1
|
Matson AW, Hullsiek R, Dixon KJ, Wang S, Lindstedt AJ, Friess RR, Phung SK, Freedman TS, Felices M, Truckenbrod EN, Wu J, Miller JS, Walcheck B. Enhanced IL-15-mediated NK cell activation and proliferation by an ADAM17 function-blocking antibody involves CD16A, CD137, and accessory cells. J Immunother Cancer 2024; 12:e008959. [PMID: 39053944 DOI: 10.1136/jitc-2024-008959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/27/2024] [Indexed: 07/27/2024] Open
Abstract
BACKGROUND Natural killer (NK) cells are being extensively studied as a cell therapy for cancer. These cells are activated by recognition of ligands and antigens on tumor cells. Cytokine therapies, such as IL-15, are also broadly used to stimulate endogenous and adoptively transferred NK cells in patients with cancer. These stimuli activate the membrane protease ADAM17, which cleaves various cell-surface receptors on NK cells as a negative feedback loop to limit their cytolytic function. ADAM17 inhibition can enhance IL-15-mediated NK cell proliferation in vitro and in vivo. In this study, we investigated the underlying mechanism of this process. METHODS Peripheral blood mononuclear cells (PBMCs) or enriched NK cells from human peripheral blood, either unlabeled or labeled with a cell proliferation dye, were cultured for up to 7 days in the presence of rhIL-15±an ADAM17 function-blocking antibody. Different fully human versions of the antibody were generated; Medi-1 (IgG1), Medi-4 (IgG4), Medi-PGLALA, Medi-F(ab')2, and TAB16 (anti-ADAM17 and anti-CD16 bispecific) to modulate CD16A binding. Flow cytometry was used to assess NK cell proliferation and phenotypic markers, immunoblotting to examine CD16A signaling, and IncuCyte-based live cell imaging to measure NK cell antitumor activity. RESULTS The ADAM17 function-blocking monoclonal antibody (mAb) Medi-1 markedly increased early NK cell activation by IL-15. By using different engineered versions of the antibody, we demonstrate involvement by CD16A, an activating Fcγ receptor and well-described ADAM17 substrate. Hence, Medi-1 when bound to ADAM17 on NK cells is engaged by CD16A and blocks its shedding, inducing and prolonging its signaling. This process did not promote evident NK cell fratricide or dysfunction. Synergistic signaling by Medi-1 and IL-15 enhanced the upregulation of CD137 on CD16A+ NK cells and augmented their proliferation in the presence of PBMC accessory cells or an anti-CD137 agonistic mAb. CONCLUSIONS Our data reveal for the first time that CD16A and CD137 underpin Medi-1 enhancement of IL-15-driven NK cell activation and proliferation, respectively, with the latter requiring PBMC accessory cells. The use of Medi-1 represents a novel strategy to enhance IL-15-driven NK cell proliferation, and it may be of therapeutic importance by increasing the antitumor activity of NK cells in patients with cancer.
Collapse
Affiliation(s)
- Anders W Matson
- Graduate Program in Comparative and Molecular Biosciences, University of Minnesota, Saint Paul, Minnesota, USA
| | - Rob Hullsiek
- Graduate Program in Microbiology, Immunology, and Cancer Biology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Kate J Dixon
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Minneapolis, Minnesota, USA
| | - Sam Wang
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Minneapolis, Minnesota, USA
| | - Anders J Lindstedt
- Graduate Program in Microbiology, Immunology, and Cancer Biology, University of Minnesota, Minneapolis, Minnesota, USA
- Medical Scientist Training Program, University of Minnesota, Minneapolis, Minnesota, USA
| | - Ryan R Friess
- Graduate Program in Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota, USA
| | - Shee Kwan Phung
- Graduate Program in Comparative and Molecular Biosciences, University of Minnesota, Saint Paul, Minnesota, USA
| | - Tanya S Freedman
- Center for Immunology, University of Minnesota, Minneapolis, Minnesota, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
- Department of Pharmacology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Martin Felices
- Department of Medicine, Division of Hematology, Oncology, and Transplantation, University of Minnesota, Minneapolis, Minnesota, USA
| | - Emily N Truckenbrod
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Minneapolis, Minnesota, USA
| | - Jianming Wu
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Minneapolis, Minnesota, USA
| | - Jeffrey S Miller
- Center for Immunology, University of Minnesota, Minneapolis, Minnesota, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
- Department of Medicine, Division of Hematology, Oncology, and Transplantation, University of Minnesota, Minneapolis, Minnesota, USA
| | - Bruce Walcheck
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Minneapolis, Minnesota, USA
- Center for Immunology, University of Minnesota, Minneapolis, Minnesota, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
2
|
Matson AW, Hullsiek RH, Dixon KJ, Wang S, Lindstedt AJ, Friess RR, Phung SK, Freedman TS, Felices M, Truckenbrod EN, Wu J, Miller JS, Walcheck B. Enhanced IL-15-mediated NK cell activation and proliferation by an ADAM17 function-blocking antibody involves CD16A, CD137, and accessory cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.09.593347. [PMID: 38798522 PMCID: PMC11118905 DOI: 10.1101/2024.05.09.593347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Background NK cells are being extensively studied as a cell therapy for cancer. Their effector functions are induced by the recognition of ligands on tumor cells and by various cytokines. IL-15 is broadly used to stimulate endogenous and adoptively transferred NK cells in cancer patients. These stimuli activate the membrane protease ADAM17, which then cleaves assorted receptors on the surface of NK cells as a negative feedback loop to limit their activation and function. We have shown that ADAM17 inhibition can enhance IL-15-mediated NK cell proliferation in vitro and in vivo . In this study, we investigated the underlying mechanism of this process. Methods PBMCs or enriched NK cells from human peripheral blood, either unlabeled or labeled with a cell proliferation dye, were cultured for up to 7 days in the presence of rhIL-15 +/- an ADAM17 function-blocking antibody. Different versions of the antibody were generated; Medi-1 (IgG1), Medi-4 (IgG4), Medi-PGLALA, Medi-F(ab') 2 , and TAB16 (anti-ADAM17 and anti-CD16 bispecific) to modulate CD16A engagement on NK cells. Flow cytometry was used to assess NK cell proliferation and phenotypic markers, immunoblotting to examine CD16A signaling, and IncuCyte-based live cell imaging to measure NK cell anti-tumor activity. Results The ADAM17 function-blocking mAb Medi-1 markedly increased initial NK cell activation by IL-15. Using different engineered versions of the antibody revealed that the activating Fcγ receptor CD16A, a well-described ADAM17 substrate, was critical for enhancing IL-15 stimulation. Hence, Medi-1 bound to ADAM17 on NK cells can be engaged by CD16A and block its shedding, inducing and prolonging its signaling. This process did not promote evident NK cell fratricide, phagocytosis, or dysfunction. Synergistic activity by Medi-1 and IL-15 enhanced the upregulation of CD137 on CD16A + NK cells and augmented their proliferation in the presence of PBMC accessory cells. Conclusions Our data reveal for the first time that CD16A and CD137 underpin Medi-1 enhancement of IL-15-driven NK cell activation and proliferation, respectively. The use of Medi-1 represents a novel strategy to enhance IL-15-driven NK cell proliferation, and it may be of therapeutic importance by increasing the anti-tumor activity of NK cells in cancer patients. What is already known on this topic NK cell therapies are being broadly investigated to treat cancer. NK cell stimulation by IL-15 prolongs their survival in cancer patients. Various stimuli including IL-15 activate ADAM17 in NK cells, a membrane protease that regulates the cell surface density of various receptors as a negative feedback mechanism. What this study adds Treating NK cells with the ADAM17 function-blocking mAb Medi-1 markedly enhanced their activation and proliferation. Our study reveals that the Fc and Fab regions of Medi-1 function synergistically with IL-15 in NK cell activation. Medi-1 treatment augments the upregulation of CD137 by NK cells, which enhances their proliferation in the presence of PBMC accessory cells. How this study might affect research practice or policy Our study is of translational importance as Medi-1 treatment in combination with IL-15 could potentially augment the proliferation and function of endogenous or adoptively transferred NK cells in cancer patients. Graphical abstract
Collapse
|
3
|
Gulen AE, Rudraboina R, Tarique M, Ulker V, Shirwan H, Yolcu ES. A novel agonist of 4-1BB costimulatory receptor shows therapeutic efficacy against a tobacco carcinogen-induced lung cancer. Cancer Immunol Immunother 2023; 72:3567-3579. [PMID: 37605009 PMCID: PMC10991934 DOI: 10.1007/s00262-023-03507-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 07/23/2023] [Indexed: 08/23/2023]
Abstract
Immunotherapy utilizing checkpoint inhibitors has shown remarkable success in the treatment of cancers. In addition to immune checkpoint inhibitors, immune co-stimulation has the potential to enhance immune activation and destabilize the immunosuppressive tumor microenvironment. CD137, also known as 4-1BB, is one of the potent immune costimulatory receptors that could be targeted for effective immune co-stimulation. The interaction of the 4-1BB receptor with its natural ligand (4-1BBL) generates a strong costimulatory signal for T cell proliferation and survival. 4-1BBL lacks costimulatory activity in soluble form. To obtain co-stimulatory activity in soluble form, a recombinant 4-1BBL protein was generated by fusing the extracellular domains of murine 4-1BBL to a modified version of streptavidin (SA-4-1BBL). Treatment with SA-4-1BBL inhibited the development of lung tumors in A/J mice induced by weekly injections of the tobacco carcinogen NNK for eight weeks. The inhibition was dependent on the presence of T cells and NK cells; depletion of these cells diminished the SA-4-1BBL antitumor protective effect. The number of lung tumor nodules was significantly reduced by the administration of SA-4-1BBL to mice during ongoing exposure to NNK. The data presented in this paper suggest that utilizing an immune checkpoint stimulator as a single agent generate a protective immune response against lung cancer in the presence of a carcinogen. More broadly, this study suggests that immune checkpoint stimulation can be extended to a number of other cancer types, including breast and prostate cancers, for which improved diagnostics can detect disease at the preneoplastic stage.
Collapse
Affiliation(s)
- Ayse Ece Gulen
- Department of Child Health, University of Missouri, Columbia, MO, USA
- NextGen Precision Health, University of Missouri, Columbia, MO, USA
| | - Rakesh Rudraboina
- Department of Child Health, University of Missouri, Columbia, MO, USA
- NextGen Precision Health, University of Missouri, Columbia, MO, USA
| | - Mohammad Tarique
- Department of Child Health, University of Missouri, Columbia, MO, USA
- NextGen Precision Health, University of Missouri, Columbia, MO, USA
| | - Vahap Ulker
- Department of Child Health, University of Missouri, Columbia, MO, USA
- NextGen Precision Health, University of Missouri, Columbia, MO, USA
| | - Haval Shirwan
- Department of Child Health, University of Missouri, Columbia, MO, USA.
- Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, MO, USA.
- NextGen Precision Health, University of Missouri, Columbia, MO, USA.
| | - Esma S Yolcu
- Department of Child Health, University of Missouri, Columbia, MO, USA.
- Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, MO, USA.
- NextGen Precision Health, University of Missouri, Columbia, MO, USA.
| |
Collapse
|
4
|
Dunn ZS, Qu Y, MacMullan M, Chen X, Cinay G, Wang P. Secretion of 4-1BB Ligand Crosslinked to PD-1 Checkpoint Inhibitor Potentiates Chimeric Antigen Receptor T Cell Solid Tumor Efficacy. Hum Gene Ther 2023; 34:1145-1161. [PMID: 36851890 DOI: 10.1089/hum.2022.068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy has transformed the treatment of hematological malignancies but has yet to achieve similar success in solid tumors due to a lack of persistence and function in the tumor microenvironment. We previously reported the augmentation of CAR T cell therapy in an engineered solid tumor model through the secretion of anti-PD-1 single-chain fragment variable region (scFv), as shown by enhanced CAR T cell antitumor efficacy, expansion, and vitality. We have since improved the platform to create a superior cellular product-CAR T cells secreting single-chain trimeric 4-1BB ligand fused to anti-PD-1 scFv (αPD1-41BBL). 4-1BB signaling promotes cytotoxic T lymphocyte proliferation and survival but targeting 4-1BB with agonist antibodies in the clinic has been hindered by low antitumor activity and high toxicity. CAR T cells using 4-1BB endodomain for costimulatory signals have demonstrated milder antitumor response and longer persistence compared to CAR T cells costimulated by CD28 endodomain. We have, for the first time, engineered CD28-costimulated CAR T cells to secrete a fusion protein containing the soluble trimeric 4-1BB ligand. In vitro and in vivo, CAR19.αPD1-41BBL T cells exhibited reduced inhibitory receptor upregulation, enhanced persistence and proliferation, and a less differentiated memory status compared to CAR T cells without additional 4-1BB:4-1BBL costimulation. Accordingly, CAR19.αPD1-41BBL T cell-treated mice displayed significantly improved tumor growth control and overall survival. Spurred on by our preclinical success targeting CD19 as a model antigen, we produced mesothelin-targeting CAR T cells and confirmed the enhanced solid tumor efficacy of αPD1-41BBL-secreting CAR T cells.
Collapse
Affiliation(s)
- Zachary S Dunn
- Mork Family Department of Chemical Engineering and Materials Science, Viterbi School of Engineering, University of Southern California, Los Angeles, California, USA
| | - Yun Qu
- Mork Family Department of Chemical Engineering and Materials Science, Viterbi School of Engineering, University of Southern California, Los Angeles, California, USA
| | - Melanie MacMullan
- Mork Family Department of Chemical Engineering and Materials Science, Viterbi School of Engineering, University of Southern California, Los Angeles, California, USA
| | - Xianhui Chen
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California, USA
| | - Gunce Cinay
- Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, California, USA
| | - Pin Wang
- Mork Family Department of Chemical Engineering and Materials Science, Viterbi School of Engineering, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
5
|
Salek-Ardakani S, Zajonc DM, Croft M. Agonism of 4-1BB for immune therapy: a perspective on possibilities and complications. Front Immunol 2023; 14:1228486. [PMID: 37662949 PMCID: PMC10469789 DOI: 10.3389/fimmu.2023.1228486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 08/03/2023] [Indexed: 09/05/2023] Open
Abstract
Costimulatory receptors on immune cells represent attractive targets for immunotherapy given that these molecules can increase the frequency of individual protective immune cell populations and their longevity, as well as enhance various effector functions. 4-1BB, a member of the TNF receptor superfamily, also known as CD137 and TNFRSF9, is one such molecule that is inducible on several cell types, including T cells and NK cells. Preclinical studies in animal models have validated the notion that stimulating 4-1BB with agonist reagents or its natural ligand could be useful to augment conventional T cell and NK cell immunity to protect against tumor growth and against viral infection. Additionally, stimulating 4-1BB can enhance regulatory T cell function and might be useful in the right context for suppressing autoimmunity. Two human agonist antibodies to 4-1BB have been produced and tested in clinical trials for cancer, with variable results, leading to the production of a wealth of second-generation antibody constructs, including bi- and multi-specifics, with the hope of optimizing activity and selectivity. Here, we review the progress to date in agonism of 4-1BB, discuss the complications in targeting the immune system appropriately to elicit the desired activity, together with challenges in engineering agonists, and highlight the untapped potential of manipulating this molecule in infectious disease and autoimmunity.
Collapse
Affiliation(s)
| | - Dirk M. Zajonc
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Michael Croft
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA, United States
- Department of Medicine, University of California (UC) San Diego, La Jolla, CA, United States
| |
Collapse
|
6
|
Peper-Gabriel JK, Pavlidou M, Pattarini L, Morales-Kastresana A, Jaquin TJ, Gallou C, Hansbauer EM, Richter M, Lelievre H, Scholer-Dahirel A, Bossenmaier B, Sancerne C, Riviere M, Grandclaudon M, Zettl M, Bel Aiba RS, Rothe C, Blanc V, Olwill SA. The PD-L1/4-1BB Bispecific Antibody-Anticalin Fusion Protein PRS-344/S095012 Elicits Strong T-Cell Stimulation in a Tumor-Localized Manner. Clin Cancer Res 2022; 28:3387-3399. [PMID: 35121624 PMCID: PMC9662934 DOI: 10.1158/1078-0432.ccr-21-2762] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/25/2021] [Accepted: 02/02/2022] [Indexed: 01/07/2023]
Abstract
PURPOSE While patients responding to checkpoint blockade often achieve remarkable clinical responses, there is still significant unmet need due to resistant or refractory tumors. A combination of checkpoint blockade with further T-cell stimulation mediated by 4-1BB agonism may increase response rates and durability of response. A bispecific molecule that blocks the programmed cell death 1 (PD-1)/programmed cell death 1 ligand 1 (PD-L1) axis and localizes 4-1BB costimulation to a PD-L1-positive (PD-L1+) tumor microenvironment (TME) or tumor draining lymph nodes could maximize antitumor immunity and increase the therapeutic window beyond what has been reported for anti-4-1BB mAbs. EXPERIMENTAL DESIGN We generated and characterized the PD-L1/4-1BB bispecific molecule PRS-344/S095012 for target binding and functional activity in multiple relevant in vitro assays. Transgenic mice expressing human 4-1BB were transplanted with human PD-L1-expressing murine MC38 cells to assess in vivo antitumoral activity. RESULTS PRS-344/S095012 bound to its targets with high affinity and efficiently blocked the PD-1/PD-L1 pathway, and PRS-344/S095012-mediated 4-1BB costimulation was strictly PD-L1 dependent. We demonstrated a synergistic effect of both pathways on T-cell stimulation with the bispecific PRS-344/S095012 being more potent than the combination of mAbs. PRS-344/S095012 augmented CD4-positive (CD4+) and CD8-positive (CD8+) T-cell effector functions and enhanced antigen-specific T-cell stimulation. Finally, PRS-344/S095012 demonstrated strong antitumoral efficacy in an anti-PD-L1-resistant mouse model in which soluble 4-1BB was detected as an early marker for 4-1BB agonist activity. CONCLUSIONS The PD-L1/4-1BB bispecific PRS-344/S095012 efficiently combines checkpoint blockade with a tumor-localized 4-1BB-mediated stimulation burst to antigen-specific T cells, more potent than the combination of mAbs, supporting the advancement of PRS-344/S095012 toward clinical development. See related commentary by Shu et al., p. 3182.
Collapse
Affiliation(s)
| | | | - Lucia Pattarini
- Institut de Recherches Servier, Center for Therapeutic Innovation Oncology, Croissy-sur-Seine, France
| | | | | | - Catherine Gallou
- Institut de Recherches Servier, Center for Therapeutic Innovation Oncology, Croissy-sur-Seine, France
| | | | | | - Helene Lelievre
- Institut de Recherches Internationales Servier Oncology R&D Unit, Suresnes, France
| | - Alix Scholer-Dahirel
- Institut de Recherches Internationales Servier Oncology R&D Unit, Suresnes, France
| | | | - Celine Sancerne
- Institut de Recherches Servier, Center for Therapeutic Innovation Oncology, Croissy-sur-Seine, France
| | - Matthieu Riviere
- Institut de Recherches Servier, Center for Therapeutic Innovation Oncology, Croissy-sur-Seine, France
| | - Maximilien Grandclaudon
- Institut de Recherches Servier, Center for Therapeutic Innovation Oncology, Croissy-sur-Seine, France
| | - Markus Zettl
- Pieris Pharmaceuticals GmbH, Hallbergmoos, Germany
| | | | | | - Veronique Blanc
- Institut de Recherches Servier, Center for Therapeutic Innovation Oncology, Croissy-sur-Seine, France
| | | |
Collapse
|
7
|
Warmuth S, Gunde T, Snell D, Brock M, Weinert C, Simonin A, Hess C, Tietz J, Johansson M, Spiga FM, Heiz R, Flückiger N, Wagen S, Zeberer J, Diem D, Mahler D, Wickihalder B, Muntwiler S, Chatterjee B, Küttner B, Bommer B, Yaman Y, Lichtlen P, Urech D. Engineering of a trispecific tumor-targeted immunotherapy incorporating 4-1BB co-stimulation and PD-L1 blockade. Oncoimmunology 2022; 10:2004661. [PMID: 35844969 PMCID: PMC9278964 DOI: 10.1080/2162402x.2021.2004661] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Co-stimulatory 4-1BB receptors on tumor-infiltrating T cells are a compelling target for overcoming resistance to immune checkpoint inhibitors, but initial clinical studies of 4-1BB agonist mAbs were accompanied by liver toxicity. We sought to engineer a tri-specific antibody-based molecule that stimulates intratumoral 4-1BB and blocks PD-L1/PD-1 signaling without systemic toxicity and with clinically favorable pharmacokinetics. Recombinant fusion proteins were constructed using scMATCH3 technology and humanized antibody single-chain variable fragments against PD-L1, 4-1BB, and human serum albumin. Paratope affinities were optimized using single amino acid substitutions, leading to design of the drug candidate NM21-1480. Multiple in vitro experiments evaluated pharmacodynamic properties of NM21-1480, and syngeneic mouse tumor models assessed antitumor efficacy and safety of murine analogues. A GLP multiple-dose toxicology study evaluated its safety in non-human primates. NM21-1480 inhibited PD-L1/PD-1 signaling with a potency similar to avelumab, and it potently stimulated 4-1BB signaling only in the presence of PD-L1, while exhibiting an EC50 that was largely independent of PD-L1 density. NM21-1480 exhibited high efficacy for co-activation of pre-stimulated T cells and dendritic cells. In xenograft models in syngeneic mice, NM21-1480 induced tumor regression and tumor infiltration of T cells without causing systemic T-cell activation. A GLP toxicology study revealed no evidence of liver toxicity at doses up to 140 mg/kg, and pharmacokinetic studies in non-human primates suggested a plasma half-life in humans of up to 2 weeks. NM21-1480 has the potential to overcome checkpoint resistance by co-activating tumor-infiltrating lymphocytes without liver toxicity.
Collapse
Affiliation(s)
| | - Tea Gunde
- Numab Therapeutics AG, Waedenswil, Switzerland
| | | | | | | | | | | | - Julia Tietz
- Numab Therapeutics AG, Waedenswil, Switzerland
| | | | | | - Robin Heiz
- Numab Therapeutics AG, Waedenswil, Switzerland
| | | | | | | | - Dania Diem
- Numab Therapeutics AG, Waedenswil, Switzerland
| | - Dana Mahler
- Numab Therapeutics AG, Waedenswil, Switzerland
| | | | | | | | | | | | | | | | - David Urech
- Numab Therapeutics AG, Waedenswil, Switzerland
| |
Collapse
|
8
|
Safarzadeh Kozani P, Safarzadeh Kozani P, Ahmadi Najafabadi M, Yousefi F, Mirarefin SMJ, Rahbarizadeh F. Recent Advances in Solid Tumor CAR-T Cell Therapy: Driving Tumor Cells From Hero to Zero? Front Immunol 2022; 13:795164. [PMID: 35634281 PMCID: PMC9130586 DOI: 10.3389/fimmu.2022.795164] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 04/04/2022] [Indexed: 12/21/2022] Open
Abstract
Chimeric antigen receptor T-cells (CAR-Ts) are known as revolutionary living drugs that have turned the tables of conventional cancer treatments in certain hematologic malignancies such as B-cell acute lymphoblastic leukemia (B-ALL) and diffuse large B-cell lymphoma (DLBCL) by achieving US Food and Drug Administration (FDA) approval based on their successful clinical outcomes. However, this type of therapy has not seen the light of victory in the fight against solid tumors because of various restricting caveats including heterogeneous tumor antigen expression and the immunosuppressive tumor microenvironments (TME) that negatively affect the tumor-site accessibility, infiltration, stimulation, activation, and persistence of CAR-Ts. In this review, we explore strategic twists including boosting vaccines and designing implementations that can support CAR-T expansion, proliferation, and tumoricidal capacity. We also step further by underscoring novel strategies for triggering endogenous antitumor responses and overcoming the limitation of poor CAR-T tumor-tissue infiltration and the lack of definitive tumor-specific antigens. Ultimately, we highlight how these approaches can address the mentioned arduous hurdles.
Collapse
Affiliation(s)
- Pouya Safarzadeh Kozani
- Department of Medical Biotechnology, Faculty of Paramedicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Pooria Safarzadeh Kozani
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Milad Ahmadi Najafabadi
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Fatemeh Yousefi
- Department of Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | | | - Fatemeh Rahbarizadeh
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.,Research and Development Center of Biotechnology, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
9
|
4-1BBL as a Mediator of Cross-Talk between Innate, Adaptive, and Regulatory Immunity against Cancer. Int J Mol Sci 2021; 22:ijms22126210. [PMID: 34207500 PMCID: PMC8227424 DOI: 10.3390/ijms22126210] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/27/2021] [Accepted: 05/28/2021] [Indexed: 01/25/2023] Open
Abstract
The ability of tumor cells to evade the immune system is one of the main challenges we confront in the fight against cancer. Multiple strategies have been developed to counteract this situation, including the use of immunostimulant molecules that play a key role in the anti-tumor immune response. Such a response needs to be tumor-specific to cause as little damage as possible to healthy cells and also to track and eliminate disseminated tumor cells. Therefore, the combination of immunostimulant molecules and tumor-associated antigens has been implemented as an anti-tumor therapy strategy to eliminate the main obstacles confronted in conventional therapies. The immunostimulant 4-1BBL belongs to the tumor necrosis factor (TNF) family and it has been widely reported as the most effective member for activating lymphocytes. Hence, we will review the molecular, pre-clinical, and clinical applications in conjunction with tumor-associated antigens in antitumor immunotherapy, as well as the main molecular pathways involved in this association.
Collapse
|
10
|
Recombinant Costimulatory Fusion Proteins as Functional Immunomodulators Enhance Antitumor Activity in Murine B16F10 Melanoma. Vaccines (Basel) 2020; 8:vaccines8020223. [PMID: 32423130 PMCID: PMC7349950 DOI: 10.3390/vaccines8020223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 05/04/2020] [Accepted: 05/12/2020] [Indexed: 11/23/2022] Open
Abstract
Blocking inhibitory signaling and engaging stimulatory signaling have emerged as important therapeutic modalities for cancer immunotherapy. This study aimed to investigate immunomodulatory features of three recombinant costimulatory ligand proteins in a mouse model, which are extracellular domains of OX40-ligand (OX40L), 4-1BB-ligand (4-1BBL), or two domains in tandem, fused with the transmembrane domain of diphtheria toxin (DTT), named DTT-COS1, DTT-COS2, and DTT-COS12, respectively. In vitro study showed that DTT-COS1 and DTT-COS12 had immunological activity increasing the ratio of CD8/CD4 T cells. Treatments with DTT-COS1 and DTT-COS12 dramatically generated immune protection against the B16F10 tumor challenge in both prophylactic and therapeutic efficacy. Furthermore, regarding tumor microenvironment (TME) immunomodulation, DTT-COS1 treatment increased the proportion of CD4+ effector T cells (Teff) and decreased the expression of a suppressive cytokine. Meanwhile, DTT-COS12 reduced regulatory T cells (Treg) and improved the level of stimulatory cytokines. In addition, endogenous antibodies against OX40L/4-1BBL were generated, which may help with antitumor responses. Unexpectedly, DTT-COS2 lacked antitumor effects in vitro and in vivo. Importantly, serum analysis of liver-function associated factors and pro-inflammatory cytokines demonstrated that treatments were safe formulations in mice without signs of systemic toxicity. Remarkably, DTT-COS1 and DTT-COS12 are functional immunomodulators for mouse B16F10 melanoma, creating practical preclinical value in cancer immunotherapy.
Collapse
|
11
|
Barsoumian HB, Batra L, Shrestha P, Bowen WS, Zhao H, Egilmez NK, Gomez-Gutierrez JG, Yolcu ES, Shirwan H. A Novel Form of 4-1BBL Prevents Cancer Development via Nonspecific Activation of CD4 + T and Natural Killer Cells. Cancer Res 2019; 79:783-794. [PMID: 30770367 DOI: 10.1158/0008-5472.can-18-2401] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 11/13/2018] [Accepted: 12/28/2018] [Indexed: 01/11/2023]
Abstract
Costimulation through 4-1BB (CD137) receptor generates robust CD8+ T-effector and memory responses. The only known ligand, 4-1BBL, is a trimeric transmembrane protein that has no costimulatory activity as a soluble molecule. Thus, agonistic antibodies to the receptor have been used for cancer immunotherapy in preclinical models and are currently being evaluated in the clinic. Here, we report that treatment with an oligomeric form of the ligand, SA-4-1BBL, as a single agent is able to protect mice against subsequent tumor challenge irrespective of the tumor type. Protection was long-lasting (>8 weeks) and a bona fide property of SA-4-1BBL, as treatment with an agonistic antibody to the 4-1BB receptor was ineffective in generating immune protection against tumor challenge. Mechanistically, SA-4-1BBL significantly expanded IFNγ-expressing, preexisting memory-like CD44+CD4+ T cells and NK cells in naïve mice as compared with the agonistic antibody. In vivo blockade of IFNγ or depletion of CD4+ T or NK cells, but not CD8+ T or B cells, abrogated the immunopreventive effects of SA-4-1BBL against cancer. SA-4-1BBL as a single agent also exhibited robust efficacy in controlling postsurgical recurrences. This work highlights unexpected features of SA-4-1BBL as a novel immunomodulator with implications for cancer immunoprevention and therapy. SIGNIFICANCE: This study demonstrates the unique and unexpected immunomodulatory features of SA-4-1BBL that bridge innate and adaptive immune responses with both preventive and therapeutic efficacy against cancer.
Collapse
Affiliation(s)
- Hampartsoum B Barsoumian
- Institute for Cellular Therapeutics, University of Louisville, Louisville, Kentucky.,Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky
| | - Lalit Batra
- Institute for Cellular Therapeutics, University of Louisville, Louisville, Kentucky.,Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky
| | - Pradeep Shrestha
- Institute for Cellular Therapeutics, University of Louisville, Louisville, Kentucky.,Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky
| | | | - Hong Zhao
- Institute for Cellular Therapeutics, University of Louisville, Louisville, Kentucky.,Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky
| | - Nejat K Egilmez
- Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky
| | | | - Esma S Yolcu
- Institute for Cellular Therapeutics, University of Louisville, Louisville, Kentucky. .,Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky.,FasCure Therapeutics, LLC, Louisville, Kentucky
| | - Haval Shirwan
- Institute for Cellular Therapeutics, University of Louisville, Louisville, Kentucky. .,Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky
| |
Collapse
|
12
|
A DNA Vaccine Encoding SA-4-1BBL Fused to HPV-16 E7 Antigen Has Prophylactic and Therapeutic Efficacy in a Cervical Cancer Mouse Model. Cancers (Basel) 2019; 11:cancers11010096. [PMID: 30650588 PMCID: PMC6356763 DOI: 10.3390/cancers11010096] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 01/03/2019] [Accepted: 01/08/2019] [Indexed: 12/12/2022] Open
Abstract
The SA-4-1BBL, an oligomeric novel form of the natural ligand for the 4-1BB co-stimulatory receptor of the tumor necrosis factor (TNF) superfamily, as a recombinant protein has potent pleiotropic effects on cells of innate, adaptive, and regulatory immunity with demonstrated therapeutic efficacy in several tumor models. However, the production of soluble form of SA-4-1BBL protein and quality control is time and resource intensive and face various issues pertinent to clinical development of biologics. The present study sought to take advantage of the simplicity and translatability of DNA-based vaccines for the production and delivery of SA-4-1BBL for cancer immune prevention and therapy. A chimeric HPV-16 E7 DNA vaccine (SP-SA-E7-4-1BBL) was constructed that contains the signal peptide (SP) of calreticulin (CRT), streptavidin (SA) domain of SA-4-1BBL, HPV-16 E7 double mutant gene, and the extracellular domain of mouse 4-1BBL. Immunization by gene gun with SP-SA-E7-4-1BBL induced greater prophylactic as well as therapeutic effects in C57BL/6 mice against TC-1 tumor model compared with immunization with E7wt, SP-SA-4-1BBL or reference-positive control CRT-E7wt. The therapeutic efficacy of the DNA vaccine was associated with increased frequency of E7-specific T cells producing interferon (IFN)-γ. Overall, our data suggest that this DNA-based vaccine strategy might represent a translational approach because it provides a simpler and versatile alternative to a subunit vaccine based on SA-4-1BBL and E7 proteins.
Collapse
|
13
|
Sun C, He D, Ma C, Gao Z, Chen Y, Wang S. Bifunctional Fusion Proteins Derived from Tumstatin and 4-1BBL for Targeted Cancer Therapy. Mol Pharm 2018; 16:867-876. [PMID: 30565463 DOI: 10.1021/acs.molpharmaceut.8b01190] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The therapeutic utilities of antiangiogenesis and immunotherapy have been proven in clinics, and cancer patients have benefited from respective therapy. Given that the combination of both therapeutic strategies may further improve the effectiveness, a recombinant human 4-1BBL/tumstatin fusion protein (rh4TFP) library was constructed in the present study to target both angiogenesis and T lymphocyte activation, in which the fragments of an endogenous angiogenesis inhibitor tumstatin and a T lymphocyte costimulatory 4-1BBL are coupled with different linkers. After comparison of different combinations, rh4TFP-2 was found to show a promise on potential antiangiogenic immunotherapy. On one hand, rh4TFP-2 inhibited proliferation and migration of human umbilical vein endothelial cells, exhibiting the antiangiogenic activity similar to tumstatin. On the other hand, rh4TFP-2 led to significant increase of T lymphocyte activation for the release of IL-2 and IFN-γ, showing the T lymphocyte activation by 4-1BBL. Moreover, administration of rh4TFP-2 suppressed tumor growth and prolonged survival in a B16F10 melanoma-bearing mouse model. Taken together, the present study provides a new approach of using bifunctional fusion proteins to target both angiogenesis and T lymphocyte activation for cancer therapy.
Collapse
Affiliation(s)
- Chao Sun
- State Key Laboratory of Natural Medicines and Laboratory of Chemical Biology , China Pharmaceutical University , Nanjing 210009 , China
| | - Dongyang He
- State Key Laboratory of Natural Medicines and Laboratory of Chemical Biology , China Pharmaceutical University , Nanjing 210009 , China
| | - Chao Ma
- State Key Laboratory of Natural Medicines and Laboratory of Chemical Biology , China Pharmaceutical University , Nanjing 210009 , China
| | - Zhenyue Gao
- State Key Laboratory of Natural Medicines and Laboratory of Chemical Biology , China Pharmaceutical University , Nanjing 210009 , China
| | - Yijun Chen
- State Key Laboratory of Natural Medicines and Laboratory of Chemical Biology , China Pharmaceutical University , Nanjing 210009 , China
| | - Shuzhen Wang
- State Key Laboratory of Natural Medicines and Laboratory of Chemical Biology , China Pharmaceutical University , Nanjing 210009 , China
| |
Collapse
|
14
|
A tumor-targeted trimeric 4-1BB-agonistic antibody induces potent anti-tumor immunity without systemic toxicity. Nat Commun 2018; 9:4809. [PMID: 30442944 PMCID: PMC6237851 DOI: 10.1038/s41467-018-07195-w] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 10/17/2018] [Indexed: 02/06/2023] Open
Abstract
The costimulation of immune cells using first-generation anti-4-1BB monoclonal antibodies (mAbs) has demonstrated anti-tumor activity in human trials. Further clinical development, however, is restricted by significant off-tumor toxicities associated with FcγR interactions. Here, we have designed an Fc-free tumor-targeted 4-1BB-agonistic trimerbody, 1D8N/CEGa1, consisting of three anti-4-1BB single-chain variable fragments and three anti-EGFR single-domain antibodies positioned in an extended hexagonal conformation around the collagen XVIII homotrimerization domain. The1D8N/CEGa1 trimerbody demonstrated high-avidity binding to 4-1BB and EGFR and a potent in vitro costimulatory capacity in the presence of EGFR. The trimerbody rapidly accumulates in EGFR-positive tumors and exhibits anti-tumor activity similar to IgG-based 4-1BB-agonistic mAbs. Importantly, treatment with 1D8N/CEGa1 does not induce systemic inflammatory cytokine production or hepatotoxicity associated with IgG-based 4-1BB agonists. These results implicate FcγR interactions in the 4-1BB-agonist-associated immune abnormalities, and promote the use of the non-canonical antibody presented in this work for safe and effective costimulatory strategies in cancer immunotherapy. Cancer therapy using systemically administrated 4-1BB-targeting antibodies is often associated with severe toxicity due to the nonspecific activation of autoreactive T cells. Here, the authors have developed a trimeric antibody targeting both 4-1BB and EGFR, which activates T cells effectively and shows negligible cytotoxicity.
Collapse
|
15
|
Zitzmann J, Schreiber C, Eichmann J, Bilz RO, Salzig D, Weidner T, Czermak P. Single-cell cloning enables the selection of more productive Drosophila melanogaster S2 cells for recombinant protein expression. BIOTECHNOLOGY REPORTS (AMSTERDAM, NETHERLANDS) 2018; 19:e00272. [PMID: 29998071 PMCID: PMC6037645 DOI: 10.1016/j.btre.2018.e00272] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 06/18/2018] [Accepted: 06/22/2018] [Indexed: 11/16/2022]
Abstract
The generation of monoclonal cell lines is an important early process development step for recombinant protein production. Although single-cell cloning is an established method in mammalian cell lines, straightforward protocols are not yet available for insect cells. We describe a new method for the generation of monoclonal insect cells without using fetal bovine serum and/or feeder cells pretreated by irradiation or exposure to mitomycin. Highly productive clones of Drosophila melanogaster S2 cells were prepared in a two-step procedure, comprising the establishment of a polyclonal population and subsequent single cell isolation by limiting dilution. Necessary growth factors were provided by co-cultivation of single transformants with untransfected feeder cells, which were later removed by antibiotic selection. Enhanced expression of EGFP and two target peptides was confirmed by flow cytometry and dot/western blotting. Highly productive clones were stable, showed a uniform expression profile and typically a sixfold to tenfold increase in cell-specific productivity.
Collapse
Key Words
- AMP, antimicrobial peptide/protein
- BR021, Harmonia axyridis antimicrobial peptide BR021
- BSA, bovine serum albumin
- D. melanogaster S2 cells
- DMSO, dimethyl sulfoxide
- EGFP, enhanced green fluorescent protein
- FACS, fluorescence activated cell sorting
- FBS, fetal bovine serum
- GMP, good manufacturing practice
- GmGlv, Galleria mellonella antimicrobial peptide Gloverin
- Insect cell culture
- Monoclonal cell line
- OD600, optical density at 600nm
- PBS, phosphate-buffered saline
- PCR, polymerase chain reaction
- PVDF, polyvinylidene difluoride
- RMCE, recombinase mediated cassette exchange
- Recombinant protein expression
- SDS-PAGE, sodium dodecylsulfate polyacrylamide gel electrophoresis
- SFM, serum free medium
- Sf9, clonal isolate of Spodoptera frugiperda Sf21 cells
- Single-cell cloning
- Stably transformed
- rS2, recombinant Drosophila melanogaster Schneider 2 cells
Collapse
Affiliation(s)
- Jan Zitzmann
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen, Giessen, Germany
| | - Christine Schreiber
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen, Giessen, Germany
| | - Joel Eichmann
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen, Giessen, Germany
| | - Roberto Otmar Bilz
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen, Giessen, Germany
| | - Denise Salzig
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen, Giessen, Germany
| | - Tobias Weidner
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen, Giessen, Germany
| | - Peter Czermak
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen, Giessen, Germany
- Department of Chemical Engineering, Kansas State University, Manhattan KS, USA
- Faculty of Biology and Chemistry, Justus-Liebig University of Giessen, Germany
- Fraunhofer Institute for Molecular Biology and Applied Ecology (IME), Project group Bioresources, Giessen, Germany
| |
Collapse
|
16
|
Teijeira A, Labiano S, Garasa S, Etxeberria I, Santamaría E, Rouzaut A, Enamorado M, Azpilikueta A, Inoges S, Bolaños E, Aznar MA, Sánchez-Paulete AR, Sancho D, Melero I. Mitochondrial Morphological and Functional Reprogramming Following CD137 (4-1BB) Costimulation. Cancer Immunol Res 2018; 6:798-811. [PMID: 29678874 DOI: 10.1158/2326-6066.cir-17-0767] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 03/23/2018] [Accepted: 04/19/2018] [Indexed: 12/27/2022]
Abstract
T and NK lymphocytes express CD137 (4-1BB), a costimulatory receptor of the TNFR family whose function is exploitable for cancer immunotherapy. Mitochondria regulate the function and survival of T lymphocytes. Herein, we show that CD137 costimulation provided by agonist mAb and CD137L (4-1BBL) induced mitochondria enlargement that resulted in enhanced mitochondrial mass and transmembrane potential in human and mouse CD8+ T cells. Such mitochondrial changes increased T-cell respiratory capacities and were critically dependent on mitochondrial fusion protein OPA-1 expression. Mass and function of mitochondria in tumor-reactive CD8+ T cells from cancer-bearing mice were invigorated by agonist mAb to CD137, whereas mitochondrial baseline mass and function were depressed in CD137-deficient tumor reactive T cells. Tumor rejection induced by the synergistic combination of adoptive T-cell therapy and agonistic anti-CD137 was critically dependent on OPA-1 expression in transferred CD8+ T cells. Moreover, stimulation of CD137 with CD137 mAb in short-term cultures of human tumor-infiltrating lymphocytes led to mitochondria enlargement and increased transmembrane potential. Collectively, these data point to a critical link between mitochondrial morphology and function and enhanced antitumor effector activity upon CD137 costimulation of T cells. Cancer Immunol Res; 6(7); 798-811. ©2018 AACR.
Collapse
Affiliation(s)
- Alvaro Teijeira
- Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain. .,CIBERONC, Centro Virtual de la Investigación Biomédica en red de Oncología, Madrid, Spain
| | - Sara Labiano
- Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Saray Garasa
- Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Iñaki Etxeberria
- Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Eva Santamaría
- Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain.,CIBEREHD, Centro Virtual de la Investigación Biomédica en red de Enfermedades Hepáticas y Digestivas, Madrid, Spain
| | - Ana Rouzaut
- Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain.,CIBERONC, Centro Virtual de la Investigación Biomédica en red de Oncología, Madrid, Spain
| | - Michel Enamorado
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Arantza Azpilikueta
- Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain.,CIBERONC, Centro Virtual de la Investigación Biomédica en red de Oncología, Madrid, Spain
| | - Susana Inoges
- Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Elixabet Bolaños
- Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain.,CIBERONC, Centro Virtual de la Investigación Biomédica en red de Oncología, Madrid, Spain
| | - Maria Angela Aznar
- Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Alfonso R Sánchez-Paulete
- Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain.,CIBERONC, Centro Virtual de la Investigación Biomédica en red de Oncología, Madrid, Spain
| | - David Sancho
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Ignacio Melero
- Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain. .,CIBERONC, Centro Virtual de la Investigación Biomédica en red de Oncología, Madrid, Spain
| |
Collapse
|
17
|
Bitra A, Doukov T, Wang J, Picarda G, Benedict CA, Croft M, Zajonc DM. Crystal structure of murine 4-1BB and its interaction with 4-1BBL support a role for galectin-9 in 4-1BB signaling. J Biol Chem 2018; 293:1317-1329. [PMID: 29242193 PMCID: PMC5787808 DOI: 10.1074/jbc.m117.814905] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 12/01/2017] [Indexed: 11/06/2022] Open
Abstract
4-1BB (CD137) is a TNF receptor superfamily (TNFRSF) member that is thought to undergo receptor trimerization upon binding to its trimeric TNF superfamily ligand (4-1BBL) to stimulate immune responses. 4-1BB also can bind to the tandem repeat-type lectin galectin-9 (Gal-9), and signaling through mouse (m)4-1BB is reduced in galectin-9 (Gal-9)-deficient mice, suggesting a pivotal role of Gal-9 in m4-1BB activation. Here, using sulfur-SAD phasing, we determined the crystal structure of m4-1BB to 2.2-Å resolution. We found that similar to other TNFRSFs, m4-1BB has four cysteine-rich domains (CRDs). However, the organization of CRD1 and the orientation of CRD3 and CRD4 with respect to CRD2 in the m4-1BB structure distinctly differed from those of other TNFRSFs. Moreover, we mapped two Asn residues within CRD4 that are N-linked glycosylated and mediate m4-1BB binding to Gal-9. Kinetics studies of m4-1BB disclosed a very tight nanomolar binding affinity to m4-1BBL with an unexpectedly strong avidity effect. Both N- and C-terminal domains of Gal-9 bound m4-1BB, but with lower affinity compared with m4-1BBL. Although the TNF homology domain (THD) of human (h)4-1BBL forms non-covalent trimers, we found that m4-1BBL formed a covalent dimer via 2 cysteines absent in h4-1BBL. As multimerization and clustering is a prerequisite for TNFR intracellular signaling, and as m4-1BBL can only recruit two m4-1BB monomers, we hypothesize that m4-1BBL and Gal-9 act together to aid aggregation of m4-1BB monomers to efficiently initiate m4-1BB signaling.
Collapse
Affiliation(s)
- Aruna Bitra
- From the Division of Immune Regulation, La Jolla Institute for Allergy and Immunology (LJI), La Jolla, California 92037
| | - Tzanko Doukov
- the Stanford Synchrotron Radiation Light Source, Menlo Park, California 94025
| | - Jing Wang
- From the Division of Immune Regulation, La Jolla Institute for Allergy and Immunology (LJI), La Jolla, California 92037
| | - Gaelle Picarda
- From the Division of Immune Regulation, La Jolla Institute for Allergy and Immunology (LJI), La Jolla, California 92037
| | - Chris A Benedict
- From the Division of Immune Regulation, La Jolla Institute for Allergy and Immunology (LJI), La Jolla, California 92037
| | - Michael Croft
- From the Division of Immune Regulation, La Jolla Institute for Allergy and Immunology (LJI), La Jolla, California 92037
- the Department of Medicine, University of California San Diego, La Jolla, California 92037, and
| | - Dirk M Zajonc
- From the Division of Immune Regulation, La Jolla Institute for Allergy and Immunology (LJI), La Jolla, California 92037
- the Department of Internal Medicine, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| |
Collapse
|
18
|
Jin C, Duan X, Liu Y, Zhu J, Zhang K, Zhang Y, Xia T, Fei Y, Ye J. T cell immunity induced by a bivalent Salmonella-based CEACAM6 and 4-1BBL vaccines in a rat colorectal cancer model. Oncol Lett 2017; 13:3753-3759. [PMID: 28521477 DOI: 10.3892/ol.2017.5938] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 01/31/2017] [Indexed: 01/07/2023] Open
Abstract
The present study investigated the anti-tumor mechanisms of recombinant non-specific cross-reacting antigen (CEACAM6) and 4-1BB ligand (4-1BBL) Salmonella-based vaccines, and the effect that these vaccinations have on memory T cells and T helper (Th) cell polarization. Colon tumors were induced in rats via 1,2-dimethylhydrazine (DMH) injections. Rats were then treated with injections of attenuated Salmonella typhimurium carrying pIRES-CEACAM6, pIRES-4-1BBL or pIRES-CEACAM6-4-1BBL. In total, 4 vaccine injections, one every other week, were administered during the 8 weeks subsequent to the DMH injection. Rats were sacrificed 18 weeks subsequent to the DMH injections, and the colons and spleens were collected for further analysis. Cluster of differentiation (CD) 45RO, interleukin (IL)-4 and IL-17 expression was analyzed in colon tumor tissues, and the expression of interferon (IFN)-γ, CD3+, CD4+, CD8+, CD56+, forkhead/winged-helix transcription factor box P3 (FOXP3+), IL-4 and IL-17 were analyzed in splenic tissues. Compared with the pIRES/SL3261 group, the pIRES-CEACAM6-4-1BBL/SL3261 treatment group had a significantly higher number of CD45RO+ expressing tumor infiltrating lymphocytes and lower expression levels of IL-4 and IL-17. Splenic tissues from the same treatment group exhibited significantly increased expression of IFN-γ, CD3+ and CD8+ and reduced expression levels of Foxp3, IL-4 and IL-7. CD56+ T cell expression was increased in all groups except for the group that received no vaccine. The present study concluded that the combined CEACAM6 and 4-1BBL-attenuated recombinant Salmonella vaccine was able to inhibit the growth of DMH-induced colorectal tumors. This was mediated by generating an anti-tumor immune response, increasing the number of of CD45RO+ memory T cells, decreasing the number of FOXP3+ cells and promoting Th1 polarization.
Collapse
Affiliation(s)
- Chunhui Jin
- Department of Oncology, Wuxi City Hospital of Traditional Chinese Medicine, Wuxi, Jiangsu 214000, P.R. China
| | - Xiaoqing Duan
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Yingying Liu
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Jianhong Zhu
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Ke Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Yuanting Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Tingting Xia
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Yajun Fei
- Department of Pathology, Wuxi City Hospital of Traditional Chinese Medicine, Wuxi, Jiangsu 214000, P.R. China
| | - Jianxin Ye
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| |
Collapse
|
19
|
Fellermeier S, Beha N, Meyer JE, Ring S, Bader S, Kontermann RE, Müller D. Advancing targeted co-stimulation with antibody-fusion proteins by introducing TNF superfamily members in a single-chain format. Oncoimmunology 2016; 5:e1238540. [PMID: 27999756 DOI: 10.1080/2162402x.2016.1238540] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 09/10/2016] [Accepted: 09/13/2016] [Indexed: 01/12/2023] Open
Abstract
Co-stimulation via receptors of the tumor necrosis factor superfamily (TNFSF) emerges as promising strategy to support antitumor immune responses. Targeted strategies with antibody-fusion proteins composed of a tumor-directed antibody part and the extracellular domain of a co-stimulatory ligand of the TNFSF constitute an attractive option to focus the co-stimulatory activity to the tumor site. Since TNFSF members intrinsically form functional units of non-covalently linked homotrimers, the protein engineering of suitable antibody-fusion proteins is challenging. Aiming for molecules of simple and stable configuration, we used TNFSF ligands in a single-chain format (scTNFSF), i.e., three units of the ectodomain connected by polypeptide linkers, folding into an intramolecular trimer. By fusing tumor-directed scFv antibody fragments directed against EpCAM or FAP to co-stimulatory scTNFSF molecules (sc4-1BBL, scOX40L, scGITRL or scLIGHT), a set of monomeric scFv-scTNFSF fusion proteins was generated. In comparison to the scFv-TNFSF format, defined by intermolecular homotrimerization via the TNFSF part, scFv-scTNFSF showed equal or enhanced co-stimulatory activity despite reduced avidity in antibody binding. In addition, enhanced serum stability and improved bioavailability in mice were observed. We show that the scFv-scTNFSF format can be applied to various members of the TNFSF, presenting targeting-dependent co-stimulatory activity. Hence, this format exhibits favorable properties that make it a promising choice for further therapeutic fusion protein development.
Collapse
Affiliation(s)
- Sina Fellermeier
- Institute of Cell Biology and Immunology, University of Stuttgart , Stuttgart, Germany
| | - Nadine Beha
- Institute of Cell Biology and Immunology, University of Stuttgart , Stuttgart, Germany
| | - Jan-Erik Meyer
- Institute of Cell Biology and Immunology, University of Stuttgart , Stuttgart, Germany
| | - Sarah Ring
- Institute of Cell Biology and Immunology, University of Stuttgart , Stuttgart, Germany
| | - Stefan Bader
- Institute of Cell Biology and Immunology, University of Stuttgart , Stuttgart, Germany
| | - Roland E Kontermann
- Institute of Cell Biology and Immunology, University of Stuttgart , Stuttgart, Germany
| | - Dafne Müller
- Institute of Cell Biology and Immunology, University of Stuttgart , Stuttgart, Germany
| |
Collapse
|
20
|
Lang I, Füllsack S, Wyzgol A, Fick A, Trebing J, Arana JAC, Schäfer V, Weisenberger D, Wajant H. Binding Studies of TNF Receptor Superfamily (TNFRSF) Receptors on Intact Cells. J Biol Chem 2015; 291:5022-37. [PMID: 26721880 DOI: 10.1074/jbc.m115.683946] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Indexed: 01/17/2023] Open
Abstract
Ligands of the tumor necrosis factor superfamily (TNFSF) interact with members of the TNF receptor superfamily (TNFRSF). TNFSF ligand-TNFRSF receptor interactions have been intensively evaluated by many groups. The affinities of TNFSF ligand-TNFRSF receptor interactions are highly dependent on the oligomerization state of the receptor, and cellular factors (e.g. actin cytoskeleton and lipid rafts) influence the assembly of ligand-receptor complexes, too. Binding studies on TNFSF ligand-TNFRSF receptor interactions were typically performed using cell-free assays with recombinant fusion proteins that contain varying numbers of TNFRSF ectodomains. It is therefore not surprising that affinities determined for an individual TNFSF ligand-TNFRSF interaction differ sometimes by several orders of magnitude and often do not reflect the ligand activity observed in cellular assays. To overcome the intrinsic limitations of cell-free binding studies and usage of recombinant receptor domains, we performed comprehensive binding studies with Gaussia princeps luciferase TNFSF ligand fusion proteins for cell-bound TNFRSF members on intact cells at 37 °C. The affinities of the TNFSF ligand G. princeps luciferase-fusion proteins ranged between 0.01 and 19 nm and offer the currently most comprehensive and best suited panel of affinities for in silico studies of ligand-receptor systems of the TNF family.
Collapse
Affiliation(s)
- Isabell Lang
- From the Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital Würzburg, Röntgenring 11, 97070 Würzburg, Germany
| | - Simone Füllsack
- From the Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital Würzburg, Röntgenring 11, 97070 Würzburg, Germany
| | - Agnes Wyzgol
- From the Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital Würzburg, Röntgenring 11, 97070 Würzburg, Germany
| | - Andrea Fick
- From the Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital Würzburg, Röntgenring 11, 97070 Würzburg, Germany
| | - Johannes Trebing
- From the Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital Würzburg, Röntgenring 11, 97070 Würzburg, Germany
| | - José Antonio Carmona Arana
- From the Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital Würzburg, Röntgenring 11, 97070 Würzburg, Germany
| | - Viktoria Schäfer
- From the Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital Würzburg, Röntgenring 11, 97070 Würzburg, Germany
| | - Daniela Weisenberger
- From the Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital Würzburg, Röntgenring 11, 97070 Würzburg, Germany
| | - Harald Wajant
- From the Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital Würzburg, Röntgenring 11, 97070 Würzburg, Germany
| |
Collapse
|
21
|
LI XIAOMEI, HE CHENHUI, LIU CHANGZHEN, MA JUAN, MA PAN, CUI HONGLIAN, TAO HUA, GAO BIN. Expansion of NK cells from PBMCs using immobilized 4-1BBL and interleukin-21. Int J Oncol 2015; 47:335-42. [DOI: 10.3892/ijo.2015.3005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2015] [Accepted: 04/02/2015] [Indexed: 11/05/2022] Open
|
22
|
Sharma RK, Yolcu ES, Shirwan H. SA-4-1BBL as a novel adjuvant for the development of therapeutic cancer vaccines. Expert Rev Vaccines 2014; 13:387-98. [PMID: 24521311 DOI: 10.1586/14760584.2014.880340] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Tumor associated antigen (TAA)-based therapeutic vaccines have great potential as a safe, practical, and cost-efficient alternative to standard treatments for cancer. Clinical efficacy of TAA-based vaccines, however, has yet to be realized and will require adjuvants with pleiotropic functions on immune cells. Such adjuvants need not only to generate/boost T cell responses, but also reverse intrinsic/extrinsic tumor immune evasion mechanisms for therapeutic efficacy. This review focuses on a novel agonistic ligand, SA-4-1BBL, for 4-1BB costimulatory receptor as an adjuvant of choice because of its ability to: i) serve as a vehicle to deliver TAAs to dendritic cells (DCs) for antigen uptake and cross-presentation to CD8(+) T cells; ii) augment adaptive Th1 and innate immune responses; and iii) overcome various immune evasion mechanisms, cumulatively translating into therapeutic efficacy in preclinical tumor models.
Collapse
Affiliation(s)
- Rajesh K Sharma
- Department of Microbiology and Immunology, Institute for Cellular Therapeutics, School of Medicine, University of Louisville, Louisville, KY, 40202, USA
| | | | | |
Collapse
|
23
|
Madireddi S, Eun SY, Lee SW, Nemčovičová I, Mehta AK, Zajonc DM, Nishi N, Niki T, Hirashima M, Croft M. Galectin-9 controls the therapeutic activity of 4-1BB-targeting antibodies. ACTA ACUST UNITED AC 2014; 211:1433-48. [PMID: 24958847 PMCID: PMC4076583 DOI: 10.1084/jem.20132687] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Biologics to TNF family receptors are prime candidates for therapy of immune disease. Whereas recent studies have highlighted a requirement for Fcγ receptors in enabling the activity of CD40, TRAILR, and GITR when engaged by antibodies, other TNFR molecules may be controlled by additional mechanisms. Antibodies to 4-1BB (CD137) are currently in clinical trials and can both augment immunity in cancer and promote regulatory T cells that inhibit autoimmune disease. We found that the action of agonist anti-4-1BB in suppressing autoimmune and allergic inflammation was completely dependent on Galectin-9 (Gal-9). Gal-9 directly bound to 4-1BB, in a site distinct from the binding site of antibodies and the natural ligand of 4-1BB, and Gal-9 facilitated 4-1BB aggregation, signaling, and functional activity in T cells, dendritic cells, and natural killer cells. Conservation of the Gal-9 interaction in humans has important implications for effective clinical targeting of 4-1BB and possibly other TNFR superfamily molecules.
Collapse
Affiliation(s)
- Shravan Madireddi
- Division of Immune Regulation and Division of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037
| | - So-Young Eun
- Division of Immune Regulation and Division of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037
| | - Seung-Woo Lee
- Division of Immune Regulation and Division of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037
| | - Ivana Nemčovičová
- Division of Immune Regulation and Division of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037
| | - Amit Kumar Mehta
- Division of Immune Regulation and Division of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037
| | - Dirk M Zajonc
- Division of Immune Regulation and Division of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037
| | - Nozomu Nishi
- Life Science Research Center; and Department of Immunology and Immunopathology, Faculty of Medicine; Kagawa University, Kagawa 761-0793, Japan
| | - Toshiro Niki
- Life Science Research Center; and Department of Immunology and Immunopathology, Faculty of Medicine; Kagawa University, Kagawa 761-0793, Japan GalPharma Co., Ltd., Kagawa 760-0301, Japan
| | - Mitsuomi Hirashima
- Life Science Research Center; and Department of Immunology and Immunopathology, Faculty of Medicine; Kagawa University, Kagawa 761-0793, Japan GalPharma Co., Ltd., Kagawa 760-0301, Japan
| | - Michael Croft
- Division of Immune Regulation and Division of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037
| |
Collapse
|
24
|
Kachapati K, Bednar KJ, Adams DE, Wu Y, Mittler RS, Jordan MB, Hinerman JM, Herr AB, Ridgway WM. Recombinant soluble CD137 prevents type one diabetes in nonobese diabetic mice. J Autoimmun 2013; 47:94-103. [DOI: 10.1016/j.jaut.2013.09.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Revised: 09/26/2013] [Accepted: 09/28/2013] [Indexed: 11/28/2022]
|
25
|
Sharma RK, Yolcu ES, Srivastava AK, Shirwan H. CD4+ T cells play a critical role in the generation of primary and memory antitumor immune responses elicited by SA-4-1BBL and TAA-based vaccines in mouse tumor models. PLoS One 2013; 8:e73145. [PMID: 24066030 PMCID: PMC3774737 DOI: 10.1371/journal.pone.0073145] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Accepted: 07/17/2013] [Indexed: 12/20/2022] Open
Abstract
The role of CD4+ T cells in the generation of therapeutic primary and memory immune responses in cancer diverse immunotherapy settings remains ambiguous. We herein investigated this issue using two vaccine formulations containing a novel costimulatory molecule, SA-4-1BBL, as adjuvant and HPV E7 or survivin (SVN) as tumor associated antigens (TAAs) in two mouse transplantable tumor models; the TC-1 cervical cancer expressing xenogeneic HPV E7 and 3LL lung carcinoma overexpressing autologous SVN. Single vaccination with optimized SA-4-1BBL/TAA formulations resulted in the eradication of 6-day established TC-1 and 3LL tumors in >70% of mice in both models. The in vivo depletion of CD4+ T cells one day before tumor challenge resulted in compromised vaccine efficacy in both TC-1 (25%) and 3LL (12.5%) tumor models. In marked contrast, depletion of CD4+ T cells 5 days post-tumor challenge and one day prior to vaccination did not significantly alter the therapeutic efficacy of these vaccines. However, long-term immunological memory was compromised in the 3LL, but not in TC-1 model as a significant number (85.7%) of tumor free-mice succumbed to tumor growth when rechallenged with 3LL cells 60 days after the initial tumor inoculation. Collectively, these results demonstrate the indispensable role CD4+ T cells play in the generation of therapeutic primary immune responses elicited by SA-4-1BBL/TAA-based vaccines irrespective of the nature of TAAs and establish the importance of CD4+ T cells for long-term immune memory against 3LL tumor expressing self-antigen SVN, but not TC-1 expressing xenogeneic viral antigen E7.
Collapse
Affiliation(s)
- Rajesh K. Sharma
- Institute for Cellular Therapeutics, Department of Microbiology and Immunology and James Brown Cancer Center, University of Louisville, Louisville, Kentucky, United States of America
| | - Esma S. Yolcu
- Institute for Cellular Therapeutics, Department of Microbiology and Immunology and James Brown Cancer Center, University of Louisville, Louisville, Kentucky, United States of America
| | - Abhishek K. Srivastava
- Institute for Cellular Therapeutics, Department of Microbiology and Immunology and James Brown Cancer Center, University of Louisville, Louisville, Kentucky, United States of America
| | - Haval Shirwan
- Institute for Cellular Therapeutics, Department of Microbiology and Immunology and James Brown Cancer Center, University of Louisville, Louisville, Kentucky, United States of America
- * E-mail:
| |
Collapse
|
26
|
Müller D. Antibody–Cytokine Fusion Proteins for Cancer Immunotherapy: An Update on Recent Developments. BioDrugs 2013; 28:123-31. [DOI: 10.1007/s40259-013-0069-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
27
|
Teijeira Á, Palazón A, Garasa S, Marré D, Aubá C, Rogel A, Murillo O, Martínez‐Forero I, Lang F, Melero I, Rouzaut A. CD137 on inflamed lymphatic endothelial cells enhances CCL21‐guided migration of dendritic cells. FASEB J 2012; 26:3380-92. [DOI: 10.1096/fj.11-201061] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Álvaro Teijeira
- Department of Oncology, Centre for Applied Medical ResearchUniversity of Navarra Pío XII Pamplona Spain
| | - Asís Palazón
- Hepathology and Gene Therapy Unit, Centre for Applied Medical ResearchUniversity of Navarra Pío XII Pamplona Spain
| | - Saray Garasa
- Department of Oncology, Centre for Applied Medical ResearchUniversity of Navarra Pío XII Pamplona Spain
| | - Diego Marré
- Department of Plastic Surgery, Clínica Universidad de NavarraUniversity of Navarra Pío XII Pamplona Spain
| | - Cristina Aubá
- Department of Plastic Surgery, Clínica Universidad de NavarraUniversity of Navarra Pío XII Pamplona Spain
| | - Anne Rogel
- Institut de Recherche Thérapeutique de l'Université de Nantes Nantes France
| | - Ohiana Murillo
- Hepathology and Gene Therapy Unit, Centre for Applied Medical ResearchUniversity of Navarra Pío XII Pamplona Spain
| | - Iván Martínez‐Forero
- Hepathology and Gene Therapy Unit, Centre for Applied Medical ResearchUniversity of Navarra Pío XII Pamplona Spain
| | - François Lang
- Institut de Recherche Thérapeutique de l'Université de Nantes Nantes France
| | - Ignacio Melero
- Hepathology and Gene Therapy Unit, Centre for Applied Medical ResearchUniversity of Navarra Pío XII Pamplona Spain
| | - Ana Rouzaut
- Department of Oncology, Centre for Applied Medical ResearchUniversity of Navarra Pío XII Pamplona Spain
| |
Collapse
|
28
|
Wang S, Lv J, Wang P, Yin X, Tan A, Chen Y. Recombinant human CD137L for cancer immunotherapy: effects of different fusions and linkers on its activity. Cancer Immunol Immunother 2012; 61:489-95. [PMID: 21968735 PMCID: PMC11029538 DOI: 10.1007/s00262-011-1097-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2011] [Accepted: 08/12/2011] [Indexed: 11/29/2022]
Abstract
Co-stimulatory molecules can be efficiently produced as a recombinant protein in E. coli with a large range of applications in the fields of immunotherapy. However, whether, different fusions that would affect their functions have rarely been analyzed. To explore the effects of different fusions and linkers on the molecular conformation and activity of CD137 ligand (CD137L), a recombinant human CD137L protein (rhCD137L) library, which consists of the entire extracellular domain of human CD137L fused to N- or C-terminal His-tag through different linkers, was constructed and all rhCD137Ls were, respectively, expressed in E. coli BL21 (DE3) strain carrying a chaperone plasmid pG-Tf2. After purification of the soluble rhCD137Ls, the recombinant fusion proteins could markedly promote the growth of activated T cells, but their effects on cytokine productions were different from each other. The present work indicated that, although all rhCD137Ls have desired biological activity, different fusions and linkers did affect their structures and functions. Consequently, rational design of a library is a necessary and feasible approach for fusion proteins in order to obtain a satisfactory drug candidate for further development.
Collapse
Affiliation(s)
- Shuzhen Wang
- Laboratory of Chemical Biology, China Pharmaceutical University, 24 Tongjia Street, Nanjing, Jiangsu Province 210009 People’s Republic of China
- Jiangsu Key Laboratory of Molecular Targeted Antitumor Drug Research, Nanjing, People’s Republic of China
| | - Junfang Lv
- Laboratory of Chemical Biology, China Pharmaceutical University, 24 Tongjia Street, Nanjing, Jiangsu Province 210009 People’s Republic of China
- Jiangsu Key Laboratory of Molecular Targeted Antitumor Drug Research, Nanjing, People’s Republic of China
| | - Peng Wang
- Jiangsu Simcere Pharmaceutical R&D Co., Ltd., No. 699-18, Xuanwu Avenue, Nanjing, Jiangsu Province 210042 People’s Republic of China
- Jiangsu Key Laboratory of Molecular Targeted Antitumor Drug Research, Nanjing, People’s Republic of China
| | - Xiaojin Yin
- Jiangsu Simcere Pharmaceutical R&D Co., Ltd., No. 699-18, Xuanwu Avenue, Nanjing, Jiangsu Province 210042 People’s Republic of China
- Jiangsu Key Laboratory of Molecular Targeted Antitumor Drug Research, Nanjing, People’s Republic of China
| | - Aimin Tan
- Jiangsu Simcere Pharmaceutical R&D Co., Ltd., No. 699-18, Xuanwu Avenue, Nanjing, Jiangsu Province 210042 People’s Republic of China
- Jiangsu Key Laboratory of Molecular Targeted Antitumor Drug Research, Nanjing, People’s Republic of China
| | - Yijun Chen
- Laboratory of Chemical Biology, China Pharmaceutical University, 24 Tongjia Street, Nanjing, Jiangsu Province 210009 People’s Republic of China
- Jiangsu Key Laboratory of Molecular Targeted Antitumor Drug Research, Nanjing, People’s Republic of China
| |
Collapse
|
29
|
Wang S, Tan A, Lv J, Wang P, Yin X, Chen Y. Soluble expression of recombinant human CD137 ligand in Escherichia coli by co-expression of chaperones. ACTA ACUST UNITED AC 2012; 39:471-6. [PMID: 21997275 DOI: 10.1007/s10295-011-1045-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2011] [Accepted: 09/29/2011] [Indexed: 12/31/2022]
Abstract
Abstract
CD137 ligand (CD137L) is a member of the tumor-necrosis factor superfamily that binds CD137 to provide positive co-stimulatory signals for T cells activation. Co-stimulation through CD137/CD137L has become one of the promising approaches for cancer therapy. Previous reports have shown that CD137L expressed in Escherichia coli resulted in inclusion bodies or low yield. In this study, the effects of five different chaperone teams on the soluble expression of recombinant human CD137L protein were explored and analyzed. The poor expression of CD137L in the cytoplasm of E. coli was improved significantly by co-expression of chaperone GroES-GroEL-Tf. After dual induction and affinity chromatography, purified recombinant CD137L was obtained at a yield of 3 mg protein per liter with purity greater than 98% from original undetectable level. Additionally, the purified recombinant CD137L could bind CD137-positive cells in a dose-dependent manner, markedly promote the growth of activated mice T cells, and elevate the release of IL-2. The present work provides an effective system for soluble expression of functional human co-stimulatory molecule CD137L, which will facilitate the clinical developments of recombinant protein drugs.
Collapse
Affiliation(s)
- Shuzhen Wang
- grid.254147.1 0000000097767793 Laboratory of Chemical Biology, China Pharmaceutical University 24 Tongjia Street 210009 Nanjing China
- Jiangsu Key Laboratory of Molecular Targeted Antitumor Drug Research 210009 Nanjing China
| | - Aimin Tan
- Jiangsu Simcere Pharmaceutical R&D Co., Ltd. No.699-18 Xuanwu Avenue 210042 Nanjing China
- Jiangsu Key Laboratory of Molecular Targeted Antitumor Drug Research 210009 Nanjing China
| | - Junfang Lv
- grid.254147.1 0000000097767793 Laboratory of Chemical Biology, China Pharmaceutical University 24 Tongjia Street 210009 Nanjing China
- Jiangsu Key Laboratory of Molecular Targeted Antitumor Drug Research 210009 Nanjing China
| | - Peng Wang
- Jiangsu Simcere Pharmaceutical R&D Co., Ltd. No.699-18 Xuanwu Avenue 210042 Nanjing China
- Jiangsu Key Laboratory of Molecular Targeted Antitumor Drug Research 210009 Nanjing China
| | - Xiaojin Yin
- Jiangsu Simcere Pharmaceutical R&D Co., Ltd. No.699-18 Xuanwu Avenue 210042 Nanjing China
- Jiangsu Key Laboratory of Molecular Targeted Antitumor Drug Research 210009 Nanjing China
| | - Yijun Chen
- grid.254147.1 0000000097767793 Laboratory of Chemical Biology, China Pharmaceutical University 24 Tongjia Street 210009 Nanjing China
- Jiangsu Key Laboratory of Molecular Targeted Antitumor Drug Research 210009 Nanjing China
| |
Collapse
|
30
|
Abstract
Costimulatory factors hold great promise for development into novel anticancer biotherapeutics. An agonist to 4-1BB is ranked number 8 by National Cancer Institute on the list of 20 agents with high potential for use in treating cancer. We earlier reported on a recombinant murine 4-1BB ligand fusion protein that binds 4-1BB receptor on murine T cells and stimulates their proliferation in tumor-bearing mice. To facilitate clinical translation,we constructed a corresponding recombinant human 4-1BB ligand fusion protein (hIg-h4-1BBLs) and showed its ability to activate human T cells in vitro. Using Chinese hamster ovary cells transformed with a plasmid coexpressing hIg-h4-1BBLs and rat glutamine synthetase, we generated a high-producing clone by sequential selection with methionine sulfoximine. The hIg-h4-1BBLs was partially purified by protein A column chromatography and characterized biochemically and functionally, using human 4-1BB binding and human T-cell proliferation assays, in vitro.Sodium dodecyl sulfate-polyacrylamide gel electrophoresis and Western Blot confirmed that the hIg-h4-1BBLs is expressed predominantly as a functionally active multimeric protein with the ability to specifically bind to cells expressing human 4-1BB receptor and induce significant T-cell proliferation in vitro using both human and monkey peripheral blood mononuclear cells. The hIg-h4-1BBLs can be produced in large quantities from the high producer clone and developed as a novel immune costimulatory biotherapeutic to treat, alone and in combination with other modalities, various malignant diseases in patients through T-cell activation. Process development of this clinical agent has been discussed with the Food and Drug Administration in a pre-Investigational New Drug meeting and presented to the Office of Biotechnology Activities in a public hearing.
Collapse
|
31
|
Sharma RK, Srivastava AK, Yolcu ES, MacLeod KJ, Schabowsky RH, Madireddi S, Shirwan H. SA-4-1BBL as the immunomodulatory component of a HPV-16 E7 protein based vaccine shows robust therapeutic efficacy in a mouse cervical cancer model. Vaccine 2010; 28:5794-802. [PMID: 20603135 DOI: 10.1016/j.vaccine.2010.06.073] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2010] [Revised: 06/21/2010] [Accepted: 06/24/2010] [Indexed: 12/23/2022]
Abstract
Cervical cancer is the leading cause of cancer-related deaths among women worldwide. Current prophylactic vaccines based on HPV (Human papillomavirus) late gene protein L1 are ineffective in therapeutic settings. Therefore, there is an acute need for the development of therapeutic vaccines for HPV associated cancers. The HPV E7 oncoprotein is expressed in cervical cancer and has been associated with the cellular transformation and maintenance of the transformed phenotype. As such, E7 protein represents an ideal target for the development of therapeutic subunit vaccines against cervical cancer. However, the low antigenicity of this protein may require potent adjuvants for therapeutic efficacy. We recently generated a novel chimeric form of the 4-1BBL costimulatory molecule engineered with core streptavidin (SA-4-1BBL) and demonstrated its safe and pleiotropic effects on various cells of the immune system. We herein tested the utility of SA-4-1BBL as the immunomodulatory component of HPV-16 E7 recombinant protein based therapeutic vaccine in the E7 expressing TC-1 tumor as a model of cervical cancer in mice. A single subcutaneous vaccination was effective in eradicating established tumors in approximately 70% of mice. The therapeutic efficacy of the vaccine was associated with robust primary and memory CD4(+) and CD8(+) T cell responses, Th1 cytokine response, infiltration of CD4(+) and CD8(+) T cells into the tumor, and enhanced NK cell killing. Importantly, NK cells played an important role in vaccine mediated therapy since their physical depletion compromised vaccine efficacy. Collectively, these data demonstrate the utility of SA-4-1BBL as a new class of multifunctional immunomodulator for the development of therapeutic vaccines against cancer and chronic infections.
Collapse
Affiliation(s)
- Rajesh K Sharma
- Institute for Cellular Therapeutics, Department of Microbiology and Immunology, University of Louisville, Louisville, KY 40202, USA
| | | | | | | | | | | | | |
Collapse
|
32
|
Sharma RK, Schabowsky RH, Srivastava AK, Elpek KG, Madireddi S, Zhao H, Zhong Z, Miller RW, Macleod KJ, Yolcu ES, Shirwan H. 4-1BB ligand as an effective multifunctional immunomodulator and antigen delivery vehicle for the development of therapeutic cancer vaccines. Cancer Res 2010; 70:3945-54. [PMID: 20406989 DOI: 10.1158/0008-5472.can-09-4480] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Therapeutic subunit vaccines based on tumor-associated antigens (TAA) represent an attractive approach for the treatment of cancer. However, poor immunogenicity of TAAs requires potent adjuvants for therapeutic efficacy. We recently proposed the tumor necrosis factor family costimulatory ligands as potential adjuvants for therapeutic vaccines and, hence, generated a soluble form of 4-1BBL chimeric with streptavidin (SA-4-1BBL) that has pleiotropic effects on cells of innate, adaptive, and regulatory immunity. We herein tested whether these effects can translate into effective cancer immunotherapy when SA-4-1BBL was also used as a vehicle to deliver TAAs in vivo to dendritic cells (DCs) constitutively expressing the 4-1BB receptor. SA-4-1BBL was internalized by DCs upon receptor binding and immunization with biotinylated antigens conjugated to SA-4-1BBL resulted in increased antigen uptake and cross-presentation by DCs, leading to the generation of effective T-cell immune responses. Conjugate vaccines containing human papillomavirus 16 E7 oncoprotein or survivin as a self-TAA had potent therapeutic efficacy against TC-1 cervical and 3LL lung carcinoma tumors, respectively. Therapeutic efficacy of the vaccines was associated with increased CD4(+) T and CD8(+) T-cell effector and memory responses and higher intratumoral CD8(+) T effector/CD4(+)CD25(+)Foxp3(+) T regulatory cell ratio. Thus, potent pleiotropic immune functions of SA-4-1BBL combined with its ability to serve as a vehicle to increase the delivery of antigens to DCs in vivo endow this molecule with the potential to serve as an effective immunomodulatory component of therapeutic vaccines against cancer and chronic infections.
Collapse
Affiliation(s)
- Rajesh K Sharma
- Department of Microbiology and Immunology, James Brown Cancer Center, Institute for Cellular Therapeutics, University of Louisville and ApoImmune, Inc, Louisville, Kentucky, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Won EY, Cha K, Byun JS, Kim DU, Shin S, Ahn B, Kim YH, Rice AJ, Walz T, Kwon BS, Cho HS. The structure of the trimer of human 4-1BB ligand is unique among members of the tumor necrosis factor superfamily. J Biol Chem 2010; 285:9202-10. [PMID: 20032458 PMCID: PMC2838339 DOI: 10.1074/jbc.m109.084442] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2009] [Indexed: 12/29/2022] Open
Abstract
Binding of the 4-1BB ligand (4-1BBL) to its receptor, 4-1BB, provides the T lymphocyte with co-stimulatory signals for survival, proliferation, and differentiation. Importantly, the 4-1BB-4-1BBL pathway is a well known target for anti-cancer immunotherapy. Here we present the 2.3-A crystal structure of the extracellular domain of human 4-1BBL. The ectodomain forms a homotrimer with an extended, three-bladed propeller structure that differs from trimers formed by other members of the tumor necrosis factor (TNF) superfamily. Based on the 4-1BBL structure, we modeled its complex with 4-1BB, which was consistent with images obtained by electron microscopy, and verified the binding site by site-directed mutagenesis. This structural information will facilitate the development of immunotherapeutics targeting 4-1BB.
Collapse
Affiliation(s)
- Eun-Young Won
- From the Department of Biology, Yonsei University, 134 Shinchon-dong, Seodaemun-gu, Seoul 120-749, Korea
| | - Kiweon Cha
- the Department of Biochemistry and Cell Biology/Advanced Medical Technology Cluster for Diagnosis and Prediction, School of Medicine, Kyungpook National University, Daegu 700-42, Korea
| | - Jung-Sue Byun
- From the Department of Biology, Yonsei University, 134 Shinchon-dong, Seodaemun-gu, Seoul 120-749, Korea
| | - Dong-Uk Kim
- From the Department of Biology, Yonsei University, 134 Shinchon-dong, Seodaemun-gu, Seoul 120-749, Korea
| | - Sumi Shin
- the Division of Cell and Immunobiology and R & D Center for Cancer Therapeutics, National Cancer Center, Ilsan, Goyang, Gyonggi-do 410-769, Korea
| | - Byungchan Ahn
- the Department of Biological Sciences, University of Ulsan, Ulsan 680-749, Korea, and
| | - Young Ho Kim
- the Division of Cell and Immunobiology and R & D Center for Cancer Therapeutics, National Cancer Center, Ilsan, Goyang, Gyonggi-do 410-769, Korea
| | | | - Thomas Walz
- the Department of Cell Biology and
- Howard Hughes Medical Institute, Harvard Medical School, Boston, Massachusetts 02115
| | - Byoung S. Kwon
- the Division of Cell and Immunobiology and R & D Center for Cancer Therapeutics, National Cancer Center, Ilsan, Goyang, Gyonggi-do 410-769, Korea
| | - Hyun-Soo Cho
- From the Department of Biology, Yonsei University, 134 Shinchon-dong, Seodaemun-gu, Seoul 120-749, Korea
| |
Collapse
|
34
|
Schabowsky RH, Elpek KG, Madireddi S, Sharma RK, Yolcu ES, Bandura-Morgan L, Miller R, MacLeod KJ, Mittler RS, Shirwan H. A novel form of 4-1BBL has better immunomodulatory activity than an agonistic anti-4-1BB Ab without Ab-associated severe toxicity. Vaccine 2009; 28:512-22. [PMID: 19836479 DOI: 10.1016/j.vaccine.2009.09.127] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2009] [Revised: 09/29/2009] [Accepted: 09/30/2009] [Indexed: 01/25/2023]
Abstract
Agonistic Abs to select costimulatory members of CD28 and TNFR family have shown efficacy in various preclinical cancer immunotherapeutic settings. However, the use of agonistic Abs is often associated with severe toxicity due to non-specific activation of lymphocytes. We hypothesized that natural costimulatory ligands may serve as more potent and safer alternative to agonistic Abs for immunotherapy. In this communication, we focused on 4-1BBL as the molecule of choice because of the pleiotropic effects of 4-1BB signaling in the immune system and the demonstrated therapeutic efficacy of 4-1BB agonistic Abs in preclinical cancer and infection models. We report that a novel form of soluble ligand, SA-4-1BBL, delivered more potent and qualitatively different signals to T cells than an agonistic Ab. Importantly, while treatment of naïve mice with the agonistic Ab resulted in severe toxicity, as assessed by enlarged spleen and peripheral LNs, non-specific T cell proliferation, hepatitis, and systemic inflammatory cytokine production, treatment with SA-4-1BBL lacked these immune anomalies. Agonistic Ab treatment produced full toxicity in FcgammaR(-/-) or complement C1q(-/-) or C3(-/-) knockout mice, suggesting lack of involvement of stimulatory FcgammaRs or complement system in the observed toxicity. Naïve and memory T cells served as direct targets of anti-4-1BB Ab-mediated toxicity. Potent immunostimulatory activity combined with lack of toxicity rationalizes further development of soluble SA-4-1BBL as an immunomodulatory component of therapeutic vaccines against cancer and chronic infections.
Collapse
Affiliation(s)
- Rich-Henry Schabowsky
- Institute for Cellular Therapeutics, Department of Microbiology and Immunology, and James Brown Cancer Center, University of Louisville, KY 40202, United States
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Wyzgol A, Müller N, Fick A, Munkel S, Grigoleit GU, Pfizenmaier K, Wajant H. Trimer Stabilization, Oligomerization, and Antibody-Mediated Cell Surface Immobilization Improve the Activity of Soluble Trimers of CD27L, CD40L, 41BBL, and Glucocorticoid-Induced TNF Receptor Ligand. THE JOURNAL OF IMMUNOLOGY 2009; 183:1851-61. [DOI: 10.4049/jimmunol.0802597] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
36
|
Sharma RK, Elpek KG, Yolcu ES, Schabowsky RH, Zhao H, Bandura-Morgan L, Shirwan H. Costimulation as a platform for the development of vaccines: a peptide-based vaccine containing a novel form of 4-1BB ligand eradicates established tumors. Cancer Res 2009; 69:4319-26. [PMID: 19435920 DOI: 10.1158/0008-5472.can-08-3141] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Vaccines represent an attractive treatment modality for the management of cancer primarily because of their specificity and generation of immunologic memory important for controlling recurrences. However, the efficacy of therapeutic vaccines may require formulations that not only generate effective immune responses but also overcome immune evasion mechanisms employed by progressing tumor. Costimulatory molecules play critical roles in modulating innate, adaptive, and regulatory immunity and have potential to serve as effective immunomodulatory components of therapeutic vaccines. In this study, we tested the function of a novel soluble form of 4-1BB ligand (4-1BBL) costimulatory molecule in modulating innate, adaptive, and regulatory immunity and assessed its therapeutic efficacy in the HPV-16 E7-expressing TC-1 cervical cancer and survivin-expressing 3LL lung carcinoma mouse models. Vaccination with 4-1BBL activated dendritic cells and enhanced antigen uptake, generated CD8(+) T-cell effector/memory responses, and endowed T effector cells refractory to suppression by CD4(+)CD25(+)FoxP3(+) T regulatory cells. Immunization with 4-1BBL in combination with an E7 peptide or survivin protein resulted in eradication of TC-1 and 3LL tumors, respectively. 4-1BBL was more effective than TLR agonists LPS, MPL, and CpG and an agonistic 4-1BB antibody as a component of E7 peptide-based therapeutic vaccine for the generation of immune responses and eradication of TC-1 established tumors in the absence of detectable toxicity. Therapeutic efficacy was associated with reversal of tumor-mediated nonresponsiveness/anergy as well as establishment of long-term CD8(+) T-cell memory. Potent pleiotropic immunomodulatory activities combined with lack of toxicity highlight the potential of 4-1BBL molecule as an effective component of therapeutic cancer vaccines.
Collapse
Affiliation(s)
- Rajesh K Sharma
- Institute for Cellular Therapeutics, Department of Microbiology and Immunology, and James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA
| | | | | | | | | | | | | |
Collapse
|
37
|
Abstract
Costimulation is an essential step in T-cell activation and hence, represents an important aspect in cancer immunotherapy. 4-1BB, a member of the tumor necrosis factor receptor family, has gained particular interest as a costimulatory molecule. Here, we investigated the potential of a targeted activation of 4-1BB-mediated costimulation at the tumor site by generating a recombinant antibody-cytokine fusion protein composed of a single-chain antibody fragment (scFv36) specific for the tumor stromal antigen fibroblast activation protein (FAP) and the extracellular domain of the 4-1BB ligand (4-1BBL). The scFv36-4-1BBL fusion protein is a homotrimeric molecule that binds specifically to FAP and the receptor 4-1BB. T-cell costimulation was demonstrated by interferon-gamma release of peripheral blood mononuclear cells cocultured with FAP-expressing HT1080 cells upon T-cell receptor triggering by monoclonal anti-CD3 antibody. Costimulatory activity of the scFv36-4-1BBL fusion protein was concentration dependent, ligand-specific, and substantially constrained to FAP-expressing target cell binding. Furthermore, scFv36-4-1BBL enhanced T-cell activation when the bispecific antibody scDb33CD3 (specific for FAP and CD3) was used as primary stimulus. Thus, target cell-dependent costimulation with scFv36-4-1BBL constitutes a new option to enhance T-cell activation by bispecific antibodies or antigen-dependent T-cell receptor triggering and should be useful to improve T cell-mediated antitumor responses.
Collapse
|
38
|
Lynch DH. The promise of 4-1BB (CD137)-mediated immunomodulation and the immunotherapy of cancer. Immunol Rev 2009; 222:277-86. [PMID: 18364008 DOI: 10.1111/j.1600-065x.2008.00621.x] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The continuing efforts in biomedical research to develop new therapies for cancer are entering an exciting new phase. Research over the past two to three decades has yielded a much more detailed understanding of the complexities of the cellular and molecular interactions involved in the generation and regulation of immune responses. We are also gaining insights into the mechanisms by which tumors evade or escape immune recognition and by which they become resistant to various existing chemotherapeutic and/or radiotherapeutic strategies. A clear conclusion that can be drawn from these studies is that effective treatments of cancer will become much more multifaceted and will include immunotherapeutic approaches. The identification and molecular cloning of genes encoding the receptors and ligands that play crucial roles in the generation and regulation of immune responses provides exciting new opportunities to induce and enhance effective endogenous immune responses to cancer. In this regard, the genes that comprise the tumor necrosis factor and tumor necrosis factor receptor superfamilies show particular promise. One receptor:ligand pair (4-1BB/CD137 and 4-1BBL/CD137L) is emerging as a target with important potential in its ability to enhance the generation of effective tumor-specific immune responses in situ. The results of the studies cited in this review highlight the potentials of 4-1BB-mediated immunotherapy.
Collapse
Affiliation(s)
- David H Lynch
- Bainbridge Biopharma Consulting, Bainbridge Island, WA, USA.
| |
Collapse
|
39
|
ProtEx technology for the generation of novel therapeutic cancer vaccines. Exp Mol Pathol 2009; 86:198-207. [PMID: 19454266 DOI: 10.1016/j.yexmp.2009.01.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2009] [Indexed: 01/15/2023]
Abstract
Therapeutic vaccines present an attractive alternative to conventional treatments for cancer. However, tumors have evolved various immune evasion mechanisms to modulate innate, adaptive, and regulatory immunity for survival. Therefore, successful vaccine formulations may require a non-toxic immunomodulator or adjuvant that not only induces/stimulates innate and adaptive tumor-specific immune responses, but also overcomes immune evasion mechanisms. Given the paramount role costimulation plays in modulating innate, adaptive, and regulatory immune responses, costimulatory ligands may serve as effective immunomodulating components of therapeutic cancer vaccines. Our laboratory has developed a novel technology designated as ProtEx that allows for the generation of recombinant costimulatory ligands with potent immunomodulatory activities and the display of these molecules on the cell surface in a rapid and efficient manner as a practical and safe alternative to gene therapy for immunomodulation. Importantly, the costimulatory ligands not only function when displayed on tumor cells, but also as soluble proteins that can be used as immunomodulatory components of conventional vaccine formulations containing tumor-associated antigens (TAAs). We herein discuss the application of the ProtEx technology to the development of effective cell-based as well as cell-free conventional therapeutic cancer vaccines.
Collapse
|
40
|
Schrama D, Hauschild A, Becker J. Immunmodulierende Antikörper in der Dermatoonkologie. Hautarzt 2008; 59:806-13. [DOI: 10.1007/s00105-008-1539-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
41
|
Müller N, Wyzgol A, Münkel S, Pfizenmaier K, Wajant H. Activity of soluble OX40 ligand is enhanced by oligomerization and cell surface immobilization. FEBS J 2008; 275:2296-304. [DOI: 10.1111/j.1742-4658.2008.06382.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
42
|
Olofsson PS, Söderström LA, Wågsäter D, Sheikine Y, Ocaya P, Lang F, Rabu C, Chen L, Rudling M, Aukrust P, Hedin U, Paulsson-Berne G, Sirsjö A, Hansson GK. CD137 Is Expressed in Human Atherosclerosis and Promotes Development of Plaque Inflammation in Hypercholesterolemic Mice. Circulation 2008; 117:1292-301. [DOI: 10.1161/circulationaha.107.699173] [Citation(s) in RCA: 173] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Background—
Atherosclerosis is a multifactorial disease in which inflammatory processes play an important role. Inflammation underlies lesion evolution at all stages, from establishment to plaque rupture and thrombosis. Costimulatory molecules of the tumor necrosis factor superfamily such as CD40/CD40L and OX40/OX40L have been implicated in atherosclerosis.
Methods and Results—
This study shows that the tumor necrosis factor superfamily members CD137 and CD137 ligand (CD137L), which play a major role in several autoimmune diseases, may constitute a pathogenic pair in atherogenesis. We detected CD137 protein in human atherosclerotic lesions not only on T cells but also on endothelial cells and showed that CD137 in cultured endothelial cells and smooth muscle cells was induced by proinflammatory cytokines implicated in atherosclerosis. Activation of CD137 by CD137L induced adhesion molecule expression on endothelial cells and reduced smooth muscle cell proliferation. In addition, treatment of atherosclerosis-prone apolipoprotein E–deficient mice with a CD137 agonist caused increased inflammation. T-cell infiltration, mainly of CD8
+
cells, and expression of the murine major histocompatibility complex class II molecule I-A
b
increased significantly in atherosclerotic lesions, as did the aortic expression of proinflammatory cytokines.
Conclusions—
Taken together, these observations suggest that CD137-CD137L interactions in the vasculature may contribute to the progression of atherosclerosis via augmented leukocyte recruitment, increased inflammation, and development of a more disease-prone phenotype.
Collapse
Affiliation(s)
- Peder S. Olofsson
- From the Center for Molecular Medicine (P.S.O., L.A.S., Y.S., U.H., G.P.-B., G.K.H.) and Center for Metabolism and Endocrinology (M.R.), Department of Medicine, and Department of Anesthesiology and Intensive Care Medicine (P.S.O.), Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden; Division of Biomedicine, Department of Clinical Medicine, University of Örebro, Örebro, Sweden (D.W., P.O., A.S.); Division of Nuclear Medicine/PET and Noninvasive Cardiovascular Imaging Program,
| | - Leif Å. Söderström
- From the Center for Molecular Medicine (P.S.O., L.A.S., Y.S., U.H., G.P.-B., G.K.H.) and Center for Metabolism and Endocrinology (M.R.), Department of Medicine, and Department of Anesthesiology and Intensive Care Medicine (P.S.O.), Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden; Division of Biomedicine, Department of Clinical Medicine, University of Örebro, Örebro, Sweden (D.W., P.O., A.S.); Division of Nuclear Medicine/PET and Noninvasive Cardiovascular Imaging Program,
| | - Dick Wågsäter
- From the Center for Molecular Medicine (P.S.O., L.A.S., Y.S., U.H., G.P.-B., G.K.H.) and Center for Metabolism and Endocrinology (M.R.), Department of Medicine, and Department of Anesthesiology and Intensive Care Medicine (P.S.O.), Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden; Division of Biomedicine, Department of Clinical Medicine, University of Örebro, Örebro, Sweden (D.W., P.O., A.S.); Division of Nuclear Medicine/PET and Noninvasive Cardiovascular Imaging Program,
| | - Yuri Sheikine
- From the Center for Molecular Medicine (P.S.O., L.A.S., Y.S., U.H., G.P.-B., G.K.H.) and Center for Metabolism and Endocrinology (M.R.), Department of Medicine, and Department of Anesthesiology and Intensive Care Medicine (P.S.O.), Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden; Division of Biomedicine, Department of Clinical Medicine, University of Örebro, Örebro, Sweden (D.W., P.O., A.S.); Division of Nuclear Medicine/PET and Noninvasive Cardiovascular Imaging Program,
| | - Pauline Ocaya
- From the Center for Molecular Medicine (P.S.O., L.A.S., Y.S., U.H., G.P.-B., G.K.H.) and Center for Metabolism and Endocrinology (M.R.), Department of Medicine, and Department of Anesthesiology and Intensive Care Medicine (P.S.O.), Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden; Division of Biomedicine, Department of Clinical Medicine, University of Örebro, Örebro, Sweden (D.W., P.O., A.S.); Division of Nuclear Medicine/PET and Noninvasive Cardiovascular Imaging Program,
| | - François Lang
- From the Center for Molecular Medicine (P.S.O., L.A.S., Y.S., U.H., G.P.-B., G.K.H.) and Center for Metabolism and Endocrinology (M.R.), Department of Medicine, and Department of Anesthesiology and Intensive Care Medicine (P.S.O.), Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden; Division of Biomedicine, Department of Clinical Medicine, University of Örebro, Örebro, Sweden (D.W., P.O., A.S.); Division of Nuclear Medicine/PET and Noninvasive Cardiovascular Imaging Program,
| | - Catherine Rabu
- From the Center for Molecular Medicine (P.S.O., L.A.S., Y.S., U.H., G.P.-B., G.K.H.) and Center for Metabolism and Endocrinology (M.R.), Department of Medicine, and Department of Anesthesiology and Intensive Care Medicine (P.S.O.), Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden; Division of Biomedicine, Department of Clinical Medicine, University of Örebro, Örebro, Sweden (D.W., P.O., A.S.); Division of Nuclear Medicine/PET and Noninvasive Cardiovascular Imaging Program,
| | - Lieping Chen
- From the Center for Molecular Medicine (P.S.O., L.A.S., Y.S., U.H., G.P.-B., G.K.H.) and Center for Metabolism and Endocrinology (M.R.), Department of Medicine, and Department of Anesthesiology and Intensive Care Medicine (P.S.O.), Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden; Division of Biomedicine, Department of Clinical Medicine, University of Örebro, Örebro, Sweden (D.W., P.O., A.S.); Division of Nuclear Medicine/PET and Noninvasive Cardiovascular Imaging Program,
| | - Mats Rudling
- From the Center for Molecular Medicine (P.S.O., L.A.S., Y.S., U.H., G.P.-B., G.K.H.) and Center for Metabolism and Endocrinology (M.R.), Department of Medicine, and Department of Anesthesiology and Intensive Care Medicine (P.S.O.), Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden; Division of Biomedicine, Department of Clinical Medicine, University of Örebro, Örebro, Sweden (D.W., P.O., A.S.); Division of Nuclear Medicine/PET and Noninvasive Cardiovascular Imaging Program,
| | - Pål Aukrust
- From the Center for Molecular Medicine (P.S.O., L.A.S., Y.S., U.H., G.P.-B., G.K.H.) and Center for Metabolism and Endocrinology (M.R.), Department of Medicine, and Department of Anesthesiology and Intensive Care Medicine (P.S.O.), Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden; Division of Biomedicine, Department of Clinical Medicine, University of Örebro, Örebro, Sweden (D.W., P.O., A.S.); Division of Nuclear Medicine/PET and Noninvasive Cardiovascular Imaging Program,
| | - Ulf Hedin
- From the Center for Molecular Medicine (P.S.O., L.A.S., Y.S., U.H., G.P.-B., G.K.H.) and Center for Metabolism and Endocrinology (M.R.), Department of Medicine, and Department of Anesthesiology and Intensive Care Medicine (P.S.O.), Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden; Division of Biomedicine, Department of Clinical Medicine, University of Örebro, Örebro, Sweden (D.W., P.O., A.S.); Division of Nuclear Medicine/PET and Noninvasive Cardiovascular Imaging Program,
| | - Gabrielle Paulsson-Berne
- From the Center for Molecular Medicine (P.S.O., L.A.S., Y.S., U.H., G.P.-B., G.K.H.) and Center for Metabolism and Endocrinology (M.R.), Department of Medicine, and Department of Anesthesiology and Intensive Care Medicine (P.S.O.), Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden; Division of Biomedicine, Department of Clinical Medicine, University of Örebro, Örebro, Sweden (D.W., P.O., A.S.); Division of Nuclear Medicine/PET and Noninvasive Cardiovascular Imaging Program,
| | - Allan Sirsjö
- From the Center for Molecular Medicine (P.S.O., L.A.S., Y.S., U.H., G.P.-B., G.K.H.) and Center for Metabolism and Endocrinology (M.R.), Department of Medicine, and Department of Anesthesiology and Intensive Care Medicine (P.S.O.), Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden; Division of Biomedicine, Department of Clinical Medicine, University of Örebro, Örebro, Sweden (D.W., P.O., A.S.); Division of Nuclear Medicine/PET and Noninvasive Cardiovascular Imaging Program,
| | - Göran K. Hansson
- From the Center for Molecular Medicine (P.S.O., L.A.S., Y.S., U.H., G.P.-B., G.K.H.) and Center for Metabolism and Endocrinology (M.R.), Department of Medicine, and Department of Anesthesiology and Intensive Care Medicine (P.S.O.), Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden; Division of Biomedicine, Department of Clinical Medicine, University of Örebro, Örebro, Sweden (D.W., P.O., A.S.); Division of Nuclear Medicine/PET and Noninvasive Cardiovascular Imaging Program,
| |
Collapse
|
43
|
Stephan MT, Ponomarev V, Brentjens RJ, Chang AH, Dobrenkov KV, Heller G, Sadelain M. T cell-encoded CD80 and 4-1BBL induce auto- and transcostimulation, resulting in potent tumor rejection. Nat Med 2007; 13:1440-9. [PMID: 18026115 DOI: 10.1038/nm1676] [Citation(s) in RCA: 222] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2007] [Accepted: 10/02/2007] [Indexed: 01/22/2023]
Abstract
To reject tumors, T cells must overcome poor tumor immunogenicity and an adverse tumor microenvironment. Providing agonistic costimulatory signals to tumor-infiltrating T cells to augment T cell function remains a challenge for the implementation of safe and effective immunotherapy. We hypothesized that T cells overexpressing selected costimulatory ligands could serve as cellular vehicles mediating powerful, yet constrained, anatomically targeted costimulation. Here, we show that primary human T cells expressing CD80 and 4-1BB ligand (4-1BBL) vigorously respond to tumor cells lacking costimulatory ligands and provoke potent rejection of large, systemic tumors in immunodeficient mice. In addition to showing costimulation of bystander T cells (transcostimulation), we show the effect of CD80 and 4-1BBL binding to their respective receptors in the immunological synapse of isolated single cells (autocostimulation). This new strategy of endowing T cells with constitutively expressed costimulatory ligands could be extended to other ligand-receptor pairs and used to enhance any targeted adoptive transfer therapy.
Collapse
Affiliation(s)
- Matthias T Stephan
- Center for Cell Engineering, Molecular Pharmacology and Chemistry Program, Memorial Sloan-Kettering Cancer Center (MSKCC), New York, New York 10021, USA
| | | | | | | | | | | | | |
Collapse
|
44
|
Elpek KG, Yolcu ES, Franke DDH, Lacelle C, Schabowsky RH, Shirwan H. Ex Vivo Expansion of CD4+CD25+FoxP3+ T Regulatory Cells Based on Synergy between IL-2 and 4-1BB Signaling. THE JOURNAL OF IMMUNOLOGY 2007; 179:7295-304. [DOI: 10.4049/jimmunol.179.11.7295] [Citation(s) in RCA: 108] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
45
|
Morris NP, Peters C, Montler R, Hu HM, Curti BD, Urba WJ, Weinberg AD. Development and characterization of recombinant human Fc:OX40L fusion protein linked via a coiled-coil trimerization domain. Mol Immunol 2007; 44:3112-21. [PMID: 17374396 PMCID: PMC1950941 DOI: 10.1016/j.molimm.2007.02.004] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2007] [Revised: 02/06/2007] [Accepted: 02/07/2007] [Indexed: 01/28/2023]
Abstract
OX40 (CD134) is a potent costimulatory molecule found on the surface of activated CD4(+) and CD8(+) T cells. Immunotherapy with OX40 agonists administered in vivo has demonstrated efficacy in several murine tumor models. A phase I clinical trial is currently underway in patients with advanced cancer using a mouse anti-CD134 monoclonal antibody. Therapy with this antibody will likely be limited to one cycle because patients develop neutralizing human anti-mouse antibody (HAMA). Therefore, we developed a humanized OX40 agonist that links the extracellular domain of human OX40L to the Fc domain of human IgG(1) via a trimerizing isoleucine zipper domain (ILZ). Physical characterization by velocity sedimentation revealed that this novel construct, hFcILZOX40L, was assembled into hexamers in which the Fc domains formed three disulfide-bonded dimers and the ILZ-OX40L domains formed two trimers. Trimerization of the ILZ domain was necessary to achieve appropriate assembly. In vitro biologic activity of the hFcILZOX40L hexamer was equivalent to the activity of agonist antibodies in plate-bound assays and was superior when the agonists were tested as soluble agents. Our ultimate goal is to use this recombinant molecule in a future clinical trial, and we feel that the OX40L hexamer will have equivalent or superior agonist activity in vivo when compared to an anti-OX40 antibody.
Collapse
|
46
|
Abstract
We identified 1113 articles (103 reviews, 1010 primary research articles) published in 2005 that describe experiments performed using commercially available optical biosensors. While this number of publications is impressive, we find that the quality of the biosensor work in these articles is often pretty poor. It is a little disappointing that there appears to be only a small set of researchers who know how to properly perform, analyze, and present biosensor data. To help focus the field, we spotlight work published by 10 research groups that exemplify the quality of data one should expect to see from a biosensor experiment. Also, in an effort to raise awareness of the common problems in the biosensor field, we provide side-by-side examples of good and bad data sets from the 2005 literature.
Collapse
Affiliation(s)
- Rebecca L Rich
- Center for Biomolecular Interaction Analysis, University of Utah, Salt Lake City, UT 84132, USA
| | | |
Collapse
|