Abstract
Carcinogenesis is the uncontrolled growth of cells gaining the potential to invade and disrupt vital tissue functions. This malignant process includes the occurrence of ‘unwanted’ gene mutations that induce the transformation of normal cells, for example, by overactivation of pro-oncogenic pathways and inactivation of tumor-suppressive or anti-oncogenic pathways. It is now recognized that the number of major signaling pathways that control oncogenesis is not unlimited; therefore, suppressing these pathways can conceivably lead to a cancer cure. However, the clinical application of cancer intervention has not matched up to scientific expectations. Increasing numbers of studies have revealed that many oncogenic-signaling elements show double faces, in which they can promote or suppress cancer pathogenesis depending on tissue type, cancer stage, gene dosage and their interaction with other players in carcinogenesis. This complexity of oncogenic signaling poses challenges to traditional cancer therapy and calls for considerable caution when designing an anticancer drug strategy. We propose future oncology interventions with the concept of integrative cancer therapy.
Keywords: cell signaling, cell survival, integrative cancer therapy, oncogene, tumor suppressor
Goal of cancer therapy
Carcinogenesis includes ‘unwanted’ gene mutations that induce the transformation of normal cells, for example, by overactivation of protooncogenes, such as ras, src or abl, and inactivation of tumor suppressors, such as p53 and PTEN (Table1) [1–5]. Tumorigenic gene mutations can also be induced by harmful environmental factors, such as tobacco overusage and UV exposure [6–8]. On the other hand, cancer development can be viewed from the point of evolution, in which case, genetic drifting followed by selection acting upon growth-control genes by chance may be sufficient to introduce oncogenic mutations that cause malignant transformation [9,10]. During the repetitive repairing of damaged tissues by stem cells, deviation from recapitulation of normal developmental processes may drive cells towards malignancy [11,12]. Regardless of the underlying causes, the consequence is the same: the activation of pro-oncogenic signaling pathways and the downregulation of anti-oncogenic pathways that normally keep cellular behaviors in check. The outcome of the overgrowth of cells capable of metastasizing to vital organs is the disruption of normal functions of the body, ultimately resulting in death. The goal of cancer therapy is to suppress or eliminate the cancer growth and metastasis by reversing oncogenic signaling pathways. However, increasing amounts of literature have demonstrated considerable complexities in the regulation of pro- and anti-oncogenic pathways, which pose huge challenges for traditional cancer therapies.
Table 1. Pro- and anti-oncogenic functions of oncogenes and tumor suppressors in carcinogenesis.
Name | Functions | Pro-oncogenic | Anti-oncogenic | Mutations† | Ref. |
---|---|---|---|---|---|
C-myc | Cell proliferation | + | + | ND | [199] |
E2F1 | Transcription factor | + | + | ND | [45–47,200,201] |
K-ras | Kirsten ras oncogene homolog | + | + | Mutations in codons 12 or 13 | [202–205] |
Src | Tyrosine kinase | + | + | Transformation-defective mutants | [1] |
Abl | Tyrosine kinase | + | + | SH3 domain, ATP-binding domain mutation | [206–208] |
Rb | Cell proliferation | + | + | C-terminal caspase cleavage site mutation; loss of heterozygosity | [44,209,210] |
Aurora A | A serine/threonine kinase in mitotic regulation | + | + | ND | [211,212] |
β-catenin | Key player in cell–cell adhesion and also in the Wnt signaling pathways | + | + | Mutations in codon 32 or 3 | [213–220] |
APC | Mitotic regulation | + | + | Truncating mutation; loss of heterozygosity | [210,221–226] |
p120ctn | Regulate E-cadherin stability and Rho small GTPases | + | + | ND | [69–72,227, 228–233] |
δ-catenin | Neural gene misregulated in cancer | + | + | 5′-UTR mutation | [66–68,73,75,79, 234,235] |
Raf | Functions downstream of the Ras family GTPases | + | + | Mutations in codon 600 | [129–139] |
miRNAs | Function in the negative regulation of gene expression | + | + | ND | [236–248] |
EBNA1 | Human herpes virus | + | + | ND | [249] |
TGF-β | Growth factor initially growth inhibitory and then becoming tumor promoting | + | + | ND | [250–254] |
IRF-4 | A hematopoietic cell-restricted transcription factor | + | + | ND | [255] |
GSK-3 | A key serine/threonine kinase in Wnt signaling pathway | + | + | ND | [256] |
Notch | Signaling pathway for cell–cell communication and gene regulation | + | + | Int3 (insertion site) | [153–165, 177,178] |
PI3K | A family of lipid kinases that propagates intracellular signaling cascades | + | + | Somatic mutations in PIK3CA | [257–265] |
AKT | Serine/threonine-specific protein kinase | + | + | AKT1 E17K mutation | [266–270] |
p53 | Tumor suppressor called the guardian of the genome | + | + | DNA-binding domain mutations | [142–145, 147–152,271] |
PTEN | Negatively regulates PI3K/AKT signaling pathways | + | + | Gene loss | [82,105,106, 128,140,141] |
Here we only show some mutation sites or domains as examples.
+: Literature cited to support the findings; ND: Mutations not discussed in this article; UTR: Untranslated region.
Progresses in detecting & treating cancers
Early detection
In 2007, the steepest decline in cancer deaths in the USA was recorded. This significant milestone reflects the outcome of many years of investment in cancer research and care, and it is one of the most encouraging signs of progress since the war on cancer began in the 1970s. It is clear that early detection saves lives and increases treatment options. For example, mammography can detect breast cancer at an early stage when treatment can be more effective and survival is more likely. Clinical data indicate that, on average, mammography can detect approximately 80–90% of breast cancers in women without alarming symptoms [13,14]. Indeed, the recent decline in breast cancer mortality rates among women can be attributed to a combination of early detection and improvements in treatments. Another good example is the early detection of prostate cancer in men.
Although there are insufficient data to recommend for or against prostate cancer screening in men at average risk of developing the disease, the American Cancer Society recommends the prostate-specific antigen (PSA) blood test and the digital rectal examination for men at average risk, beginning at the age of 50 years. Individuals at higher risk of developing prostate cancer (e.g., African–Americans or men with a strong family history) are encouraged to begin screening at the age of 45 years. Despite the controversial clinical trial outcomes regarding PSA reliability and utility, and the clear need to provide additional viable biomarkers in prostate cancer diagnosis and prognosis, over the past 20 years the availability of the PSA test has led to significant advances in the early detection of many prostate cancers [15,16].
Effective therapies
There have been remarkable successes in the treatment of certain cancers. Pioneering chemotherapy drugs, such as platinum compounds are good examples. Cisplatin suppresses cancer cell proliferation by disrupting DNA synthesis and is extremely effective in inhibiting germ-cell-derivative cancers. It was originally found to elicit anticancer properties in tumor-bearing mice in 1968 [17–19]. The results from the preliminary studies were so promising that patients were treated with cisplatin for the first time in 1971, and cisplatin received US FDA approval shortly thereafter [20,21]. Before cisplatin was available, metastatic testicular cancer had a mortality rate of 95%. Cisplatin-based regimens resulted in a 60% cure rate for this disease [22,23].
Unlike cisplatin, Gleevec® (imatinib mesylate; Novartis, Basel, Switzerland) is effective for the treatment of certain cancer types by interfering with cell-signaling pathways [24–26]. By blocking the abnormal protein tyrosine kinase BCR-ABL, Gleevec induces leukemia cells to undergo apoptosis. The FDA approved Gleevec in 2001 for the treatment of chronic myeloid leukemia (CML) and for the treatment of a rare form of stomach cancer, gastrointestinal stromal tumor in 2002 [27,28]. The outcome of Gleevec clinical trials was remarkable. In 1999, a small-scale clinical study demonstrated that with an effective daily dose, all patients had their blood counts return to normal with minimal side effects. Such dramatic results were uncommon in any early clinical cancer study [29]. A 2001 study further reported that Gleevec restored normal blood counts in 53 out of 54 chemotherapy-resistant CML patients, a response rate rarely seen in oncology trials with a single antineoplastic agent [30]. Almost all of the patients were still doing well after 1 year on Gleevec, and few complained about side effects. Before Gleevec, efforts on the discovery of anticancer drugs by targeting tyrosine kinases were largely futile. The success of Gleevec and the newer generation inhibitors renewed hopes that inhibition of specific signaling elements of carcinogenesis can produce effective drugs.
Challenges on most fronts of anticancer efforts
Lack of reliable cancer biomarkers
Despite the successes in certain areas of cancer diagnosis and therapy, we still face tremendous challenges in treating most cancer types. In colorectal cancers, when cancers are detected at an early, localized stage, the 5-year survival rate is 90%; however, fewer than half of all colorectal cancer cases are diagnosed at this stage, mostly owing to low screening rates [31]. After the cancer has spread regionally to involve adjacent organs or lymph nodes, the 5-year survival rate for the disease drops to 68%. For patients with distant metastases, the 5-year survival rate drops precipitously to only 10%. For lung cancers, the 5-year survival rate for all stages combined is only 15%. The survival rate is 49% for cases detected when the disease is still localized; however, only 16% of lung cancer cases are diagnosed at this early stage. For pancreatic cancer, at the present time, there is still no method for early detection at all, and fewer than 10% of cases are diagnosed at an early stage. For all stages combined, the 1- and 5-year relative survival rates are only 24 and 5%, respectively. Even for patients diagnosed with local disease occurrence, the 5-year survival rate for pancreatic cancer is only 20% [301]. Additional frustration at the inability to detect and combat these types of cancers is exemplified because the diseases are not uncommon among the working-age population when productivity is high.
From the previous discussions, it is clear that despite the billions of dollars invested in research and development, we are still far from achieving the mission: a cure for cancer. What are the modern strategies for drug discovery against cancer? Most anticancer strategies focus on attacking specific elements of a given biological pathway of cancer cells to achieve target specificity with minimized toxicity. This approach, although it has clear merits, has not produced the desired numbers of viable drugs because most drug leads that are successful in vitro or in animal studies do not show the same efficacy in human trials. Some leads, even when proven to be effective in human trials, either encounter resistance during treatment or lose efficacy owing to mutations of the target genes. Other potential leads may be abandoned during early discovery stages owing to low efficacy seen in selected screening models in vitro, while they could be effective in human trials. Consequently, few viable cancer drugs have entered the market in recent years, leading to skyrocketing research and development costs. Clearly, we should re-examine our anticancer drug-discovery approaches in current literature and re-evaluate our strategies for future anticancer therapy.
Complex roles of pro- & anti-oncogenic pathways
As we further explored cancer cell-signaling events dictated by pro- and anti-oncogenic pathways, more and more surprising research results were found [32,33]. When oncogenes were first identified in the 1970s, the perception was that if we inhibited the functions of certain oncogenes, we could stop the disease. It was also thought that we could increase the function of tumor suppressors to inhibit disease progression [34–36]. However, we now know that many genes and signaling pathways can be pro-oncogenic under one context but anti-oncogenic under a different context, at different stages or in different tissue types of cancer development [37–42]. In this article, we will discuss some common examples (for additional review, see Lu et al. [43]).
Retinoblastoma/E2F pathway
Cancer cells often contain mutations that lead to the loss of retinoblastoma (Rb) tumor suppressor function and the activation of E2F-dependent transcription (Table 1). As a result, cell proliferation is deregulated, and the sensitivity of responses to apoptotic stimuli is increased. Rb protein regulates cell proliferation and cell death. Animal model studies suggest that Rb tumor-suppressor function, that is, inhibition of proliferation, is inactivated by phosphorylation, whereas Rb tumor-promoting function, that is, inhibition of apoptosis, is inactivated by caspase cleavage [44]. On the other hand, Johnson showed that E2F1 can function as both an oncogene and a tumor suppressor gene, and both positive and negative effects on tumorigenesis can be observed depending on whether E2F1 is absent or overexpressed [42]. In a prostate tissue-recombination model, homozygous deletion of Rb elicited increased E2F activity, activation of E2F-target genes and susceptibility to hormonal carcinogenesis [45]. A closer tissue microarray examination of E2F1 expression in human specimens found that E2F1 levels were low in benign and localized prostate cancer, modestly elevated in metastatic lymph nodes from hormone-naive patients, and significantly elevated in metastatic tissues from hormone-resistant prostate cancer patients [46]. By contrast, strong androgen receptor expression was detected in benign prostate, localized prostate cancer and lymph node metastasis, but decreased in metastatic hormone-resistant prostate cancer. Since E2F1 can repress androgen receptor transcription, elevated E2F1 levels may contribute to the progression of hormone-refractory prostate cancer [47]. These studies suggest that in the early stages of prostate cancer development, E2F1 levels are low, probably to allow its oncogenic effects to be best exerted with the cooperation of other oncogenic mutations, such as Ras, to avoid its potential tumor suppressive effects. Therefore, E2F1 is equally capable of inducing cell proliferation and apoptosis [48–50].
TGF pathway
TGF-β1 is a cytokine that displays context-dependent tumor suppressive and tumor-promoting activity in carcinogenesis. For most normal epithelial cells and at the early stages of tumor development, TGF-β1 is believed to function as a tumor suppressor [51]. This process occurs largely through the activation of surface TGF-β1 receptor complex and phosphorylation of Smads, although other signaling pathways are also involved [52]. By contrast, elevated expression of TGF-β1 may enhance the malignant properties of tumor cells through effects on cell invasion, metastasis, epithelial to mesenchymal transition or antitumor immunity. This alteration in the functions of TGF-β1 in cancer development is probably due to its interaction with other signaling pathways, such as ras oncogene, that becomes activated during tumor progression [53]. Indeed, oncogenic Ras and TGF-β1 can perhaps cooperate to promote epithelial to mesenchymal transition and the invasive behavior of neoplastic cells [54–55].
Two-stage chemical carcinogenesis of the mouse epidermis is a widely used model to study the interaction of TGF-β1 and oncogenic Ras. TGF-β signaling can suppress squamous tumor formation and progression initiated by an activated cellular or viral ras oncogene [56–59]. However, other studies showed that overexpression of TGF-β1 in normal skin can lead to the formation of highly malignant spindle cell carcinomas, as well as enhancing a metastatic phenotype of benign squamous tumor cells [60,61]. It is suggested that the different roles of TGF-β1 in skin carcinogenesis may depend on the interaction of Smad2 and Smad3 with Ras signaling. Smad2 and Smad3 expression are lost during progression of chemically induced tumors, and the expression of v-ras in Smad3-null mouse epidermal keratinocytes causes rapid progression to squamous cell carcinoma in a skin graft system [62,63]. Studies have shown that Smad3 is required for tumor formation in the two-stage carcinogenesis model, suggesting that context-dependent interactions between Ras and Smads are critical for tumor suppression or progression in this tissue model [64].
Cadherin/catenin pathway
Recent studies suggested that cell–cell junction proteins, such as p120ctn (CTNND1) and δ-catenin/NPRAP/neurojungin (CTNND2) may also play pro-oncogenic roles in one context and anti-oncogenic roles in another. δ-catenin, a close member of the p120ctn subgroup of the armadillo/β-catenin superfamily, shares more than 30% amino acid sequence identity in the armadillo domains with p120ctn, and binds to the same juxtamembrane region on E-cadherin as p120ctn [65,66]. Unlike p120ctn, which is ubiquitously expressed, δ-catenin is primarily expressed in the CNS [67,68]. p120ctn is frequently downregulated in many cancer types, although its expression remains paradoxically high in most cancer-derived cell lines [69]. Accumulating evidence indicates that p120ctn could play the role of tumor suppressor or metastasis promoter depending on whether it is downregulated before or after the loss of E-cadherin. If E-cadherin loss precedes p120ctn loss, p120ctn remains stranded in the cytoplasm and can promote cell invasion and metastasis through the regulation of Rho GTPases [70,71], which control the formation of actin-rich lamellipodia, filopodia, stress fibers and many other structures associated with cell motility, morphology and invasiveness. Thus, p120ctn may function as a metastasis promoter under these conditions. If p120ctn loss precedes E-cadherin loss, then it may become the initial event, ultimately leading to the inactivation of the cadherin complex. This is strongly suggested by studies that the loss of p120ctn destabilizes E-cadherin, which, in turn, may reduce levels of α- and β-catenin at the cell–cell junction [72].
In many carcinomas, p120ctn is downregulated while δ-catenin is overexpressed [73]. It is interesting and also paradoxical that δ-catenin expression levels in cancer-derived cell lines are generally moderate or minimal [74,75]. Its overexpression in prostate cancer cells altered tumor suppressor E-cadherin and p120ctn distribution at the cell–cell junction [73], disrupted the normal monolayer in cell culture [76] and promoted prostate tumorigenesis in mice [76]. Furthermore, a recent study identified the association of δ-catenin specifically with only pathological (but not physiological) angiogenesis, such as with cancer [77]. Thus, it behaves like a cancer-promoting protein. However, in normal fibroblast cells in culture, ectopic expression of δ-catenin results in the cessation of cell division to promote cellular protrusions [78]. Furthermore, in noncancerous mammary epithelial cells, δ-catenin appears to act as a tumor suppressor inhibiting cell-colony formation [79]. These studies raise intriguing possibilities that δ-catenin can exert context-dependent effects on normal epithelial cells as a tumor suppressor while acting as a tumor promoter on certain types of transformed cancer cells.
PI3K/PTEN/Akt pathway
Recent research on a widely studied oncogenic protein family, PI3Ks, indicated that multiple pathways, of which PI3Ks play critical roles, may function in opposite directions in carcinogenesis. The family of PI3Ks regulates signaling for diverse cellular functions, including cell division, migration, apoptosis and angiogenesis [80,81]. Disruption of this tightly regulated pathway by gene loss (PTEN), mutation (PIK3CA, AKT1 or, less commonly, PIK3R1) or amplification (PIK3CA) is one of the most common alterations in human cancers (Table 1) [82]. PIK3CA mutations lead to constitutive activation of p110 (the catalytic subunit of PI3K), increasing its lipid kinase activity and resulting in an increase in Akt activation [83–85]. Cultured cells expressing PIK3CA mutations exhibit features of cell transformation, promote cell survival and show increased angiogenesis [80,83,84,86–89]. However, in normal, nontransformed cells, Akt activation has an opposite effect through the induction of cellular senescence [90,91]. In animal models, differences in tumorigenic effects have also been observed. For example, while an increased PIK3CA activity induces tumorigenesis in xenografts [86,88], the expression of activated Akt1 is insufficient to induce tumors in mouse mammary epithelium [92,93]. Furthermore, although p110 expression induces mouse mammary tumors, they are generally microscopic with low frequency [94].
In human breast cancers, the PIK3CA mutation frequency ranges variably from 8 to 40% [95–100]. The majority of mutations in breast cancer occur at three hotspots: E542K and E545K at exon 9, which encode the helical domain, and H1047R at exon 20, which encodes the kinase domain [97–99]. In cell-based assays, these rare mutations confer a gain of function as measured by lipid kinase activity, constitutive activation of Akt, and cellular transformation, with a different range of oncogenic potency [83]. Therefore, the frequency of PIK3CA mutation is consistent with the significance of PI3Ks in breast cancer pathogenesis.
However, Kalinsky and colleagues recently identified a positive prognostic significance of PIK3CA mutations [101], indicating that PIK3CA can even be a ‘good’ activating mutation in breast cancer [102]. They analyzed the prognostic value of PI3KCA mutations in paraffin-embedded tumor tissue from 590 breast cancer patients with a median follow-up of 12.8 years. In this large cohort with optimal follow-up, they found PIK3CA mutations in 32.5% of invasive breast primary tumors, 24.1% occurring at the three ‘hot spot’ sites and the remaining 8.5% in the combined rare PIK3CA mutations. PI3KCA mutations were significantly more likely to occur in postmenopausal patients, aged 60–70 years, with hormone-receptor-positive, HER2-negative, and low-grade and -stage breast cancer at diagnosis. Patients with PI3KCA-mutated tumors showed a marginally significant longer progression-free survival and a significantly improved breast cancer-specific and overall survival [101]. Thus, this study proposes the positive prognostic significance of PIK3CA mutations and is clinically relevant, because it will significantly affect the design of clinical trials planned for PI3K-targeted therapy [102]. These considerations are also clinically important because the presence of activated PI3KCA in cancer has led to the development of PI3K inhibitors as a novel targeted therapeutic strategy. PI3K inhibitors have been demonstrated to be active in preclinical models [88], with many agents currently in the early stages of clinical development. These agents have different degrees of potency and target specificity. Some agents inhibit both PI3K and mTOR, whereas others are PI3K-specific inhibitors. This approach may be fruitful in targeting breast cancer, because a neoadjuvant study using an mTOR inhibitor in combination with an antiestrogen therapy exhibited silencing of the PI3K pathway and reduction in its activity in patients harboring PI3K mutations [90]. These agents also hold promise in combination with chemotherapy and in HER2-amplified breast cancer patients harboring either PI3KCA mutations or PTEN deletions [91].
Besides PI3K and mTOR inhibitors, targeting of other components of the pathway, such as AKT and Rictor, is also under investigation [103–106]. 17-allyl-aminogeldanamycin is a geldanamycin derivative currently employed in clinical trials [103]. In addition to downregulating Akt expression, 17-allyl-aminogeldanamycin also causes a rapid inhibition of Akt kinase activity prior to proteosomal degradation of HER2 and Akt [104]. CCI-779 and 40-O-(2-hydroxyethyl)-rapamycin are esterified rapamycin derivatives that improve drug solubility and oral bioavailability. Recent data suggest that tumor cell lines or murine tumors lacking PTEN are particularly sensitive to CCI-779 [105]. Phase II/III trials are underway in the treatment of several cancers including prostate cancer. While we cannot conclude for the efficacy for the CCI-779 from these trials, it would not be surprising if the outcomes are variable for different cancer types and cancers at the different stages. In consideration of the current therapeutic strategy with PI3K/AKT pathways, the study by Kalinsky and colleagues thus cautions that because tumors with PI3KCA mutations have a more favorable clinical outcome and these mutations are quite frequent, the presence of PI3KCA mutations will need to be taken into consideration when analyzing the effects of hormonal therapy.
MAPK pathway
The current consensus is that tumorigenesis requires the deregulation of at least six cellular processes [107], and cancer cells must acquire the following capabilities [108]:
Independence of proliferation signals;
Evasion of apoptosis;
Insensitivity to antigrowth signals;
Unlimited replicative potential;
The ability to invade and metastasize;
The ability to attract and sustain angiogenesis for nutrient supply.
Abnormalities in MAPK signaling impinge on most of these processes and play a critical role in the development and progression of cancer [109,110]. To date, six distinct groups of MAPKs have been characterized in mammals; ERK1/2, ERK3/4, ERK5, ERK7/8, JNK1/2/3 and the p38 isoforms α/β/γ(ERK6)/δ [111–114].
ERK is a downstream component of an evolutionarily conserved signaling module that is activated by the Raf serine/threonine kinase. Raf activates the MEK1/2 dual-specificity protein kinases, which then activate ERK1/2 [115]. The mutational activation of Raf plays important roles in human oncogenesis (Table 1) [116]. In addition, the Raf–MEK–ERK pathway is a key downstream effector of the Ras small GTPase, which is the most frequently mutated oncoprotein in human cancers. Ras is a key downstream effector of EGF receptor (EGFR), which is mutationally activated and/or overexpressed in a wide variety of human cancers [117,118]. ERK activation also promotes upregulation of EGFR ligands, promoting an autocrine growth loop critical for tumor growth. In addition, MAPK signaling pathway often plays key roles in the response of tumor cells to cancer therapies [119]. Thus, the EGFR–Ras–Raf–MEK–ERK signaling network has been the subject of intense research and pharmaceutical scrutiny to identify novel target-based approaches for cancer treatment.
For example, second-generation MEK inhibitors, such as PD184352 (also known as CI1040), have been developed and are currently in clinical trials [120–123]. However, how the activation of Ras/Raf/MEK/ERK leads to a cellular decision to proliferate or differentiate remains highly context dependent. A classical example is the neuronal differentiation of pheochromocytoma (PC12) cells. It is well documented that EGF-mediated activation of Ras/Raf/MEK/ERK signaling promotes cell proliferation, while NGF activates the same pathway to induce neuronal differentiation [124–126]. It was found that EGF stimulation actually results in a transient activation of ERK, while NGF elicits a sustained ERK activation, which leads to a neuronal differentiation program. However, this model may not be simply extended to other cancer cells under cytotoxic treatment. For example, the anticancer activity of ethyl-4-isothiocyanatobutanoate can lead to a rapid activation of the ERK signaling pathway coupled with delayed transition through the cell cycle and cell cycle arrest, which results in diminished mitochondrial membrane potential, culminating in apoptosis in leukemic HL-60 cells [127].
The role of the Ras/Raf/MEK/ERK pathway in the development of specific cancer is also not without controversy [128]. Take Raf activation as an example; overexpression of activated Raf proteins is associated with cell growth, cell cycle arrest or even apoptosis [129–131]. However, the fate of the cells depends on the expression level and isoform of Raf kinase. Overexpression of Raf proteins is associated with proliferation in hematopoietic cells [129], erythroid progenitor cells [132] and A10 smooth muscle cells [133]. However, overexpression of activated Raf proteins is also associated with cell cycle arrest in Schwann cells, PC12 cells, HL-60 cells, small-cell lung cancer cells and some hematopoietic cells [134–136]. Depending on the Raf isoform, overexpression of Raf can either lead to cell proliferation (A-Raf or Raf-1) or cell-growth arrest (B-Raf) in NIH-3T3 fibroblasts and FDC-P1 hematopoietic cells [129,137,138]. It is not clear why overexpression of the Raf gene can lead to such diverse and conflicting results. One possibility is that the opposite outcomes may be determined by the amount or activity of the particular Raf oncoprotein [138,139]. Therefore, cancer therapies targeted at the inhibition of Raf kinase expression and/or activity should consider the potential off-target effects in different types of cancer cells.
Furthermore, crosstalks between Ras/Raf/MEK/ERK and PI3K/PTEN/AKT signaling pathways can be observed in some advanced prostate cancer cells. These cells express increased levels of activated Akt [140,141], which may suppress Raf activation. Introduction of activated forms of Akt increased the drug resistance of advanced prostate cancer cells. However, introduction of activated forms of Raf did not increase the drug resistance of the prostate cancer cells. In contrast to the results observed in hematopoietic cells, Raf may normally promote differentiation in prostate cells, which is suppressed in advanced prostate cancer owing to increased expression of activated Akt, resulting from PTEN mutation. Thus in advanced prostate cancer, it may be advantageous to induce Raf expression to promote differentiation, while in hematopoietic cancers, it may be better to inhibit Raf/MEK/ERK-induced proliferation to achieve optimal effects [128].
p53 functions
Mutations in p53 are found in most tumor types and contribute to the complex network of molecular events leading to tumor formation (Table 1). p53 is perhaps one of the most consistent players performing roles as a tumor suppressor and is called the ‘guardian of the genome’ [142]. Wild-type p53 is believed to be continuously monitoring the integrity of the DNA molecule. When changes in the DNA are detected, p53 protein binds to a gene to promote the transcription of p21cip1, which, in turn, enters the metabolic process of the cell to shut down the cell cycle at the G1 phase [143]. This block of cell cycle progression allows time for the cell to repair the damaged DNA before it enters the DNA replication phase. If repair is not possible, p53 stimulates the cell to enter a pathway leading to apoptosis. In total, p53 targets approximately 150 genes to prevent the proliferation of damaged cells [144,145]. In carcinogenesis, p53 is mutated and can no longer bind DNA in an effective way. As a consequence, the p21cip1 protein is not available to act as the ‘stop signal’ for cell division. With the DNA structure now unprotected against defects, mutations accumulate, checks and balances on the cell cycle fail to operate, and cell growth proceeds unchecked, which result in tumorigenesis. Recent studies showed that mutant p53 proteins not only represent the loss of wild-type p53 tumor suppressor activity, but also gain new oncogenic properties favoring the insurgence, maintenance, spreading and chemoresistance of malignant tumors [146].
There have been hundreds of clinical trials targeted at restoring p53 function [147]. Gene therapy involves restoring the function of a defective gene by the addition of a normal gene into the DNA structure. Roth et al. attached wild-type p53 gene to an adenovirus and injected it directly into tumors, which led to tumor regression [148]. The function of p53 protein is completely dependent upon maintaining the correct 3D conformation of the molecule. Pfizer conducted studies in which they inserted small synthetic molecules into the DNA-binding region of mutated p53 protein [149]. This technique stabilized the active conformation of newly synthesized mutated p53, and has the ability to inhibit tumor growth in mice. In addition, studies have shown that some components, such as resveratrol or vitamin A, activate or increase the expression of p53, leading to cell cycle arrest and apoptosis [150–152].
Notch/γ-secretase pathway
The diverse roles that Notch signaling plays during the development and maintenance of normal tissues are recapitulated and associated with both tumor-suppressive and oncogenic functions in different forms of cancer. Depending on the tumor type, Notch can variously promote or limit tumor growth through either cell-autonomous or -nonautonomous effects on differentiation, cellular metabolism, cell cycle progression, angiogenesis, and possibly self-renewal and immune function.
The first data describing the oncogenic consequences of aberrant Notch signaling in solid tumors was derived from animal studies characterizing a frequent insertion site, named int3, in the mouse mammary tumor virus (Table 1) [153]. Studies showed that aberrant Notch signaling is significant for human solid cancers, such as breast cancer, medulloblastoma, colorectal cancer and pancreatic cancer, as well as melanoma and leukemia [154–160].
Notch can also act as a tumor suppressor in certain contexts. Instead of maintaining progenitor cells in an undifferentiated state or influencing their cell fate decisions, Notch can induce differentiation in some tissues, which is associated with growth suppression. The best-studied example is the role of Notch in the skin. Conditional Notch1 knockout mice develop cutaneous basal cell carcinoma-like lesions that have increased levels of Hedgehog and Wnt signaling [161]. In vitro data from both human and mouse keratinocytes suggest that Notch signaling induces differentiation, which is accompanied by cell cycle arrest. Negative crosstalk between Notch and p63 regulates the balance between self-renewal and differentiation, and the dysregulation of this Notch function could contribute to keratinocyte transformation [162–164]. Another property of Notch1 activation is the induction of early differentiation markers, including Keratin1/10 and involucrin, and the downmodulation of integrin expression [164]. Conditional inactivation of signaling components of the Notch cascade in mouse skin results in hyperproliferation of the skin and epidermal cyst formation. Over time, Notch1-deficient animals develop spontaneous, highly vascularized basal cell carcinoma-like tumors [161,165].
Another feature of the complex nature of Notch signaling in cancer can be reflected by the opposite outcomes of the Notch-activated downstream elements, such as Hairy and enhancer of split (HES)1 protein, the human homolog of Drosophila Hes1 and a basic helix–loop–helix transcriptional repressor. The Notch–HES1 axis in signaling is intricately modulated to control proliferation, differentiation and apoptosis [166]. Notch activation involves its proteolytic cleavage by γ-secretase and the generation of an intracellular Notch fragment to enter nuclei and activate transcriptions. HES1 is a target of Notch1 signaling that is aberrantly activated in a variety of human cancers, including prostate, lung, colorectal, osteogenic and breast carcinomas [167–171]. HES1 expression is downregulated in the nonmetastatic cancer cell line LNCaP compared with that in metastatic cancer cells (C4-2B) and, in this case, may act as a tumor suppressor for primary prostate tumorigenesis [172,173]. In breast and pancreatic endocrine tumors, HES1 is also downregulated [174,175]. However, in some tumors, HES1 is upregulated during tumorigenesis, such as osteosarcomas [167,176]. As the tumorigenesis is further underway, gene expression in cancer cells is deregulated and seems to be very complex and multifaceted in nature. HES1 may thus act as a tumor suppressor in one context and as an oncogene in another depending on the tumor type and the stage of cancer progression.
As outlined earlier, while a rationale can now be developed for the treatment of several human tumors with Notch inhibitors, and the availability of γ-secretase inhibitors makes this feasible [177], suppression of the Notch and γ-secretase pathways may have unintended effects. Current γ-secretase inhibitors turn off all Notch receptors, which have significant acute toxicities [178], and their long-term effects are yet to be discovered.
Conclusion: integrative cancer therapies
The previously described analyses argue that many oncogenes and tumor suppressors elicit context-dependent effects (sometimes opposite to their definitions) in carcinogenesis. It is conceivable that the signaling pathways that they are responsible for initiating also display complex regulation. To improve the success of anticancer therapy, we propose to develop and implement an integrative treatment strategy (integrative cancer therapy [ICT]). ICT emphasizes the need to not only categorize tumor signatures but also incorporate the stage dimension as well as the personalized care strategy. There is increasing emphasis on personalized medicine to categorize the specific gene signatures in given tumors to guide anticancer treatment. It is also important to develop better cancer biomarkers to assist in categorizing the specific stages of given cancer types to guide stage-dependent anticancer treatment. Additionally, emerging research reveals that there are underinvestigated relationships between stress, low immunity, psychosocial intervention and cancer [179–183]. Some epidemiological evidence suggests a reduced incidence of many common types of nonsmoking-related cancers in individuals with Parkinson's disease [184–186], and an inverse relationship between Alzheimer's disease and cancer [187]. Clearly, further investigation into how the alterations of system physiology affect cancer incidence and treatment outcomes can contribute significantly to the rational strategy of intervention.
Future perspective
The term ‘targeted therapy’ refers to anticancer drugs designed to interfere with a specific molecular target (whether at the RNA or protein level) in a biological pathway that is believed to play a critical role in tumorigenesis or progression. The identification of appropriate targets is based on a detailed understanding of the molecular mechanisms underlying cancer progression [188–191]. This approach is in contrast to the more empirical approach used to develop cytotoxic chemotherapeutics – the mainstay of cancer drug development in past decades. Recent development of target-specific therapy, such as Gleevec [25–27] and the humanized anti-HER2 monoclonal antibody trastuzumab, has the potential to revolutionize cancer therapy by individualizing treatment [192]. With the better understanding of the dichotomy of oncogenic and tumor-suppressive pathways, they can be applied to best exert their anticancer efficacy.
Cytotoxic therapy with protective adjuvants could be effective for symptom relief in advanced cancer [193], although many of these compounds lack tumor selectivity [194]. Cytotoxic therapy, or chemotherapy as it is commonly known, has been the cornerstone of cancer treatment for many years and involves the use of drugs that are toxic to all cells, including normal, non-cancer cells. While relatively effective, this leads to the unpleasant side effects commonly associated with cytotoxic treatment, such as vomiting, nausea, alopecia and fatigue.
Currently as well as for the future, the chemotherapeutic emphasis of oncology is still towards the use of adjuvant drugs to improve patient survival by delaying micrometastasis [195]. This treatment philosophy can be extended by expanding the previously discussed knowledge of the oncogenic pathways underlying tumor progression, as well as by the development of molecularly targeted drugs that modulate these pathways. More and more combined cytotoxic and targeted anticancer drugs have been incorporated into practice and have improved both survival outcomes and quality of life [196,197]. Monoclonal antibodies, which have the capability to bind to specific markers on the surface of tumor cells, offer an attractive therapy that is tumor specific and thus less toxic [194]. For example, pancreatic cancer is a common, highly lethal disease with a rising incidence. Mucin 1 (MUC1) is a tumor-associated antigen that is overexpressed in pancreatic adenocarcinoma. Most recent studies showed that active immunotherapy that targets MUC1 could have great treatment value and may be used as a new adjuvant strategy against pancreatic cancer [198].
There are many time-proven therapies that have not reached their full potential owing to toxicity; there are also many underexplored areas that may prove promising in cooperation with anticancer therapy. Many existing target-specific therapies can manifest greater potential in limiting cancer if they are applied to the most appropriate stages in carcinogenesis. Future oncology will rely on an integrative approach of applying target-specific cancer therapies and cytotoxic therapies with protective adjuvant, as well as reliable cancer biomarkers for treatment guidance.
Executive summary.
Goal of cancer therapy: our progress in detecting & treating cancers
-
Early detection and effective therapies:
Breast cancer and prostate cancer, two areas in which early detection is possible;
Pioneering chemoagent platinum compounds offered earlier hopes for cancer cure;
Signaling-targeted anticancer drug Gleevec® (Novartis, Basel, Switzerland) renewed hopes for target-based cancer therapies.
Challenges on most fronts of anticancer efforts
-
Lack of reliable cancer biomarkers:
We lack reliable biomarkers for some cancers to reduce false-detection rates;
Many cancers are detected at a later stage when there are fewer treatment options.
-
Complex roles of pro- and anti-oncogenic pathways:
Rb–E2F axis showed both pro-proliferation and pro-apoptosis features;
TGF-β1 signaling suppresses tumorigenesis at the earlier stage but promotes tumor progression at the later stage;
Cell–cell junction proteins from the p120ctn/δ-catenin family can have both oncogenic and tumor-suppressive functions depending on the status of E-cadherin;
PI3K signaling activation can be pro-oncogenic as well as anti-oncogenic;
Inhibition of the Raf–MEK–ERK pathway at different tumor stages or in different cancer types produces different effects;
Hundreds of clinical trials are exploring restoration of wild-type, tumor-suppressive p53 functions or inhibition of mutant, oncogenic p53 functions;
Complex Notch signaling pathway with both oncogenic and tumor-suppressive outcomes can now be explored with the availability of γ-secretase inhibitors.
Conclusion: integrative cancer therapies
It is important to recognize the roles of personalized medicine (e.g., categorize the specific gene signatures in given tumors to guide anticancer treatment).
It is also important to develop better cancer biomarkers to assist in categorizing the specific stages of given cancer types to guide stage-dependent anticancer treatment to circumvent the unintended effects of targeting given anti- and pro-oncogenic pathways.
It is important to recognize the underinvestigated relationships between stress, low immunity, psychosocial intervention, Alzheimer's disease, Parkinson's disease and certain cancers. Further investigation into how the alterations of system physiology affect cancer incidence and treatment outcomes can contribute significantly to the rationale of intervention strategy.
Future perspective
Target-specific cancer therapy, such as monoclonal antibody-based drugs, showed long-term benefits to minimize toxicity towards noncancer cells.
Cytotoxic therapy with protective adjuvant strategy has the potential to expand and extend the application of existing US FDA-approved drugs. Both strategies in conjunction with reliable, stage-specific biomarkers will provide greater opportunities for cancer management.
Strategies of applying integrative cancer therapy will have great potential in succeeding in the war against cancer.
Acknowledgments
The authors wish to thank the reviewers for their constructive comments and suggestions. We also apologize for the large amount of outstanding research on cancer cell signaling that has not been included here owing to space limitation.
Footnotes
Financial & competing interests disclosure:
This study is supported in part by grants from NIH CA111891 (Qun Lu), AG026630 (Qun Lu), ES016888 (Yan-Hua Chen) and Department of Defense PC040569 (Qun Lu). The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.
No writing assistance was utilized in the production of this manuscript.
Contributor Information
Jiao Zhang, Department of Anatomy & Cell Biology, The Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA.
Yan-Hua Chen, Department of Anatomy & Cell Biology, Leo Jenkins Cancer Center, The Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA.
Qun Lu, Associate Professor, Department of Anatomy & Cell Biology, Leo Jenkins Cancer Center, The Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA, Tel.: +1 252 744 2844, Fax: +1 252 744 2850, luq@ecu.edu.
Bibliography
Papers of special note have been highlighted as:
▪ of interest
▪▪ of considerable interest
- 1.Donner L, Turek LP, Ruscetti SK, Fedele LA, Sherr CJ. Transformation-defective mutants of feline sarcoma virus which express a product of the viral Src gene. J Virol. 1980;35(1):129–140. doi: 10.1128/jvi.35.1.129-140.1980. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Semczuk A, Postawski K, Przadka D, et al. K-ras gene point mutations and p21ras immunostaining in human ovarian tumors. Eur J Gynaecol Oncol. 2004;25(4):484–488. [PubMed] [Google Scholar]
- 3.Ni Z, Liu Y, Keshava N, et al. Analysis of K-ras and p53 mutations in mesotheliomas from humans and rats exposed to asbestos. Mutat Res. 2000;468(1):87–92. doi: 10.1016/s1383-5718(00)00043-7. [DOI] [PubMed] [Google Scholar]
- 4.Karakosta A, Golias C, Charalabopoulos A, et al. Genetic models of human cancer as a multistep process. Paradigm models of colorectal cancer, breast cancer, and chronic myelogenous and acute lymphoblastic leukaemia. J Exp Clin Cancer Res. 2005;24(4):505–514. [PubMed] [Google Scholar]
- 5.Leibovitch SA, Leibovitch MP, Guillier M, Hillion J, Harel J. Differential expression of protooncogenes related to transformation and cancer progression in rat myoblasts. Cancer Res. 1986;46(8):4097–4103. [PubMed] [Google Scholar]
- 6.Radiloff DR, Rinella ES, Threadgill DW. Modeling cancer patient populations in mice: complex genetic and environmental factors. Drug Discov Today Dis Models. 2008;4(2):83–88. doi: 10.1016/j.ddmod.2007.06.004. [DOI] [PMC free article] [PubMed] [Google Scholar]; ▪▪ Emphasizes the point that more sophisticated models treating cancer as a complex disease present within heterogenous patient populations will be needed.
- 7.Actis AB, Eynard AR. Influence of environmental and nutritional factors on salivary gland tumorigenesis with a special reference to dietary lipids. Eur J Clin Nutr. 2000;54(11):805–810. doi: 10.1038/sj.ejcn.1601077. [DOI] [PubMed] [Google Scholar]
- 8.Ehrhardt A, Bartels T, Klocke R, Paul D, Halter R. Increased susceptibility to the tobacco carcinogen 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone in transgenic mice overexpressing c-myc and epidermal growth factor in alveolar type II cells. J Cancer Res Clin Oncol. 2003;129(2):71–75. doi: 10.1007/s00432-002-0400-z. [DOI] [PubMed] [Google Scholar]
- 9.Gatenby RA, Vincent TL. An evolutionary model of carcinogenesis. Cancer Res. 2003;63(19):6212–6220. [PubMed] [Google Scholar]
- 10.Toren A, Amariglio N, Rechavi G. Curable and non-curable malignancies: lessons from paediatric cancer. Med Oncol. 1996;13(1):15–21. doi: 10.1007/BF02988837. [DOI] [PubMed] [Google Scholar]
- 11.Rich JN, Eyler CE. Cancer stem cells in brain tumor biology. Cold Spring Harb Symp Quant Biol. 2008;73:411–420. doi: 10.1101/sqb.2008.73.060. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Padron Velazquez JL. Stem cell fusion as an ultimate line of defense against xenobiotics. Med Hypotheses. 2006;67(2):383–387. doi: 10.1016/j.mehy.2006.01.031. [DOI] [PubMed] [Google Scholar]
- 13.Coldman A, Phillips N, Warren L, Kan L. Breast cancer mortality after screening mammography in British Columbia women. Int J Cancer. 2007;120(5):1076–1080. doi: 10.1002/ijc.22249. [DOI] [PubMed] [Google Scholar]
- 14.Doi K, Giger ML, Nishikawa RM, Schmidt RA. Computer-aided diagnosis of breast cancer on mammograms. Breast Cancer. 1997;4(4):228–233. doi: 10.1007/BF02966511. [DOI] [PubMed] [Google Scholar]
- 15.Ilic D, O'Connor D, Green S, Wilt T. Screening for prostate cancer. Cochrane Database Syst Rev. 2006;3 doi: 10.1002/14651858.CD004720.pub2. CD004720. [DOI] [PubMed] [Google Scholar]
- 16.Twombly R. Preventive Services Task Force recommends against PSA screening after age 75. J Natl Cancer Inst. 2008;100(22):1571–1573. doi: 10.1093/jnci/djn422. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Lipp HP, Hartmann JT. Platinum compounds: metabolism, toxicity and supportive strategies. Praxis (Bern 1994) 2005;94(6):187–198. doi: 10.1024/0369-8394.94.6.187. [DOI] [PubMed] [Google Scholar]
- 18.Gouyette A, Deniel A, Pico JL, et al. Clinical pharmacology of high-dose etoposide associated with cisplatin. Pharmacokinetic and metabolic studies. Eur J Cancer Clin Oncol. 1987;23(11):1627–1632. doi: 10.1016/0277-5379(87)90441-x. [DOI] [PubMed] [Google Scholar]
- 19.Pera MF, Jr, Sessford D, Roberts JJ. Toxicity of cisplatin and hydroxymalonatodiammine platinum (II) towards mouse bone marrow and B16 melanoma in relation to DNA binding in vivo. Biochem Pharmacol. 1982;31(13):2273–2278. doi: 10.1016/0006-2952(82)90114-9. [DOI] [PubMed] [Google Scholar]
- 20.Zinner RG, Herbst RS. Pemetrexed in the treatment of advanced non-small-cell lung cancer: a review of the clinical data. Clin Lung Cancer. 2004;5(Suppl. 2):S67–S74. doi: 10.3816/clc.2004.s.006. [DOI] [PubMed] [Google Scholar]
- 21.Cohen MH, Williams GA, Sridhara R, et al. United States Food and Drug Administration Drug Approval summary: gefitinib (ZD1839; Iressa) tablets. Clin Cancer Res. 2004;10(4):1212–1218. doi: 10.1158/1078-0432.ccr-03-0564. [DOI] [PubMed] [Google Scholar]
- 22.Huddart RA, Birtle AJ. Recent advances in the treatment of testicular cancer. Expert Rev Anticancer Ther. 2005;5(1):123–138. doi: 10.1586/14737140.5.1.123. [DOI] [PubMed] [Google Scholar]
- 23.Raghavan D. Testicular cancer: maintaining the high cure rate. Oncology. 2003;17(2):218–228. discussion 228–229, 234–234, passim. [PubMed] [Google Scholar]
- 24.Arora A, Scholar EM. Role of tyrosine kinase inhibitors in cancer therapy. J Pharmacol Exp Ther. 2005;315(3):971–979. doi: 10.1124/jpet.105.084145. [DOI] [PubMed] [Google Scholar]
- 25.Weisberg E, Manley PW, Cowan-Jacob SW, Hochhaus A, Griffin JD. Second generation inhibitors of BCR-ABL for the treatment of imatinib-resistant chronic myeloid leukaemia. Nat Rev Cancer. 2007;7(5):345–356. doi: 10.1038/nrc2126. [DOI] [PubMed] [Google Scholar]
- 26.Hahn EA, Glendenning GA. Quality of life on imatinib. Semin Hematol. 2003;40(2 Suppl. 2):31–36. doi: 10.1053/shem.2003.50039. [DOI] [PubMed] [Google Scholar]
- 27.Pytel D, Sliwinski T, Poplawski T, Ferriola D, Majsterek I. Tyrosine kinase blockers: new hope for successful cancer therapy. Anticancer Agents Med Chem. 2009;9(1):66–76. doi: 10.2174/187152009787047752. [DOI] [PubMed] [Google Scholar]
- 28.Chang BS, Yang T, Cibas ES, Fletcher JA. An in vitro cytologic assay for evaluation of the KIT signaling pathway in gastrointestinal stromal tumors. Mod Pathol. 2007;20(5):579–583. doi: 10.1038/modpathol.3800779. [DOI] [PubMed] [Google Scholar]
- 29.Mauro MJ, Druker BJ. STI571: targeting BCR-ABL as therapy for CML. Oncologist. 2001;6(3):233–238. doi: 10.1634/theoncologist.6-3-233. [DOI] [PubMed] [Google Scholar]; ▪▪ One of the original discussions of the therapeutic agent STI571 as a successfully developed, potent and selective inhibitor for Abl tyrosine kinases, including Bcr-Abl, as well c-Kit and the PDGF receptor tyrosine kinases.
- 30.Druker B. Signal transduction inhibition: results from Phase I clinical trials in chronic myeloid leukemia. Semin Hematol. 2001;38(3 Suppl. 8):9–14. doi: 10.1016/s0037-1963(01)90112-x. [DOI] [PubMed] [Google Scholar]
- 31.Booth RA. Minimally invasive biomarkers for detection and staging of colorectal cancer. Cancer Lett. 2007;249(1):87–96. doi: 10.1016/j.canlet.2006.12.021. [DOI] [PubMed] [Google Scholar]
- 32.Watanabe N. Oncogene and tumor suppressor gene. Rinsho Byori Suppl. 2002;123:131–136. [PubMed] [Google Scholar]
- 33.Nagai H, Harada H, Emi M. Oncogene and tumor suppressor gene. Nippon Rinsho. 2000;58(Suppl):25–29. [PubMed] [Google Scholar]
- 34.Zhang WW. Antisense oncogene and tumor suppressor gene therapy of cancer. J Mol Med. 1996;74(4):191–204. doi: 10.1007/BF00204749. [DOI] [PubMed] [Google Scholar]
- 35.Fearon ER, Dang CV. Cancer genetics: tumor suppressor meets oncogene. Curr Biol. 1999;9(2):R62–R65. doi: 10.1016/s0960-9822(99)80012-8. [DOI] [PubMed] [Google Scholar]
- 36.Sung J, Turner J, McCarthy S, et al. Oncogene regulation of tumor suppressor genes in tumorigenesis. Carcinogenesis. 2005;26(2):487–494. doi: 10.1093/carcin/bgh318. [DOI] [PubMed] [Google Scholar]; ▪ Demonstrates an epigenetic link between oncogenes and tumor-suppressive genes in tumorigenesis using a high-throughput model.
- 37.Menke AL, van der Eb AJ, Jochemsen AG. The Wilms' tumor 1 gene: oncogene or tumor suppressor gene? Int Rev Cytol. 1998;181:151–212. doi: 10.1016/s0074-7696(08)60418-0. [DOI] [PubMed] [Google Scholar]
- 38.Stiewe T, Putzer BM. Role of p73 in malignancy: tumor suppressor or oncogene? Cell Death Differ. 2002;9(3):237–245. doi: 10.1038/sj.cdd.4400995. [DOI] [PubMed] [Google Scholar]
- 39.Frank JL, Bur ME, Garb JL, et al. p53 tumor suppressor oncogene expression in squamous cell carcinoma of the hypopharynx. Cancer. 1994;73(1):181–186. doi: 10.1002/1097-0142(19940101)73:1<181::aid-cncr2820730131>3.0.co;2-3. [DOI] [PubMed] [Google Scholar]
- 40.Kossatz U, Malek NP. p27: tumor suppressor and oncogene …? Cell Res. 2007;17(10):832–833. doi: 10.1038/cr.2007.86. [DOI] [PubMed] [Google Scholar]; ▪ Research highlight discussing the tumor-promoting effects versus tumor-suppressor functions of p27.
- 41.Damm K. ErbA: tumor suppressor turned oncogene? FASEB J. 1993;7(10):904–909. doi: 10.1096/fasebj.7.10.8102105. [DOI] [PubMed] [Google Scholar]
- 42.Johnson DG. The paradox of E2F1: oncogene and tumor suppressor gene. Mol Carcinog. 2000;27(3):151–157. doi: 10.1002/(sici)1098-2744(200003)27:3<151::aid-mc1>3.0.co;2-c. [DOI] [PubMed] [Google Scholar]
- 43.Lu Q, Zhang J, Chen YH. Prostate cancer cell growth and death: complex roles of pro- and anti-oncogenic protein signaling. In: Meridith AT, editor. Handbook of Prostate Cancer Cell Research: Growth, Signalling and Survival. Nova Science Pub Inc.; NY, USA: 2009. pp. 431–447. [Google Scholar]; ▪▪ Discusses the dichotomy of oncogenes and tumor suppressors in prostate cancer cell growth and death.
- 44.Borges HL, Bird J, Wasson K, et al. Tumor promotion by caspase-resistant retinoblastoma protein. Proc Natl Acad Sci USA. 2005;102(43):15587–15592. doi: 10.1073/pnas.0503925102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Wang D, Russell JL, Johnson DG. E2F4 and E2F1 have similar proliferative properties but different apoptotic and oncogenic properties in vivo. Mol Cell Biol. 2000;20(10):3417–3424. doi: 10.1128/mcb.20.10.3417-3424.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Davis JN, Wojno KJ, Daignault S, et al. Elevated E2F1 inhibits transcription of the androgen receptor in metastatic hormone-resistant prostate cancer. Cancer Res. 2006;66(24):11897–11906. doi: 10.1158/0008-5472.CAN-06-2497. [DOI] [PubMed] [Google Scholar]
- 47.Kaseb AO, Chinnakannu K, Chen D, et al. Androgen receptor and E2F-1 targeted thymoquinone therapy for hormone-refractory prostate cancer. Cancer Res. 2007;67(16):7782–7788. doi: 10.1158/0008-5472.CAN-07-1483. [DOI] [PubMed] [Google Scholar]
- 48.Rogoff HA, Kowalik TF. Life, death and E2F: linking proliferation control and DNA damage signaling via E2F1. Cell Cycle. 2004;3(7):845–846. doi: 10.4161/cc.3.7.976. [DOI] [PubMed] [Google Scholar]
- 49.Nakajima Y. Interaction of E2F/Rb family members with factor binding to co-repressor element on B-myb and E2F1 promoters. Kokubyo Gakkai Zasshi. 1998;65(2):172–188. doi: 10.5357/koubyou.65.172. [DOI] [PubMed] [Google Scholar]
- 50.Rogoff HA, Pickering MT, Debatis ME, Jones S, Kowalik TF. E2F1 induces phosphorylation of p53 that is coincident with p53 accumulation and apoptosis. Mol Cell Biol. 2002;22(15):5308–5318. doi: 10.1128/MCB.22.15.5308-5318.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.de Caestecker MP, Piek E, Roberts AB. Role of transforming growth factor-β signaling in cancer. J Natl Cancer Inst. 2000;92(17):1388–1402. doi: 10.1093/jnci/92.17.1388. [DOI] [PubMed] [Google Scholar]
- 52.Schnaper HW, Hayashida T, Hubchak SC, Poncelet AC. TGF-β signal transduction and mesangial cell fibrogenesis. Am J Physiol Renal Physiol. 2003;284(2):F243–F2452. doi: 10.1152/ajprenal.00300.2002. [DOI] [PubMed] [Google Scholar]
- 53.Glick AB. TGF-β1, back to the future: revisiting its role as a transforming growth factor. Cancer Biol Ther. 2004;3(3):276–283. doi: 10.4161/cbt.3.3.849. [DOI] [PubMed] [Google Scholar]; ▪▪ Excellent discussion pointing out the stage-specific duality of function as the emerging paradigm for the role of TGF-β1 in cancer.
- 54.Oft M, Akhurst RJ, Balmain A. Metastasis is driven by sequential elevation of H-ras and Smad2 levels. Nat Cell Biol. 2002;4(7):487–494. doi: 10.1038/ncb807. [DOI] [PubMed] [Google Scholar]
- 55.Shi Y, Massague J. Mechanisms of TGF-β signaling from cell membrane to the nucleus. Cell. 2003;113(6):685–700. doi: 10.1016/s0092-8674(03)00432-x. [DOI] [PubMed] [Google Scholar]
- 56.Amendt C, Schirmacher P, Weber H, Blessing M. Expression of a dominant negative type II TGF-β receptor in mouse skin results in an increase in carcinoma incidence and an acceleration of carcinoma development. Oncogene. 1998;17(1):25–34. doi: 10.1038/sj.onc.1202161. [DOI] [PubMed] [Google Scholar]
- 57.Blessing M, Nanney LB, King LE, Hogan BL. Chemical skin carcinogenesis is prevented in mice by the induced expression of a TGF-β related transgene. Teratog Carcinog Mutagen. 1995;15(1):11–21. doi: 10.1002/tcm.1770150103. [DOI] [PubMed] [Google Scholar]
- 58.Glick AB, Lee MM, Darwiche N, et al. Targeted deletion of the TGF-β1 gene causes rapid progression to squamous cell carcinoma. Genes Dev. 1994;8(20):2429–2440. doi: 10.1101/gad.8.20.2429. [DOI] [PubMed] [Google Scholar]
- 59.Wang XJ, Greenhalgh DA, Bickenbach JR, et al. Expression of a dominant-negative type II transforming growth factor β (TGF-β) receptor in the epidermis of transgenic mice blocks TGF-β-mediated growth inhibition. Proc Natl Acad Sci USA. 1997;94(6):2386–2391. doi: 10.1073/pnas.94.6.2386. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Cui W, Fowlis DJ, Bryson S, et al. TGF-β1 inhibits the formation of benign skin tumors, but enhances progression to invasive spindle carcinomas in transgenic mice. Cell. 1996;86(4):531–542. doi: 10.1016/s0092-8674(00)80127-0. [DOI] [PubMed] [Google Scholar]
- 61.Weeks BH, He W, Olson KL, Wang XJ. Inducible expression of transforming growth factor β1 in papillomas causes rapid metastasis. Cancer Res. 2001;61(20):7435–7443. [PubMed] [Google Scholar]
- 62.He W, Cao T, Smith DA, Myers TE, Wang XJ. Smads mediate signaling of the TGF-β superfamily in normal keratinocytes but are lost during skin chemical carcinogenesis. Oncogene. 2001;20(4):471–483. doi: 10.1038/sj.onc.1204117. [DOI] [PubMed] [Google Scholar]
- 63.Vijayachandra K, Lee J, Glick AB. Smad3 regulates senescence and malignant conversion in a mouse multistage skin carcinogenesis model. Cancer Res. 2003;63(13):3447–3452. [PubMed] [Google Scholar]
- 64.Li AG, Lu SL, Zhang MX, Deng C, Wang XJ. Smad3 knockout mice exhibit a resistance to skin chemical carcinogenesis. Cancer Res. 2004;64(21):7836–7845. doi: 10.1158/0008-5472.CAN-04-1331. [DOI] [PubMed] [Google Scholar]
- 65.Peifer M, Berg S, Reynolds AB. A repeating amino acid motif shared by proteins with diverse cellular roles. Cell. 1994;76(5):789–791. doi: 10.1016/0092-8674(94)90353-0. [DOI] [PubMed] [Google Scholar]
- 66.Lu Q, Paredes M, Medina M, et al. δ-catenin, an adhesive junction-associated protein which promotes cell scattering. J Cell Biol. 1999;144(3):519–532. doi: 10.1083/jcb.144.3.519. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Paffenholz R, Franke WW. Identification and localization of a neurally expressed member of the plakoglobin/armadillo multigene family. Differentiation. 1997;61(5):293–304. doi: 10.1046/j.1432-0436.1997.6150293.x. [DOI] [PubMed] [Google Scholar]
- 68.Zhou J, Liyanage U, Medina M, et al. Presenilin 1 interaction in the brain with a novel member of the armadillo family. Neuroreport. 1997;8(6):1489–1494. doi: 10.1097/00001756-199704140-00033. [DOI] [PubMed] [Google Scholar]
- 69.Thoreson MA, Reynolds AB. Altered expression of the catenin p120 in human cancer: implications for tumor progression. Differentiation. 2002;70:9–10. 583–589. doi: 10.1046/j.1432-0436.2002.700911.x. [DOI] [PubMed] [Google Scholar]; ▪ One of the earlier reviews discussing the potential roles of p120 catenin as a tumor suppressor or metastasis promoter.
- 70.Anastasiadis PZ, Moon SY, Thoreson MA, et al. Inhibition of RhoA by p120 catenin. Nat Cell Biol. 2000;2(9):637–644. doi: 10.1038/35023588. [DOI] [PubMed] [Google Scholar]
- 71.Noren NK, Liu BP, Burridge K, Kreft B. p120 catenin regulates the actin cytoskeleton via Rho family GTPases. J Cell Biol. 2000;150(3):567–580. doi: 10.1083/jcb.150.3.567. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Ireton RC, Davis MA, van Hengel J, et al. A novel role for p120 catenin in E-cadherin function. J Cell Biol. 2002;159(3):465–476. doi: 10.1083/jcb.200205115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Lu Q, Dobbs LJ, Gregory CW, et al. Increased expression of δ-catenin/neural plakophilin-related armadillo protein is associated with the down-regulation and redistribution of E-cadherin and p120ctn in human prostate cancer. Hum Pathol. 2005;36(10):1037–1048. doi: 10.1016/j.humpath.2005.07.012. [DOI] [PubMed] [Google Scholar]
- 74.Lu Q, Mukhopadhyay NK, Griffin JD, et al. Brain armadillo protein δ-catenin interacts with Abl tyrosine kinase and modulates cellular morphogenesis in response to growth factors. J Neurosci Res. 2002;67(5):618–624. doi: 10.1002/jnr.10151. [DOI] [PubMed] [Google Scholar]
- 75.Kim K, Oh M, Ki H, et al. Identification of E2F1 as a positive transcriptional regulator for δ-catenin. Biochem Biophys Res Commun. 2008;369(2):414–420. doi: 10.1016/j.bbrc.2008.02.069. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Zeng Y, Abdallah A, Lu JP, et al. δ-catenin promotes prostate cancer cell growth and progression by altering cell cycle and survival gene profiles. Mol Cancer. 2009;8:19. doi: 10.1186/1476-4598-8-19. [DOI] [PMC free article] [PubMed] [Google Scholar]; ▪ Reports a primarily neural-specific protein that promotes prostate cancer cell expansion.
- 77.Debusk LM, Boelte K, Min Y, Lin PC. Heterozygous deficiency of δ-catenin impairs pathological angiogenesis. J Exp Med. 2010;207(1):77–84. doi: 10.1084/jem.20091097. [DOI] [PMC free article] [PubMed] [Google Scholar]; ▪ Interesting article showing that the paucity of δ-catenin disrupted only vessel growth associated with tumors and healing wounds but not normal vessels.
- 78.Kim K, Sirota A, Chen Yh YH, et al. Dendrite-like process formation and cytoskeletal remodeling regulated by δ-catenin expression. Exp Cell Res. 2002;275(2):171–184. doi: 10.1006/excr.2002.5503. [DOI] [PubMed] [Google Scholar]
- 79.Westbrook TF, Martin ES, Schlabach MR, et al. A genetic screen for candidate tumor suppressors identifies REST. Cell. 2005;121(6):837–848. doi: 10.1016/j.cell.2005.03.033. [DOI] [PubMed] [Google Scholar]; ▪ Interesting article identifying δ-catenin as a potential tumor suppressor for mammary epithelial cells.
- 80.Bader AG, Kang S, Zhao L, Vogt PK. Oncogenic PI3K deregulates transcription and translation. Nat Rev Cancer. 2005;5(12):921–929. doi: 10.1038/nrc1753. [DOI] [PubMed] [Google Scholar]
- 81.Engelman JA, Luo J, Cantley LC. The evolution of phosphatidylinositol 3-kinases as regulators of growth and metabolism. Nat Rev Genet. 2006;7(8):606–619. doi: 10.1038/nrg1879. [DOI] [PubMed] [Google Scholar]
- 82.Brugge J, Hung MC, Mills GB. A new mutational AKTivation in the PI3K pathway. Cancer Cell. 2007;12(2):104–107. doi: 10.1016/j.ccr.2007.07.014. [DOI] [PubMed] [Google Scholar]
- 83.Gymnopoulos M, Elsliger MA, Vogt PK. Rare cancer-specific mutations in PIK3CA show gain of function. Proc Natl Acad Sci USA. 2007;104(13):5569–5574. doi: 10.1073/pnas.0701005104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Ikenoue T, Kanai F, Hikiba Y, et al. Functional analysis of PIK3CA gene mutations in human colorectal cancer. Cancer Res. 2005;65(11):4562–4567. doi: 10.1158/0008-5472.CAN-04-4114. [DOI] [PubMed] [Google Scholar]
- 85.Kang S, Bader AG, Vogt PK. Phosphatidylinositol 3-kinase mutations identified in human cancer are oncogenic. Proc Natl Acad Sci USA. 2005;102(3):802–807. doi: 10.1073/pnas.0408864102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Bader AG, Kang S, Vogt PK. Cancer-specific mutations in PIK3CA are oncogenic in vivo. Proc Natl Acad Sci USA. 2006;103(5):1475–1479. doi: 10.1073/pnas.0510857103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Isakoff SJ, Engelman JA, Irie HY, et al. Breast cancer-associated PIK3CA mutations are oncogenic in mammary epithelial cells. Cancer Res. 2005;65(23):10992–11000. doi: 10.1158/0008-5472.CAN-05-2612. [DOI] [PubMed] [Google Scholar]
- 88.Zhao JJ, Liu Z, Wang L, et al. The oncogenic properties of mutant p110α and p110β phosphatidylinositol 3-kinases in human mammary epithelial cells. Proc Natl Acad Sci USA. 2005;102(51):18443–18448. doi: 10.1073/pnas.0508988102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Samuels Y, Diaz LA, Jr, Schmidt-Kittler O, et al. Mutant PIK3CA promotes cell growth and invasion of human cancer cells. Cancer Cell. 2005;7(6):561–573. doi: 10.1016/j.ccr.2005.05.014. [DOI] [PubMed] [Google Scholar]
- 90.Miyauchi H, Minamino T, Tateno K, et al. Akt negatively regulates the in vitro lifespan of human endothelial cells via a p53/p21-dependent pathway. EMBO J. 2004;23(1):212–220. doi: 10.1038/sj.emboj.7600045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Oyama K, Okawa T, Nakagawa H, et al. AKT induces senescence in primary esophageal epithelial cells but is permissive for differentiation as revealed in organotypic culture. Oncogene. 2007;26(16):2353–2364. doi: 10.1038/sj.onc.1210025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Blanco-Aparicio C, Pérez-Gallego L, Pequeno B, Leal JF, Renner O, Carnero A. Mice expressing myrAKT1 in the mammary gland develop carcinogen-induced ER-positive mammary tumors that mimic human breast cancer. Carcinogenesis. 2007;28(3):584–594. doi: 10.1093/carcin/bgl190. [DOI] [PubMed] [Google Scholar]
- 93.Hutchinson JN, Jin J, Cardiff RD, Woodgett JR, Muller WJ. Activation of Akt-1 (PKB-α) can accelerate ErbB-2-mediated mammary tumorigenesis but suppresses tumor invasion. Cancer Res. 2004;64(9):3171–3178. doi: 10.1158/0008-5472.can-03-3465. [DOI] [PubMed] [Google Scholar]
- 94.Renner O, Blanco-Aparicio C, Grassow M, et al. Activation of phosphatidylinositol 3-kinase by membrane localization of p110α predisposes mammary glands to neoplastic transformation. Cancer Res. 2008;68(23):9643–9653. doi: 10.1158/0008-5472.CAN-08-1539. [DOI] [PubMed] [Google Scholar]
- 95.Buttitta F, Felicioni L, Barassi F, et al. PIK3CA mutation and histological type in breast carcinoma: high frequency of mutations in lobular carcinoma. J Pathol. 2006;208(3):350–355. doi: 10.1002/path.1908. [DOI] [PubMed] [Google Scholar]
- 96.Campbell IG, Russell SE, Choong DY, et al. Mutation of the PIK3CA gene in ovarian and breast cancer. Cancer Res. 2004;64(21):7678–7681. doi: 10.1158/0008-5472.CAN-04-2933. [DOI] [PubMed] [Google Scholar]
- 97.Li SY, Rong M, Grieu F, Iacopetta B. PIK3CA mutations in breast cancer are associated with poor outcome. Breast Cancer Res Treat. 2006;96(1):91–95. doi: 10.1007/s10549-005-9048-0. [DOI] [PubMed] [Google Scholar]
- 98.Maruyama N, Miyoshi Y, Taguchi T, et al. Clinicopathologic analysis of breast cancers with PIK3CA mutations in Japanese women. Clin Cancer Res. 2007;13(2 Pt 1):408–414. doi: 10.1158/1078-0432.CCR-06-0267. [DOI] [PubMed] [Google Scholar]
- 99.Saal LH, Holm K, Maurer M, et al. PIK3CA mutations correlate with hormone receptors, node metastasis, and ERBB2, and are mutually exclusive with PTEN loss in human breast carcinoma. Cancer Res. 2005;65(7):2554–2559. doi: 10.1158/0008-5472-CAN-04-3913. [DOI] [PubMed] [Google Scholar]
- 100.Samuels Y, Wang Z, Bardelli A, et al. High frequency of mutations of the PIK3CA gene in human cancers. Science. 2004;304(5670):554. doi: 10.1126/science.1096502. [DOI] [PubMed] [Google Scholar]
- 101.Kalinsky K, Jacks LM, Heguy A, et al. PIK3CA mutation associates with improved outcome in breast cancer. Clin Cancer Res. 2009;15(16):5049–5059. doi: 10.1158/1078-0432.CCR-09-0632. [DOI] [PubMed] [Google Scholar]; ▪ One of the recent articles describing PIK3CA mutations as being associated with better prognosis in some breast cancer cases.
- 102.Di Cosimo S, Baselga J. Phosphoinositide 3-kinase mutations in breast cancer: a “good” activating mutation? Clin Cancer Res. 2009;15(16):5017–5019. doi: 10.1158/1078-0432.CCR-09-1173. [DOI] [PubMed] [Google Scholar]
- 103.Pritchard JR, Cosgrove BD, Hemann MT, Griffith LG, Wands JR, Lauffenburger DA. Three-kinase inhibitor combination recreates multipathway effects of a geldanamycin analogue on hepatocellular carcinoma cell death. Mol Cancer Ther. 2009;8(8):2183–2192. doi: 10.1158/1535-7163.MCT-08-1203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Basso AD, Solit DB, Munster PN, Rosen N. Ansamycin antibiotics inhibit Akt activation and cyclin D expression in breast cancer cells that overexpress HER2. Oncogene. 2002;21(8):1159–1166. doi: 10.1038/sj.onc.1205184. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Neshat MS, Mellinghoff IK, Tran C, et al. Enhanced sensitivity of PTEN-deficient tumors to inhibition of FRAP/mTOR. Proc Natl Acad Sci USA. 2001;98(18):10314–10319. doi: 10.1073/pnas.171076798. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Gulhati P, Cai Q, Li J, et al. Targeted inhibition of mammalian target of rapamycin signaling inhibits tumorigenesis of colorectal cancer. Clin Cancer Res. 2009;15(23):7207–7216. doi: 10.1158/1078-0432.CCR-09-1249. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Johnson R, Spiegelman B, Hanahan D, Wisdom R. Cellular transformation and malignancy induced by ras require c-jun. Mol Cell Biol. 1996;16(8):4504–4511. doi: 10.1128/mcb.16.8.4504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Hanahan D, Weinberg RA. The hallmarks of cancer. Cell. 2000;100(1):57–70. doi: 10.1016/s0092-8674(00)81683-9. [DOI] [PubMed] [Google Scholar]
- 109.Downward J. Targeting RAS signalling pathways in cancer therapy. Nat Rev Cancer. 2003;3(1):11–22. doi: 10.1038/nrc969. [DOI] [PubMed] [Google Scholar]
- 110.Torii S, Yamamoto T, Tsuchiya Y, Nishida E. ERK MAP kinase in G cell cycle progression and cancer. Cancer Sci. 2006;97(8):697–702. doi: 10.1111/j.1349-7006.2006.00244.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Schaeffer HJ, Weber MJ. Mitogen-activated protein kinases: specific messages from ubiquitous messengers. Mol Cell Biol. 1999;19(4):2435–2444. doi: 10.1128/mcb.19.4.2435. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Chen N, Nomura M, She QB, et al. Suppression of skin tumorigenesis in c-Jun NH(2)-terminal kinase-2-deficient mice. Cancer Res. 2001;61(10):3908–3912. [PubMed] [Google Scholar]
- 113.Kyriakis JM, Avruch J. Mammalian mitogen-activated protein kinase signal transduction pathways activated by stress and inflammation. Physiol Rev. 2001;81(2):807–869. doi: 10.1152/physrev.2001.81.2.807. [DOI] [PubMed] [Google Scholar]
- 114.Krens SF, He S, Spaink HP, Snaar-Jagalska BE. Characterization and expression patterns of the MAPK family in zebrafish. Gene Expr Patterns. 2006;6(8):1019–1026. doi: 10.1016/j.modgep.2006.04.008. [DOI] [PubMed] [Google Scholar]
- 115.Friday BB, Adjei AA. Advances in targeting the Ras/Raf/MEK/Erk mitogen-activated protein kinase cascade with MEK inhibitors for cancer therapy. Clin Cancer Res. 2008;14(2):342–346. doi: 10.1158/1078-0432.CCR-07-4790. [DOI] [PubMed] [Google Scholar]; ▪▪ Summarizes some recent progresses on the development of small-molecule inhibitors of MAPK/MEK, a key intermediary of MAPK signaling, in clinical trials.
- 116.Leicht DT, Balan V, Kaplun A, et al. Raf kinases: function, regulation and role in human cancer. Biochim Biophys Acta. 2007;1773(8):1196–1212. doi: 10.1016/j.bbamcr.2007.05.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Young A, Lyons J, Miller AL, et al. Ras signaling and therapies. Adv Cancer Res. 2009;102:1–17. doi: 10.1016/S0065-230X(09)02001-6. [DOI] [PubMed] [Google Scholar]; ▪▪ Discusses the complexities of Ras signaling and their effects on targeting the Ras pathway in the future.
- 118.Benvenuti S, Sartore-Bianchi A, Di Nicolantonio F, et al. Oncogenic activation of the RAS/RAF signaling pathway impairs the response of metastatic colorectal cancers to anti-epidermal growth factor receptor antibody therapies. Cancer Res. 2007;67(6):2643–2648. doi: 10.1158/0008-5472.CAN-06-4158. [DOI] [PubMed] [Google Scholar]
- 119.Dhillon AS, Hagan S, Rath O, Kolch W. MAP kinase signalling pathways in cancer. Oncogene. 2007;26(22):3279–3290. doi: 10.1038/sj.onc.1210421. [DOI] [PubMed] [Google Scholar]
- 120.Yu C, Krystal G, Varticovksi L, et al. Pharmacologic mitogen-activated protein/extracellular signal-regulated kinase kinase/mitogen-activated protein kinase inhibitors interact synergistically with STI571 to induce apoptosis in Bcr/Abl-expressing human leukemia cells. Cancer Res. 2002;62(1):188–199. [PubMed] [Google Scholar]
- 121.Allen LF, Sebolt-Leopold J, Meyer MB. CI-1040 (PD184352), a targeted signal transduction inhibitor of MEK (MAPKK) Semin Oncol. 2003;30(5 Suppl. 16):105–116. doi: 10.1053/j.seminoncol.2003.08.012. [DOI] [PubMed] [Google Scholar]
- 122.Milella M, Kornblau SM, Estrov Z, et al. Therapeutic targeting of the MEK/MAPK signal transduction module in acute myeloid leukemia. J Clin Invest. 2001;108(6):851–859. doi: 10.1172/JCI12807. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Wong KK. Recent developments in anti-cancer agents targeting the Ras/Raf/MEK/ERK pathway. Recent Pat Anticancer Drug Discov. 2009;4(1):28–35. doi: 10.2174/157489209787002461. [DOI] [PubMed] [Google Scholar]
- 124.Greene LA, Tischler AS. Establishment of a noradrenergic clonal line of rat adrenal pheochromocytoma cells which respond to nerve growth factor. Proc Natl Acad Sci USA. 1976;73(7):2424–2428. doi: 10.1073/pnas.73.7.2424. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Huff K, End D, Guroff G. Nerve growth factor-induced alteration in the response of PC12 pheochromocytoma cells to epidermal growth factor. J Cell Biol. 1981;88(1):189–198. doi: 10.1083/jcb.88.1.189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Marshall CJ. Specificity of receptor tyrosine kinase signaling: transient versus sustained extracellular signal-regulated kinase activation. Cell. 1995;80(2):179–185. doi: 10.1016/0092-8674(95)90401-8. [DOI] [PubMed] [Google Scholar]
- 127.Bodo J, Duraj J, Jakubikova J, Sedlak J. Isothiocyanate E-4IB induces MAPK activation, delayed cell cycle transition and apoptosis. Cell Prolif. 2007;40(3):316–326. doi: 10.1111/j.1365-2184.2007.00437.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.McCubrey JA, Steelman LS, Abrams SL, et al. Roles of the RAF/MEK/ERK and PI3K/PTEN/AKT pathways in malignant transformation and drug resistance. Adv Enzyme Regul. 2006;46:249–279. doi: 10.1016/j.advenzreg.2006.01.004. [DOI] [PubMed] [Google Scholar]; ▪▪ Provides examples of complexities of Raf signaling and the interactions with PI3K/AKT pathway.
- 129.Hoyle PE, Moye PW, Steelman LS, et al. Differential abilities of the Raf family of protein kinases to abrogate cytokine dependency and prevent apoptosis in murine hematopoietic cells by a MEK1-dependent mechanism. Leukemia. 2000;14(4):642–656. doi: 10.1038/sj.leu.2401720. [DOI] [PubMed] [Google Scholar]
- 130.Chang F, McCubrey JA. p21(Cip1) induced by Raf is associated with increased Cdk4 activity in hematopoietic cells. Oncogene. 2001;20(32):4354–4364. doi: 10.1038/sj.onc.1204564. [DOI] [PubMed] [Google Scholar]
- 131.Chang F, Steelman LS, McCubrey JA. Raf-induced cell cycle progression in human TF-1 hematopoietic cells. Cell Cycle. 2002;1(3):220–226. [PubMed] [Google Scholar]
- 132.Sanders MR, Lu H, Walker F, Sorba S, Dainiak N. The Raf-1 protein mediates insulin-like growth factor-induced proliferation of erythroid progenitor cells. Stem Cells. 1998;16(3):200–207. doi: 10.1002/stem.160200. [DOI] [PubMed] [Google Scholar]
- 133.Cioffi CL, Garay M, Johnston JF, et al. Selective inhibition of A-Raf and C-Raf mRNA expression by antisense oligodeoxynucleotides in rat vascular smooth muscle cells: role of A-Raf and C-Raf in serum-induced proliferation. Mol Pharmacol. 1997;51(3):383–389. [PubMed] [Google Scholar]
- 134.Yen A, Williams M, Platko JD, Der C, Hisaka M. Expression of activated RAF accelerates cell differentiation and RB protein down-regulation but not hypophosphorylation. Eur J Cell Biol. 1994;65(1):103–113. [PubMed] [Google Scholar]
- 135.Ravi RK, Weber E, McMahon M, et al. Activated Raf-1 causes growth arrest in human small cell lung cancer cells. J Clin Invest. 1998;101(1):153–159. doi: 10.1172/JCI831. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Lloyd AC, Obermuller F, Staddon S, et al. Cooperating oncogenes converge to regulate cyclin/cdk complexes. Genes Dev. 1997;11(5):663–677. doi: 10.1101/gad.11.5.663. [DOI] [PubMed] [Google Scholar]
- 137.Woods D, Parry D, Cherwinski H, et al. Raf-induced proliferation or cell cycle arrest is determined by the level of Raf activity with arrest mediated by p21Cip1. Mol Cell Biol. 1997;17(9):5598–5611. doi: 10.1128/mcb.17.9.5598. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Shelton JG, Chang F, Lee JT, et al. B-raf and insulin synergistically prevent apoptosis and induce cell cycle progression in hematopoietic cells. Cell Cycle. 2004;3(2):189–196. [PubMed] [Google Scholar]
- 139.Shelton JG, Steelman LS, White ER, McCubrey JA. Synergy between PI3K/Akt and Raf/MEK/ERK pathways in IGF-1R mediated cell cycle progression and prevention of apoptosis in hematopoietic cells. Cell Cycle. 2004;3(3):372–379. [PubMed] [Google Scholar]
- 140.Lee JT, Jr, Steelman LS, McCubrey JA. Modulation of Raf/MEK/ERK kinase activity does not affect the chemoresistance profile of advanced prostate cancer cells. Int J Oncol. 2005;26(6):1637–1644. [PubMed] [Google Scholar]
- 141.Lee JT, Jr, McCubrey JA. The Raf/MEK/ERK signal transduction cascade as a target for chemotherapeutic intervention in leukemia. Leukemia. 2002;16(4):486–507. doi: 10.1038/sj.leu.2402460. [DOI] [PubMed] [Google Scholar]
- 142.Lane DP. Cancer – p53, guardian of the genome. Nature. 1992;358(6381):15–16. doi: 10.1038/358015a0. [DOI] [PubMed] [Google Scholar]
- 143.Smith ML, Lancia JK, Mercer TI, Ip C. Selenium compounds regulate p53 by common and distinctive mechanisms. Anticancer Res. 2004;24(3A):1401–1408. [PubMed] [Google Scholar]
- 144.Beaudry GA, Bertelsen AH, Sherman MI. Therapeutic targeting of the p53 tumor suppressor gene. Curr Opin Biotechnol. 1996;7(6):592–600. doi: 10.1016/s0958-1669(96)80069-3. [DOI] [PubMed] [Google Scholar]
- 145.Milner J. Structures and functions of the tumor suppressor p53. Pathol Biol (Paris) 1997;45(10):797–803. [PubMed] [Google Scholar]
- 146.Strano S, Dell'Orso S, Di Agostino S, et al. Mutant p53: an oncogenic transcription factor. Oncogene. 2007;26(15):2212–2219. doi: 10.1038/sj.onc.1210296. [DOI] [PubMed] [Google Scholar]; ▪ Reports that p53 can also play roles as an oncogenic factor.
- 147.Wiman KG. Restoration of wild-type p53 function in human tumors: strategies for efficient cancer therapy. Adv Cancer Res. 2007;97:321–338. doi: 10.1016/S0065-230X(06)97014-6. [DOI] [PubMed] [Google Scholar]
- 148.Roth JA, Swisher SG, Meyn RE. p53 tumor suppressor gene therapy for cancer. Oncology. 1999;13(10 Suppl. 5):148–154. [PubMed] [Google Scholar]
- 149.Foster BA, Coffey HA, Morin MJ, Rastinejad F. Pharmacological rescue of mutant p53 conformation and function. Science. 1999;286(5449):2507–2510. doi: 10.1126/science.286.5449.2507. [DOI] [PubMed] [Google Scholar]
- 150.Mrass P, Rendl M, Mildner M, et al. Retinoic acid increases the expression of p53 and proapoptotic caspases and sensitizes keratinocytes to apoptosis: a possible explanation for tumor preventive action of retinoids. Cancer Res. 2004;64(18):6542–6548. doi: 10.1158/0008-5472.CAN-04-1129. [DOI] [PubMed] [Google Scholar]
- 151.Hsu SL, Hsu JW, Liu MC, Chen LY, Chang CD. Retinoic acid-mediated G1 arrest is associated with induction of p27(Kip1) and inhibition of cyclin-dependent kinase 3 in human lung squamous carcinoma CH27 cells. Exp Cell Res. 2000;258(2):322–331. doi: 10.1006/excr.2000.4933. [DOI] [PubMed] [Google Scholar]
- 152.Falasca L, Favale A, Gualandi G, Maietta G, Conti Devirgiliis L. Retinoic acid treatment induces apoptosis or expression of a more differentiated phenotype on different fractions of cultured fetal rat hepatocytes. Hepatology. 1998;28(3):727–737. doi: 10.1002/hep.510280319. [DOI] [PubMed] [Google Scholar]
- 153.Gallahan D, Kozak C, Callahan R. A new common integration region (int-3) for mouse mammary tumor virus on mouse chromosome 17. J Virol. 1987;61(1):218–220. doi: 10.1128/jvi.61.1.218-220.1987. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Efstratiadis A, Szabolcs M, Klinakis A. Notch, Myc and breast cancer. Cell Cycle. 2007;6(4):418–429. doi: 10.4161/cc.6.4.3838. [DOI] [PubMed] [Google Scholar]
- 155.Callahan R, Egan SE. Notch signaling in mammary development and oncogenesis. J Mammary Gland Biol Neoplasia. 2004;9(2):145–163. doi: 10.1023/B:JOMG.0000037159.63644.81. [DOI] [PubMed] [Google Scholar]
- 156.Weeraratna AT, Jiang Y, Hostetter G, et al. Wnt5a signaling directly affects cell motility and invasion of metastatic melanoma. Cancer Cell. 2002;1(3):279–288. doi: 10.1016/s1535-6108(02)00045-4. [DOI] [PubMed] [Google Scholar]
- 157.Callahan R, Raafat A. Notch signaling in mammary gland tumorigenesis. J Mammary Gland Biol Neoplasia. 2001;6(1):23–36. doi: 10.1023/a:1009512414430. [DOI] [PubMed] [Google Scholar]
- 158.Hu C, Dievart A, Lupien M, et al. Overexpression of activated murine Notch1 and Notch3 in transgenic mice blocks mammary gland development and induces mammary tumors. Am J Pathol. 2006;168(3):973–990. doi: 10.2353/ajpath.2006.050416. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Miyamoto Y, Maitra A, Ghosh B, et al. Notch mediates TGF-α-induced changes in epithelial differentiation during pancreatic tumorigenesis. Cancer Cell. 2003;3(6):565–576. doi: 10.1016/s1535-6108(03)00140-5. [DOI] [PubMed] [Google Scholar]
- 160.Koch U, Radtke F. Notch and cancer: a double-edged sword. Cell Mol Life Sci. 2007;64(21):2746–2762. doi: 10.1007/s00018-007-7164-1. [DOI] [PMC free article] [PubMed] [Google Scholar]; ▪▪ Discusses the point that Notch signaling is associated with oncogenic and growth-promoting roles but, depending on the tissue type, it can also function as a tumor suppressor.
- 161.Nicolas M, Wolfer A, Raj K, et al. Notch1 functions as a tumor suppressor in mouse skin. Nature Genet. 2003;33(3):416–421. doi: 10.1038/ng1099. [DOI] [PubMed] [Google Scholar]
- 162.Nguyen BC, Lefort K, Mandinova A, et al. Cross-regulation between Notch and p63 in keratinocyte commitment to differentiation. Genes Dev. 2006;20(8):1028–1042. doi: 10.1101/gad.1406006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Lowell S, Jones P, Le Roux I, Dunne J, Watt FM. Stimulation of human epidermal differentiation by δ-Notch signalling at the boundaries of stem-cell clusters. Curr Biol. 2000;10(9):491–500. doi: 10.1016/s0960-9822(00)00451-6. [DOI] [PubMed] [Google Scholar]
- 164.Rangarajan A, Talora C, Okuyama R, et al. Notch signaling is a direct determinant of keratinocyte growth arrest and entry into differentiation. EMBO J. 2001;20(13):3427–3436. doi: 10.1093/emboj/20.13.3427. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Vauclair S, Nicolas M, Barrandon Y, Radtke F. Notch1 is essential for postnatal hair follicle development and homeostasis. Dev Biol. 2005;284(1):184–193. doi: 10.1016/j.ydbio.2005.05.018. [DOI] [PubMed] [Google Scholar]
- 166.Weinmaster G. The ins and outs of Notch signalling. Mol Cell Neurosci. 1997;9(2):91–102. doi: 10.1006/mcne.1997.0612. [DOI] [PubMed] [Google Scholar]
- 167.Engin F, Bertin T, Ma O, et al. Notch signaling contributes to the pathogenesis of human osteosarcomas. Hum Mol Genet. 2009;18(8):1464–1470. doi: 10.1093/hmg/ddp057. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Villaronga MA, Bevan CL, Belandia B. Notch signaling: a potential therapeutic target in prostate cancer. Curr Cancer Drug Targets. 2008;8(7):566–580. doi: 10.2174/156800908786241096. [DOI] [PubMed] [Google Scholar]
- 169.Katoh M. Notch signaling in gastrointestinal tract (review) Int J Oncol. 2007;30(1):247–251. [PubMed] [Google Scholar]
- 170.Leong KG, Gao WQ. The Notch pathway in prostate development and cancer. Differentiation. 2008;76(6):699–716. doi: 10.1111/j.1432-0436.2008.00288.x. [DOI] [PubMed] [Google Scholar]
- 171.Zhang Y, Lian JB, Stein JL, van Wijnen AJ, Stein GS. The Notch-responsive transcription factor Hes-1 attenuates osteocalcin promoter activity in osteoblastic cells. J Cell Biochem. 2009;108(3):651–659. doi: 10.1002/jcb.22299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Zayzafoon M, Abdulkadir SA, McDonald JM. Notch signaling and ERK activation are important for the osteomimetic properties of prostate cancer bone metastatic cell lines. J Biol Chem. 2004;279(5):3662–3670. doi: 10.1074/jbc.M308158200. [DOI] [PubMed] [Google Scholar]
- 173.Scorey N, Fraser SP, Patel P, et al. Notch signalling and voltage-gated Na+ channel activity in human prostate cancer cells: independent modulation of in vitro motility. Prostate Cancer Prostatic Dis. 2006;9(4):399–406. doi: 10.1038/sj.pcan.4500894. [DOI] [PubMed] [Google Scholar]
- 174.Strom A, Arai N, Leers J, Gustafsson JA. The hairy and enhancer of split homologue-1 (HES-1) mediates the proliferative effect of 17β-estradiol on breast cancer cell lines. Oncogene. 2000;19(51):5951–5953. doi: 10.1038/sj.onc.1203990. [DOI] [PubMed] [Google Scholar]
- 175.Johansson T, Lejonklou MH, Ekeblad S, Stalberg P, Skogseid B. Lack of nuclear expression of hairy and enhancer of split-1 (HES1) in pancreatic endocrine tumors. Horm Metab Res. 2008;40(5):354–359. doi: 10.1055/s-2008-1076695. [DOI] [PubMed] [Google Scholar]
- 176.Zhang P, Yang Y, Zweidler-McKay PA, Hughes DP. Critical role of Notch signaling in osteosarcoma invasion and metastasis. Clin Cancer Res. 2008;14(10):2962–2969. doi: 10.1158/1078-0432.CCR-07-1992. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 177.Nickoloff BJ, Osborne BA, Miele L. Notch signaling as a therapeutic target in cancer: a new approach to the development of cell fate modifying agents. Oncogene. 2003;22(42):6598–6608. doi: 10.1038/sj.onc.1206758. [DOI] [PubMed] [Google Scholar]
- 178.Wong GT, Manfra D, Poulet FM, et al. Chronic treatment with the γ-secretase inhibitor LY-411,575 inhibits β-amyloid peptide production and alters lymphopoiesis and intestinal cell differentiation. J Biol Chem. 2004;279(13):12876–12882. doi: 10.1074/jbc.M311652200. [DOI] [PubMed] [Google Scholar]
- 179.Spiegel D, Sephton SE. Psychoneuroimmune and endocrine pathways in cancer: effects of stress and support. Semin Clin Neuropsychiatry. 2001;6(4):252–265. doi: 10.1053/scnp.2001.26995. [DOI] [PubMed] [Google Scholar]
- 180.McGregor BA, Antoni MH. Psychological intervention and health outcomes among women treated for breast cancer: a review of stress pathways and biological mediators. Brain Behav Immun. 2009;23(2):159–166. doi: 10.1016/j.bbi.2008.08.002. [DOI] [PMC free article] [PubMed] [Google Scholar]; ▪ Reviews and calls on psychological intervention studies to focus on more newly discovered stress–tumor pathways and follow larger cohorts of the more vulnerable patients over longer periods to evaluate the biobehavioral mechanisms and lasting effects of these interventions on health and quality of life.
- 181.Andersen BL, Farrar WB, Golden-Kreutz D, et al. Stress and immune responses after surgical treatment for regional breast cancer. J Natl Cancer Inst. 1998;90(1):30–36. doi: 10.1093/jnci/90.1.30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Coker AL, Sanderson M, Ellison GL, Fadden MK. Stress, coping, social support, and prostate cancer risk among older African American and Caucasian men. Ethn Dis. 2006;16(4):978–987. [PubMed] [Google Scholar]
- 183.Giese-Davis J, Wilhelm FH, Conrad A, et al. Depression and stress reactivity in metastatic breast cancer. Psychosom Med. 2006;68(5):675–683. doi: 10.1097/01.psy.0000238216.88515.e5. [DOI] [PubMed] [Google Scholar]
- 184.Vanacore N, Spila-Alegiani S, Raschetti R, Meco G. Mortality cancer risk in parkinsonian patients: a population-based study. Neurology. 1999;52(2):395–398. doi: 10.1212/wnl.52.2.395. [DOI] [PubMed] [Google Scholar]
- 185.D'Amelio M, Ragonese P, Morgante L, et al. Tumor diagnosis preceding Parkinson's disease: a case–control study. Mov Disord. 2004;19(7):807–811. doi: 10.1002/mds.20123. [DOI] [PubMed] [Google Scholar]
- 186.Olsen JH, Friis S, Frederiksen K, et al. Atypical cancer pattern in patients with Parkinson's disease. Br J Cancer. 2005;92(1):201–205. doi: 10.1038/sj.bjc.6602279. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Roe CM, Fitzpatrick AL, Xiong C, et al. Cancer linked to Alzheimer disease but not vascular dementia. Neurology. 2010;74:106–112. doi: 10.1212/WNL.0b013e3181c91873. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Buchler P, Reber HA, Roth MM, et al. Target therapy using a small molecule inhibitor against angiogenic receptors in pancreatic cancer. Neoplasia. 2007;9(2):119–127. doi: 10.1593/neo.06616. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Pasetto LM, Bortolami A, Falci C, Sinigaglia G, Monfardini S. Recent progress in target therapy in colorectal cancer. Anticancer Res. 2006;26(5B):3973–3981. [PubMed] [Google Scholar]
- 190.Midgley RS, Kerr DJ. Ras as a target in cancer therapy. Crit Rev Oncol Hematol. 2002;44(2):109–120. doi: 10.1016/s1040-8428(01)00189-5. [DOI] [PubMed] [Google Scholar]
- 191.Kelley JR, Brown JM, Frasier MM, et al. The cancer-associated Sm-like oncogene: a novel target for the gene therapy of pancreatic cancer. Surgery. 2000;128(2):353–360. doi: 10.1067/msy.2000.107605. [DOI] [PubMed] [Google Scholar]
- 192.Jimeno A, Daw NC, Amador ML, et al. Analysis of biologic surrogate markers from a Children's Oncology Group Phase I trial of gefitinib in pediatric patients with solid tumors. Pediatr Blood Cancer. 2007;49(3):352–357. doi: 10.1002/pbc.20753. [DOI] [PubMed] [Google Scholar]
- 193.Epstein RJ. Maintenance therapy to suppress micrometastasis: the new challenge for adjuvant cancer treatment. Clin Cancer Res. 2005;11(15):5337–5341. doi: 10.1158/1078-0432.CCR-05-0437. [DOI] [PubMed] [Google Scholar]
- 194.Chari RV. Targeted cancer therapy: conferring specificity to cytotoxic drugs. Acc Chem Res. 2008;41(1):98–107. doi: 10.1021/ar700108g. [DOI] [PubMed] [Google Scholar]
- 195.Gelber RD, Bonetti M, Castiglione-Gertsch M, Coates AS, Goldhirsch A, International Breast Cancer Study Group (IBCSG) Tailoring adjuvant treatments for the individual breast cancer patient. Breast. 2003;12(6):558–568. doi: 10.1016/s0960-9776(03)00166-8. [DOI] [PubMed] [Google Scholar]
- 196.Hamberg P, Verweij J, Seynaeve C. Cytotoxic therapy for the elderly with metastatic breast cancer: a review on safety, pharmacokinetics and efficacy. Eur J Cancer. 2007;43(10):1514–1528. doi: 10.1016/j.ejca.2007.03.017. [DOI] [PubMed] [Google Scholar]
- 197.Briest S, Stearns V. Chemotherapeutic strategies for advanced breast cancer. Oncology. 2007;21(11):1325–1335. discussion 1338–1340. [PubMed] [Google Scholar]
- 198.Rong Y, Jin D, Wu W, et al. Induction of protective and therapeutic anti-pancreatic cancer immunity using a reconstructed MUC1 DNA vaccine. BMC Cancer. 2009;9:191. doi: 10.1186/1471-2407-9-191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199.Taghavi P, Verhoeven E, Jacobs JJ, et al. In vitro genetic screen identifies a cooperative role for LPA signaling and c-Myc in cell transformation. Oncogene. 2008;27:6806–6816. doi: 10.1038/onc.2008.294. [DOI] [PubMed] [Google Scholar]
- 200.Iaquinta PJ, Lees JA. Life and death decisions by the E2F transcription factors. Curr Opin Cell Biol. 2007;19(6):649–657. doi: 10.1016/j.ceb.2007.10.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201.Pierce AM, Schneider-Broussard R, Gimenez-Conti IB, et al. E2F1 has both oncogenic and tumor-suppressive properties in a transgenic model. Mol Cell Biol. 1999;19(9):6408–6414. doi: 10.1128/mcb.19.9.6408. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202.Fisher GH, Wellen SL, Klimstra D, et al. Induction and apoptotic regression of lung adenocarcinomas by regulation of a K-Ras transgene in the presence and absence of tumor suppressor genes. Genes Dev. 2001;15(24):3249–3262. doi: 10.1101/gad.947701. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203.Macias-Perez I, Borza C, Chen X, et al. Loss of integrin α1β1 ameliorates Kras-induced lung cancer. Cancer Res. 2008;68(15):6127–6135. doi: 10.1158/0008-5472.CAN-08-1395. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204.Zhang Z, Wang Y, Vikis HG, et al. Wildtype Kras2 can inhibit lung carcinogenesis in mice. Nat Genet. 2001;29(1):25–33. doi: 10.1038/ng721. [DOI] [PubMed] [Google Scholar]
- 205.Cho NY, Choi M, Kim BH, et al. BRAF and KRAS mutations in prostatic adenocarcinoma. Int J Cancer. 2006;119(8):1858–1862. doi: 10.1002/ijc.22071. [DOI] [PubMed] [Google Scholar]
- 206.Allen PB, Wiedemann LM. An activating mutation in the ATP binding site of the ABL kinase domain. J Biol Chem. 1996;271(32):19585–19591. doi: 10.1074/jbc.271.32.19585. [DOI] [PubMed] [Google Scholar]
- 207.Jackson P, Baltimore D. N-terminal mutations activate the leukemogenic potential of the myristoylated form of c-abl. EMBO J. 1989;8(2):449–456. doi: 10.1002/j.1460-2075.1989.tb03397.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208.Carter TA, Wodicka LM, Shah NP, et al. Inhibition of drug-resistant mutants of ABL, KIT, EGF receptor kinases. Proc Natl Acad Sci USA. 2005;102(31):11011–11016. doi: 10.1073/pnas.0504952102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209.Cordon-Cardo C, Reuter VE. Alterations of tumor suppressor genes in bladder cancer. Semin Diagn Pathol. 1997;14(2):123–132. [PubMed] [Google Scholar]
- 210.Sakata K. Alterations of tumor suppressor genes and the H-ras oncogene in oral squamous cell carcinoma. J Oral Pathol Med. 1996;25(6):302–307. doi: 10.1111/j.1600-0714.1996.tb00266.x. [DOI] [PubMed] [Google Scholar]
- 211.Zhou H, Kuang J, Zhong L, et al. Tumour amplified kinase STK15/BTAK induces centrosome amplification, aneuploidy and transformation. Nat Genet. 1998;20(2):189–193. doi: 10.1038/2496. [DOI] [PubMed] [Google Scholar]
- 212.Lu LY, Wood JL, Ye L, et al. Aurora a is essential for early embryonic development and tumor suppression. J Biol Chem. 2008;283(46):31785–31790. doi: 10.1074/jbc.M805880200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 213.Weinberg BD, Ai H, Blanco E, Anderson JM, Gao J. Antitumor efficacy and local distribution of doxorubicin via intratumoral delivery from polymer millirods. J Biomed Mater Res A. 2007;81(1):161–170. doi: 10.1002/jbm.a.30914. [DOI] [PubMed] [Google Scholar]
- 214.Polakis P. Wnt signaling and cancer. Genes Dev. 2000;14(15):1837–1851. [PubMed] [Google Scholar]
- 215.Voeller HJ, Truica CI, Gelmann EP. β-catenin mutations in human prostate cancer. Cancer Res. 1998;58(12):2520–2523. [PubMed] [Google Scholar]
- 216.Chesire DR, Ewing CM, Sauvageot J, Bova GS, Isaacs WB. Detection and analysis of β-catenin mutations in prostate cancer. Prostate. 2000;45(4):323–334. doi: 10.1002/1097-0045(20001201)45:4<323::aid-pros7>3.0.co;2-w. [DOI] [PubMed] [Google Scholar]
- 217.Kim K, Pang KM, Evans M, Hay ED. Overexpression of β-catenin induces apoptosis independent of its transactivation function with LEF-1 or the involvement of major G1 cell cycle regulators. Mol Biol Cell. 2000;11(10):3509–3523. doi: 10.1091/mbc.11.10.3509. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 218.Terry S, Yang X, Chen MW, Vacherot F, Buttyan R. Multifaceted interaction between the androgen and Wnt signaling pathways and the implication for prostate cancer. J Cell Biochem. 2006;99(2):402–410. doi: 10.1002/jcb.20983. [DOI] [PubMed] [Google Scholar]
- 219.Yang X, Chen MW, Terry S, et al. Complex regulation of human androgen receptor expression by Wnt signaling in prostate cancer cells. Oncogene. 2006;25(24):3436–3444. doi: 10.1038/sj.onc.1209366. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220.Yu X, Wang Y, Jiang M, et al. Activation of β-catenin in mouse prostate causes HGPIN and continuous prostate growth after castration. Prostate. 2009;69(3):249–262. doi: 10.1002/pros.20877. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 221.Kinzler KW, Vogelstein B. Lessons from hereditary colorectal cancer. Cell. 1996;87(2):159–170. doi: 10.1016/s0092-8674(00)81333-1. [DOI] [PubMed] [Google Scholar]
- 222.Hanson CA, Miller JR. Non-traditional roles for the adenomatous polyposis coli (APC) tumor suppressor protein. Gene. 2005;361:1–12. doi: 10.1016/j.gene.2005.07.024. [DOI] [PubMed] [Google Scholar]
- 223.Dobosy JR, Roberts JL, Fu VX, Jarrard DF. The expanding role of epigenetics in the development, diagnosis and treatment of prostate cancer and benign prostatic hyperplasia. J Urol. 2007;177(3):822–831. doi: 10.1016/j.juro.2006.10.063. [DOI] [PubMed] [Google Scholar]
- 224.Kawasaki Y, Sato R, Akiyama T. Mutated APC and Asef are involved in the migration of colorectal tumour cells. Nat Cell Biol. 2003;5(3):211–215. doi: 10.1038/ncb937. [DOI] [PubMed] [Google Scholar]
- 225.Tighe A, Johnson VL, Taylor SS. Truncating APC mutations have dominant effects on proliferation, spindle checkpoint control, survival and chromosome stability. J Cell Sci. 2004;117(Pt 26):6339–6353. doi: 10.1242/jcs.01556. [DOI] [PubMed] [Google Scholar]
- 226.Caldwell CM, Green RA, Kaplan KB. APC mutations lead to cytokinetic failures in vitro and tetraploid genotypes in Min mice. J Cell Biol. 2007;178(7):1109–1120. doi: 10.1083/jcb.200703186. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 227.Kallakury BV, Sheehan CE, Ross JS. Co-downregulation of cell adhesion proteins α- and β-catenins, p120CTN, E-cadherin, and CD44 in prostatic adenocarcinomas. Hum Pathol. 2001;32(8):849–855. doi: 10.1053/hupa.2001.26463. [DOI] [PubMed] [Google Scholar]
- 228.Nagafuchi A, Takeichi M, Tsukita S. The 102 kd cadherin-associated protein: similarity to vinculin and posttranscriptional regulation of expression. Cell. 1991;65(5):849–857. doi: 10.1016/0092-8674(91)90392-c. [DOI] [PubMed] [Google Scholar]
- 229.Papkoff J. Regulation of complexed and free catenin pools by distinct mechanisms. Differential effects of Wnt-1 and v-Src. J Biol Chem. 1997;272(7):4536–4543. [PubMed] [Google Scholar]
- 230.Anastasiadis PZ, Reynolds AB. Regulation of Rho GTPases by p120-catenin. Curr Opin Cell Biol. 2001;13(5):604–610. doi: 10.1016/s0955-0674(00)00258-1. [DOI] [PubMed] [Google Scholar]
- 231.Noren NK, Liu BP, Burridge K, Kreft B. p120 catenin regulates the actin cytoskeleton via Rho family GTPases. J Cell Biol. 2000;150(3):567–580. doi: 10.1083/jcb.150.3.567. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 232.Reynolds AB, Jenkins NA, Gilbert DJ, et al. The gene encoding p120cas, a novel catenin, localizes on human chromosome 11q11 (CTNND) and mouse chromosome 2 (Catns) Genomics. 1996;31(1):127–129. doi: 10.1006/geno.1996.0020. [DOI] [PubMed] [Google Scholar]
- 233.Thoreson MA, Anastasiadis PZ, Daniel JM, et al. Selective uncoupling of p120ctn from E-cadherin disrupts strong adhesion. J Cell Biol. 2000;148(1):189–202. doi: 10.1083/jcb.148.1.189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 234.Wang T, Chen YH, Hong H, et al. Increased nucleotide polymorphic changes in the 5′-untranslated region of δ-catenin (CTNND2) gene in prostate cancer. Oncogene. 2009;28(4):555–564. doi: 10.1038/onc.2008.399. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 235.Burger MJ, Tebay MA, Keith PA, et al. Expression analysis of δ-catenin and prostate-specific membrane antigen: their potential as diagnostic markers for prostate cancer. Int J Cancer. 2002;100(2):228–237. doi: 10.1002/ijc.10468. [DOI] [PubMed] [Google Scholar]
- 236.Sylvestre Y, De Guire V, Querido E, et al. An E2F/miR-20a autoregulatory feedback loop. J Biol Chem. 2007;282(4):2135–2143. doi: 10.1074/jbc.M608939200. [DOI] [PubMed] [Google Scholar]
- 237.Galardi S, Mercatelli N, Giorda E, et al. miR-221 and miR-222 expression affects the proliferation potential of human prostate carcinoma cell lines by targeting p27Kip1. J Biol Chem. 2007;282(32):23716–23724. doi: 10.1074/jbc.M701805200. [DOI] [PubMed] [Google Scholar]
- 238.Shi XB, Tepper CG, White RW. MicroRNAs and prostate cancer. J Cell Mol Med. 2008;12(5A):1456–1465. doi: 10.1111/j.1582-4934.2008.00420.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 239.Musiyenko A, Bitko V, Barik S. Ectopic expression of miR-126*, an intronic product of the vascular endothelial EGF-like 7 gene, regulates prostein translation and invasiveness of prostate cancer LNCaP cells. J Mol Med. 2008;86(3):313–322. doi: 10.1007/s00109-007-0296-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 240.Lin SL, Chiang A, Chang D, Ying SY. Loss of mir-146a function in hormone-refractory prostate cancer. RNA. 2008;14(3):417–424. doi: 10.1261/rna.874808. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 241.Yoon S, De Micheli G. Prediction of regulatory modules comprising microRNAs and target genes. Bioinformatics. 2005;21(Suppl. 2):II93–II100. doi: 10.1093/bioinformatics/bti1116. [DOI] [PubMed] [Google Scholar]
- 242.Bonci D, Coppola V, Musumeci M, et al. The miR-15a-miR-16–11 cluster controls prostate cancer by targeting multiple oncogenic activities. Nat Med. 2008;14(11):1271–1277. doi: 10.1038/nm.1880. [DOI] [PubMed] [Google Scholar]
- 243.Prueitt RL, Yi M, Hudson RS, et al. Expression of microRNAs and protein-coding genes associated with perineural invasion in prostate cancer. Prostate. 2008;68(11):1152–1164. doi: 10.1002/pros.20786. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 244.O'Donnell KA, Wentzel EA, Zeller KI, Dang CV, Mendell JT. c-Myc-regulated microRNAs modulate E2F1 expression. Nature. 2005;435(7043):839–843. doi: 10.1038/nature03677. [DOI] [PubMed] [Google Scholar]
- 245.Murakami Y, Yasuda T, Saigo K, et al. Comprehensive analysis of microRNA expression patterns in hepatocellular carcinoma and non-tumorous tissues. Oncogene. 2006;25(17):2537–2545. doi: 10.1038/sj.onc.1209283. [DOI] [PubMed] [Google Scholar]
- 246.Chan JA, Krichevsky AM, Kosik KS. MicroRNA-21 is an antiapoptotic factor in human glioblastoma cells. Cancer Res. 2005;65(14):6029–6033. doi: 10.1158/0008-5472.CAN-05-0137. [DOI] [PubMed] [Google Scholar]
- 247.Ciafre SA, Galardi S, Mangiola A, et al. Extensive modulation of a set of microRNAs in primary glioblastoma. Biochem Biophys Res Commun. 2005;334(4):1351–1358. doi: 10.1016/j.bbrc.2005.07.030. [DOI] [PubMed] [Google Scholar]
- 248.Iorio MV, Ferracin M, Liu CG, et al. MicroRNA gene expression deregulation in human breast cancer. Cancer Res. 2005;65(16):7065–7070. doi: 10.1158/0008-5472.CAN-05-1783. [DOI] [PubMed] [Google Scholar]
- 249.Liu JY, Chuang TC, Way TD, et al. The N-terminal domain of EBNA1 acts as a suppressor of the HER2/neu oncogene. Cancer Lett. 2008;273(2):273–280. doi: 10.1016/j.canlet.2008.08.013. [DOI] [PubMed] [Google Scholar]
- 250.Criswell TL, Dumont N, Barnett JV, Arteaga CL. Knockdown of the transforming growth factor-β type III receptor impairs motility and invasion of metastatic cancer cells. Cancer Res. 2008;68(18):7304–7312. doi: 10.1158/0008-5472.CAN-07-6777. [DOI] [PubMed] [Google Scholar]
- 251.Yang J, Song K, Krebs TL, Jackson MW, Danielpour D. Rb/E2F4 and Smad2/3 link survivin to TGF-β-induced apoptosis and tumor progression. Oncogene. 2008;27(40):5326–5338. doi: 10.1038/onc.2008.165. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 252.Barrack ER. TGF- β in prostate cancer: a growth inhibitor that can enhance tumorigenicity. Prostate. 1997;31(1):61–70. doi: 10.1002/(sici)1097-0045(19970401)31:1<61::aid-pros10>3.0.co;2-m. [DOI] [PubMed] [Google Scholar]
- 253.Jakowlew SB. Transforming growth factor-β in cancer and metastasis. Cancer Metastasis Rev. 2006;25(3):435–457. doi: 10.1007/s10555-006-9006-2. [DOI] [PubMed] [Google Scholar]
- 254.Bello-DeOcampo D, Tindall DJ. TGF-βl/Smad signaling in prostate cancer. Curr Drug Targets. 2003;4(3):197–207. doi: 10.2174/1389450033491118. [DOI] [PubMed] [Google Scholar]
- 255.Acquaviva J, Chen X, Ren R. IRF-4 functions as a tumor suppressor in early B-cell development. Blood. 2008;112(9):3798–3806. doi: 10.1182/blood-2007-10-117838. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 256.Wang Z, Smith KS, Murphy M, et al. Glycogen synthase kinase 3 in MLL leukaemia maintenance and targeted therapy. Nature. 2008;455(7217):1205–1209. doi: 10.1038/nature07284. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 257.Chalhoub N, Baker SJ. PTEN and the PI3-kinase pathway in cancer. Annu Rev Pathol. 2009;4:127–150. doi: 10.1146/annurev.pathol.4.110807.092311. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 258.DeGraffenried LA, Fulcher L, Friedrichs WE, et al. Reduced PTEN expression in breast cancer cells confers susceptibility to inhibitors of the PI3 kinase/Akt pathway. Ann Oncol. 2004;15(10):1510–1516. doi: 10.1093/annonc/mdh388. [DOI] [PubMed] [Google Scholar]
- 259.Wong KK, Engelman JA, Cantley LC. Targeting the PI3K signaling pathway in cancer. Curr Opin Genet Dev. 2009;20(1):87–90. doi: 10.1016/j.gde.2009.11.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 260.Sun X, Huang J, Homma T, et al. Genetic alterations in the PI3K pathway in prostate cancer. Anticancer Res. 2009;29(5):1739–1743. [PubMed] [Google Scholar]
- 261.Sarker D, Reid AH, Yap TA, de Bono JS. Targeting the PI3K/AKT pathway for the treatment of prostate cancer. Clin Cancer Res. 2009;15(15):4799–4805. doi: 10.1158/1078-0432.CCR-08-0125. [DOI] [PubMed] [Google Scholar]
- 262.Morgan TM, Koreckij TD, Corey E. Targeted therapy for advanced prostate cancer: inhibition of the PI3K/Akt/mTOR pathway. Curr Cancer Drug Targets. 2009;9(2):237–249. doi: 10.2174/156800909787580999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 263.Chen M, Cassidy A, Gu J, et al. Genetic variations in PI3K–AKT–mTOR pathway and bladder cancer risk. Carcinogenesis. 2009;30(12):2047–2052. doi: 10.1093/carcin/bgp258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 264.Yuan TL, Cantley LC. PI3K pathway alterations in cancer: variations on a theme. Oncogene. 2008;27(41):5497–5510. doi: 10.1038/onc.2008.245. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 265.Georgescu MM. NHERF1: molecular brake on the PI3K pathway in breast cancer. Breast Cancer Res. 2008;10(2):106. doi: 10.1186/bcr1992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 266.Cohen Y, Shalmon B, Korach J, et al. AKT1 pleckstrin homology domain E17K activating mutation in endometrial carcinoma. Gynecol Oncol. 2010;116(1):88–91. doi: 10.1016/j.ygyno.2009.09.038. [DOI] [PubMed] [Google Scholar]
- 267.Askham JM, Platt F, Chambers PA, et al. AKT1 mutations in bladder cancer: identification of a novel oncogenic mutation that can co-operate with E17K. Oncogene. 2010;29(1):150–155. doi: 10.1038/onc.2009.315. [DOI] [PubMed] [Google Scholar]
- 268.Boormans JL, Hermans KG, van Leenders GJ, Trapman J, Verhagen PC. An activating mutation in AKT1 in human prostate cancer. Int J Cancer. 2008;123(11):2725–2726. doi: 10.1002/ijc.23787. [DOI] [PubMed] [Google Scholar]
- 269.Mohamedali A, Lea NC, Feakins RM, et al. AKT1E17K mutation in pancreatic cancer. Technol Cancer Res Treat. 2008;7(5):407–408. doi: 10.1177/153303460800700509. [DOI] [PubMed] [Google Scholar]
- 270.Bleeker FE, Felicioni L, Buttitta F, et al. AKT1E17K in human solid tumours. Oncogene. 2008;27:5648–5650. doi: 10.1038/onc.2008.170. [DOI] [PubMed] [Google Scholar]
- 271.Zupnick A, Prives C. Mutational analysis of the p53 core domain L1 loop. J Biol Chem. 2006;281(29):20464–20473. doi: 10.1074/jbc.M603387200. [DOI] [PubMed] [Google Scholar]
Website
- 301.American Cancer Society. Cancer Facts and Figures. www.cancer.org/downloads/STT/CAFF2007PWSecured.pdf.