12 results on '"Haley Neff-LaFord"'
Search Results
2. 438 Synergy between SEA-CD40 and chemotherapeutics drives curative antitumor activity in preclinical models
- Author
-
Michael Schmitt, Weiping Zeng, Celine Jacquemont, Haley Neff-LaFord, and Sahar Ansari
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2020
- Full Text
- View/download PDF
3. EP064/#192 Potential mechanism for ocular adverse events observed with tisotumab vedotin
- Author
-
Ignace Vergote, Stella Kim, Paul Ursell, Sandro Pignata, Antonio Gonzalez-Martin, Leslie Randall, Bohuslav Melichar, Kristine Madsen, Sven Mahner, Domenica Lorusso, Stéphanie Henry, Lydia Gaba, Theodora Salcedo, Haley Neff-Laford, Jeffrey Harris, Joy Jiang, Ibrahima Soumaoro, Shweta Jain, Bradley Monk, and Robert L Coleman
- Published
- 2022
4. 1357 Reversible chemical modulation of antibody effector function maintains anti-tumor activity while mitigating peripheral immune activation
- Author
-
Matthew Levengood, Chris Leiske, Noah Bindman, Xinqun Zhang, Nicole Duncan, Weiping Zeng, Serena Wo, Abbie Wong, Clark Henderson, Karalyne Crowder, Haley Neff-LaFord, Django Sussman, Shyra Gardai, and Philip Moquist
- Published
- 2022
5. Abstract 5668: Using clinical utility index (CUI) to determine the optimal biological dose of a nonfucosylated anti-TIGIT antibody: A proposed alternative to maximum tolerated dose (MTD)
- Author
-
Gabriela Patilea-Vrana, John Harrold, Joseph A. Ware, Shaparak Lonning, Hun Lee, Lisa Brooks, Haley Neff-LaFord, William D. Hanley, and Andres Forero-Torres
- Subjects
Cancer Research ,Oncology - Abstract
SEA-TGT is a human nonfucosylated monoclonal antibody (mAb) targeting the T cell immunoreceptor with immunoglobulin (Ig) and ITIM domains (TIGIT) protein. TIGIT is an immunoregulatory receptor expressed on activated and memory T cells, Tregs, and NK cells. TIGIT binding to CD155 and CD112 on tumor cells drives an inhibitory signal resulting in decreased T cell functionality. TIGIT targeting has been reported to release these inhibitory signals, drive Treg depletion, augment CD8+ T cell generation, and promote anti-tumor responses. SGNTGT-001 (NCT04254107) is a phase 1 clinical trial that evaluated the safety and tolerability of SEA-TGT as monotherapy in solid tumors and lymphomas at doses ranging from 0.01 to 6 mg/kg. Because an MTD was not identified in dose escalation, pharmacokinetic (PK) and pharmacodynamic (PD) endpoints were measured to assess biological activity and inform dose selection. The biological activity of SEA-TGT as monotherapy was compared across different dose levels using a clinical utility index (CUI), which mathematically integrated multiple clinically-meaningful PK and PD endpoints into a single readout. PD endpoints included NK and CD8+ T cell proliferation, maintenance of overall peripheral CD8+ T cell numbers, depletion of peripheral regulatory T cells, and peripheral target engagement. The PK endpoint was pharmacokinetic linearity. Surrogates of predicted tumor efficacy metrics included tumor target engagement and formation of the TIGIT:SEA-TGT:Fc receptor gamma (FcγRIIIa) trimer complexes in the tumor. An increase in the CUI score, representing an increase in biological activity, was observed during dose escalation, with an apparent plateau between the 0.3 and 6.0 mg/kg levels. Based on safety signals at 6 mg/kg and PK variability at lower doses, 1 and 3 mg/kg were of most interest for further evaluation. Both 1 and 3 mg/kg represented biologically active dose levels as they showed PK and PD activity that were within desirable ranges and had similarly high overall CUI scores relative to all doses evaluated. Dose selection for SEA-TGT was based on biological activity as assessed via PK/PD endpoints and integrated into a CUI model. Based on the totality of clinical data and the CUI results from monotherapy dose escalation from SGNTGT-001, the SEA-TGT dose of 1 mg/kg was determined to be the lowest biologically active dose with acceptable safety and tolerability. Citation Format: Gabriela Patilea-Vrana, John Harrold, Joseph A. Ware, Shaparak Lonning, Hun Lee, Lisa Brooks, Haley Neff-LaFord, William D. Hanley, Andres Forero-Torres. Using clinical utility index (CUI) to determine the optimal biological dose of a nonfucosylated anti-TIGIT antibody: A proposed alternative to maximum tolerated dose (MTD). [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 5668.
- Published
- 2023
6. 783 SGN-PDL1V, a novel, investigational PD-L1-directed antibody-drug conjugate for the treatment of solid tumors
- Author
-
Steven Jin, Shawna Hengel, Christina L. Zuch de Zafra, Byron Hua Kwan, Sean Allred, Andres Forero-Torres, Megan Ramirez, Haley Neff-LaFord, Changpu Yu, Serena Wo, Jessica K. Simmons, Heather Van Epps, Priyanka Gupta, and Kelly Hensley
- Subjects
Pharmacology ,Cancer Research ,Antibody-drug conjugate ,business.industry ,Immunology ,Enfortumab vedotin ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Immune checkpoint ,Polatuzumab vedotin ,chemistry.chemical_compound ,Oncology ,Monomethyl auristatin E ,chemistry ,Tolerability ,Cancer research ,Molecular Medicine ,Immunology and Allergy ,Immunogenic cell death ,Medicine ,business ,Brentuximab vedotin ,RC254-282 ,medicine.drug - Abstract
BackgroundPD-1/PD-L1 immune checkpoint inhibitors have transformed oncology, but a significant unmet need persists for patients with relapsed/refractory tumors following PD-1/PD-L1 treatment. PD-L1 is expressed in patients across a broad spectrum of tumor types and displays limited normal tissue expression, highlighting the potential of PD-L1 as a target for antibody-drug conjugates (ADCs) in addition to its role as an immune checkpoint. SGN-PDL1V is a PD-L1-directed ADC currently under preclinical investigation, which is comprised of an anti-PD-L1 antibody conjugated to the vedotin drug-linker. The vedotin drug-linker, consists of the microtubule disrupting agent, monomethyl auristatin E (MMAE), and a protease-cleavable peptide linker, which has been clinically validated in multiple ADC programs including brentuximab vedotin, enfortumab vedotin and polatuzumab vedotin.1–3 The proposed SGN-PDL1V primary mechanism of action is direct cytotoxicity against PD-L1-expressing malignant cells through delivery of the MMAE payload. Additionally, MMAE induces immunogenic cell death, leading to subsequent immune activation in the tumor microenvironment.4 Here, we characterize the preclinical activity and tolerability of SGN-PDL1V.MethodsSGN-PDL1V cytotoxicity was evaluated using PD-L1 expressing tumor cell lines in vitro and xenograft tumor models in vivo. Inhibition of the PD-1/PD-L1 immune checkpoint was assessed in a luminescent reporter system in vitro and a syngeneic tumor model in vivo. The tolerability and safety profile of SGN-PDL1V was determined in a non-human primate study.ResultsIn vitro, SGN-PDL1V demonstrated internalization and potent cytotoxic activity against PD-L1 expressing tumor cells. In vivo, SGN-PDL1V achieved tumor regressions in multiple tumor xenograft models at doses as low as 1 mg/kg when dosed weekly for a total of three doses. This activity was observed in immunocompromised mice, which lack responses to PD-1/PD-L1 inhibition. Notably, activity was observed even in xenograft models with low, heterogeneous PD-L1 expression, supporting the possibility to treat patients across a wide range of PD-L1 expression levels. Additionally, SGN-PDL1V exhibited potential to inhibit the PD-1/PD-L1 checkpoint in vitro and in vivo. The tolerability and safety profile of SGN-PDL1V were assessed in a non-human primate study and found to be comparable to other FDA-approved vedotin ADCs.ConclusionsSGN-PDL1V is a promising PD-L1 directed ADC with a unique cytotoxic mechanism of action among other PD-L1-targeted therapeutics. SGN-PDL1V demonstrated robust activity in multiple preclinical models and comparable tolerability and safety profile to other vedotin ADCs in non-human primates. Collectively, these data support further evaluation of SGN-PDL1V in a planned, first-in-human Phase 1 study.AcknowledgementsWe would like to thank Kerry Klussman for assay support and Jamie Mitchell for conjugation support.Trial RegistrationN/AReferencesSenter PD, Sievers EL. The discovery and development of brentuximab vedotin for use in relapsed Hodgkin lymphoma and systemic anaplastic large cell lymphoma. Nat Biotechnol 2012;30(7):631–7. Epub 2012/07/12. doi: 10.1038/nbt.2289. PubMed PMID: 22781692.Rosenberg JE, O'Donnell PH, Balar AV, McGregor BA, Heath EI, Yu EY, et al. Pivotal trial of enfortumab vedotin in urothelial carcinoma after platinum and anti-programmed death 1/Programmed death ligand 1 therapy. J Clin Oncol 2019;37(29):2592–600. Epub 2019/07/30. doi: 10.1200/JCO.19.01140. PubMed PMID: 31356140; PubMed Central PMCID: PMC6784850.Tilly H, Morschhauser F, Bartlett NL, Mehta A, Salles G, Haioun C, et al. Polatuzumab vedotin in combination with immunochemotherapy in patients with previously untreated diffuse large B-cell lymphoma: an open-label, non-randomised, phase 1b-2 study. Lancet Oncol 2019;20(7):998–1010. Epub 2019/05/19. doi: 10.1016/S1470-2045(19)30091-9. PubMed PMID: 31101489.Klussman K, Tenn E, Higgins S, Mazahreh R, Snead K, Hamilton J, Grogan B, Sigurjonsson J, Cao A, Gardai S, Liu B. 618 Vedotin ADCs induce ER stress and elicit hallmarks of ICD across multiple cancer indications. J Immunother Cancer 2020;8(Suppl 3):A372. DOI:10.1136/jitc-2020-SITC2020.0618.Ethics ApprovalAll animal studies were conducted in accordance with protocols reviewed and approved by the Institutional Animal Care and Use Committee at Seagen or the external testing facility that conducted the studies.
- Published
- 2021
7. Abstract 5535: SEA-CD40 is a non-fucosylated anti-CD40 antibody with potent pharmacodynamic activity in preclinical models and patients with advanced solid tumors
- Author
-
Olivier Rixe, Timothy M. Kuzel, David Smith, Martin Gutierrez, Sahar Ansari, John A. Thompson, Sanjay Goel, Andrew L. Coveler, Michael W. Schmitt, Thomas F. Gajewski, David L. Bajor, Haley Neff-LaFord, Shyra Gardai, Elisabeth I. Heath, Brendan D. Curti, Svetomir N. Markovic, Celine Jacquemont, and Juneko E. Grilley-Olson
- Subjects
0301 basic medicine ,Antibody-dependent cell-mediated cytotoxicity ,Cancer Research ,CD40 ,biology ,business.industry ,medicine.drug_class ,T cell ,Monoclonal antibody ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Cell killing ,Immune system ,medicine.anatomical_structure ,Oncology ,Cancer research ,biology.protein ,Medicine ,Antibody ,business ,Antigen-presenting cell ,030215 immunology - Abstract
CD40 is a co-stimulatory receptor of the TNF receptor superfamily expressed on antigen presenting cells (APCs). Antibodies targeting CD40 may have therapeutic benefit via multiple mechanisms including innate immune activation that can support generation of antigen-specific, antitumor T cell responses, and binding to CD40-expressing cancer cells leading to antibody-mediated target cell killing. Multiple CD40-directed antibodies are in clinical development and differ by immunoglobulin isotype, affinity to CD40, and selectivity for FcγR-binding. These alterations could lead to differences in pharmacodynamic and antitumor activity. SEA-CD40 is an agonistic non-fucosylated, humanized IgG1 monoclonal antibody directed against CD40. SEA-CD40 has enhanced FcγRIIIa binding (~10x greater than parent IgG1 antibody) that drives increased effector function, resulting in more potent immune stimulatory activity than antibodies with muted or selective FcγR binding. The enhanced effector function of SEA-CD40 may confer greater immune stimulation and antitumor activity relative to other CD40-directed therapeutics. Preclinically, SEA-CD40 exposure results in a distinct signature of responses including activation of APCs, CD8+ and CD4+ T cells and NK cells, and targeted depletion of CD40+ B cells. SEA-CD40 demonstrates superior activity compared to other CD40-targeted antibodies in vitro and in vivo, suggesting that the enhanced effector function is critical for optimal immune cell agonism. For example, SEA-CD40 drove in vitro ADCC activity 100-fold above the parent antibody and exhibited robust ADCC with the low and high affinity FcγRIIIA genotype. At matched dose levels in cynomolgus monkeys, SEA-CD40 induced circulating cytokines and sustained B cell depletion that were up to 50-fold above that induced with the parent antibody. The SEA-CD40 signature of activation translates to increased antitumor activity as a single agent and in combination with standard of care treatments in preclinical models, suggesting the potential for beneficial combination therapy in the clinic. The SEA-CD40 immune signature was confirmed by pharmacodynamic changes in an ongoing phase 1 clinical trial in patients with relapsed/refractory metastatic solid tumors (NCT02376699). SEA-CD40 treatment induced dose-dependent increases in circulating cytokines and chemokines associated with myeloid and lymphoid immune activation and trafficking. SEA-CD40 treatment also resulted in activation of CD4+ and CD8+ T cells and CD40-targeted B cell depletion in the periphery. These findings support continued clinical evaluation of SEA-CD40. The ongoing phase 1 clinical trial is actively enrolling and includes a cohort in pancreatic cancer assessing the combination of SEA-CD40, gemcitabine, nab-paclitaxel, and pembrolizumab. Citation Format: Haley Neff-LaFord, Juneko E. Grilley-Olson, David C. Smith, Brendan Curti, Sanjay Goel, Timothy M. Kuzel, Svetomir N. Markovic, Olivier Rixe, David L. Bajor, Thomas F. Gajewski, Martin Gutierrez, Elisabeth I. Heath, John Thompson, Sahar Ansari, Shyra Gardai, Celine Jacquemont, Michael Schmitt, Andrew L. Coveler. SEA-CD40 is a non-fucosylated anti-CD40 antibody with potent pharmacodynamic activity in preclinical models and patients with advanced solid tumors [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 5535.
- Published
- 2020
8. Abstract 70: Elucidating the roles of antibody pharmacokinetics and maleimide stability in the toxicology of antibody-drug conjugates
- Author
-
Wendi Schultz, Cindy Balasubramanian, Paul Pittmen, Shawna Hengel, Nagendra Chemuturi, Franciso Zapata, Jocelyn R. Setter, Russell J. Sanderson, Haley Neff-LaFord, and Robert P. Lyon
- Subjects
Drug ,Cancer Research ,Biodistribution ,media_common.quotation_subject ,Cmax ,Pharmacology ,Toxicology ,chemistry.chemical_compound ,Oncology ,Tolerability ,Pharmacokinetics ,chemistry ,In vivo ,Maleimide ,Conjugate ,media_common - Abstract
Antibody-drug conjugates (ADCs) continue to emerge as effective therapeutics in a variety of oncology indications, with two agents currently approved and many more in late-stage clinical trials. These ADCs employ drug-linkers that were developed many years ago, and are now recognized to have properties that may adversely impact the activity and toxicology of the ADCs prepared with them. Two such properties that are now well appreciated are the reversibility of maleimide-based drug conjugation, and the impact of drug conjugation on the pharmacokinetics of the ADC. We recently reported advances in drug-linker design that independently address both of these properties, resulting in the irreversible conjugation of drugs which have minimal impact on antibody pharmacokinetics, even at high levels of drug loading (Nature Biotechnology 32, 1059-1062 (2014), Nature Biotechnology 33, 733-735 (2015)). We have now prepared drug-linkers of monomethylauristatin E (MMAE) that orthogonally employ these features to enable a systematic evaluation of the relative contributions of maleimide instability and accelerated plasma clearance on the in vivo behavior of MMAE ADCs. Biodistribution studies with these molecules have revealed that the concentration of released MMAE in normal tissues is greatly impacted by the rate of ADC clearance (fast clearance results in greater Cmax of free drug), while stabilization of the maleimide has a relatively small effect. These differences in observed free drug concentrations were paralleled in tolerability studies, with ADC clearance rates exerting a greater impact on hematology parameters than maleimide stability. Collectively, these results suggest that ADC pharmacokinetics dominate the biodistribution and toxicology profiles for a given drug payload, with conjugate stability playing a relatively minor role. Citation Format: Haley Neff-LaFord, Franciso Zapata, Wendi Schultz, Cindy Balasubramanian, Paul Pittmen, Shawna Hengel, Russell Sanderson, Nagendra Chemuturi, Jocelyn Setter, Robert P. Lyon. Elucidating the roles of antibody pharmacokinetics and maleimide stability in the toxicology of antibody-drug conjugates [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 70. doi:10.1158/1538-7445.AM2017-70
- Published
- 2017
9. Reducing hydrophobicity of homogeneous antibody-drug conjugates improves pharmacokinetics and therapeutic index
- Author
-
Svetlana O. Doronina, Haley Neff-LaFord, Martha Anderson, Jocelyn R. Setter, Joshua H. Hunter, Mechthild Jonas, Robert P. Lyon, Tim D. Bovee, Patrick J. Burke, and Peter D. Senter
- Subjects
Drug ,Models, Molecular ,Immunoconjugates ,media_common.quotation_subject ,Chemistry, Pharmaceutical ,Biomedical Engineering ,Bioengineering ,Mice, SCID ,Pharmacology ,Applied Microbiology and Biotechnology ,Cell Line ,Mice ,Therapeutic index ,Pharmacokinetics ,In vivo ,parasitic diseases ,Potency ,Animals ,Humans ,media_common ,Chemistry ,fungi ,In vitro ,body regions ,Models, Chemical ,Pharmaceutical Preparations ,Drug delivery ,Molecular Medicine ,Hydrophobic and Hydrophilic Interactions ,Biotechnology ,Conjugate - Abstract
The in vitro potency of antibody-drug conjugates (ADCs) increases with the drug-to-antibody ratio (DAR); however, ADC plasma clearance also increases with DAR, reducing exposure and in vivo efficacy. Here we show that accelerated clearance arises from ADC hydrophobicity, which can be modulated through drug-linker design. We exemplify this using hydrophilic auristatin drug linkers and PEGylated ADCs that yield uniform, high-DAR ADCs with superior in vivo performance.
- Published
- 2014
10. Abstract 4994: SEA-CD40: from bench to bedside
- Author
-
Angela Epp, Thomas Manley, Haley Neff-LaFord, Shyra Gardai, Jing Yang, and Che-Leung Law
- Subjects
Cancer Research ,CD40 ,biology ,medicine.drug_class ,business.industry ,Monocyte ,medicine.medical_treatment ,CXCR3 ,Monoclonal antibody ,medicine.anatomical_structure ,Immune system ,Cytokine ,Oncology ,medicine ,biology.protein ,Cancer research ,Antibody ,business ,Receptor - Abstract
SEA-CD40 is a non-fucosylated, humanized IgG1 monoclonal antibody directed against human CD40, a co-stimulatory receptor of the TNF receptor superfamily. The consequence of enhanced SEA-CD40/FcγRIIIa binding is potent immune stimulatory activity. CD40 receptor ligation induces multiple pathways; to pave the road for identification of a specific activity signature in the clinical setting, in vitro preclinical assays were developed to monitor the immune modulatory activity of SEA-CD40. Human PBMCs stimulated with increasing concentrations of SEA-CD40 were assessed for immune changes including cytokine production, cellular activation, and modulation of cellular subsets. SEA-CD40 PBMC stimulation elicited a unique set of cytokines including MIP-1β, MCP-1, and IL-8. In addition to inducing cytokines, specific immune cell changes were also observed including up-regulation of stimulatory molecules on monocyte/ macrophages, activation of NK cells, and changes in cellular subsets such as deletion of B-cells. While some of these changes were common across the other CD40 therapeutic antibodies being tested in the clinic, SEA-CD40-specific changes were identified. These changes included a reduction in the immune dampening cytokine IL-10, induction of Th1 CXCR3 positive cells, and reduction of T-regulatory cells, all potentially contributing to an antitumor immune response. The specific in vitro SEA-CD40 signature was also observed in vivo in cynomolgus monkeys. SEA-CD40 treatment induced the same signature cytokines observed in vitro and elicited the same cellular changes including depletion of B-cells, and activation of CD8+ T-cells. A CD40 receptor occupancy assay was also created to correlate receptor engagement with activity. Interestingly, while SEA-CD40 is rapidly cleared from plasma, it is detectable on the surface of antigen-presenting cells for up to 3 weeks. The initial starting dose for SEA-CD40 clinical trials was calculated using the minimal anticipated biological effect level (MABEL). SEA-CD40 cytokine induction was the most sensitive preclinical marker of biologic activity and was, therefore, used for MABEL dose calculation. In the ongoing phase 1 First-In-Human clinical trial, this preclinical SEA-CD40 activity signature is being monitored in adult patients with advanced solid tumors (study NCT02376699). Establishing a clear immune biomarker strategy from pre-clinical research to clinical trials is vital for tracking the activity of our immuno-oncology drugs in patients and for identifying a safe and efficacious regimen. Citation Format: Shyra J. Gardai, Haley Neff-LaFord, Angela Epp, Jing Yang, Thomas Manley, Che-Leung Law. SEA-CD40: from bench to bedside. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 4994.
- Published
- 2016
11. Abstract 2472: SEA-CD40, a sugar engineered non-fucosylated anti-CD40 antibody with improved immune activating capabilities
- Author
-
Angela Epp, Lori Westendorf, Stanford L. Peng, Haley Neff-LaFord, May Kung Sutherland, Che-Leung Law, Germein Linares, and Shyra Gardai
- Subjects
Antibody-dependent cell-mediated cytotoxicity ,Cancer Research ,CD40 ,biology ,medicine.drug_class ,Dacetuzumab ,hemic and immune systems ,Monoclonal antibody ,Molecular biology ,Tumor antigen ,Oncology ,Antigen ,biology.protein ,medicine ,Antibody ,Antigen-presenting cell ,medicine.drug - Abstract
SEA-CD40 is a non-fucosylated, humanized IgG1 monoclonal antibody directed against human CD40, a co-stimulatory receptor of the TNF receptor superfamily. SEA-CD40 is derived from dacetuzumab, a humanized IgG1 previously developed and studied for B-lineage malignancies. Glycosylation of the antibody Fc is essential for Fc receptor-mediated activity and non-fucosylated antibodies show improved efficacy, particularly via increased binding to low affinity FcγRIIIa. Enhanced functionality of SEA-CD40 was determined through FcγRIIIa binding affinity, antibody-dependent cellular cytotoxicity (ADCC) activity, activation of the immune response, and induction of antigen-specific T-cells. While SEA-CD40 and the parent antibody dacetuzumab bind to CD40 with similar affinity, the non-fucosylated SEA-CD40 binds equally well to the low (158F) and high (158V) affinity versions of FcγRIIIa with higher affinity than dacetuzumab. The consequence of enhanced SEA-CD40/FcγRIIIa binding is potent ADCC activity against a CD40+ lymphoma B cell line and improved agonistic signaling to antigen presenting cells (APCs). SEA-CD40 treatment of human PBMCs elicits a robust immune response as measured by increased cytokine production and up-regulation of maturation markers on APCs with maintained activity at antibody concentrations as low as 10 ng/ml. The immune stimulatory properties of SEA-CD40 were observed in vivo as increased activity in xenograft and syngenic tumor models as well as induction of cytokine production in cynomolgus monkeys. Both in-vitro and in-vivo activity of SEA-CD40 was significantly greater than with dacetuzumab. The increased functionality of SEA-CD40 occurs through the non-fucosylated Fc domain as a F(ab’)2 version loses the ability to induce ADCC, stimulate cytokines, or up-regulate APC activation markers. SEA-CD40 induction of antigen specific T-cells was assessed using human peripheral blood mononuclear cells (PBMCs) exposed to influenza antigen. In the presence of SEA-CD40 influenza specific T-cells mount a robust antigen-specific response characterized by tetramer staining and elevated production of IFNγ. SEA-CD40 activity on PBMCs from donors with melanoma, pancreatic, or breast cancer was assessed and tumor antigen specific T-cell proliferation and IFNγ production was observed. SEA-CD40 is a non-fucosylated agonistic anti-CD40 antibody that shows enhanced binding to FcγRIIIa resulting in amplified cytokine production, co-stimulatory molecule up regulation, and ultimately stimulation of antigen specific T-cell responses to viral and tumor antigens. Citation Format: Shyra J. Gardai, Angela Epp, Germein Linares, Lori Westendorf, May Sutherland, Haley Neff-LaFord, Stanford L. Peng, Che-Leung Law. SEA-CD40, a sugar engineered non-fucosylated anti-CD40 antibody with improved immune activating capabilities. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 2472. doi:10.1158/1538-7445.AM2015-2472
- Published
- 2015
12. A sugar engineered non-fucosylated anti-CD40 antibody, SEA-CD40, with enhanced immune stimulatory activity alone and in combination with immune checkpoint inhibitors
- Author
-
Jonathan G. Drachman, Haley Neff-LaFord, Germein Linares, Shyra Gardai, Angela Epp, Loir Westendorf, Stanford L. Peng, Che-Leung Law, and May Kung Sutherland
- Subjects
Cancer Research ,CD40 ,medicine.drug_class ,Immune checkpoint inhibitors ,Dacetuzumab ,hemic and immune systems ,Biology ,Monoclonal antibody ,Immune system ,Oncology ,Immunology ,medicine ,biology.protein ,Anti cd40 ,Antibody ,medicine.drug - Abstract
3074 Background: SEA-CD40 is a non-fucosylated, humanized IgG1 monoclonal antibody directed against human CD40. It is derived from dacetuzumab, a humanized IgG1 previously developed for B-lineage m...
- Published
- 2015
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.