Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
Filter
  • Li, Zhiying  (8)
  • Wang, Hongyu  (8)
  • 1
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2023
    In:  Cancer Research Vol. 83, No. 7_Supplement ( 2023-04-04), p. 5198-5198
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 83, No. 7_Supplement ( 2023-04-04), p. 5198-5198
    Abstract: Introduction: GARP (Glycoprotein-A repetitions predominant) is a cell surface receptor which is coded by Lrrc32 (Leucine rich repeat containing protein 32) and involved in the activation of TGF-beta. The expression of GARP has been widely found in various cells including Treg, activated B cells, mesenchymal stromal cells and many types of cancer cells. In the tumor microenvironment (TME), GARP from Treg promotes the growth of tumor via secretion of immune suppressive TGF-beta. Result: We established GARP humanized mouse model (hGARP) on C57BL/6 background in which the signal peptide and extracellular domain of mouse GARP (mGARP) was replaced by human GARP (hGARP). Similar expression level of hGARP on Treg cells in splenocytes from C57BL/6-hGARP mice was observed compared with mGARP in wildtype mice. After treating with anti-hGARP antibody, tumor growth was inhibited in the cohort engrafted with MC38 colorectal cancer cell line. Flow cytometry analysis of the tumor infiltrating lymphocytes revealed an abundant accumulation of hGARP on the Treg cells suggesting that blocking of hGARP in Treg cells contributed to the therapeutic efficiency. In addition, hGARP expression was also found on platelet cells from C57BL/6-hGARP mice, which is similar to human. After treating with anti-hGARP, we also found a significant reduction of platelet cells in C57BL/6-hGARP mice. In vivo assay suggested that the reduction of platelet cells from C57BL/6-hGARP mice correlated well with the binding of anti-hGARP. Conclusion: C57BL/6-hGARP is a well-established mouse model for the pre-clinical evaluation of both therapeutic effect and toxicity of anti-hGARP therapy. Citation Format: Xing Liu, Meirong Wu, Huiyi Wang, Weiwei Yu, Jianming Xu, Hongyan Sun, Cunxiang Ju, Hongyu Wang, Santi Suryani Chen, Zhiying Li, Mark Wade Moore, Jing Zhao, Xiang Gao. Humanized mice for preclinical efficacy evaluation of drugs targeting GARP. [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 5198.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2023
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 83, No. 7_Supplement ( 2023-04-04), p. 3242-3242
    Abstract: Antibody-drug conjugates (ADCs) are effective anti-tumor therapies with high specificity, with little toxicity and side effects. Xenograft tumor models based on immunodeficient mice have been used for evaluating the human anti-tumor efficacy of ADCs. However, the antibody-mediated immune response of ADCs is not well evaluated in immunodeficient mice. To this end, we have developed BALB/c-hPD1 humanized mouse model to evaluate the efficacy of ADCs in combination with immunotherapy. Trastuzumab deruxtecan (T-DXd, DS-8201a) is a HER2-targeting ADC. In vitro efficacy studies showed that T-DXd effectively bound to the HER2-positive cell line SK-BR-3, resulting in strong endocytosis and induction of apoptosis. It has also been shown to kill HER2-negative MDA-MB-468 cells through a bystander effect both in vitro and in vivo. In vivo efficacy studies have demonstrated that ADCs can effectively inhibit human tumor cell growth in immunodeficient mice. In addition to human tumor cell lines, the efficacy of ADCs was also studied in BALB/c-hPD1mice engrafted with murine colon cancer cells model, and the results showed that T-DXd and Keytruda could significantly inhibit CT26-hHER2 tumor growth, with the tumor growth inhibition rates being 57% and 66%, respectively, Tumor growth inhibition rate of the combined treatment group was about 91%, which was superior compared to the efficacy of single drug treatment. The evaluation of Fc segment-mediated effect (such as ADCC, CDC and ADCP) of ADCs antibodies was carried out in humanized immune system mouse models, such as NCG-hIL15 and NCG-hSGM3. In summary, we have established a drug evaluation platform using various humanized mouse models to study the synergistic anti-tumor effects of ADCs combined with immune checkpoint blockade antibodies. Citation Format: Fang Zhu, Hongyan Sun, Qian Lu, Yuan Fang, Jianming Xu, Cunxiang Ju, Hongyu Wang, Santi Suryani Chen, Zhiying Li, Mark Wade Moore, Jing Zhao, Xiang Gao. Efficacy evaluation by novel humanized mouse models for preclinical study of ADCs combined with immunotherapy [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 3242.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2023
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 83, No. 7_Supplement ( 2023-04-04), p. 5199-5199
    Abstract: Immune checkpoint molecule B7-H3, also known as CD276, is a member of the B7-CD28 family of immunomodulatory proteins. It is a type I membrane protein with sequence similarities to the extracellular domain of programmed death-ligand 1 (PD-L1). B7-H3 is highly expressed in most human cancers, but has limited distribution in normal tissues, remaining elusive of its receptor. Due to its promising safety as a dominant tumor target, various strategies have been developed to modulate the effect of B7-H3 via monoclonal antibodies, bispecific antibodies, ADC, or CAR-T. To study the effect of these therapies in an immunocompetent mouse model, we established a double humanized B7-H3 and PD-L1 mouse model on BALB/c background (BALB/c-hB7-H3/hPD-L1). In this model, the extracellular domain of murine fragments was replaced by the human counterparts while the trans-membrane and cytoplasmic domain were kept intact. When engrafted with CT26 colon cancer cells, which stably overexpress human B7-H3 and PD-L1 while endogenous murine counterparts were knocked out, the tumor growth was inhibited certain degree by anti-B7-H3 antibody (8H9 Biosimilar, 20mpk, TGI=18.56%) treatment while inhibited significantly after the monotherapy of anti-PD-L1 (Tecentriq, 3 mpk, TGI=55.89%, p & lt;0.001). The same dosage combination of anti-B7-H3 and anti-PD-L1 had a significant inhibition on tumor growth (TGI=76.85%, p & lt;0.001), and had a synergistic effect (CDI & lt;0.7) compared with the monotherapy. Analysis of tumor-infiltrating lymphocytes (TILs) at the end of the efficacy study showed that the proportion of CD45+ immune cells was significantly increased in all of the treated groups. The NK cells were significantly increased and the Treg cells were significantly decreased especially in the combined treatment group. Based on the above, the B7-H3 and PD-L1 double humanized mouse model is suitable for the pre-clinical evaluation of mono or combined immune checkpoint blockade with anti-human B7-H3 and PD-L1 therapy. Citation Format: Shiying Guo, Lingyu Song, Xin Qin, Jianming Xu, Hongyan Sun, Cunxiang Ju, Hongyu Wang, Santi Suryani Chen, Zhiying Li, Mark Wade Moore, Jing Zhao, Xiang Gao. B7-H3 & PD-L1 double-humanized BALB/c mouse: a novel animal model for preclinical studies of human B7-H3 antibodies or bispecific antibodies. [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 5199.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2023
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 4
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 83, No. 7_Supplement ( 2023-04-04), p. 5197-5197
    Abstract: Interleukin 2 is a key molecule that promotes the expansion and activation of lymphocytes, including T regulatory cells (Tregs) and natural killer (NK) cells. Tregs are specialized subpopulation of T cells that suppress the immune response by inhibiting T cell proliferation and cytokine production. NK cells are the predominant innate immune subset that mediates anti-tumor and anti-viral responses. Both Tregs and NK cells are two critical immune cells that play an important role in cancer immunotherapy, however, there is a lack of models that supports the in vivo status of these two immune subsets. To address this, we developed a severe immunodeficient mouse model, NOD/ShiLtJGpt-Prkdcem26Cd52Il2rgem26Cd22/Gpt (NCG-hIL2), by knocking in the human IL-2 gene on the NCG background. The ability of this model to support Treg and NK cells was then compared to NCG and tested with the reconstitution of human peripheral blood mononuclear cells (PBMC) and human hematopoietic stem cells (HSC). In the PBMC engrafted mice, the presence of IL-2 supported the development of T cells, especially the Tregs, with 53.74% in Tregs in NCG-IL2 compared to 4.81% in NCG mice (test at 2 weeks of reconstruction). Similarly, the NK cells are also supported with those cohort engrafted with human HSC (91.63% in NK cells in NCG-IL2 compared to 0.53% in NCG mice, test at 10 weeks of reconstruction). Notably, reconstituted NK cells expressed various NK receptors such as NKp30, NKp44, NKp46, NKG2D, and CD94 in NCG-IL2 mice. They produced comparable levels of granzyme when compared with human peripheral blood-derived NK cells, and a considerable amount of perforin protein was detected in the plasma of huHSC-NCG-hIL2 mice. In conclusion, compared with NCG mice, humanized NCG-hIL2 mice can reconstruct more abundant immune cells and have a wider application in anti-tumor research. It is an ideal model for the preclinical study of anti-tumor therapeutics. Citation Format: Xinxin Zhang, Meirong Wu, Huiyi Wang, Weiwei Yu, Fang Zhu, Jianming Xu, Hongyan Sun, Hongyu Wang, Cunxiang Ju, Santi Suryani Chen, Zhiying Li, Mark Wade Moore, Jing Zhao, Xiang Gao. NCG-hIL2 mice: a novel model to support the development of T regulatory and NK cells. [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 5197.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2023
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 5
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 83, No. 7_Supplement ( 2023-04-04), p. 6653-6653
    Abstract: Despite the success of programmed death ligand 1 (PD-L1) and programmed death 1 (PD-1) blockage as cancer therapies, the overall clinical response rates are still unsatisfying, with only a 25% response rate in most cancers. The underlying mechanisms of non-responsiveness towards anti-PD-L1 treatment have been extensively studied and associated with many factors, including PD1/PD-L1 expression level, tumor mutation burden, IFN signaling, loss of MHC-I, tumor microenvironment (immune infiltration, suppressive immune system), and so on. Immunodeficient mice such as the NSG and NCG are commonly used to study the efficacy of cancer immunotherapies. These mice, reconstituted with either human PBMC or HSC, act as an avatar to provide a window of the potential agent efficacy at the preclinical level. However, due to the challenge of obtaining an HSC/PBMC donor, and often cost and time can also be a limiting factor, many studies only use 2-5 human donors. Here we present data demonstrating the significant variation observed between donor-to-donor, highlighting the importance of testing multiple donors. We reconstituted immune system and tumor tissue humanized mouse models using peripheral blood mononuclear cells (PBMCs), and cell line-derived xenografts (CDX) in severe immunodeficiency NCG mice. Anti-tumor efficacy of anti-PD-L1 (Tecentriq) in huPBMC-NCG mice engrafted with several tumor cells including MDA-MB-231, HCC827, NUGC4, and some other CDX cell lines was tested in our system. In the cases of MDA-MB-231, HCC827, and NUGC4, different donors were enrolled in the same study, and tumor growth inhibition measured by tumor volume (TGITV) of 3% to 52.40% (MDA-MB-231), 5% to 33% (HCC827) and 4.75% to 20.74% (NUGC4) were observed. Especially, In the case of MDA-MB-231, 6 donors were enrolled in this study at the same time. About 40% of immune infiltration was first observed in tumors. High expression of PD1 on immune cells and PDL1 on tumor cells were also detected from different donors simultaneously. Although the similar immune infiltration and PD1/PDL1 expression, the TGITV of 6 donors varied between 3% and 52.40%, suggesting the donor-dependent response to anti-PD-L1 treatment, which is similar to the clinical trials. Our data showed diverse in vivo responses when multiple human PBMC donors were used for preclinical agent evaluation. While unsurprising, it showed the critical need to include various human PBMC donors when an agent is in preclinical status because the donors' immune profile may dictate the study's outcome. This can be divided into two parts, on the one hand, donor screening for favorable efficacy response donors in advance is an alternative approach to ensure the positive efficacy of anti-PDL1 blockade relative therapy. On the other hand, the low-efficacy response donors also can be candidates for anti-PD1/PDL1 combination therapy. Citation Format: Tingting Gu, Weiwei Yu, Hongyan Sun, Fang Zhu, Huiyi Wang, Mengting Wang, Shuai Li, Jianming Xu, Santi Chen, Zhiying Li, Mark Wade Moore, Cunxiang Ju, Hongyu Wang, Jing Zhao, Xiang Gao. Analysis of selective response of anti-PD-L1 treatment in huPBMC-NCG mice reconstituted with different donors [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 6653.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2023
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 6
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 83, No. 7_Supplement ( 2023-04-04), p. 4472-4472
    Abstract: Hematological malignancies, including leukemia, lymphoma and multiple myeloma, account for about 7% of all newly diagnosed cancers. Allogeneic hematopoietic stem cell transplantation (HSCT) is a traditional treatment for hematologic malignancies. Currently, CAR-T cell therapy has made a major breakthrough in the treatment of hematologic malignancies. However, the occurrence of acute Graft-versus-Host Disease (GvHD) represents a major obstacle for HSCT and limits the application of CAR-T cell therapy. GemPharmatech established and characterized two murine models for anti-GvHD drug evaluation. In the first model, bone marrow from C57BL/6 donor mice was transplanted into a sublethally-irradiated BALB/c recipient mouse. In this model, T cell infiltration, tissue damage, and systemic inflammation were observed, similar to the clinical observations of patients receiving allogeneic HSCT. Efficacy evaluation of anti-GVHD drugs have been carried out based on this model. Our results showed that treatment with the anti-GVHD drug Ibrutinib, the survival rate and the lifespan of the mice was significantly prolonged, and the GvHD score was reduced by about 70% compared with the placebo group. In the second model, human Peripheral Blood Mononuclear cells (PBMCs) were transplanted into irradiated severe immunodeficient NCG mice. The cohort was irradiated to accelerate GvHD occurrence. The reconstructed human T cells will recognize and attack mouse tissues to cause GvHD. Our results showed that hCD45+ cells infiltrated into heart, liver, and spleen of huPBMC-NCG mice. This model has also been successfully used for anti-GvHD efficacy evaluation. Upon treatment with an anti-GVHD drug, the lifespan of huPBMC-NCG mice was significantly extended and weight loss was alleviated. Overall, the above-mentioned GvHD models show great potential for preclinical evaluation of anti-GvHD drug development in allogeneic HSCT. Citation Format: Meirong Wu, Shiru Zhang, Fang Zhu, Hongyan Sun, Jianming Xu, Cunxiang Ju, Hongyu Wang, Santi Suryani Chen, Zhiying Li, Mark Wade Moore, Jing Zhao, Xiang Gao. A mouse model for GvHD: an excellent tool for evaluating the efficacy of anti-GvHD drugs and testing mechanisms of immunoregulation. [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 4472.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2023
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 7
    Online Resource
    Online Resource
    The American Association of Immunologists ; 2023
    In:  The Journal of Immunology Vol. 210, No. 1_Supplement ( 2023-05-01), p. 237.12-237.12
    In: The Journal of Immunology, The American Association of Immunologists, Vol. 210, No. 1_Supplement ( 2023-05-01), p. 237.12-237.12
    Abstract: Immunodeficient NCG mice were unable to reconsituted human NK cells, but after knockin of human IL-15 (hIL-15), the NCG-hIL15 mice could assist the reconstitution of human NK cells. Compared with NCG, the level of hIL-15 was significantly increased in heterozygous and homozygous NCG-hIL15 mice. After transplanting human PBMC (huPBMC-NCG-hIL15) or NK cells into NCG-hIL15 (huNK-NCG-IL15), the level of peripheral blood hCD56+ NK cells in huNK-NCG-hIL15 mice were much higher than that in huPBMC-NCG-hIL15 mice. However, the reconstituted proportion of human CD3+ T cells in huNK-NCG-hIL15 mice was not comparable in huPBMC-NCG-hIL15 mice. The level of NK cell reconstitution is also highly dependent on the donor NK cells. The expression of perforin in the peripheral blood of huNK-NCG-hIL15 mice was significantly higher than that in huPBMC-NCG-hIL15 mice. In vitro functional analysis of NK cells from huPBNK-NCG-hIL15 and huPBMC-NCG-hIL15 showed Granzyme B expression in peripheral blood were comparable and human NK cells purified from splenocytes of huPBNK-NCG-hIL15 mice were cytotoxic upon coculture with Raji cells in the presence of Rituximab. Based on these in vitro data, we established huNK-NCG-hIL15 mice subcutaneously engrafted with Raji cells, and the in vivo efficacy of rituximab was evaluated. Efficacy study data showed that rituximab significantly inhibited the Raji tumor cells growth in huNK-NCG-hIL15 mice. Both huPBMC-NCG-hIL15 and huNK-NCG-hIL15 mouse model can rapidly reconstitute functional human NK cells compared to CD34+ HSC engrafted NCG-hIL15 mice. The development of huNK-NCG-hIL15 is an ideal mouse model to specifically evaluate the anti-tumor efficacy of drugs targeting human NK cells.
    Type of Medium: Online Resource
    ISSN: 0022-1767 , 1550-6606
    RVK:
    RVK:
    Language: English
    Publisher: The American Association of Immunologists
    Publication Date: 2023
    detail.hit.zdb_id: 1475085-5
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 8
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 83, No. 7_Supplement ( 2023-04-04), p. 3970-3970
    Abstract: Introduction: IL-15 is a four α-helix bundle cytokine which is produced by dendritic cells, monocytes, and epithelial cells. IL-15 is necessary for the development, survival, and activation of natural killer (NK) cells. NK cells are unable to persist in immunodeficient NCG mice reconsituted with human NK cells, but after knocking in human IL-15 (hIL-15), NCG-hIL15 mice can support the reconstitution and survival of human NK cells. Results: Compared with NCG mice, the level of hIL-15 was significantly increased in NCG-hIL15 mice. After transplanting human PBMC (huPBMC) or NK (huNK) cells into NCG-hIL15 mice, the peripheral blood level of NK cells in huNK-NCG-hIL15 mice was much higher than in huPBMC-NCG-hIL15 mice. The expression of perforin in peripheral blood of huNK-NCG-hIL15 mice was significantly higher than that in huPBMC-NCG-hIL15 mice. Considering the obvious ADCC of rituximab acting on Raji cells in vitro, we evaluated the efficacy of rituximab in huNK-NCG-hIL15 mice subcutaneously engrafted with Raji cells. Rituximab significantly inhibited the tumor growth in huNK-NCG-hIL15 mice. Conclusion: NCG-hIL15 is a great mouse model to evaluate the anti-tumor efficacy of drugs targeting human NK cells without the interference of T cells. Citation Format: Xing Liu, Meirong Wu, Huiyi Wang, Weiwei Yu, Jianming Xu, Hongyan Sun, Cunxiang Ju, Hongyu Wang, Santi Suryani Chen, Zhiying Li, Mark Wade Moore, Jing Zhao, Xiang Gao. NCG-hIL15 humanized mice: an ideal model for human immune reconstitution of NK cells. [abstract] . In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 3970.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2023
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. Further information can be found on the KOBV privacy pages