GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

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

Proceed reservation?

Export
  • 1
    In: Antibody Therapeutics, Oxford University Press (OUP), Vol. 1, No. 2 ( 2018-09-17), p. 65-73
    Abstract: Programmed cell death 1 (PD-1) is an inhibitory immune checkpoint expressed on activatedT cells. Upon the formation of T cell receptor (TCR)-pMHC complexes, concomitant PD-1 ligation to its ligands programmed death-ligand 1 (PD-L1) or programmed death-ligand 2 (PD-L2) downregulates TCR signaling and effector function. Here we describe the preclinical characterization of Sintilimab, a fully human IgG4 antibody that potently blocks PD-1 interactions with PD-L1 and PD-L2. Methods The binding affinity and blockade function were detected by using surface plasmon resonance (SPR), Enzyme-linked immunosorbent assay (ELISA) and flow cytometry. The biology function properties were measured with luciferase assay and mixed lymphocyte reaction assay. In vivo anti-tumor function and preclinical pharmacokinetic (PK) were identified with human PD-1 transgenic mice and non-human primates separately. Results Sintilimab can specifically and strongly bind to human PD-1 (hPD-1) and cynomolgus PD-1 and the affinity of Sintilimab to human PD-1 was measured at 0.3 nm via surface SPR, and displayed slow dissociation kinetics. Sintilimab can block the interaction of PD-1 to PD-L1 and PD-L2 and induce high secretion levels of interferon (IFN)-γ and interleukin (IL)-2 in primary T cell assays. In humanized hPD-1 knock-in mouse models, Sintilimab showed potent anti-tumor activity and increased tumor-infiltrating CD8/CD4 T cell and CD8/ Treg ratios. Preclinical experimentation in non-human primates following a single intravenous infusion of Sintilimab at 1, 6 and 30 mg/kg presented with no signs of drug-related toxicity, and showed typical PK characteristics of an IgG antibody. Conclusions Sintilimab has desirable preclinical attributes that supports its clinical development for cancer treatment.
    Type of Medium: Online Resource
    ISSN: 2516-4236
    Language: English
    Publisher: Oxford University Press (OUP)
    Publication Date: 2018
    detail.hit.zdb_id: 3031893-2
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Biomedicine & Pharmacotherapy, Elsevier BV, Vol. 67, No. 5 ( 2013-6), p. 437-444
    Type of Medium: Online Resource
    ISSN: 0753-3322
    RVK:
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2013
    detail.hit.zdb_id: 392415-4
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 12_Supplement ( 2022-06-15), p. 5527-5527
    Abstract: Background: TIGIT (T-cell immunoglobulin and ITIM domain), which is primarily expressed on activated and 'exhausted' T and NK cells, is one of the promising 'next generation' immune checkpoint molecules. Engagement of TIGIT to its ligands (i.e., PVR and PVRL2) leads to inhibitory signaling in T cells, promoting functional exhaustion of tumor-infiltrating T lymphocytes. Anti-TIGIT monoclonal antibodies have shown clinical benefit when combined with anti-PD-L1 agents in NSCLC. However, the single-agent efficacy of anti-TIGIT therapies have been limited. PVRIG (PVR-related immunoglobulin domain containing), which is another coinhibitory receptor of the DNAM/TIGIT/CD96 nectin family, binds with high affinity to PVRL2 and suppresses T-cell function, and shows nonredundant inhibitory effects alongside the TIGIT/PVR/PVRL2 axis. Here, we report a fully-human anti-TIGIT × PVRIG bispecific antibody (anti-TIGIT × PVRIG biAb), which blocks both the PVRIG/PVRL2 and TIGIT/PVR/PVRL2 pathways, that maintains the efficacy of the combination of the two mono-agents. The anti-TIGIT × PVRIG biAb is also highly efficacious when combined with PD1/PD-L1 inhibitors in mouse tumor models. Methods: An anti-TIGIT × PVRIG biAb was generated through the fusing of a fully-human IgG targeting TIGIT with a wild type G1-Fc to a fully-human scFv at the c-terminus targeting PVRIG. Binding affinity and specificity analyses were studied by flow cytometry and biolayer interferometry. The co-binding of the anti-TIGIT × PVRIG biAb to TIGIT and PVRIG was detected by ELISA. The immunomodulatory functions of the anti-TIGIT × PVRIG biAb were evaluated using a luciferase reporter cell assay in vitro and human PBMC-based tumor models in vivo. Results: The anti-TIGIT × PVRIG biAb binds with high affinity to the extracellular domain of human TIGIT/PVRIG and can bind to TIGIT and PVRIG simultaneously. In a competition assay, the anti-TIGIT × PVRIG biAb efficiently blocked the interaction between TIGIT and PVR/PVRRL2, and PVRIG with PVRL2. In a luciferase reporter cell system, the anti-TIGIT × PVRIG biAb induced high levels of luciferase activity compared with the anti-TIGIT or anti-PVRIG mAbs alone. In vivo, the anti-TIGIT × PVRIG biAb demonstrated stronger anti-tumor efficacy than the anti-TIGIT and anti-PVRIG mAbs as monotherapies or combined with anti-PD-1 mAb. Conclusion: Our anti-TIGIT × PVRIG biAb, a fully human bispecific antibody, either alone or in combination with anti-PD-1 mAb promotes immune cell activation both in vitro and in vivo, supporting its clinical development for the treatment of human cancers. The molecule is currently under GLP-toxicity evaluation in NHP, and a first-in-human study is expected to begin in 2022. Citation Format: Shuang Dai, Weifeng Huang, Zhijun Yuan, Shaogang Peng, Jiayi Si, Chao Wang, Xiaoniu Miao, Yingda Xu, Joanne Sun, Xiaolin Liu, Andy Tsun, Tianhang Zhai. A novel fully human anti-TIGIT and PVRIG bispecific antibody that elicits potent anti-tumor efficacy in pre-clinical studies [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 5527.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2022
    detail.hit.zdb_id: 2036785-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2022
    In:  Cancer Research Vol. 82, No. 12_Supplement ( 2022-06-15), p. 5555-5555
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 12_Supplement ( 2022-06-15), p. 5555-5555
    Abstract: Background: Gastric cancer is one of the most common cancers worldwide, which is estimated to have 32.1 and 13.2 cases per 100,000 individuals in males and females, respectively. Claudin-18 isoform 2 (CLDN18.2), a membrane-bound protein involved in the formation of tight junctions, has been identified to be highly expressed in various types of cancers such as gastric and pancreatic cancer. Targeting CLDN18.2 with the monoclonal antibody (mAb) Zolbetuximab has achieved moderate clinical benefit with controllable toxicity. 4-1BB is a potent stimulator of T cells and NK cells, and when activated, can improve effector and/or memory responses. However, inherent on-target related toxic effects in the liver was observed during the clinical development of the monoclonal agonist antibody Urelumab. Here, we report the discovery of a humanized bispecific antibody (biAb) anti-CLDN18.2 x 4-1BB, which activates immune cells such as T cells via CLDN18.2-mediated crosslinking of 4-1BB. This bispecific antibody induces potent anti-tumor efficacy with limited toxicity. Methods: The anti-CLDN18.2 x 4-1BB biAb was generated by the fusion of an anti-4-1BB VHH targeting the CRD4 domain for 4-1BB, to the C-terminus of an anti-CLDN18.2 mAb via a G4S linker. Binding affinity and specificity testing of each arm were studied by flow cytometry and on the Retrogenix Cell Microarray Technology platform, a human membrane/secreted protein binding array. The immunomodulatory functions were evaluated using luciferase reporter cell assays in vitro and human 4-1BB KI mouse models in vivo. Results: The CLDN18.2 antibody interacted specifically to CLDN18.2 on the Retrogenix Cell Microarray platform among over 5,500 proteins. Next, in vitro studies showed how the molecule specifically induced T cell activation and cytokine release against CLDN18.2-positive cells. More importantly, such activation was correlated to the expression level of CLDN18.2, which indicates that the molecule may preferentially recognize CLDN18.2-high-expressing cancer cells. In mouse models, the anti-CLDN18.2 x 4-1BB biAb induced superior anti-tumor efficacy compared to anti-CLDN18.2 and anti-4-1BB mAbs alone with acceptable PK attributes. Furthermore, the anti-CLDN18.2 x 4-1BB biAb induced strong immunological memory where tumors could not grow in re-challenged tumor-free mice. Finally, the CLDN18.2 x 4-1BB biAb showed a safe profile in a GLP NHP study of which no obvious signs of toxicity were observed at 50 mg/kg, 100 mg/kg or 150 mg/kg doses. Conclusion: In summary, a bispecific antibody targeting CLDN18.2 and 4-1BB was developed, which displayed potent anti-tumor efficacy with strong immunological memory and has shown good safety in NHPs. Moreover, the molecule has high selectivity to CLDN18.2. The molecule shall enter clinical trials by early 2022. Citation Format: Junying Chen, Weifeng Huang, Zhiquan Liang, Xiaoniu Miao, Shaogang Peng, Chao Wang, Tiantian Dong, Andy Tsun, Yi Luo. A bispecific antibody targeting CLDN18.2 and 4-1BB induces potent anti-tumor efficacy with an extraordinary safety profile [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 5555.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2022
    detail.hit.zdb_id: 2036785-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2022
    In:  Cancer Research Vol. 82, No. 12_Supplement ( 2022-06-15), p. 6115-6115
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 12_Supplement ( 2022-06-15), p. 6115-6115
    Abstract: Background: Although the discovery and development of first-generation of immune checkpoint inhibitors (towards PD1 and CTLA-4) was a major milestone for cancer therapy, current clinical response rates are still considered very limited. Combination treatments are predicted to improve on these current immunotherapies including the targeting of the adenosine pathway (CD39, CD73 or A2AR), which has shown a lot of promise in preclinical and early clinical studies. CD73 is an ecto-5′-nucleotidase which transforms adenosine monophosphate (AMP) to adenosine. Adenosine is a soluble immunosuppressive metabolite that can suppress natural killer cells and cytotoxic CD8+ T cells. Blockade of CD73-mediated conversion of AMP to adenosine may therefore recover anti-tumor immunity through preventing the enrichment of adenosine in the tumor microenvironment. Method: In this campaign, two humanized and Fc-silenced IgG antibodies were generated named 7002-01 and 7002-04. The target binding epitopes of these candidates were revealed by binning experiments through bio-layer interferometry. Cell binding experiments were tested on human and cynomolgus CD73 overexpression CHO cell lines by flow cytometry. Cellular CD73 enzyme inhibition experiments were tested using A375, MDA-MB-231, H2030 and BT549 tumor cell lines via the CellTiter-Glo method. Soluble CD73 enzymatic tests were carried out on patient sera or recombinant CD73 protein using a similar method. T cell proliferation assays were performed using PBMC. In vivo efficacy studies were tested in B-NDG B2M-KO mice that were injected subcutaneously with A375 tumor cells and human PBMC. Results: Two candidates, 7002-01 and 7002-04, were selected based on their functional activity, that recognize different non-overlapping binding epitopes on CD73. Both candidates selectively bind to and can inhibit the activities of both membrane-bound and soluble human CD73 to high levels and seem to maintain inhibition at high dose-ranges without a hook effect. Both candidates can potently rescue adenosine-mediated T cell regulation. Additionally, 7002-01 and 7002-04 have combination synergy or additive effects for CD73 inhibition on soluble CD73, tumor cell lines that express CD73, and PBMC. 7002-01 and 7002-04 have single-agent anti-tumor efficacy and combination synergy with anti-PD1 antibodies in mice. 7002-01 has a typical antibody-like PK profile when rhesus monkeys were administered with a single intravenous dose at 25 or 50 mg/kg. No drug-related toxicities have been observed in GLP toxicity studies with dosages at 50, 250, and 500 mg/kg (QW, 4 weeks). Conclusion: Highly differentiating anti-CD73 antibodies were discovered that show maximal inhibition of both membranous and soluble CD73 without hook effects at high concentrations. 7002-01 was chosen as the lead molecule for its better overall activity profile and should be entering clinical trials by early 2022. Citation Format: Zhenqing Zhang, Yunli Jia, Xiaoniu Miao, Weifeng Huang, Chao Wang, Zhijun Yuan, Wenchao Jiang, Zhiyuan Li, Liandi Chen, Andy Tsun. Development of functionally differentiating anti-CD73 antibodies for cancer therapy [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 6115.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2022
    detail.hit.zdb_id: 2036785-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: mAbs, Informa UK Limited, Vol. 12, No. 1 ( 2020-01-01)
    Type of Medium: Online Resource
    ISSN: 1942-0862 , 1942-0870
    Language: English
    Publisher: Informa UK Limited
    Publication Date: 2020
    detail.hit.zdb_id: 2537838-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Journal for ImmunoTherapy of Cancer, BMJ, Vol. 9, No. 6 ( 2021-06), p. e002131-
    Abstract: The discovery of checkpoint inhibitors towards cytotoxic T-lymphocyte protein 4 (CTLA-4) and programmed cell death protein 1 (PD-1) has been revolutionary for the treatment of cancers. These therapies have only offered an average of 20%–30% response rates across the tumor spectrum and the combination of agonists towards the tumor-necrosis superfamily members, such as 4-1BB and CD40, has shown potent efficacy in preclinical studies; however, these agonists have exhibited high degrees of toxicity with limited efficacy in human trials. In this study, we have generated a single-domain antibody towards a unique epitope of 4-1BB that limits its potential on-target toxicity while maintaining sufficient potency. This 4-1BB binder is ideal for use in the engineering of multispecific antibodies to localize 4-1BB activation within the tumor microenvironment, as shown here by a anti-PD-L1/4-1BB bispecific candidate (PM1003). Methods To determine the functional activity of the 4-1BB- and PD-L1-binding elements of PM1003, in vitro luciferase reporter and primary cell assays were used to test the potency of programmed cell death 1 ligand 1 (PD-L1) blockade and PD-L1-mediated 4-1BB activation via cross-bridging. X-ray crystallography was conducted to resolve the binding epitopes of the respective binding arms, and accurate binding kinetics were determined using standard affinity measurement techniques. Human 4-1BB and/or PD-L1 knock-in mice were used in cancer models for testing the in vivo antitumor efficacy of PM1003, and safety was evaluated further. Results PM1003 shows potent activation of 4-1BB and blockade of PD-L1 in cell-based assays. 4-1BB activation was exerted through the bridging of PD-L1 on target cells and 4-1BB on effector cells. No PD-L1-independent activation of 4-1BB was observed. Through X-ray crystallography, a unique binding epitope in the cysteine-rich domain 4 (CRD4) region was resolved that provides high potency and potentially low on-target toxicity as determined by primary immune cell assays and toxicity evaluation in vivo . Conclusions A unique single-domain antibody was discovered that binds to the CRD4 domain of 4-1BB. When incorporated into a 4-1BB/PD-L1 bispecific (PM1003), we have shown the potent inhibition of PD-L1 activity with 4-1BB agonism upon cross-bridging with PD-L1 in vitro . Antitumor activity with minimal toxicity was found in vivo . Thus, PM1003 is a uniquely differentiating and next generation therapeutic agent for cancer therapy.
    Type of Medium: Online Resource
    ISSN: 2051-1426
    Language: English
    Publisher: BMJ
    Publication Date: 2021
    detail.hit.zdb_id: 2719863-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2022
    In:  Cancer Research Vol. 82, No. 12_Supplement ( 2022-06-15), p. 5554-5554
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 12_Supplement ( 2022-06-15), p. 5554-5554
    Abstract: Background: The carcinoembryonic antigen-related adhesion molecules (CEACAM), containing seven members, are involved in many biological processes including cell adhesion, proliferation, differentiation, and tumor suppression. Some variants of CEACAMs such as CEACAM5 and CEACAM6 have long been recognized to be overexpressed in many cancers and highly associated with cancer development. Immunotherapies targeting CEACAM have achieved some clinical benefit, e.g. Tusamitamab Ravtansine (anti-CEACAM5 ADC), that has shown decent disease control in NSCLC. Recently, with the success of CAR-T and T cell engager (TCE) therapies against blood cancers, TCEs against solid tumors have also been developed. For instance, Cibisatamab, targeting CEACAM5 and CD3, is currently under clinical investigation in CRC patients. Here, we report the discovery of a bispecific antibody (biAb) anti-CEACAM5/6 x CD3 that mediates direct T cell killing against CEACAM5- and/or CEACAM6-expressing cancer cells. Methods: Anti-CEACAM5/6 x CD3 TCE consists of two CEACAM5/6 binding Fabs for the preferential recognition of CEACAM5/6-high-expressing cancer cells, and one CD3 binding domain that permits CEACAM5/6-mediated CD3 crosslinking and T cell activation. To reduce off-target T cell stimulation there is no CH2 in this format to eliminate Fc receptor binding, while an anti-HSA VHH was included into this construct for half-life extension. Binding affinity and specificity were studied by flow cytometry, IHC and on the Retrogenix Cell Microarray Technology platform, a human membrane/secreted protein binding array. The immunomodulatory functions were evaluated using luciferase reporter cell assays, cell killing assays in vitro and via PBMC-based Hu-NSG mouse models in vivo. Results: Our CEACAM5/6 binding antibody only interacted with CEACAM5 and CEACAM6 on the Retrogenix Cell Microarray against over 5,500 proteins. Importantly, it showed a similar sensitivity to recognize CRC tumor tissues in an IHC array as the benchmark (BMK), but significantly better selectivity at recognition of tumor tissues vs. normal tissues. Next, in vitro testing illustrated CEACAM5/6-mediated T cell activation by the TCE, and such stimulation also correlated with the binding affinity of the anti-CD3 domain. The lead candidate with the optimized CD3 binding affinity, showed better anti-tumor efficacy than the BMK in mice. Furthermore, this molecule displayed a typical PK as a conventional antibody with a mean T1/2 of around 115hrs. Finally, we show high tolerance in NHP of which no obvious signs of toxicity were observed at 2.5 mg/kg. Conclusion: In summary, a bispecific TCE with high selectivity to CEACAM5/6 overexpressing cancer cells was engineered. It displays potent anti-tumor efficacy, and support from target selectivity testing shows promising safety traits. Citation Format: Liandi Chen, Weifeng Huang, Xiaoniu Miao, Jiayi Si, Chao Wang, Tiantian Dong, Andy Tsun, Yi Luo. A bispecific T cell engager targeting CEACAM5/6 exhibits strong anti-tumor efficacy in pre-clinical studies [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 5554.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2022
    detail.hit.zdb_id: 2036785-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2024
    In:  Cancer Research Vol. 84, No. 6_Supplement ( 2024-03-22), p. 5083-5083
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 84, No. 6_Supplement ( 2024-03-22), p. 5083-5083
    Abstract: Background: The epidermal growth factor receptor (EGFR) family, consisting of EGFR, HER2, HER3, and HER4, plays a critical role in tumorigenesis and tumor progression. Inhibition of ERBBs using either monoclonal antibodies (mAb) or small-molecule tyrosine kinase inhibitors have been approved for use for the treatment of various types of cancers. Despite relative success, many patients acquire resistance to long-term benefit due to alternative compensatory signaling pathways. Accordingly, activating mutations or overexpression of HER3 has been reported in many cancers, such as breast, ovarian, lung, colorectal, melanoma, head and neck, cervical and prostate cancers. Here, we report the discovery of a humanized anti-EGFR x HER3 IgG-like bispecific antibody-drug conjugate (ADC). The ADC takes a asymmetric 1+1 form through HC-HC and HC-LC charge-based heterodimerization, which is then conjugated to a topoisomerase 1 inhibitor (TOP1i) payload via a cleavable linker. The affinity of EGFR and HER3 was optimized to improve the therapeutical window, and has shown potent anti-tumor efficacy in various tumor models with limited toxicity supporting further development in clinic. Methods: An anti-EGFR x HER3 bispecific ADC (PM1300) was generated by introducing unique mutations to the CH1-CL domains of each binding arm to avoid HC-LC mispairing. KIH mutations were also introduced to each CH3 domain to support HC/HC heterodimerization and generate an IgG-like bispecific with one arm binding to EGFR and the other to HER3. TOP1i payload was conjugated to the IgG-like bispecific by the endogenous cysteine residues at DAR 8 via a clearable linker. Results: Through affinity optimization, PM300 was found to preferentially bind to EGFR/HER3 double-positive cells rather than EGFR single-positive cells. PM1300 showed significantly increased internalization and anti-proliferation activity against EGFR/HER3 double-positive cells, with limited activity towards single positive-cells. PM1300 used an optimized linker for better payload release and serum stability. PM1300 induced a potent anti-tumor efficacy in various CDX models representing different EGFR and HER3 expressing levels and mutations. Importantly, PM1300 exhibited a good safety profile in NHP consistent with the optimal affinity for EGFR and HER3. These results demonstrate that PM1300 has a promising efficacy/safety therapeutical window in preclinical models supporting further development in the clinic. Conclusion: A bispecific ADC targeting EGFR x HER3 was discovered, named PM1300, which has an asymmetric 1+1 IgG-like structure and optimized affinity. This allows for preferential binding to EGFR/HER3 double-positive cancer cells, rather than EGFR single-positive cells. This may significantly contribute to minimizing safety risk that is common for EGFR-targeting agents. Citation Format: Liandi Chen, Shaogang Peng, Chao Wang, Xiaoniu Miao, Yao Yan, Jun Xing, Weifeng Huang, Andy Tsun, Yingda Xu, Xiaolin Liu, Yi Luo. Discovery of an asymmetric IgG-like bispecific ADC targeting EGFR/HER3 [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2024; Part 1 (Regular Abstracts); 2024 Apr 5-10; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2024;84(6_Suppl):Abstract nr 5083.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2024
    detail.hit.zdb_id: 2036785-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2017
    In:  Cancer Research Vol. 77, No. 13_Supplement ( 2017-07-01), p. 2661-2661
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 77, No. 13_Supplement ( 2017-07-01), p. 2661-2661
    Abstract: The rise of therapeutic antibodies directed towards T cell checkpoint inhibitors has paved the way to a novel and exciting frontier of cancer treatments. Spearheaded by anti-CTLA-4 and PD-1/PD-L1 inhibitors, many investigators have strived to uncover combinatory therapies that could synergize with CTLA-4 and PD-1 inhibitors to improve the overall survival of cancer patients. The TNFR-superfamily has been seen as an ideal combinatory target in providing co-stimulatory T cell signals in sync with the unleashing of T cells from their CTLA-4 and PD-1 brakes. The ideal outcome of these combinations is the induction of T cell memory and effector function towards tumour cells. Current clinical-stage agents against TNFR-superfamily targets include conventional or Fc-engineered therapeutic antibodies and multimeric ligands that aim to cluster and activate targets such as OX40, 4-1BB, GITR, and CD27. For OX40 and GITR, wildtype IgG1 variants offer target cell depletion of OX40/GITR-high expressing Treg cells, and also provide agonism through FcgR-mediated crosslinking. We have taken a broad approach of screening IgG1, IgG2, Fc-engineered and tetravalent antibodies to select for potent therapeutic antibody candidates to TNFR-superfamily targets, and conducted humanized mouse studies as a screen for candidate potency. Citation Format: Cheng Chen, Xiaoniu Miao, Yao Yan, Liang Tang, Bingliang Chen, Junjian Liu, Xiaolin Liu, Michael Yu, Andy Tsun. A broad approach for the selection of therapeutic mAbs to TNFR-superfamily members for use in combination with an anti-PD-1 mAb [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 2661. doi:10.1158/1538-7445.AM2017-2661
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2017
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...