Skip to main content
Erschienen in: BMC Cancer 1/2011

Open Access 01.12.2011 | Research article

Major histocompatibility complex class I-related chain A/B (MICA/B) expression in tumor tissue and serum of pancreatic cancer: Role of uric acid accumulation in gemcitabine-induced MICA/B expression

verfasst von: Xiulong Xu, Geetha S Rao, Veronika Groh, Thomas Spies, Paolo Gattuso, Howard L Kaufman, Janet Plate, Richard A Prinz

Erschienen in: BMC Cancer | Ausgabe 1/2011

Abstract

Background

Major histocompatibility complex class I-related chain A and B (MICA/B) are two stress-inducible ligands that bind the immunoreceptor NKG2D and play an important role in mediating the cyotoxicity of NK and T cells. In this study, we sought to study MICA/B expression in pancreatic cancer and to determine whether and how genotoxic drugs such as gemcitabine can affect MICA/B expression and natural killer cytotoxity.

Methods

Seven pancreatic cancer cell lines were analyzed for MICA/B expression by flow cytometry and for their sensitivity to NK-92 cell killing by a 51Cr release assay. MICA/B expression in tumor tissues and sera of pancreatic cancer was analyzed by immunohistochemical staining (IHC) and ELISA, respectively.

Results

Two MICA/B-positive cell lines were sensitive to the cytotoxic activity of NK-92 cells. Other two MICA/B-positive cell lines and three MICA/B-negative cell lines were resistant to NK-92 cell killing. MICA/B expression was positive in 17 of 25 (68%) pancreatic ductal adenocarcinomas but not in normal pancreatic ductal epithelial cells. Serum MICA/B levels were significantly elevated in patients with pancreatic adenocarcinomas but did not correlate with the stage of pancreatic cancer and patient survival. Gemcitabine therapy led to increased serum MICA levels in 6 of 10 patients with detectable serum MICA. Allopurinol, an inhibitor of xanthine oxidoreductase that converts xanthine to uric acid, blocked uric acid production, MICA/B expression, and sensitivity to NK-92 cell killing toward a PANC-1 cancer cell line exposed to radiation and two genotoxic drugs, gemcitabine and 5-fluorouracil.

Conclusions

The levels of MICA/B expression in serum and tissue of pancreatic cancer are elevated. DNA damage-induced MICA/B expression is mediated through increased uric acid production.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1471-2407-11-194) contains supplementary material, which is available to authorized users.

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

All authors read and approved the final manuscript.
XX, GR, PG, J, & RAP: Conducted basic, pathological and clinical studies.
XX, VG, TS, HLK, JP, & RAP, conceived and executed the project, writing and/or critically reading the manuscript
Abkürzungen
5-FU
5-fluorouracil
Gem
emcitabine
IHC
immunohistochemistry
MHC
major histocompatibility complex
MICA/B
major histocompatibility complex class I-related chain A and B
MMP
matrix metalloproteinase
NK
natural killer
sMICA
soluble MICA.

Background

Pancreatic cancer remains one of the most lethal human cancers and causes > 30,000 deaths per year in the United States [1]. Conventional treatments, such as surgery, radiation, and chemotherapy, or combination of these approaches, have had little impact on patient survival [1]. Over the past decade, improved understanding and knowledge of the immune system have generated novel strategies for immunotherapy [2]. While the success of tumor immunotherapy primarily relies on the identification of tumor antigens, the expression of transformation-associated stress genes commonly provokes innate immune reactions. These responses could be exploited to develop immunotherapeutic approaches to treat cancer [3].
MICA is a glycosylated, polymorphic and membrane-anchored non-classical MHC class I molecule [4, 5]. The structure of MICA resembles other MHC class I heavy chains. However, MICA is not associated with β2 microglobulin, lacks a CD8 binding site and does not present any antigens [4, 5]. MICA is broadly expressed in a variety of malignancies, including melanoma, breast, colon and hepatocellular cancers [68]. MICA can be cleaved by matrix metalloproteinases and ADAM proteinase, and released into the blood stream or tissue culture medium as a soluble molecule (sMICA) [7, 911]. MICA functions as a ligand for NKG2D, an important immunoreceptor expressed on NK cells, CD8 and γδ T cells [5]. The interaction of MICA and NKG2D plays an important role in immune surveillance by both innate immunity and adaptive immunity. In vitro studies have provided strong evidence that MICA is critical for the susceptibility of target cells to NK cells, CD8 cytotoxic T cells, and γδ T cells [5]. Antibodies that block the interaction of MICA and NKG2D can inhibit NK and T cell-mediated cytolysis [5]. Tumor cells stably expressing NKG2D ligands at high level are rejected by CD8 T cells and/or NK cells [12]. Mice immunized with NKG2/D ligand-transfected tumor cells develop adaptive immunity against re-challenge with the parental tumor cell lines [13].
Gemcitabine is a first-line chemotherapy drug for pancreatic cancer [14]. Gemcitabine alone or in combination with 5-fluorouracil (5-FU) or radiation treatment can prolong the survival of pancreatic cancer patients [14]. We have recently characterized the change of immune cells in pancreatic cancer patients treated with gemcitabine [15]. Our data suggest that gemcitabine therapy may decrease memory T-cells and promote naive T-cell activation, and that gemcitabine therapy is not immunosuppressive but rather may enhance antitumor immunity induced by tumor vaccine [15]. Our present study aims at analyzing MICA/B expression in pancreatic tumor tissues and sera, and determining if gemcitabine can stimulate antitumor immunity by inducing MICA/B expression on pancreatic cancer cells.

Materials and methods

Reagents and cell lines

Anti-MICA/B mAbs (Clones 6D4 and SR99) have been previously described [6, 1618]. Both antibodies can be used in flow cytometric analysis to detect MICA/B cell surface expression. SR99 but not 6D4 can be used in immunohistochemical staining to monitor MICA/B expression in sections of paraffin-embedded tumor blocks. A polyclonal anti-MICA rabbit IgG was purchased from Santa Cruz Biotechnology, Inc. (Santa Cruz, CA). CAPAN-1, CAPAN-2, COLO-587, HPAF-II, MiaPaCa, PANC-1, and MPANC-96 cell lines were purchased from the American Type Culture Collection (Manassas, VA). MRO87, originally thought to be a thyroid cancer cell line but later identified as a colon adenocarcinoma cell line, was included as a positive control [19]. CAPAN-1, CAPAN-2, and HPAF-II cells were grown in complete MEM medium containing 10% FBS, non-essential amino acids, sodium pyruvate, HEPES, penicillin and streptomycilin. COLO-587 and MRO87 cells were grown in complete RPMI 1640 medium containing 10% FBS. PANC-1 cells were grown in complete RPMI 1640 medium containing 10% FBS, non-essential amino acids, sodium pyruvate, HEPES, penicillin and streptomycilin. MPANC-96 and MiaPaCa were grown in complete DMEM medium containing 10% FBS. NK-92 cells were grown in MyeloCult H5100 medium (StemCell Technologies, Vancouver, British Columbia, Canada) supplemented with IL-2 (100 units/ml) (R&D Systems).

Patient information, tumor specimens and blood samples

The use of specimens from human subjects was approved by the Institutional Review Board of Rush University Medical Center. Paraffin-embedded tissue blocks derived from patients with either primary pancreatic adenocarcinomas and/or metastases were obtained from the Department of Pathology. The stage of the pancreatic adenocarcinomas was classified according to the TNM scheme used by the AJCC. Blood samples were collected from 61 patients with pancreatic ductal adenocarcinoma. Ten of them were treated with gemcitabine (Gemzar®; Eli Lilly, Indianapolis, IN) reconstituted from lyophilized powder in 200-mg and 1000-mg aliquots. Patients were treated at a dose of 1000 mg/m2 weekly by an intravenous infusion in 250 mL over 30 minutes. Patients were treated for 3 weeks. Blood samples were drawn before (day 0) and on day 3, 7, 14, 21 after gemcitabine treatment. Serum samples were prepared, aliquoted, and stored at -80°C until analysis of sMICA using a sandwich ELISA as described below.

Flow cytometric analysis

The monolayers of MRO87 and 7 pancreatic cancer cell lines were harvested when they reached approximately 80% confluence. To determine whether DNA damage stimulated MICA/B expression in pancreatic cancer cells, PANC-1 cells grown in 6-well plates were preincubated with allopurinol (250 μg/ml) for 1 hr, followed by treatment with 5-FU (10 μM), gemcitabine (2 μM) or radiation (40 Gy). PANC-1 cells were also treated with uric acid crystals (200 μg/ml) and analyzed for MICA/B expression. Cell surface MICA/B was stained with an anti-MICA/B mAb (Clone 6D4) and analyzed on live cells, which were gated on the forward scattering, in a FACScan flow cytometer (Becton Dickinson, Palo Alto, CA) as previous described [20].

Chromium release assay

Pancreatic cancer cell lines were grown in T-25 flasks. Upon 90% confluence, the cell monolayers were washed and treated with Cell Dissociation Solution (Sigma, St. Louis, MO). Single cell suspensions were labeled with 51Cr ( 50 μ Ci per 1 × 106 cells) at 37°C for 1 hr. The cells (5000 per well) were aliquoted in triplicate in a 96-well U-bottom plate in the absence or presence of NK-92 cells with a ratio of effector to target at 25:1, 5:1, or 1:1. Cells were incubated at 37°C in a humidified CO2 incubator for 4 hr. The supernatants were collected, transferred to Ready-Caps, and the radioactivity was measured in a scintillation counter (Becton Dickinson, Palo Alto, CA). To determine whether genotoxic drugs and radiation can sensitize pancreatic cancer cells for NK-92 cell killing, PANC-1 cells grown in 6-well plates were preincubated with 5-FU (10 μM), gemcitabine (2 μM) or radiation (40 Gy) in the absence or presence of allopurinol (250 μg/ml). The experiments were conducted in triplicate and repeated at least twice with similar results.

Immunohistochemical (IHC) analysis

Tissue sections were de-paraffinized with xylene and rehydrated. MICA/B were stained with an anti-MICA/B mAb (SR99) as previously described [20]. Two investigators (X.X. and P. G.) graded MICA/B expression in a blinded fashion. MICA/B expression was confirmed with a rabbit polyclonal anti-MICA/B IgG with a similar immunohistochemical staining procedure as previously published [20]. Negative MICA/B expression was graded as no MICA/B signal (-)with weak signal (+) in less than 20% of tumor cells. MICA/B-positive tumors were defined as weak (+), moderate (++) or strong (+++) MICA/B signal in more than 20% of tumor cells.

ELISA

A MICA ELISA kit (IMMATICS Biotechnologies, Tubigen, Germany) was used to detect sMICA in sera following a protocol described previously [21]. Plates were coated with the AMO1 capture mAb against MICA in PBS. After incubation overnight at 4°C, the plates were blocked with PBS containing 5% bovine serum albumin for 1 hr at 37°C and washed. Standard recombinant sMICA and serum samples (diluted 1:3) were added to the plates and incubated for 2 hrs at 37°C. After 3x wash, detection mAb BAMO3 (IgG2a specific for MICA/B) was added and incubated at 37°C for 2 hrs. The plates were washed and incubated at 37°C with HRP- conjugated anti mouse IgG2a. Color was developed using ABTS (2,2-Azino-bis-(3-ethylbenzenthiazoline-6-sulfnoic acid). Absorbance was measured at 450 nm. A standard curve of the logarithimic relationship between concentration and absorbance was used to calculate the sMICA concentration in serum samples.

Quantification of uric acid

PANC-1 cells were left untreated or treated with 5-FU (10 μM), gemcitabine (2 μM) or irradiated with 40 G in the absence or presence of allopurinol (250 μg/ml) for 24 hr. Cells were collected by trypsinization and counted with Trypan Blue staining in a hemacytometer. The cells were then washed 3 times with PBS and lysed at 5 × 105 cells/100 μl in the buffer containing Tris-HCl, 50 mM, pH 8.0; 2 mM EDTA, 1% Triton X-100 and followed by a 10-second homogenization. The lysates were incubated on ice for 30 min then centrifuged at 2000 g for 15 min. The supernatants were analyzed for uric acid concentration by using a uric acid kit (BioAssay Systems, Hayward, CA).

Statistical analysis

Chi-square tests were performed to analyze the difference in the clinicopathologic parameters of MICA/B-positive and MICA/B-negative pancreatic ductal adenocarcinomas for significant association. Student t test was used to analyze the difference in NK-92 cell-mediated cytotoxicity. Mann-Whitney U test was used to compare serum MICA levels between the control group and pancreatic cancer patients. Paired student t test was used to determine whether serum MICA/B levels were significantly changed before (day 0) and on day 3 after gemcitabine treatment. The survival of pancreatic cancer patients with serum MICA/B activity at a cutoff of 200 pg/ml was determined according to the Kaplan-Meier method, and the difference was evaluated by the log-rank test. A p value less than 0.05 was considered statistically significant. All statistics was conducted by using SigmaStat 3 software (Richmond, CA).

Results

MICA/B expression in pancreatic cancer cell lines

MICA/B expression was analyzed in 7 pancreatic cancer cell lines by flow cytometry. MICA/B expression was detected in a positive control cell line (MRO87) and in 4 pancreatic cancer cell lines (PANC-1, MPANC96, HPAF-II, and CAPAN-1) but not in the other 3 cell lines (COLO-587, MiaPaCa, and CAPAN-2) (Figure 1A). Next, MICA/B expression was correlated with their sensitivity to cytotoxic activity mediated by NK-92 cells, a natural killer cell line that expresses NKG2D receptor [20]. Three MICA/B-negative cell lines (COLO-587, MiaPaCa, and CAPAN-2) were not sensitive to NK-92 cell killing (Figure 1B). Percent cytolysis of these cell lines by NK-92 cells assayed with a ratio of effector to target cells of 5:1 or 1:1 were less than 10%. Two of four MICA/B-positive cell lines, CAPAN-1 and PANC-1, were sensitive to NK-92 cell-mediated killing, with a percent cytolysis of approximately 20% when the ratio of effector to target cells was 5:1 or 1:1. Two MICA/B-positive cell lines, MPANC96 and CAPAN-1, were resistant to NK-92 cell killing.

MICA/B expression in pancreatic ductal adenocarcinomas

Twenty-five pancreatic cancer specimens from 25 patients (15 male and 10 female) with a mean age of 65 ± 11 (median age: 65, range: 38-81 years) were analyzed for MICA/B expression by immunohistochemical staining according our previous publication [18]. These 25 samples included 23 primary adenocarcinomas and 3 metastatic implants (one in the peritoneum, one in the liver, and one in the gallbladder). IHC staining with an anti-MICA/B mAb (SR99) (Figure 2,A-C) revealed the strong dark brown signal of MICA/B expression present in the tumor cells (Figure 2C) but not in normal ductal cells (Figure 2A) nor in a cyst adenoma (Figure 2B). Negative control IgG did not show any signal (Figure 2D). MICA/B expression was confirmed with a polyclonal anti-MICA/B antibody (Figure 2E). A MICA/B-negative pancreatic adenocarcinoma is shown in Figure 2F. Eighteen of (68%) of 25 pancreatic adenocarcinomas were positive for MICA/B (Table 1). Among these MICA/B-positive tumors, MICA/B expression was graded as weak, moderate, and strong in 2, 5, 11 samples, respectively. Clinicopathologic analyses did not reveal that MICA/B expression was associated with patient gender, tumor stage, and lymph node metastasis, and differentiation (Table 1).
Table 1
MICA/B expression in 25 pancreatic cancers and clinicopatholgoic significance
Gender
Total
MICA/B+
p value
Female
10
7
 
Male
15
11
0.66
Tumor stage
   
T1
3
1
 
T2
9
7
 
T3
8
6
> 0.05
T4
1
1
 
Metastases
3
3
 
Lymph node invasion
   
Yes
14
10
 
No
8
5
1.00
Tumor differenation
   
Poorly differentiated
3
2
 
Well differentiated
22
16
0.47

Increased serum MICA levels in pancreatic cancer patients

The median and mean serum MICA levels in 61 patients were 228 pg/ml and 1107 ± 46 pg/ml respectively. The median and mean sMICA concentrations in 26 healthy control subjects were 30 and 211 ± 18 pg/ml, respectively (Table 2). Statistical analyses revealed that serum MICA levels were significantly increased in pancreatic cancer patients, compared to those in the healthy controls (p = 0.002) (Table 2).
Table 2
Increased serum sMICA levels and its clinicopathologic significance
 
Number
Median
Mean ± SE
p value
Healthy donors
26
30
211 ± 18
 
Pancreatic cancer patients
61
228
1107 ± 46
0.002
Female
25
220
461 ± 173
 
Male
36
238
1556 ± 582
0.901
with surgery
24
145
390 ± 177
 
without surgery
37
267
1572 ± 565
0.018
Stage I-III
15
144
437 ± 40
 
Stage IV
46
231
1325 ± 69
0.178
No difference in serum MICA levels amongst male and female patients was revealed. The mean and median serum MICA levels in 25 female patients were 220 and 461 ± 173 pg/ml respectively; whereas the mean and median serum MICA levels in 36 male patients were 238 and 1556.2 ± 582 pg/ml, respectively (Table 2). Mann-Whitney test revealed that there was no significant difference in serum MICA levels between female and male patients (p = 0.901). The association between serum MICA levels with different tumor stages was also determined. The median and mean serum MICA levels in 15 patients with tumor stages I-III were 144 and 437 ± 40 pg/ml respectively, compared to 231 and 1325 ± 69 pg/ml in 46 patients with a stage IV tumor (Table 2). Statistical analyses revealed that MICA levels in patients with tumor stages I-III were not significantly lower than those with a stage IV tumor (p = 0.178). Surgical resection is an option for patients with earlier stage disease, hence serum MICA levels in 24 patients who underwent surgery were lower than those who did not. The median and mean serum levels in 24 patients who underwent Whipple procedure (9 cases) or tumor resection (15 cases) were 145 and 390 ± 177 pg/respectively, whereas the median and mean serum levels in 37 patients (14 cases with palliative surgery and 23 cases without any surgery) were 267 and 1572 ± 565 pg/ml, respectively (Table 2). Mann-Whitney test revealed a p value of 0.018, indicating that serum MICA levels in patients who were eligible for tumor resecton is significantly lower than those with unresectable tumors.
Pancreatic cancer patients were divided into two groups according to a cutoff value of 200 pg/ml of serum MICA levels. The mean survival of these 16 pancreatic cancer patients with serum MICA levels < 200 pg/ml was 10.3 ± 7.6 months; whereas the mean survival of 45 pancreatic cancer patients with serum MICA levels > 200 pg/ml was 10.3 ± 4.9 months (Figure 3A). Serum MICA/B levels was not associated with survival in pancreatic cancer patients (p = 0.233). We next tested whether gemcitabine treatment led to increased serum MICA levels in pancreatic cancer. Blood samples were taken from 10 patients before (day 0) and on day 3, 7, 14, and 28 after drug administration. Serum MICA levels were increased in 6 of 10 patients with detectable serum MICA levels (Figure 3B). Paired student t test analysis revealed that serum sMICA levels were significantly higher 3 days after gemcitabine treatment (day 0) than those before treatment (p = 0.011). Serum MICA levels in 4 patients with undetectable serum MICA level before gemcitabine administration remained undetectable during 4 weeks of gemcitabine treatment.

Allopurinol blocks DNA damage-induced MICA/B expression, uric acid production, and sensitivity of PANC-1 cells to NK cell killing

The ability of gemcitabine and other DNA damage reagents to induce MICA/B expression was verified in vitro. Gemcitabine, 5-FU and radiation all led to increased cell surface MICA/B expression in PANC-1 cells (Figure 4A). Allopurinol, an inhibitor of xanthine oxidoreductase that converts xanthine to uric acid, was added to PANC-1 cells 1 hr prior to the addition of gemcitabine, 5-FU, or irradiation. Allopurinol was able to block the induction of MICA/B expression in the cells exposed to 5-FU, gemcitabine, or radiation (Figure 4A). To confirm a role of uric acid in mediating DNA damage-induced MICA/B expression, exogenous uric acid crystals were added and successfully induced MICA/B expression in PANC-1 cells (Figure 4A). Analysis of uric acid concentrations in PANC-1 cell lysates revealed that uric acid levels were increased by approximately 3-fold in PANC-1 cells treated with gemcitabine, 5-FU, and radiation (Figure 4B), compared to untreated controls. Allopurinol blocked the induction of uric acid accumulation (Figure 4B). Of note, Trypan blue staining revealed that 5-FU, gemcitabine, or radiation treatment during 24 hr incubation did not lead to a significant number of dead cells (< 5% of stained cells), suggesting that increased uric acid concentrations in DNA-damaged cells were not due to the variation caused by uncounted dead cells.
Pre-treatment of PANC-1 cells with gemcitabine, 5-FU, or radiation led to a significant increase of the percent 51Cr release (19-27%) in DNA-damaged cells, compared to that in untreated or allopurinol-treated cells (7-8%) (Figure 5A & 5B). Allopurinol alone did not have any effect on the sensitivity of PANC-1 to NK-92 cell killing. However, allopuinol blocked DNA damaged-induced sensitization of PANC-1 cells to NK-92 cytotoxicity (Figure 5A & 5B). These observations suggest that uric acid production and increased MICA/B expression play an important role in sensitizing pancreatic cancer cells to NK-92 cell-mediated cytotoxicity.

Discussion

It has been well documented that MICA/B expression is increased in several types of malignancy. A very recent study showed that MICA/B expression was detected in 92 of 103 pancreatic ductal adenocarcinomas from a cohort of Chinese patients [22]. Marten et al. reported that MICA/B was expressed in three pancreatic cancer cell lines, including PANC-1, DNA-G, and PatSci [21]. Our study confirms the expression of MICA/B in pancreatic adenocarcinomas, showing that 17 of 25 pancreatic adenocarcinomas (68%) were positive for MICA/B expression; and that MICA/B expression was detected in 4 of 7 pancreatic cancer cell lines. The molecular mechanisms of increased MICA/B expression in pancreatic cancer are unknown. Our prior study suggests that MAP kinase activation due to BRAF gene mutation in thyroid cancer may contribute to increased MICA/B expression [20]. KRAS is mutated in more than 95% of pancreatic cancers [14]. We speculate that MAP kinase activation due to RAS gene mutation may in part contribute to increased MICA/B expression. Further epigenetic changes may be required for MICA/B overexpression since MICA/B promoter methylation regulates MICA/B gene expression in hepatomas [23].
Ectopic expression of NKG2D ligands Rae1β or H60 in several murine tumor cell lines leads to the sensitization of these cells to immune cell-mediated cytolysis and tumor rejection [13]. In contrast to murine NKG2D ligands, tumor-derived soluble MICA/B molecules can induce endocytosis and degradation of NKG2D on both tumor-infiltrating and peripheral-blood lymphocytes from patients with cancer [7, 24]. Thus, serum MICA/B molecules released from tumor cells act as a negative force to counteract the effect of membrane-bound MICA/B in immune surveillance and sensitization for immune cell killing. Cell surface levels of NKG2D in NK and T cells from cancer patients are decreased, subsequently leading to the loss of cytotoxic activity [22, 25]. Therefore, MICA/B expression in pancreatic cancer and subsequent release as soluble molecules may have a very intriguing impact in clinical outcome. In this study, we found that elevated serum MICA levels were not associated with a prolonged or shorter survival. In contrast, Duan et al. [22] recently reported that the mean survival of pancreatic cancer patients with low serum sMICA levels are significantly longer than those with high serum MICA levels, whereas the mean survival of patients with low MICA/B expression in pancreatic tumor tissues are significantly shorter than those with high MICA/B expression in tumor tissues. We have no explanation for this discrepancy. We found that serum sMICA levels in patients with stage IV pancreatic cancer were higher than those with stage I to III cancer, but this was not statistically significant. An earlier study showed that sMICA levels in patients with stage IV pancreatic cancer are significantly higher than those with stage III cancer [21].
NK-92 cells have a superior effect over lymphokine activated killer cells and other NK cell lines such as the YT cell line [26], largely because of an abundant expression of perforin and granzyme B [27]. Romanski et al. [28] showed that NK-92 cells can selectively kill a panel of leukemia cell lines in a MICA/B-dependent manner. NK-92 cells moderately express the NKG2D receptor and two other NK activation receptors, NKp30 and NKp46 [27]. NK-92 cells preferentially kill a panel of MICA/B-positive tumor cell lines [20]. In this study, we demonstrated that NK-92 cells were able to kill two MICA/B positive pancreatic cancer cell lines (PANC-1 & CAPAN-1) but had only minimal effect on other two MICA/B-positive cell lines (HPAF-II and MPANC-96). Marten et al. [21] found that NK-92 cells are able to kill HLA-ABC-positive PANC-1 cells with similar potency. The resistance of MICA/B-positive pancreatic cancer to NK-92 cell killing could be due to the inhibitory effect of co-expressed HLA-G antigen that suppresses NK cell function [2931]. Three MICA/B-negative cell lines, CAPAN-2, COLO-587, and MiaPaCa, displayed very low to moderate sensitivity to NK-92 cells. It is likely that other NK activation receptors or other members of MICA/B such as ULBP1-4 and Letal, a recently identified NKG2D ligand [32, 33], may also participate in NK-92 cell-mediated cytotoxic activity against MICA/B-negative tumor cell lines [21].
Activation of the DNA damage pathway leads to increased expression of NKG2D ligands in several murine tumor cell lines [34]. Other non-genotoxic anticancer drugs can also increase MICA/B and other NKG2D ligand expression in various tumor cell lines and in patients [3537]. Mechanistic studies suggest that increased cell surface MICA/B levels by anticancer drugs could be due to transcriptional up-regulation of MICA/B gene expression or due to the suppression of proteinases that cleave MICA/B [3537]. Our prior study showed that MAP kinase activation due to RAS or BRAF oncogene activation contributed to increased MICA/B expression [20]. Additional study showed that DNA damage induced by genotoxic drugs and radiation leads to uric acid accumulation and MAP kinase activation, subsequently resulting in increased MICA/B expression in MRO87 and HeLa cell lines (Manuscript under review). Our in vitro study showed that uric acid crystals were able to induce MICA/B expression in PANC-1 cells, and that the blockade of uric acid production by allopurinol abrogated DNA damage-induced uric acid production and MICA/B expression. Our clinical study showed that gemcitabine treatment led to a transient increase of serum sMICA levels in 6 of 10 pancreatic cancer patients. These observations collectively suggest that uric acid accumulation in DNA-damaged pancreatic cancer cells plays an important role in mediating genotoxic drug- and radiation-induced MICA/B expression. Consistent with this notion, we were able to detect the expression of xanthine oxidoreductase, a metabolic enzyme that generates uric acid, in PANC-1 cell line and in pancreatic adenocarcinomas (X. Xu, unpublished observations).
Since MICA/B expression in PANC-1 cells plays a critical role in NK-92 cell-mediated cytotoxicity [21], it is highly likely that increased MICA/B expression contributes significantly to DNA damage-enhanced sensitivity of PANC-1 cells to NK-92 cell killing. Increased MICA/B expression in HeLa and MRO87 cells by uric acid crystals also leads to increased sensitivity to NK-92 cell killing. It should be noted that other NKG2D ligands or Fas can be up-regulated by DNA damage too and may also sensitize tumor cells to NK cell killing. Our clinical study showed that gemcitabine treatment led to a transient increase of serum MICA levels in pancreatic cancer patients. The highest levels of serum sMICA were on day 3 after gemcitabine administration. Serum sMICA declined slightly thereafter, which could be due to tumoricidal effect of gemcitabine or due to the suppression of proteinase gene expression. Since soluble MICA/B can antagonize membrane-bound MICA/B-mediated antitumor immunity, the impact of genotoxic drug- or radiation-induced MICA/B expression in pancreatic tumor cells in a clinical setting remains unknown. Recent clinical trials revealed that pancreatic cancer patients treated with chemotherapy followed by immunization with a GM-CSF-transfected pancreatic cancer cell line developed a strong antitumor immunity and prolonged patient survival [38, 39]. Combinational use of proteinase inhibitors that block MICA/B cleavage or use of chemotherapeutic drugs that can also suppress proteinase expression may further improve the therapeutic outcome of immunotherapy for pancreactic cancer.

Conclusions

Our study has demonstrated that serum MICA levels and MICA/B expression in pancreatic tumor tissues are elevated. Radiation and genotoxic drugs are able to induce MICA/B expression in pancreatic cancer cells through the accumulation of uric acid. Gemcitabine therapy leads to increased serum MICA levels, probably as a result of increased MICA expression in tumor tissues.

Acknowledgements

The authors would like to thank Dr. Sophie Caillat-Zucman (INSERM U561, Equipe Avenir, Hôpital Saint-Vincent de Paul, Paris) for kindly providing the anti-MICA/B mAb (Clone SR99), Dr. Hans-G. Klingemann (Tufts-New England Medical Center, Boston) for kindly providing the NK-92 cell line. This work was supported in part by a grant from American Cancer Society, IL Division (X. Xu) and by the Eli Lilly Research Laboratories (J. Plate).
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 read and approved the final manuscript.
XX, GR, PG, J, & RAP: Conducted basic, pathological and clinical studies.
XX, VG, TS, HLK, JP, & RAP, conceived and executed the project, writing and/or critically reading the manuscript
Literatur
1.
Zurück zum Zitat Li D, Xie K, Wolff R, Abbruzzese JL: Pancreatic cancer. Lancet. 2004, 363: 1049-1057. 10.1016/S0140-6736(04)15841-8.CrossRefPubMed Li D, Xie K, Wolff R, Abbruzzese JL: Pancreatic cancer. Lancet. 2004, 363: 1049-1057. 10.1016/S0140-6736(04)15841-8.CrossRefPubMed
2.
Zurück zum Zitat Waldmann TA: Immunotherapy: past, present and future. Nat Med. 2003, 9: 269-277. 10.1038/nm0303-269.CrossRefPubMed Waldmann TA: Immunotherapy: past, present and future. Nat Med. 2003, 9: 269-277. 10.1038/nm0303-269.CrossRefPubMed
5.
Zurück zum Zitat Raulet DH: Roles of the NKG2D immunoreceptor and its ligands. Nat Rev Immunol. 2003, 3: 781-790. 10.1038/nri1199.CrossRefPubMed Raulet DH: Roles of the NKG2D immunoreceptor and its ligands. Nat Rev Immunol. 2003, 3: 781-790. 10.1038/nri1199.CrossRefPubMed
6.
Zurück zum Zitat Groh V, Rhinehart R, Secrist H, Bauer S, Grabstein KH, Spies T: Broad tumor-associated expression and recognition by tumor-derived gamma delta T cells of MICA and MICB. Proc Natl Acad Sci USA. 1999, 96: 6879-6884. 10.1073/pnas.96.12.6879.CrossRefPubMedPubMedCentral Groh V, Rhinehart R, Secrist H, Bauer S, Grabstein KH, Spies T: Broad tumor-associated expression and recognition by tumor-derived gamma delta T cells of MICA and MICB. Proc Natl Acad Sci USA. 1999, 96: 6879-6884. 10.1073/pnas.96.12.6879.CrossRefPubMedPubMedCentral
7.
Zurück zum Zitat Groh V, Wu J, Yee C, Spies T: Tumour-derived soluble MIC ligands impair expression of NKG2D and T-cell activation. Nature. 2002, 419: 734-738. 10.1038/nature01112.CrossRefPubMed Groh V, Wu J, Yee C, Spies T: Tumour-derived soluble MIC ligands impair expression of NKG2D and T-cell activation. Nature. 2002, 419: 734-738. 10.1038/nature01112.CrossRefPubMed
8.
Zurück zum Zitat Pende D, Rivera P, Marcenaro S, Chang CC, Biassoni R, Conte R, Kubin M, Cosman D, Ferrone S, Moretta L, Moretta A: Major histocompatibility complex class I-related chain A and UL16-binding protein expression on tumor cell lines of different histotypes: analysis of tumor susceptibility to NKG2D-dependent natural killer cell cytotoxicity. Cancer Res. 2002, 62: 6178-6186.PubMed Pende D, Rivera P, Marcenaro S, Chang CC, Biassoni R, Conte R, Kubin M, Cosman D, Ferrone S, Moretta L, Moretta A: Major histocompatibility complex class I-related chain A and UL16-binding protein expression on tumor cell lines of different histotypes: analysis of tumor susceptibility to NKG2D-dependent natural killer cell cytotoxicity. Cancer Res. 2002, 62: 6178-6186.PubMed
9.
Zurück zum Zitat Salih HR, Rammensee HG, Steinle A: Cutting edge: down-regulation of MICA on human tumors by proteolytic shedding. J Immunol. 2002, 169: 4098-4102.CrossRefPubMed Salih HR, Rammensee HG, Steinle A: Cutting edge: down-regulation of MICA on human tumors by proteolytic shedding. J Immunol. 2002, 169: 4098-4102.CrossRefPubMed
10.
Zurück zum Zitat Waldhauer I, Goehlsdorf D, Gieseke F, Weinschenk T, Wittenbrink M, Ludwig A, Stevanovic S, Rammensee HG, Steinle A: Tumor-associated MICA is shed by ADAM proteases. Cancer Res. 2008, 68: 6368-6376. 10.1158/0008-5472.CAN-07-6768.CrossRefPubMed Waldhauer I, Goehlsdorf D, Gieseke F, Weinschenk T, Wittenbrink M, Ludwig A, Stevanovic S, Rammensee HG, Steinle A: Tumor-associated MICA is shed by ADAM proteases. Cancer Res. 2008, 68: 6368-6376. 10.1158/0008-5472.CAN-07-6768.CrossRefPubMed
11.
Zurück zum Zitat Liu G, Atteridge CL, Wang X, Lundgren AD, Wu JD: The membrane type matrix metalloproteinase MMP14 mediates constitutive shedding of MHC class I chain-related molecule A independent of A disintegrin and metalloproteinases. J Immunol. 184: 3346-3350. Liu G, Atteridge CL, Wang X, Lundgren AD, Wu JD: The membrane type matrix metalloproteinase MMP14 mediates constitutive shedding of MHC class I chain-related molecule A independent of A disintegrin and metalloproteinases. J Immunol. 184: 3346-3350.
12.
Zurück zum Zitat Hayakawa Y, Kelly JM, Westwood JA, Darcy PK, Diefenbach A, Raulet D, Smyth MJ: Cutting edge: tumor rejection mediated by NKG2D receptor-ligand interaction is dependent upon perforin. J Immunol. 2002, 169: 5377-5381.CrossRefPubMed Hayakawa Y, Kelly JM, Westwood JA, Darcy PK, Diefenbach A, Raulet D, Smyth MJ: Cutting edge: tumor rejection mediated by NKG2D receptor-ligand interaction is dependent upon perforin. J Immunol. 2002, 169: 5377-5381.CrossRefPubMed
13.
Zurück zum Zitat Diefenbach A, Jensen ER, Jamieson AM, Raulet DH: Rae1 and H60 ligands of the NKG2D receptor stimulate tumour immunity. Nature. 2001, 413: 165-171. 10.1038/35093109.CrossRefPubMedPubMedCentral Diefenbach A, Jensen ER, Jamieson AM, Raulet DH: Rae1 and H60 ligands of the NKG2D receptor stimulate tumour immunity. Nature. 2001, 413: 165-171. 10.1038/35093109.CrossRefPubMedPubMedCentral
14.
Zurück zum Zitat Hidalgo M: Pancreatic cancer. N Engl J Med. 362: 1605-1617. Hidalgo M: Pancreatic cancer. N Engl J Med. 362: 1605-1617.
15.
Zurück zum Zitat Plate JM, Plate AE, Shott S, Bograd S, Harris JE: Effect of gemcitabine on immune cells in subjects with adenocarcinoma of the pancreas. Cancer Immunol Immunother. 2005, 54: 915-925. 10.1007/s00262-004-0638-1.CrossRefPubMed Plate JM, Plate AE, Shott S, Bograd S, Harris JE: Effect of gemcitabine on immune cells in subjects with adenocarcinoma of the pancreas. Cancer Immunol Immunother. 2005, 54: 915-925. 10.1007/s00262-004-0638-1.CrossRefPubMed
16.
Zurück zum Zitat Groh V, Bahram S, Bauer S, Herman A, Beauchamp M, Spies T: Cell stress-regulated human major histocompatibility complex class I gene expressed in gastrointestinal epithelium. Proc Natl Acad Sci USA. 1996, 93: 12445-12450. 10.1073/pnas.93.22.12445.CrossRefPubMedPubMedCentral Groh V, Bahram S, Bauer S, Herman A, Beauchamp M, Spies T: Cell stress-regulated human major histocompatibility complex class I gene expressed in gastrointestinal epithelium. Proc Natl Acad Sci USA. 1996, 93: 12445-12450. 10.1073/pnas.93.22.12445.CrossRefPubMedPubMedCentral
17.
Zurück zum Zitat Groh V, Steinle A, Bauer S, Spies T: Recognition of stress-induced MHC molecules by intestinal epithelial gammadelta T cells. Science. 1998, 279: 1737-1740. 10.1126/science.279.5357.1737.CrossRefPubMed Groh V, Steinle A, Bauer S, Spies T: Recognition of stress-induced MHC molecules by intestinal epithelial gammadelta T cells. Science. 1998, 279: 1737-1740. 10.1126/science.279.5357.1737.CrossRefPubMed
18.
Zurück zum Zitat Hue S, Monteiro RC, Berrih-Aknin S, Caillat-Zucman S: Potential role of NKG2D/MHC class I-related chain A interaction in intrathymic maturation of single-positive CD8 T cells. J Immunol. 2003, 171: 1909-1917.CrossRefPubMed Hue S, Monteiro RC, Berrih-Aknin S, Caillat-Zucman S: Potential role of NKG2D/MHC class I-related chain A interaction in intrathymic maturation of single-positive CD8 T cells. J Immunol. 2003, 171: 1909-1917.CrossRefPubMed
19.
Zurück zum Zitat Schweppe RE, Klopper JP, Korch C, Pugazhenthi U, Benezra M, Knauf JA, Fagin JA, Marlow LA, Copland JA, Smallridge RC, Haugen BR: Deoxyribonucleic acid profiling analysis of 40 human thyroid cancer cell lines reveals cross-contamination resulting in cell line redundancy and misidentification. J Clin Endocrinol Metab. 2008, 93: 4331-4341. 10.1210/jc.2008-1102.CrossRefPubMedPubMedCentral Schweppe RE, Klopper JP, Korch C, Pugazhenthi U, Benezra M, Knauf JA, Fagin JA, Marlow LA, Copland JA, Smallridge RC, Haugen BR: Deoxyribonucleic acid profiling analysis of 40 human thyroid cancer cell lines reveals cross-contamination resulting in cell line redundancy and misidentification. J Clin Endocrinol Metab. 2008, 93: 4331-4341. 10.1210/jc.2008-1102.CrossRefPubMedPubMedCentral
20.
Zurück zum Zitat Xu X, Rao G, Gaffud MJ, Ding HG, Maki G, Klingemann HG, Groh V, Spies T, Caillat-Zucman S, Gattuso P, Plate J, Prinz RA: Clinicopathologic significance of major histocompatibility complex class I-related chain A and B (MICA/B) expression in thyroid cancer. J Clin Endocrinol Metab. 2006 Xu X, Rao G, Gaffud MJ, Ding HG, Maki G, Klingemann HG, Groh V, Spies T, Caillat-Zucman S, Gattuso P, Plate J, Prinz RA: Clinicopathologic significance of major histocompatibility complex class I-related chain A and B (MICA/B) expression in thyroid cancer. J Clin Endocrinol Metab. 2006
21.
Zurück zum Zitat Marten A, von Lilienfeld-Toal M, Buchler MW, Schmidt J: Soluble MIC is elevated in the serum of patients with pancreatic carcinoma diminishing gammadelta T cell cytotoxicity. Int J Cancer. 2006, 119: 2359-2365. 10.1002/ijc.22186.CrossRefPubMed Marten A, von Lilienfeld-Toal M, Buchler MW, Schmidt J: Soluble MIC is elevated in the serum of patients with pancreatic carcinoma diminishing gammadelta T cell cytotoxicity. Int J Cancer. 2006, 119: 2359-2365. 10.1002/ijc.22186.CrossRefPubMed
22.
Zurück zum Zitat Duan X, Deng L, Chen X, Lu Y, Zhang Q, Zhang K, Hu Y, Zeng J, Sun W: Clinical significance of the immunostimulatory MHC class I chain-related molecule A and NKG2D receptor on NK cells in pancreatic cancer. Med Oncol. Duan X, Deng L, Chen X, Lu Y, Zhang Q, Zhang K, Hu Y, Zeng J, Sun W: Clinical significance of the immunostimulatory MHC class I chain-related molecule A and NKG2D receptor on NK cells in pancreatic cancer. Med Oncol.
23.
Zurück zum Zitat Jinushi M, Takehara T, Tatsumi T, Kanto T, Groh V, Spies T, Kimura R, Miyagi T, Mochizuki K, Sasaki Y, Hayashi N: Expression and role of MICA and MICB in human hepatocellular carcinomas and their regulation by retinoic acid. Int J Cancer. 2003, 104: 354-361. 10.1002/ijc.10966.CrossRefPubMed Jinushi M, Takehara T, Tatsumi T, Kanto T, Groh V, Spies T, Kimura R, Miyagi T, Mochizuki K, Sasaki Y, Hayashi N: Expression and role of MICA and MICB in human hepatocellular carcinomas and their regulation by retinoic acid. Int J Cancer. 2003, 104: 354-361. 10.1002/ijc.10966.CrossRefPubMed
24.
Zurück zum Zitat Groh V, Bruhl A, El-Gabalawy H, Nelson JL, Spies T: Stimulation of T cell autoreactivity by anomalous expression of NKG2D and its MIC ligands in rheumatoid arthritis. Proc Natl Acad Sci USA. 2003, 100: 9452-9457. 10.1073/pnas.1632807100.CrossRefPubMedPubMedCentral Groh V, Bruhl A, El-Gabalawy H, Nelson JL, Spies T: Stimulation of T cell autoreactivity by anomalous expression of NKG2D and its MIC ligands in rheumatoid arthritis. Proc Natl Acad Sci USA. 2003, 100: 9452-9457. 10.1073/pnas.1632807100.CrossRefPubMedPubMedCentral
25.
Zurück zum Zitat Wu JD, Higgins LM, Steinle A, Cosman D, Haugk K, Plymate SR: Prevalent expression of the immunostimulatory MHC class I chain-related molecule is counteracted by shedding in prostate cancer. J Clin Invest. 2004, 114: 560-568.CrossRefPubMedPubMedCentral Wu JD, Higgins LM, Steinle A, Cosman D, Haugk K, Plymate SR: Prevalent expression of the immunostimulatory MHC class I chain-related molecule is counteracted by shedding in prostate cancer. J Clin Invest. 2004, 114: 560-568.CrossRefPubMedPubMedCentral
26.
Zurück zum Zitat Tonn T, Becker S, Esser R, Schwabe D, Seifried E: Cellular immunotherapy of malignancies using the clonal natural killer cell line NK-92. J Hematother Stem Cell Res. 2001, 10: 535-544. 10.1089/15258160152509145.CrossRefPubMed Tonn T, Becker S, Esser R, Schwabe D, Seifried E: Cellular immunotherapy of malignancies using the clonal natural killer cell line NK-92. J Hematother Stem Cell Res. 2001, 10: 535-544. 10.1089/15258160152509145.CrossRefPubMed
27.
Zurück zum Zitat Maki G, Klingemann HG, Martinson JA, Tam YK: Factors regulating the cytotoxic activity of the human natural killer cell line, NK-92. J Hematother Stem Cell Res. 2001, 10: 369-383. 10.1089/152581601750288975.CrossRefPubMed Maki G, Klingemann HG, Martinson JA, Tam YK: Factors regulating the cytotoxic activity of the human natural killer cell line, NK-92. J Hematother Stem Cell Res. 2001, 10: 369-383. 10.1089/152581601750288975.CrossRefPubMed
28.
Zurück zum Zitat Romanski A, Bug G, Becker S, Kampfmann M, Seifried E, Hoelzer D, Ottmann OG, Tonn T: Mechanisms of resistance to natural killer cell-mediated cytotoxicity in acute lymphoblastic leukemia. Exp Hematol. 2005, 33: 344-352. 10.1016/j.exphem.2004.11.006.CrossRefPubMed Romanski A, Bug G, Becker S, Kampfmann M, Seifried E, Hoelzer D, Ottmann OG, Tonn T: Mechanisms of resistance to natural killer cell-mediated cytotoxicity in acute lymphoblastic leukemia. Exp Hematol. 2005, 33: 344-352. 10.1016/j.exphem.2004.11.006.CrossRefPubMed
29.
Zurück zum Zitat Paul P, Rouas-Freiss N, Khalil-Daher I, Moreau P, Riteau B, Le Gal FA, Avril MF, Dausset J, Guillet JG, Carosella ED: HLA-G expression in melanoma: a way for tumor cells to escape from immunosurveillance. Proc Natl Acad Sci USA. 1998, 95: 4510-4515. 10.1073/pnas.95.8.4510.CrossRefPubMedPubMedCentral Paul P, Rouas-Freiss N, Khalil-Daher I, Moreau P, Riteau B, Le Gal FA, Avril MF, Dausset J, Guillet JG, Carosella ED: HLA-G expression in melanoma: a way for tumor cells to escape from immunosurveillance. Proc Natl Acad Sci USA. 1998, 95: 4510-4515. 10.1073/pnas.95.8.4510.CrossRefPubMedPubMedCentral
30.
Zurück zum Zitat Wiendl H, Mitsdoerffer M, Hofmeister V, Wischhusen J, Bornemann A, Meyermann R, Weiss EH, Melms A, Weller M: A functional role of HLA-G expression in human gliomas: an alternative strategy of immune escape. J Immunol. 2002, 168: 4772-4780.CrossRefPubMed Wiendl H, Mitsdoerffer M, Hofmeister V, Wischhusen J, Bornemann A, Meyermann R, Weiss EH, Melms A, Weller M: A functional role of HLA-G expression in human gliomas: an alternative strategy of immune escape. J Immunol. 2002, 168: 4772-4780.CrossRefPubMed
31.
Zurück zum Zitat Menier C, Riteau B, Carosella ED, Rouas-Freiss N: MICA triggering signal for NK cell tumor lysis is counteracted by HLA-G1-mediated inhibitory signal. Int J Cancer. 2002, 100: 63-70. 10.1002/ijc.10460.CrossRefPubMed Menier C, Riteau B, Carosella ED, Rouas-Freiss N: MICA triggering signal for NK cell tumor lysis is counteracted by HLA-G1-mediated inhibitory signal. Int J Cancer. 2002, 100: 63-70. 10.1002/ijc.10460.CrossRefPubMed
32.
Zurück zum Zitat Conejo-Garcia JR, Benencia F, Courreges MC, Gimotty PA, Khang E, Buckanovich RJ, Frauwirth KA, Zhang L, Katsaros D, Thompson CB, Levine B, Coukos G: Ovarian carcinoma expresses the NKG2D ligand Letal and promotes the survival and expansion of CD28- antitumor T cells. Cancer Res. 2004, 64: 2175-2182. 10.1158/0008-5472.CAN-03-2194.CrossRefPubMed Conejo-Garcia JR, Benencia F, Courreges MC, Gimotty PA, Khang E, Buckanovich RJ, Frauwirth KA, Zhang L, Katsaros D, Thompson CB, Levine B, Coukos G: Ovarian carcinoma expresses the NKG2D ligand Letal and promotes the survival and expansion of CD28- antitumor T cells. Cancer Res. 2004, 64: 2175-2182. 10.1158/0008-5472.CAN-03-2194.CrossRefPubMed
33.
Zurück zum Zitat Conejo-Garcia JR, Benencia F, Courreges MC, Khang E, Zhang L, Mohamed-Hadley A, Vinocur JM, Buckanovich RJ, Thompson CB, Levine B, Coukos G, Letal : A tumor-associated NKG2D immunoreceptor ligand, induces activation and expansion of effector immune cells. Cancer Biol Ther. 2003, 2: 446-451.CrossRefPubMed Conejo-Garcia JR, Benencia F, Courreges MC, Khang E, Zhang L, Mohamed-Hadley A, Vinocur JM, Buckanovich RJ, Thompson CB, Levine B, Coukos G, Letal : A tumor-associated NKG2D immunoreceptor ligand, induces activation and expansion of effector immune cells. Cancer Biol Ther. 2003, 2: 446-451.CrossRefPubMed
34.
Zurück zum Zitat Gasser S, Orsulic S, Brown EJ, Raulet DH: The DNA damage pathway regulates innate immune system ligands of the NKG2D receptor. Nature. 2005, 436: 1186-1190. 10.1038/nature03884.CrossRefPubMedPubMedCentral Gasser S, Orsulic S, Brown EJ, Raulet DH: The DNA damage pathway regulates innate immune system ligands of the NKG2D receptor. Nature. 2005, 436: 1186-1190. 10.1038/nature03884.CrossRefPubMedPubMedCentral
35.
Zurück zum Zitat Kohga K, Takehara T, Tatsumi T, Miyagi T, Ishida H, Ohkawa K, Kanto T, Hiramatsu N, Hayashi N: Anticancer chemotherapy inhibits MHC class I-related chain a ectodomain shedding by downregulating ADAM10 expression in hepatocellular carcinoma. Cancer Res. 2009, 69: 8050-8057. 10.1158/0008-5472.CAN-09-0789.CrossRefPubMed Kohga K, Takehara T, Tatsumi T, Miyagi T, Ishida H, Ohkawa K, Kanto T, Hiramatsu N, Hayashi N: Anticancer chemotherapy inhibits MHC class I-related chain a ectodomain shedding by downregulating ADAM10 expression in hepatocellular carcinoma. Cancer Res. 2009, 69: 8050-8057. 10.1158/0008-5472.CAN-09-0789.CrossRefPubMed
36.
Zurück zum Zitat Kim JY, Bae JH, Lee SH, Lee EY, Chung BS, Kim SH, Kang CD: Induction of NKG2D ligands and subsequent enhancement of NK cell-mediated lysis of cancer cells by arsenic trioxide. J Immunother. 2008, 31: 475-486. 10.1097/CJI.0b013e3181755deb.CrossRefPubMed Kim JY, Bae JH, Lee SH, Lee EY, Chung BS, Kim SH, Kang CD: Induction of NKG2D ligands and subsequent enhancement of NK cell-mediated lysis of cancer cells by arsenic trioxide. J Immunother. 2008, 31: 475-486. 10.1097/CJI.0b013e3181755deb.CrossRefPubMed
37.
Zurück zum Zitat Armeanu S, Krusch M, Baltz KM, Weiss TS, Smirnow I, Steinle A, Lauer UM, Bitzer M, Salih HR: Direct and natural killer cell-mediated antitumor effects of low-dose bortezomib in hepatocellular carcinoma. Clin Cancer Res. 2008, 14: 3520-3528. 10.1158/1078-0432.CCR-07-4744.CrossRefPubMed Armeanu S, Krusch M, Baltz KM, Weiss TS, Smirnow I, Steinle A, Lauer UM, Bitzer M, Salih HR: Direct and natural killer cell-mediated antitumor effects of low-dose bortezomib in hepatocellular carcinoma. Clin Cancer Res. 2008, 14: 3520-3528. 10.1158/1078-0432.CCR-07-4744.CrossRefPubMed
38.
Zurück zum Zitat Laheru D, Lutz E, Burke J, Biedrzycki B, Solt S, Onners B, Tartakovsky I, Nemunaitis J, Le D, Sugar E, Hege K, Jaffee E: Allogeneic granulocyte macrophage colony-stimulating factor-secreting tumor immunotherapy alone or in sequence with cyclophosphamide for metastatic pancreatic cancer: a pilot study of safety, feasibility, and immune activation. Clin Cancer Res. 2008, 14: 1455-1463. 10.1158/1078-0432.CCR-07-0371.CrossRefPubMedPubMedCentral Laheru D, Lutz E, Burke J, Biedrzycki B, Solt S, Onners B, Tartakovsky I, Nemunaitis J, Le D, Sugar E, Hege K, Jaffee E: Allogeneic granulocyte macrophage colony-stimulating factor-secreting tumor immunotherapy alone or in sequence with cyclophosphamide for metastatic pancreatic cancer: a pilot study of safety, feasibility, and immune activation. Clin Cancer Res. 2008, 14: 1455-1463. 10.1158/1078-0432.CCR-07-0371.CrossRefPubMedPubMedCentral
39.
Zurück zum Zitat Emens LA, Asquith JM, Leatherman JM, Kobrin BJ, Petrik S, Laiko M, Levi J, Daphtary MM, Biedrzycki B, Wolff AC, Stearns V, Disis ML, Ye X, Piantadosi S, Fetting JH, Davidson NE, Jaffee EM: Timed sequential treatment with cyclophosphamide, doxorubicin, and an allogeneic granulocyte-macrophage colony-stimulating factor-secreting breast tumor vaccine: a chemotherapy dose-ranging factorial study of safety and immune activation. J Clin Oncol. 2009, 27: 5911-5918. 10.1200/JCO.2009.23.3494.CrossRefPubMedPubMedCentral Emens LA, Asquith JM, Leatherman JM, Kobrin BJ, Petrik S, Laiko M, Levi J, Daphtary MM, Biedrzycki B, Wolff AC, Stearns V, Disis ML, Ye X, Piantadosi S, Fetting JH, Davidson NE, Jaffee EM: Timed sequential treatment with cyclophosphamide, doxorubicin, and an allogeneic granulocyte-macrophage colony-stimulating factor-secreting breast tumor vaccine: a chemotherapy dose-ranging factorial study of safety and immune activation. J Clin Oncol. 2009, 27: 5911-5918. 10.1200/JCO.2009.23.3494.CrossRefPubMedPubMedCentral
Metadaten
Titel
Major histocompatibility complex class I-related chain A/B (MICA/B) expression in tumor tissue and serum of pancreatic cancer: Role of uric acid accumulation in gemcitabine-induced MICA/B expression
verfasst von
Xiulong Xu
Geetha S Rao
Veronika Groh
Thomas Spies
Paolo Gattuso
Howard L Kaufman
Janet Plate
Richard A Prinz
Publikationsdatum
01.12.2011
Verlag
BioMed Central
Erschienen in
BMC Cancer / Ausgabe 1/2011
Elektronische ISSN: 1471-2407
DOI
https://doi.org/10.1186/1471-2407-11-194

Weitere Artikel der Ausgabe 1/2011

BMC Cancer 1/2011 Zur Ausgabe

Umsetzung der POMGAT-Leitlinie läuft

03.05.2024 DCK 2024 Kongressbericht

Seit November 2023 gibt es evidenzbasierte Empfehlungen zum perioperativen Management bei gastrointestinalen Tumoren (POMGAT) auf S3-Niveau. Vieles wird schon entsprechend der Empfehlungen durchgeführt. Wo es im Alltag noch hapert, zeigt eine Umfrage in einem Klinikverbund.

CUP-Syndrom: Künstliche Intelligenz kann Primärtumor finden

30.04.2024 Künstliche Intelligenz Nachrichten

Krebserkrankungen unbekannten Ursprungs (CUP) sind eine diagnostische Herausforderung. KI-Systeme können Pathologen dabei unterstützen, zytologische Bilder zu interpretieren, um den Primärtumor zu lokalisieren.

Sind Frauen die fähigeren Ärzte?

30.04.2024 Gendermedizin Nachrichten

Patienten, die von Ärztinnen behandelt werden, dürfen offenbar auf bessere Therapieergebnisse hoffen als Patienten von Ärzten. Besonders gilt das offenbar für weibliche Kranke, wie eine Studie zeigt.

Adjuvante Immuntherapie verlängert Leben bei RCC

25.04.2024 Nierenkarzinom Nachrichten

Nun gibt es auch Resultate zum Gesamtüberleben: Eine adjuvante Pembrolizumab-Therapie konnte in einer Phase-3-Studie das Leben von Menschen mit Nierenzellkarzinom deutlich verlängern. Die Sterberate war im Vergleich zu Placebo um 38% geringer.

Update Onkologie

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