Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Discov ; 13(3): 552-569, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36576322

ABSTRACT

Twenty-five years ago, we reported that agonist anti-CD137 monoclonal antibodies eradicated transplanted mouse tumors because of enhanced CD8+ T-cell antitumor immunity. Mouse models indicated that anti-CD137 agonist antibodies synergized with various other therapies. In the clinic, the agonist antibody urelumab showed evidence for single-agent activity against melanoma and non-Hodgkin lymphoma but caused severe liver inflammation in a fraction of the patients. CD137's signaling domain is included in approved chimeric antigen receptors conferring persistence and efficacy. A new wave of CD137 agonists targeting tumors, mainly based on bispecific constructs, are in early-phase trials and are showing promising safety and clinical activity. SIGNIFICANCE: CD137 (4-1BB) is a costimulatory receptor of T and natural killer lymphocytes whose activity can be exploited in cancer immunotherapy strategies as discovered 25 years ago. Following initial attempts that met unacceptable toxicity, new waves of constructs acting agonistically on CD137 are being developed in patients, offering signs of clinical and pharmacodynamic activity with tolerable safety profiles.


Subject(s)
Anniversaries and Special Events , Neoplasms , Animals , Mice , Neoplasms/drug therapy , Immunotherapy , Tumor Necrosis Factor Receptor Superfamily, Member 9/therapeutic use , CD8-Positive T-Lymphocytes , Signal Transduction
2.
Clin Cancer Res ; 27(11): 3167-3177, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33785484

ABSTRACT

PURPOSE: The induction of 4-1BB signaling by agonistic antibodies can drive the activation and proliferation of effector T cells and thereby enhance a T-cell-mediated antitumor response. Systemic administration of anti-4-1BB-agonistic IgGs, although effective preclinically, has not advanced in clinical development due to their severe hepatotoxicity. EXPERIMENTAL DESIGN: Here, we generated a humanized EGFR-specific 4-1BB-agonistic trimerbody, which replaces the IgG Fc region with a human collagen homotrimerization domain. It was characterized by structural analysis and in vitro functional studies. We also assessed pharmacokinetics, antitumor efficacy, and toxicity in vivo. RESULTS: In the presence of a T-cell receptor signal, the trimerbody provided potent T-cell costimulation that was strictly dependent on 4-1BB hyperclustering at the point of contact with a tumor antigen-displaying cell surface. It exhibits significant antitumor activity in vivo, without hepatotoxicity, in a wide range of human tumors including colorectal and breast cancer cell-derived xenografts, and non-small cell lung cancer patient-derived xenografts associated with increased tumor-infiltrating CD8+ T cells. The combination of the trimerbody with a PD-L1 blocker led to increased IFNγ secretion in vitro and resulted in tumor regression in humanized mice bearing aggressive triple-negative breast cancer. CONCLUSIONS: These results demonstrate the nontoxic broad antitumor activity of humanized Fc-free tumor-specific 4-1BB-agonistic trimerbodies and their synergy with checkpoint blockers, which may provide a way to elicit responses in most patients with cancer while avoiding Fc-mediated adverse reactions.


Subject(s)
Breast Neoplasms/pathology , Breast Neoplasms/therapy , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/therapy , ErbB Receptors , Immunotherapy/methods , Lung Neoplasms/pathology , Lung Neoplasms/therapy , Tumor Necrosis Factor Receptor Superfamily, Member 9/therapeutic use , Animals , Breast Neoplasms/immunology , Carcinoma, Non-Small-Cell Lung/immunology , Cell Line , Disease Models, Animal , Female , Lung Neoplasms/immunology , Lymphocyte Activation/genetics , Lymphocyte Activation/physiology , Mice, Transgenic , T-Lymphocytes/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism
3.
Front Immunol ; 10: 2566, 2019.
Article in English | MEDLINE | ID: mdl-31787971

ABSTRACT

We show here that soluble CD137 (sCD137), the alternately spliced gene product of Tnfsfr9, effectively treats acute type 1 diabetes (T1D) in nonobese diabetic (NOD) mice. sCD137 significantly delayed development of end-stage disease, preserved insulin+ islet beta cells, and prevented progression to end-stage T1D in some mice. We demonstrate that sCD137 induces CD4+ T cell anergy, suppressing antigen-specific T cell proliferation and IL-2/IFN-γ secretion. Exogenous IL-2 reversed the sCD137 anergy effect. sCD137 greatly reduces inflammatory cytokine production by CD8 effector memory T cells, critical mediators of beta cell damage. We demonstrate that human T1D patients have decreased serum sCD137 compared to age-matched controls (as do NOD mice compared to NOD congenic mice expressing a protective Tnfsfr9 allele), that human sCD137 is secreted by regulatory T cells (Tregs; as in mice), and that human sCD137 induces T cell suppression in human T cells. These findings provide a rationale for further investigation of sCD137 as a treatment for T1D and other T cell-mediated autoimmune diseases.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Clonal Anergy/immunology , Diabetes Mellitus, Type 1/therapy , Tumor Necrosis Factor Receptor Superfamily, Member 9/therapeutic use , Animals , Cell Cycle , Cytokines/metabolism , Diabetes Mellitus, Type 1/immunology , Female , Immunologic Memory , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Interleukin-2/immunology , Lymphocyte Activation , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Mice, Inbred NOD , Signal Transduction , T-Lymphocyte Subsets/metabolism , Tumor Necrosis Factor Receptor Superfamily, Member 9/antagonists & inhibitors , Tumor Necrosis Factor Receptor Superfamily, Member 9/genetics , Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology
5.
Tumour Biol ; 39(3): 1010428317695924, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28347235

ABSTRACT

The 4-1BB is a surface glycoprotein that pertains to the tumor necrosis factor-receptor family. There is compelling evidence suggesting important roles for 4-1BB in the immune response, including cell activation and proliferation and also cytokine induction. Because of encouraging results of different agonistic monoclonal antibodies against 4-1BB in the treatment of cancer, infectious, and autoimmune diseases, 4-1BB has been suggested as an attractive target for immunotherapy. In this study, single chain variable fragment phage display libraries, Tomlinson I+J, were screened against specific synthetic oligopeptides (peptides I and II) designed from 4-1BB extracellular domain. Five rounds of panning led to selection of four 4-1BB specific single chain variable fragments (PI.12, PI.42, PII.16, and PII.29) which showed specific reaction to relevant peptides in phage enzyme-linked immunosorbent assay. The selected clones were successfully expressed in Escherichia coli Rosetta-gami 2, and their expression was confirmed by western blot analysis. Enzyme-linked immunosorbent assay experiments indicated that these antibodies were able to specifically recognize 4-1BB without any cross-reactivity with other antigens. Flow cytometry analysis demonstrated an acceptable specific binding of the single chain variable fragments to 4-1BB expressed on CCRF-CEM cells, while no binding was observed with an irrelevant antibody. Anti-4-1BB single chain variable fragments enhanced surface CD69 expression and interleukin-2 production in stimulated CCRF-CEM cells which confirmed the agonistic effect of the selected single chain variable fragments. The data from this study have provided a rationale for further experiments involving the biological functions of anti-4-1BB single chain variable fragments in future studies.


Subject(s)
Antibodies, Anti-Idiotypic/immunology , Immunotherapy , Leukemia/immunology , Single-Chain Antibodies/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology , Antibodies, Anti-Idiotypic/therapeutic use , Antigens, CD/biosynthesis , Antigens, Differentiation, T-Lymphocyte/biosynthesis , Antigens, Neoplasm/immunology , Cell Line, Tumor , Escherichia coli/genetics , Flow Cytometry , Humans , Immunity, Innate , Interleukin-2/biosynthesis , Lectins, C-Type/biosynthesis , Leukemia/therapy , Peptide Library , Peptides/immunology , Peptides/therapeutic use , Protein Domains/immunology , Single-Chain Antibodies/isolation & purification , Single-Chain Antibodies/therapeutic use , Tumor Necrosis Factor Receptor Superfamily, Member 9/isolation & purification , Tumor Necrosis Factor Receptor Superfamily, Member 9/therapeutic use
6.
Hum Gene Ther ; 28(5): 437-448, 2017 05.
Article in English | MEDLINE | ID: mdl-27530312

ABSTRACT

T cells engineered to express CD19-specific chimeric antigen receptors (CARs) have shown breakthrough clinical successes in patients with B-cell lymphoid malignancies. However, similar therapeutic efficacy of CAR T cells in solid tumors is yet to be achieved. In this study we systematically evaluated a series of CAR constructs targeting glypican-3 (GPC3), which is selectively expressed on several solid tumors. We compared GPC3-specific CARs that encoded CD3ζ (Gz) alone or with costimulatory domains derived from CD28 (G28z), 4-1BB (GBBz), or CD28 and 4-1BB (G28BBz). All GPC3-CARs rendered T cells highly cytotoxic to GPC3-positive hepatocellular carcinoma, hepatoblastoma, and malignant rhabdoid tumor cell lines in vitro. GBBz induced the preferential production of Th1 cytokines (interferon γ/granulocyte macrophage colony-stimulating factor) while G28z preferentially induced Th2 cytokines (interleukin-4/interleukin-10). Inclusion of 4-1BB in G28BBz could only partially ameliorate the Th2-polarizing effect of CD28. 4-1BB induced superior expansion of CAR T cells in vitro and in vivo. T cells expressing GPC3-CARs incorporating CD28, 4-1BB, or both induced sustained tumor regressions in two xenogeneic tumor models. Thus, GBBz CAR endows T cells with superior proliferative potential, potent antitumor activity, and a Th1-biased cytokine profile, justifying further clinical development of GBBz CAR for immunotherapy of GPC3-positive solid tumors.


Subject(s)
CD28 Antigens/genetics , Glypicans/genetics , Lymphoma, B-Cell/therapy , Receptors, Antigen, T-Cell/genetics , Tumor Necrosis Factor Receptor Superfamily, Member 9/genetics , Animals , Antigens, CD19/genetics , Antigens, CD19/therapeutic use , CD28 Antigens/therapeutic use , Cell Polarity/immunology , Glypicans/therapeutic use , Humans , Immunotherapy , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/immunology , Mice , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/therapeutic use , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes, Cytotoxic/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 9/therapeutic use , Xenograft Model Antitumor Assays
7.
Cancer J ; 20(2): 134-40, 2014.
Article in English | MEDLINE | ID: mdl-24667959

ABSTRACT

In addition to T-cell receptor signals, T lymphocytes require costimulatory signals for robust activation. Among these, those mediated by 4-1BB (CD137, TNFRSF9) are critical for tumor immunity. 4-1BB is expressed in T-cell receptor-activated lymphocytes as well as natural killer cells and other hematopoietic and nonhematopoietic cells. 4-1BB ligation induces a signaling cascade that results in cytokine production, expression of antiapoptotic molecules, and enhanced immune responses. In line with the described function of 4-1BB, its addition to CD3ζ chimeric antigen receptors (CARs) increases their capacity to provoke T-cell expansion and antitumor activity. The results of preclinical studies with 4-1BB CARs have been corroborated by encouraging results from clinical trials. Advantages and disadvantages of 4-1BB CARs versus CARs bearing other costimulatory components remain to be fully elucidated. In this review, we discuss the properties of 4-1BB, the design of 4-1BB CARs, and the function of T lymphocytes and natural killer cells expressing them.


Subject(s)
Immunotherapy, Adoptive , Neoplasms/therapy , Receptors, Antigen, T-Cell/therapeutic use , Tumor Necrosis Factor Receptor Superfamily, Member 9/therapeutic use , CD8-Positive T-Lymphocytes/immunology , Humans , Lymphocyte Activation/immunology , Neoplasms/immunology , Neoplasms/pathology , Receptors, Antigen, T-Cell/immunology , Signal Transduction/genetics , Signal Transduction/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology
8.
Cytotherapy ; 14(7): 830-40, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22458956

ABSTRACT

BACKGROUND AIMS: Retroviral transduction of anti-CD19 chimeric antigen receptors significantly enhances the cytotoxicity of natural killer (NK) cells against B-cell malignancies. We aimed to validate a more practical, affordable and safe method for this purpose. METHODS: We tested the expression of a receptor containing CD3ζ and 4-1BB signaling molecules (anti-CD19-BB-ζ) in human NK cells after electroporation with the corresponding mRNA using a clinical-grade electroporator. The cytotoxic capacity of the transfected NK cells was tested in vitro and in a mouse model of leukemia. RESULTS: Median anti-CD19-BB-ζ expression 24 h after electroporation was 40.3% in freshly purified (n =18) and 61.3% in expanded (n = 31) NK cells; median cell viability was 90%. NK cells expressing anti-CD19-BB-ζ secreted interferon (IFN)-γ in response to CD19-positive target cells and had increased cytotoxicity. Receptor expression was detectable 6 h after electroporation, reaching maximum levels at 24-48 h; specific anti-CD19 cytotoxicity was observed at 96 h. Levels of expression and cytotoxicities were comparable with those achieved by retroviral transduction. A large-scale protocol was developed and applied to expanded NK cells (median NK cell number 2.5 × 10(8), n = 12). Median receptor expression after 24 h was 82.0%; NK cells transfected under these conditions exerted considerable cytotoxicity in xenograft models of B-cell leukemia. CONCLUSIONS: The method described here represents a practical way to augment the cytotoxicity of NK cells against B-cell malignancies. It has the potential to be extended to other targets beyond CD19 and should facilitate the clinical use of redirected NK cells for cancer therapy.


Subject(s)
Antigens, CD19 , Cell- and Tissue-Based Therapy , Killer Cells, Natural/cytology , Lymphoma, Non-Hodgkin , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Animals , Antigens, CD19/genetics , Antigens, CD19/immunology , Antigens, CD19/therapeutic use , CD3 Complex/genetics , CD3 Complex/therapeutic use , Cell Line, Tumor , Cytotoxicity, Immunologic/genetics , Gene Expression Regulation, Leukemic , Humans , Killer Cells, Natural/metabolism , Lymphocyte Activation , Lymphoma, Non-Hodgkin/therapy , Mice , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , RNA, Messenger/genetics , RNA, Messenger/therapeutic use , Tumor Necrosis Factor Receptor Superfamily, Member 9/genetics , Tumor Necrosis Factor Receptor Superfamily, Member 9/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...