10 results on '"Pallavur Sivakumar"'
Search Results
2. The IL-20 Cytokine Family in Rheumatoid Arthritis and Spondyloarthritis
- Author
-
Tue W. Kragstrup, Thomas Andersen, Line D. Heftdal, Malene Hvid, Jens Gerwien, Pallavur Sivakumar, Peter C. Taylor, Ladislav Senolt, and Bent Deleuran
- Subjects
cytokine ,rheumatoid arthritis ,spondyloarthritis ,interleukin ,IL-10 family ,fibroblast ,Immunologic diseases. Allergy ,RC581-607 - Abstract
This review describes the IL-20 family of cytokines in rheumatoid arthritis (RA) and spondyloartrhitits (SpA) including psoriatic arthritis. The IL-20 receptor (R) cytokines IL-19, IL-20, and IL-24 are produced in both the peripheral blood and the synovial joint and are induced by Toll-like receptor ligands and autoantibody-associated immune complexes in monocytes. IL-19 seems to have anti-inflammatory functions in arthritis. In contrast, IL-20 and IL-24 increase the production of proinflammatory molecules such as monocyte chemoattractant protein 1 and are associated with bone degradation and radiographic progression. IL-22 is also associated with progression of bone erosions. This suggests that the IL-22RA1 subunit shared by IL-20, IL-22, and IL-24 is important for bone homeostasis. In line with this, the IL-22RA1 has been found on preosteoclasts in early RA. IL-26 is produced in high amounts by myofibroblasts and IL-26 stimulation of monocytes is an important inducer of Th17 cells in RA. This indicates a role for IL-26 as an important factor in the interactions between resident synovial cells and infiltrating leukocytes. Clinical trials that investigate inhibitors of IL-20 (fletikumab) and IL-22 (fezakinumab) in psoriasis and RA have been terminated. Instead, it seems that the strategy for modulating the IL-20 cytokine family should take the overlap in cellular sources and effector mechanisms into account. The redundancy encourages inhibition of more than one cytokine or one of the shared receptors. All IL-20 family members utilize the Janus kinase signaling pathway and are therefore potentially inhibited by drugs targeting these enzymes. Effects and adverse effects in ongoing clinical trials with inhibitors of IL-22 and the IL-22RA1 subunit and recombinant IL-22 fusion proteins will possibly provide important information about the IL-20 subfamily of cytokines in the future.
- Published
- 2018
- Full Text
- View/download PDF
3. Promise & challenges in discovering cellular immunotherapies &T cell engagers for solid tumor indications
- Author
-
Pallavur Sivakumar
- Published
- 2022
4. Discovery and Preclinical Characterization of CC-95251, an Anti-SIRPα Antibody That Enhances Macrophage-Mediated Phagocytosis of Non-Hodgkin Lymphoma (NHL) Cells When Combined with Rituximab
- Author
-
Mahan Abbasian, Konstantinos Mavrommatis, David Mikolon, Pallavur Sivakumar, Gustavo Fenalti, Roberto Guzman, Trout Christina, Preston Adams, Kandasamy Hariharan, Lawrence Dearth, Chan Henry, and Brian Fox
- Subjects
biology ,business.industry ,Phagocytosis ,Immunology ,Cell Biology ,Hematology ,Biochemistry ,biology.protein ,Cancer research ,Macrophage ,Medicine ,Hodgkin lymphoma ,Rituximab ,Antibody ,business ,medicine.drug - Abstract
Introduction: The transmembrane protein CD47 is ubiquitously expressed on normal cells and often overexpressed on cancer cells, including NHL. Binding of CD47 to signal regulatory protein-α (SIRPα) on macrophages induces an anti-phagocytic signal enabling tumor cells to escape the innate immune response. Macrophage antitumor activity can be restored by simultaneously blocking the CD47-SIRPα signaling axis and inducing a pro-phagocytic signal with the use of tumor-opsonizing antibodies. Targeting SIRPα on monocytes and macrophages rather than the ubiquitously expressed CD47 may overcome some toxicities associated with anti-CD47 therapies. Here, we report the discovery and characterization of a fully human anti-SIRPα antibody and its preclinical activity in combination with the opsonizing antibody rituximab in CD20+ diffuse large B-cell lymphoma (DLBCL) cell lines. Methods: A total of ~ 10 10 fully human immunoglobulin G antibodies were screened for binding to the extracellular domain of recombinant human SIRPα using a yeast display platform. Surface plasmon resonance was used to determine CC-95251 binding coverage across SIRPα haplotypes. The ability of CC-95251 to block CD47-SIRPα interaction was measured using Octet ® and Biacore™ assays. We determined the crystal structure of SIRPα in complex with the CC-95251 Fab to characterize its epitope and to define the structural basis for CD47-SIRPα interaction blockade. To identify tumor types likely susceptible to CD47-SIRPα axis disruption, expression levels of CD47-SIRPα and CD163 were assessed in bulk tumor samples using The Cancer Genome Atlas (TCGA) data. The antitumor effects of CC-95251 in combination with rituximab were examined by measuring the percentage of phagocytic macrophages in co-culture experiments of differentiated macrophages and CD20+ DLBCL cell lines (OCI-Ly3, RIVA, Pfeiffer, and Karpas 422). To confirm CC-95251 binding to monocytes, immunophenotyping of peripheral blood mononuclear cells from healthy donors and cynomolgus monkeys was performed using multiparameter flow cytometry. Lastly, pharmacokinetics and hematologic effects were analyzed in cynomolgus monkeys after treatment with 10, 30, or 100 mg/kg CC-95251. Results: Initial screening by yeast display yielded ~ 350 candidates. The top 24 clones were characterized fully and CC-95251 was selected as the lead monoclonal antibody exhibiting high binding affinity across the 6 most prevalent SIRPα human haplotypes. CC-95251 potently blocked CD47-SIRPα binding in a dose-dependent manner, with a concentration of 100 nM inhibiting CD47 binding almost completely. Co-crystallization modeling showed that CC-95251 engages SIRPα in a region overlapping the CD47 binding site, demonstrating a mechanism for CD47-SIRPα blockade. DLBCL was identified as a suitable tumor type for CC-95251 treatment based on CD47-SIRPα expression and macrophage infiltration. Co-culture experiments of donor macrophages and several DLBCL cell lines showed that CC-95251 monotherapy had weak-to-moderate antitumor activity. However, when combined with rituximab, the levels of phagocytic macrophages were markedly increased in a CC-95251 dose-dependent manner, suggesting that inhibition of the CD47-SIRPα anti-phagocytic axis with CC-95251 and activation of pro-phagocytic signaling with rituximab provide an enhanced antitumor effect in DLBCL cell lines. CC-95251 predominantly bound to cells of myeloid origin, including monocytes and, to a lesser extent, myeloid dendritic cells, whereas no binding to natural killer cells was observed. Toxicology studies in cynomolgus monkeys showed safe intravenous delivery of CC-95251 at therapeutic doses, with no evidence of white blood cell, monocyte, lymphocyte, or red blood cell depletion. Conclusions: CC-95251 is a novel, high-affinity, fully human monoclonal anti-SIRPα antibody that blocks the binding of CD47 to SIRPα. When combined with the therapeutic opsonizing antibody rituximab, CC-95251 enhances macrophage phagocytic activity against DLBCL cell lines in co-culture models. These results support the clinical evaluation of CC-95251 + rituximab for relapsed or refractory NHL. A phase 1 dose-escalation and -expansion study of CC-95251 for the treatment of advanced solid and hematologic malignancies is underway (NCT03783403). Disclosures Chan: Bristol Myers Squibb: Current Employment. Trout: Bristol Myers Squibb: Ended employment in the past 24 months. Mikolon: Bristol Myers Squibb: Current Employment. Adams: Bristol Myers Squibb: Current Employment. Guzman: Bristol Myers Squibb: Current Employment, Current equity holder in publicly-traded company. Fenalti: Bristol Myers Squibb: Current Employment. Mavrommatis: Bristol Myers Squibb: Current Employment, Current equity holder in publicly-traded company. Abbasian: Bristol Myers Squibb: Current equity holder in publicly-traded company, Ended employment in the past 24 months. Dearth: Bristol Myers Squibb: Current Employment. Fox: Bristol Myers Squibb: Current Employment, Current equity holder in publicly-traded company. Sivakumar: Bristol Myers Squibb: Current Employment, Current equity holder in publicly-traded company. Hariharan: Bristol Myers Squibb: Current Employment.
- Published
- 2021
5. 200 TGFβ-armoring boosts potency and persistence of engineered TCR T cells, unlocking superior efficacy against HPV-positive solid tumors
- Author
-
Stephanie Busch, Jianguo Huang, Ruth A. Salmon, Gail Turner, Teresa M. Foy, Cédric Cleyrat, Yeonjoo Oh, Pallavur Sivakumar, Jenna Bailey, Andreia Costa, Cyr De Imus, and Gabriela Diaz
- Subjects
Pharmacology ,Cancer Research ,Tumor microenvironment ,Adoptive cell transfer ,T cell ,Immunology ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Biology ,Chimeric antigen receptor ,medicine.anatomical_structure ,Oncology ,Antigen ,NSG mouse ,medicine ,Cancer research ,Molecular Medicine ,Immunology and Allergy ,Cytotoxic T cell ,RC254-282 ,CD8 - Abstract
BackgroundAdoptive transfer of chimeric antigen receptor (CAR)-expressing T cells targeting cell surface antigens has shown remarkable success in hematological malignancies. However, only limited success has been achieved to date with CAR T cells, or their engineered T cell receptor (eTCR) counterparts, in the context of solid tumors. This is largely due to: 1) challenges in identifying highly expressed, tumor-specific antigens and; 2) the immune-suppressive tumor microenvironment mediated by cellular and secreted factors such as TGFβ, known to suppress intra-tumoral immunity and notably elevated in many human cancers, including in human papilloma virus (HPV)-associated cancers (e.g. head and neck squamous cell carcinoma and cervical cancers).Here, we describe the generation of highly potent, TGFβ-armored, engineered T cells expressing a novel fully human, natural TCRαβ sequence that is HLA-A*02:01-restricted, CD8 coreceptor-independent and targets the tumor-restricted HPV-16 E7(11–19) onco-peptide.MethodsDonor-derived T cells were genetically engineered using high efficiency CRISPR-Cas9 editing as follows: 1) TRAC domain knock-out (KO) to prevent endogenous TCR expression; 2) knock-in of an HPV-specific eTCR at the TRAC locus; and 3) KO of TGFBR2 to prevent TGFβ signaling. Functional evaluation of edited T cells was performed in vitro using 3D serial spheroid stimulation as well as in vivo using NSG mouse tumor xenografts and against two cancer lines, SCC-152 and CasKi.ResultsUnder chronic antigen stimulation and in the presence of high TGFβ at optimal effector-to-target (E:T) ratio, HPV eTCR WT (control) and HPV eTCR TGFBR2 KO cells demonstrated robust and comparable cytotoxic functions in vitro. However, when tested at suboptimal E:T ratio, HPV eTCR TGFBR2 KO cells demonstrated superior expansion (>5-fold difference), cytotoxicity and an improved functional phenotype, suggesting that TGFβ-Armoring may decouple T cell expansion and the onset of exhaustion. In vivo studies demonstrated significant inhibition of tumor growth (p ConclusionsPharmacology studies demonstrate that the HPV eTCR armoring strategy aimed at overcoming TGFβ-mediated immune-suppression is highly effective in suboptimal conditions. Additionally, TGFβ-armored eTCR cells presented with improved pharmacodynamic and phenotypic characteristics, paving the way for effective clinical applications in solid tumors.AcknowledgementsRibonucleoprotein complexes designed specifically for the editing of human TRAC and TGFBR2 loci were provided by Editas Medicine.
- Published
- 2021
6. Integrative network modeling reveals mechanisms underlying T cell exhaustion
- Author
-
Andrew Dervan, Matthew Trotter, Christopher Mark Hill, Brian Fox, Hamid Bolouri, Alexander V. Ratushny, Anne-Renee van der Vuurst de Vries, Joshua Beilke, Pallavur Sivakumar, Paul Shannon, Rebecca Johnson, Mary Young, Lu Huang, Douglas Bassett, and C.C. Santini
- Subjects
0301 basic medicine ,Computer science ,T cell ,Datasets as Topic ,lcsh:Medicine ,CD8-Positive T-Lymphocytes ,Lymphocyte Activation ,Network topology ,Article ,Mice ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Animals ,Humans ,Computer Simulation ,Enhancer of Zeste Homolog 2 Protein ,Gene Regulatory Networks ,RNA-Seq ,lcsh:Science ,Oligonucleotide Array Sequence Analysis ,Network model ,Multidisciplinary ,Node (networking) ,lcsh:R ,Models, Immunological ,Translational research ,030104 developmental biology ,medicine.anatomical_structure ,Preclinical research ,030220 oncology & carcinogenesis ,lcsh:Q ,Immunologic Memory ,Neuroscience ,Signal Transduction - Abstract
Failure to clear antigens causes CD8+ T cells to become increasingly hypo-functional, a state known as exhaustion. We combined manually extracted information from published literature with gene expression data from diverse model systems to infer a set of molecular regulatory interactions that underpin exhaustion. Topological analysis and simulation modeling of the network suggests CD8+ T cells undergo 2 major transitions in state following stimulation. The time cells spend in the earlier pro-memory/proliferative (PP) state is a fixed and inherent property of the network structure. Transition to the second state is necessary for exhaustion. Combining insights from network topology analysis and simulation modeling, we predict the extent to which each node in our network drives cells towards an exhausted state. We demonstrate the utility of our approach by experimentally testing the prediction that drug-induced interference with EZH2 function increases the proportion of pro-memory/proliferative cells in the early days post-activation.
- Published
- 2019
- Full Text
- View/download PDF
7. Abstract 2696: Genetic and pharmacologic evaluation of the ubiquitin ligase CBL-B as a small-molecule, tumor immunotherapy target
- Author
-
Austin Tenn-McClellan, Hiroko Tanaka, Joseph Juan, Christoph W. Zapf, Katherine Kurylo, Pallavur Sivakumar, Anne-Renee van der Vuurst de Vries, Ryan Rountree, Andria Christianson, Jennifer McKinnell, Ketki Dhamnaskar, Christopher Karim, Thomas Cummins, Frederick Cohen, Paul A. Barsanti, May Tan, Jilliane Bruffey, Asad M. Taherbhoy, Mario G. Cardozo, Chenbo Wang, Szerenke Kiss von Soly, Julie Sheung, Dahlia Weiss, Mark Gallop, Anjanabha Saha, Neil F. Bence, Dane Karr, Jennifa Gosling, and Kathleen A. Boyle
- Subjects
Cancer Research ,biology ,Chemistry ,CD3 ,T cell ,CD28 ,Molecular biology ,Ubiquitin ligase ,medicine.anatomical_structure ,Oncology ,biology.protein ,medicine ,Cytokine secretion ,Ligase activity ,IL-2 receptor ,CD8 - Abstract
E3 ubiquitin ligases play critical roles in directing cellular protein fate by controlling the specificity of ubiquitin conjugation to substrate proteins and targeting them for cellular relocalization or degradation by the ubiquitin proteasome system. The E3 ubiquitin ligase CBL-B is expressed in immune cell lineages and negatively regulates activity of the T-cell receptor (TCR) by imposing a requirement for a costimulatory signal to mount a productive immune response upon TCR engagement. Mice deficient in Cbl-b, and more specifically in the RING Zn-finger ligase domain of Cbl-b, demonstrate a tumor rejection phenotype mediated by CD8+ T cells (Paolino et al., JI, 2011). We have reproduced these results and demonstrate that Cbl-b deficient mice show enhanced anti-tumor activity. In addition, we show that CD4+ and CD8+ T cells from mice deficient in Cbl-b have 5 to 10-fold enhanced secretion of IL-2 and IFN γ when stimulated ex vivo. These data provide a genetic rationale for the development of a small molecule inhibitor of CBL-B ligase activity for use in patients with tumor-mediated immune suppression of effector T cells. We have identified a series of small molecule inhibitors of CBL-B activity with biochemical potency at low nanomolar concentrations. CBL-B inhibitors increased cytokine secretion in vitro at low nanomolar concentrations, as measured by IL-2 and IFN γ secretion, in primary human and mouse T cells stimulated with CD3/CD28 or CD3 alone. The compounds also stimulated proliferation and elevated levels of the T cell surface activation markers CD25 and CD69. CBL-B inhibitors enhanced an antigen recall response in human PBMCs ex vivo, as measured by approximately 5-fold higher secretion of GM-CSF, TNF-α and RANTES, and demonstrated effects in an ex vivo model of exhausted T cell function. Oral dosing of an optimized CBL-B inhibitor enhanced anti-CD3 stimulated T cell activation in mouse CD4+ and CD8+ T cells, demonstrating a dose proportional pharmacodynamic effect. Oral administration over 28 days in the syngeneic CT-26 tumor model was well tolerated and resulted in single agent tumor growth inhibition. These data support the continued advancement of small molecule oral CBL-B inhibitors for future development in immuno-oncology. Citation Format: Jennifa Gosling, Ryan Rountree, Asad Taherbhoy, Chenbo Wang, Thomas Cummins, Frederick Cohen, Hiroko Tanaka, Dahlia Weiss, Mario Cardozo, Christopher Karim, May Tan, Joseph Juan, Austin Tenn-McClellan, Szerenke Kiss von Soly, Julie Sheung, Kathleen Boyle, Ketki Dhamnaskar, Katherine Kurylo, Jilliane Bruffey, Jennifer McKinnell, Dane Karr, Andria Christianson, Anne-Renee Van Der Vuurst de Vries, Pallavur Sivakumar, Mark Gallop, Paul A. Barsanti, Anjanabha Saha, Neil F. Bence, Christoph W. Zapf. Genetic and pharmacologic evaluation of the ubiquitin ligase CBL-B as a small-molecule, tumor immunotherapy target [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 2696.
- Published
- 2019
8. IL-15: Insights from Characterizing IL-15-Deficient Mice
- Author
-
Mary K. Kennedy, Sandra N. Brown, Anne Renee Van der Vuurst de Vries, Pallavur Sivakumar, Ananda W. Goldrath, and Joanne L. Viney
- Subjects
Interleukin 15 ,Deficient mouse ,Molecular biology - Published
- 2003
9. Interleukin-21 Enhances Rituximab Activity in a Cynomolgus Monkey Model of B Cell Depletion and in Mouse B Cell Lymphoma Models
- Author
-
Wayne R. Kindsvogel, Kim Waggie, Lay Chin, Felecia Wagener, Ken M. Bannink, Don Foster, Rick D. Holly, Pallavur Sivakumar, Julie Hill, Rafael Ponce, Rebecca Johnson, Richard Roque, Fariba Barahmand-pour, Steven D. Hughes, Hong-Ping Ren, Chris Clegg, Jane Heffernan, Faith Shiota, Cecile M. Krejsa, Mark Heipel, and Katherine Henderson
- Subjects
Male ,lcsh:Medicine ,Antibodies, Monoclonal, Murine-Derived ,Mice ,Interleukin 21 ,Drug Metabolism ,immune system diseases ,hemic and lymphatic diseases ,lcsh:Science ,B-cell lymphoma ,Immune Response ,Drug Distribution ,CD20 ,Antibody-dependent cell-mediated cytotoxicity ,B-Lymphocytes ,Multidisciplinary ,biology ,Drug Synergism ,hemic and immune systems ,medicine.anatomical_structure ,Oncology ,Medicine ,Female ,Rituximab ,Antibody ,Veterinary Pathology ,Research Article ,medicine.drug ,Drugs and Devices ,Lymphoma, B-Cell ,Drug Research and Development ,Immunology ,Antineoplastic Agents ,chemical and pharmacologic phenomena ,Immunopathology ,Immunomodulation ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Pharmacokinetics ,Biology ,B cell ,Interleukins ,lcsh:R ,Antibody-Dependent Cell Cytotoxicity ,medicine.disease ,Survival Analysis ,Immunity, Innate ,Lymphoma ,Disease Models, Animal ,Macaca fascicularis ,Pharmacodynamics ,biology.protein ,Veterinary Science ,lcsh:Q - Abstract
Rituximab, a monoclonal antibody targeting CD20 on B cells, is currently used to treat many subtypes of B cell lymphomas. However, treatment is not curative and response rates are variable. Recombinant interleukin-21 (rIL-21) is a cytokine that enhances immune effector function and affects both primary and transformed B cell differentiation. We hypothesized that the combination of rIL-21 plus rituximab would be a more efficacious treatment for B cell malignancies than rituximab alone. We cultured human and cynomolgus monkey NK cells with rIL-21 and found that their activity was increased and proteins associated with antibody dependent cytotoxicity were up-regulated. Studies in cynomolgus monkeys modeled the effects of rIL-21 on rituximab activity against CD20 B cells. In these studies, rIL-21 activated innate immune effectors, increased ADCC and mobilized B cells into peripheral blood. When rIL-21 was combined with rituximab, deeper and more durable B cell depletion was observed. In another series of experiments, IL-21 was shown to have direct antiproliferative activity against a subset of human lymphoma cell lines, and combination of murine IL-21 with rituximab yielded significant survival benefits over either agent alone in xenogeneic mouse tumor models of disseminated lymphoma. Therefore, our results do suggest that the therapeutic efficacy of rituximab may be improved when used in combination with rIL-21.
- Published
- 2013
10. Interleukin-21 Elicits Durable T and NK Cytotoxicity: Basic Biology to Clinical Trials
- Author
-
Pallavur Sivakumar
- Subjects
Pharmacology ,Clinical trial ,Cancer Research ,Interleukin 21 ,business.industry ,Immunology ,Immunology and Allergy ,Medicine ,business ,Cytotoxicity - Published
- 2004
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.