GET THE APP

Chemoresistance in Cancer Stem Cells and Strategies to Overcome R
Chemotherapy: Open Access

Chemotherapy: Open Access
Open Access

ISSN: 2167-7700

+44 1223 790975

Review Article - (2014) Volume 3, Issue 1

Chemoresistance in Cancer Stem Cells and Strategies to Overcome Resistance

Margaret Lois Thomas1, Krysta Mila Coyle1, Mohammad Sultan1, Ahmad Vaghar-Kashani1 and Paola Marcato2*
1Department of Biology, Uppsala University, Sweden
2Department of Pathology, Dalhousie University, Halifax, NS, Canada
*Corresponding Author: Paola Marcato, Department of Pathology, Dalhousie University, Rm 11-C1, 5850 College Street, Halifax, NS, B3H 4R2, NS, Canada, Tel: (902) 494-4231 Email:

Abstract

In cancers, there exists a subpopulation of cells which are referred to as cancer stem cells (CSCs) or tumorinitiating cells that have enhanced tumor-initiating capacity and metastatic potential, and drive tumor progression.Since the initial identification of acute myeloid leukemia CSCs in 1997, CSCs have been found in many types ofcancer and have intrinsic resistance to the current chemotherapeutic strategies. With increased levels of detoxifyingenzymes, enhanced DNA repair abilities, impressive efflux capacity, and a slower cell-cycle; CSCs present aformidable obstacle against effective chemotherapy. Several methods of specifically targeting CSCs have beendeveloped in recent years, and these compounds have potential as adjuvant therapies. The following is a review ofthe mechanisms responsible for chemoresistance in CSCs, with an emphasis on potential strategies to overcomethis resistance .

Keywords: Cancer stem cells, Chemoresistance, Targeted therapies

Introduction to Cancer Stem Cells

For many years, tumors had been thought of as monoclonal populations of rapidly dividing cells, and that all cells had equivalent cancer-initiating abilities. Over time, it has become evident that tumors are heterogeneous in nature and that certain cells have increased tumor-initiating abilities. These tumor-initiating cells are also referred to as cancer stem cells (CSCs) and are hypothesized to self-renew (maintaining a population of CSCs) and to differentiate into less tumorigenic Non-CSCs [1]. First identified by Bonnet & Dick as the tumor-initiating cells of acute myeloid leukemia, CSCs were later isolated from solid tumors by Al-hajj et al. in breast cancer, as well as in brain tumours by Singh et al. [2-4]. Since these seminal publications, CSCs have been isolated from many cancers, including colon, pancreatic, liver, and prostate, lung, head and neck, ovarian, and stomach cancers [5-12].

CSCs are functionally defined by their ability to initiate new tumors in severely immunocompromised mice [1]. A number of biomarkers are associated with more tumorigenic cells and can be used in combination to identify or isolate CSCs. These biomarkers are cancer-type specific and are often cell surface markers or based on increased aldehyde dehydrogenase (ALDH) activity as measured by the Aldefluor assay. For example, in breast cancer, cells sorted based on CD44+CD24- are enriched for CSCs, and high ALDH activity is also found in this CSC-enriched population [3,13]. Throughout this paper, CSC biomarkers will be referred to these biomarkers are either well-established identifiers for the cancer type discussed, or were verified by the authors to be prevalent in the population of cells that initiated tumors in immunocompromised mice.

As research on CSCs gained notoriety [14-16], it became clear that these seemingly ubiquitous tumor-initiating cells were resistant to radiation and chemotherapy. The presence of these cells in tumors contributes to a patient's likelihood of recurrence post-treatment, and may be the cause of resistance in tumors that do not respond to anti-cancer therapies. Herein, we will review the literature with regards to chemotherapeutics that CSCs are resistant to, mechanisms of CSC chemotherapeutic resistance and finally we discuss novel targeted therapies that are being developed which show efficacy towards killing CSCs.

Drug Resistance in Cancer Stem Cells

The success of most chemotherapeutics is judged on the drug's ability to decrease tumor size or induce short-term remission. While this measure of success is intuitive and many drugs evaluated by these criteria are used in effective chemotherapeutic regimens, it is becoming increasingly evident that in some cases, eliminating the bulk of cancer cells may effectively select for resistant cells. As we discuss below, CSCs have a higher intrinsic resistance to chemotherapy than do normal cancer cells, and may be the source of post-therapy relapse [17] (Figure 1).

chemotherapy-conventional-therapies

Figure 1: Treatment of tumors with conventional therapies fails to effectively target CSCs, potentially leading to increased chance of recurrence. (A) Conventional chemotherapies and radiation induce tumor regression; however, there is a long-term risk of relapse as the surviving resistant CSCs can initiate a new tumor. (B) Conventional therapies with the addition of CSC targeted therapy lead to both tumor regression and elimination of CSCs, resulting in a decreased chance of recurrence.

Cancer cells may acquire resistance to chemotherapy, or may have a high basal level of resistance through a variety of mechanisms (Table 1). These mechanisms have been well studied in cancer cells, and the same concept is being applied to CSCs. As discussed in detail later, there is evidence of increased drug inactivation through increased expression of detoxifying ALDH enzymes, enhanced DNA repair activity which thwarts platinum and alkylating agents, reduced drug activation via quiescence, and increased drug efflux by upregulation of ABC transporters.

Mechanisms of Chemoresistance
Increases drug export
Increased drug inactivation
Reduced Drug Uptake
Reduced Drug Activation
Changes in Drug Target interaction
Enhanced EGFR and MAPK/ERK signalling
Enhanced DNA repair
Inhibition of apoptosis

Table 1: General mechanisms of chemoresistance in cancer cells Footnote: Mechanisms of chemoresistance that are more prevalent in CSCs are in bold.

Chemoresistance of Cancer Stem Cells Due to Aldehyde Dehydrogenase Activity

The Aldefluor assay (Stem Cell Technologies, Inc.) was originally designed to isolate viable hematopoietic stem cells in human umbilical cord blood by identifying cells with high ALDH enzymatic activity [18]. The population of Aldefluor+ cells is often referred to as ALDH+ or ALDH bright. Highly tumorigenic cells isolated based on high Aldefluor activity were first identified in breast carcinomas and leukemia [19,20] and since then, Aldefluor+ cells have efficiently initiated xenograft tumors of liver, head and neck, stomach, lung, pancreatic, cervical, thyroid, prostate, colon, bladder, and ovarian cancers [7,21-29].

ALDHs are a super-family of enzymes involved in oxidizing aldehydes to carboxylic acids, and increased activity of some isoforms is associated with detoxification capabilities [30]. Due to the general function of ALDH enzymes in detoxification, it has been hypothesized that Aldefluor activity associated with CSCs would confer resistance to chemotherapeutics as well. Indeed, in a breast cancer study, tumor samples with high ALDH protein levels were associated with patient resistance to paclitaxel and epirubicin [31]. Additionally, Aldefluor+ cells from lung cancer cells lines demonstrated a high resistance to multiple chemotherapeutic agents (cisplatin, gemcitabine, vinorelbine, docetaxel, doxorubicin and daunorubicin) when compared to Aldefluor- cells [22]. Though none of the aforementioned drugs seem to be metabolized directly by ALDH; other chemotherapeutics, such as the alkylating agent cyclophosphamide, are detoxified by the enzyme.

Abundant in the environment, and unavoidable in living cells, alkylating agents are a family of compounds defined by their ability to add alkyl groups to a variety of molecules [32]. This process damages DNA by generating covalent adducts that lead to mutations in the sequence. These mutations may result in apoptosis or replication failure. The DNA damaging effects of alkylating agents are utilized in chemotherapy for a variety of cancers; examples of alkylating agents include cyclophosphamide, melphalan, ifosfamide, carmustine, procarbazine, and temzolomide. While these agents are usually effective against most non-CSCs, it seems that CSCs are resistant to these drugs via ALDH detoxification and through increased DNA repair.

Cyclophosphamide is used to treat breast, lung and ovarian cancers, as well as acute myeloid leukemia, chronic myeloid leukemia, neuroblastoma, and lymphoma [33-38]. Cyclophosphamide is an inactive prodrug that is converted to 4-hydroxycyclophosphamide and aldophosphamide once inside of the cell, and eventually results in phosphoramide mustard which forms the DNA crosslinks [39]. Despite widespread effectiveness in many cancer types [40], cyclophosphamide is less effective in the presence of ALDH which interferes with the drug's decomposition to aldophosphamide[39]. Metabolism of cyclophosphamide by ALDH has been suspected for decades [41], and the enzyme's role in chemotherapy resistance was first elucidated for leukemia. Studies using the murine leukemia cell line L1210 found that 4-hydroxycyclophosphamide was detoxified by ALDH and that cell line resistance could be attributed to ALDH activity [42-44].

There are 19 isoforms of ALDH present in the human genome [30], and the specific isoforms responsible for cyclophosphamide detoxification in cancer cells is not fully known, however some studies indicate the involvement of at least two isoforms. Induced expression of ALDH1A1 in L1210 cells led to increased resistance to cyclophosphamide [45]. Moreb et al. determined that siRNA knockdown of isoforms ALDH1A1 and ALDH3A1 resulted in an 84% increase in cyclophosphamide toxicity in lung adenocarcinoma cell line A549 [46]. In breast cancer patient tumors, immunohistological staining determined that increased expression of ALDH1A1 and ALDH3A1 was found in tumors that did not respond to cyclophosphamide therapy, and in tumors that had undergone cyclophosphamide therapy [47]. These results implicate ALDH1A1 and ALDH3A1 in resistance to cyclophosphamide. Notably, expression of the ALDH1A1 isoform is associated with the Aldefluor activity of the CSCs of many cancers [48-51]. Therefore, there is a direct link with ALDH1A1 expression, CSCs and

Chemoresistance of Cancers Stem Cells by Enhanced DNA Repair Mechanisms

The platinum group of chemotherapeutic agents (including the common analogues cisplatin, carboplatin, and oxaliplatin) induce tumor regression by causing DNA damage. Cancer cells often have defective DNA repair pathways, and due to rapid proliferation, these cells are often in S-phase which is a vulnerable phase for DNA damage [52]. Thus, these DNA-damaging chemotherapeutics are selectively deleterious to cancer cells in S-phase, which due to impaired repair mechanisms, are not able to recover from the damage. When the DNA repair cascades are unable to adequately fix the damage, cell-cycle checkpoint components are activated which can recruit additional DNA repair components or activate apoptosis. Data from many studies imply that CSCs have elevated levels of DNA repair [53-61], these provide one explanation for the resistance of some tumor types to platinum agents.

In vitro evidence suggests that CSCs in lung, ovarian and breast cancer cell lines are resistant to DNA-damaging agent cisplatin, since these cells were enriched post-treatment [62]. Similar CSC enrichment was observed in mice bearing breast tumor xenografts post-cisplatin treatment [63]. Perhaps more importantly, primary clinical samples support the hypothesis that CSCs are more resistant to treatment with platinum analogues. Patients with advanced ovarian cancer showed an elevated percentage of CSCs in sampled ascites when they had been treated with cisplatin compared to chemotherapy-naïve patients [64]. In glioblastoma, resistance of CD133+ CSCs to chemotherapeutics was attributed to increased expression of DNA repair and anti-apoptosis proteins [65]. Furthermore, the authors also showed that patients with recurrent glioblastoma had higher expression of CD133 in their tumors post-treatment with chemotherapeutics.

Chemoresistance of Cancer Stem Cells Due to Quiescence

Anti-mitotic drugs target the reorganization of microtubules essential for proper cell division and proliferation (Gascoigne & Taylor, 2011 for review of mechanism) [66]. The two classes of antimitotics currently approved for cancer therapy are vinca alkaloids (vincristine, vinblastine, vindesine and vinorelbine) which prevent the polymerization of microtubules, and taxanes (paclitaxel, docetaxel) that stabilize existing microtubules. Both classes effectively inhibit the formation of the mitotic spindle, inhibiting the mitotic phase of the cell cycle. Many other antimitotic agents are in development; however, they have not yet been approved for clinical use [67]. The relation of CSCs to progression through the cell cycle is inconclusive; however, there is evidence to suggest that CSCs may be more quiescent or slower-cycling than their associated non-CSCs. Quiescence and a slower progression through the cell cycle in CSCs would likely render these cells less susceptible to cell-cycle targeted therapies such as the antimitotic class of chemotherapeutics [68].

In glioblastoma, CD133+- identified CSCs were resistant to a variety of chemotherapeutic agents, including paclitaxel [65]. A later study, using CD133+ cells derived from patients with treatment-refractory recurrent gliomas, demonstrated that these CSCs had gene expression profiles consistent with quiescent cells [69]. Similarly, Pece et al. determined that the gene expression profiles of high-grade breast tumors matched the profiles generated from quiescent mammary stem cells [70]. Additionally, mammospheres formed from higher-grade cells retained high levels of the quiescence marker, PKH26. Mammospheres are an in vitro measure of CSC tumorigenicity; thus, this data suggests that the CSCs are quiescent when compared to non-CSCs. The authors suggest that tumor progression can be associated with an increase in the number of quiescent cells, which maintain their stem-like tumorigenicity. Inducing cell-cycle entry in these cells may be an interesting option for CSC-targeted therapy, and data from a leukemia model [71], demonstrates that stimulating quiescent CSCs to divide improves the efficacy of cell-cycle dependent chemotherapy.

Chemoresistance of Cancer Stem Cells by Enhanced Drug Efflux Mechanisms

CSCs are enriched in the side population (SP) of tumor cells which have high efflux of Hoescht dye [72-77]. The efflux capacity of the SP is attributed to increased expressed of ATP-binding cassette (ABC) transport proteins ABCB1, ABCC1, and ABCG2 [76,78-81]. These ABC transporters are able to efflux a wide array of chemotherapeutic drugs (e.g. colchicine, doxorubicin, etoposide, vinblastine, and paclitaxel) and their expression is a major cause of multi drug resistance in cancers [82]. Upregulation of these three ABC transporters is often seen in CSCs, and contributes to chemoresistance. For example, increased expression of ABCB1 was shown in the CD44+CD24- identified breast CSCs, which were also comparatively resistant to doxorubicin [83].

Targeting Cancer Stem Cells to Overcome Chemoresistance

CSCs exhibit dysfunctional signalling via three key embryonic pathways: the Wnt/β-catenin, Notch, and Hedgehog (Hh) pathways. The reliance of CSCs on these dysregulated signalling paradigms have generated potential targets for anti-CSC-directed therapies. Here we review several strategies for CSC focused therapy: targeting the Wnt, Notch, and Hh signaling pathways; inhibition of ALDH; and inhibition of ABC transport proteins.

Targeting Wnt Signalling

Wnt signaling is essential for controlled cell proliferation, cell fate decisions during development, and adult stem cell maintenance. Briefly, the binding of extracellular Wnt ligands to membrane-bound frizzled receptors results in the recruitment of disheveled proteins that block glycogen synthase kinase 3 from interacting with its substrates, which include β-catenin [84].

Enhanced Wnt signaling has been observed in the CSCs of many different cancer types. In chronic myeloid leukemia, deletion of β-catenin in combination with imatinib depleted leukemic CSCs; however, deletion of β-catenin alone did not prolong survival in mice [85]. In a breast cancer model with spontaneous lung metastasis, Malanchi et al. determined that periostin, a key regulator of metastatic colonization, recruits Wnt ligands and likely promotes the maintenance of CSCs in their niche [86]. Additional evidence supports aberrant Wnt signaling in lung, colon, and gastric CSCs [87-89]. Strikingly, Teng et al. observed increased β-catenin and OCT-4 (a marker of stemness, see Pesce, 2001) [90] expression in cisplatin-selected A549 lung adenocarcinoma cells.

Resveratrol, a natural polyphenol, has anti-oxidant properties, may have a role in somatic cell reprogramming and can be used in reprogramming mouse embryonic fibroblasts into induced pluripotent stem cells [91]. Furthermore, is hypothesized to exhibit anti-CSC properties via inhibition of Wnt signaling. Resveratrol inhibits fatty acid synthase (FASN) in breast cancer cell-line-derived CSCs, thus suppressing their proliferation [92]. Overexpression of FASN has been associated with increased stability of β-catenin [93] thus, inhibition of FASN likely results in decreased Wnt signaling. More recent findings suggest that resveratrol may be able to inhibit CSC migration and invasion in pancreatic cancer [94]. However, a phase 1 trial of resveratrol treatment in colon cancer inhibited the Wnt pathway in normal colonic mucosa, but not in cancerous colon tissue [95], suggesting toxicity may be a concern with using reservatrol as anti-cancer therapeutic, despite its potential anti-CSC activity.

An isoflavone, genistein, primarily acts as a specific tyrosine-kinase inhibitor [96] and has been demonstrated to inhibit the tumorigenicity of CSCs in prostate, gastric and breast cancers [97-100]. However, it is unclear whether this is due to attenuation of Wnt signaling [101,102] or of the Hedgehog pathway [98,100]. A phase 2 study of genistein on localized prostate cancer patients revealed a decrease in serum PSA in patients treated with genistein [103], as more focal cancer was observed among genistein-treated patients, although effects on CSCs is unclear at this time.

OncoMed Pharmaceuticals, Inc. (OncoMed) and Bayer have initiated a Phase 1b dose-escalating clinical trial of a decoy receptor for Wnt ligand, Fzd8-Fc. This trial, using Fzd8-Fc (OMP-54F28) in combination with paclitaxel and gemcitabine for patients with first-line Stage 4 pancreatic cancer, follows a Phase 1 trial in patients with solid tumors (NCT01608867). In addition, OncoMed has developed an anti-frizzled monoclonal antibody, vantictumab. Results from a Phase 1a study were presented at the European Cancer Congress (2013), suggesting that vantictumab is well tolerated. As well, prolonged stable disease was observed in 3 patients with neuroendocrine tumors. Phase 1b trials of vantictumab in combination with standard chemotherapeutic regimens are ongoing in untreated stage 4 pancreatic cancer (NCT02005315), previously-treated NSCLC (NCT01957007), and locally recurrent or metastatic breast cancer (NCT01973309).

Prism Pharma Co., Ltd. and iNVentiv Health Clinical are investigating PRI-724 in clinical trials. PRI-724 blocks the recruitment of β-catenin to Wnt-responsive elements in the genome, thus preventing activated transcription. Ongoing trials include those in patients with advanced solid tumors, pancreatic cancer, or myeloid malignancies (NCT01606579, NCT01764477, NCT01302405). Therefore, Wnt targeted treatment has potential as an adjuvant therapy in a wide range of cancer types, and presents a very promising avenue for future anti-CSC targeted therapy research.

Targeting Notch Signalling

The Notch signalling pathway is essential for cell-fate determination and pattern formation throughout vertebrate development; its absence results in lethal hyperplasia of the nervous system [104,105]. Notch signalling is initiated by ligand binding of transmembrane receptors Notch1, Notch2, Notch3, and Notch4, which induce proteolytic cleavage of the receptors' intracellular domains by the presenillin-γ–secretase complex [106,107]. The intracellular domains enter the nucleus, and regulate transcription of target genes. Aberrant Notch pathway signalling has been demonstrated in the CSCs of a number of cancers. Notch pathway inhibition depleted CD133+ glioblastoma cells and inhibited tumor growth neurosphere formation [108]. Activating Notch1 mutations are seen in ~50% of T cell acute lymphoblastic leukemia (T-ALL) cases [109] and Notch signalling appears to be of greater importance in CD43+ CD7+ T-ALL leukemia stem cells [110]. Other work has specifically identified Notch4 as contributing to Notch activity in breast CSCs [111].

A phase 1 study of γ-secretase inhibitor (GSI) RO4929097 (Hoffman-La Roche) in refractory metastatic disease or patients with locally advanced solid tumors had mostly favourable results and spawned a number of subsequent clinical studies (e.g. NCT01238133, NCT01154452, NCT01196416) [112]. Another GSI in clinical trials is MK-0752 (Merck), which was shown to decrease CD44+CD24- and Aldefluor+ CSC populations in breast tumor xenografts [113]. In addition, a phase 1 clinical trial of MK0752 in combination with docetaxel decreased breast CSCs over the course of treatment. Unfortunately, due to the involvement of γ-secretase throughout the gastrointestinal tract, dose-limiting gastrointestinal side-effects are of concern in using this particular mode of therapy. Additional toxicity may be observed due to goblet cell metaplasia [114,115]. A third GSI, GSI-18 been shown to deplete the Aldefluor+ CSCs and decrease colony formation and xenograft engraftment in pancreatic cancer models [116]. Similar results were seen in DAOY medulloblastoma cells [117]. We have yet to see if GSI-18 has clinical effects.

OncoMed has also targeted the Notch pathway with demcizumab (OMP-21M18), which is a monoclonal antibody against Delta-like ligand 4 (DLL4) which binds the Notch receptor. Phase 1b trials of demcizmab in combination with gemcitabine and Abraxane in pancreatic cancer (NCT01189929) demonstrated a high clinical benefit rate (Market watch report). Additional clinical trials are ongoing in non-small cell lung carcinoma (NCT01189968) and ovarian cancer (NCT01952249).

Despite a multitude of evidence suggesting reliance of CSCs on aberrant Notch signalling, recent findings indicate that not all CSCs may be reliant [118]. Thus, while targeting Notch signalling may effectively eliminate a significant proportion of CSCs, there may be some CSCs which are not susceptible to this therapy. It remains to be seen if anti-Notch therapies have long-term benefits in cancer patients over standard therapeutics alone.

Targeting Hedgehog Signalling

The Hedgehog (Hh) signalling pathway is involved in the regulation of cell differentiation and proliferation in embryonic development and in the maintenance of adult stem cells. Hh ligands sonic hedgehog (Shh), indian hedgehog (Ihh) and desert hedgehog (Dhh) bind to the cell-surface receptor Patched (PTCH). The binding of ligands to PTCH triggers an accumulation of Smoothened (SMO) within the cell membrane and activates GLI transcriptional regulators. Activated GLI proteins (activators Gli1 and Gli2, and repressor Gli3) accumulate in the nucleus and control transcription of Hh target genes. Targets of Hh signalling include JAG2 and Wnt proteins, resulting in significant cross-talk between the Hh, Wnt, and Notch pathways [119].

There is a fair amount of evidence to suggest that CSCs have higher levels of Hh signalling than their non-CSC counterparts. Hh components are more highly expressed in CD44+CD24- breast CSCs and likely contribute to maintenance of self-renewal potential in these cells [120]. Inhibition of Hh signalling decreased spherogenicity in CD133+ glioma CSCs, decreased self-renewal in Aldefluor+ B-ALL cells, and inhibited clonogenicity of multiple myeloma CSCs [121,122]. In addition to depleting Aldefluor+ cells, Hh inhibition also decreased metastatic spread in a xenograft model of pancreatic cancer and inhibited the growth of human serous ovarian tumor xenografts [123,124].

Several inhibitors of Hh signalling are in various pre-clinical and clinical stages of testing. The inhibitors of Hh ligand-PTCH interactions are perhaps the least advanced in the drug development pipeline. 5E1 is an anti-Shh antibody, and has been shown to inhibit the growth of colon cancer xenografts [125]. Robotnikinin also blocks the Shh-PTCH interaction, but it remains to be seen if it is able to exert anti-cancer effects [126].

Cyclopamine inhibits Hh signalling by binding to SMO [127]. Treatment of mice with cyclopamine or its analogues inhibited the growth of medullablatoma xenografts [128]. In a phase 1 trial in patients with refractory solid tumors, the cyclopamine-derived SMO inhibitor IPI-926 (saridegib, Infinity Pharmaceuticals), contributed to a response in eight of 28 patients [129]. Additionally, there was substantial evidence for the use of IPI-926 in patients with pancreatic cancer [130] however, the phase 2 study (NCT01130142) was stopped after an interim analysis revealed that patients on the saridegib + gemcitabine arm had a median survival of less than 6 months, which is less than the historical gemcitabine-treatment mean of 6 months. There have been a number of other trials of IPI-926 in other malignancies (NCT01310816, NCT01371617), and we await the findings of these studies to properly evaluate the promise of saridegib.

Another inhibitor of SMO, GDC-0449 (vismodegib, Genentech), reduced growth of several lung cancer cell lines via inhibition of the Hoescht-excluding side populations [131]. A phase I trial of GDC-0449 resulted in a number of partial and complete responses among patients with basal-cell carcinoma [132]. Similarly, a case study of GDC-0449 treatment in one patient with refractory metastatic medulloblastoma resulted in rapid regression of the tumor; however, this response was incomplete and transient [133].

With targets further down the Hh pathway, GANT58 and GANT61 inhibit Gli-mediated transcription and blocked xenograft growth of prostate cancer cells [134]. In particular, GANT58 decreased viability of T-ALL cells and functioned synergistically with the AKT inhibitor GSK690693 to induce cell death [135]. Targeting Gli-mediated transcription may be able to reduce cell migration [136] and may also attenuate drug resistance in some cancer types [137] ; however, clinical data is required to definitively answer these questions.

Aldehyde Dehydrogenase Inhibitors

As discussed earlier, increased ALDH activity is a common biomarker of CSCs and is involved in detoxifying certain chemotherapeutics, making ALDH inhibitors a promising avenue for anti-CSC targeted therapy development. Known inhibitors of ALDHs include chloral hydrate, cyanamide, DEAB, gossypol, molinate, pargyline, and disulfiram [138]. Disulfiram has been used for decades to treat alcohol abuse, and recently its potential in cancer treatment has been investigated [139]. The anti-cancer mechanisms of disulfiram are not limited to ALDH inhibition; principally, disulfiram is used to inhibit the proteasome and E3 ligases, and may also be a DNA-demethylating compound [140-143]. Disulfiram inhibits both ALDH1A1 and ALDH2 isoforms [138,144], and has shown anti-CSC effects in breast cancer and GBM [145,146]. Though the anti-CSC effects in the breast cancer studies have been mostly attributed to disulfiram's proteasome inhibition [146-148] in glioblastoma, ALDH inhibition by disulfiram re-sensitized Aldefluor+ cells to gemcitabine cytotoxicity [149]. After showing potential as a chemotherapy-enhancing drug, disulfiram was approved for a phase II clinical trial as a treatment for GBM (NCT0177919).

ABC Transporter Inhibitors

The upregulation of ABC transporters in CSCs and the increased efflux capacity that accompanies that upregulation has prompted investigation of ABC inhibitors as adjuvant therapy. ABC inhibitors have already been tested in various clinical trials; however, they were originally positioned as a broad strategy to increase the efficacy of chemotherapeutics on all cells within a tumor. ABC transporter inhibition should instead be conceptualized as a way to re-sensitize the small population of CSCs with pre-existing intrinsic resistance [150]. ABCB1 inhibitors such as verapamil and cyclosporine A were among the first to be investigated and were effective in treating acute myeloid leukemia, non-small cell lung carcinoma, and breast cancer [151-153]. More recently, ABCG2 inhibition by axitinib has been investigated, and in cell lines from many cancer types, it effectively re-sensitized the side population of CSCs to topotecan and mitoxantrone [154]. The current approach is to inhibit a single ABC transporter; however there are three efflux proteins that are important to chemoresistance in CSCs (ABCB1, ABCC1, and ABCG2). Several compound have been found to inhibit the three key ABC transporters: cyclosporine A, biricodar, PK11195, and curcumin [155]. While not all of these drugs have undergone clinical trials, inhibiting the key ABC transporters in CSCs is an avenue for future research.

Conclusions

The bulk of a tumor is composed of non-CSCs; however, the presence of CSCs represents an important hurdle to effective cancer therapy. These tumor-initiating cells are more resistant to conventional chemotherapeutics than the majority of cancer cells, and survival of CSCs likely contributes to tumor recurrence. Mechanisms by which CSCs are resistant to chemotherapy include enhanced DNA repair, increased detoxification capacity, and quiescence. Though CSCs are a great challenge to chemotherapy, they may be surmounted by the use of ABC transporter inhibitors, ALDH inhibitors, and through targeting CSC-specific Wnt, Hh, and Notch pathways. Theoretically, eliminating CSCs through targeted therapies would increase the efficacy of our existing treatments and lead to more favourable long-term prognoses for many cancer types.

Financial Support

MLT is supported by a studentship from the Canadian Breast Cancer Foundation Atlantic-Chapter, administered by the Beatrice Hunter Cancer Research Institute. KMC is supported by a trainee award from the Beatrice Hunter Cancer Research Institute with funds provided by the Canadian Imperial Bank of Commerce as part of the Terry Fox Strategic Health Research Training Program in Cancer Research at the Canadian Institutes of Health Research (CIHR). Publication of this manuscript is supported by a grant to PM from the CIHR (MOP-130304).

References

  1. Dalerba P, Cho RW, Clarke MF (2007) Cancer stem cells: models and concepts. Annu Rev Med 58: 267-284.
  2. Bonnet D, Dick JE (1997) Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nat Med 3: 730-737.
  3. Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke MF (2003) Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci U S A 100: 3983-3988.
  4. Singh SK, Hawkins C, Clarke ID, Squire JA, Bayani J, et al. (2004) Identification of human brain tumour initiating cells. Nature 432: 396-401.
  5. O'Brien CA, Pollett A, Gallinger S, Dick JE (2007) A human colon cancer cell capable of initiating tumour growth in immunodeficient mice. Nature 445: 106-110.
  6. Hermann PC, Huber SL, Herrler T, Aicher A, Ellwart JW, et al. (2007) Distinct populations of cancer stem cells determine tumor growth and metastatic activity in human pancreatic cancer. Cell Stem Cell 1: 313-323.
  7. Ma S, Chan KW, Lee TK, Tang KH, Wo JY, et al. (2008) Aldehyde dehydrogenase discriminates the CD133 liver cancer stem cell populations. Mol Cancer Res 6: 1146-1153.
  8. Eramo A, Lotti F, Sette G, Pilozzi E, Biffoni M, et al. (2008) Identification and expansion of the tumorigenic lung cancer stem cell population. Cell Death Differ 15: 504-514.
  9. Zhang S, Balch C, Chan MW, Lai HC, Matei D, et al. (2008) Identification and characterization of ovarian cancer-initiating cells from primary human tumors. Cancer Res 68: 4311-4320.
  10. Takaishi S, Okumura T, Tu S, Wang SS, Shibata W, et al. (2009) Identification of gastric cancer stem cells using the cell surface marker CD44. Stem Cells 27: 1006-1020.
  11. Collins AT1, Berry PA, Hyde C, Stower MJ, Maitland NJ (2005) Prospective identification of tumorigenic prostate cancer stem cells. Cancer Res 65: 10946-10951.
  12. Prince ME Sivanandan R, Kaczorowski A, Wolf GT, Kaplan MJ, et al. (2007) Identification of a subpopulation of cells with cancer stem cell properties in head and neck squamous cell carcinoma. Proc Natl Acad Sci U S A 104: 973-978.
  13. Marcato P, Dean CA, Pan D, Araslanova R, Gillis M, et al. (2011) Aldehyde dehydrogenase activity of breast cancer stem cells is primarily due to isoform ALDH1A3 and its expression is predictive of metastasis. Stem Cells 29: 32-45.
  14. Clevers H (2011) The cancer stem cell: premises, promises and challenges. Nat Med 17: 313-319.
  15. Visvader JE, Lindeman GJ (2008) Cancer stem cells in solid tumours: accumulating evidence and unresolved questions. Nat Rev Cancer 8: 755-768.
  16. Reya T, Morrison SJ, Clarke MF, Weissman IL (2001) Stem cells, cancer, and cancer stem cells. Nature 414: 105-111.
  17. Blagosklonny MV (2005) Why therapeutic response may not prolong the life of a cancer patient: selection for oncogenic resistance. Cell Cycle 4: 1693-1698.
  18. Hess DA, Meyerrose TE, Wirthlin L (2004) Functional characterization of highly purified human hematopoietic repopulating cells isolated according to aldehyde dehydrogenase activity. Blood 104:1648-1655.
  19. Ginestier C, Hur MH, Charafe-Jauffret E, Monville F, Dutcher J, et al. (2007) ALDH1 is a marker of normal and malignant human mammary stem cells and a predictor of poor clinical outcome. Cell Stem Cell 1: 555-567.
  20. Cheung A, Wan T, Leung J (2007) Aldehyde dehydrogenase activity in leukemic blasts defines a subgroup of acute myeloid leukemia with adverse prognosis and superior NOD/SCID engrafting potential. Leukemia. 21:1423-1430.
  21. Chen YC, Chen YW, Hsu HS, Tseng LM, Huang PI, et al. (2009) Aldehyde dehydrogenase 1 is a putative marker for cancer stem cells in head and neck squamous cancer. Biochem Biophys Res Commun 385: 307-313.
  22. Jiang F, Qiu Q, Khanna A, Todd NW, Deepak J, et al. (2009) Aldehyde dehydrogenase 1 is a tumor stem cell-associated marker in lung cancer. Mol Cancer Res 7: 330-338.
  23. Kim MP, Fleming JB, Wang H, Abbruzzese JL, Choi W, et al. (2011) ALDH activity selectively defines an enhanced tumor-initiating cell population relative to CD133 expression in human pancreatic adenocarcinoma. PLoS One 6: e20636.
  24. Rao QX, Yao TT, Zhang BZ, Lin RC, Chen ZL, et al. (2012) Expression and functional role of ALDH1 in cervical carcinoma cells. Asian Pac J Cancer Prev 13: 1325-1331.
  25. Todaro M, Iovino F, Eterno V, Cammareri P, Gambara G, et al. (2010) Tumorigenic and metastatic activity of human thyroid cancer stem cells. Cancer Res 70: 8874-8885.
  26. Van den Hoogen C, van der Horst G, Cheung H, Buijs JT, Lippitt JM, et al. (2010) High aldehyde dehydrogenase activity identifies tumor-initiating and metastasis-initiating cells in human prostate cancer. Cancer Res 70: 5163-5173.
  27. Huang EH, Hynes MJ, Zhang T, Ginestier C, Dontu G, et al. (2009) Aldehyde dehydrogenase 1 is a marker for normal and malignant human colonic stem cells (SC) and tracks SC overpopulation during colon tumorigenesis. Cancer Res 69: 3382-3389.
  28. Landen CN Jr, Goodman B, Katre AA, Steg AD, Nick AM, et al. (2010) Targeting aldehyde dehydrogenase cancer stem cells in ovarian cancer. Mol Cancer Ther 9: 3186-3199.
  29. Su Y, Qiu Q, Zhang X, Jiang Z, Leng Q, et al. (2010) Aldehyde dehydrogenase 1 A1-positive cell population is enriched in tumor-initiating cells and associated with progression of bladder cancer. Cancer Epidemiol Biomarkers Prev 19: 327-337.
  30. Vasiliou V, Nebert DW (2005) Analysis and update of the human aldehyde dehydrogenase (ALDH) gene family. Hum Genomics 2: 138-143.
  31. Tanei T, Morimoto K, Shimazu K (2009) Association of breast cancer stem cells identified by aldehyde dehydrogenase 1 expression with resistance to sequential paclitaxel and epirubicin-based chemotherapy for breast cancers. Clinical Cancer Research. 15:4234-4241.
  32. Fu D, Calvo JA, Samson LD (2012) Balancing repair and tolerance of DNA damage caused by alkylating agents. Nat Rev Cancer 12: 104-120.
  33. Bonadonna G, Valagussa P, Moliterni A, Zambetti M, Brambilla C (1995) Adjuvant cyclophosphamide, methotrexate, and fluorouracil in node-positive breast cancer: the results of 20 years of follow-up. N Engl J Med 332: 901-906.
  34. von Pawel J, Schiller JH, Shepherd FA, Fields SZ, Kleisbauer JP, et al. (1999) Topotecan versus cyclophosphamide, doxorubicin, and vincristine for the treatment of recurrent small-cell lung cancer. J Clin Oncol 17: 658-667.
  35. Di Re F, Bohm S, Oriana S, Spatti GB, Zunino F (1990) Efficacy and safety of high-dose cisplatin and cyclophosphamide with glutathione protection in the treatment of bulky advanced epithelial ovarian cancer. Cancer Chemother Pharmacol 25: 355-360.
  36. Socié G, Clift RA, Blaise D, Devergie A, Ringden O, et al. (2001) Busulfan plus cyclophosphamide compared with total-body irradiation plus cyclophosphamide before marrow transplantation for myeloid leukemia: long-term follow-up of 4 randomized studies. Blood 98: 3569-3574.
  37. THURMAN WG, FERNBACH DJ, SULLIVAN MP (1964) CYCLOPHOSPHAMIDE THERAPY IN CHILDHOOD NEUROBLASTOMA. N Engl J Med 270: 1336-1340.
  38. Luce JK, Gamble JF, Wilson HE, Monto RW, Isaacs BL, et al. (1971) Combined cyclophosphamide vincristine, and prednisone therapy of malignant lymphoma. Cancer 28: 306-317.
  39. Emadi A, Jones RJ, Brodsky RA (2009) Cyclophosphamide and cancer: golden anniversary. Nat Rev Clin Oncol 6: 638-647.
  40. de Jonge ME, Huitema AD, Rodenhuis S, Beijnen JH (2005) Clinical pharmacokinetics of cyclophosphamide. Clin Pharmacokinet 44: 1135-1164.
  41. Hipkens JH, Struck RF, Gurtoo HL (1981) Role of aldehyde dehydrogenase in the metabolism-dependent biological activity of cyclophosphamide. Cancer Res 41: 3571-3583.
  42. Hilton J (1984) Role of aldehyde dehydrogenase in cyclophosphamide-resistant L1210 leukemia. Cancer Res 44: 5156-5160.
  43. Russo JE, Hilton J (1988) Characterization of cytosolic aldehyde dehydrogenase from cyclophosphamide resistant L1210 cells. Cancer Res 48: 2963-2968.
  44. Jones RJ, Barber JP, Vala MS, Collector MI, Kaufmann SH, et al. (1995) Assessment of aldehyde dehydrogenase in viable cells. Blood 85: 2742-2746.
  45. Magni M, Shammah S, Schiró R, Mellado W, Dalla-Favera R, et al. (1996) Induction of cyclophosphamide-resistance by aldehyde-dehydrogenase gene transfer. Blood 87: 1097-1103.
  46. Moreb JS, Muhoczy D, Ostmark B, Zucali JR (2007) RNAi-mediated knockdown of aldehyde dehydrogenase class-1A1 and class-3A1 is specific and reveals that each contributes equally to the resistance against 4-hydroperoxycyclophosphamide. Cancer Chemother Pharmacol. 59:127-136.
  47. Sládek NE, Kollander R, Sreerama L, Kiang DT (2002) Cellular levels of aldehyde dehydrogenases (ALDH1A1 and ALDH3A1) as predictors of therapeutic responses to cyclophosphamide-based chemotherapy of breast cancer: A retrospective study. Cancer Chemother Pharmacol. 49:309-321.
  48. Storms RW, Trujillo AP, Springer JB, Shah L, Colvin OM, et al. (1999) Isolation of primitive human hematopoietic progenitors on the basis of aldehyde dehydrogenase activity. Proc Natl Acad Sci U S A 96: 9118-9123.
  49. Luo Y, Dallaglio K, Chen Y, Robinson WA, Robinson SE, et al. (2012) ALDH1A isozymes are markers of human melanoma stem cells and potential therapeutic targets. Stem Cells 30: 2100-2113.
  50. Neumeister V, Agarwal S, Bordeaux J, Camp RL, Rimm DL (2010) In situ identification of putative cancer stem cells by multiplexing ALDH1, CD44, and cytokeratin identifies breast cancer patients with poor prognosis. Am J Pathol 176: 2131-2138.
  51. Resetkova E, Reis-Filho JS, Jain RK, Mehta R, Thorat MA, et al. (2010) Prognostic impact of ALDH1 in breast cancer: a story of stem cells and tumor microenvironment. Breast Cancer Res Treat 123: 97-108.
  52. Jackson SP, Bartek J (2009) The DNA-damage response in human biology and disease. Nature 461: 1071-1078.
  53. Bao S, Wu Q, McLendon RE, Hao Y, Shi Q, et al. (2006) Glioma stem cells promote radioresistance by preferential activation of the DNA damage response. Nature 444: 756-760.
  54. Eyler CE, Foo WC, LaFiura KM, McLendon RE, Hjelmeland AB, et al. (2008) Brain cancer stem cells display preferential sensitivity to Akt inhibition. Stem Cells 26: 3027-3036.
  55. McCord AM, Jamal M, Williams ES, Camphausen K, Tofilon PJ (2009) CD133+ glioblastoma stem-like cells are radiosensitive with a defective DNA damage response compared with established cell lines. Clin Cancer Res 15: 5145-5153.
  56. Ropolo M, Daga A, Griffero F, Foresta M, Casartelli G, et al. (2009) Comparative analysis of DNA repair in stem and nonstem glioma cell cultures. Mol Cancer Res 7: 383-392.
  57. Bartucci M, Svensson S, Romania P, Dattilo R, Patrizii M, et al. (2012) Therapeutic targeting of Chk1 in NSCLC stem cells during chemotherapy. Cell Death Differ 19: 768-778.
  58. Gallmeier E, Hermann PC, Mueller MT, Machado JG, Ziesch A, et al. (2011) Inhibition of ataxia telangiectasia- and Rad3-related function abrogates the in vitro and in vivo tumorigenicity of human colon cancer cells through depletion of the CD133(+) tumor-initiating cell fraction. Stem Cells 29: 418-429.
  59. Sciuscio D, Diserens AC, van Dommelen K, Martinet D, Jones G, et al. (2011) Extent and patterns of MGMT promoter methylation in glioblastoma- and respective glioblastoma-derived spheres. Clin Cancer Res 17: 255-266.
  60. Mathews LA, Cabarcas SM, Farrar WL (2011) DNA repair: the culprit for tumor-initiating cell survival? Cancer Metastasis Rev 30: 185-197.
  61. Mathews LA, Cabarcas SM, Hurt EM, Zhang X, Jaffee EM, et al. (2011) Increased expression of DNA repair genes in invasive human pancreatic cancer cells. Pancreas 40: 730-739.
  62. Levina V, Marrangoni AM, DeMarco R, Gorelik E, Lokshin AE (2008) Drug-selected human lung cancer stem cells: cytokine network, tumorigenic and metastatic properties. PLoS One 3: e3077.
  63. Shafee N, Smith CR, Wei S, Kim Y, Mills GB, et al. (2008) Cancer stem cells contribute to cisplatin resistance in Brca1/p53-mediated mouse mammary tumors. Cancer Res 68: 3243-3250.
  64. Rizzo S, Hersey JM, Mellor P, Dai W, Santos-Silva A, et al. (2011) Ovarian cancer stem cell-like side populations are enriched following chemotherapy and overexpress EZH2. Mol Cancer Ther 10: 325-335.
  65. Liu G, Yuan X, Zeng Z, Tunici P, Ng H, et al. (2006) Analysis of gene expression and chemoresistance of CD133+ cancer stem cells in glioblastoma. Mol Cancer 5: 67.
  66. Gascoigne KE, Taylor SS (2009) How do anti-mitotic drugs kill cancer cells? J Cell Sci 122: 2579-2585.
  67. Marzo I, Naval J (2013) Antimitotic drugs in cancer chemotherapy: promises and pitfalls. Biochem Pharmacol 86: 703-710.
  68. Viale A, De Franco F, Orleth A, Cambiaghi V, Giuliani V, et al. (2009) Cell-cycle restriction limits DNA damage and maintains self-renewal of leukaemia stem cells. Nature 457: 51-56.
  69. Liu Q, Nguyen DH, Dong Q, Shitaku P, Chung K, et al. (2009) Molecular properties of CD133+ glioblastoma stem cells derived from treatment-refractory recurrent brain tumors. J Neurooncol 94: 1-19.
  70. Pece S, Tosoni D, Confalonieri S, Mazzarol G, Vecchi M, et al. (2010) Biological and molecular heterogeneity of breast cancers correlates with their cancer stem cell content. Cell 140: 62-73.
  71. Saito Y, Uchida N, Tanaka S, Suzuki N, Tomizawa-Murasawa M, et al. (2010) Induction of cell cycle entry eliminates human leukemia stem cells in a mouse model of AML. Nat Biotechnol 28: 275-280.
  72. Britton KM, Eyre R, Harvey IJ, Stemke-Hale K, Browell D, et al. (2012) Breast cancer, side population cells and ABCG2 expression. Cancer Lett 323: 97-105.
  73. Singh A, Wu H, Zhang P, Happel C, Ma J, et al. (2010) Expression of ABCG2 (BCRP) is regulated by Nrf2 in cancer cells that confers side population and chemoresistance phenotype. Mol Cancer Ther 9: 2365-2376.
  74. Hu L, McArthur C, Jaffe RB (2010) Ovarian cancer stem-like side-population cells are tumourigenic and chemoresistant. Br J Cancer 102: 1276-1283.
  75. Ho MM, Ng AV, Lam S, Hung JY (2007) Side population in human lung cancer cell lines and tumors is enriched with stem-like cancer cells. Cancer Res 67: 4827-4833.
  76. Chiba T, Kita K, Zheng YW, Yokosuka O, Saisho H, et al. (2006) Side population purified from hepatocellular carcinoma cells harbors cancer stem cell-like properties. Hepatology 44: 240-251.
  77. Hadnagy A, Gaboury L, Beaulieu R, Balicki D (2006) SP analysis may be used to identify cancer stem cell populations. Exp Cell Res 312: 3701-3710.
  78. Haraguchi N, Utsunomiya T, Inoue H, Tanaka F, Mimori K, et al. (2006) Characterization of a side population of cancer cells from human gastrointestinal system. Stem Cells 24: 506-513.
  79. Loebinger MR, Giangreco A, Groot KR, Prichard L, Allen K, et al. (2008) Squamous cell cancers contain a side population of stem-like cells that are made chemosensitive by ABC transporter blockade. Br J Cancer 98: 380-387.
  80. Zhu Z, Hao X, Yan M, Yao M, Ge C, et al. (2010) Cancer stem/progenitor cells are highly enriched in CD133+CD44+ population in hepatocellular carcinoma. Int J Cancer 126: 2067-2078.
  81. Okamoto A, Chikamatsu K, Sakakura K, Hatsushika K, Takahashi G, et al. (2009) Expansion and characterization of cancer stem-like cells in squamous cell carcinoma of the head and neck. Oral Oncol 45: 633-639.
  82. Dean M (2009) ABC transporters, drug resistance, and cancer stem cells. J Mammary Gland Biol Neoplasia 14: 3-9.
  83. Wright MH, Calcagno AM, Salcido CD, Carlson MD, Ambudkar SV, et al. (2008) Brca1 breast tumors contain distinct CD44+/CD24- and CD133+ cells with cancer stem cell characteristics. Breast Cancer Res 10: R10.
  84. Wagner U, Brownlees J, Irving NG, Lucas FR, Salinas PC, et al. (1997) Overexpression of the mouse dishevelled-1 protein inhibits GSK-3beta-mediated phosphorylation of tau in transfected mammalian cells. FEBS Lett 411: 369-372.
  85. Heidel FH, Mar BG, Armstrong SA (2011) Self-renewal related signaling in myeloid leukemia stem cells. Int J Hematol 94: 109-117.
  86. Malanchi I, Santamaria-Martínez A, Susanto E, Peng H, Lehr HA, et al. (2011) Interactions between cancer stem cells and their niche govern metastatic colonization. Nature 481: 85-89.
  87. Teng Y, Wang X, Wang Y, Ma D (2010) Wnt/beta-catenin signaling regulates cancer stem cells in lung cancer A549 cells. Biochem Biophys Res Commun 392: 373-379.
  88. Vermeulen L, De Sousa E Melo F, van der Heijden M, Cameron K, de Jong JH, et al. (2010) Wnt activity defines colon cancer stem cells and is regulated by the microenvironment. Nat Cell Biol 12: 468-476.
  89. Cai C, Zhu X (2012) The Wnt/β-catenin pathway regulates self-renewal of cancer stem-like cells in human gastric cancer. Mol Med Rep 5: 1191-1196.
  90. Pesce M, Schöler HR (2001) Oct-4: gatekeeper in the beginnings of mammalian development. Stem Cells 19: 271-278.
  91. Ding DF, Li XF, Xu H, Wang Z, Liang QQ, et al. (2013) Mechanism of resveratrol on the promotion of induced pluripotent stem cells. J Integr Med 11: 389-396.
  92. Pandey PR, Okuda H, Watabe M, Pai SK, Liu W, et al. (2011) Resveratrol suppresses growth of cancer stem-like cells by inhibiting fatty acid synthase. Breast Cancer Res Treat 130: 387-398.
  93. Fiorentino M, Zadra G, Palescandolo E, Fedele G, Bailey D, et al. (2008) Overexpression of fatty acid synthase is associated with palmitoylation of Wnt1 and cytoplasmic stabilization of beta-catenin in prostate cancer. Lab Invest 88: 1340-1348.
  94. Shankar S, Nall D, Tang S (2011) Resveratrol inhibits pancreatic cancer stem cell characteristics in human and KrasG12D transgenic mice by inhibiting pluripotency maintaining factors and epithelial-mesenchymal transition. PLoS One 6:e16530.
  95. Nguyen AV, Martinez M, Stamos MJ (2009)Results of a phase I pilot clinical trial examining the effect of plant-derived resveratrol and grape powder on wnt pathway target gene expression in colonic mucosa and colon cancer. Cancer management and research 1:25.
  96. Akiyama T, Ishida J, Nakagawa S, Ogawara H, Watanabe S, et al. (1987) Genistein, a specific inhibitor of tyrosine-specific protein kinases. J Biol Chem 262: 5592-5595.
  97. Montales MTE, Rahal OM, Kang J (2012) Repression of mammosphere formation of human breast cancer cells by soy isoflavone genistein and blueberry polyphenolic acids suggests diet-mediated targeting of cancer stem-like/progenitor cells. Carcinogenesis 33:652-660.
  98. Zhang L, Li L, Jiao M, Wu D, Wu K, et al. (2012) Genistein inhibits the stemness properties of prostate cancer cells through targeting Hedgehog-Gli1 pathway. Cancer Lett 323: 48-57.
  99. Fan P, Fan S, Wang H, Mao J, Shi Y, et al. (2013) Genistein decreases the breast cancer stem-like cell population through Hedgehog pathway. Stem Cell Res Ther 4: 146.
  100. Yu D, Shin HS, Lee YS, Lee D, Kim S1, et al. (2014) Genistein attenuates cancer stem cell characteristics in gastric cancer through the downregulation of Gli1. Oncol Rep 31: 673-678.
  101. Zhang Y, Chen H (2011) Genistein attenuates WNT signaling by up-regulating sFRP2 in a human colon cancer cell line. Exp Biol Med (Maywood) 236: 714-722.
  102. Hirata H, Ueno K, Nakajima K, Tabatabai ZL, Hinoda Y, et al. (2013) Genistein downregulates onco-miR-1260b and inhibits Wnt-signalling in renal cancer cells. Br J Cancer 108: 2070-2078.
  103. Lazarevic B, Boezelijn G, Diep LM, (2011) Efficacy and safety of short-term genistein intervention in patients with localized prostate cancer prior to radical prostatectomy: A randomized, placebo-controlled, double-blind phase 2 clinical trial. Nutr Cancer 63:889-898.
  104. Djiane A, Krejci A, Bernard F, Fexova S, Millen K, et al. (2013) Dissecting the mechanisms of Notch induced hyperplasia. EMBO J 32: 60-71.
  105. Le Borgne R, Bardin A, Schweisguth F (2005) The roles of receptor and ligand endocytosis in regulating Notch signaling. Development 132: 1751-1762.
  106. De Strooper B, Annaert W, Cupers P, Saftig P, Craessaerts K, et al. (1999) A presenilin-1-dependent gamma-secretase-like protease mediates release of Notch intracellular domain. Nature 398: 518-522.
  107. Schroeter EH, Kisslinger JA, Kopan R (1998) Notch-1 signalling requires ligand-induced proteolytic release of intracellular domain. Nature 393: 382-386.
  108. Fan X, Khaki L, Zhu TS, Soules ME, Talsma CE, et al. (2010) NOTCH pathway blockade depletes CD133-positive glioblastoma cells and inhibits growth of tumor neurospheres and xenografts. Stem Cells 28: 5-16.
  109. Weng AP, Ferrando AA, Lee W, Morris JP 4th, Silverman LB, et al. (2004) Activating mutations of NOTCH1 in human T cell acute lymphoblastic leukemia. Science 306: 269-271.
  110. Gerby B, Clappier E, Armstrong F, Deswarte C, Calvo J, et al. (2011) Expression of CD34 and CD7 on human T-cell acute lymphoblastic leukemia discriminates functionally heterogeneous cell populations. Leukemia 25: 1249-1258.
  111. Harrison H, Farnie G, Howell SJ, Rock RE, Stylianou S, et al. (2010) Regulation of breast cancer stem cell activity by signaling through the Notch4 receptor. Cancer Res 70: 709-718.
  112. Tolcher AW, Messersmith WA, Mikulski SM, Papadopoulos KP, Kwak EL, et al. (2012) Phase I study of RO4929097, a gamma secretase inhibitor of Notch signaling, in patients with refractory metastatic or locally advanced solid tumors. J Clin Oncol 30: 2348-2353.
  113. Schott AF, Landis MD, Dontu G, Griffith KA, Layman RM, et al. (2013) Preclinical and clinical studies of gamma secretase inhibitors with docetaxel on human breast tumors. Clin Cancer Res 19: 1512-1524.
  114. van Es JH, van Gijn ME, Riccio O, van den Born M, Vooijs M, et al. (2005) Notch/gamma-secretase inhibition turns proliferative cells in intestinal crypts and adenomas into goblet cells. Nature 435: 959-963.
  115. Alison MR, Lim SM, Nicholson LJ (2011) Cancer stem cells: problems for therapy? J Pathol 223: 147-161.
  116. Mullendore ME, Koorstra JB, Li YM, Offerhaus GJ, Fan X, et al. (2009) Ligand-dependent Notch signaling is involved in tumor initiation and tumor maintenance in pancreatic cancer. Clin Cancer Res 15: 2291-2301.
  117. Fan X, Matsui W, Khaki L, Stearns D, Chun J, et al. (2006) Notch pathway inhibition depletes stem-like cells and blocks engraftment in embryonal brain tumors. Cancer Res 66: 7445-7452.
  118. Wong NK, Fuller M, Sung S, Wong F, Karsan A (2012) Heterogeneity of breast cancer stem cells as evidenced with Notch-dependent and Notch-independent populations. Cancer Med 1: 105-113.
  119. Katoh Y, Katoh M (2009) Hedgehog target genes: mechanisms of carcinogenesis induced by aberrant hedgehog signaling activation. Curr Mol Med 9: 873-886.
  120. Liu S, Dontu G, Mantle ID, Patel S, Ahn NS, et al. (2006) Hedgehog signaling and Bmi-1 regulate self-renewal of normal and malignant human mammary stem cells. Cancer Res 66: 6063-6071.
  121. Clement V, Sanchez P, de Tribolet N, Radovanovic I, Ruiz i Altaba A (2007) HEDGEHOG-GLI1 signaling regulates human glioma growth, cancer stem cell self-renewal, and tumorigenicity. Curr Biol 17: 165-172.
  122. Peacock CD, Wang Q, Gesell GS, Corcoran-Schwartz IM, Jones E, et al. (2007) Hedgehog signaling maintains a tumor stem cell compartment in multiple myeloma. Proc Natl Acad Sci U S A 104: 4048-4053.
  123. Feldmann G, Dhara S, Fendrich V, Bedja D, Beaty R, et al. (2007) Blockade of hedgehog signaling inhibits pancreatic cancer invasion and metastases: a new paradigm for combination therapy in solid cancers. Cancer Res 67: 2187-2196.
  124. McCann CK, Growdon WB, Kulkarni-Datar K, Curley MD, Friel AM, et al. (2011) Inhibition of Hedgehog signaling antagonizes serous ovarian cancer growth in a primary xenograft model. PLoS One 6: e28077.
  125. Yauch RL, Gould SE, Scales SJ, Tang T, Tian H, et al. (2008) A paracrine requirement for hedgehog signalling in cancer. Nature 455: 406-410.
  126. Stanton BZ, Peng LF, Maloof N, Nakai K, Wang X, et al. (2009) A small molecule that binds Hedgehog and blocks its signaling in human cells. Nat Chem Biol 5: 154-156.
  127. Chen JK, Taipale J, Cooper MK, Beachy PA (2002) Inhibition of Hedgehog signaling by direct binding of cyclopamine to Smoothened. Genes Dev 16: 2743-2748.
  128. Berman DM, Karhadkar SS, Hallahan AR, Pritchard JI, Eberhart CG, et al. (2002) Medulloblastoma growth inhibition by hedgehog pathway blockade. Science 297: 1559-1561.
  129. Jimeno A, Weiss GJ, Miller WH Jr, Gettinger S, Eigl BJ, et al. (2013) Phase I study of the Hedgehog pathway inhibitor IPI-926 in adult patients with solid tumors. Clin Cancer Res 19: 2766-2774.
  130. Olive KP, Jacobetz MA, Davidson CJ, Gopinathan A, McIntyre D, et al. (2009) Inhibition of Hedgehog signaling enhances delivery of chemotherapy in a mouse model of pancreatic cancer. Science 324: 1457-1461.
  131. Tian F, Mysliwietz J, Ellwart J, Gamarra F, Huber RM, et al. (2012) Effects of the Hedgehog pathway inhibitor GDC-0449 on lung cancer cell lines are mediated by side populations. Clin Exp Med 12: 25-30.
  132. Von Hoff DD, LoRusso PM, Rudin CM, Reddy JC, Yauch RL, et al. (2009) Inhibition of the hedgehog pathway in advanced basal-cell carcinoma. N Engl J Med 361: 1164-1172.
  133. Rudin CM, Hann CL, Laterra J, Yauch RL, Callahan CA, et al. (2009) Treatment of medulloblastoma with hedgehog pathway inhibitor GDC-0449. N Engl J Med 361: 1173-1178.
  134. Lauth M, Bergström A, Shimokawa T, Toftgård R (2007) Inhibition of GLI-mediated transcription and tumor cell growth by small-molecule antagonists. Proc Natl Acad Sci U S A 104: 8455-8460.
  135. Hou X, Chen X, Zhang P, Fan Y, Ma A, et al. (2014) Inhibition of hedgehog signaling by GANT58 induces apoptosis and shows synergistic antitumor activity with AKT inhibitor in acute T cell leukemia cells. Biochimie .
  136. Thomas Z, Gibson W, Sexton JZ, Aird KM, Ingram SM, et al. (2011) Targeting GLI1 expression in human inflammatory breast cancer cells enhances apoptosis and attenuates migration. Br J Cancer 104: 1575-1586.
  137. Mozzetti S, Martinelli E, Raspaglio G, Prislei S, De Donato M, et al. (2012) Gli family transcription factors are drivers of patupilone resistance in ovarian cancer. Biochem Pharmacol 84: 1409-1418.
  138. Koppaka V, Thompson DC, Chen Y, Ellermann M, Nicolaou KC, et al. (2012) Aldehyde dehydrogenase inhibitors: a comprehensive review of the pharmacology, mechanism of action, substrate specificity, and clinical application. Pharmacol Rev 64: 520-539.
  139. Cvek B (2011) Targeting malignancies with disulfiram (Antabuse): multidrug resistance, angiogenesis, and proteasome. Curr Cancer Drug Targets 11: 332-337.
  140. R Kona, Buac D, M Burger (2011) Disulfiram, and disulfiram derivatives as novel potential anticancer drugs targeting the ubiquitin proteasome system in both preclinical and clinical studies. Current cancer drug targets 11:338-346.
  141. Chen D, Cui QC, Yang H, Dou QP (2006) Disulfiram, a clinically used anti-alcoholism drug and copper-binding agent, induces apoptotic cell death in breast cancer cultures and xenografts via inhibition of the proteasome activity. Cancer Res 66: 10425-10433.
  142. Lövborg H, Oberg F, Rickardson L, Gullbo J, Nygren P, et al. (2006) Inhibition of proteasome activity, nuclear factor-KappaB translocation and cell survival by the antialcoholism drug disulfiram. Int J Cancer 118: 1577-1580.
  143. Lin J, Haffner MC, Zhang Y, Lee BH, Brennen WN, et al. (2011) Disulfiram is a DNA demethylating agent and inhibits prostate cancer cell growth. Prostate 71: 333-343.
  144. Moreb JS, Ucar D, Han S, Amory JK, Goldstein AS, et al. (2012) The enzymatic activity of human aldehyde dehydrogenases 1A2 and 2 (ALDH1A2 and ALDH2) is detected by Aldefluor, inhibited by diethylaminobenzaldehyde and has significant effects on cell proliferation and drug resistance. Chem Biol Interact 195: 52-60.
  145. Liu P, Brown S, Goktug T (2012) Cytotoxic effect of disulfiram/copper on human glioblastoma cell lines and ALDH-positive cancer-stem-like cells. Br J Cancer.
  146. Yip NC, Fombon IS, Liu P, Brown S, Kannappan V, et al. (2011) Disulfiram modulated ROS-MAPK and NFκB pathways and targeted breast cancer cells with cancer stem cell-like properties. Br J Cancer 104: 1564-1574.
  147. Lipsky JJ, Shen ML, Naylor S (2001) Overview--in vitro inhibition of aldehyde dehydrogenase by disulfiram and metabolites. Chem Biol Interact 130-132: 81-91.
  148. Guo X, Xu B, Pandey S, Goessl E, Brown J, et al. (2010) Disulfiram/copper complex inhibiting NFkappaB activity and potentiating cytotoxic effect of gemcitabine on colon and breast cancer cell lines. Cancer Lett 290: 104-113.
  149. Triscott J, Lee C, Hu K, Fotovati A, Berns R, et al. (2012) Disulfiram, a drug widely used to control alcoholism, suppresses the self-renewal of glioblastoma and over-rides resistance to temozolomide. Oncotarget 3: 1112-1123.
  150. Shukla S, Ohnuma S, Ambudkar SV (2011) Improving cancer chemotherapy with modulators of ABC drug transporters. Curr Drug Targets 12: 621-630.
  151. Sonneveld P, Suciu S, Weijermans P, Beksac M, Neuwirtova R, et al. (2001) Cyclosporin A combined with vincristine, doxorubicin and dexamethasone (VAD) compared with VAD alone in patients with advanced refractory multiple myeloma: an EORTC-HOVON randomized phase III study (06914). Br J Haematol 115: 895-902.
  152. Millward MJ, Cantwell BM, Munro NC, Robinson A, Corris PA, et al. (1993) Oral verapamil with chemotherapy for advanced non-small cell lung cancer: a randomised study. Br J Cancer 67: 1031-1035.
  153. Belpomme D, Gauthier S, Pujade-Lauraine E, Facchini T, Goudier MJ, et al. (2000) Verapamil increases the survival of patients with anthracycline-resistant metastatic breast carcinoma. Ann Oncol 11: 1471-1476.
  154. Wang F, Mi YJ, Chen XG, Wu XP, Liu Z, et al. (2012) Axitinib targeted cancer stemlike cells to enhance efficacy of chemotherapeutic drugs via inhibiting the drug transport function of ABCG2. Mol Med 18: 887-898.
  155. Wu CP, Calcagno AM, Ambudkar SV (2008) Reversal of ABC drug transporter-mediated multidrug resistance in cancer cells: evaluation of current strategies. Curr Mol Pharmacol 1: 93-105.
Citation: Thomas ML, Coyle KM, Sultan M, Ahmad VK, Marcato P (2014) Chemoresistance in Cancer Stem Cells and Strategies to Overcome Resistance. Chemotherapy 3:125.

Copyright: © 2014 Thomas ML, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Top