Local delivery of recombinant adenovirus expressing hepatitis B virus X protein and interleukin-12 results in antitumor effects via inhibition of hepatoma cell growth and intervention of tumor microenvironment

  • Authors:
    • Hong He
    • Ping Fan
    • Tao Yin
    • Qiaoling Chen
    • Huashan Shi
    • Sijia Liu
    • Haoyu Li
    • Qiuping  Jing
    • Yingying Yan
    • Hailong Zhang
    • Li Yang
    • Yanjun Wen
    • Yuhua Li
    • Ping Cheng
  • View Affiliations

  • Published online on: June 12, 2012     https://doi.org/10.3892/ijmm.2012.1027
  • Pages: 599-605
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Hepatocellular carcinoma (HCC) is a typical hypervascular tumor. Our previous studies have demonstrated that hepatitis B virus X protein (HBx) was able to inhibit the growth of HCC cells via inducing apoptosis and inhibiting tumor angiogenesis. Interleukin-12 (IL-12) is a disulfide-linked heterodimeric cytokine with potent immunostimulatory activity and anti-angiogenic properties. In this study, to further investigate the regulatory effect of IL-12 on HBx-mediated intervention of hepatoma microenvironment especially on intervention of neovessels and immune microenvironment, we constructed the recombinant adenovirus expressing HBx and mouse IL-12 named Ad-HBx-mIL-12. HBx-mIL-12 could effectively suppress tumor growth and induce apoptosis in vivo. Moreover, treatment with Ad-HBx-mIL-12 not only induced a massive accumulation of immune cells (CD8+ T leukocytes, macrophages and dendritic cells) in tumors in situ, also apparently reduced the number of angiogenic blood vessels within tumor tissues. These results suggest that HBx-mIL-12 can not only induce cell cycle arrest and apoptosis in HCC cells, but also effectively shift the tumor microenvironment from pro-oncogenic to antitumor through recruitment of immune cells and inhibiting stromal cell growth, such as vascular endothelial cells.

Introduction

Hepatocellular carcinoma (HCC) is one of the most common and recurrent malignancies worldwide (1). Although the surgical techniques and various nonsurgical treatment modalities have improved, none of these therapies has significantly improved the extremely poor prognosis and the overall 5-year survival rate worldwide is only 2% (2). It is imperative that novel strategies against HCC are identified.

Hepatitis B virus X protein (HBx) is an HBV-encoded protein with multiple functions in transcription pathways, signal transduction, cell cycle progress and HBV replication (3). Studies have demonstrated that HBx could inhibit hepatocarcinoma cell growth via inducing apoptosis. In our previous studies, we showed that treatment with Ad-HBx intratumorally could not only induce apoptosis but also suppress angiogenesis (4).

HCC is a typical hypervascular tumor and tumor angiogenesis is required for both growth and metastasis of HCC (5,6). Most patients show disease recurrence that rapidly progresses to the advanced stages with vascular invasion and multiple intrahepatic metastases (7). Our laboratory demonstrated that implantation of mesenchymal stem cells (MSCs) that are genetically modified with adenovirus vector encoding interleukin-12 (IL-12) could significantly suppress HCC tumor growth in vivo via inhibiting tumor angiogenesis (8).

Not only neovascular endothelial cells that are critical components of the tumor microenvironment, but also the immunocytes in the tumor stroma can affect tumor prognosis through participating innate and adaptive immunity (9). Many studies showed that HBx could elicit cytotoxic T lymphocyte (CTL) antitumor immune responses. Vaccines based on HBx full-length sequence or specific epitopes could trigger significant immune reactions (1013). Our previous studies showed that immunotherapy with Ad-HBx vaccine induced a massive accumulation of CD8+ T cells at the tumor site and accordingly elicit the host CTL response against HBV-associated HCC (11). Intratumoral injections with Ad-HBx induced not only apoptosis of HCC cells but also lymphocytes infiltration in the tumor stroma.

IL-12 as a heterodimeric cytokine, has potent anti-tumor activity and anti-metastatic effect. A large number of animal experiments demonstrated that IL-12 could inhibit tumor angiogenesis and participate in immunoregulation (14,15). IL-12 significantly promoted the NK cell and T cell proliferation, induced the production of IFN-γ and stimulated naive CD4+ T cells to differentiate toward the Th1 phenotype (1618). Recently, combination IL-12 with other treatment options was proved to be a better anti-tumor effect with lower side-effect (1921).

In this study, to further investigate the regulatory effect of IL-12 on HBx mediated intervention of hepatoma microenvironment especially on intervention of neovessels and immune microenvironment, we constructed the recombinant adenovirus carrying HBx and mIL-12 named Ad-HBx-mIL-12.

Materials and methods

Cell lines and adenovirus preparation

Hepa1-6 (mouse HCC) cell line, which was purchased from the American Type Culture Collection, was cultured in DMEM medium (Gibco-BRL) with 10% fetal bovine serum (FBS; Gibco-BRL) and 10 μg/ml gentamicin sulfate at 37°C in 5% CO2. Ad-HBx-mIL-12 is an E1, E3-deleted recombinant adenoviruse (rAd) expressing the HBx and murine IL-12. Ad-HBx, Ad-mIL-12 and Ad-null are E1, E3-deleted rAd expressing the HBx, murine IL-12 and no transgene, respectively. All recombinant adenoviruses were constructed and conserved by our laboratory.

Hepa1-6 cell cycle and apoptosis assay in vitro

Hepa1-6 cells were plated into 6-well plates at 5.0×105 cells/well, overnight. Then, cells were left untreated or infected with Ad-null, Ad-mIL-12, Ad-HBx and Ad-HBx-mIL-12. After 48 h, cell cycle profiles (MOI 10) and apoptosis (MOI 30) were analyzed by flow cytometer (Beckman Coulter) after propidium iodide (PI) staining or Annexin V-phycoerythrin/PI staining, respectively.

In vivo studies

Female syngeneic C57BL/6 mice, 6–8 weeks old, were obtained from Beijing Hua Fu Kang biological technology company and maintained in pathogen-free conditions. All procedures were reviewed and approved by the Institute Animal Care and Use Committee. Mice were challenged subcutaneously with 5×106 Hepa1-6 cells in the right flank. When tumors had reached the desired size (100 mm3), mice were grouped randomly (n=10) and treatment was initiated. Ad-null, Ad-HBx, Ad-mIL-12 and Ad-HBx-mIL-12, diluted in 100 μl sterile 1x PBS (Sigma-Aldrich), were administered 4 times, 5 days apart, intratumorally, at the dose of 108 plaque-forming units (pfu). Tumor dimensions were measured with calipers every 3 days and tumor volumes were calculated according to the formula: V = length × width2 × π/6.

Tumor microenvironment analysis

For FACS analysis, we prepared single-cell suspensions of tumors from untreated or treated mice. Briefly, tumors were minced using a sterile razor blade and digested with collagenase I. For extracellular staining of immune markers, 1×106 of freshly prepared cells were stained with different combinations of fluorochrome-coupled antibodies to CD4, CD8, CD11b, CD11c, F4/80 and Gr1 (BD Biosciences). Fluorescence data were collected on FACScalibur and analyzed using cell quest software (BD Biosciences).

Immunohistochemistry

Three days after the completion of treatment, the mice were sacrificed for histological analysis. The tissues were fixed in 4% paraformaldehyde. Primary tumors were embedded in paraffin and cut into 3–5 μm sections. The apoptotic cells within the tumor sections were evaluated by TUNEL staining, using DeadEnd™ Fluorometric TUNEL System (Promega). Apoptosis index was determined by counting the number of apoptotic cells and dividing by the total number of cells in the field (5 high power fields/slide). Frozen tumor tissues were stained for the quantification of microvessel density (MVD) using a monoclonal rabbit anti-mouse CD31-phcoerythrin conjugate (Santa Cruz Biotechnology, Inc.).

Toxicity evaluation

To investigate potential side effects or toxicity in the treated mice, they were observed continuously for relevant indexes such as weight loss, ruffled fur, diarrhea, anorexia, cachexia, skin ulceration or toxic deaths. After fixing in 4% neutral buffered formalin solution more than 24 h, the tissues of heart, liver, spleen, lung, kidney and brain were embedded in paraffin. Slices of 3–5 mm were stained with hematoxylin and eosin (H&E) and observed in double blinded manner.

Statistical analysis

SPSS 17.0 was used for statistical analysis. Data were expressed as the mean ± SE. The statistical analysis in all the experiments was performed using one-way analysis of variance (ANOVA) or unpaired Student’s t-test. P-values <0.05 were considered significant.

Results

HBx-mIL-12 induces HCC cells to cell cycle arrest and apoptosis in vitro

Hepa1-6 cells were left untreated or infected by Ad-null, Ad-mIL-12, Ad-HBx and Ad-HBx-mIL-12. After 48 h, cell cycle profiles (MOI 10) and apoptosis (MOI 30) were analyzed by flow cytometer. Results showed that Ad-HBx and Ad-HBx-mIL-12 induced a significant accumulation of cells in G2/M phase accompanied by a decrease in the percentage of cells in G0/G1 phase and S phase (Fig. 1A). The percentages of Ad-HBx and Ad-HBx-mIL-12 infected Hepa1-6 cells in G2/M phase were 22.25±1.8% and 26.15±2.1%, respectively, and higher than those of untreated, Ad-null and Ad-mIL-12 infected cells (P<0.01) (Fig. 1B). With increasing MOI, flow cytometric analysis revealed that Ad-HBx and Ad-HBx-mIL-12 infected cells underwent significant apoptosis (Fig. 1C). The percentages of apoptosis in Ad-HBx and Ad-HBx-mIL-12 infected groups were 63.17±5.2% and 61.68±3.1%, respectively, and higher than those of untreated, Ad-null and Ad-mIL-12 infected groups (P<0.01) (Fig. 1D). According to our flow cytometric analysis, the cell cycle arrest and apoptosis in Ad-HBx and Ad-HBx-mIL-12 infected groups have no significant deviation. These results indicate that HBx could induce HCC cells to G2/M arrest and apoptosis, whereas mIL-12 has no function in inducing cell cycle arrest and apoptosis of HCC cells in vitro.

HBx-mIL-12 inhibits the growth of HCC in vivo

A murine hepatic carcinoma model was established to investigate the anti-tumor effect of Ad-HBx-mIL-12 in vivo. When tumors had reached the desired size (100 mm3), Ad-null, Ad-HBx, Ad-mIL-12, Ad-HBx-mIL-12 or PBS was administered intratumorally. Alterations in tumor growth were monitored every 3 days (Fig. 2A). The Ad-HBx-mIL-12 group exhibited effective inhibition of tumor growth, compared to the other groups (P<0.01). On Day 54 after the Hepa1-6 cells were challenged, the mice were sacrificed and the tumor weights were measured (Fig. 2B). We observed that treatment with Ad-HBx-mIL-12 resulted in average tumor weight reductions of 85.6±4.9%, 85.1±2.8%, 75.8±6.1% and 72.8±5.6% compared to PBS, Ad-null, Ad-mIL-12 and Ad-HBx, respectively (P<0.01). These results indicate that Ad-HBx-mIL-12 can effectively suppress tumor growth.

HBx-mIL-12 induces apoptosis of HCC cells in vivo

In order to estimate the apoptosis in tumor tissues, TUNEL staining was applied to the tumor sections. We observed more apoptotic cells in the tumor sections of mice treated with Ad-HBx-mIL-12 than with PBS, Ad-null, Ad-mIL-12 and Ad-HBx (Fig. 3A). The apoptosis index also showed that the Ad-HBx-mIL-12 displayed the highest indices among all the groups (Fig. 3B).

HBx-mIL-12 inhibits vascular endothelial cell growth in vivo

We further investigated whether Ad-HBx-mIL-12 also had inhibitory effects on tumor-stromal cells besides HCC cells within treated tumor tissue. Angiogenic blood vessels within tumor tissue were detected by CD31 immunohistochemistry. The frozen sections were stained for angiogenic blood vessels using a monoclonal rabbit anti-mouse CD-31-phcoerythrin conjugate and counterstained with DAPI (Fig. 4A). Tumors of the control groups (including PBS, Ad-null, Ad-mIL-12 and Ad-HBx groups) showed larger microvessel count than those of Ad-HBx-mIL-12 group (Fig. 4B).

HBx-mIL-12 causes a significant increase in tumor-infiltrating CD8+ T-cell

To further assess the role of immune modulation in the observed antitumor effects mediated by Ad-HBx-mIL-12 treatment, we analyzed the modulation of tumor microenvironment. The ratio of T cells within the tumor microenvironment is considered indicative of the effect of adaptive immune responses on tumor progression and metastasis (19,20). We investigated the subsets of tumor-infiltrating T-cell populations by staining with anti-CD4-PE and CD8-fluorescein isothiocyanate. Although Ad-HBx-mIL-12 treatment did not induce significant change in the ratio of CD4+ T-cells, we observed an increase in the infiltration of total CD8+ T cells in the tumor stroma, as shown by flow cytometric analysis (Fig. 5A). These effects probably result from HBx and mIL-12 mediating host CTL response.

HBx-mIL-12 induces an increase in tumor-infiltrating DCs and macrophages

It has been documented that tumor-infiltrating dendritic cells (DC) and macrophages are potentially able to induce an antitumor innate and adaptive immune response (9). We therefore assessed DCs (CD11b+CD11c+) and tumor-infiltrating macrophage (F4/80+CD11b+) populations by flow cytometry, which showed an increase in DCs and macrophages in tumors after local treatment with Ad-HBx-mIL-12 and Ad-mIL-12 (Fig. 5B and C). However, the increased ratios of DCs and macrophages in Ad-mIL-12 and Ad-HBx-mIL-12 treated tumors have no significant deviation. The numbers of tumor-infiltrating DCs and macrophages in Ad-HBx and Ad-null treated tumors have no significant increase compared with that in PBS group. These results indicate that the increased ratios of tumor-infiltrating DCs and macrophages potentially result from IL-12 mediating the immune responses.

HBx-mIL-12 has no impact on the percentage of MDSCs in tumor microenvironment

Myeloid-derived suppressor cells (MDSCs) in the tumor microenvironment produce growth factors and angiogenic factors critical for tumor progression (20,21). Thus, a large number of MDSCs in tumor microenvironment have probably suppressive actions in the treatment of tumor. We therefore assessed MDSCs (Gr1+CD11b+) populations by flow cytometry, which showed the percentage of MDSCs in Ad-HBx-mIL-12 treated tumors did not have significant changes compared with control groups (including PBS, Ad-null, Ad-mIL-12 and Ad-HBx groups) (Fig. 5D). These result indicate that Ad-HBx-mIL-12 have no effect on decreasing the percentage of MDSCs in tumor microenvironment.

Discussion

In the present study, we demonstrated that HBx-mIL-12 has more anti-tumor effect when compared with HBx or IL-12, as shown in our previous studies (4). In vitro, we found that Hepa1-6 cells infected with Ad-HBx and Ad-HBx-mIL-12 underwent G2/M arrest and apoptosis. However IL-12 did not affect the function of HBx. In vivo, we established a mouse tumor model and performed intratumoral injection of Ad-HBx-mIL-12. We found that although IL-12 did not affect the function of HBx in vitro, combination of IL-12 with HBx induced higher apoptotic effect than HBx or IL-12 in vivo. Combination of IL-12 with HBx led to an almost complete eradication of HCC. The anti-tumor mechanisms of HBx-mIL-12 are due not only to induction of cell cycle arrest and apoptosis in HCC cells, but also effectively shift the tumor microenvironment from pro-oncogenic to anti-tumor.

Although tumors are capable of autonomous growth, their progression is highly influenced by their stromal component (22). T cells, tumor-infiltrating DC, tumor-associated macrophages and myeloid cells are critical components of the tumor microenvironment and the cells may exert either inhibiting or promoting effects on tumor growth (23) through participating innate and adaptive immunity (21,24,25). The ratio of T cells within the tumor microenvironment is considered indicative of the effect of adaptive immune responses on tumor progression and metastasis (19). Abundant CD8+ T-cell infiltration in the tumor stroma is associated with a favorable course of malignancy in cancer (2629). Previous studies have demonstrated that HBx could induce infiltration of CD8+ T lymphocytes in HBx-positive tumors (11). In addition, IL-12 can induce an efficient antitumor CD8+ CTL response which is regarded as the major mediators of the natural host response against developing tumors (30,31). Thus, there is a hypothesis that IL-12 can enhance CD8+ CTL response elicited by HBx. In our study, increased T cell infiltration, especially CD8+ cells, can be detected in tumor tissue of mice treated with Ad-HBx-mIL-12 compared to control groups. This observation is consistent with other studies, indicating that a higher infiltration CD8+ ratio within the tumor microenvironment has been correlated with a favorable response to the treatment, as compared with unchanged ratios in non-responders.

Many studies have definitively shown that recruitment of various antigen-presenting cells, such as dendritic cells (DCs) and macrophages, within tumors can initiate a cascade of innate, adaptive or humoral immune responses against growing tumors (20,32,33). Our study showed an increase in the infiltration of DCs and macrophages in the tumor stroma, after local treatment with Ad-HBx-mIL-12 and Ad-mIL-12. However, the increased ratios of DCs and macrophages in Ad-mIL-12 and Ad-HBx-mIL-12 treated tumors have no significant deviation. These results indicate that the increased ratios of tumor-infiltrating DCs and macrophages potentially result from IL-12 mediating the immunological reaction. Indeed, our findings support the view that IL-12, a key cytokine in cancer immunoprevention, seems to induce infiltration of activated DCs and macrophages to tumor and different organs (34,35).

In our study, treatment with Ad-HBx-mIL-12 reduced the number of angiogenic blood vessels within tumor tissues compared with control group. Studies have shown that MDSCs in the tumor microenvironment produce growth factors and angiogenic factors critical for tumor progression (21). However, the percentage of MDSCs in treated tumors did not decrease compared with control groups. The anti-angiogenic property of IL-12 was well documented in different in vitro and in vivo studies (36,37). In our previous study, treatment with Ad-HBx apparently reduced the number of angiogenic blood vessels within HCC tumor tissue (4). Thus, the mechanism of the anti-angiogenic effect of Ad-HBx-mIL-12 in the present study may result from IL-12 and HBx mediated the anti-angiogenic effects.

In our study, treatment with Ad-HBx-mIL-12 not only induced cell cycle arrest and apoptosis in HCC cells, also induced a massive accumulation of immune cells (CD8+ T leukocytes, macrophages and DC) and significant reduction of vascular endothelial cell growth tumors in situ. HBx-mIL-12 could shift the tumor microenvironment from pro-oncogenic to antitumor not only through recruitment immune cells but also inhibiting stromal cell growth. Collectively, our data in the present study suggest that local delivery of Ad-HBx-mIL-12 could result in antitumor effects via inhibition of hepatoma cell growth and intervention of tumor microenvironment.

Acknowledgements

This study is supported by the National Natural Science Foundation of China (Grant no. 81101728), the National Science and Technology Major Projects of New Drugs (Grant no. 2012ZX09103301-036), the National Science and Technology Major Project for Infectious Diseases Control (Grant no. 2012ZX10002014-004) and the Research Fund for the Doctoral Program of Higher Education of China (Grant no. 20110181120086).

References

1. 

RP BeasleyRocks along the road to the control of HBV and HCCAnn Epidemiol19231234200910.1016/j.annepidem.2009.01.01719344859

2. 

K OkudaHepatocellular carcinomaJ Hepatol32225237200010.1016/S0168-8278(00)80428-6

3. 

H TangN OishiS KanekoS MurakamiMolecular functions and biological roles of hepatitis B virus x proteinCancer Sci97977983200610.1111/j.1349-7006.2006.00299.x16984372

4. 

P ChengY LiL YangHepatitis B virus X protein (HBx) induces G2/M arrest and apoptosis through sustained activation of cyclin B1-CDK1 kinaseOncol Rep22110111072009

5. 

T TianKJ NanSH WangPTEN regulates angiogenesis and VEGF expression through phosphatase-dependent and -independent mechanisms in HepG2 cellsCarcinogenesis3112111219201010.1093/carcin/bgq08520430845

6. 

M FernandezD SemelaJ BruixI ColleM PinzaniJ BoschAngiogenesis in liver diseaseJ Hepatol50604620200910.1016/j.jhep.2008.12.011

7. 

L LiuY CaoC ChenSorafenib blocks the RAF/MEK/ERK pathway, inhibits tumor angiogenesis, and induces tumor cell apoptosis in hepatocellular carcinoma model PLC/PRF/5Cancer Res661185111858200610.1158/0008-5472.CAN-06-137717178882

8. 

X ChenX LinJ ZhaoA tumor-selective biotherapy with prolonged impact on established metastases based on cytokine gene-engineered MSCsMol Ther16749756200810.1038/mt.2008.318362930

9. 

C GuiducciAP VicariS SangalettiG TrinchieriMP ColomboRedirecting in vivo elicited tumor infiltrating macrophages and dendritic cells towards tumor rejectionCancer Res6534373446200515833879

10. 

E ChunJ LeeHS CheongKY LeeTumor eradication by hepatitis B virus X antigen-specific CD8+ T cells in xenografted nude miceJ Immunol17011831190200310.4049/jimmunol.170.3.118312538674

11. 

Y LiP ChengY WenT lymphocyte responses against hepatitis B virus-related hepatocellular carcinoma induced by adenovirus vaccine encoding HBxInt J Mol Med268698762010

12. 

FX DingF WangYM LuMultiepitope peptide-loaded virus-like particles as a vaccine against hepatitis B virus-related hepatocellular carcinomaHepatology4914921502200910.1002/hep.2281619206147

13. 

YJ WangY HouH HuangGR LiuAP WhiteSL LiuTwo oral HBx vaccines delivered by live attenuated Salmonella: both eliciting effective anti-tumor immunityCancer Lett2636776200810.1016/j.canlet.2007.12.02218226855

14. 

A HombachC HeuserH AbkenSimultaneous targeting of IL2 and IL12 to Hodgkin’s lymphoma cells enhances activation of resting NK cells and tumor cell lysisInt J Cancer1152412472005

15. 

C HalinS RondiniF NilssonEnhancement of the antitumor activity of interleukin-12 by targeted delivery to neovasculatureNat Biotechnol20264269200210.1038/nbt0302-26411875427

16. 

LS PengML PenichetSL MorrisonA single-chain IL-12 IgG3 antibody fusion protein retains antibody specificity and IL-12 bioactivity and demonstrates antitumor activityJ Immunol1632502581999

17. 

JA HendrzakMJ BrundaAntitumor and antimetastatic activity of interleukin-12Curr Top Microbiol Immunol21365831996

18. 

G TrinchieriImmunobiology of interleukin-12Immunol Res17269278199810.1007/BF02786451

19. 

Y NasuCH BangmaGW HullCombination gene therapy with adenoviral vector-mediated HSV-tk+ GCV and IL-12 in an orthotopic mouse model for prostate cancerProstate Cancer Prostatic Dis44451200110.1038/sj.pcan.450049412497062

20. 

CM CouglinKE SalhangM WysockaInterleukin-12 and interleukin-18 synergistically induce murine tumor regression which involves inhibition of angiogenesisJ Clin Invest10114411452199810.1172/JCI15559502787

21. 

BM PützerM HittWJ MullerInterleukin 12 and B7-1 co-stimulatory molecule expressed by an adenovirus vector act synergistically to facilitate tumor regressionProc Natl Acad Sci USA94108891089519979380730

22. 

JD BuiR UppaluriCS HsiehRD SchreiberComparative analysis of regulatory and effector T cells in progressively growing versus rejecting tumors of similar originsCancer Res6673017309200610.1158/0008-5472.CAN-06-055616849580

23. 

M KortylewskiP SwiderskiA HerrmannIn vivo delivery of siRNA to immune cells by conjugation to a TLR9 agonist enhances antitumor immune responsesNat Biotechnol27925932200910.1038/nbt.156419749770

24. 

M KujawskiM KortylewskiH LeeA HerrmannH KayH YuStat3 mediates myeloid cell-dependent tumor angiogenesis in miceJ Clin Invest11833673377200810.1172/JCI3521318776941

25. 

LM CoussensZ WerbInflammation and cancerNature420860867200210.1038/nature0132212490959

26. 

A MantovaniS SozzaniM LocatiP AllavenaA SicaMacrophage polarization: tumor-associated macrophages as a paradigm for polarized M2 mononuclear phagocytesTrends Immunol23549555200210.1016/S1471-4906(02)02302-512401408

27. 

PC RodriguezMS ErnstoffC HernandezArginase I-producing myeloid-derived suppressor cells in renal cell carcinoma are a subpopulation of activated granulocytesCancer Res6915531560200910.1158/0008-5472.CAN-08-192119201693

28. 

LA NorianPC RodriguezLA O’MaraTumor-infiltrating regulatory dendritic cells inhibit CD8+ T cell function via L-arginine metabolismCancer Res6930863094200910.1158/0008-5472.CAN-08-282619293186

29. 

SJ PiersmaES JordanovaMI van PoelgeestHigh number of intraepithelial CD8+ tumor-infiltrating lymphocytes is associated with the absence of lymph node metastases in patients with large early-stage cervical cancerCancer Res67354361200717210718

30. 

AG MenonGJ FleurenEA AlphenaarA basal membrane-like structure surrounding tumour nodules may prevent intraepithelial leucocyte infiltration in colorectal cancerCancer Immunol Immunother521211262003

31. 

E SatoSH OlsonJ AhnIntraepithelial CD8+ tumor-infiltrating lymphocytes and a high CD8+/regulatory T cell ratio are associated with favorable prognosis in ovarian cancerProc Natl Acad Sci USA10218538185432005

32. 

L ZhangJR Conejo-GarciaD KatsarosIntratumoral T cells, recurrence, and survival in epithelial ovarian cancerN Engl J Med348203213200310.1056/NEJMoa02017712529460

33. 

O SimmaE ZebedinN NeugebauerIdentification of an indispensable role for tyrosine kinase 2 in CTL-mediated tumor surveillanceCancer Res69203211200910.1158/0008-5472.CAN-08-170519118004

34. 

MN TorreroX XiaW HenkS YuS LiStat1 deficiency in the host enhances interleukin-12-mediated tumor regressionCancer Res6644614467200610.1158/0008-5472.CAN-05-355416618773

35. 

H KanzlerFJ BarratEM HesselRL CoffmanTherapeutic targeting of innate immunity with Toll-like receptor agonists and antagonistsNat Med13552559200710.1038/nm158917479101

36. 

W BarchetV WimmenauerM SchleeG HartmannAccessing the therapeutic potential of immunostimulatory nucleic acidsCurr Opin Immunol20389395200810.1016/j.coi.2008.07.00718652893

37. 

CS TannenbaumN WickerD ArmstrongCytokine and chemokine expression in tumors of mice receiving systemic therapy with IL-12J Immunol15669369919968543822

38. 

S AdrisE ChuluyanA BravoMice vaccination with interleukin 12-transduced colon cancer cells potentiates rejection of syngeneic non-organ-related tumor cellsCancer Res6066966703200011118055

39. 

CL NastalaHD EdingtonTG McKinneyRecombinant IL-12 administration induces tumor regression in association with IFN-gamma productionJ Immunol1531697170619947913943

40. 

AD LusterP LederIP-10, a -C-X-C- chemokine, elicits a potent thymus-dependent antitumor response in vivoJ Exp Med17810571065199310.1084/jem.178.3.10578350046

Related Articles

Journal Cover

September 2012
Volume 30 Issue 3

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
He H, Fan P, Yin T, Chen Q, Shi H, Liu S, Li H, Jing Q, Yan Y, Zhang H, Zhang H, et al: Local delivery of recombinant adenovirus expressing hepatitis B virus X protein and interleukin-12 results in antitumor effects via inhibition of hepatoma cell growth and intervention of tumor microenvironment. Int J Mol Med 30: 599-605, 2012
APA
He, H., Fan, P., Yin, T., Chen, Q., Shi, H., Liu, S. ... Cheng, P. (2012). Local delivery of recombinant adenovirus expressing hepatitis B virus X protein and interleukin-12 results in antitumor effects via inhibition of hepatoma cell growth and intervention of tumor microenvironment. International Journal of Molecular Medicine, 30, 599-605. https://doi.org/10.3892/ijmm.2012.1027
MLA
He, H., Fan, P., Yin, T., Chen, Q., Shi, H., Liu, S., Li, H., Jing, Q., Yan, Y., Zhang, H., Yang, L., Wen, Y., Li, Y., Cheng, P."Local delivery of recombinant adenovirus expressing hepatitis B virus X protein and interleukin-12 results in antitumor effects via inhibition of hepatoma cell growth and intervention of tumor microenvironment". International Journal of Molecular Medicine 30.3 (2012): 599-605.
Chicago
He, H., Fan, P., Yin, T., Chen, Q., Shi, H., Liu, S., Li, H., Jing, Q., Yan, Y., Zhang, H., Yang, L., Wen, Y., Li, Y., Cheng, P."Local delivery of recombinant adenovirus expressing hepatitis B virus X protein and interleukin-12 results in antitumor effects via inhibition of hepatoma cell growth and intervention of tumor microenvironment". International Journal of Molecular Medicine 30, no. 3 (2012): 599-605. https://doi.org/10.3892/ijmm.2012.1027