Proceedings of the 2nd International Conference for Cancer Metabolism and Therapy, October 13–15, 2017, Wenzhou, China
Gary Guishan Xiao,Mengtao Zhou,Bin Lv,Bichen Chen,Hongxia Wang,Xiangdong Wang,Jianhua Wang,Ge Gao,Wai-Nang Paul Lee,Laszlo G. Boros,Ping Gao,Jianyuan Luo,Yue Pan,Xingguo Liu,Vay Liang W. Go
DOI: https://doi.org/10.1097/mpa.0000000000001046
2018-01-01
Pancreas
Abstract:The 2nd International Conference for Cancer Metabolism and Therapy was successfully held in Wenzhou Awailou Resort from October 13 to 15, 2017 with the theme of "Cancer Metabolism and Therapy". The conference was organized by The First Affiliated Hospital of Wenzhou Medical with sponsorship from the Chemical Engineering and Environmental-Biological schools, Dalian University of Technology, the Hirshberg Foundation for Pancreatic Cancer (http://pancreatic.org/), UCLA Agi Hirshberg Center for Pancreas Diseases and participating industries, Bruker Daltonics, Agilent, Bio-Rad Laboratories, and Beijing Ancomol Biotechnologies Co Ltd. Cancer metabolism is intimately linked to drug resistance, which is currently one of the most important challenges in cancer therapy. Cell phenotype may be altered by proteome phenotype that results from altered genomic phenotype that may be regulated by metabolic phenotype initiated by early signals. Due to newly developed technology, we can readily use many high-tech diagnostic tools, new anticancer drugs, molecular targeting chemotherapy, less invasive and/or less expensive and effective therapy that were unavailable in the near past. Recent advancement of novel high-throughput technologies, such as transcriptomics, proteomics and metabolomics, has significantly enhanced our understanding of metabolic properties related to malignancy, paving the way for selection of molecular targets for therapeutic interventions. Cancer therapy and research are in a new era of rapid and significant developments not only in the West but also in Asia as well. As the burden of cancer is increasing rampantly and posing challenges in early diagnosis and treatment, this international cancer metabolism and therapy conference is planned to be held every other year with the aim of identifying the genuine needs in the area and implementation for patient welfare. This conference covers a wide range of topics in cancer such as etiology, epidemiology, metabolic reprogramming, environment altered cellular metabolism, cancer prevention and vaccines, new drug development, multidisciplinary treatment i.e. surgical therapy, chemotherapy, and radiotherapy and cancer stem cell therapy. Scientists from all over the world shared their new ideas and thoughts with each other, especially young scientists who are able to communicate openly with the global leaders in the field, investigators, medical and surgical oncologists through presentations and vigorous discussion. The next meeting is planned to be held October 12–October 14, 2018 in Shanghai, China. The meeting venue will be in the Shanghai Baohua Marriott, Shanghai, China. ABSTRACTS PRESENTED Metabolic Regulation of Tumor Microenvironment Heterogeneity G. G. Xiao. School of Pharmaceutical Science, Dalian University of Technology, Dalian, China; Creighton University Medical Center, Omaha, NE. Background: Tumor Microenvironment is the cellular environment in which the tumor exists, including surrounding blood vessels, immune cells, fibroblasts, bone marrow-derived inflammatory cells, lymphocytes, signaling molecules and the extracellular matrix. The tumor microenvironment contributes to tumor heterogeneity. Exact mechanism of causing tumor heterogeneity is not clearly understood yet. Results and Discussion: Data from our previous research and others suggest that tumor heterogeneity may be resulted from unsynchronized differentiation of cancer progenitor cells. We then hypothesized that cell phenotype may be altered by proteome phenotype that is resulted from altered genomic phenotype that may be regulated by metabolic phenotype initiated by early signals. In another words, heterogeneity of tumor microenvironment may be resulted from dysregulated metabolic phenotypes. To test our hypothesis, we developed analytical methods to measure quantitatively the signals and its initiated metabolic phenotypes in a cell. The methods developed in our group at both UCLA and Creighton University include stable isotopomer-based flux analysis and dynamic measurement of protein turnover in a cell, which extensively used in cancer research. In this oral communication, the details on how these methods can be used will be delivered and an example based on the methods will be also illustrated. Conclusion: Understanding the mechanism underlying tumor microenvironment heterogeneity regulated metabolically is key for development of anticancer drugs with minimum toxicity and maximal effectiveness. Ubiquitylation of Autophagy Receptors: Multiple Modes of Regulation on Cellular Autophagic Flux R. Hu. State Key Lab of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences (CAS), Shanghai, China. Background: Ubiquitin (Ub) signaling regulates myriad of fundamental cellular activities. The alterations in cellular Ub homeostasis, known as Ub stress, feature and affect cellular responses in multiple conditions, yet the underlying mechanisms are incompletely understood. Method: By biochemical and cell biology method we found that p62/SQSTM1 was polyubiquitylated in Ub+ stress conditions, and we identified its E2 enzyme Ube2d2 and Ube2d3 in yeast two hybrid screening, then we mapped the E2-dependant ubiquitylation sites by mass spectrum analysis. We further confirmed that polyubiquitylated p62 form large complex than unmodified p62 by electro-microscopy and dynamic light scattering analysis. Results: Ub+ stress in cells induced by over-expression of Ub, prolonged proteasomal inhibition and heat shock could efficiently induce autophagy in dependence of autophagy receptor, p62/SQSTM1, p62 was increasingly ubiquitylated during those conditions. Subsequently, p62 was found to specifically interact with two Ub conjugating enzymes, Ube2d2 or Ube2d3, using a common E2-interacting region (EIR), and these E2s could support p62 ubiquitylation both in vitro and in the cell. Multiple Lys (K) residues in p62 were mapped as the sites for this E2-supported ubiquitylation, which include K420 in the UBA domain of p62. As previously reported, UBA domains in p62 formed stable dimers, which would prevent p62 from binding to polyUb chain in autophagy cargos; our in vitro polyUb-binding data showed that polyubiquitylated p62 in the presence of the E2s bind to polyUb chains much more efficiently than unmodified p62. Consistently, electro-microscopy and dynamic light scattering analyses indicated that polyubiquitylated p62 might predominantly form large complex with polyUb chains (~ two fold lager in diameter) than unmodified p62, suggesting that p62 might have indeed adopted an open conformation upon E2-supported polyubiquitylation. Conclusion: By discovering E2-supported ubiquitylation of p62 as a novel mechanism for activating its autophagy receptor function under Ub+ stress conditions, our work has thus revealed a unique “sensor” function of p62 in modulating autophagy as part of the cellular responses to prolonged proteasomal inhibition, heat shock or Ub overexpression. Identifying Clinically Actionable Alterations in the Era of P4plus Medicine: Challenges and Opportunities B. Shen. Center for Systems Biology, Soochow University, Suzhou, China. Background: The Chinese government recently launched two projects, i.e. “Healthy China 2030” and "Genetic testing for Chinese public health: science and social engineering". The programs center on chronic disease prevention and promoting personalized and positive lifestyle choices. Scientifically the identification of actionable alterations in gene, lifestyle or environmental factors will be the key for the diagnosis and prognosis of diseases and the improving of population healthcare. Method: I will present our researches on the identification of actionable biomarkers for diagnosis of complex diseases and talk about our strategies for the biomarker discovery from big data to small data, based on the statistical analysis of the reported biomarkers, the mechanistic and functional analysis of biological networks. Results: We first studied the network properties of gene society, and then applied the ideas to the identification of microRNA biomarkers for cancers, such as prostate cancer and leukemia. We cooperated with medical doctors to verify our findings by collect disease samples and experimental confirmation. Conclusion: The future challenges and opportunities for the P4 medicine, i.e. predictive, preventive, personalized and participatory medicine include the data privacy, lifestyle, universal and experimental data characterization as well as the standardization of diverse unstructured data. The Role of Hypermethylated in Cancer 1 (HIC1) in Tumorigenesis and Development M. Hao,1,2 Y. Wang,1 X. Weng,3 J. Wang.31Department of Biochemistry and Molecular & Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China; 2Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH; 3Fudan University Shanghai Cancer Center, Shanghai, China. Background: Hypermethylated in cancer 1 (HIC1) is a tumor suppressor gene and a transcriptional inhibitory factor, whose promoter is usually silenced in many solid tumor tissues and precancerous lesions (such as smokers’ lung, colorectal polyps and other tissues). HIC1+/- mice studies also suggest that the gene may be associated with tumorigenesis, tumor development and metastasis. Method: A variety of solid tumor tissues (including prostate cancer, breast cancer, lung cancer, etc.) were collected for analyzing HIC1 expression and methylation status. In vitro and in vivo (HIC1 cKO and transplant mice model) experiments were utilized to study the phenotypes as well as mechanisms upon HIC1 loss or reexpression. Results: In recent years, our team have conducted a series of studies on HIC1 and found that hyper methylation of HIC1 promoters in a variety of solid tumor tissues (including prostate cancer, breast cancer, lung cancer, etc.) may lead to HIC1 silence, resulting in suppressive function loss or attenuation in these tumors. In vitro and in vivo experiments show that the recovery of HIC1 expression could significantly reduce the tumor proliferation, invasion, and metastasis. Mechanism studies disclose that HIC1 expression is widely modulated including ubiquitination, SUMOylation, as well as methylation. Conclusion: These studies suggest that HIC1 may act as a potential tumor biomarker, which provides new ideas for the early diagnosis and treatment of tumors. RP-MDM2-p53 Pathway Regulates Lipid Metabolism and Acts as the Checkpoint of Myc-induced Lymphomagenesis Y. Liu, E.Macias, Y. Zhang. Cancer Institute, Xuzhou Medical University, Xuzhou, China. Background: The tumor suppressor p53 has recently been shown to regulate lipid metabolism through multiple mechanisms. However, the in vivo signaling pathways related to p53-mediated metabolic regulation remain largely uncharacterized. Results: By using mice bearing a single amino acid substitution at cysteine residue 305 of mouse double minute 2 (Mdm2C305F), which renders Mdm2 deficient in binding ribosomal proteins (RPs) RPL11 and RPL5, we show that the RP–Mdm2–p53 signaling pathway is critical for sensing nutrient deprivation and maintaining liver lipid homeostasis. Nutrient deprivation inhibits rRNA biosynthesis and triggers ribosomal stress, which increases RP–Mdm2 interaction, and induces p53-mediated transactivation of malonyl-CoA decarboxylase (MCD), leading to increased fatty acid oxidation. By contrast, Mdm2 mutant mice demonstrate attenuated MCD induction and increased lipid accumulation in the liver. Furthermore, to test whether RP-Mdm2-p53 pathway involved in the tumor suppressive function of p53, we crossed Mdm2C305F mouse with Eμ-myc mouse, and found that interruption of RP-MDM2 interaction strikingly accelerates oncogenic Myc-induced lymphomagenesis. Conclusion: Thus, the RP–Mdm2–p53 pathway appears to function as an endogenous stress sensor responsible for breaking down lipid storage for survival. However, in response to oncogene over-activation, this pathway acts as the checkpoint of excessive ribosome biogenesis. miR-1207-5p Induced Metastasis by Regulating Tryptase-alpha-1 in Oral Cancer M. Cao, Q. Yang, X.Miao, J. Liu, Y. Pan, G.G. Xiao. School of Pharmaceutical Science and Technology, Dalian University of Technology, Dalian, China. Background: Oral cancer metastasis is a life threatening health problem worldwide. Development of an effective strategy for treatment of oral cancer is a key for reducing the death rate of the disease. In recent years, microRNAs (miRNAs) as a group of small non-coded RNAs play a vital role in tumor metastasis. The goal of this study is to determine the regulatory role of miR-1207-5p and the mechanism underlying in oral cancer metastasis. Methods: In this study, we systematically evaluated a large scale of miRNA profiles from current 24 oral pathological tissues, which including 8 benign samples, 7 local carcinoma samples and 9 distal metastasis carcinoma samples. The miRNAs expressed differentially in tumor cells were identified by using DChip technology, and peptides expressed remarkably in tumor core were identified by using LTQ/MS. The quantitative real-time PCR was applied to validate the level of miR-1207-5p and the target gene. Western blot analysis was used to verify the target protein expression of miR-1207-5p. Results: The miR-1207-5p was significantly up-regulated in oral local carcinoma and distal metastasis carcinoma tissues, while tryptase-alpha-1 (TPSAB1) was dramatically up-regulated in oral local carcinoma tissues. By prediction analysis and reciprocally expression of both miR-1207-5p and its target TPSAB1 in oral tumor tissue, TPSAB1 was further confirmed as a target of miR-1207-5p. The regulatory role of miR-1207-5p in oral metastasis is still under investigation. Conclusion: The data above indicate that miR-1207-5p may be potential biomarker or target of oral cancer metastasis and induce oral cancer metastasis by targeted Tryptase alpha-1. The Expression of MALAT1 in Endometriosis G. Gao, H. Wang, Y. Cao, C. Liu, R. Hu. Xiangya Medical College, Chinese Southern University, Changsha, China. Objective: The article aimed to explore the expression and the meaning of metastasis-associated lung adenocarcinoma transcript1 (MALAT1) in endometriosis (EMs) and the serum. Methods: The differentiate gene was found between endometriosis tissues and normal tissues through GCBI, and the MALAT1 was chosen among them. Extract the RNA from ovarian endometriosis and non-endometriosis as well as the serum, real-time PCR was applied to detect the relationship of MALAT1 expression with menstruing cycle; the diagnostic efficacy of serum MALAT1 was analyzed by the receiver operating characteristic curve (ROC). Results: The MALAT1 in EMs was down-regulated about 1.35 times; and the relative expression level of MALAT1 in ectopic and eutopic endometrium with ovary endometriosis were significantly lower than those in non-endometriosis patients. The MALAT1 in ectopic endometrium was lower than the eutopic endometrium with ovary endometriosis; The relative expression level of serum MALAT1 in ovary endometriosis was significantly lower than that in non-endometriosis. The diagnostic sensitivity and specificity of endometriosis were satisfactory. Conclusion:MALAT1 played a critical role in the process of endometriosis, and the level of MALAT1 in serum contributed to the diagnosis of endometriosis. Tid1-S Regulates Metabolic Reprogramming in Clear Cell Renal Cell Carcinoma via SIRT3 L. Lan, F. Shangguan, J. Lu, Y. Liu, B. Lu. Institute of Biophysics, Attardi Institute of Mitochondrial Biomedicine and Zhejiang Provincial, Key Laboratory of Medical Genetics, School of Life Sciences, Wenzhou Medical University, Wenzhou, China. Background: Human Tid1 (hTid1) is a DnaJ chaperone protein and human homolog of Drosophila tumor suppressor Tid56. It has been reported that overexpression of hTid1-S, the short alternatively spliced isoform of hTid1, suppresses apoptosis induced by apoptotic stimuli. Altered expression of hTid1 is observed in various types of human cancers. However, the clinical significance of hTid1-S expression in clear cell renal cell carcinoma (ccRCC) remains unknown. Methods: Western blot and qRT-PCR were used to detect protein and mRNA alteration of indicated molecules. Seahorse XF96 was performed to determine aerobic glycolysis and oxygen consumption rate. IHC was used to analyze Tid1-S protein level in ccRCC tumor tissues and adjacent non-cancerous tissues. Kaplan-Meier survival analysis was applied to analyze survival rate post-operation. Results: In the present study, we found the mRNA and protein levels of hTid1-S were significantly lower in tumor tissues than adjacent non-cancerous tissues of ccRCC patients. Upregulation of hTid1-S in renal cancer cells significantly inhibits cell proliferation by increasing reactive oxygen species (ROS) production. In addition, we found that the interaction of Tid1-S with SIRT3 lead to the inhibition of mTOR/MAPK signaling and activation of AMPK-a signaling, they are both crucial processes in cell metabolism. Moreover, overexpressed Tid1-S represses aerobic glycolysis while mitochondrial respiration was enhanced, which indicates a switch of Warburg effect. Immunohistochemistry analysis showed that hTid1-S protein expression was decreased in tumor tissues. We further showed that hTid1-S expression level was much lower in later clinical stage (Stage III) than in early clinical stage (I and II) tumor tissues. Moreover, the expression level of hTid1-S was significantly correlated with patient’s T stage (P = 0.008) and TNM stage (P = 0.008). We also demonstrated that hTid1-S was an independent prognostic factor for overall survival of ccRCC. Conclusion: Our findings suggest that hTid1-S could potentially be useful as a diagnostic and prognostic biomarker, as well as a novel therapeutic target candidate for ccRCC. Deacetylation of Tumor Suppressor MST1 in Hippo Pathway Induces its Degradation Through HBXIP-elevated HDAC6 in Promotion of Breast Cancer Growth L. Li,1 R. Fang,1 B. Liu,1 H. Shi,1 Y. Wang,1 W. Zhang,1 X. Zhang,2 L. Ye.11State Key Laboratory ofMedicinal Chemical Biology, Department of Biochemistry, College of Life Sciences, Nankai University, Tianjin, China; 2State Key Laboratory of Medicinal Chemical Biology, Department of Cancer Research, College of Life Sciences, Nankai University, Tianjin, China. Background: Reduction or loss of tumor suppressor mammalian STE20-like kinase 1 (MST1) in Hippo pathway contributes to the tumorigenesis. However, the mechanism leading to reduction of MST1 in cancers remains poorly understood. Methods: We used western blot, confocal, immunohistochemistry, and xenograft mice, et al. to investigate the mechanism of MST1 expression reduction in breast cancer. Result: In this study, we explored the hypothesis that the oncoprotein hepatitis B X-interacting protein (HBXIP) is involved in the reduction of MST1 in breast cancer. Immunohistochemical analysis of tissue microarrays revealed that the expression of HBXIP was negatively associated with that of MST1 in 98 clinical breast tissue samples. Then, we found that HBXIP could post-translationally down-regulate MST1 in breast cancer cells. Mechanistically, we identified that MST1 could be acetylated in the cells. Strikingly, treatment with trichostatin A (TSA), an inhibitor of histone deacetylases (HDACs), markedly increased MST1 acetylation and protein level in the cells. Interestingly, oncoprotein HBXIP could significantly inhibit acetylation of MST1, resulting in the reduction of MST1 protein. Notably, we revealed that histone deacetylase 6 (HDAC6) could reduce protein level of MST1 though deacetylation of MST1 in the cells. Then, our data revealed that HBXIP up-regulated HDAC6 at the levels of mRNA and protein by activating transcription factor NF-κB. Furthermore, we demonstrated that MST1 deacetylation promoted the interaction of MST1 with HSC70 in the cells, resulting in a lysosome-dependent degradation of MST1 via chaperone-mediated autophagy (CMA). Functionally, the reduction of tumor suppressor MST1 mediated by HBXIP promoted the growth of breast cancer cells in vitro and in vivo. Conclusion: We conclude that the deacetylation of MST1 mediated by HBXIP-enhanced HDAC6 results in MST1 degradation in a CMA manner in promotion of breast cancer growth. Our finding provides new insights into the mechanism of MST1 reduction in Hippo pathway in breast cancer. Correlation Between miR-21 and Pancreatic Cancer C. Liu, H. Wang, T. Hu, Y. Cao, X. Chen, G. Gao. Xiangya Medical College, Central Southern University, Changsha, China. Objective: The article aimed to analysis the differentiate gene expression of pancreatic cancer to gain a core gene, namely, miR-21, clarifying the correlation between miR-21 and pancreatic cancer as well as regulating TGFβ. Methods: The miR-21 was screened by literature analysis and pancreatic cancer gene analysis. miR-21 was clinically verified by RT-PCR. RT-PCR and Western blot experiments were carried out to study the relationship between miR-21 and TGFβ by overexpression and suppression of miR-21 in pancreatic cancer cells. Results: miR-21 was highly expression in pancreatic cancer. The overexpression of miR-21 in PANC-1 cannot lead to the variation of TGFβ; the inhibition of miR-21 can cause the highly expression of TGFβ. Conclusion: miR-21 was overexpression in pancreatic cancer cells. The expression of miR-21 can regulate the expression of TGFβ, which may be a mechanism of miR-21 in pancreatic cancer. OLA1 Expression Varied in Pancreatic Cancer of Gemcitabine Resistance J. Liu, J. Huang, X. Miao,M. Cao, Q. Yang, P. Yue, G.G. Xiao. School of Pharmaceutical Science and Technology, Dalian University of Technology, Dalian, China. Background: Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive malignant disease with a 5-year survival rate less than 5%. Gemcitabine alone or in combination with other therapeutics has been the main therapy for advanced pancreatic cancer, however the acquired drug resistance becomes a limiting factor in its clinical application. Since the resistance of pancreatic cancer negatively affects the therapeutic effects of gemcitabine, finding novel target that increase tumor sensitivity and overcome drug resistance to gemcitabine are needed. OLA1 (Obg like ATPase 1) is relative with resistance of gemcitabine in pancreatic tumor. The goal of this study is to determine the efficacy of OLA1 on overcoming drug resistance and the possible mechanism. Methods: The sensitive of gemcitabine on viability of BxPC-3 and PANC-1 cell was measured with methyl thiazolyl tetrazolium (MTT) assay and microscopy examination. Flow cytometric assay (FCA) was used to detect apoptosis of these two cell lines with different dose and time. mRNA level of OLA1 was detected by Q-RT-PCR. The protein expression of OLA1 was determined by Western blot analysis. Tissue chip was carried to reveal the relationship between pancreatitis and acquired resistance of different tumors. Results: Gemcitabine inhibited the proliferation of pancreatic carcinoma and induced apoptosis in a dose-dependent manner. BxPC-3 was more sensitive to gemcitabine than PANC-1 validated by morphology and MTT assay. The mRNA and protein level of OLA1 in gemcitabine resistance cells (PANC-1) is high-expressed. It's interesting that the pancreatitis tissue expressed more OLA1 compared with tumor by tissue chip. Conclusion: Results of this project may lead to the establishment of a new anti-cancer strategy based on OLA1, an important post-translational regulator of many celluar process including proliferation, metastasis and invasion. Further studies are needed to clarify which signal pathway are relevant to OLA1 in regulating pancreatic cancer of gemcitabine resistance. Involvement of the Fragile X Protein in Translation of STAT3 Promotes Hepatocellular Carcinoma Metastasis S. Lu, Y. Han, G. Zhang, Z. Shen. School of Life Sciences, Wenzhou Medical University, Wenzhou, China. Background: The fragile X mental retardation protein (FMRP) is an RNA-binding protein that plays important roles in mRNA stability, mRNA export, location and translation. The most study of FMRP is focused on nerve diseases such as FXS caused by FMRP dysfunction, while how does it affect cancer progression is still unknown. Method: We used RNA IP assay and FISH combined with IF to verify the relationship between FMRP and STAT3. We also stably knocked down the FMRP protein using siRNA system, then, RNA extraction, RT‐qPCR array and western blotting were conduct to test the expression of Stat3. Using Co-immunoprecipitation, transwell, and immunohistochemical analysis to find the role of FMRP in HCCLM3 cells and human liver cancer tissue. Results: FMRP protein colocalized with STAT3 (Signal Transducer and Activator of Transcription 3) mRNA in HCCLM3 cells using FISH and IF assay. Meanwhile, FMRP associated with STAT3 mRNA by RNA Immunoprecipitation assay. Interestingly, the protein level of STAT3 is increased while the expression of STAT3 mRNA is constant in the FMRP KD cells. Importantly, our results have shown that FMRP interacts with many translational complexes, such as TDP-43, eIF4E, and FMRP interact with protein kinase PI3K. Finally, we find that FMRP is overexpressed in liver cancer tissue compared to para-carcinoma tissues, and the invasion ability of HCCLM3 cell is decreased in FMRP KD cell. Conclusion: In this study, we identified STAT3 mRNA as a new FMRP binding target which involved STAT3 mRNA translation. FMRP knock down decrease the HCCLM3 metastasis, and the FMRP protein level is elevated in Hepatocellular Carcinoma tissue, suggesting that FMRP may affect the Hepatocellular Carcinoma Metastasis though regulating expression of STAT3. Exosomes: A Breakthrough in Pancreatic Cancer Multidrug Resistance X.Y.Miao, J.Z. Liu, G.G.Xiao. School of Pharmaceutical Science and Technology, Dalian University of Technology, Dalian, China. Background: Pancreatic cancer is one of the most lethal malignancies on a global scale and its morbidity increases year by year. Prognosis for patients with locally advanced or metastatic disease is poor(5-year survival ratio<5%). This occurs not only in that it lacks a reliable marker for early detection, but also in that it can easily develop resistance to the anticancer drug. Discussion: Gemcitabine has been considered standard of care for treatment of advanced pancreatic cancer since 1997. Compared to 5-fluorouracil, Gemcitabine is found obviously to prolong patients’ survivals and improve clinical benefit response, however, Clinical treatment of pancreatic cancer soon develops resistance against gemcitabine. There are many studies on multidrug resistance of pancreatic cancer currently and lots of biological molecules have been found to be associated with drug resistance of pancreatic cancer, but It is still not clear how drug resistance occurs and spreads. As a kind of cell secretions, exosomes are composed of protein, RNA and lipid molecules. It is widely distributed and exists in various body fluids. It carries and transmits important information molecules, which is an important part of cell information transmission. Because of this property, it can be a good marker for early cancer detection and an entrance to the research of chemoresistance. MicroRNAs or alterations in intercellular communication play a dominant role in chemoresistance development, and recently several reports have found that in pancreatic cancer cells with drug resistance, the composition of exosomes have undergone significant changes. This is of great importance for the research of the production and spread of drug resistance in pancreatic cancer. In addition, exosomes are considered good drug carriers, which means it has great potential in terms of drug delivery. Conclusion: In this review, we have compiled the current research on the chemoresistance of pancreatic cancer and introduced the origin of the exosomes, its composition and its role in the cell growth. We discuss the relationship between exosomes and pancreatic cancer drug resistance, and we believe that further study on the composition of exosomes and Its mode of action in the intercellular will help the diagnosis and treatment of pancreatic cancer. Monoclonal Antibody Against Protein Disulphide Isomerase Anterior Gradient-2 as a Novel Agent in Suppressing Tumor Growth and Metastasis H. Negi,1 S.B. Merugu,1 Y. Tianhong,1 H.B. Mangukiya,1 S. Kamle,1 Q. Sehar,1 D. Li.1,21School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China, 2Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, Shanghai, Shanghai, China. Background: Anterior gradient 2 (AGR2) is a protein disulfide isomerase-like protein widely expressed in many normal tissues as well as cancers. AGR2 is overexpressed in multiple cancers, particularly those arising from lung, breast and prostate tissues,and higher levels of AGR2 are associated with earlier patient death. Metastasis is one of the areas which need immediate attention. There have been lots of proteins reported to have helped in the progress of metastasis. Anterior Gradient -2 (AGR2) is one of them. AGR2 protein has been demonstrated to interact with C4.4A and DAG-1 proteins which are associated with metastasis formation since these transmembrane proteins are involved in cell and matrix interactions between cancer and normal cells. Method: Knowing the importance of AGR2 in the cancer development, we designed, synthesized and purified a novel monoclonal antibody 18A4 against AGR2. We investigated the anti-metastatic and anti-tumorigenic activity of MAb 18A4 in experimental pulmonary lung melanoma metastasis model and non-small cell lung carcinoma (NSCLC) xenograft models.