Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Huamao Wang is active.

Publication


Featured researches published by Huamao Wang.


Journal of Biological Chemistry | 2011

Growth suppression of human hepatocellular carcinoma xenografts by a monoclonal antibody CH12 directed to epidermal growth factor receptor variant III

Hua Jiang; Huamao Wang; Zhonghua Tan; Suwen Hu; Hai Wang; Bizhi Shi; Lin Yang; Peiyong Li; Jianren Gu; Wang H; Zonghai Li

Human hepatocellular carcinoma (HCC) is considered difficult to cure because it is resistant to radio- and chemotherapy and has a high recurrence rate after curative liver resection. Epidermal growth factor receptor variant III (EGFRvIII) has been reported to express in HCC tissues and cell lines. This article describes the efficacy of an anti-EGFRvIII monoclonal antibody (mAb CH12) in the treatment of HCC xenografts with EGFRvIII expression and the underlying mechanism of EGFRvIII as an oncogene in HCC. The results demonstrated that CH12 bound preferentially to EGFRvIII with a dissociation constant (Kd) of 1.346 nm/liter. In addition, CH12 induces strong antibody-dependent cellular cytotoxicity and complement-dependent cytotoxicity in Huh7-EGFRvIII (with exogenous expression of EGFRvIII) and SMMC-7721 (with endogenous expression of EGFRvIII) cells. Notably, CH12 significantly inhibited the growth of Huh7-EGFRvIII and SMMC-7721 xenografts in vivo with a growth inhibition ratio much higher than C225, a U. S. Food and Drug Administration-approved anti-EGFR antibody. Treatment of the two HCC xenografts with CH12 significantly suppressed tumor proliferation and angiogenesis. Mechanistically, in vivo treatment with CH12 reduced the phosphorylation of constitutively active EGFRvIII, Akt, and ERK. Down-regulation of the apoptotic protectors Bcl-xL, Bcl-2, and the cell cycle regulator cyclin D1, as well as up-regulation of the cell-cycle inhibitor p27, were also observed after in vivo CH12 treatment. Collectively, these results indicate that the monoclonal antibody CH12 is a promising therapeutic agent for HCC with EGFRvIII expression.


Cancer Immunology, Immunotherapy | 2010

Identification and characterization of Ch806 mimotopes

Lin Yang; Hua Jiang; Bizhi Shi; Huamao Wang; Jinjun Li; Hai Wang; Ming Yao; Zonghai Li

The chimeric antibody 806 (Ch806) is a promising antitumor agent that recognizes both the epidermal growth factor receptor variant III (EGFRvIII) and the overexpressed epidermal growth factor receptor (EGFR) in cancer tissues but does not recognize the wild type EGFR in normal tissues. However, passive antibody immunization could not produce effective antitumor titers unless the immunization was administered repeatedly over long periods. To overcome this limitation, we generated epitope mimics that bind to Ch806 and tested whether the peptide mimics could induce the production of similar antibodies when actively immunizing mice with the peptides. We used the PH.D-12 phage display peptide library to identify peptides that bind to the monoclonal antibody (mAb) 12H23, which also recognizes similar epitopes of Ch806. Two mimotopes (WHTEILKSYPHE and LPAFFVTNQTQD) were shown to mimic the mAb 12H23 and Ch806 epitope using immunoassays. The mimotopes were conjugated to immunogenic carrier proteins and used to intraperitoneally immunize BALB/c mice. Interestingly, sera from the mice immunized with the isolated mimotopes not only recognize the recombinant or synthetic 806 eptitope, but can also recognize EGFR that is overexpressed in A431 cells and EGFRvIII expressed in Huh7-EGFRvIII cells, whereas sera from mice immunized with the control peptide-KLH (keyhole limpet hemocyanin) and carrier KLH alone failed to show a similar reactivity. Furthermore, in an antibody-dependent cellular cytotoxicity assay (ADCC), the mimotope-induced antibodies specifically lysed human Huh-7-EGFRvIII cells. Our data indicate that the isolated mimotopes reported here may potentially be used as new alternative agents for treating cancer with EGFRvIII expression or EGFR overexpression.


Biomarkers | 2010

Dickkopf-1 autoantibody is a novel serological biomarker for non-small cell lung cancer

Xiao Yao; Hua Jiang; Cuizhen Zhang; Huamao Wang; Lin Yang; Yongfeng Yu; Junming Yu; Bizhi Shi; Zhijun Shen; Huiping Gao; Zhiwei Chen; Shujun Tian; Shun Lu; Zonghai Li; Jianren Gu

Objective: We investigated whether or not there are autoantibodies for DKK1 (Dickkopf-1) in patients with non-small cell lung cancer (NSCLC) and whether this autoantibody can be used for cancer detection. Methods:  The levels of DKK1 autoantibodies were determined in 93 NSCLC patients and 87 healthy controls. Results: We found that, in the sera, the presence of autoantibody against DKK1 was highly correlated with NSCLC. High anti-DKK1 autoantibody titres were found in the sera of NSCLC patients, whereas low or negative titres were found in the control group. The ROC curve results showed that autoantibody immunoassay exhibited 62% sensitivity and 84% specificity. The sensitivity for the detection of NSCLC in stage I also reach 64.3%. Furthermore, a combined ELISA assays for both DKK1 and autoantibody DKK1 increased sensitivity and classified 81.7% (76/93) of the NSCLC patients as positive, whereas only 13.8 % (12/87) of healthy volunteers were falsely diagnosed as positive. Conclusions: Our results suggest that the detection of circulating DKK1 autoantibody could potentially serve as a useful non-invasive marker for determining lung cancer status.


Cancer Immunology, Immunotherapy | 2017

Development of T cells carrying two complementary chimeric antigen receptors against glypican-3 and asialoglycoprotein receptor 1 for the treatment of hepatocellular carcinoma.

Cheng Chen; Kesang Li; Hua Jiang; Fei Song; Huiping Gao; Xiaorong Pan; Bizhi Shi; Yanyu Bi; Huamao Wang; Wang H; Zonghai Li

Adoptive immunotherapy leveraging chimeric antigen receptor-modified T (CAR-T) cells holds great promise for the treatment of cancer. However, tumor-associated antigens often have low expression levels in normal tissues, which can cause on-target, off-tumor toxicity. Recently, we reported that GPC3-targeted CAR-T cells could eradicate hepatocellular carcinoma (HCC) xenografts in mice. However, it remains unknown whether on-target, off-tumor toxicity can occur. Therefore, we proposed that dual-targeted CAR-T cells co-expressing glypican-3 (GPC3) and asialoglycoprotein receptor 1 (ASGR1) (a liver tissue-specific protein)-targeted CARs featuring CD3ζ and 28BB (containing both CD28 and 4-1BB signaling domains), respectively, may have reduced on-target, off-tumor toxicity. Our results demonstrated that dual-targeted CAR-T cells caused no cytotoxicity to ASGR1+GPC3− tumor cells, but they exhibited a similar cytotoxicity against GPC3+ASGR1− and GPC3+ASGR1+ HCC cells in vitro. We found that dual-targeted CAR-T cells showed significantly higher cytokine secretion, proliferation and antiapoptosis ability against tumor cells bearing both antigens than single-targeted CAR-T cells in vitro. Furthermore, the dual-targeted CAR-T cells displayed potent growth suppression activity on GPC3+ASGR1+ HCC tumor xenografts, while no obvious growth suppression was seen with single or double antigen-negative tumor xenografts. Additionally, the dual-targeted T cells exerted superior anticancer activity and persistence against single-targeted T cells in two GPC3+ASGR1+ HCC xenograft models. Together, T cells carrying two complementary CARs against GPC3 and ASGR1 may reduce the risk of on-target, off-tumor toxicity while maintaining relatively potent antitumor activities on GPC3+ASGR1+ HCC.


Cancer Research | 2013

A Novel EGFR Isoform Confers Increased Invasiveness to Cancer Cells

Min Zhou; Hai Wang; Keke Zhou; Xiaoying Luo; Xiaorong Pan; Bizhi Shi; Hua Jiang; Jiqin Zhang; Kesang Li; Huamao Wang; Huiping Gao; Shun Lu; Ming Yao; Ying Mao; Wang H; Shengli Yang; Jianren Gu; Chuan-Yuan Li; Zonghai Li

As a validated therapeutic target in several human cancers, the EGF receptor (EGFR) provides a focus to gain deeper insights into cancer pathophysiology. In this study, we report the identification of a naturally occurring and widely expressed EGFR isoform termed EGFRvA, which substitutes a Ser/Thr-rich peptide for part of the carboxyl-terminal regulatory domain of the receptor. Intriguingly, EGFRvA expression relates more closely to histopathologic grade and poor prognosis in patients with glioma. Ectopic expression of EGFRvA in cancer cells conferred a higher invasive capacity than EGFR in vitro and in vivo. Mechanistically, EGFRvA stimulated expression of STAT3, which upregulated heparin-binding EGF (HB-EGF). Reciprocally, HB-EGF stimulated phosphorylation of EGFRvA at Y845 along with STAT3, generating a positive feedback loop that may reinforce invasive function. The significance of EGFRvA expression was reinforced by findings that it is attenuated by miR-542-5p, a microRNA that is a known tumor suppressor. Taken together, our findings define this newfound EGFR isoform as a key theranostic molecule.


Molecular Cancer | 2017

EGFR modulates monounsaturated fatty acid synthesis through phosphorylation of SCD1 in lung cancer

Jiqin Zhang; Fei Song; Xiaojing Zhao; Hua Jiang; Xiuqi Wu; Biao Wang; Min Zhou; Mi Tian; Bizhi Shi; Huamao Wang; Yuanhui Jia; Hai Wang; Xiaorong Pan; Zonghai Li

BackgroundEpidermal growth factor receptor (EGFR), a well-known oncogenic driver, contributes to the initiation and progression of a wide range of cancer types. Aberrant lipid metabolism including highly produced monounsaturated fatty acids (MUFA) is recognized as a hallmark of cancer. However, how EGFR regulates MUFA synthesis in cancer remains elusive. This is the focus of our study.MethodsThe interaction between EGFR and stearoyl-CoA desaturase-1 (SCD1) was detected byco-immunoprecipitation. SCD1 protein expression, stability and phosphorylation were tested by western blot. The synthesis of MUFA was determined by liquid chromatography-mass spectrometry. The growth of lung cancer was detected by CCK-8 assay, Annexin V/PI staining, colony formation assay and subcutaneous xenograft assay. The expression of activated EGFR, phosphorylated and total SCD1 was tested by immunohistochemistry in 90 non-small cell lung cancersamples. The clinical correlations were analyzed by Chi-square test, Kaplan-Meier survival curve analysis and Cox regression.ResultsEGFR binds to and phosphorylates SCD1 at Y55. Phosphorylation of Y55 is required for maintaining SCD1 protein stability and thus increases MUFA level to facilitate lung cancer growth. Moreover, EGFR-stimulated cancer growth depends on SCD1 activity. Evaluation of non-small cell lung cancersamples reveals a positive correlation among EGFR activation, SCD1 Y55 phosphorylation and SCD1 protein expression. Furthermore, phospho-SCD1 Y55 can serve as an independent prognostic factor for poor patient survival.ConclusionsOurstudy demonstrates that EGFR stabilizes SCD1 through Y55 phosphorylation, thereby up-regulating MUFA synthesis to promote lung cancer growth. Thus, we provide the first evidence that SCD1 can be subtly controlled by tyrosine phosphorylation and uncover a previously unknown direct linkage between oncogenic receptor tyrosine kinase and lipid metabolism in lung cancer. We also propose SCD1 Y55 phosphorylation as a potential diagnostic marker for lung cancer.


Cancer Letters | 2014

The monoclonal antibody CH12 augments 5-fluorouracil-induced growth suppression of hepatocellular carcinoma xenografts expressing epidermal growth factor receptor variant III.

Hua Jiang; Qiongna Dong; Xiaoying Luo; Bizhi Shi; Huamao Wang; Huiping Gao; Juan Kong; Jiqin Zhang; Zonghai Li

5-Fluorouracil (5-FU) is one of the most common chemotherapeutic agents used for the treatment of hepatocellular carcinoma (HCC). However, chemoresistance has precluded the use of 5-FU alone in clinical regimens. Combination therapies with 5-FU and other anticancer agents are considered to be a therapeutic option for patients with HCC. We previously reported that the expression of epidermal growth factor receptor variant III (EGFRvIII) can decrease the sensitivity of HCC cells to 5-FU. To overcome this problem, in this study, we elucidated the mechanism underlying EGFRvIII-mediated 5-FU resistance. We observed that EGFRvIII expression can induce miR-520d-3p downregulation and the ensuing upregulation of the transcription factor E2F-1 and the enzyme thymidylate synthase (TS), which may lead to drug resistance. Intriguingly, we found that CH12, a monoclonal antibody directed against EGFRvIII, and 5-FU together had an additive antitumor effect on EGFRvIII-positive HCC xenografts and significantly improved survival in all mice with established tumors when compared with either 5-FU or CH12 alone. Mechanistically, compared with 5-FU alone, the combination more noticeably downregulated EGFR phosphorylation and Akt phosphorylation as well as the expression of the apoptotic protector Bcl-xL and the cell cycle regulator cyclin D1. Additionally, the combination upregulated the expression of the cell cycle inhibitor p27 in in vivo treatment. More interestingly, CH12 treatment upregulated miR-520-3p and downregulated E2F-1 and TS at the mRNA and protein levels. Collectively, these observations suggest that the combination of 5-FU with mAb CH12 is a potential means of circumventing EGFRvIII-mediated 5-FU resistance in HCC.


BioTechniques | 2007

Protein III-based single-chain antibody phage display using bacterial cells bearing an additional genome of a gene-III-lacking helper phage.

Bizhi Shi; Huamao Wang; Shengrong Guo; Yuhong Xu; Zonghai Li; Jianren Gu

We present herein a novel method of pIII-based antibody phage display using Hpd3cells--bacterial cells bearing the genome of a gene-III-lacking helper phage (VCSM13d3). A high level of single-chain variable fragments (scFvs) was displayed in the consequent phagemid particles using Hpd3cells to rescue the phagemid encoding scFv-pIII. Hpd3cells considerably improved the specific enrichment factor when used for constructing an immunized antibody library. In addition, using Hpd3cells could overcome pill resistance and can contribute to the efficient enrichment of specific binding antibodies from a phage display library, thereby increasing the chance of obtaining more diverse antibodies specific for target antigens.


Cancer immunology research | 2018

Selective Targeting of Glioblastoma with EGFRvIII/EGFR Bitargeted Chimeric Antigen Receptor T Cell

Hua Jiang; Huiping Gao; Juan Kong; Bo Song; Peng Wang; Bizhi Shi; Huamao Wang; Zonghai Li

CAR T cells targeting EGFR- and EGFRvIII-overexpressing tumor cells exhibit antitumor activity without toxicity toward normal EGFR-expressing cells in mouse glioblastoma models. This strategy may provide an avenue for future therapeutic development in EGFR- and EGFRvIII-overexpressing cancers. The heterogeneous expression of EGFRvIII [variant III mutant of epidermal growth factor receptor (EGFR)] in glioblastoma has significant impact on the clinical response to the treatment of EGFRvIII-specific chimeric antigen receptor–engineered T (CAR T) cells. We hypothesized that CAR T cells that could target both EGFRvIII and the form of EGFR expressed on tumor cells, but not EGFR on normal cells, would greatly improve efficacy without inducing on-target, off-tumor toxicity. Therefore, we developed a humanized single-chain antibody, M27, with a single specificity that binds to an epitope found both on wild-type EGFR- and EGFRvIII-overexpressing tumor cells, but not EGFR-expressing normal cells, including primary keratinocytes, on which wild-type EGFR is highly expressed. M27-derived CAR T cells effectively lysed EGFRvIII- or EGFR-overexpressing tumor cells, but showed no observable toxicity on normal cells. Inclusion of the CD137 (4-1BB) costimulatory intracellular domain in the M27-28BBZ CAR provided CAR T cells with higher tumor lysis activity than when not included (as in the M27-28Z CAR). The growth of established EGFR- or EGFRvIII-overexpressing glioma xenografts was suppressed by M27-28BBZ CAR T cells as well. The growth of heterogeneic xenograft tumors, created by mixing EGFR- and EGFR-overexpressing glioblastoma cells, was also effectively inhibited by M27-28BBZ CAR T cells. The survival of mice in the orthotopic models was significantly prolonged after M27-28BBZ CAR T-cell infusion. These results suggested that tumor-selective, bitargeted anti-EGFR/EGFRvIII CAR T cells may be a promising modality for the treatment of patients with EGFR/EGFRvIII-overexpressing glioblastoma. Cancer Immunol Res; 6(11); 1314–26. ©2018 AACR.


Cancer Research | 2011

Abstract 4587: Identification of a monoclonal antibody to treat HCC xenografts with EGFRvIII expression

Zonghai Li; Hua Jiang; Huamao Wang; Bizhi Shi

Purposes:To identify monoclonal antibodies targeting Epidermal Growth Factor Receptor Variant III (EGFRvIII) and their anti-tumor effect on Hepatocellular carcinoma (HCC) with EGFRvIII expression. Procedures: Hybridoma technology was used to prepare monoclonal antibody recognizing EGFRvIII. Anti-tumor effects of several hybridoma clones were examined on HCC xenografts with EGFRvIII expression. Monoclonal antibody with highest growth-suppression effect on HCC was selected. Binding specificity of the antibody was tested by FACS assay. The targeting ability of this antibody was tested by injected the cyanin 5.5 labeled antibody into mice carrying EGFRvIII positive and negative xenografts. Findings: Among the tested monoclonal antibodies, mAb 12H23 has the highest tumor-suppression effect on HCC with EGFRvIII expression. mAb 12H23 bind preferentially to HCC cells with EGFRvIII expression. Cy5.5 labeled mAb 12H23 can significantly accumulate into EGFRvIII positive tumors but not the negative tumors. Conclusion: These data indicate mAb 12H23 is a potential biotherapeutic agent for treating HCC with EGFRvIII expression. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 4587. doi:10.1158/1538-7445.AM2011-4587

Collaboration


Dive into the Huamao Wang's collaboration.

Top Co-Authors

Avatar

Zonghai Li

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Hua Jiang

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Bizhi Shi

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Jianren Gu

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Huiping Gao

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Hai Wang

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Juan Kong

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Ming Yao

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Shengli Yang

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Wang H

Second Military Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge