G Model

BIOPHA 3475 1–6 Biomedicine & Pharmacotherapy xxx (2014) xxx–xxx

Available online at

ScienceDirect www.sciencedirect.com

1 2 3 4 5 6 7

Review

Colon cancer: Cancer stem cells markers, drug resistance and treatment Q1 Zuzana a b

Kozovska a,1,*, Veronika Gabrisova b,1, Lucia Kucerova a

Department of Oncology, Cancer Research Institute, Slovak Academy of Sciences, Vlarska 7, 83391 Bratislava, Slovakia Department of Cellular and Molecular Biology of Drugs, Faculty of Pharmacy, Comenius University in Bratislava, Odbojarov 10, 83232 Bratislava, Slovakia

A R T I C L E I N F O

A B S T R A C T

Article history: Received 12 August 2014 Accepted 15 October 2014

Malignant tumours consist of heterogeneous populations of tumour cells. Cancer stem cells (CSC) represent a population of cells within a tumour with highly tumorigenic and chemoresistant properties. These cells may be identified by the expression of CSC markers. There are several key stem cells markers specified for colon cancer: CD133, CD44, ALDH1, ALCAM. These days, a major obstacle to effective cancer management is development of a multidrug resistance (MDR). The principal mechanism responsible for development of MDR phenotype is the over-expression of ABC transporters. Tumours and relapsing tumours after therapy are drived by subpopulations of tumour cells with aggressive phenotype resistant to chemotherapeutics. These cells are called CSC or tumour-initiating cells (TIC). Here we outline recent information about MDR of colon cancer and CSC markers. We have focused on novel therapeutic strategies which have been developed to prevent or overcome MDR. One such strategy is a combination of chemotherapy and modulators of MDR pumps or chemotherapy and monoclonal antibodies against vascular endothelial growth factor VEGF. Colon cancer is characterized by the presence of colon CSC expressing specific stem cell markers. The divergent presence of these markers can help to adjust personalized therapy. The review provides a detailed overview of resistance of colon cancer cells and discusses how the presence of CSC markers can influence therapy and prognosis of patients. ß 2014 Published by Elsevier Masson SAS.

Keywords: Colon cancer Cancer stem cells markers Multidrug resistance Aldehyd dehydrogenase ALDH1

8 9

1. Introduction

10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25

Cancer is a genetic disease characterized by uncontrolled cell growth and the ability to invade other parts of the body by forming metastases. Malignant tumours consist of heterogeneous populations of cancer cells. CSC form a subpopulation of cells within a tumour which display the capacity of self-renewal and differentiation. These cells, also described as TIC, are presumed to promote growth and metastases of tumours [1]. There were 14.1 million new cancer cases, 8.2 million cancer deaths and 32.6 million people living with cancer (within 5 years of diagnosis) in 2012 worldwide (http://globocan.iarc.fr/Pages/fact_sheets_cancer.aspx). GLOBOCAN 2012 (IARC), Section of Cancer Information (16/6/2014). The chemotherapy failure is attributed to the development of a MDR. The MDR phenotype accounts for unsatisfactory low response rate of solid tumours to chemotherapy in patients treated chronically with antineoplastic agents. This clinical

* Corresponding author. Tel.: +421 2 59327418; fax: +421 2 59327250. E-mail address: [email protected] (Z. Kozovska). 1 These authors contributed equally to this work.

phenomenon results from the ability of cancer cells to become simultaneously resistant to many structurally dissimilar and functionally divergent drugs used in anticancer therapy [2]. The over-expression of ABC transporters represents the principal mechanism by which cancer cells develop MDR. Increased drug efflux mediated by ABC transporters leads to decreased intracellular drug accumulation, hence to the decreased bioavailability. Combination therapy using more than one medication at the same time may be used to overcome MDR [3].

26 27 28 29 30 31 32 33 34

2. Progression of cancer

35

Several types of pre-malignant lesions, such as dysplasia and hyperplasia, can be detected in diverse organs prior to the appearance of fully malignant invasive tumours. An early stage of cancer characterised by the absence of invasion of tumour cells into the surrounding tissue, before penetration through the basement membrane, is called carcinoma in situ. Accumulation of genetic alterations occurs in one (or a few) of the pre-malignant cells, and the cells convert into malignant ones of clonal origin and produce a primary tumour. After the initial transformation and growth of cells, neoangiogenesis arises. At the early stage of

36 37 38 39 40 41 42 43 44 45

http://dx.doi.org/10.1016/j.biopha.2014.10.019 0753-3322/ß 2014 Published by Elsevier Masson SAS.

Please cite this article in press as: Kozovska Z, et al. Colon cancer: Cancer stem cells markers, drug resistance and treatment. Biomed Pharmacother (2014), http://dx.doi.org/10.1016/j.biopha.2014.10.019

G Model

BIOPHA 3475 1–6 2

46 47 48 49 50 51 52 53

Z. Kozovska et al. / Biomedicine & Pharmacotherapy xxx (2014) xxx–xxx

primary tumour expansion, the cells are not invasive and metastatic. Invasiveness and metastatic ability appears as a result of further accumulation of genetic alterations in the cells. Diagnosis and treatment of cancer occurs generally late in the course of disease, in some types of cancer perhaps more than 10 years prior to the acquisition of the invasive phenotype [4]. At this stage, a high proportion of patients have obvious or occult metastases.

54

3. Metastases

55 56 57 58 59 60 61 62 63 64 65 66 67 68

Despite the significant improvements in diagnosis, surgical techniques, specificity of cancer treatment, patient care and adjuvant therapies, yet there is no cancer treatment that is able to cure majority of the patients with metastatic disease. The major obstacles to effective treatment include the biologic heterogeneity of tumour cells, the specific organ microenvironment, which influences the biologic behaviour of metastatic cells, including their response to systemic therapy and the development of drug resistance phenotype in metastatic cancer cells [5–7]. New and very promising approach for treatment of the metastases is a gene therapy using genetically modified mesenchymal stromal cells (MSC), which are able to convert a nontoxic prodrug to a highly toxic chemotherapeutic at the site of the tumour (so-called genedirected enzyme/prodrug therapy; suicide gene therapy) [8–12].

69

4. Cancer stem cell theory

70 71 72 73 74 75 76 77 78 79 80 81 82 83 84 85 86 87 88 89 90 91 92 93 94 95 96 97 98 99 100 101 102 103 104 105 106

Cancer is known to result from the accumulation of multiple genetic mutations in a single target cell, sometimes over a period of many years [13]. Although monoclonal in origin, most tumours are recognised as a morphologically heterogeneous population of cancer cells [14]. This tumour heterogeneity can be explained by variations in tumour microenvironment and genomic instability [15–17]. CSC theory brings additional explanation, stating that intratumour heterogeneity can result from a functional diversity among cells in different states of differentiation [18]. Cancer cells within tumours may display different phenotypes, somewhat reminiscent of the normal tissue from which they originate, and have varying proliferation potential [19]. The CSC theory proposes that tumours have a cellular hierarchy that is a caricature of their normal tissue counterpart because they reflect the pluripotency of the originally transformed cell [20]. According to this hypothesis, only a subpopulation of cells within a cancer, made up of so-called CSC or TIC, have the exclusive capacity to regenerate a tumour and sustain its growth. Purified CSC are known to be potently tumourigenic [21]. They are able to regenerate the tumour from which they were derived when a limited number of cells (sometimes as few as 102 cells) are injected into an orthotopic in vivo microenvironment [20,22]. The other cancer cells have only a limited capacity to replicate and thus contribute to the tumour bulk but not to tumour maintenance [20]. CSC demonstrate some properties of normal stem cells, such as self-renewing abilities and differentiation capacity. Self-renewal is a unique replication process that allows stem cells to divide and give rise to either one or two stem cells [23]. In normal tissues, the expansion of the stem cell pool is restricted to prevent uncontrolled growth [24], whereas extensive and indefinite selfrenewal is observed in CSC. CSC either originate from normal cells that became malignant or committed cells that acquired selfrenewal capacity. Asymmetric division enables CSC to self-renew, thus forming another CSC, and a more committed progenitor cell. Subsequently, progenitor cell divides and differentiates to form a malignant progeny [25]. CSC are able to self-renew in vivo after serial transplantation in secondary and tertiary recipients when

phenotypically identical and heterogeneous tumour is observed [21]. Differentiation is the second function of a stem cell and involves the potential to form tissue-specific specialized cells. Tumours arising from CSC demonstrate differentiation capacity in vivo by creating a phenocopy of the original human tumour.

107 108 109 110 111

4.1. Colorectal CSC

112

There is now accumulating evidence for the existence of colorectal CSC in human colorectal cancer (CRC) [18,26–29]. Using the CD133 surface marker in two separate studies [26,27], highly tumourigenic and self-renewing colorectal CSC population in human colon cancers have been successfully enriched and CD133+ human cancer cells were found capable of inducing tumour formation while CD133 cancer cells failed to do so [27]. The CD133+ cells produced passageable sphere-forming cells that remained undifferentiated for more than one year and were able to initiates in vivo tumours that were phenotypically similar to the original tumour [27]. In a renal capsule xenograft model, CD133+ cells could generate tumours, but CD133 cells were unable to do so [26]. Moreover, CD133high populations displayed a much more aggressive phenotype than CD133/low cells, in terms of metastases, implicating CD133 as a marker of increased tumourigenicity [30]. In addition, CD133+ single cell clinical tumour-derived cultures were shown to possess multilineage differentiation potential and were capable of tumour initiation in vivo [31]. CD133+ overexpressing tumours were more resistant to 5FU-based chemotherapy and CD133 expression was associated with poor prognosis [32]. RNA interference approach showed that knockdown of CD133 had no effect on in vitro clonogenicity or in vivo xenograft tumour formation, demonstrating that CD133 is rather a passive marker of colorectal CSC [33]. Another subset of tumourigenic human colon cancer cells was identified based on ESAhighCD44+ CD166+Lineage colon cancer phenotype [18]. Knockdown of CD44 (hyaluronate receptor, P-gp 1) resulted in limited colony formation and dramatically reduced tumour formation in xenografts, strongly implying a functional role of CD44 in CRC tumourigenesis [33]. Aldehyde dehydrogenase 1 (ALDH1) is a promising new marker for identification of colorectal CSC. ALDH was found to be a specific marker for identification, isolation and tracking of malignant human colonic CSC and for quantifying the number of CSC over the course of CRC development [28]. Flow cytometric isolation of cancer cells based on enzymatic activity of ALDH (ALDEFLUORassay) and implantation of these cells in NOD-SCID mice:

113 114 115 116 117 118 119 120 121 122 123 124 125 126 127 128 129 130 131 132 133 134 135 136 137 138 139 140 141 142 143 144 145 146 147 148 149

 generated xenograft tumours (Aldefluor cells did not);  generated them after implanting as few as 25 cells, and;  generated them dose dependently [28].

151 152 153

ALDH is an intracellular enzyme that oxidizes aldehydes, possess a detoxifying role, converts retinol to retinoic acid [34], mediates control over differentiation pathways [35], plays an important role in self-protection of normal stem cells [36] and confers resistance to alkylating agents. ALDH is now widely used as a marker for identification and isolation of various types of normal stem cells and CSC. ALDH can confer resistance to selected anticancer agents by metabolic inactivation [37] and it is speculated to be a cause of relapse in cancer patients [38– 40]. ALDH1A1 is one of the 19 ALDH isoforms expressed in humans. In many cancers, high ALDH1 expression is associated with metastases development and correlates with poor clinical outcome [41,42]. Recent evidence suggests that ALDH activity may protect against cell death caused by reactive oxygen species (ROS) [43]. ALDH activity can be inhibited pharmacologically by

154 155 156 157 158 159 160 161 162 163 164 165 166 167 168

Please cite this article in press as: Kozovska Z, et al. Colon cancer: Cancer stem cells markers, drug resistance and treatment. Biomed Pharmacother (2014), http://dx.doi.org/10.1016/j.biopha.2014.10.019

G Model

BIOPHA 3475 1–6 Z. Kozovska et al. / Biomedicine & Pharmacotherapy xxx (2014) xxx–xxx

169 170 171 172 173 174 175 176 177

4-diethylaminobenzaldehyde (DEAB) (Fig. 1). Because chemotherapy is known to induce oxidative stress [44], it is possible that inhibition of ALDH activity using DEAB may sensitize ALDHhi CSC to therapy through enhanced exposure to ROS and induction of apoptosis [41]. We were able to increase susceptibility of colorectal cancer cells (HT-29) to chemotherapy after incubation with DEAB in our preliminary study. We would like to confirm this observation by molecular inhibition of ALDH with specific siRNA (small interfering RNA) towards ALDH1 isoforms in our research.

178

4.2. Clinical implications of the CSC model

179 180 181 182 183 184 185 186 187 188 189 190 191 192 193 194 195 196 197 198 199 200 201 202

Nowadays, clinical treatment regimens operate under the assumption that all cancer cells have equal malignant potential [20]. However, since CSC are potentially more resistant to antineoplastic therapy and probably cause disease recurrence and/or metastases [45], therapeutic approaches that do not eradicate CSC are likely to fail. They might kill the bulk tumour population and induce temporary regression of gross tumour lesions but fail to prevent disease relapse and metastatic dissemination. Even though these drugs help to maintain longterm remissions, they are not curative. It is, therefore, necessary to design new and more effective antitumour treatments that would specifically target the CSC subpopulation [46]. Targeted therapies, selective to CSC and non-toxic to normal stem cells, hold a great promise for the effective and curative therapy of many human cancers. Antitumour treatments designed and selected for broad cytotoxic activity may kill the majority of cancer cells within a specific tumour tissue and induce dramatic, even complete regression of large tumor masses; however, if any of the CSCs are spared, tumour tissues can be regenerated and cause disease relapse. In contrast, antitumour treatments specifically designed to target CSCs, although theoretically unable to cause rapid shrinkage of tumour lesions, might nonetheless achieve long-term disease eradication by exhausting the self-renewal and growth potential of cancer tissues [47].

203

5. Treatment

204 205 206 207 208 209 210 211 212 213 214 215 216 217 218 219 220 221

Management of CRC relies primarily on resection of the bowel with the adjacent lymph nodes. The need for neoadjuvant or adjuvant chemotherapy, with or without concurrent irradiation, depends on tumour location and a stage of the disease. Early stages of CRC are treated primarily by surgery-laparoscopic colectomy, if possible. A combination of surgical and other therapeutic modalities is indicated if regional or distant metastases are discovered at the time of diagnosis. Adjuvant treatment for CRC is indicated for curative-intent resections of stage IV disease and for stage III disease [48]. The utility of adjuvant therapy in stage II disease remains unclear and controversial [49]. High-risk patients might be considered for such treatment. On the other hand, patients with poorer prognosis may benefit more from the palliative care instead of chemotherapy. After decades of 5-FUbased treatment, the arrival of new, effective agents has significantly changed the way this cancer is treated. Although 5FU remains the backbone of most regimens, the new agents have become an important part of a front-line treatment providing

Fig. 1. DEAB structure.

3

improvements for patients in both response and survival. Synergic or additive action and decreased toxicity and side effects of drugs represent the major benefits of combination treatment.

222 223 224

5.1. 5-fluorouracil (5-FU)

225

Antimetabolite 5-FU, used in the treatment of a range of cancers including CRC, acts by incorporation into DNA and RNA and thus inhibiting nucleotide biosynthesis. 5-FU is a prodrug that enters the cells using the same facilitated transport system as uracil [50]. It is intracellularly converted into three active metabolites: fluorodeoxyuridine monophosphate (FdUMP), fluorodeoxyuridine triphosphate (FdUTP) and fluorouridine triphosphate (FUTP).

226 227 228 229 230 231 232

5.1.1. Anticancer agents used for modulation of 5-FU action 5.1.1.1. Increased TS binding activity of 5-FdUMP by leucovorin. Leucovorin (calcium folinate) increases the level of reduced folate (CH2THF; N5,N-10-methylenetetrahydrofolate), which enables 5fluorodeoxyuridine monophosphate (5-FdUMP) to bind to TS and to form stable tertiary complex resulting in increased in vitro [51,52] and in vivo [53,54] toxicity of 5-FU.

233 234 235 236 237 238 239

5.1.1.2. Decreased 5-FU degradation by capecitabine. Capecitabine is an oral fluoropyrimidine prodrug that is converted to 50 -deoxy5-fluorouridine (5´DFUR) in the liver. 5´DFUR is then converted to 5FU by thymidine phosphorylase (TP) and/or uridine phosphorylase (UP) ([55,56] at the site of the tumour. Capecitabine is used to avoid dihydropyrimidine dehydrogenase (DPD)-mediated 5-FU degradation in the liver.

240 241 242 243 244 245 246

5.1.1.3. Increased 5-FU activation by methotrexate. Methotrexate is an antifolate inhibitor of dihydrofolate reductase. Inhibition of purine biosynthesis by MTX increases the levels of phosphoribosyl pyrophosphate (PRPP), the cofactor of 5-FU conversion to fluorouridine monophosphate (FUMP) by orotate phosphoribosyl transferase (OPRT). The increased PRPP levels induced by MTX promote conversion of 5-FU to FUMP [57].

247 248 249 250 251 252 253

5.1.1.4. Irinotecan. Camptothecin derivate, irinotecane is a prodrug whose active metabolite is a potent inhibitor of the nuclear enzyme topoisomerase I, which facilitates the uncoiling of DNA during replication [58].

254 255 256 257

5.1.1.5. Oxaliplatin. Oxaliplatin is a platinum compound that forms cross-linking DNA adducts, thereby blocking DNA replication and transcription [59].

258 259 260

5.1.2. Monoclonal antibodies used in colorectal cancer treatment 5.1.2.1. Bevacizumab. Bevacizumab is a humanized monoclonal antibody that inhibits blood-vessel formation by binding to the vascular endothelial growth factor-A (VEGF-A) [60]. It was the first anti-angiogenic drug approved for metastatic colon cancer. It prevents the binding of VEGF-A to the VEGFR and, consequently, inhibits angiogenesis, tumour growth and metastatic development [3].

261 262 263 264 265 266 267 268

5.1.2.2. Ziv-aflibercept. Ziv-aflibercept is a recombinant fusion protein, which acts as a soluble receptor that binds to VEGF-A, VEGF-B and PlGF (phosphatidylinositol-glycan biosynthesis class F protein) and inhibits them, resulting in a decreased neovascularization and a decreased vascular permeability [3].

269 270 271 272 273

5.1.2.3. Cetuximab. Cetuximab is a chimeric (human/mouse) monoclonal antibody that binds to the extracellular domain of the epidermal growth factor receptor (EGFR) and blocks ligand-induced receptor signalling [60]. These anti-EGFR

274 275 276 277

Please cite this article in press as: Kozovska Z, et al. Colon cancer: Cancer stem cells markers, drug resistance and treatment. Biomed Pharmacother (2014), http://dx.doi.org/10.1016/j.biopha.2014.10.019

G Model

BIOPHA 3475 1–6 4

Z. Kozovska et al. / Biomedicine & Pharmacotherapy xxx (2014) xxx–xxx

278 279

therapies have found their place in the treatment of stage IV colon cancer [3].

 induction of mechanisms that repair DNA damage [74];  alterations in the cell cycle and checkpoints [71].

337 338

280 281

5.1.2.4. Panitumumab. Panitumumab is a fully human monoclonal antibody to EGFR, with similar activity to cetuximab [60].

6.1. Overview of ABC transporters

339

282 283 284 285 286

5.1.2.5. Regorafenib. It is an oral tumour deactivation agent that potently blocks multiple protein kinases, including kinases involved in tumour angiogenesis (VEGFR1, -2, -3, TIE2), oncogenesis (KIT, RET, RAF-1, BRAF, BRAFV600E), and the tumour microenvironment (PDGFR, FGFR) [3].

287

5.2. Cisplatin (CisPt)

288 289 290 291 292 293 294 295 296 297 298 299 300 301 302 303 304 305 306 307 308 309 310 311 312 313 314 315 316 317 318 319

A platinum-based compound, cisplatin ( cis-diamminedichloroplatinum(II)) displays clinical activity against a wide variety of solid tumours, including colorectal carcinoma and malignant melanoma. The primary cellular target for cisplatin is DNA [61]. Its cytotoxic mode of action is based on DNA adduct formation, which affects many DNA-dependent cellular functions, including inhibition of replication and transcription, cell cycle arrest, and DNA damage leading to cell death and apoptosis [62–65]. The cytotoxic effect is ascribed to interaction with nucleophilic N7-sites of purine bases in DNA to form DNA–protein, DNA–DNA interstrand, but mainly intrastrand cross-links [66]. Damage recognition proteins (DRPs) of the mismatch repair (MMR) complex bind to physical distortions in the DNA and initiate the sequence of events extending from the formation of DNA adducts to the completion of the apoptosis [67]. Cisplatin resistant cells exhibit a variety of mechanisms of resistance, which arise from intracellular changes that either prevent cisplatin from interacting with DNA, interfere with DNA damage signals from activating the apoptotic machinery, or both [67]. Reduction of the extent of DNA damage increases cisplatin resistance. Reduced intracellular accumulation of the drug is a significant mechanism of resistance. Cisplatin resistant cells were shown to have a pleiotropic defect resulting in reduced plasma membrane receptors and transporters and reduced endocytosis [68] and passive drug diffusion [69], with no evidence for an energy-dependent efflux pump for cisplatin [70,71]. Another significant factor in cisplatin resistance is the increased inactivation by conjugation reaction between cisplatin and thiol-containing molecules (e.g.: GSH). Increased DNA damage repair, which attenuates the apoptotic process, is also responsible for the development of resistance.

Forty-eight ABC transporters identified in the human genome are grouped into seven subfamilies (ABCA – ABCG), based on phylogenetic analysis – similarity in the gene structure (half versus full transporters), order of the domains, and sequence homology in the nucleotide-binding domains (NBDs) and transmembrane domains (TMDs) [75]. Several ABC transporters have been found to be overexpressed in MDR cell lines. The three major proteins involved in cancer MDR are MDR1 (P-gp, ABCB1), MRP1 (ABCC1) and ABCG2 (BCRP, MXR). Numerous studies have found a very good correlation between ABC transporters´ expression levels and poor prognosis and response to chemotherapy [76,77]. In our work, we were able to show functional relationship between resistance to 5-FU based therapy and ABCC11 expression [8].

340 341 342 343 344 345 346 347 348 349 350 351 352 353

6.2. Overcoming MDR with inhibitors

354

320

6. Multidrug resistance (MDR)

321 322 323 324 325 326 327 328

Simultaneous resistance of tumour cells to the cytostatic or cytotoxic actions of multiple structurally unrelated drugs that do not have a common mechanism of action, a phenomenon that is known as MDR [71], might explain why treatment regimens that combine multiple agents with different targets are not more effective [72]. MDR cancer cells are endowed with multiple mechanisms for survival, which can result from:

330 331 332 333 334 335 336

      

Due to the promiscuity of the ABC-transporters, a large number of high affinity substrates, which increase the cytotoxic action of MDR-related anticancer drugs by preventing their efflux from the target cells, have been found. The first generation of P-gp inhibitors (modulators) were drugs already in use for treatment of other medical conditions. It included compounds of diverse structure and function such as calcium channel blockers (verapamil), immunosuppressants (cyclosporin A), antibiotics (erythromycin), antimalarials (quinine) or anti-steroids (tamoxifen) [78]. Since these were not specifically developed for inhibiting MDR, they suffered from the dual problems of high toxicity and low efficacy at tolerable doses. Second-generation inhibitors, such as valspodar, biricodar and dexniguldipine, were synthesized around first-generation pharmacophores to increase the affinity to P-gp while reducing doselimiting toxicities. Nevertheless, reduced clearance of the anticancer drug led to increased toxicity in cases where both the treatment drug and the modulator were substrates for cytochrome P450 3A. Third-generation inhibitors, such as zosuquidar, tariquidar, elacridar and ontogen, were specifically designed to have high affinity to P-gp and low pharmacokinetic interactions [79]. It was thought that the targeted agents would be so specific that resistance would not develop. The disappointing fact is that most of these therapies improve progression-free survival but still fail to induce cures. The main goal of these modulators is to maximize therapeutic outcome by synergic or additive action without increasing cytotoxicity. The drugs used in combination should act by different mechanisms, have some efficacy by themselves, have different toxicity profiles and different mechanisms of resistance development. Another very promising strategy is the development of the drug delivery system based on liposomes to enhance drug delivery to tumour cells and to reduce plasma protein binding [80]. Novel systematic computational tool DrugComboRanker to prioritize synergistic drug combinations and uncover their mechanisms of action in cancer treatment was described [81]. Gene therapy can be used in combination with conventional chemotherapy for primary tumours and also for treatment of metastases. Development of therapies targeting CSC features holds hope for improvement of survival of cancer patients, especially patients with metastatic disease or with aggressive tumours, like glioblastoma, where in addition disseminated tumour cells in brain

355 356 357 358 359 360 361 362 363 364 365 366 367 368 369 370 371 372 373 374 375 376 377 378 379 380 381 382 383 384 385 386 387 388 389 390 391 392 393 394 395 396 397

limited drug uptake; enhanced drug efflux; affected membrane lipids (e.g. ceramides) [73]; increased drug metabolism; altered drug targets; detoxification by compartmentalization; blocked programmed cell death (apoptosis);

Please cite this article in press as: Kozovska Z, et al. Colon cancer: Cancer stem cells markers, drug resistance and treatment. Biomed Pharmacother (2014), http://dx.doi.org/10.1016/j.biopha.2014.10.019

G Model

BIOPHA 3475 1–6 Z. Kozovska et al. / Biomedicine & Pharmacotherapy xxx (2014) xxx–xxx

398 399 400 401

tissue have to be attacked. Prodrug cancer gene therapy driven by engineered MSCs might be one out of several treatment modalities fulfilling these requirements [82]. It remains to be evaluated if this type of treatment can target cells with CSC properties.

402

7. Conclusions

403 404 405 406 407 408 409 410 411 412 413 414 415 416 417 418 419 420 421 422

We summarized our knowledge about current approaches to colon cancer treatment, colon CSCs and their drug resistance. Only precise identification of colon CSC and their properties can help to design effective anticancer therapy. Efficient cancer therapy which can target CSCs together with conventional chemotherapy should be applied at the same time. This strategy together with the gene therapy using genetically modified MSC, which are able to convert nontoxic prodrug to highly toxic chemotherapeutic at the site of the tumour (so-called gene-directed enzyme/prodrug therapy; suicide gene therapy), seems to be the anticancer therapy of the near future. Combinations of traditional, targeted and stem celldirected gene therapy could significantly advance the treatment of cancer [8–11,82]. There is experimental evidence that the CSC marker expression is linked to increased drug resistance [83]. Our data confirmed the same phenomenon in thyroid cancer cells linking CD133+ marker and their resistance to 5-FU [84]. It remains to be further examined whether decreased expression of CSC markers also leads to decreased expression of MDR efflux pumps and at the same time increases sensitivity of the tumour to chemotherapy.

423

Disclosure of interest

424 425

The authors declare that they have no conflicts of interest concerning this article.

426

Acknowledgements

427 428 429 430 431 432 433 434 435 436 437 438 439 440 441 442 443 444 445 446 447 448 449 450 451 452 453 454 455 456 457 458 459

We thank Dr. M. Cihova for reading the manuscript. The studies and experiments mentioned in this study were performed with the Q2 kind support provided by the Slovak Research and Development Agency under the contract No. APVV-0230-11 and APVV-0052-12; Slovak Cancer Research Foundation; VEGA grants No. 2/0171/13 and 2/0130/13. References [1] Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell 2011;144:646–74. [2] Marin JJ, Monte MJ, Blazquez AG, Macias RI, Serrano MA, Briz O. The role of reduced intracellular concentrations of active drugs in the lack of response to anticancer chemotherapy. Acta Pharmacol Sin 2014;35:1–10. [3] Recondo GJ, Diaz-Canton E, de la Vega M, Greco M, Recondo GS, Valsecchi ME. Advances and new perspectives in the treatment of metastatic colon cancer. World J Gastrointest Oncol 2014;6:211–24. [4] Hong WK, Research AAfC, Hait W. Holland Frei Cancer Medicine Eight. People’s Medical Publishing House; 2010. [5] Dutour A, Leclers D, Monteil J, Paraf F, Charissoux JL, Rousseau R, et al. Noninvasive imaging correlates with histological and molecular characteristics of an osteosarcoma model: application for early detection and follow-up of MDR phenotype. Anticancer Res 2007;27:4171–8. [6] Langley RR, Fidler IJ. The biology of brain metastasis. Clin Chem 2013;59: 180–9. [7] La Porta CA. Drug resistance in melanoma: new perspectives. Curr Med Chem 2007;14:387–91. [8] Matuskova M, Baranovicova L, Kozovska Z, Durinikova E, Pastorakova A, Hunakova L, et al. Intrinsic properties of tumour cells have a key impact on the bystander effect mediated by genetically engineered mesenchymal stromal cells. J Gene Med 2012;14:776–87. [9] Altanerova V, Cihova M, Babic M, Rychly B, Ondicova K, Mravec B, et al. Human adipose tissue-derived mesenchymal stem cells expressing yeast cytosinedeaminase: uracil phosphoribosyltransferase inhibit intracerebral rat glioblastoma. Int J Cancer 2012;130:2455–63.

5

[10] Kucerova L, Poturnajova M, Tyciakova S, Matuskova M. Increased proliferation and chemosensitivity of human mesenchymal stromal cells expressing fusion yeast cytosine deaminase. Stem Cell Res 2012;8:247–58. [11] Matuskova M, Hlubinova K, Pastorakova A, Hunakova L, Altanerova V, Altaner C, et al. HSV-tk expressing mesenchymal stem cells exert bystander effect on human glioblastoma cells. Cancer Lett 2010;290:58–67. [12] Durinikova E, Kucerova L, Matuskova M. Mesenchymal stromal cells retrovirally transduced with prodrug-converting genes are suitable vehicles for cancer gene therapy. Acta Virol 2014;58:1–13. [13] Fearon ER, Vogelstein B. A genetic model for colorectal tumorigenesis. Cell 1990;61:759–67. [14] Poste G, Fidler IJ. The pathogenesis of cancer metastasis. Nature 1980;283:139–46. [15] Heng HH, Stevens JB, Liu G, Bremer SW, Ye KJ, Reddy PV, et al. Stochastic cancer progression driven by non-clonal chromosome aberrations. J Cell Physiol 2006;208:461–72. [16] Grady WM, Markowitz S. Genomic instability and colorectal cancer. Curr Opin Gastroenterol 2000;16:62–7. [17] Diaz-Cano SJ. Tumor heterogeneity: mechanisms and bases for a reliable application of molecular marker design. Int J Mol Sci 2012;13:1951–2011. [18] Dalerba P, Dylla SJ, Park IK, Liu R, Wang X, Cho RW, et al. Phenotypic characterization of human colorectal cancer stem cells. Proc Natl Acad Sci U S A 2007;104:10158–63. [19] Heppner GH. Tumor heterogeneity. Cancer Res 1984;44:2259–65. [20] Lobo NA, Shimono Y, Qian D, Clarke MF. The biology of cancer stem cells. Annu Rev Cell Dev Biol 2007;23:675–99. [21] Ward RJ, Dirks PB. Cancer stem cells: at the headwaters of tumor development. Annu Rev Pathol 2007;2:175–89. [22] Cho RW, Clarke MF. Recent advances in cancer stem cells. Curr Opin Genet Dev 2008;18:48–53. [23] Morrison SJ, Kimble J. Asymmetric and symmetric stem-cell divisions in development and cancer. Nature 2006;441:1068–74. [24] Morrison SJ, Qian D, Jerabek L, Thiel BA, Park IK, Ford PS, et al. A genetic determinant that specifically regulates the frequency of hematopoietic stem cells. J Immunol 2002;168:635–42. [25] Moitra K, Lou H, Dean M. Multidrug efflux pumps and cancer stem cells: insights into multidrug resistance and therapeutic development. Clin Pharmacol Ther 2011;89:491–502. [26] O’Brien CA, Pollett A, Gallinger S, Dick JE. A human colon cancer cell capable of initiating tumour growth in immunodeficient mice. Nature 2007;445:106–10. [27] Ricci-Vitiani L, Lombardi DG, Pilozzi E, Biffoni M, Todaro M, Peschle C, et al. Identification and expansion of human colon-cancer-initiating cells. Nature 2007;445:111–5. [28] Huang EH, Hynes MJ, Zhang T, Ginestier C, Dontu G, Appelman H, et al. Aldehyde dehydrogenase 1 is a marker for normal and malignant human colonic stem cells (SC) and tracks SC overpopulation during colon tumorigenesis. Cancer Res 2009;69:3382–9. [29] Ke J, Wu X, He X, Lian L, Zou Y, Wang H, et al. A subpopulation of CD24(+) cells in colon cancer cell lines possess stem cell characteristics. Neoplasma 2012;59:282–8. [30] Wilson BJ, Schatton T, Frank MH, Frank NY. Colorectal Cancer Stem Cells: Biology and Therapeutic Implications. Curr Colorectal Cancer Rep 2011;7:128–35. [31] Vermeulen L, Todaro M, de Sousa Mello F, Sprick MR, Kemper K, Perez Alea M, et al. Single-cell cloning of colon cancer stem cells reveals a multi-lineage differentiation capacity. Proc Natl Acad Sci U S A 2008;105:13427–32. [32] Ong CW, Kim LG, Kong HH, Low LY, Iacopetta B, Soong R, et al. CD133 expression predicts for non-response to chemotherapy in colorectal cancer. Mod Pathol 2010;23:450–7. [33] Du L, Wang H, He L, Zhang J, Ni B, Wang X, et al. CD44 is of functional importance for colorectal cancer stem cells. Clin Cancer Res 2008;14:6751–60. [34] Vasiliou V, Nebert DW. Analysis and update of the human aldehyde dehydrogenase (ALDH) gene family. Hum Genomics 2005;2:138–43. [35] Landen Jr CN, Goodman B, Katre AA, Steg AD, Nick AM, Stone RL, et al. Targeting aldehyde dehydrogenase cancer stem cells in ovarian cancer. Mol Cancer Ther 2010;9:3186–99. [36] Ma I, Allan AL. The role of human aldehyde dehydrogenase in normal and cancer stem cells. Stem Cell Rev 2011;7:292–306. [37] Koppaka V, Thompson DC, Chen Y, Ellermann M, Nicolaou KC, Juvonen RO, et al. Aldehyde dehydrogenase inhibitors: a comprehensive review of the pharmacology, mechanism of action, substrate specificity, and clinical application, Pharmacol Rev; 2012;64:520–39. [38] Su Y, Qiu Q, Zhang X, Jiang Z, Leng Q, Liu Z, et al. Aldehyde dehydrogenase 1 A1positive cell population is enriched in tumor-initiating cells and associated with progression of bladder cancer. Cancer Epidemiol Biomarkers Prev 2010;19:327–37. [39] Kim MP, Fleming JB, Wang H, Abbruzzese JL, Choi W, Kopetz S, et al. ALDH activity selectively defines an enhanced tumor-initiating cell population relative to CD133 expression in human pancreatic adenocarcinoma. PLoS ONE 2011;6:e20636. [40] Honoki K, Fujii H, Kubo A, Kido A, Mori T, Tanaka Y, et al. Possible involvement of stem-like populations with elevated ALDH1 in sarcomas for chemotherapeutic drug resistance. Oncol Rep 2010;24:501–5. [41] Croker AK, Allan AL. Inhibition of aldehyde dehydrogenase (ALDH) activity reduces chemotherapy and radiation resistance of stem-like ALDHhiCD44(+) human breast cancer cells. Breast Cancer Res Treat 2012;133:75–87.

Please cite this article in press as: Kozovska Z, et al. Colon cancer: Cancer stem cells markers, drug resistance and treatment. Biomed Pharmacother (2014), http://dx.doi.org/10.1016/j.biopha.2014.10.019

460 461 462 463 464 465 466 467 468 469 470 471 472 473 474 475 476 477 478 479 480 481 482 483 484 485 486 487 488 489 490 491 492 493 494 495 496 497 498 499 500 501 502 503 504 505 506 507 508 509 510 511 512 513 514 515 516 517 518 519 520 521 522 523 524 525 526 527 528 529 530 531 532 533 534 535 536 537 538 539 540 541 542 543 544 545

G Model

BIOPHA 3475 1–6 6

546 547 548 549 550 551 552 553 554 555 556 557 558 559 560 561 562 563 564 565 566 567 568 569 570 571 572 573 574 575 576 577 578 579 580 581 582 583 584 585 586 587 588 589 590 591 592 593 594 595 596 597 598 599 600 601 602 603 604

Z. Kozovska et al. / Biomedicine & Pharmacotherapy xxx (2014) xxx–xxx

[42] Kang EJ, Jung H, Woo OH, Park KH, Woo SU, Yang DS, et al. Association of aldehyde dehydrogenase 1 expression and biologically aggressive features in breast cancer. Neoplasma 2014;61:352–62. [43] Zhang M, Shoeb M, Goswamy J, Liu P, Xiao TL, Hogan D, et al. Overexpression of aldehyde dehydrogenase 1A1 reduces oxidation-induced toxicity in SH-SY5Y neuroblastoma cells. J Neurosci Res 2010;88:686–94. [44] Lawenda BD, Kelly KM, Ladas EJ, Sagar SM, Vickers A, Blumberg JB. Should supplemental antioxidant administration be avoided during chemotherapy and radiation therapy? J Natl Cancer Inst 2008;100:773–83. [45] Donnenberg VS, Donnenberg AD. Multiple drug resistance in cancer revisited: the cancer stem cell hypothesis. J Clin Pharmacol 2005;45:872–7. [46] Zeimet AG, Reimer D, Sopper S, Boesch M, Martowicz A, Roessler J, et al. Ovarian cancer stem cells. Neoplasma 2012;59:747–55. [47] Dalerba P, Cho RW, Clarke MF. Cancer stem cells: models and concepts. Annu Rev Med 2007;58:267–84. [48] Popek S, Tsikitis VL. Neoadjuvant vs adjuvant pelvic radiotherapy for locally advanced rectal cancer: Which is superior? World J Gastroenterol 2011;17: 848–54. [49] Benson 3rd AB, Schrag D, Somerfield MR, Cohen AM, Figueredo AT, Flynn PJ, et al. American Society of Clinical Oncology recommendations on adjuvant chemotherapy for stage II colon cancer. J Clin Oncol 2004;22:3408–19. [50] Wohlhueter RM, McIvor RS, Plagemann PG. Facilitated transport of uracil and 5-fluorouracil, and permeation of orotic acid into cultured mammalian cells. J Cell Physiol 1980;104:309–19. [51] Matherly LH, Czajkowski CA, Muench SP, Psiakis JT. Role for cytosolic folatebinding proteins in the compartmentation of endogenous tetrahydrofolates and the 5-formyl tetrahydrofolate-mediated enhancement of 5-fluoro-2’deoxyuridine antitumor activity in vitro. Cancer Res 1990;50:3262–9. [52] Park JG, Collins JM, Gazdar AF, Allegra CJ, Steinberg SM, Greene RF, et al. Enhancement of fluorinated pyrimidine-induced cytotoxicity by leucovorin in human colorectal carcinoma cell lines. J Natl Cancer Inst 1988;80:1560–4. [53] Nadal JC, Van Groeningen CJ, Pinedo HM, Peters GJ. In vivo potentiation of 5fluorouracil by leucovorin in murine colon carcinoma. Biomed Pharmacother 1988;42:387–93. [54] Wright JE, Dreyfuss A, el-Magharbel I, Trites D, Jones SM, Holden SA, et al. Selective expansion of 5,10-methylenetetrahydrofolate pools and modulation of 5-fluorouracil antitumor activity by leucovorin in vivo. Cancer Res 1989;49:2592–6. [55] Miwa M, Ura M, Nishida M, Sawada N, Ishikawa T, Mori K, et al. Design of a novel oral fluoropyrimidine carbamate, capecitabine, which generates 5fluorouracil selectively in tumours by enzymes concentrated in human liver and cancer tissue. Eur J Cancer 1998;34:1274–81. [56] Cao D, Russell RL, Zhang D, Leffert JJ, Pizzorno G. Uridine phosphorylase (/) murine embryonic stem cells clarify the key role of this enzyme in the regulation of the pyrimidine salvage pathway and in the activation of fluoropyrimidines. Cancer Res 2002;62:2313–7. [57] Longley DB, Harkin DP, Johnston PG. 5-fluorouracil: mechanisms of action and clinical strategies. Nat Rev Cancer 2003;3:330–8. [58] Hsiang YH, Hertzberg R, Hecht S, Liu LF. Camptothecin induces protein-linked DNA breaks via mammalian DNA topoisomerase I. J Biol Chem 1985;260:14873–8. [59] Woynarowski JM, Chapman WG, Napier C, Herzig MC, Juniewicz P. Sequenceand region-specificity of oxaliplatin adducts in naked and cellular DNA. Mol Pharmacol 1998;54:770–7. [60] Segal NH, Saltz LB. Evolving treatment of advanced colon cancer. Annu Rev Med 2009;60:207–19. [61] Eastman A. Characterization of the adducts produced in DNA by cis-diamminedichloroplatinum(II) and cis-dichloro(ethylenediamine)platinum(II). Biochemistry 1983;22:3927–33.

[62] Chaney SG, Campbell SL, Bassett E, Wu Y. Recognition and processing of cisplatin- and oxaliplatin-DNA adducts. Crit Rev Oncol Hematol 2005;53: 3–11. [63] Zdraveski ZZ, Mello JA, Farinelli CK, Essigmann JM, Marinus MG. MutS preferentially recognizes cisplatin- over oxaliplatin-modified DNA. J Biol Chem 2002;277:1255–60. [64] Wang D, Lippard SJ. Cellular processing of platinum anticancer drugs. Nat Rev Drug Discov 2005;4:307–20. [65] Vogelstein B, Lane D, Levine AJ. Surfing the p53 network. Nature 2000;408:307–10. [66] Eastman A. The formation, isolation and characterization of DNA adducts produced by anticancer platinum complexes. Pharmacol Ther 1987;34: 155–66. [67] Siddik ZH. Cisplatin: mode of cytotoxic action and molecular basis of resistance. Oncogene 2003;22:7265–79. [68] Shen D, Pastan I, Gottesman MM. Cross-resistance to methotrexate and metals in human cisplatin-resistant cell lines results from a pleiotropic defect in accumulation of these compounds associated with reduced plasma membrane binding proteins. Cancer Res 1998;58:268–75. [69] Kelland LR. Preclinical perspectives on platinum resistance. Drugs 2000;59(Suppl. 4):1–8 [discussion 37–38]. [70] Shen DW, Goldenberg S, Pastan I, Gottesman MM. Decreased accumulation of [14C]carboplatin in human cisplatin-resistant cells results from reduced energy-dependent uptake. J Cell Physiol 2000;183:108–16. [71] Gottesman MM. Mechanisms of cancer drug resistance. Annu Rev Med 2002;53:615–27. [72] Gottesman MM, Fojo T, Bates SE. Multidrug resistance in cancer: role of ATPdependent transporters. Nat Rev Cancer 2002;2:48–58. [73] Liu YY, Han TY, Giuliano AE, Cabot MC. Ceramide glycosylation potentiates cellular multidrug resistance. FASEB J 2001;15:719–30. [74] Synold TW, Dussault I, Forman BM. The orphan nuclear receptor SXR coordinately regulates drug metabolism and efflux. Nat Med 2001;7:584–90. [75] Dean M, Rzhetsky A, Allikmets R. The human ATP-binding cassette (ABC) transporter superfamily. Genome Res 2001;11:1156–66. [76] Polgar O, Bates SE. ABC transporters in the balance: is there a role in multidrug resistance? Biochem Soc Trans 2005;33:241–5. [77] Steinbach D, Legrand O. ABC transporters and drug resistance in leukemia: was P-gp nothing but the first head of the Hydra? Leukemia 2007;21: 1172–6. [78] Ford JM, Hait WN. Pharmacology of drugs that alter multidrug resistance in cancer. Pharmacol Rev 1990;42:155–99. [79] Glavinas H, Krajcsi P, Cserepes J, Sarkadi B. The role of ABC transporters in drug resistance, metabolism and toxicity. Curr Drug Deliv 2004;1:27–42. [80] Ye P, Zhang W, Yang T, Lu Y, Lu M, Gai Y, et al. Folate receptor-targeted liposomes enhanced the antitumor potency of imatinib through the combination of active targeting and molecular targeting. Int J Nanomedicine 2014;9:2167–78. [81] Huang L, Li F, Sheng J, Xia X, Ma J, Zhan M, et al. DrugComboRanker: drug combination discovery based on target network analysis. Bioinformatics 2014;30:i228–36. [82] Cihova M, Altanerova V, Altaner C. Stem cell based cancer gene therapy. Mol Pharm 2011;8:1480–7. [83] Todaro M, Alea MP, Di Stefano AB, Cammareri P, Vermeulen L, Iovino F, et al. Colon cancer stem cells dictate tumor growth and resist cell death by production of interleukin-4. Cell Stem Cell 2007;1:389–402. [84] Kucerova L, Feketeova L, Kozovska Z, Poturnajova M, Matuskova M, Nencka R, et al. In vivo 5FU-exposed human medullary thyroid carcinoma cells contain a chemoresistant CD133+ tumor-initiating cell subset. Thyroid 2014;24: 520–32.

Please cite this article in press as: Kozovska Z, et al. Colon cancer: Cancer stem cells markers, drug resistance and treatment. Biomed Pharmacother (2014), http://dx.doi.org/10.1016/j.biopha.2014.10.019

605 606 607 608 609 610 611 612 613 614 615 616 617 618 619 620 621 622 623 624 625 626 627 628 629 630 631 632 633 634 635 636 637 638 639 640 641 642 643 644 645 646 647 648 649 650 651 652 653 654 655 656 657 658 659 660 661 662 663

Colon cancer: cancer stem cells markers, drug resistance and treatment.

Malignant tumours consist of heterogeneous populations of tumour cells. Cancer stem cells (CSC) represent a population of cells within a tumour with h...
343KB Sizes 0 Downloads 9 Views