Abstract
The human HtrA family of serine proteases consists of four members: HtrA1, HtrA2, HtrA3 and HtrA4. Although prokaryotic HtrA proteins are well characterized in their dual roles as chaperones and proteases that degrade misfolded proteins in the periplasm, some members of mammalian HtrA proteins are described as potential modulators of programmed cell death and chemotherapy-induced cytotoxicity. Goal of this review article is to describe the molecular alterations associated with these HtrA serine proteases and how these alterations may be associated with tumor behavior and response to chemotherapy. We will also discuss evidence that chemotherapeutic drugs regulate the expression and activation of HtrA serine proteases and that these proteases contributes to programmed cell death. Finally, we will discuss the potential role of epigenetic therapy in targeting the expression and activation of HtrA serine proteases and the mechanisms by which these proteases enhance cytotoxic effect of conventional chemotherapy.
Programmed cell death is a critical component of chemotherapy-induced cytotoxicity, and a determinant in sensitivity to chemotherapeutic agents. As such, various pathways that regulate programmed cell death have been the focus of intense research interest as therapeutic targets in oncology in order to enhance sensitivity to chemotherapeutic agents or to overcome the problem of resistance [1–4]. Two major pathways that regulate programmed cell death are the extrinsic death receptors-mediated and the intrinsic mitochondrial apoptotic signal pathways which converge upon the activation of caspases and the execution of cell death programs [4]. These pathways are in turn regulated by several pro- and anti-apoptotic proteins [5]. Various agents that promote caspase-mediated apoptotic signaling or agents that antagonize survival signaling are being tested in preclinical and clinical trials to supplement conventional chemotherapy [2]. In this paper, we will briefly review current therapeutic targets in cell death program and describe potential novel therapeutic targets consisting of HtrA serine proteases that interact with caspase-mediated apoptotic pathway and facilitate sensitivity to chemotherapeutic agents.
The effectiveness of current chemotherapy regimens depends on how well anticancer drugs can induce cell death in cancer cells. Accordingly, alterations in programmed cell death or survival pathways have been reported to influence cancer cell response to chemotherapy. Thus, chemoresistance is viewed by some as the result of changes in the finely tuned balance between survival and cell death pathways. In this regard, it is important to note that the relative contribution of apoptotic defects to clinical multidrug resistance is still not completely resolved [6, 7], and tumor microenvironment, genetic constitution of the tumor, and alternate cell death programs can have considerable impact on the survival of drug resistant cells [6, 8, 9].
In tumorigenesis, cancer cells acquire a selective growth advantage through a diminished capacity to undergo programmed cell death under growth-restrictive condition, aberrant oncogene activation or genetic injuries. Defects in genes involved in cell death programs are frequently associated with various types of malignancies [10], and consequent insensitivity to cell death signaling is considered a hallmark of cancer cells [11]. Insensitivity to cell death signal is suggested to not only promote carcinogenesis by allowing perpetuation and diversification of genetic alterations and oncogene activations in cancer cells but also facilitate resistance to chemotherapy by increasing the threshold against death signals. Apoptosis, the best-characterized mechanism of programmed cell death, involves targeted proteolysis by activated caspases, and this apoptotic pathway is tightly regulated by the balance of pro-apoptotic and anti-apoptotic signaling cascades [5, 12]. In cancer, activation of anti-apoptotic survival pathways and suppression of pro-apoptotic pathways mitigate apoptosis [11].
Conventional Targets
In general, conventional chemotherapeutic agents achieve greater cytotoxicity in cancer cells by either targeting components of DNA synthesis, DNA damage, or mitotic pathways [13, 14]. Conventional chemotherapeutic agents, such as alkylating agents, anthracyclines, topoisomerase inhibitors, and anti-metabolites, target DNA synthesis pathways, whereas vinca alkaloids, taxane, and epothilone target cell cycle progression by disrupting cytoskeletal components involved in cell division [13, 15]. Other agents, such as retinoids and hormone therapies, target specific types of cancer that depend on particular growth signals [3, 16–18]. In addition to conventional therapeutic agents, newer therapeutic agents that target these pathways are being developed to enhance sensitivity and specificity towards malignant cells [15, 19].
Emerging Targets
In recent years, newer therapeutic agents are increasingly used as a combined therapy to affect greater cytotoxicity in cancer cells by specifically targeting altered biological pathways in particular patient population. These newer targeted therapeutic agents include small molecule inhibitors that targets growth factor signaling pathways (EGFR, MAP Kinases, Ras, Src, IGF1R, VEGFR), survival pathways (PI3K/Akt, NFκB, Survivin, IAPs), HSP90 pathway (17-AAG), and cell cycle proteins (cdk inhibitors, flavopiridol, Ro31-7453, SB715992), monoclonal antibodies that target ErbB1, ErbB2, VEGFR, and other factors that promote proliferation or survival signaling [3, 19–29]. In addition to newer therapeutics, several novel therapeutic agents, such as immunotherapy, cytokines (INFα, INFγ, IL2, anti-TNF, and TRAIL), attenuated oncolytic viruses and viral antigen-targeted therapy (Measles, H-1PV, EBV), BH3 mimetic (ABT-737), epigenetic therapies (TSA, SAHA, CBHA, LBH), and metabolic pathway inhibitors (RAD001, rapamycin) are also being tested in preclinical and clinical trials [3, 21, 30–44].
Apoptotic regulators as targets of novel therapeutics
Targeting apoptosis is a logical strategy for cancer drug discovery [45]. One of the most promising discoveries in the developmental therapeutics is the identification of agents that affect programmed cell death pathways regulated by pro-apoptotic and anti-apoptotic proteins belonging to Bcl-2 family, anti-apoptotic survival proteins belonging to inhibitors of apoptosis proteins (IAPs) family, and pro-apoptotic death receptors and caspases. Bcl-2 is an anti-apoptotic protein and is frequently overexpressed in human malignancies, and its overexpression is associated with tumor maintenance, progression, and resistance to chemotherapy [46]. Accordingly, several strategies, such as small molecule inhibitor (e.g., HA14-1), antisense oligodeoxynucleotides (e.g., G3139), BH3 peptidomimetics, and Bcl-2 inhibitors (e.g., ABT-737), have been developed to inhibit Bcl-2 [42, 47–49]. Another class of proteins with anti-apoptotic properties are IAPs consisting of XIAP, cIAP1, cIAP2, BIRC1, survivin, ML-IAP, and IAP-like protein 2 [50, 51]. These proteins associate with caspases and prevent them from activation; thus they represent major regulators of apoptosis. Overexpression of IAP proteins has been associated with increased resistance to pro-apoptotic stimuli in various solid tumors and hematologic malignancies. According, various IAP antagonists, such as Compound 11, Compound 3, SM-131, and Compound 8, LBW242, TPI 1396-12, and TWX 024, are being tested in preclinical and clinical trials [52]. Death receptors, which are component of extrinsic apoptotic pathway mediated by death inducing ligands, such as tumor necrosis factor (TNF), TNF-Related Apoptosis-Inducing Ligand (TRAIL), and Fas ligand (FasL), are also targeted as novel therapies. TRAIL receptors are overexpressed in various solid malignancies, and thus represent a valid target opportunity in cancer. Several TRAIL receptor agonist monoclonal antibodies have been developed to target this extrinsic signaling pathway, and therapeutic potential of these antibodies are being evaluated clinical trials [21].
Alternative pathways in programmed cell death
Although disruption of caspase activity is one of the characteristics of most cancers [12], recent studies suggest that caspase-independent cell death programs also play key roles in cellular tumorigenesis [53, 54]. Non-caspase proteases like cathepsins and serine proteases like granzymes induce cell death by targeting either distinct or overlapping caspase substrates [53, 55]. Cell death also occurs through non-apoptotic pathways such as autophagy, and these pathways are being targeted for novel therapy [56]. While the exact mechanisms behind non-apoptotic cell death programs are not fully known, involvement of serine proteases, key tumor suppressors like p53, PTEN, p16Ink4a, and anti-apoptotic proteins such as survivin suggests that there is considerable cross-talk between apoptotic and non-apoptotic pathways of cell death [54]. In particular, emerging evidence support the role of serine proteases in programmed cell death and chemotherapy-induced cytotoxicity [53, 57–64].
The HtrA family of serine proteases
The HtrA family of serine proteases was initially identified in E. coli by two phenotypes of null mutants that were unable to grow at elevated temperatures (HtrA for High temperature requirement) [65], or failed to digest misfolded protein in the periplasm (DegP) [66]. Subsequently, homologues of HtrA/DegP have been described in a variety of species, including Gram-negative and -positive bacteria, plants and mammals. These proteins normally contain two conserved core domains, a chymotrypsin-like protease domain, and at least one C-terminal PDZ domain. In contrast to other protease-chaperone systems, HtrA represents the first well-known protein quality control factor that acts in an ATP-independent manner [67, 68].
Until now, four human homologues of E. coli HtrA have been identified: HtrA1 (L56 or PRSS11) [69, 70], HtrA2/Omi [71, 72], HtrA3 (PRSP) [73] and HtrA4. All mammalian HtrA proteins, belonging to this family, share a highly conserved chymotrypsin-like serine protease domain and one PDZ domain at the C-terminus [74]. Otherwise, structure of N-terminal regions of mammalian HtrA1, 3 and 4 are distinct from that of HtrA2/Omi: mitochondrial HtrA2/Omi possess a transmembrane anchor, and a large section of the N-terminus is removed by processing, whereas the N-termini of HtrA1, 3 and 4 all contain predicted signal peptides as well as domains that are recognized as IGF binding and protease inhibitor domains [75]. It is interesting to notice that of the four human HtrA family members, HtrA/Omi2 is the only one with a clear intracellular localization [75].
The HtrA family of serine protease appears to be involved in several important biological mechanisms in mammals, such as growth, apoptosis, arthritis, embryogenesis, neurodegenerative and neuromuscular disorder, and cancer. HtrA1 is the first sequenced member of the human HtrA protein family when it was identified as a gene expressed by normal fibroblasts but not by SV40 transformed counterparts [70]. Subsequently, Hu and collaborators found the same gene during analysis of transcripts overexpressed in osteoarthritic cartilage [69]. HtrA1 has a widespread pattern of expression, and its level in human tissues is modulated both in tissues with different physiological activities [76]. Expression of HtrA1 is also modulated during the proliferating phase of endometrium and during pregnancy [77].
The second member of human HtrA protein family, HtrA2/Omi, was independently identified in a yeast two-hybrid screen for Mxi2-interacting proteins [71], and as a stress-activated protease upregulated in mammalian cells in response to stress [72]. It is localized in the mitochondrial intermembrane space, and it is released into the cytosol in response to apoptotic stimuli [62]. The N-terminal tetrapeptide motif (AVPS) of mature HtrA2 is very similar to those of the mature proapoptotic Smac protein (Second Mitochondria-derived Activator of Caspases) and recognized as IAP-binding motif (IBMs); this fact provides evidence that the two proteins function in a similar way to neutralize inhibitor apoptosis proteins (IAPs) [78]. The role of HtrA2/Omi as a promoter of cell death was confirmed independently by several other groups [63, 79–81]. It was shown that the mature HtrA2/Omi is able to induce apoptosis in human cells both in a caspase-independent manner through its protease activity and in a caspase-dependent manner via its ability to disrupt caspase-IAP interaction [82–86]. HtrA2/Omi is expressed ubiquitously in human tissue, and the proteolytic activity of the protein is substantially upregulated in mouse kidneys following ischemia/reperfusion. Gray et al., who identified HtrA2/Omi using presenilin-1 as bait in a yeast two-hybrid screen, found elevated levels of this protein in the nucleus under conditions of heat shock or ER stress induced by tunicamycin treatment [72].
HtrA3 was discovered initially as a pregnancy-related serine protease (PRSP) identified by DDPCR (Differential Display Polymerase Chain Reaction) to be differentially expressed between implantation and interimplantation sites in the mouse uterus [87]. This protein shares high homology with HtrA1 (56% amino acid similarity), and high levels of its expression were detected in the heart, ovary, testis, pregnant uterus and placenta in the mouse [87]. Mammalian HtrA1 and 3 share identical domain organization including an N-terminal signal sequence (SS), an insulin growth factor binding domain (IGFBP) and a kazal-type S protease inhibitor domain (KI) [73].
The cDNA sequence of the fourth member of the HtrA family, named HtrA4, has been also cloned (GenBank accession no. AK075205) and is mapped to 8p12. No biochemical characterization has yet been reported for this protein, but the high homology in structure and domains with HtrA1 suggests some specific expression of this protein in the placenta [75].
HtrA proteases and cancer
Alterations of HTRA1 in cancer
Meta-analyses of publicly available microarray data from Oncomine.org indicate that HtrA1 is down-regulated and shows allelic imbalance in cancer of diverse origins. Z-score normalized expression values for HtrA proteases are downloaded from Oncomine website [88], and statistical significance between two groups was analyzed by Student’s two-tailed t-test. High-resolution genomic survey of brain tumors by Kotliarov et al indicates allelic loss associated with HtrA1 in glioblastoma compared to astrocytoma, mixed oligoastrocytoma, and oliodendroglioma (Table 1, Ref. 97). HtrA1 expression is also downregulated in medulloblastoma compared to normal cerebellum, atypical teratoid/rhabdoid tumor, or primitive neuroectodermal tumor (Table 1, Ref. 98). HtrA1 expression is down-regulated in tumor stem cells derived from glioblastoma cultured in bFGF and EGF compared to serum-cultured cell lines (Table 1, Ref. 99). In addition, alterations in HtrA1 are also associated with specific tumor behavior. For example, greater allelic loss of HtrA1 in glioma is associated with poor prognosis and with increasing grade of tumors (Table 1, Ref. 97).
Table 1.
Analysis of HTRA1 alterations in cancer.
| Tumor Type | Comparison | Z-score Normalized Expression |
95% CI | p-value |
|---|---|---|---|---|
| Brain | Glioblastoma | 0.5332 | 0.4274:0.6390 | |
| (Kotliarov et al) [97] | Oligodendroglioma Malignant | 0.9292 | 0.7889:1.069 | |
| Oligoastrocytoma Astrocytoma | 0.7135 | 0.3916:1.035 | ||
| 0.9038 | 0.7378:1.07 | P<0.0001 | ||
| Brain | Normal cerebellum | 1.193 | 0.6092:1.776 | |
| (Pomeroy et al) [98] | Atypical Teratoid/Rhabdoid | 0.9014 | 0.3017:1.501 | |
| Malignant Glioblastoma Primitive | 1.75 | 1.548:1.951 | ||
| Neuroectodermal Medulloblastoma | 1.067 | 0.3858:1.748 | ||
| 0.6652 | 0.5134:0.8170 | P<0.0001 | ||
| Brain | Serum | 2.466 | 2.418:2.513 | |
| (Lee et al) [99] | Serum-free | 1.941 | 1.888:1.995 | P<0.0001 |
| Brain | GBM, >5y survival | 0.04186 | −0.1011:0.1848 | |
| (Kotliarov et al) [97] | GBM, 3–5y survival | −0.04538 | −0.2628:0.172 | |
| GBM, 1–3y survival | −0.253 | −0.3765:−0.1295 | ||
| GBM, <1y survival | −0.4041 | −0.5003:−0.3080 | P<0.0001 | |
| Brain | Grade 2 | 0.9897 | 0.8675:1.112 | |
| (Kotliarov et al) [97] | Grade 3 | 0.7385 | 0.5705:0.9066 | |
| Grade 4 | 0.5332 | 0.4274:0.6390 | P<0.0001 | |
| Ovarian | Normal Ovary | 2.583 | 2.559:2.607 | |
| (Hendrix et al) [100] | Ovarian Cancer-serous | 1.363 | 1.193:1.532 | |
| Ovarian Cancer-endometrioid | 1.506 | 1.314–1.697 | P=0.0012 | |
| Ovarian | Normal Ovary | 2.021 | 1.863:2.179 | |
| (Welsh et al) [101] | Ovarian Cancer | 1.203 | 1.059:1.346 | P=0.0034 |
| Ovarian | Normal Ovary | 1.955 | 1.677:2.233 | |
| (Lu et al) [102] | Ovarian Cancer | 1.143 | 0.9547:1.332 | P=0.0014 |
| Ovarian | Normal Ovary | 2.493 | 2.320:2.667 | |
| (Adib et al) [103] | Primary Ovarian Cancer | 1.679 | 1.274:2.084 | |
| Ovarian Cancer Metastasis | 1.863 | 1.496:2.231 | P=0.0060 | |
| Head&Neck | Normal oral mucosa | 1.06 | 0.9870:1.133 | |
| (Gino et al) [104] | Squamous cell carcinoma | 1.325 | 1.226:1.423 | P=0.0018 |
| Tongue | Normal tongue | 1.042 | 0.8885:1.195 | |
| (Talbot et al) [105] | Squamous cell carcinoma | 1.585 | 1.429:1.740 | P<0.0001 |
| Lung | Lung adenocarcinoma | 0.9551 | 0.8863:1.024 | |
| (Gordon et al) [106] | Malignant Pl. Mesothelioma | 1.726 | 1.571:1.881 | P<0.0001 |
| Breast | Apocrine tumor Basal | 1.684 | 1.462:1.907 | |
| (Farmer et al) [107] | tumor Luminal tumor | 1.446 | 1.305:1.588 | |
| 1.83 | 1.745:1.916 | P<0.0001 | ||
| Breast | ER+ ER− | 1.652 | 1.607:1.697 | |
| (Wang et al) [108] | 1.413 | 1.329:1.497 | P<0.0001 | |
| Breast | ER+ ER− | 1.539 | 1.477:1.601 | |
| (Desmedt et al) [109] | 1.276 | 1.185:1.367 | P<0.0001 | |
| Breast | ER+ ER− | −0.1226 | −0.3459:0.1008 | |
| (vandeVijver) [110] | −1.234 | −1.669:−0.7998 | P<0.0001 | |
| Breast | ER+ ER− | 2.313 | 2.218:2.408 | |
| (Chin et al) [111] | 1.922 | 1.763:2.082 | P<0.0001 | |
| Breast | Lymphocytic infiltration neg | −0.06162 | −0.3218:0.1986 | |
| (vant'Veer et al) [112] | Lymphocytic infiltration pos | −2.238 | −2.805:−1.670 | P<0.0001 |
| Breast | Grade 1 | 1.786 | 1.718:1.853 | |
| (Ivshina et al) [113] | Grade 2 | 1.662 | 1.606:1.719 | |
| Grade 3 | 1.471 | 1.381:1.560 | P<0.0001 | |
| Breast | Grade 1 | 1.248 | 1.206:1.291 | |
| (Miller et al) [114] | Grade 2 | 1.17 | 1.136:1.204 | |
| Grade 3 | 1.055 | 0.9996:1.110 | P<0.0001 | |
| Breast | Grade 1 | 0.4186:1.307 | 0.8626 | |
| (vant'Veer et al) [112] | Grade 2 | 0.1831 | −0.2182:0.5844 | |
| Grade 3 | −1.07 | −1.434:−0.7050 | P<0.0001 | |
| Breast | Grade 1 | 1.771 | 1.717:1.825 | |
| (Sotiriou et al) [115] | Grade 2 | 1.599 | 1.515:1.684 | |
| Grade 3 | 1.543 | 1.463:1.623 | P<0.0001 | |
| Breast | Grade 1 | 1.583 | 1.487:1.679 | |
| (Desmedt et al) [109] | Grade 2 | 1.554 | 1.477:1.631 | |
| Grade 3 | 1.309 | 1.221:1.396 | P<0.0001 | |
| Breast | p53 wildtype p53 | 1.21 | 1.183:1.237 | |
| (Ivshina et al) [113] | mutant | 1.055 | 1.005:1.105 | P<0.0001 |
| Breast | p53 wildtype p53 | 1.711 | 1.668:1.754 | |
| (Miller et al) [114] | mutant | 1.472 | 1.377:1.568 | P<0.0001 |
| Leukemia | B-cell acute lymphoblastic | 0.1766 | −0.0524:0.4056 | |
| (Raetz et al) [116] | T-cell acute lymphoblastic | 0.03617 | −0.2615:0.3339 | |
| T-cell lymphoblastic lymphoma | 0.7623 | 0.5628:0.9617 | P=0.0002 | |
| Leukemia | Precursor-B ALL (Remission) | 0.279 | 0.05617:0.5019 | |
| (Mullighan et al) [117] | Precursor-B ALL (ETV6-RUNX1 positive) | −0.6435 | −0.8601:−0.4269 | P<0.0001 |
| Leukemia | B-cell ALL (Remission) B-cell | 0.3381 | 0.08658:0.5895 | |
| (Mullighan et al) [117] | ALL (B-cell ALL with Pseudodiploidy) | −0.8315 | −1.143:−0.5199 | P=0.0005 |
| Prostate | Normal prostate | 1.191 | 1.022:1.360 | |
| (Welsh et al) [118] | Prostate carcinoma | 0.7086 | 0.6041:0.8132 | P=0.0001 |
| Prostate | Prostate cancer | 0.0925 | −0.0039:0.1889 | |
| (Holzbeierlein et al) [119] | Post neoadjuvant therapy | 0.451 | 0.3350:0.5671 | P<0.0001 |
| Prostate | Normal prostate Prostate | 0.6178 | 0.4904:0.7451 | |
| (Yu et al) [120] | carcinoma Metastatic prostate | 0.4366 | 0.3794:0.4938 | |
| cancer | 0.2052 | 0.1037:0.3067 | P<0.0001 | |
| Prostate | Benign prostate | 1.383 | 1.266:1.499 | |
| (Varambally et al) [121] | Prostate carcinoma | 1.191 | 1.104:1.278 | |
| Hormone-refractory metastatic | 0.8841 | 0.6830:1.085 | P<0.0001 | |
| Sarcoma | Primary | 1.755 | 1.554:1.957 | |
| (Segal et al) [122] | Metastatic | 0.96 9 | 0.7547:1.179 | P<0.0001 |
| Melanoma | Normal skin | 1.777 | 1.575:1.979 | |
| (Talantov et al) [123] | Benign nevus | 1.247 | 1.124:1.370 | |
| Melanoma | 0.8427 | 0.7484:0.9370 | P<0.0001 | |
| Melanoma | Disomy 3 | 0.1371 | −0.1139:0.3881 | |
| (Tschentscher et al) [124] | Monosomy 3 | 1.113 | 0.8287:1.398 | P=0.0002 |
| Kidney | Normal fetal kidney Clear | 1.069 | 0.5929:1.544 | |
| (Cutcliffe et al) [125] | cell sarcoma Wilms tumor | 1.57 | 1.258:1.883 | |
| 0.7488 | 0.6710:0.8266 | P<0.0001 | ||
| Bladder | Normal bladder | 1.534 | 1.466:1.603 | |
| (Sanchez et al) [126] | Bladder carcinoma | 1.266 | 1.176:1.356 | P=0.0001 |
Significant value in each comparison is highlighted by bold.
Several studies indicate that HtrA1 expression is also down-regulated in epithelial ovarian tumors and metastases compared to normal ovary or normal ovarian surface epithelium (Table 1, Refs. 100–103). Consistent with these results, expression analyses by our research group using real-time PCR, RT-PCR, Northern blot analysis, Western blot analysis, and immunohistochemical analysis of tissue microarrays all indicate that HtrA1 is down-regulated in ovarian cancer of different histologies [64, 89]. Moreover, we also identified epigenetic silencing and allelic loss as potential mechanisms associated with HtrA1 down-regulation in ovarian cancer [89]. We also showed that HtrA1 expression is regulated by chemotherapeutic drugs. Most importantly, we showed that expression of HtrA1 primary tumors correlates with better response to cisplatin-based chemotherapy in ovarian cancer and gastric cancer [64]. HtrA1 is activated during drug treatment in vitro, and active HtrA1 increases caspase 3/7 activity and participates in chemotherapy-induced cytotoxicity [64].
In breast cancer, several studies indicate that HtrA1 expression is lower in ER-negative tumors compared to ER-positive tumors and that down-regulation of HtrA1 is significantly correlated with higher Elston grade of breast carcinoma (Table 1, Refs. 108–111, 112–115, 109). Moreover, poorly differentiated breast tumors and those with mutant p53 or with lymphocytic infiltration have significantly lower levels of HtrA1 expression (Table 1, Ref. 112).
Microarray studies indicate that HtrA1 is down-regulated in prostate cancer compared to normal controls (Table 1, Ref. 118). In detail, HtrA1 appear to be down-regulated in metastatic prostate cancer compared to localized prostate cancer or normal controls and in hormone-refractory metastatic prostate cancer compared to prostate carcinoma or benign prostate (Table 1, Refs. 118, 120–121) but is upregulated in prostate tumors post neoadjuvant therapy compared to prostate tumors that did not receive neoadjuvant therapy (Table 1, Ref. 119).
HtrA1 is also down-regulated in malignant melanoma compared to benign melanocytic lesions or normal controls (Table 1, Ref. 123). These results are consistent with our studies indicating that HtrA1 is down-regulated in metastatic melanoma lesions compared to primary melanoma lesions from the same patients [90]. Moreover, ectopic expression of HtrA1 in melanoma cell lines inhibited cell growth [90].
In hematologic malignancies, genome-wide analysis of genetic alterations in acute lymphoblastic leukemia indicates that ETV6-RUNX1 positive precursor-B ALL in remission and B-ALL with Pseudodiploidy in remission have higher levels of HtrA1 compared to ETV6-RUNX1 Positive Precursor-B ALL and B-ALL with Pseudodiploidy without remission (Table 1, Ref. 117). Finally, HtrA1 is also down-regulated in metastatic sarcoma compared to primary sarcoma (Table 1, Ref. 122). Interestingly, recent data from our research group indicates that HtrA1 is down-regulated in lung carcinomas and in mesotheliomas [91, 92].
Collectively, these results highlight the important role of HtrA1 down-regulation in the cancer progression and in the acquisition of aggressive tumor traits in tumors of diverse origins, and provide a rationale for targeting this protein for cancer therapy due to its pro-apoptotic, anti-growth properties and its regulation by therapeutic drugs.
Alterations of HtrA2/Omi in cancer
HtrA2 expression in tumors is somewhat variable according to tumor types. For example, HtrA2 is upregulated in lung adenocarcinoma, superficial or invasive transitional cell carcinoma, and in oligodendroglioma compared to normal controls (Table 2, Refs. 127–128). HtrA2 is also upregulated in squamous cell carcinoma of head and neck compared to normal oral mucosa (Table 2, Ref. 104). Interestingly, HtrA2 is down-regulated in chronic lymphocytic leukemia (CLL) samples compared to normal B-cells (Table 2, Ref. 132). In contrast, studies by Andersson et al indicate that HtrA2 is upregulated in B-Cell Acute Lymphoblastic Leukemia and T-Cell Acute Lymphoblastic Leukemia compared to Acute Myeloid Leukemia (Table 2, Ref. 133), suggesting differential expression of HtrA1 in different types of leukemia. HtrA2 is also upregulated in Wilm’s tumors compared to normal fetal kidney or clear cell sarcoma (Table 2, Ref. 125). On the other hand, HtrA2 is down-regulated in renal carcinoma treated with mTOR inhibitor CCI-779 (8 weeks following treatment) compared to untreated renal carcinoma (Table 2, Ref. 134). When comparison was made between microsatellite stable and instable colorectal carcinoma, HtrA2 is upregulated in microsatellite instable colorectal carcinoma compared to the former group (Table 2, Ref. 136). However, in ovarian cancer, HtrA2 is consistently down-regulated in ovarian tumor of different histologies (Table 2, Ref. 100). Two independent validation analyses of gene expression in breast cancer by Sorlie et al indicate that HtrA2 expression is down-regulated with increasing tumor staging (Table 2, Refs.137–138). HtrA2 is down- regulated in lymphoma positive for Ig-Myc translocation compared to Myc fusion-negative lymphoma (Table 2, Ref. 139). In the study by Dhanasekaran et al, HtrA2 is down-regulated in metastatic prostate cancer compared to primary prostate cancer or normal prostate or benign prostate (Table 2, Ref. 140). Finally, in studies by Korkola et al, HtrA2 expression is down-regulated adult male germ cell tumor compared to normal testis (Table 2, Ref. 141). Collectively, the results from these studies indicate that unlike HtrA1 expression in tumors, expression of HtrA2 is variable, upregulated in some tumor types and downregulated in others.
Table 2.
Analysis of HTRA2 alterations in cancer.
| Tumor Type | Comparison | Z-score Normalized Expression |
95% CI | p-value | |
|---|---|---|---|---|---|
| Lung | Normal lung | 0.5791 | 0.5250:0.6332 | ||
| (Stearman et al) [127] | Lung adenocarcinoma | 0.8055 | 0.7514:0.8595 | P<0.0001 | |
| Lung | Carcinoid | 0.9593 | 0.8339:1.085 | ||
| (Bhattacharjee et al) [128] | Lung adenocarcinoma | 0.9483 | 0.8939:1.003 | ||
| Small cell lung cancer | 1.178 | 0.9053:1.451 | |||
| Squamous cell lung cancer | 0.6416 | 0.5254:0.7579 | P<0.0001 | ||
| Bladder | Normal bladder | 0.5172 | 0.4563:0.5781 | ||
| (Dyrskjot et al) [129] | Superficial carcinoma | 0.7156 | 0.6084:0.8228 | ||
| Invasive carcinoma | 0.8069 | 0.6853:0.9284 | P=0.0041 | ||
| Bladder | Superficial carcinoma | 0.1761 | 0.1165:0.2357 | ||
| (Stransky et al) [130] | Invasive carcinoma | 0.4017 | 0.3310:0.4723 | P<0.0001 | |
| Brain | Normal brain | 0.9783 | 0.9350:1.022 | ||
| (Sun et al) [131] | Oligodendroglioma | 1.116 | 1.073:1.159 | P<0.0001 | |
| Head&Neck | Normal oral mucosa | 0.1767 | 0.04534:0.3080 | ||
| (Ginos et al) [104] | Squamous cell carcinoma | 0.5354 | 0.4769:0.5940 | P<0.0001 | |
| Leukemia | Normal B-cells | 0.8633 | 0.7867:0.9399 | ||
| (Haslinger et al) [132] | Chronic Lymphocytic | 0.5815 | 0.5457:0.6174 | P<0.0001 | |
| Leukemia | Acute Myeloid | −0.5786 | −0.8323:−0.3249 | ||
| (Andersson et al) [133] | B-cell acute lymphoblastic | 0.1679 | −0.03911:0.3748 | ||
| T-cell acute lymphoblastic | 0.2809 | −0.1135:0.6753 | P<0.0001 | ||
| Sarcoma | Clear cell sarcoma of kidney | 0.2173 | 0.1059:0.3288 | ||
| (Cutcliffe et al) [125] | Wilms tumor | 0.5851 | 0.5104:0.6598 | P<0.0001 | |
| Kidney | Renal cell carcinoma (0wk) | 1.053 | 0.9978:1.108 | ||
| (Boni et al) [134] | Renal cell carcinoma (8wk) | 0.8908 | 0.8443:0.9374 | P<0.0001 | |
| Renal cell carcinoma (16wk) | 0.9413 | 0.8739:1.009 | |||
| Breast | GFP-transfected control | 0.3907 | 0.2868:0.4945 | ||
| (Bild et al) [135] | H-Ras transfected | 0.7573 | 0.7150:0.7995 | P<0.0001 | |
| (Human mammary epithelial cells) | |||||
| Colon | Stable | 0.9233 | 0.8591:0.9875 | ||
| (Watanabe et al) [136] | Instable | 1.154 | 1.072:1.235 | P<0.0001 | |
| (Microsattelite status) | |||||
| Ovary | Normal ovary | 0.7325 | 0.6578:0.8071 | ||
| (Hendrix et al) [100] | Ovarian cancer -endometrioid | 0.3219 | 0.2557:0.3882 | P=0.0023 | |
| Ovarian cancer -mucinous | 0.2262 | 0.1394:0.3130 | P=0.0039 | ||
| Ovarian cancer -serous | 0.4153 | 0.3508:0.4799 | P=0.0034 | ||
| Ovarian cancer -clear cell | 0.1361 | −0.0295:0.3016 | P=0.0040 | ||
| Breast | Stage 1 | 0.789 | 0.5009:1.077 | ||
| (Sorlie et al) [137] | Stage 2 | 0.5419 | 0.1423:0.9416 | ||
| Stage 3 | −0.06275 | −0.3027:0.1771 | |||
| Stage 4 | −0.5341 | −0.7977:−0.2718 | P<0.0001 | ||
| Breast | Stage 1 | 0.8231 | 0.5382:1.108 | ||
| (Sorlie et al) [138] | Stage 2 | 0.5781 | 0.3016:0.8558 | ||
| Stage 3 | 0.2434 | 0.08463:0.4022 | |||
| Stage 4 | −0.0657 | −0.2724:0.1410 | P=0002 | ||
| Lymphoma | Negative | 1.175 | 1.143:1.207 | ||
| (Hummel et al) [139] | Ig-Myc | 1.024 | 0.9802:1.067 | P<0.0001 | |
| (Myc Fusion) | |||||
| Prostate | Normal or BPH | 1.181 | 0.8113:1.551 | ||
| (Dhanasekaran et al) [140] | Primary prostate cancer | 0.5549 | 0.3905:0.7194 | ||
| Metastatic prostate cancer | −0.1939 | −0.3891:0.00138 | P<0.0001 | ||
| Germ cell | Normal testis | 1.113 | 1.065:1.161 | ||
| (Korkola et al) [141] | Adult seminoma | 0.9188 | 0.8698:0.9678 | P=0.0057 | |
Significant value in each comparison is highlighted by bold.
Expression analyses of HtrA3 in cancer
Similar to HtrA2, expression of HtrA3 is variable accordin to the tumor type. Studies by Hao et al indicate that HtrA3 expression is upregulated in esophageal adenocarcinoma compared to normal or Barretts esophagus (Table 3, Ref. 142). Similarly, studies by Iacobuzio-Donahue et al indicate that HtrA3 upregulated in pancreatic adenocarcinoma compared to normal pancreas (Table 3, Ref. 143). Studies by Korkola et al also indicate upregulation of HtrA3 in seminoma compared to normal testis (Table 3, Ref. 141). In contrast, HtrA3 expression is variable in hematologic malignancies depending on specific molecular alterations. For example, studies by Andersson et al indicate that HtrA3 expression is down-regulated in B-Cell Acute Lymphoblastic Leukemia, T-Cell Acute Lymphoblastic Leukemia, and Acute Myeloid Leukemia compared to normal bone marrow (Table 3, Ref. 133). However, in Acute Lymphoblastic Leukemia, studies by Fine et al indicate increased expression of HtrA3 in leukemia positive for MLL/AF4 translocation compared to leukemia positive for BCR/ABL or TEL/AML1 translocation (Table 3, Ref. 145). Studies by Andersson et al also indicate that HtrA3 is overexpressed in acute lymphoblastic leukemia (ALL) positive for 11q23/MLL or TCF3/PBX1 translocation compared to normal control or ALL with 47–50 chromosomes, BCR/ABL1, Hyperdiploid Greater Than 50, IGH/MYC, or TEL/AML (Table 3, Ref133). In Acute Myeloid Leukemia, studies by Heuser et al indicate that HtrA3 is down-regulated malignancies positive for partial tandem duplication of MLL compared to MLL mutation-negative malignancies (Table 3, Ref. 146). In the study by Fine et al, HtrA3 is upregulated in pro-B acute lymphoblastic leukemia (ALL) compared to Common or Mixed Lineage ALL or Pre-B ALL (Table 3, Ref. 145). Finally, in the study by Schmidt et al, HtrA3 is upregulated in ALL with hyperploidy greater than 50 compared to normal controls or ALL with low hyperploidy, TEL/AML, or t(8:14)(q24:11) (Table 3, Ref. 147). These results highlight the tumor type-specific variation in HtrA3 expression, and as such point to the need to better understand the underlying molecular alterations in cancer before they can be targeted by novel therapeutics.
Table 3.
Analysis of HTRA3 alterations in cancer
| Tumor Type | Comparison | Z-score Normalized Expression |
95% CI | p-value | |
|---|---|---|---|---|---|
| Esophagus | Normal esophagus | −1.896 | −2.426:−1.366 | ||
| (Hao et al) [142] | Barretts esophagus | −1.152 | −1.773:−0.5303 | ||
| Adenocarcinoma | 0.9189 | 0.6080:1.230 | P<0.0001 | ||
| Pancreas | Normal | 0.2267 | −0.5146:0.9681 | ||
| (Iacobuzio-Donahue et al) [143] | Pancreatic cancer | 2.606 | 1.822:3.391 | P=0.0019 | |
| Germ cell | Normal testis | 0.2704 | 0.1583:0.3826 | ||
| (Korkola et al) [141] | Seminoma | 0.5982 | 0.5418:0.6546 | P=0.0008 | |
| Leukemia | Normal bone marrow | 2.478 | 1.958:2.997 | ||
| (Andersson et al) [133] | B-cell ALL | −0.3214 | −0.820:0.177 | ||
| T-cell ALL | −1.843 | −2.814:−0.8715 | |||
| AML | 0.3605 | −0.4062:1.127 | P=0.0008 | ||
| Leukemia | Normal | 0.285 | −0.061:0.631 | ||
| (Ross et al) [144] | BCR-ABL | 0.7115 | 0.529:0.893 | ||
| Hyperdiploid | 0.5671 | 0.410:0.723 | |||
| MLL | 1.228 | 0.9728:1.484 | |||
| Pseudodiploid | 0.6376 | 0.2539:0.9549 | |||
| T Lineage | 0.384 | 0.2539:0.5142 | |||
| TEL-AML | 0.3475 | 0.1059:0.5892 | |||
| E2A-PBX | 1.36 | 1.241:1.479 | P<0.0001 | ||
| Leukemia | BCR/ABL | −1.123 | −1.68:−0.562 | ||
| (Fine et al) [145] | TEL/AML | −1.859 | −2.320:−1.398 | ||
| MLL/AF4 | 3.239 | 2.239:4.239 | P<0.0001 | ||
| Leukemia | Normal | −1.062 | −1.97:−0.149 | ||
| (Andersson et al) [144] | 11q23/MLL | 4.146 | 1.708:6.585 | ||
| 47–50 Chr | −0.3508 | −4.227:3.525 | |||
| BCR/ABL1 | 0.09373 | −4.971:5.158 | |||
| >50 Chr | −0.7515 | −1.537:0.03378 | |||
| IgH/Myc | −2.335 | −3.186:−1.484 | |||
| TEL/AML | −1.106 | −1.980:−0.2318 | |||
| TCF/PBX1 | 3.161 | 2.344:3.978 | P<0.0001 | ||
| Leukemia | Negative | 0.09801 | −0.1550:0.3511 | ||
| (Heuser et al) [146] | Partial tandem dup | −0.7226 | −0.9707:−0.4745 | ||
| P=0.0012 (MLL mutation) | |||||
| Leukemia | Common | −1.493 | −1.968:−1.019 | ||
| (Fine et al) [145] | Mixed | −1.11 | −6.258:4.038 | ||
| Pre | −0.9608 | −3.202:1.280 | |||
| Pro | 3.248 | 2.125:4.370 | P<0.0001 | ||
| (Immunophenotype) | |||||
| Leukemia | Normal | −0.08158 | −0.188:0.025 | ||
| (Schmidt et al) [147] | Low hyperploid | −0.03072 | −0.147:0.0855 | ||
| TEL/AML | −0.1678 | −0.241:−0.0939 | |||
| t(8:14)(q24:11) | −0.1595 | −0.242:−0.0765 | |||
| >50 Chr | 0.1393 | 0.07798:0.2006 | P=0.0013 | ||
| Breast | Negative | −0.3053 | −0.5682:−0.04247 | ||
| (vant'Veer et al) [112] | Positive | −1.416 | −1.747:−1.084 | P<0.0001 | |
| (Lymphocytic infiltration) | |||||
| Brain | Glioblastoma | −0.2522 | −0.3295:−0.1750 | P<0.0001 | |
| (Maser et al) [148] | (DNA copy) | ||||
Significant value in each comparison is highlighted by bold.
Alterations of HtrA4 in cancer
Analysis of microarray studies by Sun et al in brain tumors indicates that HtrA4 is upregulated in glioblastoma multiforme compared to control brain from epilepsy patients (Table 4, Ref. 131). Similarly, studies by Richardson et al indicate HtrA4 is upregulated in breast carcinoma compared to normal breast samples (Table 4, Ref. 149). Studies by Varambally et al indicate that HtrA4 is down-regulated in hormone refractory metastatic prostate cancer compared to primary prostate carcinoma (Table 4, Ref. 121). Finally, studies by Maser et al indicate that allelic ratio of HtrA4 is lower in glioblastoma and pancreatic cancer compared to normal DNA control (Table 4, Ref. 148). It should be noted however that these genomic analyses do not indicate specific loss of HtrA4 alleles. Rather, these results indicate a large region of allelic loss in pancreatic tumors. As such, loss of HtrA4 alleles in pancreatic cancer could have been the result of secondary deletion.
Table 4.
Analysis of HTRA4 alterations in cancer.
| Tumor Type | Comparison | Z-score Normalized Expression |
95% CI | p-value |
|---|---|---|---|---|
| Brain | Normal | −0.6262 | −0.7830:−0.4694 | |
| (Sun et al) [131] | Glioblastoma | −0.2154 | −0.2858:−0.1450 | P<0.0001 |
| Breast | Normal | −0.8074 | −0.9453:−0.6695 | |
| (Richardson et al) [149] | Breast carcinoma | −0.4429 | −0.5569:−0.3289 | P=0.0004 |
| Prostate | Primary | −0.6017 | −0.9403:−0.2630 | |
| (Varambally et al) [121] | Hormone-refractory | −1.574 | −2.140:−1.009 | P=0.0082 |
| Brain | Glioblastoma | −0.272 | −0.3410:−0.2029 | P<0.0001 |
| (Maser et al) [148] |
Significant value in each comparison is highlighted by bold.
Role of HtrA proteins in programmed cell death and chemotherapy-induced cytotoxicity
Studies by Bartling et al suggest that Smac/Diablo released from the mitochondria of lung cancer cells during etoposide treatment was not sufficient to induce apoptosis and may require additional mitochondrial factors such as HtrA2/Omi [93], highlighting the significance of other mitochondrial proteins, such as HtrA2/Omi, in efficienct execution of programmed cell death. Cilenti et al reported that HtrA2/Omi expression is regulated by cisplatin in renal cell lines and primary proximal tubule cells [61]. Cisplatin treatment upregulates HtrA2/Omi expression and sensitize cells the cisplatin. RNAi mediated down-regulation of HtrA2/Omi attenuated cisplatin-induced cytotoxicity. These results suggest a role of serine protease HtrA2/Omi in cisplatin- induced cytotoxicity. Studies by our laboratories also indicate that HtrA1 expression is regulated by chemotherapy [64]. HtrA1 expression is upregulated in ovarian cancer cells following cisplatin or paclitaxel treatment. Moreover, upregulation of HtrA1 resulted in autocatalytic activation of HtrA1 by removal of N-terminal regulatory domain within HtrA1. Active HtrA1 increases caspase 3/7 activity and induces cell death. Consistent with its pro-apoptotic property of HtrA1, down-regulation of HtrA1 in ovarian cancer cells by RNAi resulted in attenuation of chemotherapy-induced cytotoxicity whereas ectopic expression of HtrA1 in null cells resulted in increased sensitivity to chemotherapy-induced cell death [64]. Moreover, higher levels of HtrA1 expression in ovarian and gastric cancer is associated with better response to cisplatin-based chemotherapy in ovarian or gastric cancer patients, highlighting the potential therapeutic value of HtrA1 expression in these cancers [64]. Finally, gene expression analyses by Folgueira et al indicate that gene expression classifier that includes HtrA1 expression is associated with better response to doxorubicin-based therapy in breast cancer [94].
Epigenetic therapies and re-activated HtrA proteins as biomarkers for drug response
Epigenetic therapies targeting reactivation of pro-apoptotic proteins and tumor suppressors that are down-regulated in cancer represent a potential therapeutic strategy to overcome the problem of resistance to chemotherapy or to enhance the efficacy of conventional chemotherapeutic agents [33, 37, 39, 40, 95, 96]. Previous studies indicate that HtrA1 expression may be epigenetically regulated [89]. In ovarian cancer cell line with no expression of HtrA1, treatment with 5-aza cytidine resulted in re- expression of HtrA1 [89]. Our recent unpublished data indicate that HtrA serine proteases contain putative CpG islands in their promoter and exon 1 regions (Fig 1). Furthermore, promoter sequence analysis of bisulfite-modified DNA in ovarian cancer cells indicates that some CpG sites are differentially methylated in non-expressing cells compared to HtrA1 expressing cells (Fig 2A). Consistent with epigenetic as a mechanism of HtrA1 downregulation in cancer, pre-treatment of non-expressing cells with DNA methyltransferase inhibitor, 5-aza-2’-deoxycytidine (DAC) or histone deacetylase inhibitor (LBH589), resulted in re-activation of HtrA1 expression in initially non-expressing cells (Fig 2B). These results suggest that HtrA1 expression is epigenetically controlled in ovarian cancer. Similar analyses in other cancers are underway in our laboratories, and these studies should provide detailed understanding of epigenetic regulation of HtrA1 expression in cancer.
Figure 1.

Genomic sequence analysis of the promoter and exon 1 of HTRA genes for the presence of putative CpG islands. Each analysis contains 1000 bases of promoter and entire exon 1. Emboss CpGPlot web-based program with default settings was used to identify putative CpG islands in HTRA genes. Schematic representation of CpG sites, indicated by vertical lines, was obtained from CpG Island Searcher web program.
Figure 2.

DNA sequence analyses of bisulfite-modified genomic DNA corresponding to a putative CpG island in HTRA1 promoter. A) CpG sites are methylated in HtrA1-deficient A2780, but not methylated in HtrA1-expressing SKOV3 cells. Methylated CpG sites (underlined in A2780 sample) were resistant to bisulfite conversion and remained as Cs whereas unmethylated CpG sites (underlined in SKOV3 sample) were converted to Ts. In addition, two sinle nucleotide polymorphisms were indicated by arrowheads. B) HtrA1 expression is re-activated wit 5-aza-2’-deoxycytidine (DAC) or LBH589 treatments in two non-expressing cell lines.
Since HtrA serine proteases are variably down-regulated in specific cancer types by epigenetic mechanisms, they represent potential targets for epigenetic modulation. Future clinical trials involving the evaluation of epigenetic therapies should use epigenetic modification of these genes as therapeutic markers to assess the biological response of epigenetic therapies and should evaluate their prognostic potentials to predict tumor response to combined epigenetic and conventional chemotherapy.
Future approaches
Due to their potential interaction with caspase-mediated cell death pathways, HtrA serine protease pathway represents a potential target of therapeutic intervention. Future studies investigating the cross-talks between caspases and HtrA serine proteases are necessary to fully explore the therapeutic potential of this novel serine protease pathway. In particular, it will be of significant clinical interest in determining the mechanisms by which HtrA expression is regulated by chemotherapy. Detailed understanding of the mechanisms by which HtrA expression and protease activity are regulated is critical before this pathway can be targeted for therapeutic advantage. In addition, the identification of substrates of these proteases will also be essential in gaining insights into how best to target this novel pathway. It is our hope that this review will spark a new interest in taking a fresh look at this novel pathway by researchers of diverse cancer fields, and that combined effort will eventually lead to better and novel approaches to target cancer cells.
Acknowledgements
This work was supported by grants from FUTURA-onlus, Ministry of Health and Second University of Naples (to AB), NIH grant 1R01CA123249 (to VS and JC), and Ovarian Cancer Research Fund (to JC).
References
- 1.Fulda S, Debatin KM. Extrinsic versus intrinsic apoptosis pathways in anticancer chemotherapy. Oncogene. 2006;25(34):4798–4811. doi: 10.1038/sj.onc.1209608. [DOI] [PubMed] [Google Scholar]
- 2.Meiler J, Schuler M. Therapeutic targeting of apoptotic pathways in cancer. Curr Drug Targets. 2006;7(10):1361–1369. doi: 10.2174/138945006778559175. [DOI] [PubMed] [Google Scholar]
- 3.Nelson EC, et al. Biologic agents as adjunctive therapy for prostate cancer: a rationale for use with androgen deprivation. Nat Clin Pract Urol. 2007;4(2):82–94. doi: 10.1038/ncpuro0700. [DOI] [PubMed] [Google Scholar]
- 4.Kaufmann SH, Vaux DL. Alterations in the apoptotic machinery and their potential role in anticancer drug resistance. Oncogene. 2003;22(47):7414–7430. doi: 10.1038/sj.onc.1206945. [DOI] [PubMed] [Google Scholar]
- 5.Danial NN, Korsmeyer SJ. Cell death: critical control points. Cell. 2004;116(2):205–219. doi: 10.1016/s0092-8674(04)00046-7. [DOI] [PubMed] [Google Scholar]
- 6.Brown JM, Wouters BG. Apoptosis: mediator or mode of cell killing by anticancer agents? Drug Resist Updat. 2001;4(2):135–136. doi: 10.1054/drup.2001.0193. [DOI] [PubMed] [Google Scholar]
- 7.Brown JM, Wouters BG. Apoptosis, p53, and tumor cell sensitivity to anticancer agents. Cancer Res. 1999;59(7):1391–1399. [PubMed] [Google Scholar]
- 8.Schmitt CA, Lowe SW. Apoptosis is critical for drug response in vivo. Drug Resist Updat. 2001;4(2):132–134. doi: 10.1054/drup.2001.0188. [DOI] [PubMed] [Google Scholar]
- 9.Schmitt CA, Lowe SW. Apoptosis and chemoresistance in transgenic cancer models. J Mol Med. 2002;80(3):137–146. doi: 10.1007/s00109-001-0293-3. [DOI] [PubMed] [Google Scholar]
- 10.Reed JC. Dysregulation of Apoptosis in Cancer. Journal of Clinical Oncology. 1999;17(9):2941–2953. doi: 10.1200/JCO.1999.17.9.2941. [DOI] [PubMed] [Google Scholar]
- 11.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]
- 12.Kaufmann SH, Hengartner MO. Programmed cell death: alive and well in the new millennium. Trends Cell Biol. 2001;11(12):526–534. doi: 10.1016/s0962-8924(01)02173-0. [DOI] [PubMed] [Google Scholar]
- 13.Jiang N, et al. Advances in mitotic inhibitors for cancer treatment. Mini Rev Med Chem. 2006;6(8):885–895. doi: 10.2174/138955706777934955. [DOI] [PubMed] [Google Scholar]
- 14.Epstein RJ. Drug-induced DNA damage and tumor chemosensitivity. J Clin Oncol. 1990;8(12):2062–2084. doi: 10.1200/JCO.1990.8.12.2062. [DOI] [PubMed] [Google Scholar]
- 15.Schmidt M, Bastians H. Mitotic drug targets and the development of novel anti-mitotic anticancer drugs. Drug Resist Updat. 2007;10(4–5):162–181. doi: 10.1016/j.drup.2007.06.003. [DOI] [PubMed] [Google Scholar]
- 16.Tallmann MS. Curative therapeutic approaches to APL. Ann Hematol. 2004;83(Suppl 1):S81–S82. doi: 10.1007/s00277-004-0850-2. [DOI] [PubMed] [Google Scholar]
- 17.Brueggemeier RW. Update on the use of aromatase inhibitors in breast cancer. Expert Opin Pharmacother. 2006;7(14):1919–1930. doi: 10.1517/14656566.7.14.1919. [DOI] [PubMed] [Google Scholar]
- 18.Ramirez ML, Keane TE, Evans CP. Managing prostate cancer: the role of hormone therapy. Can J Urol. 2007;14(Suppl 1):10–18. [PubMed] [Google Scholar]
- 19.O'Connor MJ, Martin NM, Smith GC. Targeted cancer therapies based on the inhibition of DNA strand break repair. Oncogene. 2007;26(56):7816–7824. doi: 10.1038/sj.onc.1210879. [DOI] [PubMed] [Google Scholar]
- 20.Leary A, Johnston SR. Small molecule signal transduction inhibitors for the treatment of solid tumors. Cancer Invest. 2007;25(5):347–365. doi: 10.1080/07357900701259694. [DOI] [PubMed] [Google Scholar]
- 21.Rowinsky EK. Targeted induction of apoptosis in cancer management: the emerging role of tumor necrosis factor-related apoptosis-inducing ligand receptor activating agents. J Clin Oncol. 2005;23(36):9394–9407. doi: 10.1200/JCO.2005.02.2889. [DOI] [PubMed] [Google Scholar]
- 22.Shapiro GI. Cyclin-dependent kinase pathways as targets for cancer treatment. J Clin Oncol. 2006;24(11):1770–1783. doi: 10.1200/JCO.2005.03.7689. [DOI] [PubMed] [Google Scholar]
- 23.O'Connor OA. Targeting histones and proteasomes: new strategies for the treatment of lymphoma. J Clin Oncol. 2005;23(26):6429–6436. doi: 10.1200/JCO.2005.05.014. [DOI] [PubMed] [Google Scholar]
- 24.Adjei AA, Hidalgo M. Intracellular signal transduction pathway proteins as targets for cancer therapy. J Clin Oncol. 2005;23(23):5386–5403. doi: 10.1200/JCO.2005.23.648. [DOI] [PubMed] [Google Scholar]
- 25.Petricoin EF, 3rd, et al. Mapping molecular networks using proteomics: a vision for patient-tailored combination therapy. J Clin Oncol. 2005;23(15):3614–3621. doi: 10.1200/JCO.2005.02.509. [DOI] [PubMed] [Google Scholar]
- 26.Dy GK, Adjei AA. Novel targets for lung cancer therapy: part I. J Clin Oncol. 2002;20(12):2881–2894. doi: 10.1200/JCO.2002.11.145. [DOI] [PubMed] [Google Scholar]
- 27.Pennati M, Folini M, Zaffaroni N. Targeting survivin in cancer therapy. Expert Opin Ther Targets. 2008;12(4):463–476. doi: 10.1517/14728222.12.4.463. [DOI] [PubMed] [Google Scholar]
- 28.Dy GK, Haluska P, Adjei AA. Novel pharmacological agents in clinical development for solid tumours. Expert Opin Investig Drugs. 2001;10(12):2059–2088. doi: 10.1517/13543784.10.12.2059. [DOI] [PubMed] [Google Scholar]
- 29.Baird RD, Kaye SB. Drug resistance reversal--are we getting closer? Eur J Cancer. 2003;39(17):2450–2461. doi: 10.1016/s0959-8049(03)00619-1. [DOI] [PubMed] [Google Scholar]
- 30.Agarwal R, Linch M, Kaye SB. Novel therapeutic agents in ovarian cancer. Eur J Surg Oncol. 2006;32(8):875–886. doi: 10.1016/j.ejso.2006.03.041. [DOI] [PubMed] [Google Scholar]
- 31.Vannucchi S, et al. Perspectives in biomolecular therapeutic intervention in cancer: from the early to the new strategies with type I interferons. Curr Med Chem. 2007;14(6):667–679. doi: 10.2174/092986707780059616. [DOI] [PubMed] [Google Scholar]
- 32.Hutson TE, Figlin RA. Evolving role of novel targeted agents in renal cell carcinoma. Oncology(Williston Park) 2007;21(10):1175–1180. discussion 1184, 1187, 1190. [PubMed] [Google Scholar]
- 33.Glaser KB. HDAC inhibitors: Clinical update and mechanism-based potential. Biochem Pharmacol. 2007 doi: 10.1016/j.bcp.2007.04.007. [DOI] [PubMed] [Google Scholar]
- 34.McDonald CJ, et al. A measles virus vaccine strain derivative as a novel oncolytic agent against breast cancer. Breast Cancer Res Treat. 2006;99(2):177–184. doi: 10.1007/s10549-006-9200-5. [DOI] [PubMed] [Google Scholar]
- 35.Allen C, et al. Oncolytic measles virus strains in the treatment of gliomas. Expert Opin Biol Ther. 2008;8(2):213–220. doi: 10.1517/14712598.8.2.213. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Fulda S. Modulation of TRAIL-induced apoptosis by HDAC inhibitors. Curr Cancer Drug Targets. 2008;8(2):132–140. doi: 10.2174/156800908783769355. [DOI] [PubMed] [Google Scholar]
- 37.Zhou XC, et al. Epigenetic considerations for endometrial cancer prevention, diagnosis and treatment. Gynecol Oncol. 2007;107(1):143–153. doi: 10.1016/j.ygyno.2007.06.019. [DOI] [PubMed] [Google Scholar]
- 38.Swaminathan V, et al. Small molecule modulators in epigenetics: implications in gene expression and therapeutics. Subcell Biochem. 2007;41:397–428. [PubMed] [Google Scholar]
- 39.Fandy TE, Carraway H, Gore SD. DNA demethylating agents and histone deacetylase inhibitors in hematologic malignancies. Cancer J. 2007;13(1):40–48. doi: 10.1097/PPO.0b013e31803c7359. [DOI] [PubMed] [Google Scholar]
- 40.Galm O, Herman JG, Baylin SB. The fundamental role of epigenetics in hematopoietic malignancies. Blood Rev. 2006;20(1):1–13. doi: 10.1016/j.blre.2005.01.006. [DOI] [PubMed] [Google Scholar]
- 41.Sun Q, et al. Interferon-gamma expressing EBV LMP2A-specific T cells for cellular immunotherapy. Cell Immunol. 2007;246(2):81–91. doi: 10.1016/j.cellimm.2007.06.004. [DOI] [PubMed] [Google Scholar]
- 42.Adams JM, Cory S. Bcl-2-regulated apoptosis: mechanism and therapeutic potential. Curr Opin Immunol. 2007;19(5):488–496. doi: 10.1016/j.coi.2007.05.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Eberle J, et al. Overcoming apoptosis deficiency of melanoma-hope for new therapeutic approaches. Drug Resist Updat. 2007;10(6):218–234. doi: 10.1016/j.drup.2007.09.001. [DOI] [PubMed] [Google Scholar]
- 44.Mitsiades CS, et al. Novel histone deacetylase inhibitors in the treatment of thyroid cancer. Clin Cancer Res. 2005;11(10):3958–3965. doi: 10.1158/1078-0432.CCR-03-0776. [DOI] [PubMed] [Google Scholar]
- 45.Fesik SW. Promoting apoptosis as a strategy for cancer drug discovery. Nat Rev Cancer. 2005;5(11):876–885. doi: 10.1038/nrc1736. [DOI] [PubMed] [Google Scholar]
- 46.Dai Y, Grant S. Targeting multiple arms of the apoptotic regulatory machinery. Cancer Res. 2007;67(7):2908–2911. doi: 10.1158/0008-5472.CAN-07-0082. [DOI] [PubMed] [Google Scholar]
- 47.Reed JC, Pellecchia M. Apoptosis-based therapies for hematologic malignancies. Blood. 2005;106(2):408–418. doi: 10.1182/blood-2004-07-2761. [DOI] [PubMed] [Google Scholar]
- 48.Walensky LD, et al. Activation of apoptosis in vivo by a hydrocarbon-stapled BH3 helix. Science. 2004;305(5689):1466–1470. doi: 10.1126/science.1099191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Oltersdorf T, et al. An inhibitor of Bcl-2 family proteins induces regression of solid tumours. Nature. 2005;435(7042):677–681. doi: 10.1038/nature03579. [DOI] [PubMed] [Google Scholar]
- 50.Altieri DC. Survivin, cancer networks and pathway-directed drug discovery. Nat Rev Cancer. 2008;8(1):61–70. doi: 10.1038/nrc2293. [DOI] [PubMed] [Google Scholar]
- 51.Tamm I, et al. Expression and prognostic significance of IAP-family genes in human cancers and myeloid leukemias. Clin Cancer Res. 2000;6(5):1796–1803. [PubMed] [Google Scholar]
- 52.Vucic D, Fairbrother WJ. The inhibitor of apoptosis proteins as therapeutic targets in cancer. Clin Cancer Res. 2007;13(20):5995–6000. doi: 10.1158/1078-0432.CCR-07-0729. [DOI] [PubMed] [Google Scholar]
- 53.Jaattela M. Multiple cell death pathways as regulators of tumour initiation and progression. Oncogene. 2004;23(16):2746–2756. doi: 10.1038/sj.onc.1207513. [DOI] [PubMed] [Google Scholar]
- 54.Okada H, Mak TW. Pathways of apoptotic and non-apoptotic death in tumour cells. Nat Rev Cancer. 2004;4(8):592–603. doi: 10.1038/nrc1412. [DOI] [PubMed] [Google Scholar]
- 55.Leist M, Jaattela M. Four deaths and a funeral: from caspases to alternative mechanisms. Nat Rev Mol Cell Biol. 2001;2(8):589–598. doi: 10.1038/35085008. [DOI] [PubMed] [Google Scholar]
- 56.Moretti L, et al. Autophagy signaling in cancer and its potential as novel target to improve anticancer therapy. Drug Resist Updat. 2007;10(4–5):135–143. doi: 10.1016/j.drup.2007.05.001. [DOI] [PubMed] [Google Scholar]
- 57.O'Connell AR, et al. Characterization of a serine protease-mediated cell death program activated in human leukemia cells. Exp Cell Res. 2006;312(1):27–39. doi: 10.1016/j.yexcr.2005.10.003. [DOI] [PubMed] [Google Scholar]
- 58.O'Connell AR, Lee BW, Stenson-Cox C. Caspase-dependant activation of chymotrypsin-like proteases mediates nuclear events during Jurkat T cell apoptosis. Biochem Biophys Res Commun. 2006;345(2):608–616. doi: 10.1016/j.bbrc.2006.04.147. [DOI] [PubMed] [Google Scholar]
- 59.Moffitt KL, Martin SL, Walker B. The emerging role of serine proteases in apoptosis. Biochem Soc Trans. 2007;35(Pt 3):559–560. doi: 10.1042/BST0350559. [DOI] [PubMed] [Google Scholar]
- 60.de Bruin EC, et al. A serine protease is involved in the initiation of DNA damage-induced apoptosis. Cell Death Differ. 2003;10(10):1204–1212. doi: 10.1038/sj.cdd.4401296. [DOI] [PubMed] [Google Scholar]
- 61.Cilenti L, et al. Omi/HtrA2 protease mediates cisplatin-induced cell death in renal cells. Am J Physiol Renal Physiol. 2005;288(2):F371–F379. doi: 10.1152/ajprenal.00154.2004. [DOI] [PubMed] [Google Scholar]
- 62.Suzuki Y, et al. A serine protease, HtrA2, is released from the mitochondria and interacts with XIAP, inducing cell death. Mol Cell. 2001;8(3):613–621. doi: 10.1016/s1097-2765(01)00341-0. [DOI] [PubMed] [Google Scholar]
- 63.Verhagen AM, et al. HtrA2 promotes cell death through its serine protease activity and its ability to antagonize inhibitor of apoptosis proteins. J Biol Chem. 2002;277(1):445–454. doi: 10.1074/jbc.M109891200. [DOI] [PubMed] [Google Scholar]
- 64.Chien J, et al. Serine protease HtrA1 modulates chemotherapy-induced cytotoxicity. J Clin Invest. 2006;116(7):1994–2004. doi: 10.1172/JCI27698. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Lipinska B, Sharma S, Georgopoulos C. Sequence analysis and regulation of the htrA gene of Escherichia coli: a sigma 32-independent mechanism of heat-inducible transcription. Nucleic Acids Res. 1988;16(21):10053–10067. doi: 10.1093/nar/16.21.10053. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Strauch KL, Beckwith J. An Escherichia coli mutation preventing degradation of abnormal periplasmic proteins. Proc Natl Acad Sci U S A. 1988;85(5):1576–1580. doi: 10.1073/pnas.85.5.1576. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Pallen MJ, Wren BW. The HtrA family of serine proteases. Mol Microbiol. 1997;26(2):209–221. doi: 10.1046/j.1365-2958.1997.5601928.x. [DOI] [PubMed] [Google Scholar]
- 68.Spiess C, Beil A, Ehrmann M. A temperature-dependent switch from chaperone to protease in a widely conserved heat shock protein. Cell. 1999;97(3):339–347. doi: 10.1016/s0092-8674(00)80743-6. [DOI] [PubMed] [Google Scholar]
- 69.Hu SI, et al. Human HtrA, an evolutionarily conserved serine protease identified as a differentially expressed gene product in osteoarthritic cartilage. J Biol Chem. 1998;273(51):34406–34412. doi: 10.1074/jbc.273.51.34406. [DOI] [PubMed] [Google Scholar]
- 70.Zumbrunn J, Trueb B. Primary structure of a putative serine protease specific for IGF- binding proteins. FEBS Lett. 1996;398(2–3):187–192. doi: 10.1016/s0014-5793(96)01229-x. [DOI] [PubMed] [Google Scholar]
- 71.Faccio L, et al. Characterization of a novel human serine protease that has extensive homology to bacterial heat shock endoprotease HtrA and is regulated by kidney ischemia. J Biol Chem. 2000;275(4):2581–2588. doi: 10.1074/jbc.275.4.2581. [DOI] [PubMed] [Google Scholar]
- 72.Gray CW, et al. Characterization of human HtrA2, a novel serine protease involved in the mammalian cellular stress response. Eur J Biochem. 2000;267(18):5699–5710. doi: 10.1046/j.1432-1327.2000.01589.x. [DOI] [PubMed] [Google Scholar]
- 73.Nie GY, et al. Identification and cloning of two isoforms of human high-temperature requirement factor A3 (HtrA3), characterization of its genomic structure and comparison of its tissue distribution with HtrA1 and HtrA2. Biochem J. 2003;371(Pt 1):39–48. doi: 10.1042/BJ20021569. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Oka C, et al. HtrA1 serine protease inhibits signaling mediated by Tgfbeta family proteins. Development. 2004;131(5):1041–1053. doi: 10.1242/dev.00999. [DOI] [PubMed] [Google Scholar]
- 75.Clausen T, Southan C, Ehrmann M. The HtrA family of proteases: implications for protein composition and cell fate. Mol Cell. 2002;10(3):443–455. doi: 10.1016/s1097-2765(02)00658-5. [DOI] [PubMed] [Google Scholar]
- 76.De Luca A, et al. Distribution of the serine protease HtrA1 in normal human tissues. J Histochem Cytochem. 2003;51(10):1279–1284. doi: 10.1177/002215540305101004. [DOI] [PubMed] [Google Scholar]
- 77.De Luca A, et al. The serine protease HtrA1 is upregulated in the human placenta during pregnancy. J Histochem Cytochem. 2004;52(7):885–892. doi: 10.1369/jhc.3A6186.2004. [DOI] [PubMed] [Google Scholar]
- 78.Wu G, et al. Structural basis of IAP recognition by Smac/DIABLO. Nature. 2000;408(6815):1008–1012. doi: 10.1038/35050012. [DOI] [PubMed] [Google Scholar]
- 79.Hegde R, et al. Identification of Omi/HtrA2 as a mitochondrial apoptotic serine protease that disrupts inhibitor of apoptosis protein-caspase interaction. J Biol Chem. 2002;277(1):432–438. doi: 10.1074/jbc.M109721200. [DOI] [PubMed] [Google Scholar]
- 80.Martins LM, et al. The serine protease Omi/HtrA2 regulates apoptosis by binding XIAP through a reaper-like motif. J Biol Chem. 2002;277(1):439–444. doi: 10.1074/jbc.M109784200. [DOI] [PubMed] [Google Scholar]
- 81.van Loo G, et al. The serine protease Omi/HtrA2 is released from mitochondria during apoptosis. Omi interacts with caspase-inhibitor XIAP and induces enhanced caspase activity. Cell Death Differ. 2002;9(1):20–26. doi: 10.1038/sj.cdd.4400970. [DOI] [PubMed] [Google Scholar]
- 82.Bartke T, et al. Dual role of BRUCE as an antiapoptotic IAP and a chimeric E2/E3 ubiquitin ligase. Mol Cell. 2004;14(6):801–811. doi: 10.1016/j.molcel.2004.05.018. [DOI] [PubMed] [Google Scholar]
- 83.Seong YM, et al. Autocatalytic processing of HtrA2/Omi is essential for induction of caspase-dependent cell death through antagonizing XIAP. J Biol Chem. 2004;279(36):37588–37596. doi: 10.1074/jbc.M401408200. [DOI] [PubMed] [Google Scholar]
- 84.Srinivasula SM, et al. Inhibitor of apoptosis proteins are substrates for the mitochondrial serine protease Omi/HtrA2. J Biol Chem. 2003;278(34):31469–31472. doi: 10.1074/jbc.C300240200. [DOI] [PubMed] [Google Scholar]
- 85.Suzuki Y, et al. Mitochondrial protease Omi/HtrA2 enhances caspase activation through multiple pathways. Cell Death Differ. 2004;11(2):208–216. doi: 10.1038/sj.cdd.4401343. [DOI] [PubMed] [Google Scholar]
- 86.Yang QH, et al. Omi/HtrA2 catalytic cleavage of inhibitor of apoptosis (IAP) irreversibly inactivates IAPs and facilitates caspase activity in apoptosis. Genes Dev. 2003;17(12):1487–1496. doi: 10.1101/gad.1097903. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Nie GY, et al. A novel serine protease of the mammalian HtrA family is up-regulated in mouse uterus coinciding with placentation. Mol Hum Reprod. 2003;9(5):279–290. doi: 10.1093/molehr/gag036. [DOI] [PubMed] [Google Scholar]
- 88.Rhodes DR, et al. Oncomine 3.0: genes, pathways, and networks in a collection of 18,000 cancer gene expression profiles. Neoplasia. 2007;9(2):166–180. doi: 10.1593/neo.07112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Chien J, et al. A candidate tumor suppressor HtrA1 is downregulated in ovarian cancer. Oncogene. 2004;23(8):1636–1644. doi: 10.1038/sj.onc.1207271. [DOI] [PubMed] [Google Scholar]
- 90.Baldi A, et al. The HtrA1 serine protease is down-regulated during human melanoma progression and represses growth of metastatic melanoma cells. Oncogene. 2002;21(43):6684–6688. doi: 10.1038/sj.onc.1205911. [DOI] [PubMed] [Google Scholar]
- 91.Esposito V, et al. Analysis of HtrA1 serine protease expression in human lung cancer. Anticancer Res. 2006;26(5A):3455–3459. [PubMed] [Google Scholar]
- 92.Baldi A, et al. The serine protease HTRA1 is a novel prognostic factor for human mesothelioma. doi: 10.2217/14622416.9.8.1069. (submitted). [DOI] [PubMed] [Google Scholar]
- 93.Bartling B, Lewensohn R, Zhivotovsky B. Endogenously released Smac is insufficient to mediate cell death of human lung carcinoma in response to etoposide. Exp Cell Res. 2004;298(1):83–95. doi: 10.1016/j.yexcr.2004.04.007. [DOI] [PubMed] [Google Scholar]
- 94.Folgueira MA, et al. Gene expression profile associated with response to doxorubicin-based therapy in breast cancer. Clin Cancer Res. 2005;11(20):7434–7443. doi: 10.1158/1078-0432.CCR-04-0548. [DOI] [PubMed] [Google Scholar]
- 95.Balch C, et al. The epigenetics of ovarian cancer drug resistance and resensitization. Am J Obstet Gynecol. 2004;191(5):1552–1572. doi: 10.1016/j.ajog.2004.05.025. [DOI] [PubMed] [Google Scholar]
- 96.Schneider-Stock R, Ocker M. Epigenetic therapy in cancer: molecular background and clinical development of histone deacetylase and DNA methyltransferase inhibitors. IDrugs. 2007;10(8):557–561. [PubMed] [Google Scholar]
- 97.Kotliarov Y, et al. High-resolution global genomic survey of 178 gliomas reveals novel regions of copy number alteration and allelic imbalances. Cancer Res. 2006;66(19):9428–9436. doi: 10.1158/0008-5472.CAN-06-1691. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Pomeroy SL, et al. Prediction of central nervous system embryonal tumour outcome based on gene expression. Nature. 2002;415(6870):436–442. doi: 10.1038/415436a. [DOI] [PubMed] [Google Scholar]
- 99.Lee J, et al. Tumor stem cells derived from glioblastomas cultured in bFGF and EGF more closely mirror the phenotype and genotype of primary tumors than do serum-cultured cell lines. Cancer Cell. 2006;9(5):391–403. doi: 10.1016/j.ccr.2006.03.030. [DOI] [PubMed] [Google Scholar]
- 100.Hendrix ND, et al. Fibroblast growth factor 9 has oncogenic activity and is a downstream target of Wnt signaling in ovarian endometrioid adenocarcinomas. Cancer Res. 2006;66(3):1354–1362. doi: 10.1158/0008-5472.CAN-05-3694. [DOI] [PubMed] [Google Scholar]
- 101.Welsh JB, et al. Analysis of gene expression profiles in normal and neoplastic ovarian tissue samples identifies candidate molecular markers of epithelial ovarian cancer. Proc Natl Acad Sci U S A. 2001;98(3):1176–1181. doi: 10.1073/pnas.98.3.1176. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Lu KH, et al. Selection of potential markers for epithelial ovarian cancer with gene expression arrays and recursive descent partition analysis. Clin Cancer Res. 2004;10(10):3291–3300. doi: 10.1158/1078-0432.CCR-03-0409. [DOI] [PubMed] [Google Scholar]
- 103.Adib TR, et al. Predicting biomarkers for ovarian cancer using gene-expression microarrays. Br J Cancer. 2004;90(3):686–692. doi: 10.1038/sj.bjc.6601603. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Ginos MA, et al. Identification of a gene expression signature associated with recurrent disease in squamous cell carcinoma of the head and neck. Cancer Res. 2004;64(1):55–63. doi: 10.1158/0008-5472.can-03-2144. [DOI] [PubMed] [Google Scholar]
- 105.Talbot SG, et al. Gene expression profiling allows distinction between primary and metastatic squamous cell carcinomas in the lung. Cancer Res. 2005;65(8):3063–3071. doi: 10.1158/0008-5472.CAN-04-1985. [DOI] [PubMed] [Google Scholar]
- 106.Gordon GJ, et al. Translation of microarray data into clinically relevant cancer diagnostic tests using gene expression ratios in lung cancer and mesothelioma. Cancer Res. 2002;62(17):4963–4967. [PubMed] [Google Scholar]
- 107.Farmer P, et al. Identification of molecular apocrine breast tumours by microarray analysis. Oncogene. 2005;24(29):4660–4671. doi: 10.1038/sj.onc.1208561. [DOI] [PubMed] [Google Scholar]
- 108.Wang Y, et al. Gene-expression profiles to predict distant metastasis of lymph-node-negative primary breast cancer. Lancet. 2005;365(9460):671–679. doi: 10.1016/S0140-6736(05)17947-1. [DOI] [PubMed] [Google Scholar]
- 109.Desmedt C, et al. Strong time dependence of the 76-gene prognostic signature for node-negative breast cancer patients in the TRANSBIG multicenter independent validation series. Clin Cancer Res. 2007;13(11):3207–3214. doi: 10.1158/1078-0432.CCR-06-2765. [DOI] [PubMed] [Google Scholar]
- 110.van de Vijver MJ, et al. A gene-expression signature as a predictor of survival in breast cancer. N Engl J Med. 2002;347(25):1999–2009. doi: 10.1056/NEJMoa021967. [DOI] [PubMed] [Google Scholar]
- 111.Chin K, et al. Genomic and transcriptional aberrations linked to breast cancer pathophysiologies. Cancer Cell. 2006;10(6):529–541. doi: 10.1016/j.ccr.2006.10.009. [DOI] [PubMed] [Google Scholar]
- 112.van 't Veer LJ, et al. Gene expression profiling predicts clinical outcome of breast cancer. Nature. 2002;415(6871):530–536. doi: 10.1038/415530a. [DOI] [PubMed] [Google Scholar]
- 113.Ivshina AV, et al. Genetic reclassification of histologic grade delineates new clinical subtypes of breast cancer. Cancer Res. 2006;66(21):10292–10301. doi: 10.1158/0008-5472.CAN-05-4414. [DOI] [PubMed] [Google Scholar]
- 114.Miller LD, et al. An expression signature for p53 status in human breast cancer predicts mutation status, transcriptional effects, and patient survival. Proc Natl Acad Sci U S A. 2005;102(38):13550–13555. doi: 10.1073/pnas.0506230102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Sotiriou C, et al. Gene expression profiling in breast cancer: understanding the molecular basis of histologic grade to improve prognosis. J Natl Cancer Inst. 2006;98(4):262–272. doi: 10.1093/jnci/djj052. [DOI] [PubMed] [Google Scholar]
- 116.Raetz EA, et al. Gene expression profiling reveals intrinsic differences between T-cell acute lymphoblastic leukemia and T-cell lymphoblastic lymphoma. Pediatr Blood Cancer. 2006;47(2):130–140. doi: 10.1002/pbc.20550. [DOI] [PubMed] [Google Scholar]
- 117.Mullighan CG, et al. Genome-wide analysis of genetic alterations in acute lymphoblastic leukaemia. Nature. 2007;446(7137):758–764. doi: 10.1038/nature05690. [DOI] [PubMed] [Google Scholar]
- 118.Welsh JB, et al. Analysis of gene expression identifies candidate markers and pharmacological targets in prostate cancer. Cancer Res. 2001;61(16):5974–5978. [PubMed] [Google Scholar]
- 119.Holzbeierlein J, et al. Gene expression analysis of human prostate carcinoma during hormonal therapy identifies androgen-responsive genes and mechanisms of therapy resistance. Am J Pathol. 2004;164(1):217–227. doi: 10.1016/S0002-9440(10)63112-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Yu YP, et al. Gene expression alterations in prostate cancer predicting tumor aggression and preceding development of malignancy. J Clin Oncol. 2004;22(14):2790–2799. doi: 10.1200/JCO.2004.05.158. [DOI] [PubMed] [Google Scholar]
- 121.Varambally S, et al. Integrative genomic and proteomic analysis of prostate cancer reveals signatures of metastatic progression. Cancer Cell. 2005;8(5):393–406. doi: 10.1016/j.ccr.2005.10.001. [DOI] [PubMed] [Google Scholar]
- 122.Segal NH, et al. Classification of clear-cell sarcoma as a subtype of melanoma by genomic profiling. J Clin Oncol. 2003;21(9):1775–1781. doi: 10.1200/JCO.2003.10.108. [DOI] [PubMed] [Google Scholar]
- 123.Talantov D, et al. Novel genes associated with malignant melanoma but not benign melanocytic lesions. Clin Cancer Res. 2005;11(20):7234–7242. doi: 10.1158/1078-0432.CCR-05-0683. [DOI] [PubMed] [Google Scholar]
- 124.Tschentscher F, et al. Tumor classification based on gene expression profiling shows that uveal melanomas with and without monosomy 3 represent two distinct entities. Cancer Res. 2003;63(10):2578–2584. [PubMed] [Google Scholar]
- 125.Cutcliffe C, et al. Clear cell sarcoma of the kidney: up-regulation of neural markers with activation of the sonic hedgehog and Akt pathways. Clin Cancer Res. 2005;11(22):7986–7994. doi: 10.1158/1078-0432.CCR-05-1354. [DOI] [PubMed] [Google Scholar]
- 126.Sanchez-Carbayo M, et al. Defining molecular profiles of poor outcome in patients with invasive bladder cancer using oligonucleotide microarrays. J Clin Oncol. 2006;24(5):778–789. doi: 10.1200/JCO.2005.03.2375. [DOI] [PubMed] [Google Scholar]
- 127.Stearman RS, et al. Analysis of orthologous gene expression between human pulmonary adenocarcinoma and a carcinogen-induced murine model. Am J Pathol. 2005;167(6):1763–1775. doi: 10.1016/S0002-9440(10)61257-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Bhattacharjee A, et al. Classification of human lung carcinomas by mRNA expression profiling reveals distinct adenocarcinoma subclasses. Proc Natl Acad Sci U S A. 2001;98(24):13790–13795. doi: 10.1073/pnas.191502998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Dyrskjot L, et al. Gene expression in the urinary bladder: a common carcinoma in situ gene expression signature exists disregarding histopathological classification. Cancer Res. 2004;64(11):4040–4048. doi: 10.1158/0008-5472.CAN-03-3620. [DOI] [PubMed] [Google Scholar]
- 130.Stransky N, et al. Regional copy number-independent deregulation of transcription in cancer. Nat Genet. 2006;38(12):1386–1396. doi: 10.1038/ng1923. [DOI] [PubMed] [Google Scholar]
- 131.Sun L, et al. Neuronal and glioma-derived stem cell factor induces angiogenesis within the brain. Cancer Cell. 2006;9(4):287–300. doi: 10.1016/j.ccr.2006.03.003. [DOI] [PubMed] [Google Scholar]
- 132.Haslinger C, et al. Microarray gene expression profiling of B-cell chronic lymphocytic leukemia subgroups defined by genomic aberrations and VH mutation status. J Clin Oncol. 2004;22(19):3937–3949. doi: 10.1200/JCO.2004.12.133. [DOI] [PubMed] [Google Scholar]
- 133.Andersson A, et al. Microarray-based classification of a consecutive series of 121 childhood acute leukemias: prediction of leukemic and genetic subtype as well as of minimal residual disease status. Leukemia. 2007;21(6):1198–1203. doi: 10.1038/sj.leu.2404688. [DOI] [PubMed] [Google Scholar]
- 134.Boni JP, et al. Population pharmacokinetics of CCI-779: correlations to safety and pharmacogenomic responses in patients with advanced renal cancer. Clin Pharmacol Ther. 2005;77(1):76–89. doi: 10.1016/j.clpt.2004.08.025. [DOI] [PubMed] [Google Scholar]
- 135.Bild AH, et al. Oncogenic pathway signatures in human cancers as a guide to targeted therapies. Nature. 2006;439(7074):353–357. doi: 10.1038/nature04296. [DOI] [PubMed] [Google Scholar]
- 136.Watanabe T, et al. Distal colorectal cancers with microsatellite instability (MSI) display distinct gene expression profiles that are different from proximal MSI cancers. Cancer Res. 2006;66(20):9804–9808. doi: 10.1158/0008-5472.CAN-06-1163. [DOI] [PubMed] [Google Scholar]
- 137.Sorlie T, et al. Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications. Proc Natl Acad Sci U S A. 2001;98(19):10869–10874. doi: 10.1073/pnas.191367098. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Sorlie T, et al. Repeated observation of breast tumor subtypes in independent gene expression data sets. Proc Natl Acad Sci U S A. 2003;100(14):8418–8423. doi: 10.1073/pnas.0932692100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Hummel M, et al. A biologic definition of Burkitt's lymphoma from transcriptional and genomic profiling. N Engl J Med. 2006;354(23):2419–2430. doi: 10.1056/NEJMoa055351. [DOI] [PubMed] [Google Scholar]
- 140.Dhanasekaran SM, et al. Delineation of prognostic biomarkers in prostate cancer. Nature. 2001;412(6849):822–826. doi: 10.1038/35090585. [DOI] [PubMed] [Google Scholar]
- 141.Korkola JE, et al. Down-regulation of stem cell genes, including those in a 200-kb gene cluster at 12p13.31, is associated with in vivo differentiation of human male germ cell tumors. Cancer Res. 2006;66(2):820–827. doi: 10.1158/0008-5472.CAN-05-2445. [DOI] [PubMed] [Google Scholar]
- 142.Hao Y, et al. Gene expression profiling reveals stromal genes expressed in common between Barrett's esophagus and adenocarcinoma. Gastroenterology. 2006;131(3):925–933. doi: 10.1053/j.gastro.2006.04.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Iacobuzio-Donahue CA, et al. Exploration of global gene expression patterns in pancreatic adenocarcinoma using cDNA microarrays. Am J Pathol. 2003;162(4):1151–1162. doi: 10.1016/S0002-9440(10)63911-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Ross ME, et al. Classification of pediatric acute lymphoblastic leukemia by gene expression profiling. Blood. 2003;102(8):2951–2959. doi: 10.1182/blood-2003-01-0338. [DOI] [PubMed] [Google Scholar]
- 145.Fine BM, et al. Gene expression patterns associated with recurrent chromosomal translocations in acute lymphoblastic leukemia. Blood. 2004;103(3):1043–1049. doi: 10.1182/blood-2003-05-1518. [DOI] [PubMed] [Google Scholar]
- 146.Heuser M, et al. Gene-expression profiles and their association with drug resistance in adult acute myeloid leukemia. Haematologica. 2005;90(11):1484–1492. [PubMed] [Google Scholar]
- 147.Schmidt S, et al. Identification of glucocorticoid-response genes in children with acute lymphoblastic leukemia. Blood. 2006;107(5):2061–2069. doi: 10.1182/blood-2005-07-2853. [DOI] [PubMed] [Google Scholar]
- 148.Maser RS, et al. Chromosomally unstable mouse tumours have genomic alterations similar to diverse human cancers. Nature. 2007;447(7147):966–971. doi: 10.1038/nature05886. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Richardson AL, et al. X chromosomal abnormalities in basal-like human breast cancer. Cancer Cell. 2006;9(2):121–132. doi: 10.1016/j.ccr.2006.01.013. [DOI] [PubMed] [Google Scholar]
