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: Cancer Immunology Research, American Association for Cancer Research (AACR), Vol. 10, No. 10 ( 2022-10-04), p. 1190-1209
    Abstract: Assessment of immune-cell subsets within the tumor immune microenvironment is a powerful approach to better understand cancer immunotherapy responses. However, the use of biopsies to assess the tumor immune microenvironment poses challenges, including the potential for sampling error, restricted sampling over time, and inaccessibility of some tissues/organs, as well as the fact that single biopsy analyses do not reflect discordance across multiple intrapatient tumor lesions. Immuno-positron emission tomography (PET) presents a promising translational imaging approach to address the limitations and assess changes in the tumor microenvironment. We have developed 89Zr-DFO-REGN5054, a fully human CD8A-specific antibody conjugate, to assess CD8+ tumor-infiltrating lymphocytes (TIL) pre- and posttherapy. We used multiple assays, including in vitro T-cell activation, proliferation, and cytokine production, and in vivo viral clearance and CD8 receptor occupancy, to demonstrate that REGN5054 has minimal impact on T-cell activity. Preclinical immuno-PET studies demonstrated that 89Zr-DFO-REGN5054 specifically detected CD8+ T cells in lymphoid tissues of CD8-genetically humanized immunocompetent mice (VelociT mice) and discerned therapy-induced changes in CD8+ TILs in two models of response to a CD20xCD3 T-cell activating bispecific antibody (REGN1979, odronextamab). Toxicology studies in cynomolgus monkeys showed no overt toxicity, and immuno-PET imaging in cynomolgus monkeys demonstrated dose-dependent clearance and specific targeting to lymphoid tissues. This work supports the clinical investigation of 89Zr-DFO-REGN5054 to monitor T-cell responses in patients undergoing cancer immunotherapy.
    Type of Medium: Online Resource
    ISSN: 2326-6066 , 2326-6074
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2022
    detail.hit.zdb_id: 2732517-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Cancer Immunology Research, American Association for Cancer Research (AACR), Vol. 8, No. 5 ( 2020-05-01), p. 596-608
    Abstract: Patients with hematologic cancers have improved outcomes after treatment with bispecific antibodies that bind to CD3 on T cells and that redirect T cells toward cancer cells. However, clinical benefit against solid tumors remains to be shown. We made a bispecific antibody that targets both the common prostate tumor–specific antigen PSMA and CD3 (PMSAxCD3) and provide evidence for tumor inhibition in several preclinical solid tumor models. Mice expressing the human extracellular regions of CD3 and PSMA were generated to examine antitumor efficacy in the presence of an intact immune system and PSMA expression in normal tissues. PSMAxCD3 accumulated in PSMA-expressing tissues and tumors as detected by immuno-PET imaging. Although PSMAxCD3 induced T-cell activation and showed antitumor efficacy in mice with low tumor burden, PSMAxCD3 lost efficacy against larger solid tumors, mirroring the difficulty of treating solid tumors in the clinic. Costimulatory receptors can enhance T-cell responses. We show here that costimulation can enhance the antitumor efficacy of PSMAxCD3. In particular, 4-1BB stimulation in combination with PSMAxCD3 enhanced T-cell activation and proliferation, boosted efficacy against larger tumors, and induced T-cell memory, leading to durable antitumor responses. The combination of CD3 bispecific antibodies and anti-4-1BB costimulation represents a therapeutic approach for the treatment of solid tumors.
    Type of Medium: Online Resource
    ISSN: 2326-6066 , 2326-6074
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2020
    detail.hit.zdb_id: 2732517-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 78, No. 13_Supplement ( 2018-07-01), p. 3033-3033
    Abstract: Persistent antigen exposure and inflammatory signals in tumors induce expression of various co-inhibitory or immune checkpoint receptors on T cells, including programmed death protein 1 (PD-1) and Lymphocyte-Activation Gene 3 (LAG-3). Therapeutic antibodies blocking such co-inhibitory receptors have produced durable antitumor responses as single agents and in combinations. In order to monitor LAG-3 expression and potential changes in expression due to therapeutic intervention, we have developed a radionuclide-conjugated antibody to LAG-3 for immuno-PET. The fully human anti-LAG3 antibody REGN3767 was radiolabeled with the positron-emitting radionuclide Zirconium-89 (89Zr) using the bifunctional chelator p-SCN-Bn-Deferoxamine (DFO). 89Zr-REGN3767 demonstrated high radiochemical purity and immunoreactivity in cell binding assays. The ability of 89Zr-REGN3767 to successfully identify LAG-3 expression in vivo was initially assessed using MC38 mouse tumors expressing human LAG-3 (MC38/hLAG-3) implanted into immune-deficient mice. 89Zr-REGN3767 demonstrated higher uptake in MC38/hLAG-3 tumors compared to an 89Zr-isotype control antibody using immuno-PET, and specificity was confirmed by ex vivo biodistribution at day 6 post radiotracer injection (~35 and ~5 %ID/g for 89Zr-REGN3767 and 89Zr-isotype, respectively). Furthermore, a dose titration study of 89Zr-REGN3767 in immune deficient mice co-implanted subcutaneously with Raji lymphoma cells and human peripheral blood mononuclear cells (hPBMCs) demonstrated the ability of 89Zr-REGN3767 to target LAG-3-expressing intratumoral T-cells. 89Zr-REGN3767 immuno-PET and ex vivo biodistribution demonstrated specific localization to Raji/hPBMC co-implanted tumors; this uptake was significantly higher at antibody doses of 0.03 - 0.3 mg/kg than at 5 mg/kg. Doses of 0.03-0.3 mg/kg 89Zr-REGN3767 were also able to detect LAG-3 positive T cells in the spleen. This study shows the ability of 89Zr-REGN3767 to successfully image LAG-3 expressed on intratumoral and splenic T lymphocytes. This work supports the clinical translation of anti-LAG-3 immuno-PET for the assessment of LAG-3 expression, with the goal to investigate its utility for predicting and monitoring response to checkpoint blockade therapy. Citation Format: Marcus P. Kelly, Richard Tavare, Jason T. Giurleo, Sosina Makonnen, Carlos Hickey, Makenzie A. Danton, T Cody Arnold, Dangshe Ma, Jie Dai, Jerry Pei, Jessica R. Kirshner, William C. Olson, Gavin. Thurston. Immuno-PET detection of LAG-3 expressing intratumoral lymphocytes using the zirconium-89 radiolabeled fully human anti-LAG-3 antibody REGN3767 [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 3033.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2018
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 79, No. 13_Supplement ( 2019-07-01), p. 1131-1131
    Abstract: Detection and quantification of immune cell subsets and changes in the tumor immune microenvironment would be a powerful tool to correlated to response in cancer immunotherapy. Currently, biopsies used to monitor the tumor microenvironment suffer from sampling error, restricted sampling over time and to inaccessible tissues/organs, and do not reflect discordance across tumor lesions of the same patient. Immuno-PET detection of CD8-expressing T cells is a promising translational imaging approach to assess CD8+ tumor-infiltrating lymphocytes (TILs) pre- and post-therapy. We have developed a fully human anti-CD8 antibody that was radiolabeled with the positron emitting radionuclide Zirconium-89 (89Zr) using the bifunctional chelator p-SCN-Bn-Desferoxamine (DFO). CD8-genetically humanized immunocompetent mice were used to validate dose dependent targeting of 89Zr-CD8 to lymphoid tissues. Next, two models were developed to monitor the response of the T cell activating bispecific antibody REGN1979 (CD3xCD20). In the first model designed to lack a CD8 antigen sink, Raji lymphoma cells were co-implanted with human peripheral blood mononuclear cells (hPBMCs) in immune deficient NSG mice and treated with REGN1979. Here, 89Zr-CD8 detected a two-fold increase in CD8+ TILs as quantified by flow cytometry compared to control antibody treated mice. To determine if 89Zr-CD8 could detect changes in CD8+ TILs in the presence of increased circulating and splenic T cells, a different model was developed. Raji lymphoma cells were implanted subcutaneously and hPBMCs were implanted intraperitoneally in immune deficient SRG-15 mice (hSIRP KI, Rag2 KO, IL2Rg KO, hIL-15 KI). Due to the humanization of IL-15, T cells from the implanted hPBMCs rapidly expanded and produced a CD8 antigen sink. In the SRG-15 model, total tumor uptake of 89Zr-CD8 was lower and spleen uptake higher than in the NSG model due to the presence of the antigen sink. These SGR-15 mice treated with REGN1979 also demonstrated an increase in CD8+ TILs by flow cytometry that was specifically detected with 89Zr-CD8. This work demonstrates that 89Zr-CD8 can specifically detect CD8+ T cells in lymphoid tissues of CD8 humanized mice and therapy-induced alterations of CD8+ TILs. Importantly, the detection of CD8+ TILs occurs in both the presence and absence of an antigen sink. This work supports the clinical translation of 89Zr-CD8 immuno-PET to investigate its utility for predicting and monitoring response in patients undergoing immunotherapy. Citation Format: Richard Tavare, Makenzie Danton, Jason T. Giurleo, Marcus P. Kelly, Sosina Makonnen, Carlos Hickey, Tomas C. Arnold, Dangshe Ma, William C. Olson, Gavin Thurston, Jessica R. Kirshner. CD8 immuno-PET using a fully human mAb can detect increases in tumor-infiltrating T cells by a T cell activating immunotherapy [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 1131.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2019
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    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...