Network


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

Hotspot


Dive into the research topics where Jiankai Liang is active.

Publication


Featured researches published by Jiankai Liang.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Identification and characterization of alphavirus M1 as a selective oncolytic virus targeting ZAP-defective human cancers

Yuan Lin; Haipeng Zhang; Jiankai Liang; Kai Li; Wenbo Zhu; Liwu Fu; Fang Wang; Xiaoke Zheng; Huijuan Shi; Sihan Wu; Xiao Xiao; Lijun Chen; Lipeng Tang; Min Yan; Xiaoxiao Yang; Yaqian Tan; Pengxin Qiu; Yijun Huang; Wei Yin; Xinwen Su; Haiyan Hu; Jun Hu; Guangmei Yan

Significance Although oncolytic virotherapy is showing great promise in clinical trials, not all patients are benefiting. Identifying predictors of therapeutic effectiveness for each oncolytic virus would provide a good chance to increase response rate. Here, we describe an alphavirus (M1) that possesses selective and potent antitumor activity through intravenous infusion, whereas its replication is controlled by the zinc-finger antiviral protein (ZAP) gene. A survey of cancer tissue banks reveals that ZAP is commonly deficient in human cancers, suggesting extensive application prospects of M1. Our work provides an example of a potentially personalized cancer therapy using a targeted oncolytic virus that can be selectively administered to patients with ZAP-deficient tumors. We predict that such agents will form the armamentarium of cancer therapy in the future. Oncolytic virotherapy is a growing treatment modality that uses replicating viruses as selective antineoplastic agents. Safety and efficacy considerations dictate that an ideal oncolytic agent would discriminate between normal and cancer cells on the basis of common genetic abnormalities in human cancers. Here, we identify a naturally occurring alphavirus (M1) as a novel selective killer targeting zinc-finger antiviral protein (ZAP)-deficient cancer cells. In vitro, in vivo, and ex vivo studies showed potent oncolytic efficacy and high tumor tropism of M1. We showed that the selectivity depends on ZAP deficiency by systematic identification. A large-scale multicenter pathology study using tissue microarrays reveals that ZAP is commonly deficient in human cancers, suggesting extensive application prospects for M1. Additionally, M1 killed cancer cells by inducing endoplasmic reticulum stress-mediated apoptosis. Our report provides novel insights into potentially personalized cancer therapy using oncolytic viruses.


Molecular Therapy | 2016

Activation of Cyclic Adenosine Monophosphate Pathway Increases the Sensitivity of Cancer Cells to the Oncolytic Virus M1.

Kai Li; Haipeng Zhang; Jianguang Qiu; Yuan Lin; Jiankai Liang; Xiao Xiao; Liwu Fu; Fang Wang; Jing Cai; Yaqian Tan; Wenbo Zhu; Wei Yin; Bingzheng Lu; Fan Xing; Lipeng Tang; Min Yan; Jialuo Mai; Yuan Li; Wenli Chen; Pengxin Qiu; Xingwen Su; Guangping Gao; Phillip W.L. Tai; Jun Hu; Guangmei Yan

Oncolytic virotherapy is a novel and emerging treatment modality that uses replication-competent viruses to destroy cancer cells. Although diverse cancer cell types are sensitive to oncolytic viruses, one of the major challenges of oncolytic virotherapy is that the sensitivity to oncolysis ranges among different cancer cell types. Furthermore, the underlying mechanism of action is not fully understood. Here, we report that activation of cyclic adenosine monophosphate (cAMP) signaling significantly sensitizes refractory cancer cells to alphavirus M1 in vitro, in vivo, and ex vivo. We find that activation of the cAMP signaling pathway inhibits M1-induced expression of antiviral factors in refractory cancer cells, leading to prolonged and severe endoplasmic reticulum (ER) stress, and cell apoptosis. We also demonstrate that M1-mediated oncolysis, which is enhanced by cAMP signaling, involves the factor, exchange protein directly activated by cAMP 1 (Epac1), but not the classical cAMP-dependent protein kinase A (PKA). Taken together, cAMP/Epac1 signaling pathway activation inhibits antiviral factors and improves responsiveness of refractory cancer cells to M1-mediated virotherapy.


Human Gene Therapy | 2016

Naturally Existing Oncolytic Virus M1 Is Nonpathogenic for the Nonhuman Primates After Multiple Rounds of Repeated Intravenous Injections

Haipeng Zhang; Yuan Lin; Kai Li; Jiankai Liang; Xiao Xiao; Jing Cai; Yaqian Tan; Fan Xing; Jialuo Mai; Yuan Li; Wenli Chen; Longxiang Sheng; Jiayu Gu; Wenbo Zhu; Wei Yin; Pengxin Qiu; Xingwen Su; Bingzheng Lu; Xuyan Tian; Jinhui Liu; Wanjun Lu; Yunling Dou; Yijun Huang; Zhuang Kang; Guangping Gao; Zixu Mao; Shi Yuan Cheng; Ling Lu; Xuetao Bai; Guangmei Yan

Cancers figure among the leading causes of morbidity and mortality worldwide. The number of new cases is expected to rise by about 70% over the next 2 decades. Development of novel therapeutic agents is urgently needed for clinical cancer therapy. Alphavirus M1 is a Getah-like virus isolated from China with a genome of positive single-strand RNA. We have previously identified that alphavirus M1 is a naturally existing oncolytic virus with significant anticancer activity against different kinds of cancer (e.g., liver cancer, bladder cancer, and colon cancer). To support the incoming clinical trial of intravenous administration of alphavirus M1 to cancer patients, we assessed the safety of M1 in adult nonhuman primates. We previously presented the genome sequencing data of the cynomolgus macaques (Macaca fascicularis), which was demonstrated as an ideal animal species for virus infection study. Therefore, we chose cynomolgus macaques of either sex for the present safety study of oncolytic virus M1. In the first round of administration, five experimental macaques were intravenously injected with six times of oncolytic virus M1 (1 × 10(9) pfu/dose) in 1 week, compared with five vehicle-injected control animals. The last two rounds of injections were further completed in the following months in the same way as the first round. Body weight, temperature, complete blood count, clinical biochemistries, cytokine profiles, lymphocytes subsets, neutralizing antibody, and clinical symptoms were closely monitored at different time points. Magnetic resonance imaging was also performed to assess the possibility of encephalitis or arthritis. As a result, no clinical, biochemical, immunological, or medical imaging or other pathological evidence of toxicity was found during the whole process of the study. Our results in cynomolgus macaques suggested the safety of intravenous administration of oncolytic virus M1 in cancer patients in the future.


Cell Reports | 2017

The Anti-Warburg Effect Elicited by the cAMP-PGC1α Pathway Drives Differentiation of Glioblastoma Cells into Astrocytes

Fan Xing; Yizhao Luan; Jing Cai; Sihan Wu; Jialuo Mai; Jiayu Gu; Haipeng Zhang; Kai Li; Yuan Lin; Xiao Xiao; Jiankai Liang; Yuan Li; Wenli Chen; Yaqian Tan; Longxiang Sheng; Bingzheng Lu; Wanjun Lu; Mingshi Gao; Pengxin Qiu; Xingwen Su; Wei Yin; Jun Hu; Zhongping Chen; Ke Sai; Jing Wang; Furong Chen; Yinsheng Chen; Shida Zhu; Dongbing Liu; Shi Yuan Cheng

SUMMARY Glioblastoma multiforme (GBM) is among the most aggressive of human cancers. Although differentiation therapy has been proposed as a potential approach to treat GBM, the mechanisms of induced differentiation remain poorly defined. Here, we established an induced differentiation model of GBM using cAMP activators that specifically directed GBM differentiation into astroglia. Transcriptomic and proteomic analyses revealed that oxidative phosphorylation and mitochondrial biogenesis are involved in induced differentiation of GBM. Dibutyryl cyclic AMP (dbcAMP) reverses the Warburg effect, as evidenced by increased oxygen consumption and reduced lactate production. Mitochondrial biogenesis induced by activation of the CREB-PGC1α pathway triggers metabolic shift and differentiation. Blocking mitochondrial biogenesis using mdivi1 or by silencing PGC1α abrogates differentiation; conversely, overexpression of PGC1α elicits differentiation. In GBM xenograft models and patient-derived GBM samples, cAMP activators also induce tumor growth inhibition and differentiation. Our data show that mitochondrial biogenesis and metabolic switch to oxidative phosphorylation drive the differentiation of tumor cells.


Oncotarget | 2016

A classical PKA inhibitor increases the oncolytic effect of M1 virus via activation of exchange protein directly activated by cAMP 1

Kai Li; Jiankai Liang; Yuan Lin; Haipeng Zhang; Xiao Xiao; Yaqian Tan; Jing Cai; Wenbo Zhu; Fan Xing; Jun Hu; Guangmei Yan

Oncolytic virotherapy is an emerging and promising treatment modality that uses replicating viruses as selective antitumor agents. Here, we report that a classical protein kinase A (PKA) inhibitor, H89, synergizes with oncolytic virus M1 in various cancer cells through activation of Epac1 (exchange protein directly activated by cAMP 1). H89 substantially increases viral replication in refractory cancer cells, leading to unresolvable Endoplasmic Reticulum stress, and cell apoptosis. Microarray analysis indicates that H89 blunts antiviral response in refractory cancer cells through retarding the nuclear translocation of NF-κB. Importantly, in vivo studies show significant antitumor effects during M1/H89 combination treatment. Overall, this study reveals a previously unappreciated role for H89 and demonstrates that activation of the Epac1 activity can improve the responsiveness of biotherapeutic agents for cancer.


Cell Death and Disease | 2015

Far upstream element-binding protein 1 is a prognostic biomarker and promotes nasopharyngeal carcinoma progression

Zhuowei Liu; Jian Hu; Jiankai Liang; Ai Jun Zhou; Manying Li; S. M. Yan; X. Zhang; S. Gao; Liang Chen; Qian Zhong; Mu Sheng Zeng

Nasopharyngeal carcinoma (NPC) is a malignant epithelial tumor with tremendous invasion and metastasis capacities, and it has a high incidence in southeast Asia and southern China. Previous studies identified that far upstream element-binding protein 1 (FBP1), a transcriptional regulator of c-Myc that is one of the most frequently aberrantly expressed oncogenes in various human cancers, including NPC, is an important biomarker for many cancers. Our study aimed to investigate the expression and function of FBP1 in human NPC. Quantitative real-time RT-PCR (qRT-PCR), western blot and immunohistochemical staining (IHC) were performed in NPC cells and biopsies. Furthermore, the effect of FBP1 knockdown on cell proliferation, colony formation, side population tests and tumorigenesis in nude mice were measured by MTT, clonogenicity analysis, flow cytometry and a xenograft model, respectively. The results showed that the mRNA and protein levels of FBP1, which are positively correlated with c-Myc expression, were substantially higher in NPC than that in nasopharyngeal epithelial cells. IHC revealed that the patients with high FBP1 expression had a significantly poorer prognosis compared with the patients with low expression (P=0.020). In univariate analysis, high FBP1 and c-Myc expression predicted poorer overall survival (OS) and poorer progression-free survival. Multivariate analysis indicated that high FBP1 and c-Myc expression were independent prognostic markers. Knockdown of FBP1 reduced cell proliferation, clonogenicity and the ratio of side populations, as well as tumorigenesis in nude mice. These data indicate that FBP1 expression, which is closely correlated with c-Myc expression, is an independent prognostic factor and promotes NPC progression. Our results suggest that FBP1 can not only serve as a useful prognostic biomarker for NPC but also as a potential therapeutic target for NPC patients.


Science Translational Medicine | 2017

Targeting VCP enhances anticancer activity of oncolytic virus M1 in hepatocellular carcinoma

Haipeng Zhang; Kai Li; Yuan Lin; Fan Xing; Xiao Xiao; Jing Cai; Wenbo Zhu; Jiankai Liang; Yaqian Tan; Liwu Fu; Fang Wang; Wei Yin; Bingzheng Lu; Pengxin Qiu; Xingwen Su; Xuetao Bai; Jun Hu; Guangmei Yan

Inhibition of VCP sensitizes hepatocellular carcinoma cells to oncolytic virus M1–induced apoptosis. A virus and its reinforcement Oncolytic viruses can be effective against a variety of cancers, including hepatocellular carcinoma, where a viral treatment is showing evidence of efficacy in people. Zhang et al. performed a high-throughput drug screen to search for compounds to pair with an oncolytic virus called M1 to further increase its effectiveness against hepatocellular carcinoma. Through this screen, they identified inhibitors of valosin-containing protein, then used them together with M1, and demonstrated the efficacy of this regimen in mouse models of cancer. In addition, the combination was well tolerated in primates, suggesting that the drug and virus combination may translate to human patients. Oncolytic virotherapy is rapidly progressing through clinical evaluation. However, the therapeutic efficacy of oncolytic viruses in humans has been less than expected from preclinical studies. We describe an anticancer drug screen for compounds that enhance M1 oncolytic virus activity in hepatocellular carcinoma (HCC). An inhibitor of the valosin-containing protein (VCP) was identified as the top sensitizer, selectively increasing potency of the oncolytic virus up to 3600-fold. Further investigation revealed that VCP inhibitors cooperated with M1 virus–suppressed inositol-requiring enzyme 1α (IRE1α)–X-box binding protein 1 (XBP1) pathway and triggered irresolvable endoplasmic reticulum (ER) stress, subsequently promoting robust apoptosis in HCC. We show that VCP inhibitor improved the oncolytic efficacy of M1 virus in several mouse models of HCC and primary HCC tissues. Finally, this combinatorial therapeutic strategy was well tolerated in nonhuman primates. Our study identifies combined VCP inhibition and oncolytic virus as a potential treatment for HCC and demonstrates promising therapeutic potential.


Cell Death and Disease | 2018

Intravenous injections of the oncolytic virus M1 as a novel therapy for muscle-invasive bladder cancer

Cheng Hu; Ying Liu; Yuan Lin; Jiankai Liang; Wen-Wen Zhong; Ke Li; Wentao Huang; Dejuan Wang; Guangmei Yan; Wenbo Zhu; Jianguang Qiu; Xin Gao

Muscle-invasive bladder cancer (MIBC) is associated with low survival and high recurrence rates even in cases in which patients receive systemic treatments, such as surgery and chemotherapy. Here, we found that a naturally existing alphavirus, namely, M1, selectively kills bladder cancer cells but not normal cells, findings supported by our observations of changes in viral replication and MIBC and patient-derived MIBC cell apoptosis. Transcriptome analysis revealed that interferon-stimulated genes (ISGs) are expressed at low levels in sensitive bladder cancer cells and high levels in resistant cells. Knocking down ZC3HAV1 (ZAP), an antiviral factor in ISGs, restores M1 virus reactivity in resistant cells, and overexpressing ZAP partially reverses M1 virus-induced decreases in cell viability in sensitive cells. In orthotopic MIBC mice, tail vein injections of M1 significant inhibit tumor growth and prolong survival period, antitumor effects of M1 are stronger than those of the first-line chemotherapy agent cisplatin (CDDP). Treated tumors display enhanced cleaved-caspase-3 signals, which are representative of cell apoptosis, and decreased Ki-67 signals, which are representative of cell proliferation. Moreover, tissue microarray (TMA) analyses of clinical tumor specimens revealed that up to 45.6% of cases of MIBC presented with low ZAP expression, a finding that is prevalent in advanced MIBC. Our results indicate that the oncolytic virus M1 is a novel agent capable of functioning as a precise and effective therapy for MIBC.


Proceedings of the National Academy of Sciences of the United States of America | 2017

Selective replication of oncolytic virus M1 results in a bystander killing effect that is potentiated by Smac mimetics

Jing Cai; Yuan Lin; Haipeng Zhang; Jiankai Liang; Yaqian Tan; Webster K. Cavenee; Guangmei Yan

Significance Although oncolytic therapy is showing great potential in clinical trials, not all patients benefit from it. Combining oncolytic viruses with anticancer chemicals could provide a better chance to increase the response rate. Here, we report that the combination of an alphavirus (M1) that we identified previously and second mitochondria-derived activator of caspases (Smac) mimetic compounds (SMCs) shows substantial oncolytic effect in vitro, in vivo, and ex vivo (samples from patients’ tumor tissues). The combined effect is mediated by a bystander killing effect and increased replication of M1. Our work provides an example for potentiating the response rate in refractory samples by synergizing oncolytic virus with other anticancer chemicals. We predict that this treatment strategy will be a promising tool to combat cancer in the future. Oncolytic virotherapy is a treatment modality that uses native or genetically modified viruses that selectively replicate in and kill tumor cells. Viruses represent a type of pathogen-associated molecular pattern and thereby induce the up-regulation of dozens of cytokines via activating the host innate immune system. Second mitochondria-derived activator of caspases (Smac) mimetic compounds (SMCs), which antagonize the function of inhibitor of apoptosis proteins (IAPs) and induce apoptosis, sensitize tumor cells to multiple cytokines. Therefore, we sought to determine whether SMCs sensitize tumor cells to cytokines induced by the oncolytic M1 virus, thus enhancing a bystander killing effect. Here, we report that SMCs potentiate the oncolytic effect of M1 in vitro, in vivo, and ex vivo. This strengthened oncolytic efficacy resulted from the enhanced bystander killing effect caused by the M1 virus via cytokine induction. Through a microarray analysis and subsequent validation using recombinant cytokines, we identified IL-8, IL-1A, and TRAIL as the key cytokines in the bystander killing effect. Furthermore, SMCs increased the replication of M1, and the accumulation of virus protein induced irreversible endoplasmic reticulum stress- and c-Jun N-terminal kinase–mediated apoptosis. Nevertheless, the combined treatment with M1 and SMCs had little effect on normal and human primary cells. Because SMCs selectively and significantly enhance the bystander killing effect and the replication of oncolytic virus M1 specifically in cancer cells, this combined treatment may represent a promising therapeutic strategy.


Nature Communications | 2018

Inhibition of the mevalonate pathway enhances cancer cell oncolysis mediated by M1 virus

Jiankai Liang; Li Guo; Kai Li; Xiao Xiao; Wenbo Zhu; Xiaoke Zheng; Jun Hu; Haipeng Zhang; Jing Cai; Yaya Yu; Yaqian Tan; Chuntao Li; Xincheng Liu; Cheng Hu; Ying Liu; Pengxin Qiu; Xingwen Su; Songmin He; Yuan Lin; Guangmei Yan

Oncolytic virus is an attractive anticancer agent that selectively lyses cancer through targeting cancer cells rather than normal cells. Although M1 virus is effective against several cancer types, certain cancer cells present low sensitivity to it. Here we identified that most of the components in the cholesterol biosynthesis pathway are downregulated after M1 virus infection. Further functional studies illustrate that mevalonate/protein farnesylation/ras homolog family member Q (RHOQ) axis inhibits M1 virus replication. Further transcriptome analysis shows that RHOQ knockdown obviously suppresses Rab GTPase and ATP-mediated membrane transporter system, which may mediate the antiviral effect of RHOQ. Based on this, inhibition of the above pathway significantly enhances the anticancer potency of M1 virus in vitro, in vivo, and ex vivo. Our research provides an intriguing strategy for the rational combination of M1 virus with farnesyl transferase inhibitors to enhance therapeutic efficacy.Oncolytic viruses selectively kill tumour cells and induce anti-tumour immunity. Here, the AUs demonstrate the anti-viral effect of the mevalonate pathway on oncolytic virus M1 in refractory cancer cells and provide evidence for a combination strategy of targeting the mevalonate pathway for potentiating oncolytic virus therapy.

Collaboration


Dive into the Jiankai Liang's collaboration.

Top Co-Authors

Avatar

Yuan Lin

Sun Yat-sen University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jing Cai

Sun Yat-sen University

View shared research outputs
Top Co-Authors

Avatar

Wenbo Zhu

Sun Yat-sen University

View shared research outputs
Top Co-Authors

Avatar

Yaqian Tan

Sun Yat-sen University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Xiao Xiao

Sun Yat-sen University

View shared research outputs
Top Co-Authors

Avatar

Kai Li

Sun Yat-sen University

View shared research outputs
Top Co-Authors

Avatar

Jun Hu

Sun Yat-sen University

View shared research outputs
Top Co-Authors

Avatar

Pengxin Qiu

Sun Yat-sen University

View shared research outputs
Researchain Logo
Decentralizing Knowledge