Accepted Manuscript Title: Mechanisms of tumor resistance against chemotherapeutic agents in veterinary oncology Author: R. Klopfleisch, B. Kohn, A.D. Gruber PII: DOI: Reference:
S1090-0233(15)00272-5 http://dx.doi.org/doi:10.1016/j.tvjl.2015.06.015 YTVJL 4547
To appear in:
The Veterinary Journal
Accepted date:
30-6-2015
Please cite this article as: R. Klopfleisch, B. Kohn, A.D. Gruber, Mechanisms of tumor resistance against chemotherapeutic agents in veterinary oncology, The Veterinary Journal (2015), http://dx.doi.org/doi:10.1016/j.tvjl.2015.06.015. This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
1
Review
2
Mechanisms of tumor resistance against chemotherapeutic agents in veterinary oncology
3
R. Klopfleisch a, B. Kohn b, A. D. Gruber a
4 5 6 7 8 9 10 11
a
Institute of Veterinary Pathology, Freie Universität Berlin, Robert-von-Ostertag-Straße 15, 14163 Berlin, Germany b Small Animal Clinic, Freie Universität Berlin, Oertzenweg 19 b, 14163 Berlin, Germany * Corresponding author. Tel.: +49 30 83862460 E-mail address:
[email protected] (R. Klopfleisch).
1 Page 1 of 36
12 13 14 15 16 17 18 19
Highlights
Chemotherapy is a major treatment modality in veterinary oncology Efficacy of chemotherapy is often impacted by intrinsic/acquired drug resistance Chemotherapy resistance can be due to multiple mechanisms Molecular data generated in humans can be translatable to pets New therapies should account for mechanism of drug resistance Abstract
20
Several classes of chemotherapy drugs are used as first line or adjuvant treatment of the
21
majority of tumour types in veterinary oncology. However, some types of tumour are
22
intrinsically resistant to several anti-cancer drugs and others, while initially sensitive, acquire
23
resistance during treatment. Chemotherapy often significantly prolongs survival or disease
24
free interval, but is not curative. The exact mechanisms behind intrinsic and acquired
25
chemotherapy resistance are unknown for most animal tumours but there is increasing
26
knowledge on the mechanisms of drug resistance in humans and a few reports on molecular
27
changes in resistant canine tumours have emerged. In addition, approaches to overcome or
28
prevent chemotherapy resistance are becoming available in humans and, given the overlaps in
29
molecular alterations between human and animal tumours, these may also be relevant in
30
veterinary oncology.
31 32
This review provides an overview of the current state of research on general
33
chemotherapy resistance mechanisms, including drug efflux, DNA repair, apoptosis evasion
34
and tumour stem cells. The known resistance mechanisms in animal tumours and the potential
35
of these findings for improving treatment efficacy in veterinary oncology are also explored.
36 37
Keywords: Oncology; Chemotherapy; Resistance; Epigenetics; DNA repair; Cancer stem
38
cells; Epithelial mesenchymal transition; Apoptosis
39 2 Page 2 of 36
40
Introduction
41
Chemotherapy is commonly used to treat cancer in pets, and for some tumour types, such
42
as haematopoietic tumours, chemotherapy is the treatment of choice. Complete remission and
43
disease stability can often be achieved with currently available drugs and protocols. However,
44
in a significant percentage of animals, standard protocols are ineffective due to intrinsic
45
resistance of the tumour against available agents. Furthermore, tumour relapse during or after
46
treatment is observed, commonly or sporadically, due to the development of acquired
47
resistance. The mechanisms of resistance and the therapeutic strategies employed to overcome
48
this resistance are important and topical areas for focus in cancer research.
49 50
Chemotherapy research in veterinary oncology is constantly progressing, but many
51
questions on the specific mechanisms of chemoresistance in pets are still unclear. In contrast,
52
numerous studies have been published on the mechanisms of resistance against common anti-
53
cancer drugs in humans and animal models. These studies have shown that the mechanisms of
54
chemoresistance mainly consist of: (1) increased cellular efflux of chemotherapeutic drugs;
55
(2) accelerated drug inactivation or lack of drug activation; (3) changes in drug targets (e.g.
56
mutation and methylation); (4) efficient mechanisms of DNA repair; (5) deregulation of
57
apoptosis, and (6) cancer stem cells as the nucleus of resistance in tumours and epithelial
58
mesenchymal transition (EMT) (Table 1).
59 60
This article provides an overview of the current knowledge of chemotherapeutic
61
resistance and explores the mechanisms that may be relevant and so targeted in pet animals
62
with chemotherapy-resistant tumours.
63 3 Page 3 of 36
64
Increased efflux of chemotherapeutic agents from tumour cells
65
Efflux of drugs from cells is mostly based on membrane transporter proteins. Of these,
66
the ATP-binding cassette (ABC) transporter family is considered to be the most important
67
group of transmembrane proteins which drive the transport of many chemically unrelated
68
drugs across the plasma membrane (Holohan et al., 2013). Of the 49 (or more) members of
69
this family, the multi-drug resistance protein 1 (MDR1, P-glycoprotein [PGP], ABCB1), the
70
MDR-associated protein 1 (MRP1, ABCC1) and the breast cancer resistance protein (BCRP,
71
ABCG2) have been most thoroughly investigated in tumours of not only humans but also of
72
dogs and cats (Fig. 1) (Brenn et al., 2008; Choi and Yu, 2014; Gramer et al., 2013; Honscha et
73
al., 2009; Pawlowski et al., 2013). All three of these proteins, and other members of the
74
family, are capable of eliminating lipids and therefore various classes of hydrophobic
75
chemotherapy drugs,
76
compounds and microtubule inhibitors, as well as the tyrosine kinase inhibitors (TKIs)
77
(Holohan et al., 2013).
including
topoisomerase
inhibitors,
antimetabolites,
platinum
78 79
MDR1 and resistance
80
MDR1 is a membrane-bound, ATP-dependent efflux pump that is overexpressed in many
81
human tumours before the start of any chemotherapy and thus contributes to intrinsic
82
resistance (Goldstein et al., 1989). MDR1 expression is increased or even induced during
83
chemotherapy in various human tumours and contributes to resistance against platinum-
84
containing compounds, topoisomerase II inhibitors, microtubulin poisons and several TKIs
85
(Table 1) (Holohan et al., 2013).
86 87
ABC transporters in animal tumours 4 Page 4 of 36
88
MDR1, MRP1 and BCRP expression has been analyzed in several canine tumours. In
89
most studies the analysis was restricted to the presence of protein or mRNA, to its correlation
90
with the efficacy and outcome of the chemotherapy protocol used, or to general clinical
91
parameters in mammary tumours and lymphomas of dogs (Dhaliwal et al., 2013; Gramer et
92
al., 2013; Tomiyasu et al., 2014a; Zandvliet et al., 2014b).
93 94
In a few studies, the desensitizing effects of MDR1, BCRP, MRP1 activities has been
95
directly confirmed in canine mammary tumour or lymphoma cells (Table 2). For instance,
96
Pawlowski et al. (2013) were able to identify the drug specificity for different ABC
97
transporters in a canine mammary tumour cell line. By siRNA-mediated gene silencing, they
98
showed that: (1) vinblastine efflux was mediated by MDR1 and MRP1; (2) cisplatin efflux
99
was mediated by MDR1, BCRP, MRP1, and (3) cyclophosphamide resistance was mediated
100
by BCRP. In addition, doxorubicin and vincristine resistance in canine lymphoma cell lines
101
was completely reversed by an MDR1 inhibitor (Zandvliet et al., 2014a).
102 103
Only a few studies on the expression of MDR1 in feline tumour cells have been
104
published. MDR1 is constitutively expressed in feline lymphomas, mast cell tumours, lung
105
tumours and squamous cell carcinomas (Hifumi et al., 2010; Van der Heyden et al., 2011).
106
Feline mammary gland tumours showed strong membranous MDR1 labelling, especially in
107
areas with infiltrative growth and in atypical cells, although it was not correlated with the
108
general grade of malignancy of the complete tumour (Van der Heyden et al., 2011). Similarly,
109
MDR1 expression was not predictive of remission or survival time in cats with lymphoma
110
(Brenn et al., 2008). Nevertheless, one study has confirmed the impact of MDR1 on
111
adriamycin and vincristine resistance in feline lymphoma cells (Okai et al., 2000).
5 Page 5 of 36
112 113
ABC transporter modulators to overcome resistance
114
The observation that ABC transporter activity in cancer cells contributes to anti-cancer
115
drug resistance has led to intensive research on agents that either block or inactivate these
116
transporters and so increase the intracellular concentration and effect of the anti-cancer drugs
117
(Fig. 1). The first-generation ABC transporter modulators were the MDR1 inhibitors
118
verapamil, cyclosporine A and quinine (Kathawala et al., 2015). In preclinical studies, these
119
modulators showed promising effects on chemotherapy resistance but these could not be
120
confirmed in clinical trials. The second-generation ABC transporter modulators were
121
designed to increase the effectiveness and decrease toxic side effects. Valspodar, a
122
cyclosporine analogue, and biricodar indeed increased the efficacy of several classical
123
chemotherapy drugs and had decreased toxicity preclinically, but again lacked sufficient
124
efficacy in clinical trials (Nobili et al., 2012).
125 126
The third-generation ABC transporter modulators (elacridar, laniquidar, zosuquidar and
127
tariquidar) were specifically designed to overcome the problems faced by the first- and
128
second-generation modulators. They inhibit MDR1, BCRP and MRP1 function at nM
129
concentrations and are less toxic. In clinical trials, they too failed to affect clinical outcome
130
significantly, which led to the assumption that MDR1 may have less impact on drug
131
resistance or that the functional redundancy between ABC transporters impedes anti-cancer
132
drug efflux inhibition (Holohan et al., 2013).
133 134
The effectiveness of verapamil and cyclosporine A to block the ABC transporter and to
135
overcome chemotherapy resistance has also been analyzed in canine tumour cells. For canine 6 Page 6 of 36
136
mammary tumours, Krol et al. (2014) showed that verapamil and cyclosporine A were unable
137
to reverse resistance against vinblastine in MDR1-expressing canine mammary cancer cells.
138
However, verapamil blocked MDR1 efflux in dermal, but not oral or gastrointestinal mast cell
139
tumour cells (Nakaichi et al., 2007).
140 141
Tyrosine kinase inhibitors to overcome resistance
142
TKIs, including masitinib and the alkylophospholipid perifosine, appear to be promising
143
ABC-transporter inhibitor alternatives. They are capable of reversing ABC transporter-
144
mediated chemotherapy resistance via inhibition of several growth factor signalling pathways.
145
TKI efficacy has been confirmed in canine lymphomas in which masitinib reversed
146
doxorubicin resistance by inhibiting MDR1 (Zandvliet et al., 2013). Likewise, perifosine, an
147
AKT and PI3K inhibitor, has been shown to reverse vincristine in canine lymphoid tumour
148
cells (Tomiyasu et al., 2014b).
149 150
Taken together, numerous modulators of ABC transporters are available and have been
151
tested successfully in vivo, mostly in humans. It will be interesting to test these modulators in
152
canine and feline tumours to evaluate their potential for overcoming chemotherapy resistance
153
in these species.
154 155
Drug inactivation in tumour cells
156
All phases of metabolism of a chemotherapeutic drug in the body (i.e. absorption,
157
distribution, metabolism and excretion) can influence its efficacy on tumour growth. In terms
158
of chemotherapy resistance, recent research has however been mainly focused on intracellular 7 Page 7 of 36
159
drug activation and inactivation in tumour cells. The most important enzymes for drug
160
activation and inactivation include the cytochrome P450 (CYP) system, the glutathione-S-
161
transferase (GST) superfamily and the uridine diphospho-glucuronosyltransferase (UGT)
162
superfamily (Housman et al., 2014).
163 164
Cytochrome P450 (CYP) system
165
Several CYPs are important for drug resistance (Housman et al., 2014). Increased levels
166
of CYP and CYPb5 have been detected in benign canine mammary tumours when compared
167
to more malignant tumours (Kumaraguruparan et al., 2006). In addition, CYP3A12 was
168
identified as a potent enzyme for vinblastine depletion in dogs in an in vitro model (Achanta
169
and Maxwell, 2014).
170 171
Glutathione-S-transferase (GST) superfamily
172
GSTs and metallothionein are relevant for resistance to doxorubicin, alkylating agents
173
and platinum drugs through direct detoxification and inhibition of the mitogen-activated
174
protein kinase (MAPK) pathway (Karotki and Vasak, 2009; Meijer et al., 1992). Increased
175
GST expression is also associated with increased risk of lymphoma development and
176
resistance to a modified University of Wisconsin-Madison protocol in canine lymphoma
177
expression (Ginn et al., 2014; Tomiyasu et al., 2010), while increased plasma GST
178
concentrations have been found in dogs with relapsing lymphomas (Hahn et al., 1999).
179 180
In canine urinary bladder transitional cell carcinoma, the expression level of GST did not
181
correlate with survival time under cisplatin, doxorubicin and mitoxantrone treatment (Rocha
8 Page 8 of 36
182
et al., 2000). In vitro, GST-mediated resistance in canine osteosarcoma cells against cisplatin
183
could be reversed by ethacrynic acid treatment (Shoieb et al., 1998).
184 185
Platinum drug resistance can also occur through inactivation by metallothionein (Karotki
186
and Vasak, 2009; Meijer et al., 1992). Expression of metallothionein has been demonstrated
187
in canine and feline lung and melanocytic tumours, in canine cutaneous apocrine gland and in
188
canine mammary tumours (Dincer et al., 2001; Erginsoy et al., 2006; Hifumi et al., 2010;
189
Martano et al., 2012). The effect of metallothionein on platinum drug resistance has not been
190
analyzed so far in cats and dogs.
191 192
Uridine diphospho-glucuronosyltransferase (UGT) superfamily
193
The UGTs catalyse glucuronidation and regulate the formation of inactive hydrophilic
194
glucuronides of cytotoxic drugs. A widespread down-regulation of UGT1A1 transcription and
195
microsomal activity occurs in some cancers partly by epigenetic regulation (Michael and
196
Doherty, 2005). Studies on the relevance of UGT on canine or feline cancer and
197
chemotherapy resistance are lacking.
198 199
In summary, several studies indicate that effective detoxification is associated with
200
chemotherapy resistance in canine tumours. Similar mechanisms are present in human
201
tumours and have been targeted successfully using innovative approaches. A similar success
202
in veterinary oncology can be expected in the mid-term range and should be explored in detail
203
to increase the efficacy of some therapeutic interventions in pets.
204
9 Page 9 of 36
205
Changes in drug targets: Mutation and methylation
206
Quantitative and qualitative changes of drug targets represent well-known mechanisms of
207
chemotherapy resistance in human oncology. These changes can be quantitative (with
208
decreased or lost expression of the target), or a compensation of the inhibited target by other
209
proteins in the cells. Another response to the pressure of chemotherapeutic drugs is the
210
selection of tumour cells expressing a qualitatively different, mutated version of the drug
211
target, with, for instance, structural changes at the regular binding site of the drug. Research
212
on chemotherapy resistance due to changes in human drug targets is extensive and first hints
213
towards changes in drug targets in canine lymphomas and mast cell tumours have emerged.
214 215
Mutation and tyrosine kinase inhibitors
216
Loss of sensitivity of tumours towards the effects of TKIs is currently the only example
217
of genetic (e.g. mutational) adaption of tumours due to selective pressure by chemotherapy in
218
veterinary oncology. TKIs such as toceranib and masitinib are commonly used and are
219
initially very effective anti-cancer drugs for canine mast cell tumours. They target several
220
tyrosine kinases but are thought to mainly act through inhibition of the KIT receptor in mast
221
cell tumour cells (London, 2013). One of the major drawbacks of these two TKIs, besides
222
their toxicity, is the large fraction of dogs with relapsing TKI-resistant tumours under or after
223
treatment (London, 2013).
224 225
In vitro studies indicate that either overexpression or de novo expression of alternative
226
proliferative pathways under TKI treatment, such as T- and B-cell receptor signalling, ERK
227
signalling and overexpression of KIT, may contribute to TKI resistance (Klopfleisch et al.,
228
2012; Kobayashi et al., 2015). However, two studies identified additional KIT mutations in 10 Page 10 of 36
229
imatinib- and toceranib-resistant canine mast cell tumour cell lines, (Halsey et al., 2014;
230
Kobayashi et al., 2015).
231 232
Epigenetics and resistance
233
Besides genetic alterations, epigenetic changes may also contribute to chemotherapy
234
resistance (Brown et al., 2014). Epigenetic changes are defined as changes in gene expression
235
that are independent of changes in DNA sequence and that persist over many cell divisions
236
(Wilting and Dannenberg, 2012). These changes mainly consist of covalent modifications of
237
DNA and histones which modulate gene expression levels directly by promotor methylation
238
or indirectly by chromatin packaging, thereby regulating the accessibility of DNA to
239
sequence-specific transcription factors (van Steensel, 2011). DNA is predominantly
240
methylated at cytosines by DNA methyltransferases (DNMTs) in the CPG islands of gene
241
promotor regions. Initially, the hypothesis that epigenetics may be of relevance for
242
chemotherapy resistance was based on the observation that acquisition of resistance often has
243
a rapid kinetic, is reversible and lacks genetic mutations (Brown et al., 2014).
244 245
Epigenetic states favouring chemotherapy resistance have been associated with aberrant
246
transcription of efflux transporters, DNA repair enzymes and apoptotic factors in human
247
tumours (Wilting and Dannenberg, 2012). Only a few studies are available on methylation-
248
associated changes in canine tumours and these mainly focus on the methylation status of the
249
promotor regions of the tumour suppressor p16 and ABC transporters.
250 251
Epigenetic p16 inactivation seems to be a rare in canine T cell lymphoma and affects the
252
prognosis of dogs with high-grade lymphoma (Fosmire et al., 2007; Fujiwara-Igarashi et al., 11 Page 11 of 36
253
2014a,b). DNA methylation and histone H3 acetylation are involved in ABCB1 gene
254
expression and associated with a multi-drug resistant (MDR) phenotype in most canine
255
lymphoid tumour cell lines (Tomiyasu et al., 2014a). Other mechanisms including JNK,
256
MAPK/ERK and PKC signalling seem to be more relevant for ABC transporter expression
257
levels in canine lymphomas (Tomiyasu et al., 2014b, d).
258 259
Several drugs that can reverse epigenetic changes in tumours have been identified. Of
260
these, DNMT inhibitors (DNMTis) and histone deacetylase (HDAC) inhibitors (HDACis)
261
have been most intensively studied in vitro and in clinical trials in humans (Nebbioso et al.,
262
2012). Common HDACis are hydroxamic acids (trichostatin A), cyclic tetrapeptides (trapoxin
263
A and B, romidepsin), short chain and aromatic fatty acids (butyrate, valproic acid) and
264
benzamides (entinostat, mecetinostat), most of which are currently being tested in in humans
265
(Nebbioso et al., 2012). DNMTis include nucleoside analogues (azacytidine, decitabine,
266
zebularine), which covalently bind DNMT, and small molecule inhibitors (hydralazine,
267
procainamide), which competitively bind to CPG-rich DNA regions (Nebbioso et al., 2012).
268 269
The potential of these epigenetic drugs has been shown in clinical trials for several
270
human tumours as both monotherapy or combined with other anti-cancer drugs (Nebbioso et
271
al., 2012). A major drawback of the drugs is their lack of specificity for specific DNA regions
272
of interest, which leads to unwanted demethylation of unrelated or unwanted genes including
273
drug resistance genes (Glasspool et al., 2014).
274 275
Epigenetics in canine cancer chemotherapy resistance
12 Page 12 of 36
276
Pilot studies indicate that epigenetic drugs may also be beneficial in dogs. Smallwood et
277
al. (2014) showed that L-asparaginase sensitivity is strongly and negatively correlated with
278
the level of methylation of the asparagine synthetase (ASNS) promoter in canine lymphoma
279
cells. Furthermore, treatment with hypomethylating azacytidine increased resistance to L-
280
asparaginase. However, ASNS methylation and expression was not predictive for overall
281
survival (OS) or progression-free survival (PFS) in dogs with lymphoma treated with L-
282
asparaginase, indicating that resistance mechanisms in vivo may be more complex
283
(Smallwood et al., 2014).
284 285
The efficacy of azacytidine and trichostatin A on chemotherapy reversal has also been
286
shown for MDR1-based vincristine resistance in canine lymphoma cells (Tomiyasu et al.,
287
2014c) and in dogs with invasive urothelial carcinoma treated with azacitidine (Hahn et al.,
288
2012). Finally, hydroxamic acid and zebularine treatment of canine osteosarcoma cells
289
restored microRNA expression at the 14q32 locus and stabilized expression of cMYC,
290
suggesting a potential benefit as an adjuvant treatment of rapidly progressive osteosarcomas
291
(Thayanithy et al., 2012)
292 293
Increased DNA damage repair
294
Inefficient DNA damage repair systems are a major mechanism of carcinogenesis. For
295
instance, dysfunctional p53 activity or inherited mutations in the BRCA1 or -2 genes, both
296
important regulators of genomic integrity, are important risk factors for the development of
297
several cancer types in humans and are also thought to be relevant for canine and feline
298
tumours (Grosse et al., 2014).
299 13 Page 13 of 36
300
DNA damage response and resistance
301
A more differentiated view on the desirability of an intact DNA damage response (DDR)
302
is however necessary in terms of chemotherapy resistance (Fig. 2). Direct or indirect
303
induction of DNA damage is the main mechanism of action for platinum drugs, alkylating
304
agents and topoisomerase inhibitors (Holohan et al., 2013). The favoured result of this
305
treatment is tumour cell death due to overwhelming DNA damage. This requires either direct
306
breakdown of cellular metabolism due to massive DNA damage or apoptosis induction via
307
p53 and p21. Loss of function mutations of p53 therefore leads to insensitivity of the tumour
308
against induced DNA-damage, since the DNA damage signal is not submitted to the intrinsic
309
apoptosis pathway to induce apoptosis (Enoch and Norbury, 1995). Highly effective DDR is
310
nevertheless also a cause of resistance because it allows tumour cells to repair and survive
311
induced DNA damage (Bouwman and Jonkers, 2012).
312 313
Inhibition of DNA damage response
314
Inhibition of DDR together with the administration of DNA damaging agents is therefore
315
an accepted, albeit somewhat counterintuitive, therapeutic strategy. This therapeutic approach
316
takes advantage of the fact that although DNA damage may drive carcinogenesis, tumour
317
cells nevertheless require some level of genetic stability to perform basic metabolism.
318
Fortunately, tumour cells often depend on only one remaining DDR pathway, while in normal
319
cells all six DDR pathways (i.e. mismatch repair [MMR], nucleotide excision repair [NER],
320
base excision repair [BER], homologous recombination [HR], inter-strand cross-link repair
321
[ICL] and the non-homologous end-joining or NHEJ) redundantly provide optimal DDR.
322
Inhibition of the remaining pathway in tumour cells thus markedly increases the sensitivity of
323
tumour cells against DNA-damaging agents (Bouwman and Jonkers, 2012).
324 14 Page 14 of 36
325
The most prominent examples of DDR targeting are inhibitors of the single strand-break
326
DNA repair enzyme poly-ADP-ribose polymerase (PARP) such as olaparib. PARP inhibitors
327
cause lethality in tumour cells with mutations in the BRCA1 and BRCA2 genes. Both genes
328
are essential for effective homologous recombination DNA repair. Loss of homologous
329
recombination is normally compensated by single strand repair mechanisms, which in turn are
330
dependent on PARP function. Blocking PARP leads to cell death due to overwhelming
331
genetic instability in rapidly dividing tumour cells (Farmer et al., 2005). Ironically, treatment
332
of BRCA2-mutated breast cancer with PARP inhibitors may also lead to resistance to PARP
333
inhibitors due to BRCA2 mutations, which restore the function of the protein and thus the
334
primary genetic starting point of the tumour (Edwards et al., 2008).
335 336
Data on the influence of PARP and its inhibition in pet cancers are not available.
337
However, several studies have focused on BRCA expression and sequence mainly in canine
338
mammary tumours. Several BRCA1 and BRCA2 single nucleotide polymorphisms (SNPs)
339
have been detected and proposed to be relevant for canine mammary tumour development,
340
but the actual biological relevance for tumour development (as in hereditary human breast
341
cancer) has not been proven (Klopfleisch et al., 2011b). BRCA1 expression levels however
342
correlate with malignancy for canine mammary tumours, but not with chemotherapeutic
343
outcome (Klopfleisch and Gruber, 2009; Klopfleisch et al., 2010; Nieto et al., 2003).
344 345
A defective activation of the p53 DNA damage signalling has been described as the cause
346
of doxorubicin, mitoxantrone and vincristine resistance in feline mammary carcinoma stem
347
cells (Pang et al., 2013) and DNA damage repair deficiency may be responsible for
348
lymphoma development in Golden Retrievers (Thamm et al., 2013).
15 Page 15 of 36
349 350
Mismatch repair system and resistance
351
Treatment of mismatch repair (MMR) system-deficient tumours with methotrexate is
352
another promising approach currently tested in humans. MMR genes including MutL homolog
353
1 (MLH1) and MutS protein homolog 2 and 6 (MSH2, MSH6) correct DNA damage caused by
354
DNA polymerase errors. Deficiency of these genes is associated with resistance to DNA
355
damaging agents like cisplatin and carboplatin, which can be reversed with methotrexate
356
(Martin et al., 2009). MLH1, MSH2, and MSH6 expression is strong in canine mast cell
357
tumours and feline intestinal lymphoma (Aberdein et al., 2012; Munday et al., 2009). Of note,
358
the effect of methotrexate as rescue drug has not been evaluated in pets, but may be
359
unknowingly used since it is part of an established multi-agent chemotherapeutic protocol for
360
canine lymphoma (Simon et al., 2008).
361 362
Taken together, although several similarities with human tumours in terms of protein
363
expression exist, only few studies using canine tumour samples have addressed the
364
importance of altered DDR pathways in tumour progression and none in chemotherapy
365
resistance, thus leaving many questions to be addressed in the future (Grosse et al., 2014;
366
Klopfleisch et al., 2011a; Klose et al., 2011).
367 368
Apoptosis deregulation
369
If a chemotherapeutic agent has reached sufficient concentration in the tumour cell and
370
induced DNA damage, the success of treatment depends on the downstream reaction of the
371
cells. This reaction is expected to be cell death, usually inflicted by energy-dependent
372
apoptosis rather than necrosis. One of the classic hallmarks of cancer cells is however 16 Page 16 of 36
373
apoptosis deregulation, and chemotherapy may thus fail despite unchanged efflux and
374
sensitive drug targets in the tumour cell (Hanahan and Weinberg, 2000).
375 376
The accentuated anti-apoptotic status of tumour cells is often based on only few
377
dysregulated genes, which make the development of drugs targeting their gene products an
378
attractive goal. Bcl-2 and tumour necrosis factor (TNF)-related apoptosis-inducing ligand
379
(TRAIL) are the most intensely studied proteins of the numerous pro- and anti-apoptotic
380
proteins known (Holohan et al., 2013). Overexpression of the anti-apoptotic BCL-2 is able to
381
block the intrinsic mitochondria-associated pathway of apoptosis and induces resistance to
382
cytotoxic agents (Miyashita and Reed, 1992). Compounds like navitoclax, which antagonizes
383
pro-apoptotic Bcl-2 and promotes the pro-apoptotic BAX and BAK, have been developed to
384
shift tumour cells towards a more pro-apoptotic state when treated with cytotoxic agents
385
(Konopleva et al., 2006). Another clinically promising approach is recombinant TRAIL and
386
TRAIL receptor-activating antibodies to stimulate the extrinsic apoptosis pathway (Pavet et
387
al., 2011).
388 389
Apoptosis with chemotherapy in animals
390
The apoptosis-inducing effect has been confirmed for nearly all cytotoxic agents
391
currently used in veterinary oncology (Pawlak et al., 2014). In addition, the induction of
392
apoptosis is a standard read out for the evaluation of new anti-cancer compounds in in vitro
393
assays of canine and feline tumours (Pang et al., 2014). Available data on apoptosis
394
dysfunction as a basis of chemotherapy resistance in veterinary oncology are however sparse.
395
17 Page 17 of 36
396
One study on prognostic factors for radiotherapy success for canine lymphoma found
397
increased levels of the anti-apoptotic protein survivin before treatment as associated with an
398
unfavourable prognosis (Fu et al., 2014). Survivin was also identified as a negative prognostic
399
factor for early treatment failure of canine lymphoma treated with a CHOP-based protocol
400
(Rebhun et al., 2008). Furthermore, expression level of myeloid cell leucaemia sequence 1
401
(MCL1), a potent anti-apoptotic protein associated with drug resistance in various human
402
cancers, is increased when canine mast cells are exposed to specific inhibitors of mitogen-
403
activated protein kinase (MAPK) or Janus kinase-signalling pathways, but not when KIT is
404
inhibited. MCL1 expression may therefore contribute to mast cell tumour survival and drug
405
resistance at least in some chemotherapy protocols (Amagai et al., 2013).
406 407
Together these three studies indirectly indicate that an accentuated anti-apoptotic status
408
may also be an inherited or acquired mechanism of chemotherapy resistance in canine
409
tumours and that therapies involving direct targeting of canine tumours may be promising.
410 411
Cancer stem cells and epithelial mesenchymal transition (EMT)
412
Increasing evidence points towards cancer stem cells (CSC) to be of central relevance for
413
chemoresistance of tumours. CSC or tumour-initiating cells (TICs) resemble in many aspects
414
‘normal’ stem cells but are not necessarily derived from them. Normal stem cells have
415
multiple mechanisms to protect them from cytotoxic insults. These include highly active drug-
416
efflux pumps, increased detoxification enzyme levels, enhanced DNA repair efficacy,
417
apoptosis resistance and quiescence. CSCs also have all of these features, which make them
418
an area of focus for the understanding and treatment of chemoresistance (Fig. 3) (Colak and
419
Medema, 2014; Pang and Argyle, 2014). 18 Page 18 of 36
420 421
Cancer stem cell-specific therapy targets
422
The phenotypic features and growth pattern of CSCs are based on the activity of few
423
signalling pathways that regulate the balance between self-renewal and differentiation. The
424
Wnt, Notch and Hedgehog (HH) signalling pathways have been identified as essential for the
425
CSC phenotype of several solid and haematopoietic tumours (Dawood et al., 2014; Duncan et
426
al., 2005; Lai et al., 2003; Yoshimoto et al., 2011). In contrast, bone morphogenetic protein
427
(BMP) signalling inhibits stem cell expansion by suppression of Wnt signalling, induces CSC
428
differentiation, increases sensitivity to chemotherapy in vivo, and therefore seems an
429
promising therapeutic approach for CSC-based resistance (He et al., 2004; Lombardo et al.,
430
2011).
431 432
Targeting the Wnt, Notch and HHS pathways is an intensely studied approach to
433
overcome chemotherapy resistance due to CSCs (Fig. 3). Cyclopamine, an HHS pathway
434
inhibitor, has been shown to increase sensitivity against TKIs and to increase survival of mice
435
in a chronic myeloid leucaemia model (Qiu et al., 2013; Zhao et al., 2009). Furthermore,
436
inhibition of the Notch pathway with antibodies against its ligands and receptor or gamma-
437
secretase inhibitors has also been successfully applied to reduce the population of breast and
438
glioblastoma CSCs and to increase the sensitivity against of taxanes (Fan et al., 2010; Hoey et
439
al., 2009). Common Wnt signalling inhibitors are non-steroidal anti-inflammatory drugs
440
(NSAIDs), the beta-catenin antagonist ICG-001 and biological inhibitors, such as antibodies,
441
RNA interference, and recombinant proteins (Takahashi-Yanaga and Kahn, 2010). So far
442
those have not been tested in canine or feline tumours.
443 444
Cancer stem cells and epithelial-mesenchymal transition 19 Page 19 of 36
445
The interleukin 8 (IL-8) signalling and transforming growth factor‑β receptor (TGFβR)
446
pathways are stem cell-associated pathways (Liu et al., 2011). Recent evidence indicates that
447
IL-8 can induce a state of ‘stemness’ by effecting EMT, which is needed to acquire stem cell
448
characteristics at least for epithelial tumour cells (Fernando et al., 2011). EMT in general may
449
be one potential mechanism for explaining how CSCs develop from epithelial cells. During
450
EMT, epithelial cells develop a mesenchymal phenotype, which is characterized by a loss of
451
polarization and tight cell–cell junctions and a switch to fibroblast-like cell shape.
452
Chemotherapy is assumed to force EMT of tumour cells. For instance, therapy with and
453
resistance against EGFR inhibitors is tightly associated with EMT in vitro and in vivo (Byers
454
et al., 2013; Fuchs et al., 2008). Mutations in the downstream effectors of the receptor with
455
consecutive activation of TGFβR signalling were identified as potential molecular mediators
456
of both EMT and chemotherapy resistance (Huang et al., 2012).
457 458
Drugs inhibiting IL-8 signalling are thought to halt EMT and thus to prevent development
459
of stem cell characteristics by differentiated tumour cells (Fernando et al., 2011). For instance,
460
repertaxin, a non-competitive inhibitor of IL-8 signalling, reduces the number and activity of
461
cancer stem cells and increases the efficacy of docetaxel in breast cancer cells (Ginestier et
462
al., 2010) but has not been tested in veterinary oncology.
463 464
Stem cells are also characterized by a high DNA repair activity and resistance to
465
apoptosis induction by anti-cancer drugs and radiation (Baumann et al., 2008; Bertolini et al.,
466
2009; Colak et al., 2014). This efficient DNA repair may also be their Achilles heel and an
467
interesting approach to target CSCs. For example, inhibition of the activity DNA repair genes
468
with the cell cycle checkpoint inhibitor staurosporin drastically increases the sensitivity of
469
lung and glioblastoma CSCs towards irradiation and chemotherapy (Bao et al., 2006; Signore
470
et al., 2014). 20 Page 20 of 36
471 472
Immunotoxins directly targeting surface markers is another approach of directly targeting
473
CSCs (Fig. 3). Antibodies against the stem cell marker CD133 conjugated to paclitaxel and
474
oncolytic CD133-specific measles viruses have been successfully tested in vitro and in mouse
475
models (Bach et al., 2013; Swaminathan et al., 2013). Finally, direct apoptosis induction via
476
caspase 9 activation efficiently kills colon CSCs, while inhibition of the anti-apoptotic
477
proteins BCL2, BCLXL and BCLW by small molecule inhibitors shifts the balance a more
478
pro-apoptotic state in tumour cells to overcome the anti-apoptotic state in dasatinib-resistant
479
chronic myeloic CSCs in vivo (Goff et al., 2013; Kemper et al., 2012).
480 481
One of the major challenges of targeting CSCs is their ability to switch into a state of
482
non-proliferative quiescence. Uncontrolled and rapid proliferation is a common feature of
483
tumour cells, but of only few non-neoplastic cells (Colak and Medema, 2014). Classic
484
chemotherapeutic agents were developed to target highly proliferative tumour cells but not
485
quiescent cells. Quiescent CSCs are thus believed to be a major cause for tumour relapse after
486
treatment with microtubule inhibitors or DNA damaging drugs.
487 488
Besides their lack of proliferation, quiescent CSCs also have an increased potential to
489
repair DNA damage induced by DNA-damaging agents or radiotherapy (Bao et al., 2006).
490
The rationale to target quiescent CSCs is to force them back into a proliferative state and to
491
overcome their efficient DNA damage repair systems. Initial clinical trials have shown that
492
administration of granulocyte colony-stimulating factor (G-CSF) induces proliferation of
493
quiescent CSCs and reverses imatinib and cytarabine resistance in acute myeloid leucaemia
494
(Fig. 3) (Lowenberg et al., 2003; Pabst et al., 2012).
495
21 Page 21 of 36
496
Research on CSCs is still a mostly uncharted territory in veterinary oncology. Moreover,
497
many questions remain on the characteristics of these cells in non-human species. The
498
relevance of these cells for chemotherapy resistance, however, is undoubtedly an important
499
area to address in order to increase success of chemotherapies in veterinary oncology.
500 501
Conclusions
502
There is significant progress in the understanding of the general mechanisms of
503
chemotherapy resistance of tumours. Strategies and drugs to overcome or prevent these
504
mechanisms are already available or in development and should be of use in veterinary
505
medicine. Increasing the efficacy of existing classic drugs remains of particular relevance in
506
veterinary medicine, since slow progress in elucidating the basic molecular mechanisms of
507
carcinogenesis of animal tumours is hampering the development of new tumour therapies.
508 509 510 511
Conflict of interest The authors of this paper have no financial or personal relationship with other people or organizations that could inappropriately influence or bias the content of the paper.
512 513 514
Acknowledgment The review was supported by the Deutsche Forschungsgemeinschaft DFG KL2240-1.
515 516
References
517 518 519
Aberdein, D., Munday, J.S., Howe, L., French, A.F., Gibson, I.R., 2012. Widespread mismatch repair expression in feline small intestinal lymphomas. Journal of Comparative Pathology 147, 24-30.
520 521
Achanta, S., Maxwell, L.K., 2014. Reaction phenotyping of vinblastine metabolism in dogs. Veterinary and Comparative Oncology DOI: 10.1111/vco.12084
22 Page 22 of 36
522 523 524
Alagoz, M., Gilbert, D.C., El-Khamisy, S., Chalmers, A.J., 2012. DNA repair and resistance to topoisomerase I inhibitors: mechanisms, biomarkers and therapeutic targets. Current Medicinal Chemistry 19, 3874-3885.
525 526 527
Amagai, Y., Tanaka, A., Matsuda, A., Oida, K., Jung, K., Nishikawa, S., Jang, H., Ishizaka, S., Matsuda, H., 2013. Increased expression of the antiapoptotic protein MCL1 in canine mast cell tumors. The Journal of Veterinary Medical Science 75, 971-974.
528 529 530
Arlt, A., Gehrz, A., Muerkoster, S., Vorndamm, J., Kruse, M.L., Folsch, U.R., Schafer, H., 2003. Role of NF-kappaB and Akt/PI3K in the resistance of pancreatic carcinoma cell lines against gemcitabine-induced cell death. Oncogene 22, 3243-3251.
531 532 533
Bach, P., Abel, T., Hoffmann, C., Gal, Z., Braun, G., Voelker, I., Ball, C.R., Johnston, I.C., Lauer, U.M., Herold-Mende, C., et al., 2013. Specific elimination of CD133+ tumor cells with targeted oncolytic measles virus. Cancer Research 73, 865-874.
534 535 536
Bao, S., Wu, Q., McLendon, R.E., Hao, Y., Shi, Q., Hjelmeland, A.B., Dewhirst, M.W., Bigner, D.D., Rich, J.N., 2006. Glioma stem cells promote radioresistance by preferential activation of the DNA damage response. Nature 444, 756-760.
537 538 539 540
Bardenheuer, W., Lehmberg, K., Rattmann, I., Brueckner, A., Schneider, A., Sorg, U.R., Seeber, S., Moritz, T., Flasshove, M., 2005. Resistance to cytarabine and gemcitabine and in vitro selection of transduced cells after retroviral expression of cytidine deaminase in human hematopoietic progenitor cells. Leukemia 19, 2281-2288.
541 542
Baumann, M., Krause, M., Hill, R., 2008. Exploring the role of cancer stem cells in radioresistance. Nature reviews. Cancer 8, 545-554.
543 544 545
Beretta, G.L., Gatti, L., Perego, P., Zaffaroni, N., 2013. Camptothecin resistance in cancer: insights into the molecular mechanisms of a DNA-damaging drug. Current Medicinal Chemistry 20, 1541-1565.
546 547 548 549
Bertolini, G., Roz, L., Perego, P., Tortoreto, M., Fontanella, E., Gatti, L., Pratesi, G., Fabbri, A., Andriani, F., Tinelli, S., et al., 2009. Highly tumorigenic lung cancer CD133+ cells display stem-like features and are spared by cisplatin treatment. Proceedings of the National Academy of Sciences of the United States of America 106, 16281-16286.
550 551
Bouwman, P., Jonkers, J., 2012. The effects of deregulated DNA damage signalling on cancer chemotherapy response and resistance. Nature Reviews Cancer 12, 587-598.
552 553 554
Brenn, S.H., Couto, S.S., Craft, D.M., Leung, C., Bergman, P.J., 2008. Evaluation of Pglycoprotein expression in feline lymphoma and correlation with clinical outcome. Veterinary and Comparative Oncology 6, 201-211.
555 556
Brown, R., Curry, E., Magnani, L., Wilhelm-Benartzi, C.S., Borley, J., 2014. Poised epigenetic states and acquired drug resistance in cancer. Nature Reviews Cancer 14, 747-753.
557 558 559 560
Byers, L.A., Diao, L., Wang, J., Saintigny, P., Girard, L., Peyton, M., Shen, L., Fan, Y., Giri, U., Tumula, P.K., et al., 2013. An epithelial-mesenchymal transition gene signature predicts resistance to EGFR and PI3K inhibitors and identifies Axl as a therapeutic target for overcoming EGFR inhibitor resistance. Clinical Cancer Research 19, 279-290. 23 Page 23 of 36
561 562 563
Cai, J., Damaraju, V.L., Groulx, N., Mowles, D., Peng, Y., Robins, M.J., Cass, C.E., Gros, P., 2008. Two distinct molecular mechanisms underlying cytarabine resistance in human leukemic cells. Cancer Research 68, 2349-2357.
564 565 566 567
Chien, W.W., Le Beux, C., Rachinel, N., Julien, M., Lacroix, C.E., Allas, S., Sahakian, P., Cornut-Thibaut, A., Lionnard, L., Kucharczak, J., Aouacheria, A., Abribat, T., Salles, G., 2015. Differential mechanisms of asparaginase resistance in B-type acute lymphoblastic leukemia and malignant natural killer cell lines. Scientific Reports 5, 8068.
568 569
Choi, Y.H., Yu, A.M., 2014. ABC transporters in multidrug resistance and pharmacokinetics, and strategies for drug development. Current Pharmaceutical Design 20, 793-807.
570 571 572 573
Chu, X.Y., Kato, Y., Niinuma, K., Sudo, K.I., Hakusui, H., Sugiyama, Y., 1997. Multispecific organic anion transporter is responsible for the biliary excretion of the camptothecin derivative irinotecan and its metabolites in rats. The Journal of Pharmacology and Experimental Therapeutics 281, 304-314.
574 575
Colak, S., Medema, J.P., 2014. Cancer stem cells--important players in tumor therapy resistance. The FEBS Journal 281, 4779-4791.
576 577 578
Colak, S., Zimberlin, C.D., Fessler, E., Hogdal, L., Prasetyanti, P.R., Grandela, C.M., Letai, A., Medema, J.P., 2014. Decreased mitochondrial priming determines chemoresistance of colon cancer stem cells. Cell Death and Differentiation 21, 1170-1177.
579 580
Dawood, S., Austin, L., Cristofanilli, M., 2014. Cancer Stem Cells: Implications for Cancer Therapy. Oncology 28.
581 582 583
Dhaliwal, R.S., Kitchell, B.E., Ehrhart, E., Valli, V.E., Dervisis, N.G., 2013. Clinicopathologic significance of histologic grade, pgp, and p53 expression in canine lymphoma. Journal of the American Animal Hospital Association 49, 175-184.
584 585 586
Dincer, Z., Jasani, B., Haywood, S., Mullins, J.E., Fuentealba, I.C., 2001. Metallothionein expression in canine and feline mammary and melanotic tumours. Journal of Comparative Pathology 125, 130-136.
587 588 589
Duncan, A.W., Rattis, F.M., DiMascio, L.N., Congdon, K.L., Pazianos, G., Zhao, C., Yoon, K., Cook, J.M., Willert, K., Gaiano, N., Reya, T., 2005. Integration of Notch and Wnt signaling in hematopoietic stem cell maintenance. Nature Immunology 6, 314-322.
590 591 592
Edwards, S.L., Brough, R., Lord, C.J., Natrajan, R., Vatcheva, R., Levine, D.A., Boyd, J., Reis-Filho, J.S., Ashworth, A., 2008. Resistance to therapy caused by intragenic deletion in BRCA2. Nature 451, 1111-1115.
593 594
Enoch, T., Norbury, C., 1995. Cellular responses to DNA damage: cell-cycle checkpoints, apoptosis and the roles of p53 and ATM. Trends in Biochemical Sciences 20, 426-430.
595 596 597
Erginsoy, S.D., Sozmen, M., Caldin, M., Furlanello, T., 2006. Metallothionein expression in benign and malignant canine mammary gland tumours. Research in Veterinary Science 81, 46-50.
598 599
Fan, X., Khaki, L., Zhu, T.S., Soules, M.E., Talsma, C.E., Gul, N., Koh, C., Zhang, J., Li, Y.M., Maciaczyk, J., et al., 2010. NOTCH pathway blockade depletes CD133-positive 24 Page 24 of 36
600 601
glioblastoma cells and inhibits growth of tumor neurospheres and xenografts. Stem Cells 28, 5-16.
602 603 604
Farmer, H., McCabe, N., Lord, C.J., Tutt, A.N., Johnson, D.A., Richardson, T.B., Santarosa, M., Dillon, K.J., Hickson, I., Knights, C., et al., 2005. Targeting the DNA repair defect in BRCA mutant cells as a therapeutic strategy. Nature 434, 917-921.
605 606 607
Fernando, R.I., Castillo, M.D., Litzinger, M., Hamilton, D.H., Palena, C., 2011. IL-8 signaling plays a critical role in the epithelial-mesenchymal transition of human carcinoma cells. Cancer Research 71, 5296-5306.
608 609
Fink, D., Aebi, S., Howell, S.B., 1998. The role of DNA mismatch repair in drug resistance. Clinical Cancer Research 4, 1-6.
610 611 612 613
Fosmire, S.P., Thomas, R., Jubala, C.M., Wojcieszyn, J.W., Valli, V.E., Getzy, D.M., Smith, T.L., Gardner, L.A., Ritt, M.G., Bell, J.S., et al., 2007. Inactivation of the p16 cyclindependent kinase inhibitor in high-grade canine non-Hodgkin's T-cell lymphoma. Veterinary Pathology 44, 467-478.
614 615 616
Fu, D.R., Kato, D., Watabe, A., Endo, Y., Kadosawa, T., 2014. Prognostic utility of apoptosis index, Ki-67 and survivin expression in dogs with nasal carcinoma treated with orthovoltage radiation therapy. The Journal of Veterinary Medical Science 76, 1505-1512.
617 618 619 620
Fuchs, B.C., Fujii, T., Dorfman, J.D., Goodwin, J.M., Zhu, A.X., Lanuti, M., Tanabe, K.K., 2008. Epithelial-to-mesenchymal transition and integrin-linked kinase mediate sensitivity to epidermal growth factor receptor inhibition in human hepatoma cells. Cancer Research 68, 2391-2399.
621 622 623
Fujiwara-Igarashi, A., Goto-Koshino, Y., Mochizuki, H., Sato, M., Fujino, Y., Ohno, K., Tsujimoto, H., 2014a. Inhibition of p16 tumor suppressor gene expression via promoter hypermethylation in canine lymphoid tumor cells. Research in Veterinary Science 97, 60-63.
624 625 626 627
Fujiwara-Igarashi, A., Goto-Koshino, Y., Sato, M., Maeda, S., Igarashi, H., Takahashi, M., Fujino, Y., Ohno, K., Tsujimoto, H., 2014b. Prognostic significance of the expression levels of the p16, p15, and p14 genes in dogs with high-grade lymphoma. The Veterinary Journal 199, 236-244.
628 629 630 631
Ginestier, C., Liu, S., Diebel, M.E., Korkaya, H., Luo, M., Brown, M., Wicinski, J., Cabaud, O., Charafe-Jauffret, E., Birnbaum, D., et al., 2010. CXCR1 blockade selectively targets human breast cancer stem cells in vitro and in xenografts. The Journal of Clinical Investigation 120, 485-497.
632 633 634
Ginn, J., Sacco, J., Wong, Y.Y., Motsinger-Reif, A., Chun, R., Trepanier, L.A., 2014. Positive association between a glutathione-S-transferase polymorphism and lymphoma in dogs. Veterinary and Comparative Oncology 12, 227-236.
635 636 637 638 639
Glasspool, R.M., Brown, R., Gore, M.E., Rustin, G.J., McNeish, I.A., Wilson, R.H., Pledge, S., Paul, J., Mackean, M., Hall, G.D., et al., 2014. A randomised, phase II trial of the DNAhypomethylating agent 5-aza-2'-deoxycytidine (decitabine) in combination with carboplatin vs carboplatin alone in patients with recurrent, partially platinum-sensitive ovarian cancer. British Journal of Cancer 110, 1923-1929. 25 Page 25 of 36
640 641 642 643
Goff, D.J., Court Recart, A., Sadarangani, A., Chun, H.J., Barrett, C.L., Krajewska, M., Leu, H., Low-Marchelli, J., Ma, W., Shih, A.Y., et al., 2013. A Pan-BCL2 inhibitor renders bonemarrow-resident human leukemia stem cells sensitive to tyrosine kinase inhibition. Cell Stem Cell 12, 316-328.
644 645 646
Goldstein, L.J., Galski, H., Fojo, A., Willingham, M., Lai, S.L., Gazdar, A., Pirker, R., Green, A., Crist, W., Brodeur, G.M., et al., 1989. Expression of a multidrug resistance gene in human cancers. Journal of the National Cancer Institute 81, 116-124.
647 648 649
Gramer, I., Kessler, M., Geyer, J., 2013. Determination of MDR1 gene expression for prediction of chemotherapy tolerance and treatment outcome in dogs with lymphoma. Veterinary and Comparative Oncology DOI: 10.1111/vco.12051
650 651
Grosse, N., van Loon, B., Rohrer Bley, C., 2014. DNA damage response and DNA repair dog as a model? BMC Cancer 14, 203.
652 653 654
Hahn, K.A., Barnhill, M.A., Freeman, K.P., Shoieb, A.M., 1999. Detection and clinical significance of plasma glutathione-S-transferases in dogs with lymphoma. In Vivo 13, 173175.
655 656 657 658
Hahn, N.M., Bonney, P.L., Dhawan, D., Jones, D.R., Balch, C., Guo, Z., Hartman-Frey, C., Fang, F., Parker, H.G., Kwon, E.M., et al., 2012. Subcutaneous 5-azacitidine treatment of naturally occurring canine urothelial carcinoma: a novel epigenetic approach to human urothelial carcinoma drug development. The Journal of Urology 187, 302-309.
659 660 661 662
Halsey, C.H., Gustafson, D.L., Rose, B.J., Wolf-Ringwall, A., Burnett, R.C., Duval, D.L., Avery, A.C., Thamm, D.H., 2014. Development of an in vitro model of acquired resistance to toceranib phosphate (Palladia(R)) in canine mast cell tumor. BMC Veterinary Research 10, 105.
663
Hanahan, D., Weinberg, R.A., 2000. The hallmarks of cancer. Cell 100, 57-70.
664 665 666
He, X.C., Zhang, J., Tong, W.G., Tawfik, O., Ross, J., Scoville, D.H., Tian, Q., Zeng, X., He, X., Wiedemann, L.M., et al., 2004. BMP signaling inhibits intestinal stem cell self-renewal through suppression of Wnt-beta-catenin signaling. Nature Genetics 36, 1117-1121.
667 668 669 670
Hifumi, T., Miyoshi, N., Kawaguchi, H., Nomura, K., Yasuda, N., 2010. Immunohistochemical detection of proteins associated with multidrug resistance to anticancer drugs in canine and feline primary pulmonary carcinoma. The Journal of Veterinary Medical Science 72, 665-668.
671 672 673
Hoey, T., Yen, W.C., Axelrod, F., Basi, J., Donigian, L., Dylla, S., Fitch-Bruhns, M., Lazetic, S., Park, I.K., Sato, A., et al., 2009. DLL4 blockade inhibits tumor growth and reduces tumorinitiating cell frequency. Cell Stem Cell 5, 168-177.
674 675
Holohan, C., Van Schaeybroeck, S., Longley, D.B., Johnston, P.G., 2013. Cancer drug resistance: an evolving paradigm. Nature Reviews Cancer 13, 714-726.
676 677 678
Honscha, K.U., Schirmer, A., Reischauer, A., Schoon, H.A., Einspanier, A., Gabel, G., 2009. Expression of ABC-transport proteins in canine mammary cancer: consequences for chemotherapy. Reproduction in Domestic Animals 44 Suppl. 2, 218-223. 26 Page 26 of 36
679 680
Housman, G., Byler, S., Heerboth, S., Lapinska, K., Longacre, M., Snyder, N., Sarkar, S., 2014. Drug resistance in cancer: an overview. Cancers 6, 1769-1792.
681 682 683
Huang, S., Holzel, M., Knijnenburg, T., Schlicker, A., Roepman, P., McDermott, U., Garnett, M., Grernrum, W., Sun, C., Prahallad, A., et al., 2012. MED12 controls the response to multiple cancer drugs through regulation of TGF-beta receptor signaling. Cell 151, 937-950.
684 685
Karotki, A.V., Vasak, M., 2009. Reaction of human metallothionein-3 with cisplatin and transplatin. Journal of Biological Inorganic Chemistry 14, 1129-1138.
686 687 688
Kathawala, R.J., Gupta, P., Ashby, C.R., Jr., Chen, Z.S., 2015. The modulation of ABC transporter-mediated multidrug resistance in cancer: A review of the past decade. Drug Resistance Updates 18C, 1-17.
689 690 691
Kavallaris, M., Tait, A.S., Walsh, B.J., He, L., Horwitz, S.B., Norris, M.D., Haber, M., 2001. Multiple microtubule alterations are associated with Vinca alkaloid resistance in human leukemia cells. Cancer Research 61, 5803-5809.
692 693
Kemper, K., Rodermond, H., Colak, S., Grandela, C., Medema, J.P., 2012. Targeting colorectal cancer stem cells with inducible caspase-9. Apoptosis 17, 528-537.
694 695
Klopfleisch, R., 2015. Personalised medicine in veterinary oncology: One to cure just one. The Veterinary Journal doi.org/10.1016/j.tvjl.2015.01.004
696 697
Klopfleisch, R., Gruber, A.D., 2009. Increased expression of BRCA2 and RAD51 in lymph node metastases of canine mammary adenocarcinomas. Veterinary Pathology 46, 416-422.
698 699 700
Klopfleisch, R., Klose, P., Gruber, A.D., 2010. The combined expression pattern of BMP2, LTBP4, and DERL1 discriminates malignant from benign canine mammary tumors. Veterinary Pathology 47, 446-454.
701 702 703
Klopfleisch, R., Lenze, D., Hummel, M., Gruber, A.D., 2011a. The metastatic cascade is reflected in the transcriptome of metastatic canine mammary carcinomas. The Veterinary Journal 190, 236-243.
704 705 706 707
Klopfleisch, R., Meyer, A., Schlieben, P., Bondzio, A., Weise, C., Lenze, D., Hummel, M., Einspanier, R., Gruber, A.D., 2012. Transcriptome and proteome analysis of tyrosine kinase inhibitor treated canine mast cell tumour cells identifies potentially kit signaling-dependent genes. BMC Veterinary Research 8, 96.
708 709 710
Klopfleisch, R., von Euler, H., Sarli, G., Pinho, S.S., Gartner, F., Gruber, A.D., 2011b. Molecular carcinogenesis of canine mammary tumors: news from an old disease. Veterinary Pathology 48, 98-116.
711 712 713 714
Klose, P., Weise, C., Bondzio, A., Multhaup, G., Einspanier, R., Gruber, A.D., Klopfleisch, R., 2011. Is there a malignant progression associated with a linear change in protein expression levels from normal canine mammary gland to metastatic mammary tumors? Journal of Proteome Research 10, 4405-4415.
715 716 717
Kobayashi, M., Kuroki, S., Tanaka, Y., Moriya, Y., Kozutumi, Y., Uehara, Y., Ono, K., Tamura, K., Washizu, T., Bonkobara, M., 2015. Molecular changes associated with the development of resistance to imatinib in an imatinib-sensitive canine neoplastic mast cell line 27 Page 27 of 36
718 719
carrying a KIT c.1523A>T mutation. European Journal of Haematology doi: 10.1111/ejh.12526
720 721 722
Konopleva, M., Contractor, R., Tsao, T., Samudio, I., Ruvolo, P.P., Kitada, S., Deng, X., Zhai, D., Shi, Y.X., Sneed, T., et al., 2006. Mechanisms of apoptosis sensitivity and resistance to the BH3 mimetic ABT-737 in acute myeloid leukemia. Cancer Cell 10, 375-388.
723 724 725
Krol, M., Pawlowski, K.M., Majchrzak, K., Mucha, J., Motyl, T., 2014. Canine mammary carcinoma cell line are resistant to chemosensitizers: verapamil and cyclosporin A. Polish Journal of Veterinary Sciences 17, 9-17.
726 727 728
Kumaraguruparan, R., Subapriya, R., Balachandran, C., Manohar, B.M., Thangadurai, A., Nagini, S., 2006. Xenobiotic-metabolizing enzymes in canine mammary tumours. The Veterinary Journal 172, 364-368.
729 730 731 732
Kwon, H.C., Roh, M.S., Oh, S.Y., Kim, S.H., Kim, M.C., Kim, J.S., Kim, H.J., 2007. Prognostic value of expression of ERCC1, thymidylate synthase, and glutathione Stransferase P1 for 5-fluorouracil/oxaliplatin chemotherapy in advanced gastric cancer. Annals of Oncology 18, 504-509.
733 734
Lai, K., Kaspar, B.K., Gage, F.H., Schaffer, D.V., 2003. Sonic hedgehog regulates adult neural progenitor proliferation in vitro and in vivo. Nature Neuroscience 6, 21-27.
735 736 737
Liu, S., Ginestier, C., Ou, S.J., Clouthier, S.G., Patel, S.H., Monville, F., Korkaya, H., Heath, A., Dutcher, J., Kleer, C.G., et al., 2011. Breast cancer stem cells are regulated by mesenchymal stem cells through cytokine networks. Cancer Research 71, 614-624.
738 739 740 741
Lombardo, Y., Scopelliti, A., Cammareri, P., Todaro, M., Iovino, F., Ricci-Vitiani, L., Gulotta, G., Dieli, F., de Maria, R., Stassi, G., 2011. Bone morphogenetic protein 4 induces differentiation of colorectal cancer stem cells and increases their response to chemotherapy in mice. Gastroenterology 140, 297-309.
742 743
London, C.A., 2013. Kinase dysfunction and kinase inhibitors. Veterinary Dermatology 24, 181-187 e139-140.
744 745 746 747
Lowenberg, B., van Putten, W., Theobald, M., Gmur, J., Verdonck, L., Sonneveld, P., Fey, M., Schouten, H., de Greef, G., Ferrant, A., et al., 2003. Effect of priming with granulocyte colony-stimulating factor on the outcome of chemotherapy for acute myeloid leukemia. The New England Journal of Medicine 349, 743-752.
748 749 750
Martano, M., Carella, F., Squillacioti, C., Restucci, B., Mazzotta, M., Lo Muzio, L., Maiolino, P., 2012. Metallothionein expression in canine cutaneous apocrine gland tumors. Anticancer Research 32, 747-752.
751 752 753 754
Martin, S.A., McCarthy, A., Barber, L.J., Burgess, D.J., Parry, S., Lord, C.J., Ashworth, A., 2009. Methotrexate induces oxidative DNA damage and is selectively lethal to tumour cells with defects in the DNA mismatch repair gene MSH2. EMBO Molecular Medicine 1, 323337.
755 756 757
Meijer, C., Mulder, N.H., Timmer-Bosscha, H., Sluiter, W.J., Meersma, G.J., de Vries, E.G., 1992. Relationship of cellular glutathione to the cytotoxicity and resistance of seven platinum compounds. Cancer Research 52, 6885-6889. 28 Page 28 of 36
758 759 760
Mhaidat, N.M., Alali, F.Q., Matalqah, S.M., Matalka, II, Jaradat, S.A., Al-Sawalha, N.A., Thorne, R.F., 2009. Inhibition of MEK sensitizes paclitaxel-induced apoptosis of human colorectal cancer cells by downregulation of GRP78. Anti-Cancer Drugs 20, 601-606.
761 762
Michael, M., Doherty, M.M., 2005. Tumoral drug metabolism: overview and its implications for cancer therapy. Journal of Clinical Oncology 23, 205-229.
763 764 765 766
Mirmohammadsadegh, A., Mota, R., Gustrau, A., Hassan, M., Nambiar, S., Marini, A., Bojar, H., Tannapfel, A., Hengge, U.R., 2007. ERK1/2 is highly phosphorylated in melanoma metastases and protects melanoma cells from cisplatin-mediated apoptosis. The Journal of Investigative Dermatology 127, 2207-2215.
767 768 769
Miyashita, T., Reed, J.C., 1992. bcl-2 gene transfer increases relative resistance of S49.1 and WEHI7.2 lymphoid cells to cell death and DNA fragmentation induced by glucocorticoids and multiple chemotherapeutic drugs. Cancer Research 52, 5407-5411.
770 771
Munday, J.S., French, A.F., Gibson, I.R., Gwynne, K., 2009. Widespread mismatch repair protein expression in canine cutaneous mast cell tumors. Veterinary Pathology 46, 227-232.
772 773 774 775
Nakaichi, M., Takeshita, Y., Okuda, M., Nakamoto, Y., Itamoto, K., Une, S., Sasaki, N., Kadosawa, T., Takahashi, T., Taura, Y., 2007. Expression of the MDR1 gene and Pglycoprotein in canine mast cell tumor cell lines. The Journal of Veterinary Medical Science 69, 111-115.
776 777
Nebbioso, A., Carafa, V., Benedetti, R., Altucci, L., 2012. Trials with 'epigenetic' drugs: an update. Molecular Oncology 6, 657-682.
778 779 780
Nieto, A., Perez-Alenza, M.D., Del Castillo, N., Tabanera, E., Castano, M., Pena, L., 2003. BRCA1 expression in canine mammary dysplasias and tumours: relationship with prognostic variables. Journal of Comparative Pathology 128, 260-268.
781 782 783
Nobili, S., Landini, I., Mazzei, T., Mini, E., 2012. Overcoming tumor multidrug resistance using drugs able to evade P-glycoprotein or to exploit its expression. Medicinal Research Reviews 32, 1220-1262.
784 785 786
Noguchi, K., Katayama, K., Sugimoto, Y., 2014. Human ABC transporter ABCG2/BCRP expression in chemoresistance: basic and clinical perspectives for molecular cancer therapeutics. Pharmacogenomics and Personalized Medicine 7, 53-64.
787 788 789
Okai, Y., Nakamura, N., Matsushiro, H., Kato, H., Setoguchi, A., Yazawa, M., Okuda, M., Watari, T., Hasegawa, A., Tsujimoto, H., 2000. Molecular analysis of multidrug resistance in feline lymphoma cells. American Journal of Veterinary Research 61, 1122-1127.
790 791 792 793
Pabst, T., Vellenga, E., van Putten, W., Schouten, H.C., Graux, C., Vekemans, M.C., Biemond, B., Sonneveld, P., Passweg, J., Verdonck, L., et al., 2012. Favorable effect of priming with granulocyte colony-stimulating factor in remission induction of acute myeloid leukemia restricted to dose escalation of cytarabine. Blood 119, 5367-5373.
794 795
Pang, L.Y., Argyle, D.J., 2014. The evolving cancer stem cell paradigm: Implications in veterinary oncology. The VeterinaryJournal doi.org/10.1016/j.tvjl.2014.12.029
29 Page 29 of 36
796 797 798
Pang, L.Y., Argyle, S.A., Kamida, A., Morrison, K.O., Argyle, D.J., 2014. The long-acting COX-2 inhibitor mavacoxib (Trocoxil) has anti-proliferative and pro-apoptotic effects on canine cancer cell lines and cancer stem cells in vitro. BMC Veterinary Research 10, 184.
799 800 801 802
Pang, L.Y., Blacking, T.M., Else, R.W., Sherman, A., Sang, H.M., Whitelaw, B.A., Hupp, T.R., Argyle, D.J., 2013. Feline mammary carcinoma stem cells are tumorigenic, radioresistant, chemoresistant and defective in activation of the ATM/p53 DNA damage pathway. The Veterinary Journal 196, 414-423.
803 804
Pavet, V., Portal, M.M., Moulin, J.C., Herbrecht, R., Gronemeyer, H., 2011. Towards novel paradigms for cancer therapy. Oncogene 30, 1-20.
805 806 807
Pawlak, A., Rapak, A., Zbyryt, I., Obminska-Mrukowicz, B., 2014. The effect of common antineoplastic agents on induction of apoptosis in canine lymphoma and leukemia cell lines. In Vivo 28, 843-850.
808 809 810
Pawlowski, K.M., Mucha, J., Majchrzak, K., Motyl, T., Krol, M., 2013. Expression and role of PGP, BCRP, MRP1 and MRP3 in multidrug resistance of canine mammary cancer cells. BMC Veterinary Research 9, 119.
811 812 813
Perez, E.A., 2009. Microtubule inhibitors: Differentiating tubulin-inhibiting agents based on mechanisms of action, clinical activity, and resistance. Molecular Cancer Therapeutics 8, 2086-2095.
814 815 816 817
Piovan, E., Yu, J., Tosello, V., Herranz, D., Ambesi-Impiombato, A., Da Silva, A.C., Sanchez-Martin, M., Perez-Garcia, A., Rigo, I., Castillo, M., et al., 2013. Direct reversal of glucocorticoid resistance by AKT inhibition in acute lymphoblastic leukemia. Cancer Cell 24, 766-776.
818 819 820 821
Qiu, M., Peng, Q., Jiang, I., Carroll, C., Han, G., Rymer, I., Lippincott, J., Zachwieja, J., Gajiwala, K., Kraynov, E., et al., 2013. Specific inhibition of Notch1 signaling enhances the antitumor efficacy of chemotherapy in triple negative breast cancer through reduction of cancer stem cells. Cancer Letters 328, 261-270.
822 823
Rabik, C.A., Dolan, M.E., 2007. Molecular mechanisms of resistance and toxicity associated with platinating agents. Cancer Treatment Reviews 33, 9-23.
824 825 826
Rebhun, R.B., Lana, S.E., Ehrhart, E.J., Charles, J.B., Thamm, D.H., 2008. Comparative analysis of survivin expression in untreated and relapsed canine lymphoma. Journal of Veterinary Internal Medicine 22, 989-995.
827 828
Rocha, T.A., Mauldin, G.N., Patnaik, A.K., Bergman, P.J., 2000. Prognostic factors in dogs with urinary bladder carcinoma. Journal of Veterinary Internal Medicine 14, 486-490.
829 830 831 832
Saez-Ayala, M., Fernandez-Perez, M.P., Montenegro, M.F., Sanchez-del-Campo, L., Chazarra, S., Pinero-Madrona, A., Cabezas-Herrera, J., Rodriguez-Lopez, J.N., 2012. Melanoma coordinates general and cell-specific mechanisms to promote methotrexate resistance. Experimental Cell Research 318, 1146-1159.
833 834 835
Sarkaria, J.N., Kitange, G.J., James, C.D., Plummer, R., Calvert, H., Weller, M., Wick, W., 2008. Mechanisms of chemoresistance to alkylating agents in malignant glioma. Clinical Cancer Research 14, 2900-2908. 30 Page 30 of 36
836 837
Schlossmacher, G., Stevens, A., White, A., 2011. Glucocorticoid receptor-mediated apoptosis: mechanisms of resistance in cancer cells. The Journal of Endocrinology 211, 17-25.
838 839 840
Shoieb, A.M., Hahn, K.A., Van Laack, R.L., Barnhill, M.A., 1998. In vitro reversal of glutathione-S-transferase-mediated resistance in canine osteosarcoma (COS31) cells. In Vivo 12, 455-462.
841 842 843 844
Signore, M., Pelacchi, F., di Martino, S., Runci, D., Biffoni, M., Giannetti, S., Morgante, L., De Majo, M., Petricoin, E.F., Stancato, L.,et al., 2014. Combined PDK1 and CHK1 inhibition is required to kill glioblastoma stem-like cells in vitro and in vivo. Cell Death and Disease 5, e1223.
845 846 847 848
Simon, D., Moreno, S.N., Hirschberger, J., Moritz, A., Kohn, B., Neumann, S., Jurina, K., Scharvogel, S., Schwedes, C., Reinacher, M., et al., 2008. Efficacy of a continuous, multiagent chemotherapeutic protocol versus a short-term single-agent protocol in dogs with lymphoma. Journal of the American Veterinary Medical Association 232, 879-885.
849 850 851
Smallwood, T.L., Small, G.W., Suter, S.E., Richards, K.L., 2014. Expression of asparagine synthetase predicts in vitro response to L-asparaginase in canine lymphoid cell lines. Leukemia and Lymphoma 55, 1357-1365.
852 853 854
Sugimoto, Y., Tsukahara, S., Oh-hara, T., Isoe, T., Tsuruo, T., 1990. Decreased expression of DNA topoisomerase I in camptothecin-resistant tumor cell lines as determined by a monoclonal antibody. Cancer Research 50, 6925-6930.
855 856 857
Swaminathan, S.K., Roger, E., Toti, U., Niu, L., Ohlfest, J.R., Panyam, J., 2013. CD133targeted paclitaxel delivery inhibits local tumor recurrence in a mouse model of breast cancer. Journal of Controlled Release 171, 280-287.
858 859
Takahashi-Yanaga, F., Kahn, M., 2010. Targeting Wnt signaling: can we safely eradicate cancer stem cells? Clinical Cancer Research 16, 3153-3162.
860 861 862
Tang, J., Xie, X., Zhang, X., Qiao, X., Jiang, S., Shi, W., Shao, Y., Zhou, X., 2012. Long term cultured HL-60 cells are intrinsically resistant to Ara-C through high CDA activity. Frontiers in Bioscience 17, 569-574.
863 864 865
Thamm, D.H., Grunerud, K.K., Rose, B.J., Vail, D.M., Bailey, S.M., 2013. DNA repair deficiency as a susceptibility marker for spontaneous lymphoma in golden retriever dogs: a case-control study. PloS One 8, e69192.
866 867 868 869
Thayanithy, V., Park, C., Sarver, A.L., Kartha, R.V., Korpela, D.M., Graef, A.J., Steer, C.J., Modiano, J.F., Subramanian, S., 2012. Combinatorial treatment of DNA and chromatinmodifying drugs cause cell death in human and canine osteosarcoma cell lines. PloS One 7, e43720.
870 871 872
Tomiyasu, H., Fujiwara-Igarashi, A., Goto-Koshino, Y., Fujino, Y., Ohno, K., Tsujimoto, H., 2014a. Evaluation of DNA methylation profiles of the CpG island of the ABCB1 gene in dogs with lymphoma. American Journal of Veterinary Research 75, 835-841.
873 874 875
Tomiyasu, H., Goto-Koshino, Y., Fujino, Y., Ohno, K., Tsujimoto, H., 2014b. Antitumour effect and modulation of expression of the ABCB1 gene by perifosine in canine lymphoid tumour cell lines. The Veterinary Journal 201, 83-90. 31 Page 31 of 36
876 877 878
Tomiyasu, H., Goto-Koshino, Y., Fujino, Y., Ohno, K., Tsujimoto, H., 2014c. Epigenetic regulation of the ABCB1 gene in drug-sensitive and drug-resistant lymphoid tumour cell lines obtained from canine patients. The Veterinary Journal 199, 103-109.
879 880 881
Tomiyasu, H., Goto-Koshino, Y., Fujino, Y., Ohno, K., Tsujimoto, H., 2014d. The regulation of the expression of ABCG2 gene through mitogen-activated protein kinase pathways in canine lymphoid tumor cell lines. The Journal of Veterinary Medical Science 76, 237-242.
882 883 884
Tomiyasu, H., Goto-Koshino, Y., Takahashi, M., Fujino, Y., Ohno, K., Tsujimoto, H., 2010. Quantitative analysis of mRNA for 10 different drug resistance factors in dogs with lymphoma. The Journal of Veterinary Medical Science 72, 1165-1172.
885 886 887
Van der Heyden, S., Chiers, K., Vercauteren, G., Daminet, S., Wegge, B., Paepe, D., Ducatelle, R., 2011. Expression of multidrug resistance-associated P-glycoprotein in feline tumours. Journal of Comparative Pathology 144, 164-169.
888 889
van Steensel, B., 2011. Chromatin: constructing the big picture. The EMBO Journal 30, 18851895.
890 891
Wilting, R.H., Dannenberg, J.H., 2012. Epigenetic mechanisms in tumorigenesis, tumor cell heterogeneity and drug resistance. Drug Resistance Updates 15, 21-38.
892 893 894
Yang, Z., Zhu, W., Gao, S., Yin, T., Jiang, W., Hu, M., 2012. Breast cancer resistance protein (ABCG2) determines distribution of genistein phase II metabolites: reevaluation of the roles of ABCG2 in the disposition of genistein. Drug Metabolism and Disposition 40, 1883-1893.
895 896 897
Yoshimoto, K., Ma, X., Guan, Y., Mizoguchi, M., Nakamizo, A., Amano, T., Hata, N., Kuga, D., Sasaki, T., 2011. Expression of stem cell marker and receptor kinase genes in glioblastoma tissue quantified by real-time RT-PCR. Brain Tumor Pathology 28, 291-296.
898 899 900
Zandvliet, M., Teske, E., Chapuis, T., Fink-Gremmels, J., Schrickx, J.A., 2013. Masitinib reverses doxorubicin resistance in canine lymphoid cells by inhibiting the function of Pglycoprotein. Journal of Veterinary Pharmacology and Therapeutics 36, 583-587.
901 902 903
Zandvliet, M., Teske, E., Schrickx, J.A., 2014a. Multi-drug resistance in a canine lymphoid cell line due to increased P-glycoprotein expression, a potential model for drug-resistant canine lymphoma. Toxicology In Vitro 28, 1498-1506.
904 905
Zandvliet, M., Teske, E., Schrickx, J.A., Mol, J.A., 2014b. A longitudinal study of ABC transporter expression in canine multicentric lymphoma. Veterinary journal.
906 907
Zhang, N., Yin, Y., Xu, S.J., Chen, W.S., 2008. 5-Fluorouracil: mechanisms of resistance and reversal strategies. Molecules 13, 1551-1569.
908 909 910
Zhao, C., Chen, A., Jamieson, C.H., Fereshteh, M., Abrahamsson, A., Blum, J., Kwon, H.Y., Kim, J., Chute, J.P., Rizzieri, D., et al., 2009. Hedgehog signalling is essential for maintenance of cancer stem cells in myeloid leukaemia. Nature 458, 776-779.
911 912 32 Page 32 of 36
913
Table 1
914 915
Specific mechanisms of resistance to chemotherapeutic agents commonly used in veterinary oncology Agent Alkylating agents Cyclophosphamide Chlorambucil Lomustine Procarbazine
Target/mechanism
Resistance mechanisms (References)
DNA-crosslinking, inhibition of DNA repair/synthesis
Increased efflux (Pawlowski et al., 2013) Increased DNA repair (Fink et al., 1998) MGMT activity (Sarkaria et al., 2008)
Unclear, maybe DNA break induction, t-RNA inhibition
Platinum containing drugs Cisplatin DNA-crosslinking, prevention of DNA-uncoiling/strand Carboplatin separation Antimetabolites Methotrexate Cytarabine
Dihydrofolate reductase DNA-Synthesis inhibition, DNA-chain termination 5-Fluorouracil (5-FU) Thymidylate synthase inhibition, replacement of cytosine, thymidine, uracil in DNA/RNA strands Gemcitabine Replaces cytidine DNAsynthesis inhibition Topoisomerase I (TOPO1) inhibitors Camptothecins Stabilization of the cleavable (Irinotecan, Topotecan) DNA-enzyme complex and thereby inducing DNA damage Indenoisoquinoline Complexing with TOPO1 Topoisomerase II (TOPO2) inhibitors Doxorubicin Inhibition of TOPO2 by DNA intercalation Mitoxantrone Microtubule poisons Paclitaxel Vinblastine Vincristine Vinorelbine Miscellaneous Agents Prednisone
L-Asparaginase
Increased efflux (Pawlowski et al., 2013), ERK activity (Mirmohammadsadegh et al., 2007) DNA repair (Kwon et al., 2007) Replicative bypass (Rabik and Dolan, 2007) Melanosomal efflux (Saez-Ayala et al., 2012) Decreased uptake (Cai et al., 2008) Decreased activation by DCK, DPD (Cai et al., 2008; Zhang et al., 2008) Detoxification by CDD, CDA, NT5C2 (Bardenheuer et al., 2005; Tang et al., 2012) Increased DNA repair (Zhang et al., 2008) Induction of anti-apoptotic molecules (Zhang et al., 2008) Activation of survival pathways (Arlt et al., 2003) TOPO1 mutations (Sugimoto et al., 1990) Reduced TOPO expression (Meijer et al., 1992) Improved DNA repair (Alagoz et al., 2012) Lack of apoptosis induction (Beretta et al., 2013) Increase efflux by MDR and BCRP (Chu et al., 1997) Unknown so far (Yang et al., 2012) Increased efflux (Zandvliet et al., 2014b) Amplification of TOPO2 and ERBB2 (Noguchi et al., 2014)
Microtubules stabilization Microtubule Depolymerisation/stabilization
Apoptosis inhibition (Mhaidat et al., 2009) Increase efflux (Pawlowski et al., 2013) Tubulin mutation (Kavallaris et al., 2001) Stathmin, MAP4, y-actin overexpression of (Perez, 2009)
Unclear, apoptosis induction
Increased efflux (Dhaliwal et al., 2013) STAT3, pSTAT3, KIT overexpression (Dhaliwal et al., 2013) PTEN loss, AKT1 activity (Piovan et al., 2013) Decreased receptor expression (Schlossmacher et al., 2011) Increased asparagine synthetase (Chien et al., 2015) Decreased cellular efflux of asparagine (Chien et al., 2015) Increased L-glutaminase activity (Chien et al., 2015) Aspartic acid synthesis (Chien et al., 2015) Activation of glutamine uptake (Chien et al., 2015)
Asparagine deprivation
Tyrosine kinase inhibitors Toceranib KIT, VEGFR2, PDGFRB Imatinib KIT, PDGFR Masitinib KIT, PDGFRA
Target mutation (Halsey et al., 2014; Kobayashi et al., 2015) Increased (MCL1) expression (Amagai et al., 2013)
33 Page 33 of 36
916 917 918 919 920
Genistein Diverse tyrosine kinases CDA, cytidine-deaminase; CDD, cytidine deaminase; DPD, dihydropyrimidine dehydrogenase; ERK, extracellular signalregulated kinase; DCK, deoxycytidine kinase; HR, homologous recombination; MCL1, Myeloid cell leucaemia sequence 1; MMR, mismatch repair; MGMT, O6-methylguanine methyltransferase; NER, nucleotide excision repair; NT5C2, cytoplasmic 5′nucleotidase; PDGFR, platelet-derived growth factor; Replicative bypass, ability of DNA polymerase to bypass DNA cross-links; VGEFR, vascular endothelial growth factor
921
34 Page 34 of 36
922
Table 2
923
Studies confirming MDR1, BCRP, MRP1-mediated chemotherapy resistance in animal
924
tumours ABCtransporter MDR1
Cancer type Canine mammary tumour
Confirmed contribution chemoresistance (agent) Cisplatin Vinblastine No effect on vinblastine No effect on cyclophosphamide Doxorubicin
to
Reference
No effect on prednisone
(Pawlowski et al., 2013) (Pawlowski et al., 2013) (Krol et al., 2014) (Pawlowski et al., 2013) (Zandvliet et al., 2013; Zandvliet et al., 2014a) (Tomiyasu et al., 2014b; Zandvliet et al., 2014a) (Zandvliet et al., 2014a)
Canine mast cells
General effect on efflux
(Nakaichi et al., 2007)
BCRP
Canine mammary tumour
Cyclophosphamide Cisplatin No effect on vinblastine Doxorubicin No effect on methotrexate
(Pawlowski et al., 2013) (Pawlowski et al., 2013) (Pawlowski et al., 2013) (Honscha et al., 2009) (Honscha et al., 2009)
MRP1
Canine mammary tumour
Cisplatin Vinblastine No effect on cyclophosphamide
(Pawlowski et al., 2013) (Pawlowski et al., 2013) (Pawlowski et al., 2013)
Canine lymphoma
Vincristine
925 926
35 Page 35 of 36
927
Figure legends
928 929 930 931 932 933 934 935
Figure 1. Mechanisms of ABC-protein mediated resistance and corresponding therapies. Each of the almost 50 ABC-transporters identified so far is able to transport a specific set of anticancer drugs out of tumour cells. These sets are however overlapping for many transporters. Specific modulators of single or few transporters therefore often lack efficacy in overcoming chemotherapy resistance. Numerous modulators of ABC transporters have been developed and tested with variable success in human clinical trials and few in vivo studies in veterinary oncology. TKI, tyrosine kinase inhibitors; MDR, multi-drug resistance gene; MRP1, MDRassociated protein 1; BCRP, breast cancer resistance protein.
936 937
Figure 2. The ambivalence of effective DNA-repair in chemotherapy resistance
938 939 940 941
Figure 3. Potential targets for the effective treatment of cancer stem cells. Chk, checkpoint kinase; G-CSF, granulocyte-colony stimulating factor; HH, hedgehog
36 Page 36 of 36