Skip to main content
Erschienen in: Journal of Hematology & Oncology 1/2011

Open Access 01.12.2011 | Review

Unfolded protein response in cancer: the Physician's perspective

verfasst von: Xuemei Li, Kezhong Zhang, Zihai Li

Erschienen in: Journal of Hematology & Oncology | Ausgabe 1/2011

Abstract

The unfolded protein response ( UPR) is a cascade of intracellular stress signaling events in response to an accumulation of unfolded or misfolded proteins in the lumen of the endoplasmic reticulum (ER). Cancer cells are often exposed to hypoxia, nutrient starvation, oxidative stress and other metabolic dysregulation that cause ER stress and activation of the UPR. Depending on the duration and degree of ER stress, the UPR can provide either survival signals by activating adaptive and antiapoptotic pathways, or death signals by inducing cell death programs. Sustained induction or repression of UPR pharmacologically may thus have beneficial and therapeutic effects against cancer. In this review, we discuss the basic mechanisms of UPR and highlight the importance of UPR in cancer biology. We also update the UPR-targeted cancer therapeutics currently in clinical trials.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1756-8722-4-8) contains supplementary material, which is available to authorized users.

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

All authors participated in the writing of this manuscript and have approved its publication.

1. The unfolded protein response: mechanism

During tumorigenesis, the high proliferation rate of cancer cells requires increased activities of ER machinery in facilitating protein folding, assembly, and transport. Other pathologic stimuli can interrupt the protein folding process and subsequently cause accumulation of unfolded or misfolded proteins in the ER, a condition referred to as "ER stress" [15]. These pathologic stimuli include those that cause ER calcium depletion, altered glycosylation, nutrient deprivation, oxidative stress, DNA damage, or energy perturbation or fluctuations. In order to handle the accumulation of the unfolded or misfolded proteins, the ER evolves a group of signal transduction pathways, collectively termed the unfolded protein response (UPR), to alter transcriptional and translational programs to maintain ER homeostasis [68].
UPR has two primary functions: 1) to initially restore normal function of the cell by halting protein translation and activating the signaling pathways that lead to increased production of molecular chaperones involved in protein folding [9, 10]; 2) to initiate apoptotic pathways to remove the stressed cells when the initial objectives are not achieved within a certain time lapse or the disruption is prolonged [11, 12].
As a part of the UPR program, ER-associated Protein Degradation (ERAD) is responsible for the degradation of aberrant or misfolded proteins in the ER, providing an important protein folding "quality control" mechanism. During the process of ERAD, molecular chaperones and associated factors recognize and target substrates for retrotranslocation to the cytoplasm, where they are polyubiquitinated and degraded by the 26S proteasome [13]. ERAD is essential for maintaining ER homeostasis, and the disruption of ERAD is closely associated with ER stress-induced apoptosis [14].
Proteasomal degradation and autophagy have been identified as two main mechanisms in charge of protein clearance in stressed cells. Proteasomal degradation digests soluble ubiquitin-conjugated proteins. Autophagy involves cytoplasmic components engulfed within a double membrane vesicle (autophagosome). The maturation of these vesicles may fuse with lysosomes, which leads in turn to the degradation of the autophagosome components by the lysosomal degradative enzymes. Conditions that induce ER stress also lead to induction of autophagy [15]. Activation of the IRE1, phosphorylation of eIF2α, and ER Ca2+ release can all regulate autophagy. Activation of autophagy after ER stress can be either cell-protective or cytotoxic. Persistent ER stress can switch the cytoprotective functions of UPR and autophagy into cell death programs. Some antitumoral agents (e.g., cannabinoids) activate ER stress and autophagy as the primary mechanism to promote cancer cell death [1618].

1.1. The unfolded protein response pathways

On aggregation of unfolded proteins, GRP78 (known also as the immunoglobulin heavy chain binding protein, or BiP), one of the most abundant ER luminal chaperones, binds to unfolded proteins and dissociates from the three membrane-bound ER stress sensors. These stress sensors include pancreatic ER kinase (PKR)-like ER kinase (PERK), activating transcription factor 6 (ATF6), and inositol-requiring enzyme 1 (IRE1). The dissociation of GRP78 from these stress sensors allows their subsequent activation (Figure 1). It has been proposed that the activation of the ER stress sensors may occur sequentially, with PERK being the first, rapidly followed by ATF6, and IRE1 may be activated last [19].
Activated PERK blocks general protein synthesis by phosphorylating eukaryotic initiation factor 2α (eIF2α), which suppress mRNA translation. Reduced global translation also leads to reduction of key regulatory proteins that are subject to rapid turnover, facilitating activation of transcription factors such as NFκ-B during cellular stress [4]. However, selective translation of some proteins is activated, including ATF4, which occurs through an alternative translation pathway. ATF4, being a transcription factor, translocates to the nucleus and induces the transcription of genes required to restore ER homeostasis. Activation of PERK is initially protective and crucial for survival during mild stress. However, it leads to the induction of CHOP (C/EBP homologous protein), an important element of the switch from pro-adaptive to pro-apoptotic signaling [2024].
PERK-mediated translational repression is transient and is followed by translational recovery and enhanced expression of genes that increase the capacity of the ER to process client proteins. P58IPK induction during the ER-stress response represses PERK activity and plays a functional role in the expression of downstream markers of PERK activity in the later phase of the ER-stress response. P58IPK, GADD34 and TRB3, are reported to be involved in switching off the PERK mediated pathway. Blocking this protective pathway can be a central element of the switch from adaptation to apoptosis [19, 25].
ATF6 is activated by regulated intramembrane proteolysis after its translocation from the ER to the Golgi apparatus [26]. Active ATF6 is also a transcription factor that regulates the expression of ER chaperones and X box-binding protein 1 (XBP1), another UPR-trans-activator. The target genes of ATF6 and XBP1 have been shown to be involved in protein folding, secretion, and degradation in the ER [27, 28].
To achieve its active form, Xbp1 mRNA must undergo a non-conventional mRNA splicing, which is carried out by IRE1α. IRE1α protein is a type I transmembrane protein that contains both a Ser/Thr kinase domain and an endoribonuclease domain. The endoribonuclease domain processes an intron from the Xbp1 mRNA. Spliced XBP1 protein (XBP1s) translocates to the nucleus to activate the transcription of the genes encoding protein chaperones or folding enzymes involved in protein folding, secretion, or ERAD. Ablation of IRE1α in mice produces an embryonic lethal phenotype. It has been demonstrated that both processes of ATF6 activation and the IRE1α-mediated splicing of XBP1 mRNA are required for full induction of the UPR [2931].

1.2. ER stress-induced apoptosis

The adaptive responses to the accumulation of unfolded or misfolded proteins in the ER provide initial protection from cell death. But persistent or excessive ER stress can trigger cell death, typically through apoptosis. Both mitochondria-dependent and -independent pathways have been proposed for ER stress-induced apoptosis [32, 33].
The mitochondria-dependent pathways involve pro-apoptotic cascades that culminate in cytochrome c release. CHOP (C/EBP homology protein) is one of the proteins involved, which heterodimerizes with several C/EBP family members to regulate their transcriptional activity [34]. CHOP is downstream of phosphorylation cascade of PERK and eIF-2α. CHOP has a role in the induction of cell death by promoting protein synthesis and oxidation in the stressed ER. It modulates the Bcl-2 family of proteins, GADD34 (growth arrest and DNA damage inducible protein 34), and TRB3 (tribbles-related protein 3), among other downstream proteins. After transcriptional activation by ATF4, CHOP directly activates GADD34, which promotes ER client protein biosynthesis by dephosphorylating phospho-Ser 51 of the α subunit of eIF-2α in stressed cells [35, 36]. Additionally, it has been suggested that CHOP upregulates pro-apoptotic members of the BCL2 family (BAK/BAD) and downregulates the anti-apoptotic members (BCL2), causing subsequent damage to the mitochondrial membrane and releasing cytochrome c into the cytosol. The released cytochrome c in turn activates cytosolic apoptotic protease activating factor1 (APAF1), which then activates the downstream caspase-9 and caspase-3-dependent cascade [37].
A number of ER stress conditions can cause calcium release from the ER to the cytosol, Increases in cytosolic calcium can also cause activation of calpain, which induces cleavage of procaspase-12 [38]. Once activated, the catalytic subunits of caspase-12 are released into the cytosol, where they activate the caspase-9 cascade in a cytochrome c independent manner [39].
It has also been suggested that activated IRE1a can recruit tumor-necrosis factor receptor associated factor 2 (TRAF2), which activates procaspase-4 as a mitochondria-independent apoptotic response. Both pathways ultimately lead to the activation of the caspase cascade mediated through caspase-9 and caspase-3, resulting in cell death [40].

2. The unfolded protein response and its effect on tumorigenesis

A broad range of cancer-types rely on ER protein folding machinery to correctly fold key signaling pathway proteins [41]. ER stress and the UPR are highly induced in various tumors. Accumulating evidence has demonstrated that the UPR is an important mechanism required for cancer cells to maintain malignancy and therapy resistance. Identifying the UPR components that are activated or suppressed in malignancy and exploring cancer therapeutic potentials by targeting the UPR are very active research areas [7].
The UPR pathways are activated in a great variety of tumor types, and have been demonstrated to be essential for tumor cells to survive the unfriendly tumor microenvironment. There are evidence of over-expression of XBP1s (excision of a 26 nucleotide unconventional intron from XBP-1 mRNA), activation of ATF6, phosphorylation of eIF-2α, induction of ATF4 and CHOP in a variety of cancer cells. The ER chaperones GRP78/BiP, glucose-regulated protein 94 (GRP94, also known as gp96 or HSP90b1) and GRP170 were also upregulated [42]. These studies were conducted in primary human tumor cells or cell lines, and animal models with breast tumor, hepatocellular carcinoma, gastric tumor, and esophageal adenocarcinoma [4252]. UPR and stress response in general have also been implicated in participating in inflammation-induced oncogenesis [53].
UPR is required for tumorigenesis. Animal study demonstrated that XBP1 was required for tumor growth in vivo. Xbp1-/- and Xbp1-knockdown cells did not form tumors in mice even though their growth rate and secretion of vascular endothelial growth factor (VEGF) in response to in vitro hypoxia treatment were not decreased [46]. ER stress can also induce anti-apoptotic responses. The activation of glycogen synthase kinase 3β (GSK 3β) leads to phosphorylation of p53, which increases its degradation [54], therefore protects cancer cells from p53 dependent apoptosis. In addition, NFκB is activated during ER stress to induce anti-apoptotic responses [55].
Heat shock proteins were reported to assist cancer cell adaptation to oncogenesis-associated stress either by repairing damaged proteins (protein refolding) or by degrading them. Heat shock proteins have also been implicated in the control of cell growth, and in resistance to various anticancer treatments that induce apoptosis. For example, HSP90 interacts with several key proteins in promoting prostate cancer progression, including wild-type and mutated AR, HER2, ErbB2, Src, Abl, Raf and Akt [56, 57]. GRP78/BiP, expressed at high levels in a variety of tumors, confers drug resistance in both proliferating and dormant cancer cells. Genetically engineered animal model with reduced GRP78 level significantly impedes tumor growth. Three major mechanisms were proposed for GRP78 mediated cancer progression: enhancement of tumor cell proliferation, protection against apoptosis, and promotion of tumor angiogenesis [5860].
ER stress has been implicated in different stages of tumor development. The proposed mechanism is, during early tumorigenesis and before angiogenesis occurs, that activation of the UPR induces a G1 cell cycle arrest and activation of p38, both of which promote a dormant state. If the apoptotic signals are induced by the UPR during this stage of tumor development, cancer cells with mutated elements of the apoptotic pathway may evade the alternative fate of death. ER stress also induces anti-apoptotic NF-κB and inhibits p53-dependent apoptotic signals. If the balance of early cancer development tilts against cell death, ER stress can further promote the aggressive growth of these cancer cells by enhancing their angiogenic ability. One example is the increased VEGF secretion through induction of GRP170, a BiP-like protein that acts as a chaperone for VEGF [37].

3. The unfolded protein response and its effect on disease prognosis

GRP78 is a marker of UPR activation. An elevated GRP78 level generally correlates with higher pathologic grade, recurrence rate, and poor survival in patients with breast, liver, prostate, colon, and gastric cancers; though there are conflicting reports on lung cancer. Neuroblastoma is an apparent exception with correlation of GRP78 abundance with earlier stage and better prognosis [59, 6164].
A retrospective cohort study of 127 stage II and III breast cancer patients who were treated with Adriamycin-based chemotherapy, showed association between GRP78 positivity and shorter time to tumor recurrence [59]. Another breast cancer study showed that the UPR is activated in the majority of breast cancers and confers resistance to chemotherapy and endocrine therapy. Estrogen is known to stimulate UPR in vitro. UPR activation interacts with estrogen response elements and may regulate tumor growth [65].
Overexpression of GRP94 and GRP78 has been observed more often in patients with poorly differentiated lung cancer than in well or moderately differentiated tumors [66]. According to a study on adenocarcinoma of the esophagus, GRP78 and GRP94 mRNA were elevated in all tumors. Increased expression of GRP78 may be responsible for controlling local tumor growth in early tumor stages, while high expression of GRP78 and GRP94 in advanced stages was believed to be dependent on other cellular stress reactions such as glucose deprivation, hypoxia, or the hosts' immune response [67]. Up-regulated expression of GRP78 and GRP94 was also reported in gastric carcinoma, which was associated with aggressive tumor growth and poor prognosis [68].
Heterozygous GRP78 mice with half of wild-type GRP78 level are comparable to WT siblings in tumor growth and development. The tumor progression was significantly impeded in these mice as exemplified by a longer latency period, reduced tumor size, and increased tumor apoptosis. Reduction of GRP78 in cancer xenograft animal model inhibited tumor formation and growth [69].
XBP1s is a trans-activator of UPR signaling. High XBP1s level is associated with increased tumor growth, resistance to anti-estrogen therapy and poor patient survival [70, 71]. In a B cell-specific XBP1s-overexpressing transgenic mouse model, multiple myeloma developed spontaneously, highlighting the importance of UPR in tumorigenesis [72].

4. Therapeutic targeting of unfolded protein response in cancer

The accumulation of unfolded proteins triggers the UPR, which mediates the inhibition of general protein synthesis but increases expression of several transcription factors that activate genes encoding ER stress-inducible molecular chaperones, transcription factors and signal pathway proteins. Most normal cells are not undergoing active "stress" response, and the UPR pathways remain in a quiescent state in these cells. This discrepancy between tumor cells and normal cells offers an advantage for the agents that target the UPR to achieve the specificity in cancer therapy. The therapeutic potential of targeting the UPR components in cancer mainly involves two approaches: induction of accumulation of misfolded protein in ER to overload the unfolded protein response, and inhibition of UPR adaptive and antiapoptotic pathways to prevent cells from adapting to stressful conditions leading to cell death. In the following paragraphs, we will discuss some examples of agents that are being developed as cancer therapeutics (Table 1).
Table 1
Examples of UPR-targeted cancer drugs in development
Drug
Classification/Mechanism
Development Stage
Disease Indication
Reference
Bortezomib
Proteasome inhibitor
FDA approved
Multiple myeloma, mantle-cell lymphoma
San et al. [97]
NPI-0052 (salinosporamide A)
Irreversible proteasome inhibitor
Phase I clinical trials
Multiple Myeloma, Advanced malignancies
Chauhan et al. [98]
Carfilzomib (PR-171)
Selective proteasome inhibitor
Phase I, II, III clinical trials
Multiple Myeloma, Waldenstrom's Macroglobulinemia
O'Connor et al. [99] Lee et al. [100]
PS-341
Selective proteasome inhibitor
Phase II
Multiple Myeloma
Richardson et al. [101]
CEP-18770
Proteasome inhibitor
Phase I, II clinical trials and preclinical studies
multiple myeloma, Non- Hodgkin's lymphoma
Piva et al. [102]
Tanespimycin (17-AAG, (17-Allylamino-17-demethoxygeldanamycin), KOS-953)
HSP90 Inhibitor
Phase I, II, III clinical trials
Gastrointestinal stromal tumors, breast cancer, gynecological, leukemia, lymphoma, melanoma, prostate, renal, thyroid carcinoma, melanoma
Richardson et al. [103, 104] Heath et al. [105] Pacey et al. [106]
Alvespimycin (KOS-1022, 17-DMAG)
HSP90 Inhibitor
Phase I clinical trials and preclinical studies
Acute myeloid leukemia, advanced carcinoma
Kummar et al. [107] Lancet et al. [108] Pamanathan et al. [109] Zismanov et al. [110]
Retaspimycin (IPI-504)
HSP90 Inhibitor
Phase II clinical trials
Gastrointestinal stromal tumors, nonsmall cell lung, prostate
Hanson et al. [111]
PU-H71
HSP90 Inhibitor
Preclinical studies
Breast cancer, myeloma, myeloproliferative disorder
Usmani et al. [84] Caldas-Lopes et al. [112] Marubayashi et al. [113]
SNX-2112
HSP-90 inhibitor
Preclinical studies
Gastric cancer
Bachleitner-Hofmann, et al. [114]
Eeyarestatin I (EerI)
Inhibitor of ER-associated degradation (ERAD)
Preclinical studies
 
Cross et al. [115]
Versipelostatin
GRP78 inhibitor
Preclinical studies
 
Matsuo et al. [87]
(-)-epigallocatechin gallate (EGCG)
GRP78 inhibitor
Preclinical studies
Breast carcinoma
Luo et al. [116]
Epidermal growth factor (EGF)-SubA
GRP78-targeting cytotoxin
Preclinical murine animal models
Prostate tumor
Backer et al. [90]
Irestatins
IRE1α inhibitor
Preclinical studies
Multiple Myeloma,
Feldman et al. [117]
Delta(9)-Tetrahydrocannabinol (THC)
Cannabinoid, activates ER stress and autophagy
Phase I clinical trial
Glioblastoma multiforme
Guzmán et al. [118]

4.1. Targeting induction of unfolded protein response

Proteasomal inhibitor

Proteasomal degradation of misfolded proteins retrotranslocated from the ER to the cytosol represents the final step in ERAD. Bortezomib (Velcade, PS-341), a boronic acid derivative, was the first proteosome inhibitor to be developed successfully for anti-cancer therapy. Although the drug probably has multiple mechanisms of action, proteasomal inhibition causes an additional burden of unfolded proteins in the ER. This explains the high efficacy of bortezomib treatment against types of cancer cells in which the ER is already predisposed with a considerable protein load. In multiple myeloma cell lines, Bortezomib rapidly induced components of the proapoptotic UPR, including PERK, the ER stress-specific eIF-2α kinase, ATF4 and its proapoptotic target, CHOP. The amount of immunoglobulin subunits retained within multiple myeloma cells correlated with their sensitivity to proteasomal inhibitors [73].
Bortezomib treatment has a cytotoxic effect on various other cancer types such as breast, colorectal, ovarian, pancreatic, prostate, lung and oral cancer. It has been approved by the FDA for the treatment of relapsed multiple myeloma, and recently for relapsed mantle cell lymphoma. Combination chemotherapy regimens with Bortezomib have been developed, leading to unprecedented high remission rates in the frontline treatment or in the relapsed setting for multiple myeloma. The combination of proteasome inhibition with novel targeted therapies is an emerging field in oncology [74].

ERAD inhibitors

As a part of ER quality control mechanism, misfolded or unassembled proteins are retained in the ER and subsequently degraded by ERAD. In the ERAD pathway, molecular chaperones and lectin-like proteins are involved in the identification of misfolded proteins. ER-resident reductases cleave disulfide bonds in these proteins to facilitate retrograde transport to the cytosol. Furthermore, the AAA(+) adenosine triphosphatase withdraws them from the retrotranslocation channel to the cytosol where they are degraded by the ubiquitin/proteasome system [75].
Defects in ERAD cause the accumulation of misfolded proteins in the ER and thus trigger ER stress and UPR. Eeyarestatin I (EerI), a chemical inhibitor that can block ERAD, has been shown to have preferential cytotoxic activity against cancer cells. EerI targets p97 (a cytosolic ATPase involved in polyubiquitinated proteins transportation) complex to inhibit deubiquitination of p97-associated ERAD substrates, which is required for the degradation process [76].

PDI inhibitors

Protein disulfide isomerase (PDI) is one of the most abundant ER proteins and maintains a sentinel function in organizing accurate protein folding. PDIs are key protein folding catalysts activated during UPR [77]. Treatment of cells with O(2)-[2,4-dinitro-5-(N-methyl-N-4-carboxyphenylamino)phenyl]1-(N,N- methylamino)diazen-1-ium-1,2-diolate (PABA/NO) resulted in a dose-dependent increase in intracellular nitric oxide that caused S-glutathionylation and therefore inhibition of PDI. PABA/NO activates the UPR and causes translational attenuation, phosphorylation and activation of PERK, and its downstream effector eIF2α in human leukemia (HL60) and ovarian cancer cells (SKOV3). There was also evidence for Xbp1 mRNA splicing and transcriptional activation of the ER resident chaperones GRP78 and GRP94. Stimulating UPR may be linked with the cytotoxic potential of PABA/NO in cancer cells [78].

4.2. Targeting ER chaperones/heat shock proteins

HSP90 inhibitor

Under conditions of cellular stress, cells upregulate chaperones to prevent protein misfolding and degradation. All three ER-membrane bound sensors are heavily reliant on the protein chaperone functions of the HSP90 complex. The interaction between the heat shock protein family and the key proteins in the UPR pathway may, in part, be mediated by their destabilizing effect on UPR proteins and increased accumulation of misfolded proteins.
Myeloma cell study demonstrated that HSP90 inhibitors, 17AAG (17-allylamino-17-demethoxygeldanamycin) and radicicol, similar to tunicamycin (TM) and thapsigargin (TG) (known UPR activators), are capable of activating all three branches of the UPR. All drugs inhibited proliferation and increased expression levels of the molecular chaperones BiP and GRP94. Unlike TG and TM, the HSP90 inhibitors activate a caspase-dependent cell death pathway [79]. 17AAG can induce the formation of 'intracellular inclusions' in breast cancer cells. In myeloma cells, these inclusions are comprised of aggregations of misfolded immunoglobulin light chains and analysis of protein samples taken from 17AAG-treated cells suggest that exposure to HSP90 inhibitors alters the expression of LC3 (microtubule-associated protein 1 light chain 3, a reliable marker for autophagosome formation), consistent with autophagosome formation [8082].
Study demonstrated analogous effects of HSP90 inhibitor, 17AAG in the colon cancer cell line HCT116 indicating that they utilize the UPR in a similar manner to multiple myeloma [41]. A recent phase II trial was done using the HSP90 inhibitor, 17-AAG in fifteen melanoma patients with measurable disease. 17-AAG was administered i.v. once weekly for 6 weeks at 450 mg/m2. No objective responses were observed. Western blot analysis of tumor biopsies showed an increase in HSP70 and a decrease in cyclin D1 expression in the posttreatment biopsies. UPR components were not analyzed in this study. More potent HSP90 inhibitor or a formulation that are soluble and can be administered chronically for a more prolonged suppression effect on UPR may be necessary to be clinically beneficial [83]. A phase III clinical trial is ongoing to evaluate the utility of 17-AAG in multiple myeloma patients. There are also Phase II clinical trails in breast cancer and non-small cell lung carcinoma. PU-H71, a novel purine scaffold HSP90 inhibitor, has shown interesting preclinical activity against myeloma [84].

Grp78/BiP inhibitor

Levels of Grp78/BiP are commonly raised in solid tumors and cancer cell lines [85]. Versipelostatin (VST) and analogues, novel macrocyclic compound and GRP78/BiP inhibitor, showed promise in solid tumors [86]. VST has demonstrated selective cytotoxicity to glucose-deprived tumor cells by preventing the unfolded protein response. It was shown to inhibit GRP78 induction and the expression of the UPR transactivators XBP1 and ATF4. Eukaryotic initiation factor 4E-binding protein 1 (4E-BP1), a negative regulator of eukaryotic initiation factor 4E-mediated protein translation, plays a role in the UPR-inhibitory action of VST. Aberrant activation of 4E-BP1 prevents induction of the GRP78 and ATF4 [7, 8789].
Treatment of glioma cells with another GRP78 inhibitor, epigallocatechin gallate (EGCG,) which targets the ATP-binding domain of GRP78 and blocks its UPR protective function, sensitizes glioma cells to chemotherapy agent temozolomide [85]. Additionally, an engineered fusion protein, epidermal growth factor-SubA (EGF-SubA), a chaperone-targeting cytotoxin, was reported to be highly toxic to growing and confluent epidermal growth factor receptor-expressing cancer cells, and its cytotoxicity is thought to be mediated by rapid cleavage of GRP78 [90].

4.3. Inhibiting IRE1α/XBP1 pathway

Inhibitors of the IRE1α/XBP1 pathway

Irestatin, an inhibitor of IRE1 and the unfolded protein response, mediates inhibition of XBP1s transcription activity. The inhibition of the IRE1 endonuclease impairs the growth of malignant myeloma cells and inhibits the survival of oxygen-starved tumor cells in vitro and subcutaneous HT1080 tumor xenografts [91].
Trierixin, a new member of the triene-ansamycin group, isolated from the fermentation broth of Streptomyces sp. AC654, was shown to be a novel inhibitor of ER-stress induced cleavage of XBP1 [92]. Future work needs to be done to evaluate its activity in cancer therapy.

4.4. Other agents affecting unfolded protein response

IPI-504, a soluble HSP90 inhibitor, can block the unfolded protein response in multiple myeloma (MM) cells. Partial UPR is constitutively activated in plasma cell-derived MM cells. IPI-504 can potently inhibit this pathway. IPI-504 achieves this by inactivating the transcription factors XBP1 and ATF6. In addition, IPI-504 also blocks the tunicamycin-induced phosphorylation of eIF2α by PERK. The inhibitory effect of IPI-504 on the UPR parallels its cytotoxic and pro-apoptotic effects on multiple myeloma cells [93].
As discussed above, autophagy is a cellular process in which cytoplasmic materials are sequestered into autophagosomes and delivered to lysosomes for degradation or recycling. It can switch from cytoprotective role to a form of programmed cell death with persistent ER stress. Tetrahydrocannabinol (THC), the main active component of marijuana, induces human glioma cell death through stimulation of autophagy. THC induced autophagy is associated with an increased phosphorylation of eIF2α [94].
Resveratrol (RES), a natural plant polyphenol, is an effective inducer of cell cycle arrest and apoptosis in a variety of carcinoma cell types. In addition, RES has been reported to inhibit tumorigenesis in several animal models. RES causes cell cycle arrest and proliferation inhibition via induction of UPR in human leukemia K562 cell line [95].
The phytoestrogen zearalenone (ZEA), one of the most active naturally occurring estrogenic compounds in food and beverages, has also been shown recently to induce human leukemic cell apoptosis via endoplasmic stress and mitochondrial pathway [96].

5. Perspectives

We have highlighted the importance of UPR in tumorigenesis and provided an overview on the potential strategy in perturbing UPR in cancer treatment. URP promotes the ability of cancer cells to adapt to and survive the hostile microenvironment through activation of stress-response pathways and upregulation of chaperones. Targeting URP pathway represents a novel targeted anti-cancer approach with initial successes in clinical studies. Further understanding of the pathway should provide additional therapeutic opportunities.
Clearly, UPR and the associated molecular components are emerging as important potential targets for drugs that may be used in the treatment of cancer in which protein-folding and protein quality control play a key role in disease pathology. This area looks set to be a very exciting one in years to come. It is worthwhile to point out that protein quality control is fundamentally important for life. Thus targeted therapy towards UPR or other arms of protein quality control is by no means cancer-specific and toxicity-free. Of particular importance is the lack of understanding of the fundamental roles and mechanisms of protein quality control in development, organ function, the evolution and fitness of organism. Thus, as more pharmacological agents are being developed clinically, attention needs to be paid to the understanding of the basic mechanism of the regulation of unfolded protein response and to the discovery of important new players in the protein quality control for disease target.

Acknowledgements

K.Z. and Z.L. are supported by NIH grants. We thank Ms. Samantha Cronin for her secretarial support.
This article is published under license to BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://​creativecommons.​org/​licenses/​by/​2.​0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

All authors participated in the writing of this manuscript and have approved its publication.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Anhänge

Authors’ original submitted files for images

Below are the links to the authors’ original submitted files for images.
Literatur
1.
Zurück zum Zitat Kaufman RJ: Stress signaling from the lumen of the endoplasmic reticulum: coordination of gene transcriptional and translational controls. Genes Dev. 1999, 13: 1211-1233. 10.1101/gad.13.10.1211.PubMedCrossRef Kaufman RJ: Stress signaling from the lumen of the endoplasmic reticulum: coordination of gene transcriptional and translational controls. Genes Dev. 1999, 13: 1211-1233. 10.1101/gad.13.10.1211.PubMedCrossRef
2.
Zurück zum Zitat McMillan DR, Gething MJ, Sambrook J: The cellular response to unfolded proteins: intercompartmental signaling. Curr Opin Biotechnol. 1994, 5: 540-545. 10.1016/0958-1669(94)90071-X.PubMedCrossRef McMillan DR, Gething MJ, Sambrook J: The cellular response to unfolded proteins: intercompartmental signaling. Curr Opin Biotechnol. 1994, 5: 540-545. 10.1016/0958-1669(94)90071-X.PubMedCrossRef
3.
Zurück zum Zitat Mori K: Tripartite management of unfolded proteins in the endoplasmic reticulum. Cell. 2000, 101: 451-454. 10.1016/S0092-8674(00)80855-7.PubMedCrossRef Mori K: Tripartite management of unfolded proteins in the endoplasmic reticulum. Cell. 2000, 101: 451-454. 10.1016/S0092-8674(00)80855-7.PubMedCrossRef
4.
Zurück zum Zitat Ron D, Walter P: Signal integration in the endoplasmic reticulum unfolded protein response. Nat Rev Mol Cell Biol. 2007, 8: 519-529. 10.1038/nrm2199.PubMedCrossRef Ron D, Walter P: Signal integration in the endoplasmic reticulum unfolded protein response. Nat Rev Mol Cell Biol. 2007, 8: 519-529. 10.1038/nrm2199.PubMedCrossRef
5.
Zurück zum Zitat Sidrauski C, Chapman R, Walter P: The unfolded protein response: an intracellular signalling pathway with many surprising features. Trends Cell Biol. 1998, 8: 245-249. 10.1016/S0962-8924(98)01267-7.PubMedCrossRef Sidrauski C, Chapman R, Walter P: The unfolded protein response: an intracellular signalling pathway with many surprising features. Trends Cell Biol. 1998, 8: 245-249. 10.1016/S0962-8924(98)01267-7.PubMedCrossRef
6.
Zurück zum Zitat Rutkowski DT WJ, Back SH, Kaufman RJ: UPR Pathways Combine to Prevent Hepatic Steatosis Caused by ER Stress-Mediated Suppression of Transcriptional Master Regulators. Dev Cell. 2008, 15 (6): 829-40. 10.1016/j.devcel.2008.10.015.PubMedCentralPubMedCrossRef Rutkowski DT WJ, Back SH, Kaufman RJ: UPR Pathways Combine to Prevent Hepatic Steatosis Caused by ER Stress-Mediated Suppression of Transcriptional Master Regulators. Dev Cell. 2008, 15 (6): 829-40. 10.1016/j.devcel.2008.10.015.PubMedCentralPubMedCrossRef
7.
Zurück zum Zitat Wang G, Yang ZQ, Zhang K: Endoplasmic reticulum stress response in cancer: molecular mechanism and therapeutic potential. Am J Transl Res. 2010, 2: 65-74.PubMedCentralPubMed Wang G, Yang ZQ, Zhang K: Endoplasmic reticulum stress response in cancer: molecular mechanism and therapeutic potential. Am J Transl Res. 2010, 2: 65-74.PubMedCentralPubMed
8.
Zurück zum Zitat Scheuner DSB, Kaufman RJ: Translational control is required for the unfolded protein response and in vivo glucose homeostasis. Mol Cell. 2001, 7 (6): 1165-76. 10.1016/S1097-2765(01)00265-9.PubMedCrossRef Scheuner DSB, Kaufman RJ: Translational control is required for the unfolded protein response and in vivo glucose homeostasis. Mol Cell. 2001, 7 (6): 1165-76. 10.1016/S1097-2765(01)00265-9.PubMedCrossRef
9.
Zurück zum Zitat Dorner AJWL, Kaufman RJ: Increased synthesis of secreted proteins induces expression of glucose-regulated proteins in butyrate-treated Chinese hamster ovary cells. J Biol Chem. 1989, 264: 20602-20607.PubMed Dorner AJWL, Kaufman RJ: Increased synthesis of secreted proteins induces expression of glucose-regulated proteins in butyrate-treated Chinese hamster ovary cells. J Biol Chem. 1989, 264: 20602-20607.PubMed
10.
Zurück zum Zitat Schroder MKR: The mammalian unfolded protein response. Annu Rev Biochem. 2005, 74: 739-789. 10.1146/annurev.biochem.73.011303.074134.PubMedCrossRef Schroder MKR: The mammalian unfolded protein response. Annu Rev Biochem. 2005, 74: 739-789. 10.1146/annurev.biochem.73.011303.074134.PubMedCrossRef
11.
Zurück zum Zitat Zhang KKR: Identification and characterization of endoplasmic reticulum stress-induced apoptosis in vivo. Methods Enzymol. 2008, 442: 395-419. full_text.PubMedCentralPubMedCrossRef Zhang KKR: Identification and characterization of endoplasmic reticulum stress-induced apoptosis in vivo. Methods Enzymol. 2008, 442: 395-419. full_text.PubMedCentralPubMedCrossRef
13.
Zurück zum Zitat Travers KPC, Wodicka L, Lockhart DL, Weissman JS, Walter P: Functional and Genomic Analyses Reveal an Essential Coordination between the Unfolded Protein Response and ER-Associated Degradation. Cell. 2000, 101 (3): 249-258. 10.1016/S0092-8674(00)80835-1.PubMedCrossRef Travers KPC, Wodicka L, Lockhart DL, Weissman JS, Walter P: Functional and Genomic Analyses Reveal an Essential Coordination between the Unfolded Protein Response and ER-Associated Degradation. Cell. 2000, 101 (3): 249-258. 10.1016/S0092-8674(00)80835-1.PubMedCrossRef
14.
Zurück zum Zitat Brodsky SSVJL: One step at a time: endoplasmic reticulum-associated degradation. Nature Reviews Molecular Cell Biology. 2008, 9: 944-957. 10.1038/nrm2546.PubMedCentralPubMedCrossRef Brodsky SSVJL: One step at a time: endoplasmic reticulum-associated degradation. Nature Reviews Molecular Cell Biology. 2008, 9: 944-957. 10.1038/nrm2546.PubMedCentralPubMedCrossRef
15.
Zurück zum Zitat Yorimitsu TNU, Yang Z, Klionsky DJ: Endoplasmic reticulum stress triggers autophagy. J Biol Chem. 2006, 281 (40): 30299-304. 10.1074/jbc.M607007200.PubMedCentralPubMedCrossRef Yorimitsu TNU, Yang Z, Klionsky DJ: Endoplasmic reticulum stress triggers autophagy. J Biol Chem. 2006, 281 (40): 30299-304. 10.1074/jbc.M607007200.PubMedCentralPubMedCrossRef
16.
Zurück zum Zitat Guzman M: Cannabinoids: potential anticancer agents. Nat Rev Cancer. 2003, 3: 745-755. 10.1038/nrc1188.PubMedCrossRef Guzman M: Cannabinoids: potential anticancer agents. Nat Rev Cancer. 2003, 3: 745-755. 10.1038/nrc1188.PubMedCrossRef
17.
Zurück zum Zitat Velasco G, Carracedo A, Blazquez C, Lorente M, Aguado T, Haro A, Sanchez C, Galve-Roperh I, Guzman M: Cannabinoids and gliomas. Mol Neurobiol. 2007, 36: 60-67. 10.1007/s12035-007-0002-5.PubMedCrossRef Velasco G, Carracedo A, Blazquez C, Lorente M, Aguado T, Haro A, Sanchez C, Galve-Roperh I, Guzman M: Cannabinoids and gliomas. Mol Neurobiol. 2007, 36: 60-67. 10.1007/s12035-007-0002-5.PubMedCrossRef
18.
Zurück zum Zitat Verfaillie T, Salazar M, Velasco G, Agostinis P: Linking ER Stress to Autophagy: Potential Implications for Cancer Therapy. Int J Cell Biol. 2010, 930509- Verfaillie T, Salazar M, Velasco G, Agostinis P: Linking ER Stress to Autophagy: Potential Implications for Cancer Therapy. Int J Cell Biol. 2010, 930509-
19.
Zurück zum Zitat Szegezdi E, Logue SE, Gorman AM, Samali A: Mediators of endoplasmic reticulum stress-induced apoptosis. EMBO Rep. 2006, 7: 880-885. 10.1038/sj.embor.7400779.PubMedCentralPubMedCrossRef Szegezdi E, Logue SE, Gorman AM, Samali A: Mediators of endoplasmic reticulum stress-induced apoptosis. EMBO Rep. 2006, 7: 880-885. 10.1038/sj.embor.7400779.PubMedCentralPubMedCrossRef
20.
Zurück zum Zitat Heather P, Harding YZ, Bertolotti Anne, Huiqing Zeng, David Ron: Perk Is Essential for Translational Regulation and Cell Survival during the Unfolded Protein Response. Molecular Cell. 2000, 5: 897-904. 10.1016/S1097-2765(00)80330-5.CrossRef Heather P, Harding YZ, Bertolotti Anne, Huiqing Zeng, David Ron: Perk Is Essential for Translational Regulation and Cell Survival during the Unfolded Protein Response. Molecular Cell. 2000, 5: 897-904. 10.1016/S1097-2765(00)80330-5.CrossRef
21.
Zurück zum Zitat Scheuner D, Song B, McEwen E, Liu C, Laybutt R, Gillespie P, Saunders T, Bonner-Weir S, Kaufman RJ: Translational control is required for the unfolded protein response and in vivo glucose homeostasis. Mol Cell. 2001, 7: 1165-1176. 10.1016/S1097-2765(01)00265-9.PubMedCrossRef Scheuner D, Song B, McEwen E, Liu C, Laybutt R, Gillespie P, Saunders T, Bonner-Weir S, Kaufman RJ: Translational control is required for the unfolded protein response and in vivo glucose homeostasis. Mol Cell. 2001, 7: 1165-1176. 10.1016/S1097-2765(01)00265-9.PubMedCrossRef
22.
Zurück zum Zitat Wek RCCD: Translational control and the unfolded protein response. Antioxid Redox Signal. 2007, 9 (12): 2357-71. 10.1089/ars.2007.1764. 2007 9:2357-2371PubMedCrossRef Wek RCCD: Translational control and the unfolded protein response. Antioxid Redox Signal. 2007, 9 (12): 2357-71. 10.1089/ars.2007.1764. 2007 9:2357-2371PubMedCrossRef
23.
Zurück zum Zitat Xu C, Bailly-Maitre B, Reed JC: Endoplasmic reticulum stress: cell life and death decisions. J Clin Invest. 2005, 115: 2656-2664. 10.1172/JCI26373.PubMedCentralPubMedCrossRef Xu C, Bailly-Maitre B, Reed JC: Endoplasmic reticulum stress: cell life and death decisions. J Clin Invest. 2005, 115: 2656-2664. 10.1172/JCI26373.PubMedCentralPubMedCrossRef
24.
25.
Zurück zum Zitat Yan W, Frank CL, Korth MJ, Sopher BL, Novoa I, Ron D, Katze MG: Control of PERK eIF2alpha kinase activity by the endoplasmic reticulum stress-induced molecular chaperone P58IPK. Proc Natl Acad Sci USA. 2002, 99: 15920-15925. 10.1073/pnas.252341799.PubMedCentralPubMedCrossRef Yan W, Frank CL, Korth MJ, Sopher BL, Novoa I, Ron D, Katze MG: Control of PERK eIF2alpha kinase activity by the endoplasmic reticulum stress-induced molecular chaperone P58IPK. Proc Natl Acad Sci USA. 2002, 99: 15920-15925. 10.1073/pnas.252341799.PubMedCentralPubMedCrossRef
26.
Zurück zum Zitat Ye J, Rawson RB, Komuro R, Chen X, Dave UP, Prywes R, Brown MS, Goldstein JL: ER stress induces cleavage of membrane-bound ATF6 by the same proteases that process SREBPs. Mol Cell. 2000, 6: 1355-1364. 10.1016/S1097-2765(00)00133-7.PubMedCrossRef Ye J, Rawson RB, Komuro R, Chen X, Dave UP, Prywes R, Brown MS, Goldstein JL: ER stress induces cleavage of membrane-bound ATF6 by the same proteases that process SREBPs. Mol Cell. 2000, 6: 1355-1364. 10.1016/S1097-2765(00)00133-7.PubMedCrossRef
27.
Zurück zum Zitat Lee AH, Iwakoshi NN, Glimcher LH: XBP-1 regulates a subset of endoplasmic reticulum resident chaperone genes in the unfolded protein response. Mol Cell Biol. 2003, 23: 7448-7459. 10.1128/MCB.23.21.7448-7459.2003.PubMedCentralPubMedCrossRef Lee AH, Iwakoshi NN, Glimcher LH: XBP-1 regulates a subset of endoplasmic reticulum resident chaperone genes in the unfolded protein response. Mol Cell Biol. 2003, 23: 7448-7459. 10.1128/MCB.23.21.7448-7459.2003.PubMedCentralPubMedCrossRef
28.
Zurück zum Zitat Yamamoto KST, Matsui T, Sato M, Okada T, Yoshida H, Harada A, Mori K: Transcriptional induction of mammalian ER quality control proteins is mediated by single or combined action of ATF6alpha and XBP1. Dev Cell. 2007, 13: 365-376. 10.1016/j.devcel.2007.07.018.PubMedCrossRef Yamamoto KST, Matsui T, Sato M, Okada T, Yoshida H, Harada A, Mori K: Transcriptional induction of mammalian ER quality control proteins is mediated by single or combined action of ATF6alpha and XBP1. Dev Cell. 2007, 13: 365-376. 10.1016/j.devcel.2007.07.018.PubMedCrossRef
29.
Zurück zum Zitat Lee K, Tirasophon W, Shen X, Michalak M, Prywes R, Okada T, Yoshida H, Mori K, Kaufman RJ: IRE1-mediated unconventional mRNA splicing and S2P-mediated ATF6 cleavage merge to regulate XBP1 in signaling the unfolded protein response. Genes Dev. 2002, 16: 452-466. 10.1101/gad.964702.PubMedCentralPubMedCrossRef Lee K, Tirasophon W, Shen X, Michalak M, Prywes R, Okada T, Yoshida H, Mori K, Kaufman RJ: IRE1-mediated unconventional mRNA splicing and S2P-mediated ATF6 cleavage merge to regulate XBP1 in signaling the unfolded protein response. Genes Dev. 2002, 16: 452-466. 10.1101/gad.964702.PubMedCentralPubMedCrossRef
30.
Zurück zum Zitat Rao RV, Bredesen DE: Misfolded proteins, endoplasmic reticulum stress and neurodegeneration. Curr Opin Cell Biol. 2004, 16: 653-662. 10.1016/j.ceb.2004.09.012.PubMedCentralPubMedCrossRef Rao RV, Bredesen DE: Misfolded proteins, endoplasmic reticulum stress and neurodegeneration. Curr Opin Cell Biol. 2004, 16: 653-662. 10.1016/j.ceb.2004.09.012.PubMedCentralPubMedCrossRef
31.
Zurück zum Zitat Yoshida H, Matsui T, Yamamoto A, Okada T, Mori K: XBP1 mRNA is induced by ATF6 and spliced by IRE1 in response to ER stress to produce a highly active transcription factor. Cell. 2001, 107: 881-891. 10.1016/S0092-8674(01)00611-0.PubMedCrossRef Yoshida H, Matsui T, Yamamoto A, Okada T, Mori K: XBP1 mRNA is induced by ATF6 and spliced by IRE1 in response to ER stress to produce a highly active transcription factor. Cell. 2001, 107: 881-891. 10.1016/S0092-8674(01)00611-0.PubMedCrossRef
32.
Zurück zum Zitat Rao RV, Castro-Obregon S, Frankowski H, Schuler M, Stoka V, del Rio G, Bredesen DE, Ellerby HM: Coupling endoplasmic reticulum stress to the cell death program. An Apaf-1-independent intrinsic pathway. J Biol Chem. 2002, 277: 21836-21842. 10.1074/jbc.M202726200.PubMedCrossRef Rao RV, Castro-Obregon S, Frankowski H, Schuler M, Stoka V, del Rio G, Bredesen DE, Ellerby HM: Coupling endoplasmic reticulum stress to the cell death program. An Apaf-1-independent intrinsic pathway. J Biol Chem. 2002, 277: 21836-21842. 10.1074/jbc.M202726200.PubMedCrossRef
33.
Zurück zum Zitat Breckenridge DG, Germain M, Mathai JP, Nguyen M, Shore GC: Regulation of apoptosis by endoplasmic reticulum pathways. Oncogene. 2003, 22: 8608-8618. 10.1038/sj.onc.1207108.PubMedCrossRef Breckenridge DG, Germain M, Mathai JP, Nguyen M, Shore GC: Regulation of apoptosis by endoplasmic reticulum pathways. Oncogene. 2003, 22: 8608-8618. 10.1038/sj.onc.1207108.PubMedCrossRef
34.
Zurück zum Zitat Ron D, Habener JF: CHOP, a novel developmentally regulated nuclear protein that dimerizes with transcription factors C/EBP and LAP and functions as a dominant-negative inhibitor of gene transcription. Genes Dev. 1992, 6: 439-453. 10.1101/gad.6.3.439.PubMedCrossRef Ron D, Habener JF: CHOP, a novel developmentally regulated nuclear protein that dimerizes with transcription factors C/EBP and LAP and functions as a dominant-negative inhibitor of gene transcription. Genes Dev. 1992, 6: 439-453. 10.1101/gad.6.3.439.PubMedCrossRef
35.
Zurück zum Zitat Zinszner H, Kuroda M, Wang X, Batchvarova N, Lightfoot RT, Remotti H, Stevens JL, Ron D: CHOP is implicated in programmed cell death in response to impaired function of the endoplasmic reticulum. Genes Dev. 1998, 12: 982-995. 10.1101/gad.12.7.982.PubMedCentralPubMedCrossRef Zinszner H, Kuroda M, Wang X, Batchvarova N, Lightfoot RT, Remotti H, Stevens JL, Ron D: CHOP is implicated in programmed cell death in response to impaired function of the endoplasmic reticulum. Genes Dev. 1998, 12: 982-995. 10.1101/gad.12.7.982.PubMedCentralPubMedCrossRef
36.
Zurück zum Zitat Marciniak SJ, Yun CY, Oyadomari S, Novoa I, Zhang Y, Jungreis R, Nagata K, Harding HP, Ron D: CHOP induces death by promoting protein synthesis and oxidation in the stressed endoplasmic reticulum. Genes Dev. 2004, 18: 3066-3077. 10.1101/gad.1250704.PubMedCentralPubMedCrossRef Marciniak SJ, Yun CY, Oyadomari S, Novoa I, Zhang Y, Jungreis R, Nagata K, Harding HP, Ron D: CHOP induces death by promoting protein synthesis and oxidation in the stressed endoplasmic reticulum. Genes Dev. 2004, 18: 3066-3077. 10.1101/gad.1250704.PubMedCentralPubMedCrossRef
37.
Zurück zum Zitat Ma Y, Hendershot LM: The role of the unfolded protein response in tumour development: friend or foe?. Nat Rev Cancer. 2004, 4: 966-977. 10.1038/nrc1505.PubMedCrossRef Ma Y, Hendershot LM: The role of the unfolded protein response in tumour development: friend or foe?. Nat Rev Cancer. 2004, 4: 966-977. 10.1038/nrc1505.PubMedCrossRef
38.
Zurück zum Zitat Nakagawa TYJ: Cross-talk between two cysteine protease families. Activation of caspase-12 by calpain in apoptosis. J Cell Biol. 2000, 150: 887-894. 10.1083/jcb.150.4.887.PubMedCentralPubMedCrossRef Nakagawa TYJ: Cross-talk between two cysteine protease families. Activation of caspase-12 by calpain in apoptosis. J Cell Biol. 2000, 150: 887-894. 10.1083/jcb.150.4.887.PubMedCentralPubMedCrossRef
39.
Zurück zum Zitat Morishima N, Nakanishi K, Takenouchi H, Shibata T, Yasuhiko Y: An endoplasmic reticulum stress-specific caspase cascade in apoptosis. Cytochrome c-independent activation of caspase-9 by caspase-12. J Biol Chem. 2002, 277: 34287-34294. 10.1074/jbc.M204973200.PubMedCrossRef Morishima N, Nakanishi K, Takenouchi H, Shibata T, Yasuhiko Y: An endoplasmic reticulum stress-specific caspase cascade in apoptosis. Cytochrome c-independent activation of caspase-9 by caspase-12. J Biol Chem. 2002, 277: 34287-34294. 10.1074/jbc.M204973200.PubMedCrossRef
40.
Zurück zum Zitat Wu JKR: From acute ER stress to physiological roles of the Unfolded Protein Response. Cell Death Differ. 2006, 13 (3): 374-84. 10.1038/sj.cdd.4401840.PubMedCrossRef Wu JKR: From acute ER stress to physiological roles of the Unfolded Protein Response. Cell Death Differ. 2006, 13 (3): 374-84. 10.1038/sj.cdd.4401840.PubMedCrossRef
41.
Zurück zum Zitat Davenport EL, Morgan GJ, Davies FE: Untangling the unfolded protein response. Cell Cycle. 2008, 7: 865-869. 10.4161/cc.7.7.5615.PubMedCrossRef Davenport EL, Morgan GJ, Davies FE: Untangling the unfolded protein response. Cell Cycle. 2008, 7: 865-869. 10.4161/cc.7.7.5615.PubMedCrossRef
42.
Zurück zum Zitat Fernandez PM, Tabbara SO, Jacobs LK, Manning FC, Tsangaris TN, Schwartz AM, Kennedy KA, Patierno SR: Overexpression of the glucose-regulated stress gene GRP78 in malignant but not benign human breast lesions. Breast Cancer Res Treat. 2000, 59: 15-26. 10.1023/A:1006332011207.PubMedCrossRef Fernandez PM, Tabbara SO, Jacobs LK, Manning FC, Tsangaris TN, Schwartz AM, Kennedy KA, Patierno SR: Overexpression of the glucose-regulated stress gene GRP78 in malignant but not benign human breast lesions. Breast Cancer Res Treat. 2000, 59: 15-26. 10.1023/A:1006332011207.PubMedCrossRef
43.
Zurück zum Zitat Fels DR, Koumenis C: The PERK/eIF2alpha/ATF4 module of the UPR in hypoxia resistance and tumor growth. Cancer Biol Ther. 2006, 5: 723-728.PubMedCrossRef Fels DR, Koumenis C: The PERK/eIF2alpha/ATF4 module of the UPR in hypoxia resistance and tumor growth. Cancer Biol Ther. 2006, 5: 723-728.PubMedCrossRef
44.
Zurück zum Zitat Haga N, Saito S, Tsukumo Y, Sakurai J, Furuno A, Tsuruo T, Tomida A: Mitochondria regulate the unfolded protein response leading to cancer cell survival under glucose deprivation conditions. Cancer Sci. 101: 1125-1132. 10.1111/j.1349-7006.2010.01525.x. Haga N, Saito S, Tsukumo Y, Sakurai J, Furuno A, Tsuruo T, Tomida A: Mitochondria regulate the unfolded protein response leading to cancer cell survival under glucose deprivation conditions. Cancer Sci. 101: 1125-1132. 10.1111/j.1349-7006.2010.01525.x.
45.
Zurück zum Zitat Koumenis C, Wouters BG: "Translating" tumor hypoxia: unfolded protein response (UPR)-dependent and UPR-independent pathways. Mol Cancer Res. 2006, 4: 423-436. 10.1158/1541-7786.MCR-06-0150.PubMedCrossRef Koumenis C, Wouters BG: "Translating" tumor hypoxia: unfolded protein response (UPR)-dependent and UPR-independent pathways. Mol Cancer Res. 2006, 4: 423-436. 10.1158/1541-7786.MCR-06-0150.PubMedCrossRef
46.
Zurück zum Zitat Romero-Ramirez L, Cao H, Nelson D, Hammond E, Lee AH, Yoshida H, Mori K, Glimcher LH, Denko NC, Giaccia AJ: XBP1 is essential for survival under hypoxic conditions and is required for tumor growth. Cancer Res. 2004, 64: 5943-5947. 10.1158/0008-5472.CAN-04-1606.PubMedCrossRef Romero-Ramirez L, Cao H, Nelson D, Hammond E, Lee AH, Yoshida H, Mori K, Glimcher LH, Denko NC, Giaccia AJ: XBP1 is essential for survival under hypoxic conditions and is required for tumor growth. Cancer Res. 2004, 64: 5943-5947. 10.1158/0008-5472.CAN-04-1606.PubMedCrossRef
47.
Zurück zum Zitat Shajahan AN, Riggins RB, Clarke R: The role of X-box binding protein-1 in tumorigenicity. Drug News Perspect. 2009, 22: 241-246. 10.1358/dnp.2009.22.5.1378631.PubMedCentralPubMedCrossRef Shajahan AN, Riggins RB, Clarke R: The role of X-box binding protein-1 in tumorigenicity. Drug News Perspect. 2009, 22: 241-246. 10.1358/dnp.2009.22.5.1378631.PubMedCentralPubMedCrossRef
48.
Zurück zum Zitat Shuda M, Kondoh N, Imazeki N, Tanaka K, Okada T, Mori K, Hada A, Arai M, Wakatsuki T, Matsubara O: Activation of the ATF6, XBP1 and grp78 genes in human hepatocellular carcinoma: a possible involvement of the ER stress pathway in hepatocarcinogenesis. J Hepatol. 2003, 38: 605-614. 10.1016/S0168-8278(03)00029-1.PubMedCrossRef Shuda M, Kondoh N, Imazeki N, Tanaka K, Okada T, Mori K, Hada A, Arai M, Wakatsuki T, Matsubara O: Activation of the ATF6, XBP1 and grp78 genes in human hepatocellular carcinoma: a possible involvement of the ER stress pathway in hepatocarcinogenesis. J Hepatol. 2003, 38: 605-614. 10.1016/S0168-8278(03)00029-1.PubMedCrossRef
49.
Zurück zum Zitat Song MS, Park YK, Lee JH, Park K: Induction of glucose-regulated protein 78 by chronic hypoxia in human gastric tumor cells through a protein kinase C-epsilon/ERK/AP-1 signaling cascade. Cancer Res. 2001, 61: 8322-8330.PubMed Song MS, Park YK, Lee JH, Park K: Induction of glucose-regulated protein 78 by chronic hypoxia in human gastric tumor cells through a protein kinase C-epsilon/ERK/AP-1 signaling cascade. Cancer Res. 2001, 61: 8322-8330.PubMed
50.
Zurück zum Zitat Sun S, Han J, Ralph WM, Chandrasekaran A, Liu K, Auborn KJ, Carter TH: Endoplasmic reticulum stress as a correlate of cytotoxicity in human tumor cells exposed to diindolylmethane in vitro. Cell Stress Chaperones. 2004, 9: 76-87.PubMedCentralPubMedCrossRef Sun S, Han J, Ralph WM, Chandrasekaran A, Liu K, Auborn KJ, Carter TH: Endoplasmic reticulum stress as a correlate of cytotoxicity in human tumor cells exposed to diindolylmethane in vitro. Cell Stress Chaperones. 2004, 9: 76-87.PubMedCentralPubMedCrossRef
51.
Zurück zum Zitat Chen Xiaoxin, Ding Yu, Chang-Gong Liu, and SM, Yang CS: Overexpression of glucose-regulated protein 94 (Grp94) in esophageal adenocarcinomas of a rat surgical model and humans. Carcinogenesis. 2002, 23: 123-130. 10.1093/carcin/23.1.123.PubMedCrossRef Chen Xiaoxin, Ding Yu, Chang-Gong Liu, and SM, Yang CS: Overexpression of glucose-regulated protein 94 (Grp94) in esophageal adenocarcinomas of a rat surgical model and humans. Carcinogenesis. 2002, 23: 123-130. 10.1093/carcin/23.1.123.PubMedCrossRef
52.
Zurück zum Zitat Ye J, Koumenis C: ATF4, an ER stress and hypoxia-inducible transcription factor and its potential role in hypoxia tolerance and tumorigenesis. Curr Mol Med. 2009, 9: 411-416. 10.2174/156652409788167096.PubMedCrossRef Ye J, Koumenis C: ATF4, an ER stress and hypoxia-inducible transcription factor and its potential role in hypoxia tolerance and tumorigenesis. Curr Mol Med. 2009, 9: 411-416. 10.2174/156652409788167096.PubMedCrossRef
53.
Zurück zum Zitat Goldstein MG, Li Z: Heat-shock proteins in infection-mediated inflammation-induced tumorigenesis. J Hematol Oncol. 2009, 2: 5-10.1186/1756-8722-2-5.PubMedCentralPubMedCrossRef Goldstein MG, Li Z: Heat-shock proteins in infection-mediated inflammation-induced tumorigenesis. J Hematol Oncol. 2009, 2: 5-10.1186/1756-8722-2-5.PubMedCentralPubMedCrossRef
54.
Zurück zum Zitat Qu L, Huang S, Baltzis D, Rivas-Estilla AM, Pluquet O, Hatzoglou M, Koumenis C, Taya Y, Yoshimura A, Koromilas AE: Endoplasmic reticulum stress induces p53 cytoplasmic localization and prevents p53-dependent apoptosis by a pathway involving glycogen synthase kinase-3beta. Genes Dev. 2004, 18: 261-277. 10.1101/gad.1165804.PubMedCentralPubMedCrossRef Qu L, Huang S, Baltzis D, Rivas-Estilla AM, Pluquet O, Hatzoglou M, Koumenis C, Taya Y, Yoshimura A, Koromilas AE: Endoplasmic reticulum stress induces p53 cytoplasmic localization and prevents p53-dependent apoptosis by a pathway involving glycogen synthase kinase-3beta. Genes Dev. 2004, 18: 261-277. 10.1101/gad.1165804.PubMedCentralPubMedCrossRef
55.
Zurück zum Zitat Kitamura M: Biphasic, bidirectional regulation of NF-kappaB by endoplasmic reticulum stress. Antioxid Redox Signal. 2009, 11: 2353-2364. 10.1089/ars.2008.2391.PubMedCrossRef Kitamura M: Biphasic, bidirectional regulation of NF-kappaB by endoplasmic reticulum stress. Antioxid Redox Signal. 2009, 11: 2353-2364. 10.1089/ars.2008.2391.PubMedCrossRef
56.
Zurück zum Zitat Jolly C, Morimoto RI: Role of the heat shock response and molecular chaperones in oncogenesis and cell death. J Natl Cancer Inst. 2000, 92: 1564-1572. 10.1093/jnci/92.19.1564.PubMedCrossRef Jolly C, Morimoto RI: Role of the heat shock response and molecular chaperones in oncogenesis and cell death. J Natl Cancer Inst. 2000, 92: 1564-1572. 10.1093/jnci/92.19.1564.PubMedCrossRef
57.
Zurück zum Zitat So A, Hadaschik B, Sowery R, Gleave M: The role of stress proteins in prostate cancer. Curr Genomics. 2007, 8: 252-261. 10.2174/138920207781386951.PubMedCentralPubMedCrossRef So A, Hadaschik B, Sowery R, Gleave M: The role of stress proteins in prostate cancer. Curr Genomics. 2007, 8: 252-261. 10.2174/138920207781386951.PubMedCentralPubMedCrossRef
58.
Zurück zum Zitat Dong D, Ni M, Li J, Xiong S, Ye W, Virrey JJ, Mao C, Ye R, Wang M, Pen L: Critical role of the stress chaperone GRP78/BiP in tumor proliferation, survival, and tumor angiogenesis in transgene-induced mammary tumor development. Cancer Res. 2008, 68: 498-505. 10.1158/0008-5472.CAN-07-2950.PubMedCrossRef Dong D, Ni M, Li J, Xiong S, Ye W, Virrey JJ, Mao C, Ye R, Wang M, Pen L: Critical role of the stress chaperone GRP78/BiP in tumor proliferation, survival, and tumor angiogenesis in transgene-induced mammary tumor development. Cancer Res. 2008, 68: 498-505. 10.1158/0008-5472.CAN-07-2950.PubMedCrossRef
59.
Zurück zum Zitat Lee AS: GRP78 induction in cancer: therapeutic and prognostic implications. Cancer Res. 2007, 67: 3496-3499. 10.1158/0008-5472.CAN-07-0325.PubMedCrossRef Lee AS: GRP78 induction in cancer: therapeutic and prognostic implications. Cancer Res. 2007, 67: 3496-3499. 10.1158/0008-5472.CAN-07-0325.PubMedCrossRef
60.
Zurück zum Zitat Li J, Lee AS: Stress induction of GRP78/BiP and its role in cancer. Curr Mol Med. 2006, 6: 45-54. 10.2174/156652406775574523.PubMedCrossRef Li J, Lee AS: Stress induction of GRP78/BiP and its role in cancer. Curr Mol Med. 2006, 6: 45-54. 10.2174/156652406775574523.PubMedCrossRef
61.
Zurück zum Zitat Al-Rawashdeh FY, Scriven P, Cameron IC, Vergani PV, Wyld L: Unfolded protein response activation contributes to chemoresistance in hepatocellular carcinoma. Eur J Gastroenterol Hepatol. 22: 1099-1105. 10.1097/MEG.0b013e3283378405. Al-Rawashdeh FY, Scriven P, Cameron IC, Vergani PV, Wyld L: Unfolded protein response activation contributes to chemoresistance in hepatocellular carcinoma. Eur J Gastroenterol Hepatol. 22: 1099-1105. 10.1097/MEG.0b013e3283378405.
62.
Zurück zum Zitat Eunjung Lee PN, Spicer Darcy, Groshen Susan, Yu Mimi C, Lee Amy S: GRP78 as a Novel Predictor of Responsiveness Chemotherapy in Breast Cancer. Cancer Res. 2006, 66: 7849-10.1158/0008-5472.CAN-06-1660.PubMedCrossRef Eunjung Lee PN, Spicer Darcy, Groshen Susan, Yu Mimi C, Lee Amy S: GRP78 as a Novel Predictor of Responsiveness Chemotherapy in Breast Cancer. Cancer Res. 2006, 66: 7849-10.1158/0008-5472.CAN-06-1660.PubMedCrossRef
63.
Zurück zum Zitat Fu Y, Lee AS: Glucose regulated proteins in cancer progression, drug resistance and immunotherapy. Cancer Biol Ther. 2006, 5: 741-744. 10.4161/cbt.5.7.2970.PubMedCrossRef Fu Y, Lee AS: Glucose regulated proteins in cancer progression, drug resistance and immunotherapy. Cancer Biol Ther. 2006, 5: 741-744. 10.4161/cbt.5.7.2970.PubMedCrossRef
64.
Zurück zum Zitat Uramoto H, Sugio K, Oyama T, Nakata S, Ono K, Yoshimastu T, Morita M, Yasumoto K: Expression of endoplasmic reticulum molecular chaperone Grp78 in human lung cancer and its clinical significance. Lung Cancer. 2005, 49: 55-62. 10.1016/j.lungcan.2004.12.011.PubMedCrossRef Uramoto H, Sugio K, Oyama T, Nakata S, Ono K, Yoshimastu T, Morita M, Yasumoto K: Expression of endoplasmic reticulum molecular chaperone Grp78 in human lung cancer and its clinical significance. Lung Cancer. 2005, 49: 55-62. 10.1016/j.lungcan.2004.12.011.PubMedCrossRef
65.
Zurück zum Zitat Scriven P, Coulson S, Haines R, Balasubramanian S, Cross S, Wyld L: Activation and clinical significance of the unfolded protein response in breast cancer. Br J Cancer. 2009, 101: 1692-1698. 10.1038/sj.bjc.6605365.PubMedCentralPubMedCrossRef Scriven P, Coulson S, Haines R, Balasubramanian S, Cross S, Wyld L: Activation and clinical significance of the unfolded protein response in breast cancer. Br J Cancer. 2009, 101: 1692-1698. 10.1038/sj.bjc.6605365.PubMedCentralPubMedCrossRef
66.
Zurück zum Zitat Wang Q, He Z, Zhang J, Wang Y, Wang T, Tong S, Wang L, Wang S, Chen Y: Overexpression of endoplasmic reticulum molecular chaperone GRP94 and GRP78 in human lung cancer tissues and its significance. Cancer Detect Prev. 2005, 29: 544-551. 10.1016/j.cdp.2005.09.010.PubMedCrossRef Wang Q, He Z, Zhang J, Wang Y, Wang T, Tong S, Wang L, Wang S, Chen Y: Overexpression of endoplasmic reticulum molecular chaperone GRP94 and GRP78 in human lung cancer tissues and its significance. Cancer Detect Prev. 2005, 29: 544-551. 10.1016/j.cdp.2005.09.010.PubMedCrossRef
67.
Zurück zum Zitat Langer R, Feith M, Siewert JR, Wester HJ, Hoefler H: Expression and clinical significance of glucose regulated proteins GRP78 (BiP) and GRP94 (GP96) in human adenocarcinomas of the esophagus. BMC Cancer. 2008, 8: 70-10.1186/1471-2407-8-70.PubMedCentralPubMedCrossRef Langer R, Feith M, Siewert JR, Wester HJ, Hoefler H: Expression and clinical significance of glucose regulated proteins GRP78 (BiP) and GRP94 (GP96) in human adenocarcinomas of the esophagus. BMC Cancer. 2008, 8: 70-10.1186/1471-2407-8-70.PubMedCentralPubMedCrossRef
68.
Zurück zum Zitat Zheng HC, Takahashi H, Li XH, Hara T, Masuda S, Guan YF, Takano Y: Overexpression of GRP78 and GRP94 are markers for aggressive behavior and poor prognosis in gastric carcinomas. Hum Pathol. 2008, 39: 1042-1049. 10.1016/j.humpath.2007.11.009.PubMedCrossRef Zheng HC, Takahashi H, Li XH, Hara T, Masuda S, Guan YF, Takano Y: Overexpression of GRP78 and GRP94 are markers for aggressive behavior and poor prognosis in gastric carcinomas. Hum Pathol. 2008, 39: 1042-1049. 10.1016/j.humpath.2007.11.009.PubMedCrossRef
69.
Zurück zum Zitat Jamora C, Dennert G, Lee AS: Inhibition of tumor progression by suppression of stress protein GRP78/BiP induction in fibrosarcoma B/C10ME. Proc Natl Acad Sci USA. 1996, 93: 7690-7694. 10.1073/pnas.93.15.7690.PubMedCentralPubMedCrossRef Jamora C, Dennert G, Lee AS: Inhibition of tumor progression by suppression of stress protein GRP78/BiP induction in fibrosarcoma B/C10ME. Proc Natl Acad Sci USA. 1996, 93: 7690-7694. 10.1073/pnas.93.15.7690.PubMedCentralPubMedCrossRef
70.
Zurück zum Zitat Davies MP, Barraclough DL, Stewart C, Joyce KA, Eccles RM, Barraclough R, Rudland PS, Sibson DR: Expression and splicing of the unfolded protein response gene XBP-1 are significantly associated with clinical outcome of endocrine-treated breast cancer. Int J Cancer. 2008, 123: 85-88. 10.1002/ijc.23479.PubMedCrossRef Davies MP, Barraclough DL, Stewart C, Joyce KA, Eccles RM, Barraclough R, Rudland PS, Sibson DR: Expression and splicing of the unfolded protein response gene XBP-1 are significantly associated with clinical outcome of endocrine-treated breast cancer. Int J Cancer. 2008, 123: 85-88. 10.1002/ijc.23479.PubMedCrossRef
71.
Zurück zum Zitat Gomez BP, Riggins RB, Shajahan AN, Klimach U, Wang A, Crawford AC, Zhu Y, Zwart A, Wang M, Clarke R: Human X-box binding protein-1 confers both estrogen independence and antiestrogen resistance in breast cancer cell lines. Faseb J. 2007, 21: 4013-4027. 10.1096/fj.06-7990com.PubMedCrossRef Gomez BP, Riggins RB, Shajahan AN, Klimach U, Wang A, Crawford AC, Zhu Y, Zwart A, Wang M, Clarke R: Human X-box binding protein-1 confers both estrogen independence and antiestrogen resistance in breast cancer cell lines. Faseb J. 2007, 21: 4013-4027. 10.1096/fj.06-7990com.PubMedCrossRef
72.
Zurück zum Zitat Carrasco DR, Sukhdeo K, Protopopova M, Sinha R, Enos M, Carrasco DE, Zheng M, Mani M, Henderson J, Pinkus GS: The differentiation and stress response factor XBP-1 drives multiple myeloma pathogenesis. Cancer Cell. 2007, 11: 349-360. 10.1016/j.ccr.2007.02.015.PubMedCentralPubMedCrossRef Carrasco DR, Sukhdeo K, Protopopova M, Sinha R, Enos M, Carrasco DE, Zheng M, Mani M, Henderson J, Pinkus GS: The differentiation and stress response factor XBP-1 drives multiple myeloma pathogenesis. Cancer Cell. 2007, 11: 349-360. 10.1016/j.ccr.2007.02.015.PubMedCentralPubMedCrossRef
73.
Zurück zum Zitat Obeng EA, Carlson LM, Gutman DM, Harrington WJ, Lee KP, Boise LH: Proteasome inhibitors induce a terminal unfolded protein response in multiple myeloma cells. Blood. 2006, 107: 4907-4916. 10.1182/blood-2005-08-3531.PubMedCentralPubMedCrossRef Obeng EA, Carlson LM, Gutman DM, Harrington WJ, Lee KP, Boise LH: Proteasome inhibitors induce a terminal unfolded protein response in multiple myeloma cells. Blood. 2006, 107: 4907-4916. 10.1182/blood-2005-08-3531.PubMedCentralPubMedCrossRef
74.
Zurück zum Zitat Sterz JSO: The potential of proteasome inhibitors in cancer therapy. Expert Opin Investig Drugs. 2008, 17: 879-895. 10.1517/13543784.17.6.879.PubMedCrossRef Sterz JSO: The potential of proteasome inhibitors in cancer therapy. Expert Opin Investig Drugs. 2008, 17: 879-895. 10.1517/13543784.17.6.879.PubMedCrossRef
75.
Zurück zum Zitat Hoseki J, Ushioda R, Nagata K: Mechanism and components of endoplasmic reticulum-associated degradation. J Biochem. 2010, 147: 19-25. 10.1093/jb/mvp194.PubMedCrossRef Hoseki J, Ushioda R, Nagata K: Mechanism and components of endoplasmic reticulum-associated degradation. J Biochem. 2010, 147: 19-25. 10.1093/jb/mvp194.PubMedCrossRef
76.
Zurück zum Zitat Wang Q, Li L, Ye Y: Inhibition of p97-dependent protein degradation by Eeyarestatin I. J Biol Chem. 2008, 283: 7445-7454. 10.1074/jbc.M708347200.PubMedCentralPubMedCrossRef Wang Q, Li L, Ye Y: Inhibition of p97-dependent protein degradation by Eeyarestatin I. J Biol Chem. 2008, 283: 7445-7454. 10.1074/jbc.M708347200.PubMedCentralPubMedCrossRef
77.
Zurück zum Zitat Lu DP, Christopher DA: Endoplasmic reticulum stress activates the expression of a sub-group of protein disulfide isomerase genes and AtbZIP60 modulates the response in Arabidopsis thaliana. Mol Genet Genomics. 2008, 280: 199-210. 10.1007/s00438-008-0356-z.PubMedCrossRef Lu DP, Christopher DA: Endoplasmic reticulum stress activates the expression of a sub-group of protein disulfide isomerase genes and AtbZIP60 modulates the response in Arabidopsis thaliana. Mol Genet Genomics. 2008, 280: 199-210. 10.1007/s00438-008-0356-z.PubMedCrossRef
78.
Zurück zum Zitat Townsend DM, Manevich Y, He L, Xiong Y, Bowers RR, Hutchens S, Tew KD: Nitrosative stress-induced s-glutathionylation of protein disulfide isomerase leads to activation of the unfolded protein response. Cancer Res. 2009, 69: 7626-7634. 10.1158/0008-5472.CAN-09-0493.PubMedCentralPubMedCrossRef Townsend DM, Manevich Y, He L, Xiong Y, Bowers RR, Hutchens S, Tew KD: Nitrosative stress-induced s-glutathionylation of protein disulfide isomerase leads to activation of the unfolded protein response. Cancer Res. 2009, 69: 7626-7634. 10.1158/0008-5472.CAN-09-0493.PubMedCentralPubMedCrossRef
79.
Zurück zum Zitat Davenport EL, Moore HE, Dunlop AS, Sharp SY, Workman P, Morgan GJ, Davies FE: Heat shock protein inhibition is associated with activation of the unfolded protein response pathway in myeloma plasma cells. Blood. 2007, 110: 2641-2649. 10.1182/blood-2006-11-053728.PubMedCrossRef Davenport EL, Moore HE, Dunlop AS, Sharp SY, Workman P, Morgan GJ, Davies FE: Heat shock protein inhibition is associated with activation of the unfolded protein response pathway in myeloma plasma cells. Blood. 2007, 110: 2641-2649. 10.1182/blood-2006-11-053728.PubMedCrossRef
80.
Zurück zum Zitat Banerji U, Judson I, Workman P: The clinical applications of heat shock protein inhibitors in cancer - present and future. Curr Cancer Drug Targets. 2003, 3: 385-390. 10.2174/1568009033481813.PubMedCrossRef Banerji U, Judson I, Workman P: The clinical applications of heat shock protein inhibitors in cancer - present and future. Curr Cancer Drug Targets. 2003, 3: 385-390. 10.2174/1568009033481813.PubMedCrossRef
81.
Zurück zum Zitat Mimnaugh EG, Xu W, Vos M, Yuan X, Isaacs JS, Bisht KS, Gius D, Neckers L: Simultaneous inhibition of hsp 90 and the proteasome promotes protein ubiquitination, causes endoplasmic reticulum-derived cytosolic vacuolization, and enhances antitumor activity. Mol Cancer Ther. 2004, 3: 551-566.PubMedCrossRef Mimnaugh EG, Xu W, Vos M, Yuan X, Isaacs JS, Bisht KS, Gius D, Neckers L: Simultaneous inhibition of hsp 90 and the proteasome promotes protein ubiquitination, causes endoplasmic reticulum-derived cytosolic vacuolization, and enhances antitumor activity. Mol Cancer Ther. 2004, 3: 551-566.PubMedCrossRef
82.
Zurück zum Zitat Mimnaugh EG, Xu W, Vos M, Yuan X, Neckers L: Endoplasmic reticulum vacuolization and valosin-containing protein relocalization result from simultaneous hsp90 inhibition by geldanamycin and proteasome inhibition by velcade. Mol Cancer Res. 2006, 4: 667-681. 10.1158/1541-7786.MCR-06-0019.PubMedCrossRef Mimnaugh EG, Xu W, Vos M, Yuan X, Neckers L: Endoplasmic reticulum vacuolization and valosin-containing protein relocalization result from simultaneous hsp90 inhibition by geldanamycin and proteasome inhibition by velcade. Mol Cancer Res. 2006, 4: 667-681. 10.1158/1541-7786.MCR-06-0019.PubMedCrossRef
83.
Zurück zum Zitat Solit DB, Osman I, Polsky D, Panageas KS, Daud A, Goydos JS, Teitcher J, Wolchok JD, Germino FJ, Krown SE: Phase II trial of 17-allylamino-17-demethoxygeldanamycin in patients with metastatic melanoma. Clin Cancer Res. 2008, 14: 8302-8307. 10.1158/1078-0432.CCR-08-1002.PubMedCentralPubMedCrossRef Solit DB, Osman I, Polsky D, Panageas KS, Daud A, Goydos JS, Teitcher J, Wolchok JD, Germino FJ, Krown SE: Phase II trial of 17-allylamino-17-demethoxygeldanamycin in patients with metastatic melanoma. Clin Cancer Res. 2008, 14: 8302-8307. 10.1158/1078-0432.CCR-08-1002.PubMedCentralPubMedCrossRef
84.
Zurück zum Zitat Usmani SZ, Bona RD, Chiosis G, Li Z: The anti-myeloma activity of a novel purine scaffold HSP90 inhibitor PU-H71 is via inhibition of both HSP90A and HSP90B1. J Hematol Oncol. 3: 40-10.1186/1756-8722-3-40. Usmani SZ, Bona RD, Chiosis G, Li Z: The anti-myeloma activity of a novel purine scaffold HSP90 inhibitor PU-H71 is via inhibition of both HSP90A and HSP90B1. J Hematol Oncol. 3: 40-10.1186/1756-8722-3-40.
85.
Zurück zum Zitat Pyrko P, Schonthal AH, Hofman FM, Chen TC, Lee AS: The unfolded protein response regulator GRP78/BiP as a novel target for increasing chemosensitivity in malignant gliomas. Cancer Res. 2007, 67: 9809-9816. 10.1158/0008-5472.CAN-07-0625.PubMedCrossRef Pyrko P, Schonthal AH, Hofman FM, Chen TC, Lee AS: The unfolded protein response regulator GRP78/BiP as a novel target for increasing chemosensitivity in malignant gliomas. Cancer Res. 2007, 67: 9809-9816. 10.1158/0008-5472.CAN-07-0625.PubMedCrossRef
86.
Zurück zum Zitat Park HRTA, Sato S, Tsukumo Y, Yun J, Yamori T, Hayakawa Y, Tsuruo T, Shin-ya K: Effect on tumor cells of blocking survival response to glucose deprivation. J Natl Cancer Inst. 2004, 96 (17): 1300-10. 10.1093/jnci/djh243.PubMedCrossRef Park HRTA, Sato S, Tsukumo Y, Yun J, Yamori T, Hayakawa Y, Tsuruo T, Shin-ya K: Effect on tumor cells of blocking survival response to glucose deprivation. J Natl Cancer Inst. 2004, 96 (17): 1300-10. 10.1093/jnci/djh243.PubMedCrossRef
87.
Zurück zum Zitat Matsuo J, Tsukumo Y, Sakurai J, Tsukahara S, Park HR, Shin-Ya K, Watanabe T, Tsuruo T, Tomida A: Preventing the unfolded protein response via aberrant activation of 4E-binding protein 1 by versipelostatin. Cancer Sci. 2008 Matsuo J, Tsukumo Y, Sakurai J, Tsukahara S, Park HR, Shin-Ya K, Watanabe T, Tsuruo T, Tomida A: Preventing the unfolded protein response via aberrant activation of 4E-binding protein 1 by versipelostatin. Cancer Sci. 2008
88.
Zurück zum Zitat Saito S, Furuno A, Sakurai J, Sakamoto A, Park HR, Shin-Ya K, Tsuruo T, Tomida A: Chemical genomics identifies the unfolded protein response as a target for selective cancer cell killing during glucose deprivation. Cancer Res. 2009, 69: 4225-4234. 10.1158/0008-5472.CAN-08-2689.PubMedCrossRef Saito S, Furuno A, Sakurai J, Sakamoto A, Park HR, Shin-Ya K, Tsuruo T, Tomida A: Chemical genomics identifies the unfolded protein response as a target for selective cancer cell killing during glucose deprivation. Cancer Res. 2009, 69: 4225-4234. 10.1158/0008-5472.CAN-08-2689.PubMedCrossRef
89.
Zurück zum Zitat Shin-Ya K: Novel antitumor and neuroprotective substances discovered by characteristic screenings based on specific molecular targets. Biosci Biotechnol Biochem. 2005, 69: 867-872. 10.1271/bbb.69.867.PubMedCrossRef Shin-Ya K: Novel antitumor and neuroprotective substances discovered by characteristic screenings based on specific molecular targets. Biosci Biotechnol Biochem. 2005, 69: 867-872. 10.1271/bbb.69.867.PubMedCrossRef
90.
Zurück zum Zitat Backer JM, Krivoshein AV, Hamby CV, Pizzonia J, Gilbert KS, Ray YS, Brand H, Paton AW, Paton JC, Backer MV: Chaperone-targeting cytotoxin and endoplasmic reticulum stress-inducing drug synergize to kill cancer cells. Neoplasia. 2009, 11: 1165-1173.PubMedCentralPubMedCrossRef Backer JM, Krivoshein AV, Hamby CV, Pizzonia J, Gilbert KS, Ray YS, Brand H, Paton AW, Paton JC, Backer MV: Chaperone-targeting cytotoxin and endoplasmic reticulum stress-inducing drug synergize to kill cancer cells. Neoplasia. 2009, 11: 1165-1173.PubMedCentralPubMedCrossRef
91.
Zurück zum Zitat Koong DFaAC: Irestatin, a potent inhibitor of IRE1 and the unfolded protein response, is a hypoxia-selective cytotoxin and impairs tumor growth. Journal of Clinical Oncology, 2007 ASCO Annual Meeting Proceedings (Post-Meeting Edition). 2007, 25: 3514- Koong DFaAC: Irestatin, a potent inhibitor of IRE1 and the unfolded protein response, is a hypoxia-selective cytotoxin and impairs tumor growth. Journal of Clinical Oncology, 2007 ASCO Annual Meeting Proceedings (Post-Meeting Edition). 2007, 25: 3514-
92.
Zurück zum Zitat Futamura Y, Tashiro E, Hironiwa N, Kohno J, Nishio M, Shindo K, Imoto M: Trierixin, a novel Inhibitor of ER stress-induced XBP1 activation from Streptomyces sp. II. structure elucidation. J Antibiot (Tokyo). 2007, 60: 582-585.CrossRef Futamura Y, Tashiro E, Hironiwa N, Kohno J, Nishio M, Shindo K, Imoto M: Trierixin, a novel Inhibitor of ER stress-induced XBP1 activation from Streptomyces sp. II. structure elucidation. J Antibiot (Tokyo). 2007, 60: 582-585.CrossRef
93.
Zurück zum Zitat Patterson J, Palombella VJ, Fritz C, Normant E: IPI-504, a novel and soluble HSP-90 inhibitor, blocks the unfolded protein response in multiple myeloma cells. Cancer Chemother Pharmacol. 2008, 61: 923-932. 10.1007/s00280-007-0546-0.PubMedCrossRef Patterson J, Palombella VJ, Fritz C, Normant E: IPI-504, a novel and soluble HSP-90 inhibitor, blocks the unfolded protein response in multiple myeloma cells. Cancer Chemother Pharmacol. 2008, 61: 923-932. 10.1007/s00280-007-0546-0.PubMedCrossRef
94.
Zurück zum Zitat Salazar M, Carracedo A, Salanueva IJ, Hernandez-Tiedra S, Lorente M, Egia A, Vazquez P, Blazquez C, Torres S, Garcia S: Cannabinoid action induces autophagy-mediated cell death through stimulation of ER stress in human glioma cells. J Clin Invest. 2009, 119: 1359-1372. 10.1172/JCI37948.PubMedCentralPubMedCrossRef Salazar M, Carracedo A, Salanueva IJ, Hernandez-Tiedra S, Lorente M, Egia A, Vazquez P, Blazquez C, Torres S, Garcia S: Cannabinoid action induces autophagy-mediated cell death through stimulation of ER stress in human glioma cells. J Clin Invest. 2009, 119: 1359-1372. 10.1172/JCI37948.PubMedCentralPubMedCrossRef
95.
Zurück zum Zitat Liu BQ, Gao YY, Niu XF, Xie JS, Meng X, Guan Y, Wang HQ: Implication of unfolded protein response in resveratrol-induced inhibition of K562 cell proliferation. Biochem Biophys Res Commun. 2009, 391: 778-782. 10.1016/j.bbrc.2009.11.137.PubMedCrossRef Liu BQ, Gao YY, Niu XF, Xie JS, Meng X, Guan Y, Wang HQ: Implication of unfolded protein response in resveratrol-induced inhibition of K562 cell proliferation. Biochem Biophys Res Commun. 2009, 391: 778-782. 10.1016/j.bbrc.2009.11.137.PubMedCrossRef
96.
Zurück zum Zitat Banjerdpongchai R, Kongtawelert P, Khantamat O, Srisomsap C, Chokchaichamnankit D, Subhasitanont P, Svasti J: Mitochondrial and endoplasmic reticulum stress pathways cooperate in zearalenone-induced apoptosis of human leukemic cells. J Hematol Oncol. 3: 50-10.1186/1756-8722-3-50. Banjerdpongchai R, Kongtawelert P, Khantamat O, Srisomsap C, Chokchaichamnankit D, Subhasitanont P, Svasti J: Mitochondrial and endoplasmic reticulum stress pathways cooperate in zearalenone-induced apoptosis of human leukemic cells. J Hematol Oncol. 3: 50-10.1186/1756-8722-3-50.
97.
Zurück zum Zitat San Miguel JF, Schlag R, Khuageva NK, Dimopoulos MA, Shpilberg O, Kropff M, Spicka I, Petrucci MT, Palumbo A, Samoilova OS: Bortezomib plus melphalan and prednisone for initial treatment of multiple myeloma. N Engl J Med. 2008, 359: 906-917. 10.1056/NEJMoa0801479.PubMedCrossRef San Miguel JF, Schlag R, Khuageva NK, Dimopoulos MA, Shpilberg O, Kropff M, Spicka I, Petrucci MT, Palumbo A, Samoilova OS: Bortezomib plus melphalan and prednisone for initial treatment of multiple myeloma. N Engl J Med. 2008, 359: 906-917. 10.1056/NEJMoa0801479.PubMedCrossRef
98.
Zurück zum Zitat Chauhan D, Singh AV, Ciccarelli B, Richardson PG, Palladino MA, Anderson KC: Combination of novel proteasome inhibitor NPI-0052 and lenalidomide trigger in vitro and in vivo synergistic cytotoxicity in multiple myeloma. Blood. 115: 834-845. 10.1182/blood-2009-03-213009. Chauhan D, Singh AV, Ciccarelli B, Richardson PG, Palladino MA, Anderson KC: Combination of novel proteasome inhibitor NPI-0052 and lenalidomide trigger in vitro and in vivo synergistic cytotoxicity in multiple myeloma. Blood. 115: 834-845. 10.1182/blood-2009-03-213009.
99.
Zurück zum Zitat O'Connor OA, Stewart AK, Vallone M, Molineaux CJ, Kunkel LA, Gerecitano JF, Orlowski RZ: A phase 1 dose escalation study of the safety and pharmacokinetics of the novel proteasome inhibitor carfilzomib (PR-171) in patients with hematologic malignancies. Clin Cancer Res. 2009, 15: 7085-7091.PubMedCentralPubMedCrossRef O'Connor OA, Stewart AK, Vallone M, Molineaux CJ, Kunkel LA, Gerecitano JF, Orlowski RZ: A phase 1 dose escalation study of the safety and pharmacokinetics of the novel proteasome inhibitor carfilzomib (PR-171) in patients with hematologic malignancies. Clin Cancer Res. 2009, 15: 7085-7091.PubMedCentralPubMedCrossRef
100.
Zurück zum Zitat Lee AFW P, Burris HA, Papadopoulos K, Sausville EA, Rosen PJ, Mendelson DS, Infante JR, Patnaik A, Gordon MS: Updated results of a phase Ib/II study of carfilzomib (CFZ) in patients (pts) with relapsed malignancies. Journal of Clinical Oncology, 2010 ASCO Annual Meeting Proceedings (Post-Meeting Edition). 2010, 28: 8147- Lee AFW P, Burris HA, Papadopoulos K, Sausville EA, Rosen PJ, Mendelson DS, Infante JR, Patnaik A, Gordon MS: Updated results of a phase Ib/II study of carfilzomib (CFZ) in patients (pts) with relapsed malignancies. Journal of Clinical Oncology, 2010 ASCO Annual Meeting Proceedings (Post-Meeting Edition). 2010, 28: 8147-
101.
Zurück zum Zitat Paul G, Richardson BB, Berenson James: Phase II study of the proteasome inhibitor PS-341 in multiple myeloma (MM) patients (pts) with relapsed/refractory disease. Proc Am Soc Clin Oncol. 2002, 21 (abstr 40): Paul G, Richardson BB, Berenson James: Phase II study of the proteasome inhibitor PS-341 in multiple myeloma (MM) patients (pts) with relapsed/refractory disease. Proc Am Soc Clin Oncol. 2002, 21 (abstr 40):
102.
Zurück zum Zitat Roberto Piva BR, Williams Michael: CEP-18770: A novel, orally active proteasome inhibitor with a tumor-selective pharmacologic profile competitive with bortezomib. Blood. 2008, 111 (5): 2765-2775. 10.1182/blood-2007-07-100651.PubMedCrossRef Roberto Piva BR, Williams Michael: CEP-18770: A novel, orally active proteasome inhibitor with a tumor-selective pharmacologic profile competitive with bortezomib. Blood. 2008, 111 (5): 2765-2775. 10.1182/blood-2007-07-100651.PubMedCrossRef
103.
Zurück zum Zitat Richardson PG, Badros AZ, Jagannath S, Tarantolo S, Wolf JL, Albitar M, Berman D, Messina M, Anderson KC: Tanespimycin with bortezomib: activity in relapsed/refractory patients with multiple myeloma. Br J Haematol. 150: 428-437. Richardson PG, Badros AZ, Jagannath S, Tarantolo S, Wolf JL, Albitar M, Berman D, Messina M, Anderson KC: Tanespimycin with bortezomib: activity in relapsed/refractory patients with multiple myeloma. Br J Haematol. 150: 428-437.
104.
Zurück zum Zitat Richardson PG, Chanan-Khan AA, Alsina M, Albitar M, Berman D, Messina M, Mitsiades CS, Anderson KC: Tanespimycin monotherapy in relapsed multiple myeloma: results of a phase 1 dose-escalation study. Br J Haematol. 150: 438-445. Richardson PG, Chanan-Khan AA, Alsina M, Albitar M, Berman D, Messina M, Mitsiades CS, Anderson KC: Tanespimycin monotherapy in relapsed multiple myeloma: results of a phase 1 dose-escalation study. Br J Haematol. 150: 438-445.
105.
Zurück zum Zitat Heath EI, Hillman DW, Vaishampayan U, Sheng S, Sarkar F, Harper F, Gaskins M, Pitot HC, Tan W, Ivy SP: A phase II trial of 17-allylamino-17-demethoxygeldanamycin in patients with hormone-refractory metastatic prostate cancer. Clin Cancer Res. 2008, 14: 7940-7946. 10.1158/1078-0432.CCR-08-0221.PubMedCentralPubMedCrossRef Heath EI, Hillman DW, Vaishampayan U, Sheng S, Sarkar F, Harper F, Gaskins M, Pitot HC, Tan W, Ivy SP: A phase II trial of 17-allylamino-17-demethoxygeldanamycin in patients with hormone-refractory metastatic prostate cancer. Clin Cancer Res. 2008, 14: 7940-7946. 10.1158/1078-0432.CCR-08-0221.PubMedCentralPubMedCrossRef
106.
Zurück zum Zitat Pacey S, Gore M, Chao D, Banerji U, Larkin J, Sarker S, Owen K, Asad Y, Raynaud F, Walton M: A Phase II trial of 17-allylamino, 17-demethoxygeldanamycin (17-AAG, tanespimycin) in patients with metastatic melanoma. Invest New Drugs. 2010, Pacey S, Gore M, Chao D, Banerji U, Larkin J, Sarker S, Owen K, Asad Y, Raynaud F, Walton M: A Phase II trial of 17-allylamino, 17-demethoxygeldanamycin (17-AAG, tanespimycin) in patients with metastatic melanoma. Invest New Drugs. 2010,
107.
Zurück zum Zitat Kummar S, Gutierrez ME, Gardner ER, Chen X, Figg WD, Zajac-Kaye M, Chen M, Steinberg SM, Muir CA, Yancey MA: Phase I trial of 17-dimethylaminoethylamino-17-demethoxygeldanamycin (17-DMAG), a heat shock protein inhibitor, administered twice weekly in patients with advanced malignancies. Eur J Cancer. 46: 340-347. 10.1016/j.ejca.2009.10.026. Kummar S, Gutierrez ME, Gardner ER, Chen X, Figg WD, Zajac-Kaye M, Chen M, Steinberg SM, Muir CA, Yancey MA: Phase I trial of 17-dimethylaminoethylamino-17-demethoxygeldanamycin (17-DMAG), a heat shock protein inhibitor, administered twice weekly in patients with advanced malignancies. Eur J Cancer. 46: 340-347. 10.1016/j.ejca.2009.10.026.
108.
Zurück zum Zitat Lancet JE, Gojo I, Burton M, Quinn M, Tighe SM, Kersey K, Zhong Z, Albitar MX, Bhalla K, Hannah AL, Baer MR: Phase I study of the heat shock protein 90 inhibitor alvespimycin (KOS-1022, 17-DMAG) administered intravenously twice weekly to patients with acute myeloid leukemia. Leukemia. 24: 699-705. 10.1038/leu.2009.292. Lancet JE, Gojo I, Burton M, Quinn M, Tighe SM, Kersey K, Zhong Z, Albitar MX, Bhalla K, Hannah AL, Baer MR: Phase I study of the heat shock protein 90 inhibitor alvespimycin (KOS-1022, 17-DMAG) administered intravenously twice weekly to patients with acute myeloid leukemia. Leukemia. 24: 699-705. 10.1038/leu.2009.292.
109.
Zurück zum Zitat Ramanathan RK, Egorin MJ, Erlichman C, Remick SC, Ramalingam SS, Naret C, Holleran JL, TenEyck CJ, Ivy SP, Belani CP: Phase I pharmacokinetic and pharmacodynamic study of 17-dimethylaminoethylamino-17-demethoxygeldanamycin, an inhibitor of heat-shock protein 90, in patients with advanced solid tumors. J Clin Oncol. 28: 1520-1526. 10.1200/JCO.2009.25.0415. Ramanathan RK, Egorin MJ, Erlichman C, Remick SC, Ramalingam SS, Naret C, Holleran JL, TenEyck CJ, Ivy SP, Belani CP: Phase I pharmacokinetic and pharmacodynamic study of 17-dimethylaminoethylamino-17-demethoxygeldanamycin, an inhibitor of heat-shock protein 90, in patients with advanced solid tumors. J Clin Oncol. 28: 1520-1526. 10.1200/JCO.2009.25.0415.
110.
Zurück zum Zitat V. Zismanov LD, Gottfried M: Targeting ER-Golgi homeostasis as a therapeutic strategy in lung cancer. J Clin Oncol. 2010, suppl; abstr e21030 V. Zismanov LD, Gottfried M: Targeting ER-Golgi homeostasis as a therapeutic strategy in lung cancer. J Clin Oncol. 2010, suppl; abstr e21030
111.
Zurück zum Zitat Hanson BE, Vesole DH: Retaspimycin hydrochloride (IPI-504): a novel heat shock protein inhibitor as an anticancer agent. Expert Opin Investig Drugs. 2009, 18: 1375-1383. 10.1517/13543780903158934.PubMedCrossRef Hanson BE, Vesole DH: Retaspimycin hydrochloride (IPI-504): a novel heat shock protein inhibitor as an anticancer agent. Expert Opin Investig Drugs. 2009, 18: 1375-1383. 10.1517/13543780903158934.PubMedCrossRef
112.
Zurück zum Zitat Caldas-Lopes E, Cerchietti L, Ahn JH, Clement CC, Robles AI, Rodina A, Moulick K, Taldone T, Gozman A, Guo Y: Hsp90 inhibitor PU-H71, a multimodal inhibitor of malignancy, induces complete responses in triple-negative breast cancer models. Proc Natl Acad Sci USA. 2009, 106: 8368-8373. 10.1073/pnas.0903392106.PubMedCentralPubMedCrossRef Caldas-Lopes E, Cerchietti L, Ahn JH, Clement CC, Robles AI, Rodina A, Moulick K, Taldone T, Gozman A, Guo Y: Hsp90 inhibitor PU-H71, a multimodal inhibitor of malignancy, induces complete responses in triple-negative breast cancer models. Proc Natl Acad Sci USA. 2009, 106: 8368-8373. 10.1073/pnas.0903392106.PubMedCentralPubMedCrossRef
113.
Zurück zum Zitat Marubayashi S, Koppikar P, Taldone T, Abdel-Wahab O, West N, Bhagwat N, Caldas-Lopes E, Ross KN, Gonen M, Gozman A: HSP90 is a therapeutic target in JAK2-dependent myeloproliferative neoplasms in mice and humans. J Clin Invest. 120: 3578-3593. 10.1172/JCI42442. Marubayashi S, Koppikar P, Taldone T, Abdel-Wahab O, West N, Bhagwat N, Caldas-Lopes E, Ross KN, Gonen M, Gozman A: HSP90 is a therapeutic target in JAK2-dependent myeloproliferative neoplasms in mice and humans. J Clin Invest. 120: 3578-3593. 10.1172/JCI42442.
114.
Zurück zum Zitat T. Bachleitner-Hofmann MYS, Chen C, Zeng Z: ntitumor activity of SNX-2112, a synthetic heat shock protein 90 inhibitor, in malignancies with amplification of the MET oncogene. J Clin Oncol. 2010, 28 (suppl; abstr e13561): T. Bachleitner-Hofmann MYS, Chen C, Zeng Z: ntitumor activity of SNX-2112, a synthetic heat shock protein 90 inhibitor, in malignancies with amplification of the MET oncogene. J Clin Oncol. 2010, 28 (suppl; abstr e13561):
115.
Zurück zum Zitat Cross BC, McKibbin C, Callan AC, Roboti P, Piacenti M, Rabu C, Wilson CM, Whitehead R, Flitsch SL, Pool MR: Eeyarestatin I inhibits Sec61-mediated protein translocation at the endoplasmic reticulum. J Cell Sci. 2009, 122: 4393-4400. 10.1242/jcs.054494.PubMedCentralPubMedCrossRef Cross BC, McKibbin C, Callan AC, Roboti P, Piacenti M, Rabu C, Wilson CM, Whitehead R, Flitsch SL, Pool MR: Eeyarestatin I inhibits Sec61-mediated protein translocation at the endoplasmic reticulum. J Cell Sci. 2009, 122: 4393-4400. 10.1242/jcs.054494.PubMedCentralPubMedCrossRef
116.
Zurück zum Zitat Luo T, Wang J, Yin Y, Hua H, Jing J, Sun X, Li M, Zhang Y, Jiang Y: (-)-Epigallocatechin gallate sensitizes breast cancer cells to paclitaxel in a murine model of breast carcinoma. Breast Cancer Res. 12: R8-10.1186/bcr2473. Luo T, Wang J, Yin Y, Hua H, Jing J, Sun X, Li M, Zhang Y, Jiang Y: (-)-Epigallocatechin gallate sensitizes breast cancer cells to paclitaxel in a murine model of breast carcinoma. Breast Cancer Res. 12: R8-10.1186/bcr2473.
117.
Zurück zum Zitat D. Feldman ACK: Irestatin, a potent inhibitor of IRE1α and the unfolded protein response, is a hypoxia-selective cytotoxin and impairs tumor growth. Journal of Clinical Oncology. 2007, ASCO Annual Meeting Proceedings Part I, 25 (18S (June 20 Supplement)): 3514-2007 D. Feldman ACK: Irestatin, a potent inhibitor of IRE1α and the unfolded protein response, is a hypoxia-selective cytotoxin and impairs tumor growth. Journal of Clinical Oncology. 2007, ASCO Annual Meeting Proceedings Part I, 25 (18S (June 20 Supplement)): 3514-2007
118.
Zurück zum Zitat Guzman M, Duarte MJ, Blazquez C, Ravina J, Rosa MC, Galve-Roperh I, Sanchez C, Velasco G, Gonzalez-Feria L: A pilot clinical study of Delta9-tetrahydrocannabinol in patients with recurrent glioblastoma multiforme. Br J Cancer. 2006, 95: 197-203. 10.1038/sj.bjc.6603236.PubMedCentralPubMedCrossRef Guzman M, Duarte MJ, Blazquez C, Ravina J, Rosa MC, Galve-Roperh I, Sanchez C, Velasco G, Gonzalez-Feria L: A pilot clinical study of Delta9-tetrahydrocannabinol in patients with recurrent glioblastoma multiforme. Br J Cancer. 2006, 95: 197-203. 10.1038/sj.bjc.6603236.PubMedCentralPubMedCrossRef
Metadaten
Titel
Unfolded protein response in cancer: the Physician's perspective
verfasst von
Xuemei Li
Kezhong Zhang
Zihai Li
Publikationsdatum
01.12.2011
Verlag
BioMed Central
Erschienen in
Journal of Hematology & Oncology / Ausgabe 1/2011
Elektronische ISSN: 1756-8722
DOI
https://doi.org/10.1186/1756-8722-4-8

Weitere Artikel der Ausgabe 1/2011

Journal of Hematology & Oncology 1/2011 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.