Network


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

Hotspot


Dive into the research topics where Baiheng Li is active.

Publication


Featured researches published by Baiheng Li.


Journal of Hematology & Oncology | 2015

Quantitative evaluation of the immunodeficiency of a mouse strain by tumor engraftments

Wei Ye; Zhiwu Jiang; Guan Xiong Li; Yiren Xiao; Simiao Lin; Yunxin Lai; Suna Wang; Baiheng Li; Bei Jia; Yin Li; Zhi Liang Huang; Jin Li; Fenglan Feng; Shuhua Li; Huihui Yao; Zixia Liu; Su Cao; Lin Xu; Yangqiu Li; Donghai Wu; Lingwen Zeng; Mei Zhong; Pentao Liu; Zhe Sheng Wen; Bing Xu; Yao Yao; Duanqing Pei; Peng Li

BackgroundThe mouse is an organism that is widely used as a mammalian model for studying human physiology or disease, and the development of immunodeficient mice has provided a valuable tool for basic and applied human disease research. Following the development of large-scale mouse knockout programs and genome-editing tools, it has become increasingly efficient to generate genetically modified mouse strains with immunodeficiency. However, due to the lack of a standardized system for evaluating the immuno-capacity that prevents tumor progression in mice, an objective choice of the appropriate immunodeficient mouse strains to be used for tumor engrafting experiments is difficult.MethodsIn this study, we developed a tumor engraftment index (TEI) to quantify the immunodeficiency response to hematologic malignant cells and solid tumor cells of six immunodeficient mouse strains and C57BL/6 wild-type mouse (WT).ResultsMice with a more severely impaired immune system attained a higher TEI score. We then validated that the NOD-scid-IL2Rg−/− (NSI) mice, which had the highest TEI score, were more suitable for xenograft and allograft experiments using multiple functional assays.ConclusionsThe TEI score was effectively able to reflect the immunodeficiency of a mouse strain.


OncoImmunology | 2017

PSCA and MUC1 in non-small-cell lung cancer as targets of chimeric antigen receptor T cells

Xinru Wei; Yunxin Lai; Jin Li; Le Qin; Youdi Xu; Ruocong Zhao; Baiheng Li; Simiao Lin; Suna Wang; Qiting Wu; Qiubin Liang; Muyun Peng; Fenglei Yu; Yangqiu Li; Zhang X; Yi-Long Wu; Pentao Liu; Duanqing Pei; Yao Yao; Peng Li

ABSTRACT In recent years, immunotherapies, such as those involving chimeric antigen receptor (CAR) T cells, have become increasingly promising approaches to non-small-cell lung cancer (NSCLC) treatment. In this study, we explored the antitumor potential of prostate stem cell antigen (PSCA)-redirected CAR T and mucin 1 (MUC1)-redirected CAR T cells in tumor models of NSCLC. First, we generated patient-derived xenograft (PDX) mouse models of human NSCLC that maintained the antigenic profiles of primary tumors. Next, we demonstrated the expression of PSCA and MUC1 in NSCLC, followed by the generation and confirmation of the specificity and efficacy of PSCA- and MUC1-targeting CAR T cells against NSCLC cell lines in vitro. Finally, we demonstrated that PSCA-targeting CAR T cells could efficiently suppress NSCLC tumor growth in PDX mice and synergistically eliminate PSCA+MUC1+ tumors when combined with MUC1-targeting CAR T cells. Taken together, our studies demonstrate that PSCA and MUC1 are both promising CAR T cell targets in NSCLC and that the combinatorial targeting of these antigens could further enhance the antitumor efficacy of CAR T cells.


Frontiers in Immunology | 2017

Anti-GPC3-CAR T Cells Suppress the Growth of Tumor Cells in Patient-Derived Xenografts of Hepatocellular Carcinoma

Zhiwu Jiang; Xiaofeng Jiang; Suimin Chen; Yunxin Lai; Xinru Wei; Baiheng Li; Simiao Lin; Suna Wang; Qiting Wu; Qiubin Liang; Qifa Liu; Muyun Peng; Fenglei Yu; Jianyu Weng; Xin Du; Duanqing Pei; Pentao Liu; Yao Yao; Ping Xue; Peng Li

Background The lack of a general clinic-relevant model for human cancer is a major impediment to the acceleration of novel therapeutic approaches for clinical use. We propose to establish and characterize primary human hepatocellular carcinoma (HCC) xenografts that can be used to evaluate the cytotoxicity of adoptive chimeric antigen receptor (CAR) T cells and accelerate the clinical translation of CAR T cells used in HCC. Methods Primary HCCs were used to establish the xenografts. The morphology, immunological markers, and gene expression characteristics of xenografts were detected and compared to those of the corresponding primary tumors. CAR T cells were adoptively transplanted into patient-derived xenograft (PDX) models of HCC. The cytotoxicity of CAR T cells in vivo was evaluated. Results PDX1, PDX2, and PDX3 were established using primary tumors from three individual HCC patients. All three PDXs maintained original tumor characteristics in their morphology, immunological markers, and gene expression. Tumors in PDX1 grew relatively slower than that in PDX2 and PDX3. Glypican 3 (GPC3)-CAR T cells efficiently suppressed tumor growth in PDX3 and impressively eradicated tumor cells from PDX1 and PDX2, in which GPC3 proteins were highly expressed. Conclusion GPC3-CAR T cells were capable of effectively eliminating tumors in PDX model of HCC. Therefore, GPC3-CAR T cell therapy is a promising candidate for HCC treatment.


Journal of Hematology & Oncology | 2017

Incorporation of a hinge domain improves the expansion of chimeric antigen receptor T cells

Le Qin; Yunxin Lai; Ruocong Zhao; Xinru Wei; Jianyu Weng; Peilong Lai; Baiheng Li; Simiao Lin; Suna Wang; Qiting Wu; Qiubin Liang; Yangqiu Li; Zhang X; Yi-Long Wu; Pentao Liu; Yao Yao; Duanqing Pei; Xin Du; Peng Li

BackgroundMultiple iterations of chimeric antigen receptors (CARs) have been developed, mainly focusing on intracellular signaling modules. However, the effect of non-signaling extracellular modules on the expansion and therapeutic efficacy of CARs remains largely undefined.MethodsWe generated two versions of CAR vectors, with or without a hinge domain, targeting CD19, mesothelin, PSCA, MUC1, and HER2, respectively. Then, we systematically compared the effect of the hinge domains on the growth kinetics, cytokine production, and cytotoxicity of CAR T cells in vitro and in vivo.ResultsDuring in vitro culture period, the percentages and absolute numbers of T cells expressing the CARs containing a hinge domain continuously increased, mainly through the promotion of CD4+ CAR T cell expansion, regardless of the single-chain variable fragment (scFv). In vitro migration assay showed that the hinges enhanced CAR T cells migratory capacity. The T cells expressing anti-CD19 CARs with or without a hinge had similar antitumor capacities in vivo, whereas the T cells expressing anti-mesothelin CARs containing a hinge domain showed enhanced antitumor activities.ConclusionsHence, our results demonstrate that a hinge contributes to CAR T cell expansion and is capable of increasing the antitumor efficacy of some specific CAR T cells. Our results suggest potential novel strategies in CAR vector design.


Journal of Hematology & Oncology | 2016

Heterogeneity of CD34 and CD38 expression in acute B lymphoblastic leukemia cells is reversible and not hierarchically organized

Zhiwu Jiang; Manman Deng; Xinru Wei; Wei Ye; Yiren Xiao; Simiao Lin; Suna Wang; Baiheng Li; Xin Liu; Gong Zhang; Peilong Lai; Jianyu Weng; Donghai Wu; Haijia Chen; Wei Wei; Yuguo Ma; Yangqiu Li; Pentao Liu; Xin Du; Duanqing Pei; Yao Yao; Bing Xu; Peng Li

The existence and identification of leukemia-initiating cells in adult acute B lymphoblastic leukemia (B-ALL) remain controversial. We examined whether adult B-ALL is hierarchically organized into phenotypically distinct subpopulations of leukemogenic and non-leukemogenic cells or whether most B-ALL cells retain leukemogenic capacity, irrespective of their immunophenotype profiles. Our results suggest that adult B-ALL follows the stochastic stem cell model and that the expression of CD34 and CD38 in B-ALL is reversibly and not hierarchically organized.


Leukemia | 2018

Toll-like receptor 2 costimulation potentiates the antitumor efficacy of CAR T Cells

Yunxin Lai; Jianyu Weng; Xinru Wei; Le Qin; Peilong Lai; Ruocong Zhao; Zhiwu Jiang; Baiheng Li; Simiao Lin; Suna Wang; Qiting Wu; Z Tang; Pentao Liu; Duanqing Pei; Yao Yao; Xin Du; Peng Li

Chimeric antigen receptor (CAR) T-cell immunotherapies have shown unprecedented success in treating leukemia but limited clinical efficacy in solid tumors. Here, we generated 1928zT2 and m28zT2, targeting CD19 and mesothelin, respectively, by introducing the Toll/interleukin-1 receptor domain of Toll-like receptor 2 (TLR2) to 1928z and m28z. T cells expressing 1928zT2 or m28zT2 showed improved expansion, persistency and effector function against CD19+ leukemia or mesothelin+ solid tumors respectively in vitro and in vivo. In a patient with relapsed B-cell acute lymphoblastic leukemia, a single dose of 5 × 104/kg 1928zT2 T cells resulted in robust expansion and leukemia eradication and led to complete remission. Hence, our results demonstrate that TLR2 signaling can contribute to the efficacy of CAR T cells. Further clinical trials are warranted to establish the safety and efficacy of this approach.


Scientific Reports | 2017

CRISPR/Cas9-Mediated Deletion of Foxn1 in NOD/SCID/IL2rg-/- Mice Results in Severe Immunodeficiency.

Xinru Wei; Yunxin Lai; Baiheng Li; Le Qin; Youdi Xu; Simiao Lin; Suna Wang; Qiting Wu; Qiubin Liang; Guohua Huang; Qiuhua Deng; Pentao Liu; Donghai Wu; Liangxue Lai; Yao Yao; Peng Li

Immunodeficient mice engrafted with either normal or cancerous human cells are widely used in basic and translational research. In particular, NOD/SCID/IL2rg−/− mice can support the growth of various types of human cancer cells. However, the hairs of these mice interfere with the observation and imaging of engrafted tissues. Therefore, novel hairless strains exhibiting comparable immunodeficiency would be beneficial. Recently, the CRISPR/Cas9 system has been used for efficient multiplexed genome editing. In the present study, we generated a novel strain of nude NOD/SCID/IL2rg−/− (NSIN) mice by knocking out Foxn1 from NOD/SCID/IL2rg−/− (NSI) mice using the CRISPR/Cas9 system. The NSIN mice were deficient in B, T, and NK cells and not only showed impaired T cell reconstitution and thymus regeneration after allogeneic bone marrow nucleated cell transplantation but also exhibited improved capacity to graft both leukemic and solid tumor cells compared with NSI, NOG, and NDG mice. Moreover, the NSIN mice facilitated the monitoring and in vivo imaging of both leukemia and solid tumors. Therefore, our NSIN mice provide a new platform for xenograft mouse models in basic and translational research.


Frontiers in Immunology | 2017

CD215+ Myeloid Cells Respond to Interleukin 15 Stimulation and Promote Tumor Progression

Shouheng Lin; Guohua Huang; Yiren Xiao; Wei Sun; Yuchuan Jiang; Qiuhua Deng; Muyun Peng; Xinru Wei; Wei Ye; Baiheng Li; Simiao Lin; Suna Wang; Qiting Wu; Qiubin Liang; Yangqiu Li; Zhang X; Yi-Long Wu; Pentao Liu; Duanqing Pei; Fenglei Yu; Zhesheng Wen; Yao Yao; Donghai Wu; Peng Li

Interleukin 15 (IL-15) regulates the development, survival, and functions of multiple innate and adaptive immune cells and plays a dual role in promoting both tumor cell growth and antitumor immunity. Here, we demonstrated that the in vivo injection of recombinant human IL-15 (200 µg/kg) or murine IL-15 (3 µg/kg) to tumor-bearing NOD-SCID-IL2Rg−/− (NSI) mice resulted in increased tumor progression and CD45+ CD11b+ Gr-1+ CD215+ cell expansion in the tumors and spleen. In B16F10-bearing C57BL/6 mice model, we found that murine IL-15 has antitumoral effect since the activation and expansion of CD8+ T cells with murine IL-15 treatment. But no enhanced or reduced tumor growth was observed in mice when human IL-15 was used. However, both murine and human IL-15 promote CD45+ CD11b+ Gr-1+ CD215+ cells expansion. In xenograft tumor models, CD215+ myeloid cells, but not CD215− cells, responded to human IL-15 stimulation and promoted tumor growth. Furthermore, we found that human IL-15 mediated insulin-like growth factor-1 production in CD215+ myeloid cells and blocking IGF-1 reduced the tumor-promoting effect of IL-15. Finally, we observed that higher IGF-1 expression is an indicator of poor prognosis among lung adenocarcinoma patients. These findings provide evidence that IL-15 may promote tumor cell progression via CD215+ myeloid cells, and IGF-1 may be an important candidate that IL-15 facilitates tumor growth.


mAbs | 2018

Establishment of peripheral blood mononuclear cell-derived humanized lung cancer mouse models for studying efficacy of PD-L1/PD-1 targeted immunotherapy

Shouheng Lin; Guohua Huang; Lin Cheng; Zhen Li; Yiren Xiao; Qiuhua Deng; Yuchuan Jiang; Baiheng Li; Simiao Lin; Suna Wang; Qiting Wu; Huihui Yao; Su Cao; Yang Li; Pentao Liu; Wei Wei; Duanqing Pei; Yao Yao; Zhesheng Wen; Zhang X; Yi-Long Wu; Zhenfeng Zhang; Shuzhong Cui; Xiaofang Sun; Xueming Qian; Peng Li

ABSTRACT Animal models used to evaluate efficacies of immune checkpoint inhibitors are insufficient or inaccurate. We thus examined two xenograft models used for this purpose, with the aim of optimizing them. One method involves the use of peripheral blood mononuclear cells and cell line-derived xenografts (PBMCs-CDX model). For this model, we implanted human lung cancer cells into NOD-scid-IL2Rg−/− (NSI) mice, followed by injection of human PBMCs. The second method involves the use of hematopoietic stem and progenitor cells and CDX (HSPCs-CDX model). For this model, we first reconstituted the human immune system by transferring human CD34+ hematopoietic stem and progenitor cells (HSPCs-derived humanized model) and then transplanted human lung cancer cells. We found that the PBMCs-CDX model was more accurate in evaluating PD-L1/PD-1 targeted immunotherapies. In addition, it took only four weeks with the PBMCs-CDX model for efficacy evaluation, compared to 10–14 weeks with the HSPCs-CDX model. We then further established PBMCs-derived patient-derived xenografts (PDX) models, including an auto-PBMCs-PDX model using cancer and T cells from the same tumor, and applied them to assess the antitumor efficacies of anti-PD-L1 antibodies. We demonstrated that this PBMCs-derived PDX model was an invaluable tool to study the efficacies of PD-L1/PD-1 targeted cancer immunotherapies. Overall, we found our PBMCs-derived models to be excellent preclinical models for studying immune checkpoint inhibitors.


Oncotarget | 2017

Defined, serum/feeder-free conditions for expansion and drug screening of primary B-acute lymphoblastic leukemia

Zhiwu Jiang; Di Wu; Wei Ye; Jianyu Weng; Peilong Lai; Pengcheng Shi; Xutao Guo; Guohua Huang; Qiuhua Deng; Yanlai Tang; Hongyu Zhao; Shuzhong Cui; Simiao Lin; Suna Wang; Baiheng Li; Qiting Wu; Yangqiu Li; Pentao Liu; Duanqing Pei; Xin Du; Yao Yao; Peng Li

Functional screening for compounds represents a major hurdle in the development of rational therapeutics for B-acute lymphoblastic leukemia (B-ALL). In addition, using cell lines as valid models for evaluating responses to novel drug therapies raises serious concerns, as cell lines are prone to genotypic/phenotypic drift and loss of heterogeneity in vitro. Here, we reported that OP9 cells, not OP9-derived adipocytes (OP9TA), support the growth of primary B-ALL cells in vitro. To identify the factors from OP9 cells that support the growth of primary B-ALL cells, we performed RNA-Seq to analyze the gene expression profiles of OP9 and OP9TA cells. We thus developed a defined, serum/feeder-free condition (FI76V) that can support the expansion of a range of clinically distinct primary B-ALL cells that still maintain their leukemia-initiating ability. We demonstrated the suitability of high-throughput drug screening based on our B-ALL cultured conditions. Upon screening 378 kinase inhibitors, we identified a cluster of 17 kinase inhibitors that can efficiently kill B-ALL cells in vitro. Importantly, we demonstrated the synergistic cytotoxicity of dinaciclib/BTG226 to B-ALL cells. Taken together, we developed a defined condition for the ex vivo expansion of primary B-ALL cells that is suitable for high-throughput screening of novel compounds.

Collaboration


Dive into the Baiheng Li's collaboration.

Top Co-Authors

Avatar

Peng Li

Guangzhou Institutes of Biomedicine and Health

View shared research outputs
Top Co-Authors

Avatar

Simiao Lin

Guangzhou Institutes of Biomedicine and Health

View shared research outputs
Top Co-Authors

Avatar

Suna Wang

Guangzhou Institutes of Biomedicine and Health

View shared research outputs
Top Co-Authors

Avatar

Yao Yao

Guangzhou Institutes of Biomedicine and Health

View shared research outputs
Top Co-Authors

Avatar

Duanqing Pei

Guangzhou Institutes of Biomedicine and Health

View shared research outputs
Top Co-Authors

Avatar

Pentao Liu

Wellcome Trust Sanger Institute

View shared research outputs
Top Co-Authors

Avatar

Qiting Wu

Guangzhou Institutes of Biomedicine and Health

View shared research outputs
Top Co-Authors

Avatar

Xinru Wei

Guangzhou Institutes of Biomedicine and Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yunxin Lai

Guangzhou Institutes of Biomedicine and Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge