
[55] Green DR, Kroemer G. The pathophysiology of mitochondrial cell deficiency increases resistance of mouse brain and neurons to exci-
death. Science 2004; 305:626-9. totoxic insults. Biochim Biophys Acta 2009; 1787: 364-70.
[56] Cheng WC, Berman SB, Ivanovska I, et al. Mitochondrial factors [83] Jang JY, Choi Y, Jeon YK, Kim CW. Suppression of adenine nu-
with dual roles in death and survival. Oncogene 2006; 25: 4697- cleotide translocase-2 by vector-based siRNA in human breast can-
705. cer cells induces apoptosis and inhibits tumor growth in vitro and
[57] Brenner C, Grimm S. The permeability transition pore complex in in vivo. Breast Cancer Res 2008; 10: R11
cancer cell death. Oncogene 2006; 25: 4744-56. [84] Gallerne C, Touat Z, Chen ZX, et al. The fourth isoform of the
[58] Cereghetti GM, Scorrano L. The many shapes of mitochondrial adenine nucleotide translocator inhibits mitochondrial apoptosis in
death. Oncogene 2006; 25: 4717-4724. cancer cells. Int J Biochem Cell Biol 2010; 42: 623-9.
[59] Moll UM, Marchenko N, Zhang XK. p53 and Nur77/TR3: Tran- [85] Schimmer AD, O’Brien S, Kantarjian H, et al. A phase I study of
scription factors that directly target mitochondria for cell death in- the pan bcl-2 family inhibitor obatoclax mesylate in patients with
duction. Oncogene 2006; 25: 4725-43. advanced hematologic malignancies. Clin Cancer Res 2008; 14:
[60] Fontenay M, Cathelin S, Amiot M, Gyan E, Solary E. Mitochon- 8295-301.
dria in hematopoiesis and hematological diseases. Oncogene 2006; [86] Belzacq AS, El Hamel C, Vieira HL, et al. Adenine nucleotide
25: 4757-67. translocator mediates the mitochondrial membrane permeabilisa-
[61] Shanmugam M, McBrayer SK, Rosen ST. Targeting the Warburg tion induced by Ionidamine, arsenite and CD437. Oncogene 2001;
effect in haematological malignancies: from PET to therapy. Curr 20: 7579-87.
Opin Oncol 2009; 21: 531-6 [87] Shanmugam M, McBrayer SK, Rosen ST. Targeting the Warburg
[62] Mathupala SP, Ko YH, Pedersen PL. Hexokinase II: Cancer's Dou- effect in haematological malignancies: from PET to Therapy. Curr
ble-Edged Sword Acting as Both Facilitator and Gatekeeper of Ma- Opin Oncol 2009; 21: 531-6.
lignancy when Bound to Mitochondria. Oncogene 2006; 25: 4777- [88] Oudard S, Carpentier A, Banu, E, et al. Phase II study of Ioni-
86. damine and diazepam in the treatment of recurrent glioblastoma
[63] Zhivotovsky B, Kroemer G. Apoptosis and genomic instability. Nat multiform. J Neurooncol 2003; 63: 81-6.
Rev Mol Cell Biol 2004; 5: 752-62. [89] Dogliott L, Berruti A, Buniva T, et al. Lonidamine significantly
[64] He Y, Wu J, Dressman DC, et al. Heteroplasmic mitochondrial increases the activity of epirubicin in patients with advanced breast
DNA mutations in normal and tumour cells. Nature 2010; 464: cancer: results from a multicenter prospective randomised trial. J
610-4. Clin Oncol 1996; 14: 1165-72.
[65] Polyak K, Li Y, Zhu H, et al. Somatic mutations of the mitochon- [90] Berruti A, Bitossi R, Gorzegno G, et al. Time to progression in
drial genome in human colorectal tumours. Nat Genet 1998; 20: metastatic breast cancer patients treated with epirubicin is not im-
291-3. proved by the addition of either cisplatin or lonidamine: final re-
[66] Hung WY, Wu CW, Yin PH, et al. Somatic mutations in mito- sults of a phase III study with a factorial design. J Clin Oncol 2002;
chondrial genome and their potential roles in the progression of 20: 4150-9.
human gastric cancer. Biochim Biophys Acta 2010; 1800: 264-70. [91] De Lena M, Lorusso V, Latorre A, et al. Paclitaxel, cisplatin and
[67] Kulawiec M, Owens KM, Singh KK. Cancer cell mitochondria lonidamine in advanced ovarian cancer. A phase II study. Eur J
confer apoptosis resistance and promote metastasis. Cancer Biol Cancer 2001; 37: 364-8.
Ther 2009; 8: 1378-85. [92] Mullauer FB, Kessler JH, Medema JP. Betulinic acid induces cyto-
[68] Dowling RJ, Topisirovic I, Fonseca BD, Sonenberg N. Dissecting chrome c release and apoptosis in a Bax/Bak-independent, perme-
the role of mTOR: lessons from mTOR inhibitors. Biochim Bio- ability transition pore dependent fashion. Apoptosis 2009; 14: 191-
phys Acta 2010; 1804: 433-9. 202.
[69] Shackelford RE, Kaufmann WK, Paules RS. Oxidative stress and [93] Eiznhamer DA, Xu ZQ. Betulinic acid: a promising anti-cancer
cell cycle checkpoint function. Free Radic Biol Med 2000; 28: candidate. IDRugs 2004; 7:359-73.
1387-1404. [94] Fulda S, Debatin KM. Betulinic acid induces apoptosis through a
[70] Bartsch H. Studies on biomarkers in cancer etiology and preven- direct effect on mitochondria in neuroectodermal tumors. Med Pe-
tion: a summary and challenge of 20 years of interdisciplinary re- diatr Oncol 2000; 35: 616-8.
search. Mutat Res 2000; 462: 255-79. [95] Fulda S, Scaffidi C, Susin S A, et al. Activation of mitochondria
[71] Marnett LJ. Oxyradicals and DNA damage. Carcinogenesis 2000; and release of mitochondrial apoptogenic factors by betulinic acid.
21: 361-70. J Biol Chem 1998; 273: 33942-8.
[72] Gogvadze V, Orrenius S, Zhivotovsky B. Mitochondria in cancer [96] Fulda S, Susin SA, Kroemer G, Debatin KM. Molecular ordering
cells: what is so special about them? Trends Cell Biol 2008; 18: of apoptosis induced by anticancer drugs in neuroblastoma cells.
165-73. Cancer Res 1998; 58: 4453-60.
[73] Modica-Napolitano JS, Singh KK. Mitochondrial dysfunction in [97] Fulda S, Friesen C, Los M, et al. Betulinic acid triggers CD95
cancer. Mitochondrion 2004; 4: 755-62. (APO-1/Fas)-and p53-independent apoptosis via activation of
[74] Bellance N, Lestienne P, Rossignol R. Mitochondria: from bio- caspases in neuroectodermal tumors. Cancer Res 1997; 57: 4956-
energetics to the metabolic regulation of carcinogenesis. Front Bio- 64.
sci 2009; 14: 4015-34. [98] Cadd VA, Hogg PJ, Harris AL, Feller SM. Molecular profiling of
[75] Baines CP, Kaiser RA, Sheiko T, Craigen WJ, Molkentin JD. Volt- signalling proteins for effects induced by the anti-cancer compound
age-dependent anion channels are dispensable for mitochondrial- GSAO with 400 antibodies. BMC Cancer 2006; 6:155
dependent cell death. Nat Cell Biol 2007; 9: 550-5. [99] Dilda PJ, Ramsay EE, Corti A, Pompella A, Hogg PJ. Metabolism
[76] Galluzzi L, Kroemer G. Mitochondrial apoptosis without VDAC. of the tumor angiogenesis inhibitor 4-(N-(S-Glutathionylacetyl)
Nat Cell Biol 2007; 9: 487-9. amino)phenylarsonous acid. J Biol Chem 2008; 283:35428-34.
[77] Kokoszka JE, Waymire KG, Levy SE, et al. The ADP/ATP trans- [100] Don AS, Kisker O, Dilda P, et al. A peptide trivalent arsenical
locator is not essential for the mitochondrial permeability transition inhibits tumor angiogenesis by perturbing mitochondrial function
pore. Nature 2004; 427: 461-5. in angiogenic endothelial cells. Cancer Cell 2003; 3: 497-509.
218 Current Pharmaceutical Design, 2014, Vol. 20, No. 2 Ubah and Wallace

[101] Dilda PJ, Decollogne S, Weerakoon L, et al. Optimisation of the ovarian cancer and primary peritoneal carcinomatosis: a Hoosier
antitumour efficacy of a synthetic mitochondrial toxin by increas- Oncology Group trial. Gynecol Oncol 2009; 115: 90-6.
ing the residence time in the cytosol. J Med Chem 2009; 52: 6209- [124] Tevaarwerk AJ, Holen KD, Alberti DB, et al. Phase I trial of 2-
16. methoxyestradiol NanoCrystal dispersion in advanced solid malig-
[102] Park D, Chiu J, Perrone GG, Dilda PJ, Hogg PJ. The tumour me- nancies. Clin Cancer Res 2009; 15: 1460-5.
tabolism inhibitors GSAO and PENAO react with cysteines 57 and [125] Juarez JC, Manuia M, Burnett ME, et al. Superoxide dismutase 1
257 of mitochondrial adenine nucleotide translocase. Cancer Cell (SOD1) is essential for H2O2-mediated oxidation and inactivation
Intl 2012; 12: 11 of phosphatises in growth factor signalling. Proc Natl Acad Sci
[103] Slack JL, Rusiniak ME. Current issues in the management of acute USA 2008; 105: 7147-52.
promyelocytic leukemia. Ann Hematol 2000; 79: 227-38. [126] Nonn L, Berggren M, Powis G. Increased expression of mitochon-
[104] Notario B, Zamora M, Vinas O, Mampel T. All-trans-retinoic acid drial peroxiredoxin-3 (thioredoxin peroxidase-2) protects cancer
binds to and inhibits adenine nucleotide translocase and induces cells against hypoxia and drug-induced hydrogen peroxide-
mitochondrial permeability transition. Mol Pharmacol 2003; 63: dependent apoptosis. Mol Cancer Res 2003; 1:682-9.
224-31. [127] Kropotov A, Gogvadze V, Shupliakov O, et al. Peroxiredoxin V is
[105] Magda D, Miller RA. Motexafin gadolinium: a novel redox active essential for protection against apoptosis in human lung carcinoma
drug for cancer therapy. Semin Cancer Biol 2006; 16: 466-76. cells. Exp Cell Res 2006; 312: 2806-15.
[106] Bradley KA, Pollack IF, Reid JM, et al. Motexafin gadolinium and [128] Song IS, Kim HK, Jeong SH, et al. Mitochondrial peroxiredoxin III
involved field radiation therapy for intrinsic pontine glioma of is apotential target for cancer therapy. Int J Mol Sci 2011; 12:
childhood: a Children’s Oncology Group phase I study. Neuro On- 7163-85.
col 2008; 10: 752-8. [129] Chung YM, Yoo YD, Park JK, Kim YT, Kim HJ. Increased ex-
[107] Costantini P, Belzacq A, La Vieira H, et al. Oxidation of a critical pression of peroxiredoxin II confers resistance to cisplatin. Anti-
thiol residue of the adenine nucleotide translocator enforces Bcl-2- cancer Res 2001; 21: 1129-33.
independent permeability transition pore opening and apoptosis. [130] Noh YH, Baek JY, Jeong W, Rhee SG, Chang TS. Sulfiredoxin
Oncogene 2000; 19: 307-14. transloaction into mitochondria play a crucial role in reducing hy-
[108] Bey EA, Bentle MS, Reinicke KE, et al. An NQO1- and PARP-1- peroxidised peroxiredoxin III. J Biol Chem 2009; 284: 8470-7.
mediated cell death pathway induced in non-small-cell lung cancer [131] Chipuk JE, Bouchier-Hayes L, Green DR. Mitochondrial outer
cells by -lapachone. Proc Natl Acad Sci USA 2007; 104: 11832-7. membrane permeabilisation during apoptosis: the innocent by-
[109] Maeda H, Ohizumi H, Segawa T, Kakehi Y, Ogawa O, Kakizuka stander scenario. Cell Death Differ 2006; 13: 1396-402.
A. Effective treatment of advanced solid tumours by the combina- [132] Gavathiotis E, Suzuki M, Davis ML, et al. BAX activation is initi-
tion of arsenic trioxide and L-buthionine-sulfoximine. Cell Death ated at a novel interaction site. Nature 2008; 455: 1076-81.
Differ 2004; 11: 737-46. [133] Bellot G, Cartron PF, Er E, et al. TOM22, a core component of the
[110] Dragovich T, Mendelson D, Wong L, et al. Phase I trial of imexon mitochondrial outer membrane protein translocation pore, is a mi-
in patients with advanced malignancy. J Clin Oncol 2007; 25: tochondrial receptor for the proapoptotic protein Bax. Cell Death
1779-84. Differ 2007; 14: 785-94.
[111] Moulder S, Dhillon N, Ng C, et al. Phase I trial of imexon, a pro- [134] Ross K, Rudel T, Kozjak-Pavlovic V. TOM-independent complex
oxidant, in combination with docetaxel for the treatment of pateints formation of Bax and Bak in mammalian mitochondria during
with advanced breast, non-small cell lung and prostate cancer. In- TNF-induced apoptosis. Cell Death Differ 2009; 16: 697-707.
vest New Drugs 2010; 28:634-40. [135] Rostovtseva TK, Boukari H, Antignani A, et al. Bax activates
[112] Tuma RS. Reactive oxygen species may have antitumor activity in endophilin B1 oligomerisation and lipid membrane vesiculation. J
metastatic melanoma. J Natl Cancer Inst 2008; 100: 11-12. Biol Chem 2009; 284: 34390-9.
[113] Kirshner JR, He S, Balasubramanyam V, et al. Elesclomol induces [136] Cipolat S, Rudka T, Hartmann D, et al. Mitochondrial rhomboid
cancer cell apoptosis through oxidative stress. Mol Cancer Ther PARL regulates cytochrome c release during apoptosis via OPA 1-
2008; 7: 2319-27. dependent cristae remodeling. Cell 2006; 126: 163-75.
[114] Berkenblit A, Eder JP Jr., Ryan DP, et al. Phase I clinical trial of [137] Frezza C, Cipolat S, Martins de Brito O, et al. OPA 1 controls
STA-4783 in combination with paclitaxel in patients with refrac- apoptotic cristae remodeling independently from mitochondrial fu-
tory solid tumors. Clin Cancer Res 2007; 13: 584-90. sion. Cell 2006; 126: 177-89.
[115] O’Day S, Gonzalez R, Lawson D, et al. Phase II, randomised, [138] Vogler M, Dinsdale D, Dyer MJ, Cohen GM. Bcl-2 inhibitors:
controlled, double-blinded trial of weekly elesclomol plus pacli- small molecules with a big impact on cancer therapy. Cell Death
taxel versus paclitaxel alone for stage IV metastatic melanoma. J Differ 2009; 16: 360-7.
Clin Oncol 2009; 27: 5452-8. [139] Vogler M, Weber K, Dinsdale D, et al. Different forms of cell
[116] Alexandre J, Nicco C, Chéreau C, et al. Improvement of the thera- death induced by putative BCL2 inhibitors. Cell Death Differ 2009;
peutic index of anticancer drugs by the superoxide dismutase 16: 1030-9.
mimic mangafodipir. J Natl Cancer Inst 2006; 98: 236-44. [140] Oltersdorf T, Elmore, SW, Shoemaker AR, et al. An inhibitor of
[117] Trachootham D, Zhou Y, Zhang H, et al. Selective killing of onco- Bcl-2 family proteins induces regression of solid tumors. Nature
genically transformed cells through a ROS-mediated mechanism by 2005; 435: 677-81.
-phenylethyl isothiocyanate. Cancer Cell 2006; 10: 241-52. [141] Kang MH, Kang YH, Szymanska B, et al. Activity of vincristine,
[118] Xiao D, Lew KL, Zeng Y, et al. Phenethyl isothiocyanate-induced L-ASP, and dexamethasone against acute lymphoblastic leukaemia
apoptosis in PC-3 human prostate cancer cells is mediated by reac- is enhanced by the BH3-mimetic ABT-737 in vitro and in vivo.
tive oxygen species -dependent disruption of the mitochondrial Blood 2007; 110: 2057-66.
membrane potential. Carcinogenesis 2006; 27: 2223-34. [142] Tagscherer KE, Fassl A, Campos B, et al. Apoptosis-based treat-
[119] Xiao D, Powolny AA, Moura MB, et al. Phenethyl isothiocyanate ment of glioblastomas with ABT-737, a novel small molecule in-
inhibits oxidative phosphorylation to trigger reactive oxygen spe- hibitor of Bcl-2 family proteins. Oncogene 2008; 27: 6646-56.
cies-mediated death of human prostate cancer cells. J Biol Chem [143] Kutuk O, Letai A. Alteration of the mitochondrial apoptotic path-
2010; 285: 26558-69. way is key to acquired paclitaxel resistance and can be reversed by
[120] Chan DK, Miskimins WK. Metformin and phenethyl isothiocy- ABT-737. Cancer Res 2008; 68: 7985-94.
anate combined treatment in vitro is cytotxic to ovarian cancer cul- [144] Hann CL, Daniel VC, Sugar EA, et al. Therapeutic efficacy of
tures. J Ovarian Res 2012; 5: 19 ABT-737, a selective inhibitor of BCL-2, in small cell lung cancer.
[121] Wood L, Leese MR, Leblond B, et al. Inhibition of superoxide Cancer Res 2008; 68: 2321-8.
dismutase by 2-methoxyoestradiol analogues and oestrogen deriva- [145] Tse C, Shoemaker AR, Adickes J, et al. ABT-263: a potent and
tives: structure-activity relationships. Anticancer Drug Des 2001; orally bioavailable Bcl-2 family inhibitor. Cancer Res 2008; 68:
16: 209-15. 3421-8.
[122] Cleveland JL, Kastan MB. Cancer: a radical approach to treatment. [146] Lynn A, Jones L. Gossypol and some other terpenoids, flavonoids,
Nature 2000; 407: 309-11. and phenols that affect quality of cottonseed protein. Am Oil
[123] Matei D, Schilder J, Sutton G, et al. Activity of 2- Chemists Soc 1979; 56: 727-30.
methoxyoestradiol (Panzem NCD) in advanced, platinum-resistant [147] Azmi AS, Mohammad RM. Non-peptidic small molecule inhibitors
against Bcl-2 for cancer therapy. J Cell Physiol 2009; 218: 13-21.
Cancer Therapy Current Pharmaceutical Design, 2014, Vol. 20, No. 2 219

[148] Liu G, Kelly WK, Wilding G, Leopold L, Brill K, Somer B. An [170] Huang Y, Stewart TM, Wu Y, et al. Novel oligoamine analogues
open-label, multicentre, phase I/II study of single-agent AT-101 in inhibit lysine-specific demethylase 1 and induce reexpression of
men with castrate-resistant prostate cancer. Clin Cancer Res 2009; epigenetically silenced genes. Clin Cancer Res 2009; 15: 7217-28.
15: 3172-219. [171] Xie SQ, Liu GC, Ma YF, et al. Synergistic antitumor effects of
[149] Kitada S, Kress CL, Krajewska M, Jia L, Pellecchia M, Reed JC. anthracenylmethyl homospermidine and alpha-
Bcl-2 antagonist apogossypol (NSC736630) displays single-agent difluoromethylornithine on promyelocytic leukemia HL60 cells.
activity in Bcl-2-transgenic mice and has superior efficacy with Toxicol In vitro 2008; 22: 352-8.
less toxicity compared with gossypol (NSC19048). Blood 2008; [172] Palmer A J, Ghani RA, Kaur N, Phanstiel O, Wallace HM. A pu-
111: 3211-9. trescine-anthracene conjugate: a paradigm for selective drug deliv-
[150] Shoemaker AR, Oleksijew A, Bauch J, et al. A small-molecule ery. Biochem J 2009; 424: 431-8.
inhibitor of Bcl-XL potentiates the activity of cytotoxic drugs in vi- [173] Guppy M, Greiner E, Brand K. The role of the Crabtree effect and
tro and in vivo. Cancer Res 2006; 66: 8731-9. an endogenous fuel in the energy metabolism of resting and prolif-
[151] Nguyen M, Marcellus RC, Roulston A, et al. Small molecule oba- erating thymocytes. Eur J Biochem 1993; 212: 95-9.
toclax (GX15-070) antagonizes MCL-1 and overcomes MCL-1- [174] DeBerardinis RJ, Mancuso A, Daikhin E, et al. Beyond aerobic
mediated resistance to apoptosis. Proc Natl Acad Sci USA 2007; glycolysis: transformed cells can engage in glutamine metabolism
104: 19512-7. that exceeds the requirement for protein and nucleotide synthesis.
[152] Trudel S, Li ZH, Rauw J, Tiedemann RE, Wen XY, Stewart AK. Proc Natl Acad Sci USA 2007; 104: 19345-50.
Preclinical studies of the pan-Bcl inhibitor obatoclax (GX015-070) [175] Gatenby RA, Gillies RJ. Why do cancers have high aerobic glyco-
in multiple myeloma. Blood 2007; 109: 5430-8. lysis? Nat Rev Cancer 2004; 4: 891-9.
[153] O'Brien SM, Claxton DF, Crump M, et al. Phase I study of obato- [176] Ko YH, Smith BL, Wang Y, et al. Advanced cancers: eradication
clax mesylate (GX15-070), a small molecule pan-Bcl-2 family an- in all cases using 3-bromopyruvate therapy to deplete ATP. Bio-
tagonist, in patients with advanced chronic lymphocytic leukemia. chem Biophysical Res Commun 2004; 324: 269-75.
Blood 2009; 113: 299-305. [177] Xu RH, Pelicano H, Zhou Y, et al. Inhibition of glycolysis in can-
[154] Schimmer A D, O'Brien S, Kantarjian H, et al. A phase I study of cer cells: a novel strategy to overcome drug resistance associated
the pan bcl-2 family inhibitor obatoclax mesylate in patients with with mitochondrial respiratory defect and hypoxia. Cancer Res
advanced hematologic malignancies. Clin Cancer Res 2008; 14: 2005; 65: 613-21.
8295-301. [178] Vali M, Liapi E, Kowalski J, et al. Intraarterial therapy with a new
[155] Parikh SA, Kantarjian H, Schimmer A, et al. Phase II study of potent inhibitor of tumor metabolism (3-bromopyruvate): identifi-
obatoclax mesylate (GX15-070), a small-molecule BCL-2 family cation of therapeutic dose and method of injection in an animal
antagonist, for patients with myelofibrosis. Clinical lymphoma, model of liver cancer. J Vasc Interv Radiol 2007; 18: 95-101.
myeloma & leukemia 2010; 10: 285-9. [179] Airley RE, Mobasheri A. Hypoxic regulation of glucose transport,
[156] Klasa RJ, Gillum AM, Klem RE, Frankel SR. Oblimersen Bcl-2 anaerobic metabolism and angiogenesis in cancer: novel pathways
antisense: facilitating apoptosis in anticancer treatment. Antisense and targets for anticancer therapeutics. Chemotherapy 2007; 53:
Nucleic Acid Drug Dev 2002; 12: 193-213. 233-56.
[157] Moulder SL, Symmans WF, Booser DJ, et al. Phase I/II study of [180] Medina RA, Owen GI. Glucose transporters: expression, regulation
G3139 (Bcl-2 antisense oligonucleotide) in combination with and cancer. Biol Res 2002; 35: 9-26.
doxorubicin and docetaxel in breast cancer. Clin Cancer Res 2008; [181] Matsushita K, Uchida K, Saigusa S, et al. Glycolysis inhibitors as a
14: 7909-16. potential therapeutic option to treat aggressive neuroblastoma ex-
[158] O'Brien S, Moore JO, Boyd TE, et al. Randomized phase III trial of pressing GLUT1. J Pediatr Surg 2012; 47: 1323-30.
fludarabine plus cyclophosphamide with or without oblimersen so- [182] Xu RH, Pelicano H, Zhang H, Giles FJ, Keating MJ, Huang P.
dium (Bcl-2 antisense) in patients with relapsed or refractory Synergistic effect of targeting mTOR by rapamycin and depleting
chronic lymphocytic leukemia. J Clin Oncol 2007; 25: 1114-1120. ATP by inhibition of glycolysis in lymphoma and leukemia cells.
[159] Pro B, Leber B, Smith M, et al. Phase II multicenter study of Leukemia 2005; 19: 2153-8.
oblimersen sodium, a Bcl2 antisense oligonucleotide, in combina- [183] Chiara F, Castellaro D, Marin O, et al. Hexokinase II detachment
tion with rituximab in patients with recurrent Bcell nonHodgkin from mitochondria triggers apoptosis through the permeability
lymphoma. Br J Haematol 2008; 143: 355-60. transition pore independent of voltage-dependent anion channels.
[160] Chanan-Khan AA, Niesvizky R, Hohl RJ, et al. Phase III random- PLoS One 2008; 3: e1852.
ised study of dexamethasone with or without oblimersen sodium [184] Goldin N, Arzoine L, Heyfets A, et al. Methyl jasmonate binds to
for patients with advanced multiple myeloma. Leuk Lymphoma and detaches mitochondria-bound hexokinase. Oncogene 2008; 27:
2009; 50: 559-65. 4636-43.
[161] Rudin CM, Salgia R, Wang X, et al. Randomized phase II Study of [185] Brown J. Effect of 2-deoxyglucose on carbohydrate metabolism:
carboplatin and etoposide with or without the bcl-2 antisense oli- review of the literature and studies in the rat. Metabolism 1962; 11:
gonucleotide oblimersen for extensive-stage small-cell lung cancer: 1098-112.
CALGB 30103. J Clin Oncol 2008; 26: 870-6. [186] Kang HT, Hwang ES. 2-Deoxyglucose: an anticancer and antiviral
[162] Wang JL, Liu D, Zhang ZJ, et al. Structure-based discovery of an therapeutic, but not anymore a low glucose mimetic. Life Sci 2006;
organic compound that binds Bcl-2 protein and induces apoptosis 78: 1392-9.
of tumor cells. Proc Natl Acad Sci USA 2000; 97: 7124-9. [187] Maher J, Krishan A, Lampidis T. Greater cell cycle inhibition and
[163] Manero F, Gautier F, Gallenne T, et al. The small organic com- cytotoxicity induced by 2-deoxy-D-glucose in tumor cells treated
pound HA14-1 prevents Bcl-2 interaction with Bax to sensitize ma- under hypoxic vs aerobic conditions. Cancer Chemother Pharmacol
lignant glioma cells to induction of cell death. Cancer Res 2006; 2004; 53: 116-22.
66: 2757-64. [188] Simons AL, Ahmad IM, Mattson DM, Dornfeld KJ, Spitz DR. 2-
[164] Oliver L, Mahé B, Gréé R, Vallette FM, Juin P. HA14-1, a small Deoxy-D-glucose combined with cisplatin enhances cytotoxicity
molecule inhibitor of Bcl-2, bypasses chemoresistance in leukae- via metabolic oxidative stress in human head and neck cancer cells.
mia cells. Leuk Res 2007; 31: 859-63. Cancer Res 2007; 67: 3364-70.
[165] Wallace HM, Fraser AV, Hughes A. A perspective of polyamine [189] Singh D, Banerji AK, Dwarakanath BS, et al. Optimizing cancer
metabolism. Biochem J 2003; 376:1-14. radiotherapy with 2-deoxy-D-glucose. Strahlentherapie und Onk-
[166] Gerner EW, Meyskens FL. Polyamines and cancer: old molecules, ologie 2005; 181: 507-14.
new understanding. Nat Rev Cancer 2004; 4: 781-92. [190] Zhong D, Liu X, Schafer-Hales K, et al. 2-Deoxyglucose induces
[167] Marton LJ, Pegg AE. Polyamines as targets for therapeutic inter- Akt phosphorylation via a mechanism independent of LKB1/AMP-
vention. Annu Rev Pharmacol Toxicol 1995; 35: 55-91. activated protein kinase signalling activation or glycolysis inhibi-
[168] Bergeron RJ, Neims AH, McManis JS, et al. Synthetic polyamine tion. Mol Cancer Ther 2008; 7: 809-17.
analogs as antineoplastics. J Med Chem 1988; 31: 1183-90. [191] Bonnet S, Archer SL, Allalunis-Turner J, et al. A Mitochondria-K+
[169] Casero RA, Marton LJ. Targeting polyamine metabolism and func- Channel Axis Is Suppressed in Cancer and Its Normalization Pro-
tion in cancer and other hyperproliferative diseases. Nat Rev Drug motes Apoptosis and Inhibits Cancer Growth. Cancer cell 11: 37-
Dis 2007; 6: 373-90. 51.
220 Current Pharmaceutical Design, 2014, Vol. 20, No. 2 Ubah and Wallace

[192] Stacpoole PW, Kerr DS, Barnes C, et al. Controlled clinical trial of [218] Chauhan D, Tian Z, Zhou B, et al. In vitro and in vivo selective
dichloroacetate for treatment of congenital lactic acidosis in chil- antitumor activity of a novel orally bioavailable proteasome inhibi-
dren. Pediatrics 2006; 117: 1519-1531. tor MLN9708 against multiple myeloma cells. Clin Cancer Res
[193] Hatzivassiliou G, Zhao F, Bauer DE, et al. ATP citrate lyase inhibi- 2011; 17: 5311-21.
tion can suppress tumor cell growth. Cancer Cell 2005; 8: 311-21. [219] Chauhan D, Singh A V, Aujay M, et al. A novel orally active pro-
[194] Wellen KE, Hatzivassiliou G, Sachdeva UM, Bui TV, Cross JR, teasome inhibitor ONX 0912 triggers in vitro and in vivo cytotoxic-
Thompson CB. ATP-citrate lyase links cellular metabolism to his- ity in multiple myeloma. Blood 2010; 116: 4906-15.
tone acetylation. Sci Signalling 2009; 324: 1076-80. [220] Papadopoulos KP, Mendelson DS, Tolcher AW, Patnaik A,
[195] Brooks P, Fuertes G, Murray RZ, et al. Subcellular localization of Burris HA, Rasco DW. A phase I, open-label, dose-
proteasomes and their regulatory complexes in mammalian cells. escalation study of the novel oral proteasome inhibitor (PI)
Biochem J 2000; 346: 155-61. ONX 0912 in patients with advanced refractory or recurrent
[196] Elliott PJ, Ross JS. The proteasome: a new target for novel drug solid tumors. J Clin Oncol 2011; 29.
therapies. Am J Clin Pathol 2001; 116: 637-46. [221] Yoshida H. ER stress response, Peroxisome prioliferation, Mito-
[197] Ahn K, Erlander M, Leturcq D, Peterson PA, Früh K, Yang Y. In chondrial unfolded protein response and Golgi stress response. Life
vivo characterization of the proteasome regulator PA28. J Biol 2009; 61: 871-9.
Chem 1996; 271: 18237-42. [222] Brodsky JL. The protective and destructive roles played by molecu-
[198] Rechsteiner M, Realini C, Ustrell V. The proteasome activator 11 S lar chaperones during ERAD (endoplasmic-reticulum-associated
REG (PA28) and class I antigen presentation. Biochem J 2000; degradation). Biochem J 2007; 404: 353-63.
345: 1-15. [223] Malhotra JD, Kaufman RJ. The endoplasmic reticulum and the
[199] Lafarga M, Fernández R, Mayo I, Berciano MT, Castaño JG. Pro- unfolded protein response. Semin Cell Dev Biol 2007 18: 716-31.
teasome dynamics during cell cycle in rat Schwann cells. Glia [224] Yoshida H. ER stress and diseases. FEBS J 2007; 274: 630-58.
2002; 38: 313-28. [225] Wlodkowic D, Skommer J, McGuinness D, Hillier C,
[200] Drexler HC. Activation of the cell death program by inhibition of Darzynkiewicz Z. ER-Golgi network - a future target for anti-
proteasome function. Proc Natl Acad Sci USA 1997; 94: 855-60. cancer therapy. Leuk Res 2009; 33: 1440-7.
[201] Drexler HC, Risau W, Konerding MA. Inhibition of proteasome [226] Wang G, Yang ZQ, Zhang K. Endoplasmic reticulum stress re-
function induces programmed cell death in proliferating endothelial sponse in cancer: molecular mechanism and therapeutic potential.
cells. FASEB J 2000; 14: 65-77. Am J Transl Res 2010; 2: 65-74.
[202] Rajkumar S V, Richardson PG, Hideshima T, Anderson KC. Pro- [227] Lee AS. GRP78 induction in cancer: therapeutic and prognostic
teasome inhibition as a novel therapeutic target in human cancer. J implications. Cancer Res 2007; 67: 3496-9.
Clin Oncol 2005; 23: 630-9. [228] Orrenius S, Zhivotovsky B, Nicotera P. Regulation of cell death:
[203] Adams J, Palombella VJ, Sausville EA, et al. Proteasome inhibi- the calcium-apoptosis link. Nat Rev Mol Cell Biol 2003; 4: 552-65.
tors: a novel class of potent and effective antitumor agents. Cancer [229] Scorrano L, Korsmeyer SJ. Mechanisms of cytochrome c release
Res 1999; 59: 2615-22. by proapoptotic BCL-2 family members. Biochem Biophys Res
[204] Richardson PG, Barlogie B, Berenson J, et al. A phase 2 study of Commun 2003; 304: 437-44.
bortezomib in relapsed, refractory myeloma. New Engl JMed 2003; [230] Healy SJ, Gorman AM, Mousavi-Shafaei P, Gupta S, Samali A.
348: 2609-17. Targeting the endoplasmic reticulum-stress response as an antican-
[205] Jagannath S, Durie BG, Wolf J, et al. Bortezomib therapy alone cer strategy. Eur J Pharmacol 2009; 625: 234-46.
and in combination with dexamethasone for previously untreated [231] Obeng EA, Carlson LM, Gutman DM, et al. Proteasome inhibitiors
symptomatic multiple myeloma. Br J Haematol 2005; 129: 776-83. induce a terminal unfolded protein response in multiple myeloma
[206] Cusack JC. Rationale for the treatment of solid tumors with the cells. Blood 2006; 107: 4907-16.
proteasome inhibitor bortezomib. Cancer Treat Rev 2003; 29: 21- [232] Davenport EL, Moore HE, Dunlop AS, et al. Heat shock protein
31. inhibition is associated with activation of the unfolded protein re-
[207] Lenz HJ. Clinical update: Proteasome inhibitors in solid tumors. sponse pathway in myeloma plasma cells. Blood 2007; 110: 2641-
Cancer Treat Rev 2003; 29: 41-8. 49.
[208] Demo SD, Kirk CJ, Aujay MA, et al. Antitumor activity of PR- [233] Gills JJ, LoPiccolo J, Tsurutani J, et al. Nelfinavir, A lead HIV
171, a novel irreversible inhibitor of the proteasome. Cancer Res protease inhibitor, is a broad-spectrum, anticancer agent that in-
2007; 67: 6383-91. duces endoplasmic reticulum stress, autophagy, and apoptosis in vi-
[209] Kuhn DJ, Chen Q, Voorhees P M, et al. Potent activity of carfil- tro and in vivo. Clinical Cancer Res 2007; 13: 5183-94.
zomib, a novel, irreversible inhibitor of the ubiquitin-proteasome [234] Pyrko P, Kardosh A, Wang W, Xiong W, Schönthal AH, Chen TC.
pathway, against preclinical models of multiple myeloma. Blood HIV-1 protease inhibitors nelfinavir and atazanavir induce malig-
2007; 110: 3281-90. nant glioma death by triggering endoplasmic reticulum stress. Can-
[210] Molineaux SM. Molecular pathways: targeting proteasomal protein cer Res 2007; 67: 10920-28.
degradation in cancer. Clin Cancer Res 2012; 18: 15-20. [235] Kraus M, Malenke E, Gogel J, et al. Ritonavir induces endoplasmic
[211] Bross PF, Kane R, Farrell AT, et al. Approval summary for borte- reticulum stress and sensitizes sarcoma cells toward bortezomib-
zomib for injection in the treatment of multiple myeloma. Clin induced apoptosis. Mol Cancer Ther 2008; 7: 1940-8.
Cancer Res 2004; 10: 3954-64. [236] Deng WG, Ruan KH, Du M, Saunders MA, Wu KK. Aspirin and
[212] Niesvizky R, Wang L, Orlowski RZ, et al. Phase Ib multicenter salicylate bind to immunoglobulin heavy chain binding protein
dose escalation study of carfilzomib plus lenalidomide and low (BiP) and inhibit its ATPase activity in human fibroblasts. FASEB
dose dexamethasone (CRd) in relapsed and refractory multiple J 15: 2463-70.
myeloma (MM). Blood 2009; 114: 128-9. [237] Ermakova SP, Kang BS, Choi BY, et al. () Epigallocatechin
[213] Moreau P, Richardson PG, Cavo M, et al. Proteasome inhibitors in Gallate Overcomes Resistance to Etoposide-Induced Cell Death by
multiple myeloma: 10 years later. Blood 2012; 120: 947-59. Targeting the Molecular Chaperone Glucose-Regulated Protein 78.
[214] Baritaki S, Berkers C, Bonavida B, et al. Marizomib, a proteasome Cancer Res 2006; 66: 9260-9.
inhibitor for all seasons: preclinical profile and a framework for [238] Zhou Y, Lee AS. Mechanism for the suppression of the mammalian
clinical trials. Curr Cancer Drug Targets 2011; 11: 254-84. stress response by genistein, an anticancer phytoestrogen from soy.
[215] Obaidat A, Weiss J, Wahlgren B, et al. Proteasome regulator mari- J Natl Cancer Inst 1998; 90: 381-8.
zomib (NPI-0052) exhibits prolonged inhibition, attenuated efflux, [239] Park HR, Furihata K, Hayakawa Y, Shin-ya K. Versipelostatin, a
and greater cytotoxicity than its reversible analogs. J Pharmacol novel GRP78/Bip molecular chaperone down-regulator of micro-
Exp Ther 2011; 337: 479-86. bial origin. Tetrahedron Lett 2002; 43: 6941-5.
[216] Singh AV, Palladino MA, Lloyd GK, Potts BC, Chauhan D, An- [240] Umeda Y, Furihata K, Sakuda S, et al. Absolute structure of prun-
derson KC. Pharmacodynamic and efficacy studies of the novel ustatin A, a novel GRP78 molecular chaperone down-regulator.
proteasome inhibitor NPI0052 (marizomib) in a human plasmacy- Organic Lett 2007; 9: 4239-42.
toma xenograft murine model. Br J Haematol 2010; 149: 550-9. [241] Park HR, Ryoo IJ, Choo SJ, et al. Glucose-deprived HT-29 human
[217] Kupperman E, Lee EC, Cao Y, et al. Evaluation of the proteasome colon carcinoma cells are sensitive to verrucosidin as a GRP78
inhibitor MLN9708 in preclinical models of human cancer. Cancer down-regulator. Toxicol 2007; 229: 253-61.
Res 2010; 70: 1970-80.
Cancer Therapy Current Pharmaceutical Design, 2014, Vol. 20, No. 2 221

[242] Hwang JH, Kim JY, Cha MR, et al. Etoposideresistant HT29 [264] Kanzawa T, Germano IM, Komata T, Ito H, Kondo Y, Kondo S.
human colon carcinoma cells during glucose deprivation are sensi- Role of autophagy in temozolomide-induced cytotoxicity for ma-
tive to piericidin A, a GRP78 downregulator. J Cell Physiol 2008; lignant glioma cells. Cell Death Differ 2004; 11: 448-57.
215: 243-50. [265] Janku F, McConkey DJ, Hong DS, Kurzrock R. Autophagy as a
[243] Choo SJ, Park HR, Ryoo IJ, et al. Deoxyverrucosidin, a novel target for anticancer therapy. Nat Rev Clin Oncol 2011; 8: 528-39.
GRP78/BiP down-regulator, produced by Penicillium sp. J Antibiot [266] Solomon VR, Lee H. Chloroquine and its analogs; a new promise
2005; 58: 210-3. of an old drug for effective and safe cancer therapies. Eur J Phar-
[244] Schönthal AH. (2012) Pharmacological targeting of endoplasmic macol 2009; 625: 220-33.
reticulum stress signalling in cancer. Biochem Pharmacol 2012; 85: [267] De Duve C. Lysosomes revisited. Eur J Biochem 1983; 137: 391-7.
653-66. [268] Castino R, Démoz M, Isidoro C. Destination ‘Lysosome’: a target
[245] Yu DH, Macdonald J, Liu G, et al. Pyrvinium targets the unfolded organelle for tumour cell killing? J Mol Recogn 2003; 16: 337-48.
protein response to hypoglycemia and its anti-tumor activity is en- [269] Borgonovo B, Cocucci E, Racchetti G, Podini P, Bachi A, Meldo-
hanced by combination therapy. PloS one 2008; 3: e3951. lesi J. Regulated exocytosis: a novel, widely expressed system. Nat
[246] Kim JY, Hwang JH, Cha MR, et al. Arctigenin blocks the unfolded Cell Biol 2002; 4: 955-62.
protein response and shows therapeutic antitumor activity. J Cell [270] Jaiswal JK, Andrews NW, Simon SM. Membrane proximal
Physiol 2010; 224: 33-40. lysosomes are the major vesicles responsible for calcium-
[247] Ni M, Zhang Y, Lee AS. Beyond the endoplasmic reticulum: atypi- dependent exocytosis in nonsecretory cells. J Cell Biol 25: 625-35.
cal GPR78 in cell viability, signalling and therapeutic targeting. [271] Kisen GØ, Tessitore L, Costelli P, et al. Reduced autophagic activ-
Biochem J 2011; 434: 181-8. ity in primary rat hepatocellular carcinoma and ascites hepatoma
[248] Wallen E, Sellers RG, Peehl DM. Brefeldin A induces p53- cells. Carcinogenesis 1993; 14: 2501-05.
independent apoptosis in primary cultures of human prostatic can- [272] Fehrenbacher N, Jäättelä M. Lysosomes as targets for cancer ther-
cer cells. J Urol 2000; 164: 836-41. apy. Cancer Res 65: 2993-5.
[249] Shao RG, Shimizu T, Pommier Y. Brefeldin A is a potent inducer [273] Reinheckel T, Deussing J, Roth W, Peters C. Towards specific
of apoptosis in human cancer cells independently of p53. Exp Cell functions of lysosomal cysteine peptidases: phenotypes of mice de-
Res 1996; 227: 190-6. ficient for cathepsin B or cathepsin L. Biol Chem 2001; 382: 735-
[250] Choi JH, Choi AY, Yoon H, et al. Baicalein protects HT22 murine 41.
hippocampal neuronal cells against endoplasmic reticulum stress- [274] Kos J, Lah TT. Cysteine proteinases and their endogenous inhibi-
induced apoptosis through inhibition of reactive oxygen species tors: target proteins for prognosis, diagnosis and therapy in cancer.
production and CHOP induction. Exp Mol Med 2010; 42: 811-22. Oncol Rep 1998; 5: 1349-61.
[251] Denmeade SR, Jakobsen CM, Janssen S, et al. Prostate-specific [275] Turk B, Turk D, Turk V. Lysosomal cysteine proteases: more than
antigen-activated thapsigargin prodrug as targeted therapy for pros- scavengers. Biochimica et Biophysica Acta 2000; 1477: 98-111.
tate cancer. J Natl Cancer Inst 2003; 95: 990-1000. [276] Koblinski JE, Ahram M, Sloane BF. Unraveling the role of prote-
[252] Denmeade SR, Mhaka AM, Rosen DM, et al. Engineering a Pros- ases in cancer. Clin Chim Acta 2000; 291: 113-35.
tate-Specific Membrane Antigen-Activated Tumor Endothelial Cell [277] Joyce JA, Baruch A, Chehade K, et al. Cathepsin cysteine prote-
Prodrug for Cancer Therapy. Sci Transl Med 2012; 4: 140ra86- ases are effectors of invasive growth and angiogenesis during mul-
140ra86. tistage tumorigenesis. Cancer cell 2004; 5: 443-53.
[253] Suh DH, Kim MK, Kim HS, Chung HH, Song YS. Unfolded pro- [278] Lakka SS, Gondi CS, Yanamandra N, et al. Inhibition of cathepsin
tein response to autophagy as a promising druggable target for anti- B and MMP-9 gene expression in glioblastoma cell line via RNA
cancer therapy. Ann N Y Acad Sci 2012; 1271: 20-32. interference reduces tumor cell invasion, tumor growth and angio-
[254] Koki AT, Masferrer JL. Celecoxib: a selective COX-2 inhibitor genesis. Oncogene 2004; 23: 4681-9.
with anticancer properties. Cancer Control 2002; 9: 28-35. [279] Cirman T, Orei K, Mazovec GD, et al. Selective disruption of
[255] Schönthal AH. Direct non-cyclooxygenase-2 targets of celecoxib lysosomes in HeLa cells triggers apoptosis mediated by cleavage of
and their potential relevance for cancer therapy. Br J Cancer 2007; Bid by multiple papain-like lysosomal cathepsins. J Biol Chem
97: 1465-8. 2004; 279: 3578-87.
[256] Dogne JM, Thiry A, Supuran CT. Carbonic anhydrase inhibition: [280] Mousavi SA, Brech A, Berg T, Kjeken R. Phosphoinositide 3-
insight into non-COX-2 pharmacological effect of some coxibs. kinase regulates maturation of lysosomes in rat hepatocytes. Bio-
Curr Pharm Des 2008; 14: 679-84. chem J 2003; 372: 861-9.
[257] Kardosh A, Golden EB, Pyrko P, et al. Aggravated endoplasmic [281] Madge LA, Li JH, Choi J, Pober JS. Inhibition of phosphatidyli-
reticulum stress as a basis for enhanced glioblastoma cell killing by nositol 3-kinase sensitises vascular endothelial cells to cytokine-
bortezomib in combination with celecoxib or its non-coxib ana- initiated cathepsin-dependent apoptosis. J Biol Chem 2003; 278:
logue, 2, 5-dimethyl-celecoxib. Cancer Res 2008; 68: 843-51. 21295-306.
[258] Schönthal AH, Chen TC, Hofman FM, Louie SG, Petasis NA. [282] Nylandsted J, Gyrd-Hansen M, Danielewicz A, et al. Heat shock
Celecoxib analogs that lack COX-2 inhibitory function: preclinical protein 70 promotes cell survival by inhibiting lysosomal mem-
development of novel anticancer drugs. Expert Opin Investig Drugs brane permeabilization. J Exp Med 2004; 200: 425-35.
2008; 17: 197-208. [283] Zang Y, Beard RL, Chandraratna RA, Kang JX. Evidence of a
[259] Tseng PH, Lin HP, Zhu J, et al. Synergistic interactions between lysosomal pathway for apoptosis induced by the synthetic retinoid
imatinib mesylate and the novel phosphoinositide-dependent CD437 in human leukemia HL-60 cells. Cell death and differentia-
kinase-1 inhibitor OSU-03012 in overcoming imatinib mesylate re- tion 2001; 8: 477-85.
sistance. Blood 2005; 105: 4021-7. [284] Neuzil J, Zhao M, Ostermann G, et al. Alpha-tocopheryl succinate,
[260] Tseng PH, Wang YC, Weng SC, et al. Overcoming trastuzumab an agent with in vivo anti-tumour activity, induces apoptosis by
resistance in HER2-overexpressing breast cancer cells by using a causing lysosomal instability. Biochem J 2002; 362: 709-15.
novel celecoxib-derived phosphoinositide-dependent kinase-1 in- [285] Salvayre R, Auge N, Benoist H, Negre-Salvayre A. Oxidized low-
hibitor. Mol Pharmacol 2006; 70: 1534-41. density lipoprotein-induced apoptosis. Biochimica et Biophysica
[261] Weng SC, Kashida Y, Kulp SK, et al. Sensitizing estrogen recep- Acta 2002; 1585: 213-21.
tor-negative breast cancer cells to tamoxifen with OSU-03012, a [286] Biasutto L, Dong L-F, Zoratti M, Neuzil J. Mitochondrially tar-
novel celecoxib-derived phosphoinositide-dependent protein geted anti-cancer agents. Mitochondrion 2010; 10: 670-81.
kinase-1/Akt signaling inhibitor. Mol Cancer Thera 2008; 7: 800-8. [287] Torchilin VP. Recent advances with liposomes as pharmaceutical
[262] Thomas S, Sharma N, Golden EB, et al. Preferential killing of carriers. Nat Rev Drug Discov 2005; 4: 145-60.
triple-negative breast cancer cells in vitro and in vivo when phar- [288] Kale AA, Torchilin VP. Enhanced transfection of tumor cells in
macological aggravators of endoplasmic reticulum stress are com- vivo using “Smart” pH-sensitive TAT-modified pegylated
bined with autophagy inhibitors. Cancer Lett 2012; 325: 63-71. liposomes. J Drug Target 2007; 15: 538-45.
[263] Briceno E, Calderon A, Sotelo J. Institutional experience with [289] Mu L, Elbayoumi TA, Torchilin VP. Mixed micelles made of
chloroquine as an adjuvant to the therapy for glioblastoma multi- poly(ethylene glycol)-phosphatidylethanolamine conjugate and d-
forme. Surg Neurol 2007; 67: 388-91. -tocopheryl polyethylene glycol 1000 succinate as pharmaceutical
nanocarriers for camtothecin. Int J Pharm 2005; 306: 142-9.
222 Current Pharmaceutical Design, 2014, Vol. 20, No. 2 Ubah and Wallace

[290] Weissig V, Cheng SM, D’Souza GG. Mitochondrial pharmaceu- ramide to mitochondria enhances its cytotoxicity in vitro and in
tics. Mitochondrion 2004; 3: 229-44. vivo. Nano Lett 2008; 8: 2559-63.
[291] Wsissig V, Boddapati SV, Cheng SM, D’Souza GG. Liposomes [294] Kogure K, Akita H, Harashima H. Multifunctional envelope-type
and liposome-like vesicles for drug and DNA delivery to mito- nano device for non-viral gene delivery: concept and application of
chondria. J Liposome Res 2006; 16: 249-64. programmed packaging. J Control Release 2007; 122: 246-51.
[292] Boddapati SV, Tongcharoensirikul P, Hanson RN, D'Souza GG, [295] Kogure K, Akita H, Yamada Y, Harashima H. Multifunctional
Torchilin VP, Weissig V. Mitochondriotropic liposomes. J envelope-type nano device (MEND) as a non-viral gene delivery
Liposome Res 2005; 15: 49-58. system. Adv Drug Deliv Rev 2008; 60: 559-71.
[293] Boddapati SV, D’Souza GG, Erdogan S, Torchilin VP, Weissig V. [296] Yamada Y, Harashima H. Mitochondrial drug delivery systems for
Organelle-targeted nanocarriers: specific delivery of liposomal ce- macromolecule and their therapeutic application to mitochondrial
diseases. Adv Drug Del Rev 2008; 60: 1439-62.

Received: March 11, 2013 Accepted: May 15, 2013

You might also like