Skip to main content

MINI REVIEW article

Front. Pharmacol., 14 March 2013
Sec. Pharmacology of Anti-Cancer Drugs
This article is part of the Research Topic Therapeutic Resistance in Cancer: Mechanisms, Insights and Progress View all 16 articles

Mechanisms and insights into drug resistance in cancer

  • Department of Pathology and Cell Biology, Institute of Research in Immunology and Cancer, Université de Montréal, Montreal, QC, Canada

Cancer drug resistance continues to be a major impediment in medical oncology. Clinically, resistance can arise prior to or as a result of cancer therapy. In this review, we discuss different mechanisms adapted by cancerous cells to resist treatment, including alteration in drug transport and metabolism, mutation and amplification of drug targets, as well as genetic rewiring which can lead to impaired apoptosis. Tumor heterogeneity may also contribute to resistance, where small subpopulations of cells may acquire or stochastically already possess some of the features enabling them to emerge under selective drug pressure. Making the problem even more challenging, some of these resistance pathways lead to multidrug resistance, generating an even more difficult clinical problem to overcome. We provide examples of these mechanisms and some insights into how understanding these processes can influence the next generation of cancer therapies.

Cancer Tale: its Treatment and Relapse

In 1961, Frei and Freireich initiated the high-dose four-drug combination clinical trial for the treatment of pediatric leukemia (Frei et al., 1965). Despite the threat imposed by administering four drugs at once, few weeks following treatment onset, children began to respond, “the bone marrow biopsies came back one after another—all without leukemia cells. Red blood cells and white blood cells and platelets sprouted up in an otherwise scorched field of bone marrow. But the leukemia did not return” (Mukherjee, 2010). Out of the 16 enrolled patients, 11 showed complete remission. This outstanding success, however, was short-lived. With the exception of a handful of children, all patients eventually relapsed, developing a more vigorous form of cancer that was no longer responsive to the treatment: leukemic cells had invaded the blood–brain barrier and colonized the brain “the only place unreachable by chemotherapy....the children died one after the other-felled by virtue of the adaptation designed to protect them… it was a consequence of the body’s defense system subverting cancer treatment” (Mukherjee, 2010). To date, this story still reflects the same tale of cancer treatment where its resistance and relapse remains a major challenge (Wilson et al., 2009). In this review we provide an overview of advances made in our understanding of the mechanisms that enable cancerous cells to adapt to and eventually overcome therapy, and how identifying these mechanisms can help circumvent resistance and improve treatment.

Despite its complex biological nature, many recent successes have been made in the treatment of cancer, including most strikingly chronic myeloid leukemia (CML) and acute promyelocytic leukemia (APL) which have met with great success as well as many cases of pediatric leukemias, Hodgkin’s lymphomas, and testicular cancers (Siegel et al., 2012). These success stories mainly relied on an increased understanding of the diverse molecular mechanisms governing tumor development. Owing to this, various anti-cancer therapies were designed to target disease-specific mechanisms that are absent in normal cells. Such strategies include (i) inhibition of a specific oncoprotein, such as targeting the oncogenic fusion proteins Bcr–Abl and PML–RARA with Gleevec and all trans retinoic acid (ATRA) with arsenic trioxide respectively or (ii) activation of a specific immune response against cancerous cells demonstrated by the use of interferon alpha alone or in combination with other anti-cancer drugs including 5-fluorouracil and cytarabine (Raderer and Scheithauer, 1995; Guilhot et al., 1997; Druker et al., 2001; Kreitman et al., 2001; Tallman et al., 2002; Goldman and Melo, 2003; O’Brien et al., 2003; Sawyers, 2004; Kreitman, 2006; Ferrantini et al., 2007; Chin and Gray, 2008; Sellers, 2011). Many of these drugs are currently being used in the clinic and have established positive impact on patient survival. However, a major impediment to their success is the development of therapeutic resistance which in some cases predates clinical intervention (Wilson et al., 2009). Based on tumor response to the initial therapy, cancer resistance can be broadly classified into two categories, primary and acquired (Meads et al., 2009; Lippert et al., 2011). While primary drug resistance exists prior to any given treatment, acquired resistance occurs after initial therapy. Unfortunately, the majority of patients will likely develop resistance at a certain point of treatment. For example, 50–70% of patients with adenocarcinoma relapse following surgery with a chemoresistant phenotype (Castells et al., 2012), and approximately 20% of adults with acute lymphoblastic leukemia suffer from primary resistance to treatment (Testi et al., 1992; Giona et al., 1994; Thomas et al., 1999; O’Connor et al., 2011). In addition, primary resistance has been recognized in nearly 50% of all cancer patients in the 1990s (Pinedo and Giaccone, 1998). Therefore, the design of anti-cancer drugs that are fully effective necessitates a better understanding of the mechanisms by which cancer cells elude treatment. Here we will discuss several features of drug resistant cells including modification of drug transport, mutation of extracellular receptors, amplification and mutation of drug targets as well as related topics. Additionally, we will briefly address the important question of how resistant cell populations emerge.

Mechanisms of Drug Resistance

Both primary and acquired resistance can be caused by alterations to drug metabolism (sequestrations or enhanced detoxification) or modifications to the drug targets (Gottesman, 2002; Gatti and Zunino, 2005; Teicher, 2006; Wilson et al., 2006; Ullah, 2008). A brief overview of these mechanisms supported with examples of clinical relevance are presented below (Figure 1).

FIGURE 1
www.frontiersin.org

FIGURE 1. A major impediment in the treatment of cancer is the development of resistance. While most tumors initially respond to the given therapy, the majority will relapse following treatment, and in some cases resistance even predates clinical intervention. Therefore cancer resistance can be classified in to two broad classes: primary or acquired. In both cases, the emergence of resistant cells could be due to, at least, two mechanisms: (A) presence of multiple initial clones some of which emerge as dominant after treatment. These subpopulations could possess stem-like characteristics and/or use their interactions with the surrounding microenvironment to enter into a dormant state, thus surviving the insult of therapy. (B) Acquisition of stochastic alterations within the cancer cells per se. In all cases, the surviving population is less likely to respond to any further therapy and will be responsible for the minimal residual disease and cancer relapse. The biochemical underpinnings of resistance include: alterations to drug metabolism, increased drug efflux, decreased drug uptake, modification of the drug targets, amplification of targeted protein, genetic rewiring, enhanced DNA repair, inactivation of apoptotic proteins, or activation of anti-apoptotic ones, among others.

Perhaps the most studied mode of resistance involves drug metabolism, including its uptake, efflux, and detoxification. The means by which drugs enter cells depend on their chemical nature, and it mainly necessitates the use of receptors, which they bind to and transmit their effects without cellular entry, or transporters, which allow their cellular entry (Gottesman, 2002). At this level, resistance can result from mutations that modify activity or reduce the expression of surface receptors and transporters. For instance, mutations or reduced expression of the extracellular receptor smoothened (Yauch et al., 2009; Atwood et al., 2012; Kasper and Toftgard, 2013), nucleoside transporters (Galmarini et al., 2001; Damaraju et al., 2003) or one or both folate transporters (Longo-Sorbello and Bertino, 2001) result in defective uptake of cyclopamine, nucleoside drugs, such as cytarabine, and toxic folate analogs, such as methotrexate, respectively. On the other hand, enhanced drug efflux is frequently caused by increased expression of ATP binding cassette (ABC) membrane transporters (Gottesman et al., 2002). Among the 48 known ABC transporters in humans, elevation of three members, P-gp (MDR1 gene product), Multidrug resistance-associated protein 1 (MRP1) and mitoxantrone resistance protein [MXR; also known as breast cancer resistance protein (BCRP) or placenta ABC protein (ABC-P)], have been correlated with cancer chemoresistance to various drugs (Gottesman, 2002; Gottesman et al., 2002). For instance, P-gp transports a wide variety of hydrophobic anti-cancer drugs such as vinblastine, doxorubicin, vincristine, and taxol, and therefore its increased expression has been correlated with resistance to these (Gottesman et al., 2002). MRP1 on the other hand, transports negatively charged natural-product drugs in addition to drugs that have been modified by the conjugation of glutathione (GSH), glucuronic acid or sulfate (Jedlitschky et al., 1996; Hipfner et al., 1999; Konig et al., 1999; Borst et al., 2000); while, MXR overexpression has been correlated with resistance to topoisomerase I inhibitors, anthracyclines, and mitoxantrone (Gottesman, 2002). As can be seen, these factors comprise a major site for the development of drug resistance.

To exert their cytotoxic effects, many anti-cancer drugs must undergo metabolic activation. For instance, cytarabine (also known as AraC), a nucleoside drug widely used for the treatment of acute myelogenous leukemia (Sampath et al., 2006), necessitates initial phosphorylation by deoxycytidine kinase to cytarabine-monophosphate which is subsequently phosphorylated to the active form cytarabine triphosphate. To circumvent the effects of these drugs, cancer cells develop resistance through decreased drug activation (Kufe and Spriggs, 1985; Bardenheuer et al., 2005). This occurs via the downregulation or mutation of enzymes involved in this metabolic pathway, such as deoxycytidine kinase in the case of cytarabine (Sampath et al., 2006). Drug inactivation can also play a major role in the development of resistance. These mechanisms include, for example, conjugation of the drug to GSH, a powerful anti-oxidant that protects the cells against the damaging effects of reactive oxygen species (Wilson et al., 2006). GSH conjugation to platinum drugs, such as oxaliplatin and cisplatin used in the treatment of various types of cancers, renders them substrates for ABC transporters which enhances drug efflux (Meijer et al., 1992; Ishikawa and Ali-Osman, 1993). Furthermore, the topoisomerase I inhibitor, irinotecan, used for treating colon cancer, have been shown to become inactivated via phase I drug metabolizing enzymes, CYP450 (Xu and Villalona-Calero, 2002). Finally, binding of platinum drugs, particularly cisplatin, to metallothionein (MT), a small cysteine-rich protein, is another means of drug inactivation (Kelley et al., 1988; Kasahara et al., 1991).

Many cancer cells develop an overreliance or dependency on an oncogene. This is referred to as oncogene addiction (Arber et al., 1997; Weinstein, 2002; Weinstein and Joe, 2006; Sharma and Settleman, 2007). Targeting such oncogenes, provided a basis for the development of targeted therapies. Examples of such targeted therapies include: (i) imatinib targeting BCR/ABL tyrosine kinase in CML (Hughes et al., 2003), (ii) gefitinib and erlotinib targeting the epidermal growth factor receptor (EGFR) tyrosine kinase domain in non-small cell lung carcinoma (Lynch et al., 2004; Shepherd et al., 2005; Taron et al., 2005), and (iii) trastuzumab targeting human epidermal growth factor receptor-2 (HER-2) receptor in breast carcinomas (Slamon et al., 2001; Piccart-Gebhart et al., 2005). Unfortunately, the long term effectiveness of these drugs is hindered by the development of drug resistance due to mutation of the targeted protein (Gioeli, 2011; Wong and Lee, 2012). In the case of BCR/ABL and EGFR inhibitors, resistance emerges as a result of mutations occurring at the gatekeeper residues of the kinase domain which disables drug binding (Gorre et al., 2001; Blencke et al., 2003; Kobayashi et al., 2005; Pao et al., 2005; Soverini et al., 2005; Balak et al., 2006; Jabbour et al., 2006, 2008; Nicolini et al., 2006; Apperley, 2007; Costa et al., 2007; Bean et al., 2008; Gioeli, 2011). Furthermore, it has been demonstrated that resistance mutations can be detected prior to treatment in small subpopulations of tumor cells suggesting that these mutant forms were selected via the targeted therapy used (Hofmann et al., 2003; Toyooka et al., 2005; Inukai et al., 2006). In essence, understanding how mutations in the target proteins confer resistance enables the development of new therapeutic approaches to surmount resistance. For instance, second generation CML inhibitors have been developed based on mutational studies of patients who have become Gleevec resistant.

Other mechanisms by which cancerous cells circumvent the effects of targeted inhibitors have also been described, including amplification of alternative oncogenes or inactivation of alternative survival pathways (le Coutre et al., 2000; Engelman et al., 2007). In some cases, targeting of one protein alone (that cells are showing dependency on) can become ineffective because another parallel pathway supports tumor survival. In this case, the two pathways develop a synthetic lethal relationship (Hartman et al., 2001; Tucker and Fields, 2003). This way, the loss/inactivation of one of these genes would be supported by the other pathway and for the most effective treatment, one would need to target both pathways (Luo et al., 2009; Nijman, 2011).

An example of new pathways emerging once another pathway is targeted comes from the work of Isoyama et al. (2012), showed that acquired resistance to phosphatidylinositol 3-kinase (PI3K) inhibitors (such as ZSTK474) was due to the upregulation of insulin-like growth factor 1 receptor (IGF1R) pathway and that inhibition of this pathway with selective IGF1R inhibitors reverses the acquired PI3Ki resistance phenotype (Isoyama et al., 2012). Additionally, resistance could result from evasion of apoptotic pathways triggered by the acquisition of either inactivating mutations in genes coding for apoptotic proteins, such as p53, or activating mutations in genes coding for anti-apoptotic proteins, such as B cell lymphoma 2 (Bcl-2; Teicher, 2006). Indeed p53 mutations have been correlated with de novo resistance to doxorubicin treatment in patients with advanced breast cancer, as well as resistance to anthracyclines in a mouse sarcoma tumor model (Aas et al., 1996; Levine, 1997).

Another excellent example of this phenomenon (i.e., synthetic lethality) is seen in breast and ovarian cancers carrying mutations in the BRCA1 and BRCA2 genes, important mediators of DNA double-strand break (DSB) repair. When the poly (ADP-ribose) polymerase (PARP) protein, which is involved in different cellular processes including DNA repair, was targeted in these tumors, selective cancer cell toxicity was achieved (Bryant et al., 2005; Farmer et al., 2005). Several PARP inhibitors (PARPi) are currently being tested in clinical trials, such as iniparib (phase III ongoing; Guha, 2011) and veliparib cancer (Trudeau et al., 2006; Palma et al., 2008, 2009; Kummar et al., 2011), among others. However, despite the promising results these inhibitors showed, whether used as a mono- or combinatorial therapy (Juvekar et al., 2012; Kummar et al., 2012; Riffell et al., 2012), cancer cells once again were capable of evolving resistance to PARPi in preclinical and clinical settings (Chiarugi, 2012; Montoni et al., 2013). The mechanisms of resistance to these inhibitors have been grouped in to at least four categories, as summarized recently (Montoni et al., 2013). But perhaps the most distinct of these, was the ability of cancer cells to revert sensitivity to PARPi by acquiring deletion of the mutation in BRCA gene, thus restoring its function and the subsequent repair of DSBs.

Development of Cross Resistance

An important feature of drug resistance, is that development of resistance to one drug can lead to resistance to other drugs (Ullah, 2008). For instance, loss of a drug transporter can lead to resistance to structurally diverse compounds that utilize it or elevation of ABC transporters resulting from one therapy will influence the efficacy of many other compounds. Since this multidrug resistance phenotype correlates with poor chemotherapy response, drug development strategies to overcome this problem are being designed. These include drugs that are not recognized by transporters and therefore evade efflux, efflux inhibitors, drugs that are selectively lethal to P-gp expressing cells, etc. (Hall et al., 2009; Kelly et al., 2011; Nobili et al., 2012). But, perhaps resistance is not useless after all, as Hall et al. (2009) proposed. The alternative strategy to treat the progeny of the drug imposed Darwinian selection process is to identify their new “Achilles’ heel,” where resistance to the first given drug conferred a hypersensitivity to an alternate cytotoxic agent to which parental cells were not sensitive to. A phenomenon referred to as “collateral sensitivity”, which could be considered as a type of synthetic lethality as well since the same genetic alteration that rendered the cells resistant to one drug now sensitizes them to another (Hall et al., 2009; Pluchino et al., 2012).

Where do Resistant Cells Come From?

The development of human cancers is a complex multistage process involving accumulation of both genetic and epigenetic alterations over time (Caulin and Maley, 2011). As a consequence, a single tumor is comprised of heterogeneous populations of cells with distinct genetic fingerprints (Heppner et al., 1978; Marusyk and Polyak, 2010; Michor and Polyak, 2010). As the tumor progresses, some cells undergo genetic alterations, with selection of those having a superior growth advantage in a given context. An excellent example of tumor heterogeneity is provided by breast cancer studies (Schvimer et al., 1995; Shen et al., 2000; Wild et al., 2000). Wild et al. (2000) demonstrated that about 97% of epithelial breast carcinomas possess high levels of intra-tumor diverseness. The relevance of this innate heterogeneity is seen in cancer resistance. Since cancer cell selection obeys the Darwinian law of evolution, hence, under therapeutic pressure, those populations that are most adaptive or resistant to treatment will be selected for. These clones will then dominate and populate the tumor rendering it highly resistant to the given therapy (Williams and Nesse, 1991; Nesse, 2001; Breivik, 2005; Crespi and Summers, 2005; Lichtenstein, 2005; Monceviciute-Eringiene, 2005; Greaves, 2007). The selection process can be rationalized by, at least, two mechanisms. First, the emergence of a dominant cellular population after drug selection since it possesses some favorable characteristics such as a mutated drug binding site (Zhang et al., 2006; O’Brien et al., 2007; Ricci-Vitiani et al., 2007). The second mechanism involves the acquisition of stochastic alterations within the cancer cells which provide a survival advantage (Campbell et al., 2008; Stratton et al., 2009; Negrini et al., 2010; The International Cancer Genome Consortium, 2010; Shen, 2011). The advantage itself, e.g., a mutation in a drug binding site or alteration in drug transporters (as just two examples) could be the same for either of these mechanisms. What is different is the underlying process to generate these biochemical differences.

Two known models, the cancer stem cell (CSC) model, and the environment-mediated drug resistance (EMDR) model, which are not mutually exclusive, could explain the origin of resistant cells. In the CSC model, rare populations of cancer stem cells possess tumor-initiating properties (Teicher, 2006; Nguyen et al., 2012). It is thought that CSCs diverge from normal tissue stem cells or from more-differentiated progenitor cells through dysregulation of self-renewal pathways. Beside modulation of molecular mechanisms, such as increased efficiency of DNA repair (Potten et al., 2002; Cai et al., 2004; Park and Gerson, 2005), changes in cell cycle parameters (Venezia et al., 2004), overexpression of anti-apoptotic proteins (Wang et al., 2003) or drug transporters (Gottesman et al., 2002; Krishnamurthy et al., 2004), etc., resistance of CSCs could be due to their quiescent nature (Teicher, 2006). Thus, in this case, the cell population is present and is difficult to target using traditional chemotherapy strategies many of which depend on active cell cycling.

In the EMDR model, resistance emerges as the cancer cells use their interactions with the surrounding microenvironment to enter into a quiescent or dormant state as a means of circumventing the effects of the given therapy. Under the drug imposed selection pressure, these cells remain in their protective shelter, undergoing genetic changes until they ultimately reach a more permanent acquired resistance phenotype and in turn, alter their surrounding microenvironment (Braun et al., 2000; Meads et al., 2009). These surviving populations, which may or may not be CSCs, can contribute to minimal residual disease (MRD) and cancer relapse (Matsunaga et al., 2003; Bidard et al., 2008; Meads et al., 2009). The EMDR model is relevant to both hematopoietic and metastatic epithelial malignancies. EMDR could be mediated by either soluble or cell adhesion-related microenvironmental factors. Soluble factor-mediated drug resistance occurs through induction of gene transcription within the tumor cells by cytokines, chemokines, or growth factors secreted by neighboring stroma-like fibroblasts (Meads et al., 2009). One of the known mediators of this resistance mechanism is interleukin-6 (IL-6), whose increased secretion has been correlated with resistance to various cytotoxins both in in vitro and in vivo models. This includes, for instance, resistance to bortezomib in multiple myeloma and to etoposide and cisplatin in hormone-independent prostate carcinomas (Borsellino et al., 1995, 1999; Frassanito et al., 2001; Voorhees et al., 2007). Further, cell adhesion-mediated drug resistance is triggered by the adhesion of integrins from tumor cells to stromal fibroblasts or to components of the surrounding extracellular matrix. Molecularly, this process could be due to many scenarios including (i) degradation of apoptotic proteins or (ii) enhanced stability or altered subcellular distribution of anti-apoptotic proteins and cell cycle regulators (Hazlehurst et al., 2001, 2007; Shain et al., 2002, 2009; Lwin et al., 2007). One example is provided by studies into melphalan resistance. In this case, the cancerous cells tend to use their adhesion to fibronectin in the surrounding microenvironment to reduce the endogenous levels of the proapoptotic BH3-only Bcl-2 family member, Bim1, thus conferring resistance by disabling apoptosis (Hazlehurst et al., 2003; Hanahan and Weinberg, 2011). From a clinical point of view, it is thought that combining current therapies with inhibitors of EMDR pathways could enhance the effectiveness of the treatment (Croix et al., 1996; Weaver et al., 1997; Hazlehurst et al., 2000; White et al., 2004; Lwin et al., 2007). A proof-of-principle example was demonstrated by the combination of melphalan, a DNA alkylating agent used in the treatment of multiple myeloma and ovarian carcinomas, with an anti-integrin α-4 antibody (natalizumab) which significantly inhibited myeloma growth and reduced tumor burden in patients (Mori et al., 2004; Engelhardt and Kappos, 2008).

Conclusion

Resistance to drugs continues to be a major problem in oncology affecting the majority of cancer patients. Here we provide many examples of how cells become resistant to various drugs including alteration in drug metabolism, modification of drug targets, and genetic rewiring of cells to bypass targeted pathways. A better understanding of oncogene networks and oncogene cooperativity will likely improve therapeutic strategies by identifying optimal combinations based on the genetic lesions in the tumors. Importantly, tumors are highly heterogenous and this heterogeneity may well substantially contribute to primary or acquired resistance. Armed with a greater understanding of the mechanisms of drug resistance will undoubtedly lead to more long term remissions and hopefully cures.

Conflict of Interest Statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Acknowledgments

This work was supported by National Institutes for Health (NIH) and The Leukemia and Lymphoma Society (USA). Hiba Zahreddine holds a scholarship from the National Council for Scientific Research (CNRS)-Lebanon. Katherine L. B. Borden holds a Canada Research Chair.

References

Aas, T., Borresen, A. L., Geisler, S., Smith-Sorensen, B., Johnsen, H., Varhaug, J. E., et al. (1996). Specific P53 mutations are associated with de novo resistance to doxorubicin in breast cancer patients. Nat. Med. 2, 811–814.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Apperley, J. F. (2007). Part I: mechanisms of resistance to imatinib in chronic myeloid leukaemia. Lancet Oncol. 8, 1018–1029.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Arber, N., Doki, Y., Han, E. K., Sgambato, A., Zhou, P., Kim, N. H., et al. (1997). Antisense to cyclin D1 inhibits the growth and tumorigenicity of human colon cancer cells. Cancer Res. 57, 1569–1574.

Pubmed Abstract | Pubmed Full Text

Atwood, S. X., Chang, A. L., and Oro, A. E. (2012). Hedgehog pathway inhibition and the race against tumor evolution. J. Cell Biol. 199, 193–197.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Balak, M. N., Gong, Y., Riely, G. J., Somwar, R., Li, A. R., Zakowski, M. F., et al. (2006). Novel D761Y and common secondary T790M mutations in epidermal growth factor receptor-mutant lung adenocarcinomas with acquired resistance to kinase inhibitors. Clin. Cancer Res. 12, 6494–6501.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Bardenheuer, W., Lehmberg, K., Rattmann, I., Brueckner, A., Schneider, A., Sorg, U. R., et al. (2005). Resistance to cytarabine and gemcitabine and in vitro selection of transduced cells after retroviral expression of cytidine deaminase in human hematopoietic progenitor cells. Leukemia 19, 2281–2288.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Bean, J., Riely, G. J., Balak, M., Marks, J. L., Ladanyi, M., Miller, V. A., et al. (2008). Acquired resistance to epidermal growth factor receptor kinase inhibitors associated with a novel T854A mutation in a patient with EGFR-mutant lung adenocarcinoma. Clin. Cancer Res. 14, 7519–7525.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Bidard, F. C., Vincent-Salomon, A., Gomme, S., Nos, C., De Rycke, Y., Thiery, J. P., et al. (2008). Disseminated tumor cells of breast cancer patients: a strong prognostic factor for distant and local relapse. Clin. Cancer Res. 14, 3306–3311.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Blencke, S., Ullrich, A., and Daub, H. (2003). Mutation of threonine 766 in the epidermal growth factor receptor reveals a hotspot for resistance formation against selective tyrosine kinase inhibitors. J. Biol. Chem. 278, 15435–15440.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Borsellino, N., Belldegrun, A., and Bonavida, B. (1995). Endogenous interleukin 6 is a resistance factor for cis-diamminedichloroplatinum and etoposide-mediated cytotoxicity of human prostate carcinoma cell lines. Cancer Res. 55, 4633–4639.

Pubmed Abstract | Pubmed Full Text

Borsellino, N., Bonavida, B., Ciliberto, G., Toniatti, C., Travali, S., and D’Alessandro, N. (1999). Blocking signaling through the Gp130 receptor chain by interleukin-6 and oncostatin M inhibits PC-3 cell growth and sensitizes the tumor cells to etoposide and cisplatin-mediated cytotoxicity. Cancer 85, 134–144.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Borst, P., Evers, R., Kool, M., and Wijnholds, J. (2000). A family of drug transporters: the multidrug resistance-associated proteins. J. Natl. Cancer Inst. 92, 1295–1302.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Braun, S., Kentenich, C., Janni, W., Hepp, F., De Waal, J., Willgeroth, F., et al. (2000). Lack of effect of adjuvant chemotherapy on the elimination of single dormant tumor cells in bone marrow of high-risk breast cancer patients. J. Clin. Oncol. 18, 80–86.

Pubmed Abstract | Pubmed Full Text

Breivik, J. (2005). The evolutionary origin of genetic instability in cancer development. Semin. Cancer Biol. 15, 51–60.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Bryant, H. E., Schultz, N., Thomas, H. D., Parker, K. M., Flower, D., Lopez, E., et al. (2005). Specific killing of BRCA2-deficient tumours with inhibitors of poly(ADP-ribose) polymerase. Nature 434, 913–917.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Cai, J., Weiss, M. L., and Rao, M. S. (2004). In search of “stemness”. Exp. Hematol. 32, 585–598.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Campbell, P. J., Stephens, P. J., Pleasance, E. D., O’Meara, S., Li, H., Santarius, T., et al. (2008). Identification of somatically acquired rearrangements in cancer using genome-wide massively parallel paired-end sequencing. Nat. Genet. 40, 722–729.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Castells, M., Thibault, B., Delord, J. P., and Couderc, B. (2012). Implication of tumor microenvironment in chemoresistance: tumor-associated stromal cells protect tumor cells from cell death. Int. J. Mol. Sci. 13, 9545–9571.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Caulin, A. F., and Maley, C. C. (2011). Peto’s Paradox: evolution’s prescription for cancer prevention. Trends Ecol. Evol. 26, 175–182.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Chiarugi, A. (2012). A snapshot of chemoresistance to PARP inhibitors. Trends Pharmacol. Sci. 33, 42–48.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Chin, L., and Gray, J. W. (2008). Translating insights from the cancer genome into clinical practice. Nature 452, 553–563.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Costa, D. B., Halmos, B., Kumar, A., Schumer, S. T., Huberman, M. S., Boggon, T. J., et al. (2007). BIM mediates EGFR tyrosine kinase inhibitor-induced apoptosis in lung cancers with oncogenic EGFR mutations. PLoS Med. 4:e315. doi: 10.1371/journal.pmed.0040315

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Crespi, B., and Summers, K. (2005). Evolutionary biology of cancer. Trends Ecol. Evol. 20, 545–552.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Croix, B. S., Rak, J. W., Kapitain, S., Sheehan, C., Graham, C. H., and Kerbel, R. S. (1996). Reversal by hyaluronidase of adhesion-dependent multicellular drug resistance in mammary carcinoma cells. J. Natl. Cancer Inst. 88, 1285–1296.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Damaraju, V. L., Damaraju, S., Young, J. D., Baldwin, S. A., Mackey, J., Sawyer, M. B., et al. (2003). Nucleoside anticancer drugs: the role of nucleoside transporters in resistance to cancer chemotherapy. Oncogene 22, 7524–7536.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Druker, B. J., Talpaz, M., Resta, D. J., Peng, B., Buchdunger, E., Ford, J. M., et al. (2001). Efficacy and safety of a specific inhibitor of the BCR–ABL tyrosine kinase in chronic myeloid leukemia. N. Engl. J. Med. 344, 1031–1037.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Engelhardt, B., and Kappos, L. (2008). Natalizumab: targeting alpha4-integrins in multiple sclerosis. Neurodegener. Dis. 5, 16–22.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Engelman, J. A., Zejnullahu, K., Mitsudomi, T., Song, Y., Hyland, C., Park, J. O., et al. (2007). MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling. Science 316, 1039–1043.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Farmer, H., Mccabe, N., Lord, C. J., Tutt, A. N., Johnson, D. A., Richardson, T. B., et al. (2005). Targeting the DNA repair defect in BRCA mutant cells as a therapeutic strategy. Nature 434, 917–921.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Ferrantini, M., Capone, I., and Belardelli, F. (2007). Interferon-alpha and cancer: mechanisms of action and new perspectives of clinical use. Biochimie 89, 884–893.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Frassanito, M. A., Cusmai, A., Iodice, G., and Dammacco, F. (2001). Autocrine interleukin-6 production and highly malignant multiple myeloma: relation with resistance to drug-induced apoptosis. Blood 97, 483–489.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Frei, E. III, Karon, M., Levin, R. H., Freireich, E. J., Taylor, R. J., Hananian, J., et al. (1965). The effectiveness of combinations of antileukemic agents in inducing and maintaining remission in children with acute leukemia. Blood 26, 642–656.

Pubmed Abstract | Pubmed Full Text

Galmarini, C. M., Mackey, J. R., and Dumontet, C. (2001). Nucleoside analogues: mechanisms of drug resistance and reversal strategies. Leukemia 15, 875–890.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Gatti, L., and Zunino, F. (2005). Overview of tumor cell chemoresistance mechanisms. Methods Mol. Med. 111, 127–148.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Gioeli, D. (2011). Targeted Therapies Mechanisms of Resistance. New York: Humana Press.

CrossRef Full Text

Giona, F., Testi, A. M., Annino, L., Amadori, S., Arcese, W., Camera, A., et al. (1994). Treatment of primary refractory and relapsed acute lymphoblastic leukaemia in children and adults: the GIMEMA/AIEOP experience. Gruppo Italiano Malattie Ematologiche Maligne dell’Adulto. Associazione Italiana Ematologia ed Ocologia Pediatrica. Br. J. Haematol. 86, 55–61.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Goldman, J. M., and Melo, J. V. (2003). Chronic myeloid leukemia – advances in biology and new approaches to treatment. N. Engl. J. Med. 349, 1451–1464.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Gorre, M. E., Mohammed, M., Ellwood, K., Hsu, N., Paquette, R., Rao, P. N., et al. (2001). Clinical resistance to STI-571 cancer therapy caused by BCR–ABL gene mutation or amplification. Science 293, 876–880.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Gottesman, M. M. (2002). Mechanisms of cancer drug resistance. Annu. Rev. Med. 53, 615–627.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Gottesman, M. M., Fojo, T., and Bates, S. E. (2002). Multidrug resistance in cancer: role of ATP-dependent transporters. Nat. Rev. Cancer 2, 48–58.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Greaves, M. (2007). Darwinian medicine: a case for cancer. Nat. Rev. Cancer 7, 213–221.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Guha, M. (2011). PARP inhibitors stumble in breast cancer. Nat. Biotechnol. 29, 373–374.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Guilhot, F., Chastang, C., Michallet, M., Guerci, A., Harousseau, J. L., Maloisel, F., et al. (1997). Interferon alfa-2b combined with cytarabine versus interferon alone in chronic myelogenous leukemia. French Chronic Myeloid Leukemia Study Group. N. Engl. J. Med. 337, 223–229.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Hall, M. D., Handley, M. D., and Gottesman, M. M. (2009). Is resistance useless? Multidrug resistance and collateral sensitivity. Trends Pharmacol. Sci. 30, 546–556.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Hanahan, D., and Weinberg, R. A. (2011). Hallmarks of cancer: the next generation. Cell 144, 646–674.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Hartman, J. L. T., Garvik, B., and Hartwell, L. (2001). Principles for the buffering of genetic variation. Science 291, 1001–1004.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Hazlehurst, L. A., Argilagos, R. F., and Dalton, W. S. (2007). Beta1 integrin mediated adhesion increases Bim protein degradation and contributes to drug resistance in leukaemia cells. Br. J. Haematol. 136, 269–275.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Hazlehurst, L. A., Damiano, J. S., Buyuksal, I., Pledger, W. J., and Dalton, W. S. (2000). Adhesion to fibronectin via beta1 integrins regulates p27kip1 levels and contributes to cell adhesion mediated drug resistance (CAM-DR). Oncogene 19, 4319–4327.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Hazlehurst, L. A., Enkemann, S. A., Beam, C. A., Argilagos, R. F., Painter, J., Shain, K. H., et al. (2003). Genotypic and phenotypic comparisons of de novo and acquired melphalan resistance in an isogenic multiple myeloma cell line model. Cancer Res. 63, 7900–7906.

Pubmed Abstract | Pubmed Full Text

Hazlehurst, L. A., Valkov, N., Wisner, L., Storey, J. A., Boulware, D., Sullivan, D. M., et al. (2001). Reduction in drug-induced DNA double-strand breaks associated with beta1 integrin-mediated adhesion correlates with drug resistance in U937 cells. Blood 98, 1897–1903.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Heppner, G. H., Dexter, D. L., Denucci, T., Miller, F. R., and Calabresi, P. (1978). Heterogeneity in drug sensitivity among tumor cell subpopulations of a single mammary tumor. Cancer Res. 38, 3758–3763.

Pubmed Abstract | Pubmed Full Text

Hipfner, D. R., Deeley, R. G., and Cole, S. P. (1999). Structural, mechanistic and clinical aspects of MRP1. Biochim. Biophys. Acta 1461, 359–376.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Hofmann, W. K., Komor, M., Wassmann, B., Jones, L. C., Gschaidmeier, H., Hoelzer, D., et al. (2003). Presence of the BCR–ABL mutation Glu255Lys prior to STI571 (imatinib) treatment in patients with Ph+ acute lymphoblastic leukemia. Blood 102, 659–661.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Hughes, T. P., Kaeda, J., Branford, S., Rudzki, Z., Hochhaus, A., Hensley, M. L., et al. (2003). Frequency of major molecular responses to imatinib or interferon alfa plus cytarabine in newly diagnosed chronic myeloid leukemia. N. Engl. J. Med. 349, 1423–1432.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Inukai, M., Toyooka, S., Ito, S., Asano, H., Ichihara, S., Soh, J., et al. (2006). Presence of epidermal growth factor receptor gene T790M mutation as a minor clone in non-small cell lung cancer. Cancer Res. 66, 7854–7858.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Ishikawa, T., and Ali-Osman, F. (1993). Glutathione-associated cis-diamminedichloroplatinum(II) metabolism and ATP-dependent efflux from leukemia cells. Molecular characterization of glutathione-platinum complex and its biological significance. J. Biol. Chem. 268, 20116–20125.

Pubmed Abstract | Pubmed Full Text

Isoyama, S., Dan, S., Nishimura, Y., Nakamura, N., Kajiwara, G., Seki, M., et al. (2012). Establishment of phosphatidylinositol 3-kinase inhibitor-resistant cancer cell lines and therapeutic strategies for overcoming the resistance. Cancer Sci. 103, 1955–1960.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Jabbour, E., Kantarjian, H., Jones, D., Breeden, M., Garcia-Manero, G., O’Brien, S., et al. (2008). Characteristics and outcomes of patients with chronic myeloid leukemia and T315I mutation following failure of imatinib mesylate therapy. Blood 112, 53–55.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Jabbour, E., Kantarjian, H., Jones, D., Talpaz, M., Bekele, N., O’Brien, S., et al. (2006). Frequency and clinical significance of BCR–ABL mutations in patients with chronic myeloid leukemia treated with imatinib mesylate. Leukemia 20, 1767–1773.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Jedlitschky, G., Leier, I., Buchholz, U., Barnouin, K., Kurz, G., and Keppler, D. (1996). Transport of glutathione, glucuronate, and sulfate conjugates by the MRP gene-encoded conjugate export pump. Cancer Res. 56, 988–994.

Pubmed Abstract | Pubmed Full Text

Juvekar, A., Burga, L. N., Hu, H., Lunsford, E. P., Ibrahim, Y. H., Balmana, J., et al. (2012). Combining a PI3K inhibitor with a PARP inhibitor provides an effective therapy for BRCA1-related breast cancer. Cancer Discov. 2, 1048–1063.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Kasahara, K., Fujiwara, Y., Nishio, K., Ohmori, T., Sugimoto, Y., Komiya, K., et al. (1991). Metallothionein content correlates with the sensitivity of human small cell lung cancer cell lines to cisplatin. Cancer Res. 51, 3237–3242.

Pubmed Abstract | Pubmed Full Text

Kasper, M., and Toftgard, R. (2013). Smoothing out drug resistance. Cancer Cell 23, 3–5.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Kelley, S. L., Basu, A., Teicher, B. A., Hacker, M. P., Hamer, D. H., and Lazo, J. S. (1988). Overexpression of metallothionein confers resistance to anticancer drugs. Science 241, 1813–1815.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Kelly, R. J., Draper, D., Chen, C. C., Robey, R. W., Figg, W. D., Piekarz, R. L., et al. (2011). A pharmacodynamic study of docetaxel in combination with the P-glycoprotein antagonist tariquidar (XR9576) in patients with lung, ovarian, and cervical cancer. Clin. Cancer Res. 17, 569–580.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Kobayashi, S., Boggon, T. J., Dayaram, T., Janne, P. A., Kocher, O., Meyerson, M., et al. (2005). EGFR mutation and resistance of non-small-cell lung cancer to gefitinib. N. Engl. J. Med. 352, 786–792.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Konig, J., Nies, A. T., Cui, Y., Leier, I., and Keppler, D. (1999). Conjugate export pumps of the multidrug resistance protein (MRP) family: localization, substrate specificity, and MRP2-mediated drug resistance. Biochim. Biophys. Acta 1461, 377–394.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Kreitman, R. J. (2006). Immunotoxins for targeted cancer therapy. AAPS J. 8, E532–E551.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Kreitman, R. J., Wilson, W. H., Bergeron, K., Raggio, M., Stetler-Stevenson, M., Fitzgerald, D. J., et al. (2001). Efficacy of the anti-CD22 recombinant immunotoxin BL22 in chemotherapy-resistant hairy-cell leukemia. N. Engl. J. Med. 345, 241–247.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Krishnamurthy, P., Ross, D. D., Nakanishi, T., Bailey-Dell, K., Zhou, S., Mercer, K. E., et al. (2004). The stem cell marker Bcrp/ABCG2 enhances hypoxic cell survival through interactions with heme. J. Biol. Chem. 279, 24218–24225.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Kufe, D. W., and Spriggs, D. R. (1985). Biochemical and cellular pharmacology of cytosine arabinoside. Semin. Oncol. 12, 34–48.

Pubmed Abstract | Pubmed Full Text

Kummar, S., Chen, A., Ji, J., Zhang, Y., Reid, J. M., Ames, M., et al. (2011). Phase I study of PARP inhibitor ABT-888 in combination with topotecan in adults with refractory solid tumors and lymphomas. Cancer Res. 71, 5626–5634.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Kummar, S., Chen, A., Parchment, R. E., Kinders, R. J., Ji, J., Tomaszewski, J. E., et al. (2012). Advances in using PARP inhibitors to treat cancer. BMC Med. 10:25. doi: 10.1186/1741-7015-10-25

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

le Coutre, P., Tassi, E., Varella-Garcia, M., Barni, R., Mologni, L., Cabrita, G., et al. (2000). Induction of resistance to the Abelson inhibitor STI571 in human leukemic cells through gene amplification. Blood 95, 1758–1766.

Pubmed Abstract | Pubmed Full Text

Levine, A. J. (1997). p53, the cellular gatekeeper for growth and division. Cell 88, 323–331.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Lichtenstein, A. V. (2005). On evolutionary origin of cancer. Cancer Cell Int. 5, 5.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Lippert, T. H., Ruoff, H. J., and Volm, M. (2011). Current status of methods to assess cancer drug resistance. Int. J. Med. Sci. 8, 245–253.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Longo-Sorbello, G. S., and Bertino, J. R. (2001). Current understanding of methotrexate pharmacology and efficacy in acute leukemias. Use of newer antifolates in clinical trials. Haematologica 86, 121–127.

Pubmed Abstract | Pubmed Full Text

Luo, J., Solimini, N. L., and Elledge, S. J. (2009). Principles of cancer therapy: oncogene and non-oncogene addiction. Cell 136, 823–837.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Lwin, T., Hazlehurst, L. A., Dessureault, S., Lai, R., Bai, W., Sotomayor, E., et al. (2007). Cell adhesion induces p27Kip1-associated cell-cycle arrest through down-regulation of the SCFSkp2 ubiquitin ligase pathway in mantle-cell and other non-Hodgkin B-cell lymphomas. Blood 110, 1631–1638.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Lynch, T. J., Bell, D. W., Sordella, R., Gurubhagavatula, S., Okimoto, R. A., Brannigan, B. W., et al. (2004). Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. N. Engl. J. Med. 350, 2129–2139.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Marusyk, A., and Polyak, K. (2010). Tumor heterogeneity: causes and consequences. Biochim. Biophys. Acta 1805, 105–117.

CrossRef Full Text

Matsunaga, T., Takemoto, N., Sato, T., Takimoto, R., Tanaka, I., Fujimi, A., et al. (2003). Interaction between leukemic-cell VLA-4 and stromal fibronectin is a decisive factor for minimal residual disease of acute myelogenous leukemia. Nat. Med. 9, 1158–1165.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Meads, M. B., Gatenby, R. A., and Dalton, W. S. (2009). Environment-mediated drug resistance: a major contributor to minimal residual disease. Nat. Rev. Cancer 9, 665–674.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Meijer, C., Mulder, N. H., Timmer-Bosscha, H., Sluiter, W. J., Meersma, G. J., and De Vries, E. G. (1992). Relationship of cellular glutathione to the cytotoxicity and resistance of seven platinum compounds. Cancer Res. 52, 6885–6889.

Pubmed Abstract | Pubmed Full Text

Michor, F., and Polyak, K. (2010). The origins and implications of intratumor heterogeneity. Cancer Prev. Res. (Phila) 3, 1361–1364.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Monceviciute-Eringiene, E. (2005). Neoplastic growth: the consequence of evolutionary malignant resistance to chronic damage for survival of cells (review of a new theory of the origin of cancer). Med. Hypotheses 65, 595–604.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Montoni, A., Robu, M., Pouliot, E., and Shah, G. M. (2013). Resistance to PARP-inhibitors in Cancer Therapy. Front. Pharmacol. 4:18. doi: 10.3389/fphar.2013.00018

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Mori, Y., Shimizu, N., Dallas, M., Niewolna, M., Story, B., Williams, P. J., et al. (2004). Anti-alpha4 integrin antibody suppresses the development of multiple myeloma and associated osteoclastic osteolysis. Blood 104, 2149–2154.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Mukherjee, S. (2010). The Emperor of All Maladies: a Biography of Cancer. New York: Scribner.

Negrini, S., Gorgoulis, V. G., and Halazonetis, T. D. (2010). Genomic instability – an evolving hallmark of cancer. Nat. Rev. Mol. Cell Biol. 11, 220–228.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Nesse, R. M. (2001). How is Darwinian medicine useful? West. J. Med. 174, 358–360.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Nguyen, L. V., Vanner, R., Dirks, P., and Eaves, C. J. (2012). Cancer stem cells: an evolving concept. Nat. Rev. Cancer 12, 133–143.

Pubmed Abstract | Pubmed Full Text

Nicolini, F. E., Corm, S., Le, Q. H., Sorel, N., Hayette, S., Bories, D., et al. (2006). Mutation status and clinical outcome of 89 imatinib mesylate-resistant chronic myelogenous leukemia patients: a retrospective analysis from the French intergroup of CML (Fi(phi)-LMC GROUP). Leukemia 20, 1061–1066.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Nijman, S. M. (2011). Synthetic lethality: general principles, utility and detection using genetic screens in human cells. FEBS Lett. 585, 1–6.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Nobili, S., Landini, I., Mazzei, T., and Mini, E. (2012). Overcoming tumor multidrug resistance using drugs able to evade P-glycoprotein or to exploit its expression. Med. Res. Rev. 32, 1220–1262.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

O’Brien, C. A., Pollett, A., Gallinger, S., and Dick, J. E. (2007). A human colon cancer cell capable of initiating tumour growth in immunodeficient mice. Nature 445, 106–110.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

O’Brien, S. G., Guilhot, F., Larson, R. A., Gathmann, I., Baccarani, M., Cervantes, F., et al. (2003). Imatinib compared with interferon and low-dose cytarabine for newly diagnosed chronic-phase chronic myeloid leukemia. N. Engl. J. Med. 348, 994–1004.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

O’Connor, D., Sibson, K., Caswell, M., Connor, P., Cummins, M., Mitchell, C., et al. (2011). Early UK experience in the use of clofarabine in the treatment of relapsed and refractory paediatric acute lymphoblastic leukaemia. Br. J. Haematol. 154, 482–485.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Palma, J. P., Rodriguez, L. E., Bontcheva-Diaz, V. D., Bouska, J. J., Bukofzer, G., Colon-Lopez, M., et al. (2008). The PARP inhibitor, ABT-888 potentiates temozolomide: correlation with drug levels and reduction in PARP activity in vivo. Anticancer. Res. 28, 2625–2635.

Pubmed Abstract | Pubmed Full Text

Palma, J. P., Wang, Y. C., Rodriguez, L. E., Montgomery, D., Ellis, P. A., Bukofzer, G., et al. (2009). ABT-888 confers broad in vivo activity in combination with temozolomide in diverse tumors. Clin. Cancer Res. 15, 7277–7290.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Pao, W., Miller, V. A., Politi, K. A., Riely, G. J., Somwar, R., Zakowski, M. F., et al. (2005). Acquired resistance of lung adenocarcinomas to gefitinib or erlotinib is associated with a second mutation in the EGFR kinase domain. PLoS Med. 2:e73. doi: 10.1371/journal.pmed.0020073

CrossRef Full Text

Park, Y., and Gerson, S. L. (2005). DNA repair defects in stem cell function and aging. Annu. Rev. Med. 56, 495–508.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Piccart-Gebhart, M. J., Procter, M., Leyland-Jones, B., Goldhirsch, A., Untch, M., Smith, I., et al. (2005). Trastuzumab after adjuvant chemotherapy in HER2-positive breast cancer. N. Engl. J. Med. 353, 1659–1672.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Pinedo, H. M., and Giaccone, G. (1998). Drug Resistance in the Treatment of Cancer. Cambridge: Cambridge University Press.

CrossRef Full Text

Pluchino, K. M., Hall, M. D., Goldsborough, A. S., Callaghan, R., and Gottesman, M. M. (2012). Collateral sensitivity as a strategy against cancer multidrug resistance. Drug Resist. Updat. 15, 98–105.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Potten, C. S., Owen, G., and Booth, D. (2002). Intestinal stem cells protect their genome by selective segregation of template DNA strands. J. Cell Sci. 115, 2381–2388.

Pubmed Abstract | Pubmed Full Text

Raderer, M., and Scheithauer, W. (1995). Treatment of advanced colorectal cancer with 5-fluorouracil and interferon-alpha: an overview of clinical trials. Eur. J. Cancer 31A, 1002–1008.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Ricci-Vitiani, L., Lombardi, D. G., Pilozzi, E., Biffoni, M., Todaro, M., Peschle, C., et al. (2007). Identification and expansion of human colon-cancer-initiating cells. Nature 445, 111–115.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Riffell, J. L., Lord, C. J., and Ashworth, A. (2012). Tankyrase-targeted therapeutics: expanding opportunities in the PARP family. Nat. Rev. Drug Discov. 11, 923–936.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Sampath, D., Cortes, J., Estrov, Z., Du, M., Shi, Z., Andreeff, M., et al. (2006). Pharmacodynamics of cytarabine alone and in combination with 7-hydroxystaurosporine (UCN-01) in AML blasts in vitro and during a clinical trial. Blood 107, 2517–2524.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Sawyers, C. (2004). Targeted cancer therapy. Nature 432, 294–297.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Schvimer, M., Lash, R. H., and Katzin, W. E. (1995). Intratumoral heterogeneity of DNA ploidy in breast carcinomas: a flow cytometric assessment of sampling techniques. Cytometry 22, 292–296.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Sellers, W. R. (2011). A blueprint for advancing genetics-based cancer therapy. Cell 147, 26–31.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Shain, K. H., Landowski, T. H., and Dalton, W. S. (2002). Adhesion-mediated intracellular redistribution of c-Fas-associated death domain-like IL-1-converting enzyme-like inhibitory protein-long confers resistance to CD95-induced apoptosis in hematopoietic cancer cell lines. J. Immunol. 168, 2544–2553.

Pubmed Abstract | Pubmed Full Text

Shain, K. H., Yarde, D. N., Meads, M. B., Huang, M., Jove, R., Hazlehurst, L. A., et al. (2009). Beta1 integrin adhesion enhances IL-6-mediated STAT3 signaling in myeloma cells: implications for microenvironment influence on tumor survival and proliferation. Cancer Res. 69, 1009–1015.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Sharma, S. V., and Settleman, J. (2007). Oncogene addiction: setting the stage for molecularly targeted cancer therapy. Genes Dev. 21, 3214–3231.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Shen, C. Y., Yu, J. C., Lo, Y. L., Kuo, C. H., Yue, C. T., Jou, Y. S., et al. (2000). Genome-wide search for loss of heterozygosity using laser capture microdissected tissue of breast carcinoma: an implication for mutator phenotype and breast cancer pathogenesis. Cancer Res. 60, 3884–3892.

Pubmed Abstract | Pubmed Full Text

Shen, Z. (2011). Genomic instability and cancer: an introduction. J. Mol. Cell Biol. 3, 1–3.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Shepherd, F. A., Rodrigues Pereira, J., Ciuleanu, T., Tan, E. H., Hirsh, V., Thongprasert, S., et al. (2005). Erlotinib in previously treated non-small-cell lung cancer. N. Engl. J. Med. 353, 123–132.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Siegel, R., Desantis, C., Virgo, K., Stein, K., Mariotto, A., Smith, T., et al. (2012). Cancer treatment and survivorship statistics, 2012. CA Cancer J. Clin. 62, 220–241.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Slamon, D. J., Leyland-Jones, B., Shak, S., Fuchs, H., Paton, V., Bajamonde, A., et al. (2001). Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N. Engl. J. Med. 344, 783–792.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Soverini, S., Martinelli, G., Rosti, G., Bassi, S., Amabile, M., Poerio, A., et al. (2005). ABL mutations in late chronic phase chronic myeloid leukemia patients with up-front cytogenetic resistance to imatinib are associated with a greater likelihood of progression to blast crisis and shorter survival: a study by the GIMEMA Working Party on Chronic Myeloid Leukemia. J. Clin. Oncol. 23, 4100–4109.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Stratton, M. R., Campbell, P. J., and Futreal, P. A. (2009). The cancer genome. Nature 458, 719–724.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Tallman, M. S., Nabhan, C., Feusner, J. H., and Rowe, J. M. (2002). Acute promyelocytic leukemia: evolving therapeutic strategies. Blood 99, 759–767.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Taron, M., Ichinose, Y., Rosell, R., Mok, T., Massuti, B., Zamora, L., et al. (2005). Activating mutations in the tyrosine kinase domain of the epidermal growth factor receptor are associated with improved survival in gefitinib-treated chemorefractory lung adenocarcinomas. Clin. Cancer Res. 11, 5878–5885.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Teicher, B. A. (2006). Cancer Drug Resistance. Totowa: Humana Press.

CrossRef Full Text

Testi, A. M., Moleti, M. L., Giona, F., Iori, A. P., Meloni, G., Miniero, R., et al. (1992). Treatment of primary refractory or relapsed acute lymphoblastic leukemia (ALL) in children. Ann. Oncol. 3, 765–767.

Pubmed Abstract | Pubmed Full Text

The International Cancer Genome Consortium. (2010). International network of cancer genome projects. Nature 464, 993–998.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Thomas, D. A., Kantarjian, H., Smith, T. L., Koller, C., Cortes, J., O’Brien, S., et al. (1999). Primary refractory and relapsed adult acute lymphoblastic leukemia: characteristics, treatment results, and prognosis with salvage therapy. Cancer 86, 1216–1230.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Toyooka, S., Kiura, K., and Mitsudomi, T. (2005). EGFR mutation and response of lung cancer to gefitinib. N. Engl. J. Med. 352, 2136; author reply 2136.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Trudeau, M. E., Crump, M., Charpentier, D., Yelle, L., Bordeleau, L., Matthews, S., et al. (2006). Temozolomide in metastatic breast cancer (MBC): a phase II trial of the National Cancer Institute of Canada – Clinical Trials Group (NCIC-CTG). Ann. Oncol. 17, 952–956.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Tucker, C. L., and Fields, S. (2003). Lethal combinations. Nat. Genet. 35, 204–205.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Ullah, M. F. (2008). Cancer multidrug resistance (MDR): a major impediment to effective chemotherapy. Asian Pac. J. Cancer Prev. 9, 1–6.

Pubmed Abstract | Pubmed Full Text

Venezia, T. A., Merchant, A. A., Ramos, C. A., Whitehouse, N. L., Young, A. S., Shaw, C. A., et al. (2004). Molecular signatures of proliferation and quiescence in hematopoietic stem cells. PLoS Biol. 2:e301. doi: 10.1371/journal.pbio.0020301

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Voorhees, P. M., Chen, Q., Kuhn, D. J., Small, G. W., Hunsucker, S. A., Strader, J. S., et al. (2007). Inhibition of interleukin-6 signaling with CNTO 328 enhances the activity of bortezomib in preclinical models of multiple myeloma. Clin. Cancer Res. 13, 6469–6478.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Wang, S., Yang, D., and Lippman, M. E. (2003). Targeting Bcl-2 and Bcl-XL with nonpeptidic small-molecule antagonists. Semin. Oncol. 30, 133–142.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Weaver, V. M., Petersen, O. W., Wang, F., Larabell, C. A., Briand, P., Damsky, C., et al. (1997). Reversion of the malignant phenotype of human breast cells in three-dimensional culture and in vivo by integrin blocking antibodies. J. Cell Biol. 137, 231–245.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Weinstein, I. B. (2002). Cancer. Addiction to oncogenes – the Achilles heal of cancer. Science 297, 63–64.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Weinstein, I. B., and Joe, A. K. (2006). Mechanisms of disease: oncogene addiction – a rationale for molecular targeting in cancer therapy. Nat. Clin. Pract. Oncol. 3, 448–457.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

White, D. E., Kurpios, N. A., Zuo, D., Hassell, J. A., Blaess, S., Mueller, U., et al. (2004). Targeted disruption of beta1-integrin in a transgenic mouse model of human breast cancer reveals an essential role in mammary tumor induction. Cancer Cell 6, 159–170.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Wild, P., Knuechel, R., Dietmaier, W., Hofstaedter, F., and Hartmann, A. (2000). Laser microdissection and microsatellite analyses of breast cancer reveal a high degree of tumor heterogeneity. Pathobiology 68, 180–190.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Williams, G. C., and Nesse, R. M. (1991). The dawn of Darwinian medicine. Q. Rev. Biol. 66, 1–22.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Wilson, T. R., Johnston, P. G., and Longley, D. B. (2009). Anti-apoptotic mechanisms of drug resistance in cancer. Curr. Cancer Drug Targets 9, 307–319.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Wilson, T. R., Longley, D. B., and Johnston, P. G. (2006). Chemoresistance in solid tumours. Ann. Oncol. 17(Suppl. 10), x315–x324.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Wong, A. L., and Lee, S. C. (2012). Mechanisms of resistance to trastuzumab and novel therapeutic strategies in HER2-positive breast cancer. Int. J. Breast Cancer 2012, 415170.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Xu, Y., and Villalona-Calero, M. A. (2002). Irinotecan: mechanisms of tumor resistance and novel strategies for modulating its activity. Ann. Oncol. 13, 1841–1851.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Yauch, R. L., Dijkgraaf, G. J., Alicke, B., Januario, T., Ahn, C. P., Holcomb, T., et al. (2009). Smoothened mutation confers resistance to a Hedgehog pathway inhibitor in medulloblastoma. Science 326, 572–574.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Zhang, P., Zuo, H., Ozaki, T., Nakagomi, N., and Kakudo, K. (2006). Cancer stem cell hypothesis in thyroid cancer. Pathol. Int. 56, 485–489.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Keywords: origin of cancer, multidrug resistance, drug metabolism, drug transporters, oncogene addiction, microenvironment, collateral sensitivity, synthetic lethality

Citation: Zahreddine H and Borden KLB (2013) Mechanisms and insights into drug resistance in cancer. Front. Pharmacol. 4:28. doi: 10.3389/fphar.2013.00028

Received: 31 December 2012; Paper pending published: 17 January 2013;
Accepted: 25 February 2013; Published online: 14 March 2013.

Edited by:

Gerald Batist, McGill University, Canada

Reviewed by:

Jian Hui Wu, McGill University, Canada
Michael Witcher, McGill University, Canada

Copyright: © 2013 Zahreddine and Borden. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits use, distribution and reproduction in other forums, provided the original authors and source are credited and subject to any copyright notices concerning any third-party graphics etc.

*Correspondence: Katherine L. B. Borden, Department of Pathology and Cell Biology, Institute of Research in Immunology and Cancer, Université de Montréal, Montreal, QC, Canada H3T 1J4. e-mail: katherine.borden@umontreal.ca

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.