7 results on '"Katrina Bykova"'
Search Results
2. 787 Natural killer cell engagers activate innate and adaptive immunity and show synergy with proinflammatory cytokines
- Author
-
Jing Qi, Ke Liu, Christine Bonzon, Rumana Rashid, John Desjarlais, Rena Bahjat, Matthew Bernett, Juan Diaz, Kendra Avery, Katrina Bykova, Matthew Faber, Noor Siddiqi, and Dong Hyun Nam
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2021
- Full Text
- View/download PDF
3. 1067 Synergistic combination of natural killer cell engagers (NKEs) with proinflammatory cytokines
- Author
-
Tian Zhang, Matthew Faber, Erik Pong, Ke Liu, Matthew Bernett, Christine Bonzon, Juan Diaz, Dong Hyun Nam, Kendra Avery, Jing Qi, Rumana Rashid, Rena Bahjat, John Desjarlais, and Katrina Bykova
- Published
- 2022
- Full Text
- View/download PDF
4. 707 IL12 Fc-fusions engineered for reduced potency and extended half-life exhibit strong anti-tumor activity and improved therapeutic index compared to wild-type IL12 agents
- Author
-
Hanh Nho Nguyen, Umesh Muchhal, Katrina Bykova, Irene Leung, John R. Desjarlais, Rumana Rashid, Matthew J. Bernett, Ke Liu, Araz Eivazi, Christine Bonzon, Connie Ardila, Kendra N. Avery, Nicole Rodriguez, Norman J. Barlow, Nargess Hassanzadeh-Kiabi, Rajat Varma, and Michael Hackett
- Subjects
Pharmacology ,Antitumor activity ,Cancer Research ,Chemistry ,Immunology ,Wild type ,Half-life ,Therapeutic index ,Oncology ,Interleukin 12 ,Molecular Medicine ,Immunology and Allergy ,Potency - Abstract
BackgroundInterleukin-12 (IL12) is a proinflammatory cytokine that induces differentiation of Th1 cells and increased cytotoxicity of T and NK cells. Stimulation by IL12 leads to production of IFNγ and an inflammatory tumor microenvironment critical for anti-tumor responses. Studies in mice revealed IL12 can dramatically shrink syngeneic tumors, however human clinical studies resulted in severe toxicity and a small therapeutic window, limiting response rates. Prior work at Xencor demonstrated that reduced-potency IL15/IL15Rα-Fc fusion proteins exhibited superior therapeutic index (TI) in non-human primates (NHP) by reducing receptor-mediated clearance. Applying similar principles to IL12, we created IL12 heterodimeric Fc-fusions (IL12-Fc) with reduced potency to improve TI.MethodsIL12 is a heterodimer of two subunits, so we engineered IL12-Fc fusions by fusing the IL12p35 subunit to one side of a heterodimeric (and inactive) Fc domain, and IL12p40 to the other side. These Fc-fusions were tuned for optimal activity by introducing amino acid substitutions at putative receptor-interface positions and screening for reductions of in vitro potency. In vitro activity was assessed on human PBMCs by measuring signaling in a STAT4 phosphorylation assay and IFNγ production in a mixed-lymphocyte reaction (MLR). In vivo anti-tumor activity of human IL12-Fc was assessed in huPBMC-NSG-DKO and huCD34+ MCF7 xenograft models. Surrogate mouse potency-reduced IL12-Fc were evaluated in syngeneic tumor models. Tolerability and pharmacodynamic activity were assessed in NHP.ResultsAn IL12-Fc potency series was created, and variants had up to a 10,000-fold reduction in STAT4 signaling and IFNγ production in an MLR assay compared to wild-type IL12-Fc. Anti-tumor activity was achieved with potency-reduced IL12-Fc as single-agents and in combination with anti-PD1, with weaker variants maintaining anti-tumor activity at higher dose levels. Analysis of peripheral lymphocytes indicated increased numbers of T and NK cells as well as activation of CD8+ T cells. Increased expression of immune checkpoints including PD1 was also observed. Analysis of serum indicated up to 200-fold increases in IFNγ levels. Surrogate potency-reduced IL12-Fc had improved tolerability and greater selectivity of IFNγ production in tumors compared to spleen and less production of IL10 compared to wild-type IL12-Fc. In NHP, potency-reduced IL12-Fc had superior exposure with slower, more sustained accumulation of IFNγ and IP10, and a more gradual dose-dependent peak response, as well as more sustained margination of T and NK cells compared to wild-type IL12-Fc.ConclusionsPotency-reduced IL12-Fc retain strong anti-tumor activity, while potentially overcoming safety and tolerability issues related to narrow TI associated with wild-type IL12 or IL12-Fc agents.
- Published
- 2021
- Full Text
- View/download PDF
5. 787 Natural killer cell engagers activate innate and adaptive immunity and show synergy with proinflammatory cytokines
- Author
-
Rena Bahjat, Rumana Rashid, Dong Hyun Nam, Matthew S. Faber, Kendra N. Avery, John R. Desjarlais, Juan Diaz, Ke Liu, Katrina Bykova, Noor Siddiqi, Jing Qi, Matthew J. Bernett, and Christine Bonzon
- Subjects
Pharmacology ,Cancer Research ,Immunology ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Biology ,Acquired immune system ,Natural killer cell ,Proinflammatory cytokine ,medicine.anatomical_structure ,Oncology ,medicine ,Molecular Medicine ,Immunology and Allergy ,RC254-282 - Abstract
BackgroundNatural Killer cell Engagers (NKEs) are multifunctional molecules that target activating receptors on the surface of NK cells, bind to tumor associated antigens and engage Fc gamma receptors expressed on effector cells of the immune system. NKEs promote tumor cell lysis by redirecting NK cells to their targets, and drive activation and proliferation of NK cells. Engagement of NK cells, an effector cell population of the innate immune system, provides an opportunity to target cancers with reduced expression of MHC molecules that are less responsive to therapies targeting the adaptive immune system. Therefore, NKEs have a potential to provide an additional treatment option to patients who respond poorly to T cell tailored immunotherapies.MethodsExpanding on Xencor's XmAb bispecific Fc platform, we developed NKE molecules targeting NKG2D, an activating receptor expressed on cytotoxic immune cells, B7H3, a pan tumor antigen, while simultaneously engaging Fc gamma receptors. Functional activity of NKEs was evaluated via assessing anti-tumor cytotoxicity and activation of NK and T cells in co-culture studies with human cancer cell lines.ResultsNKEs were engineered for synergistic effects on NK cells by the simultaneous engagement of NKG2D and Fc gamma receptors. Additionally, the NKG2D variable domains were selected for their ability to provide a co-stimulatory signal to T cells in the presence of TCR-mediated signaling. Developed NKEs showed cytotoxic activity and immune cell activation in co-culture studies of human cancer cell lines with either PBMCs, T cells or NK cells. Combination of NKEs with proinflammatory cytokines, such as IL15, showed enhancement of the cytotoxic activity against tumor cells and augmented NK cell activation.ConclusionsXmAb bispecific NKEs engineered to engage innate and adaptive immunity show encouraging tumor cell killing activity and synergistic cytotoxicity in combination with proinflammatory cytokines. These data have identified several promising candidate NKEs for future in vivo efficacy studies in mouse tumor models expressing B7H3.
- Published
- 2021
- Full Text
- View/download PDF
6. Abstract 1743: IL12 heterodimeric Fc-fusions engineered for reduced potency exhibit strong anti-tumor activity and improved therapeutic index compared to native IL12 agents
- Author
-
Rumana Rashid, Nargess Hassanzadeh-Kiabi, Irene Leung, Norm Barlow, Nicole Rodriguez, Araz Eivazi, Michael Hackett, Duc-Hanh T. Nguyen, Connie Ardila, Rajat Varma, Matthew J. Bernett, Seung Y. Chu, Christine Bonzon, John R. Desjarlais, Ke Liu, Umesh Muchhal, and Katrina Bykova
- Subjects
Antitumor activity ,Cancer Research ,Therapeutic index ,Oncology ,Chemistry ,Interleukin 12 ,Potency ,Pharmacology - Abstract
Interleukin-12 (IL12) is a proinflammatory cytokine produced by activated antigen-presenting cells that induces differentiation of Th1 cells and increased proliferation and cytotoxicity of T and NK cells. Stimulation of these cells by IL12 leads to production of high levels of IFNγ. These immune-stimulating aspects of IL12 may help to establish an inflammatory tumor microenvironment critical for anti-tumor responses. Preclinical studies in mice revealed that native IL12 can dramatically shrink syngeneic tumors, however clinical studies in humans resulted in severe toxicity and a small therapeutic window, limiting response rates. Prior work at Xencor demonstrated that reduced-potency IL15/IL15Rα-Fc fusion proteins exhibited superior pharmacokinetics, pharmacodynamics, and safety in non-human primates through reduction of receptor-mediated clearance. Applying similar principles to IL12, we created IL12 heterodimeric Fc-fusions (IL12-Fc) with reduced potency to improve tolerability, slow receptor-mediated clearance, and extend half-life. IL12 is a heterodimeric protein consisting of two subunits, so we engineered IL12-Fc fusions by fusing the IL12p35 subunit to one side of a heterodimeric (and inactive) Fc domain, and the IL12p40 subunit to the other side. These Fc-fusions were tuned for optimal activity by introducing amino acid substitutions at putative receptor-interface positions and screening for reductions of in vitro potency. In vitro activity was assessed on human PBMCs by measuring signaling in a STAT4 phosphorylation assay and IFNγ production in a mixed-lymphocyte reaction (MLR). In vivo anti-tumor activity of human IL12-Fc were assessed in a human PBMC engrafted mouse MCF7 tumor model. Surrogate mouse IL12-Fc were evaluated in additional murine tumor models. Tolerability and pharmacodynamic activity were assessed in non-human primates. IL12-Fc were produced with good yield and purity. An IL12-Fc potency series was created, and variants had up to a 10,000-fold reduction in STAT4 signaling potency and IFNγ production in an MLR assay compared to native IL12-Fc. Anti-tumor activity was achieved with potency-reduced IL12-Fc as a single-agent and in combination with anti-PD1, with weaker variants maintaining anti-tumor activity at higher dose levels. Analysis of peripheral lymphocytes indicated increased numbers of T and NK cells as well as activation of CD8+ T cells. Increased expression of immune checkpoints including PD1 was also observed. Analysis of serum indicated up to 200-fold increases in IFNγ levels. Combined, these data indicate that potency-reduced IL12-Fc retain strong anti-tumor activity, while potentially overcoming safety and tolerability issues related to narrow therapeutic index associated with recombinant native IL12 or IL12-Fc agents. Citation Format: Matthew J. Bernett, Ke Liu, Christine Bonzon, Rumana Rashid, Nicole Rodriguez, Nargess Hassanzadeh-Kiabi, Connie Ardila, Katrina Bykova, Michael Hackett, Norm Barlow, Irene Leung, Duc-Hanh Nguyen, Araz Eivazi, Seung Y. Chu, Rajat Varma, Umesh S. Muchhal, John R. Desjarlais. IL12 heterodimeric Fc-fusions engineered for reduced potency exhibit strong anti-tumor activity and improved therapeutic index compared to native IL12 agents [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr 1743.
- Published
- 2021
- Full Text
- View/download PDF
7. Abstract 1831: Affinity tuned XmAb®2+1 GPC3 x CD3 bispecific antibodies demonstrate selective activity in liver cancer models
- Author
-
Umesh Muchhal, Seung Y. Chu, Juan Diaz, Rumana Rashid, John R. Desjarlais, Nargess Hassanzadeh-Kiabi, Katrina Bykova, Alex Nisthal, Kendra N. Avery, and Gregory L. Moore
- Subjects
Cancer Research ,Bispecific antibody ,Oncology ,biology ,Chemistry ,CD3 ,biology.protein ,Cancer research ,medicine ,Liver cancer ,medicine.disease - Abstract
Bispecific T cell engagers simultaneously bind CD3 on T cells and tumor-associated antigens to promote T cell-mediated killing of tumor cells. These agents provide synthetic immunity by expanding, activating, and redirecting T cells against a target of interest. While targeting lineage-restricted antigens such as CD19 or CD20 have found clinical success in hematopoietic cancers, targeting solid tumors requires high expressing tumor associated antigens (TAAs) with minimal normal tissue expression. Glypican 3 (GPC3), a lipid-anchored cell surface protein, is over-expressed in the majority of hepatocellular carcinoma (HCC) and a smaller subset of lung squamous cell carcinoma. It is considered an ideal target because its expression is not detected in adult tissues, and its function can promote tumor growth through the Wnt/beta-catenin pathway. Although GPC3 demonstrates good differential expression, bispecific T cell engagers are powerful immunomodulatory agents and careful tuning can improve the therapeutic window on all targets. Building upon the XmAb® heterodimeric Fc platform, we generated bispecific antibodies in an XmAb 2+1 Fab2-scFv-Fc format that are bivalent for GPC3 and monovalent for CD3. Reducing the affinity of GPC3 encouraged avid binding and strong killing activity only on high GPC3 expressing cell lines while minimizing reactivity on low expressing cell lines. We found modulating the CD3 affinity, either directly through mutation or indirectly by the molecular format, contributes to the selectivity of the bispecific antibodies. To ensure biologically valid antigen densities were being considered, IHC was conducted on paraffin embedded arrays of tumor tissue, normal tissue, and cell lines. Upon matching the staining intensity between the three sample types, we identified cell lines with corresponding GPC3 antigen density that could serve as proxies of tumor and normal tissue. In vitro T cell-dependent cellular cytotoxicity (TDCC) assays on the relevant cell lines confirmed selective potency on high versus low expressing cells. Citation Format: Alex Nisthal, Nargess Hassanzadeh-Kiabi, Kendra N. Avery, Rumana Rashid, Juan E. Diaz, Umesh S. Muchhal, Seung Y. Chu, Gregory L. Moore, Katrina Bykova, John R. Desjarlais. Affinity tuned XmAb®2+1 GPC3 x CD3 bispecific antibodies demonstrate selective activity in liver cancer models [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr 1831.
- Published
- 2021
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.