Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.503
Filter
1.
MAbs ; 16(1): 2373325, 2024.
Article in English | MEDLINE | ID: mdl-38962811

ABSTRACT

T-cell engaging (TCE) bispecific antibodies are potent drugs that trigger the immune system to eliminate cancer cells, but administration can be accompanied by toxic side effects that limit dosing. TCEs function by binding to cell surface receptors on T cells, frequently CD3, with one arm of the bispecific antibody while the other arm binds to cell surface antigens on cancer cells. On-target, off-tumor toxicity can arise when the target antigen is also present on healthy cells. The toxicity of TCEs may be ameliorated through the use of pro-drug forms of the TCE, which are not fully functional until recruited to the tumor microenvironment. This can be accomplished by masking the anti-CD3 arm of the TCE with an autoinhibitory motif that is released by tumor-enriched proteases. Here, we solve the crystal structure of the antigen-binding fragment of a novel anti-CD3 antibody, E10, in complex with its epitope from CD3 and use this information to engineer a masked form of the antibody that can activate by the tumor-enriched protease matrix metalloproteinase 2 (MMP-2). We demonstrate with binding experiments and in vitro T-cell activation and killing assays that our designed prodrug TCE is capable of tumor-selective T-cell activity that is dependent upon MMP-2. Furthermore, we demonstrate that a similar masking strategy can be used to create a pro-drug form of the frequently used anti-CD3 antibody SP34. This study showcases an approach to developing immune-modulating therapeutics that prioritizes safety and has the potential to advance cancer immunotherapy treatment strategies.


Subject(s)
Antibodies, Bispecific , CD3 Complex , Immunotherapy , Prodrugs , T-Lymphocytes , Antibodies, Bispecific/immunology , Antibodies, Bispecific/pharmacology , Humans , CD3 Complex/immunology , Immunotherapy/methods , T-Lymphocytes/immunology , Prodrugs/pharmacology , Prodrugs/chemistry , Neoplasms/immunology , Neoplasms/therapy , Neoplasms/drug therapy , Protein Engineering/methods , Matrix Metalloproteinase 2/immunology
2.
Front Immunol ; 15: 1386132, 2024.
Article in English | MEDLINE | ID: mdl-38873603

ABSTRACT

The expression levels of TCRs on the surface of human T cells define the avidity of TCR-HLA/peptide interactions. In this study, we have explored which components of the TCR-CD3 complex are involved in determining the surface expression levels of TCRs in primary human T cells. The results show that there is a surplus of endogenous TCR α/ß chains that can be mobilised by providing T cells with additional CD3γ,δ,ε,ζ chains, which leads to a 5-fold increase in TCR α/ß surface expression. The analysis of individual CD3 chains revealed that provision of additional ζ chain alone was sufficient to achieve a 3-fold increase in endogenous TCR expression. Similarly, CD3ζ also limits the expression levels of exogenous TCRs transduced into primary human T cells. Interestingly, transduction with TCR plus CD3ζ not only increased surface expression of the introduced TCR, but it also reduced mispairing with endogenous TCR chains, resulting in improved antigen-specific function. TCR reconstitution experiments in HEK293T cells that do not express endogenous TCR or CD3 showed that TCRα/ß and all four CD3 chains were required for optimal surface expression, while in the absence of CD3ζ the TCR expression was reduced by 50%. Together, the data show that CD3ζ is a key regulator of TCR expression levels in human T cells, and that gene transfer of exogenous TCR plus CD3ζ improved TCR surface expression, reduced TCR mispairing and increased antigen-specific function.


Subject(s)
CD3 Complex , Humans , CD3 Complex/immunology , CD3 Complex/metabolism , CD3 Complex/genetics , HEK293 Cells , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Receptors, Antigen, T-Cell, alpha-beta/immunology , Lymphocyte Activation/immunology , Receptor-CD3 Complex, Antigen, T-Cell/immunology , Receptor-CD3 Complex, Antigen, T-Cell/metabolism , Receptor-CD3 Complex, Antigen, T-Cell/genetics
3.
Front Immunol ; 15: 1376096, 2024.
Article in English | MEDLINE | ID: mdl-38863707

ABSTRACT

Bispecific T-cell-engaging antibodies are a growing class of therapeutics with numerous molecules being tested in clinical trials and, currently, seven of them have received market approval. They are structurally complex and function as adaptors to redirect the cytotoxicity of T cells to kill tumor cells. T-cell-engaging bispecific antibodies can be generally divided into two categories: IgG/IgG-like and non-IgG-like formats. Different formats may have different intrinsic potencies and physiochemical properties, and comprehensive studies are needed to gain a better understanding of how the differences in formats impact on structural and functional characteristics. In this study, we designed and generated bispecific T-cell-engaging antibodies with IgG-like (DVD-Ig) and non-IgG (BiTE) formats. Both target the same pair of antigens (EGFR and CD3) to minimize the possible influence of targets on functional characterization. We performed a side-by-side comparison to assess differences in the physiochemical and biological properties of these two bispecific T-cell-engaging antibodies using a variety of breast and ovarian cancer cell-based functional assays to delineate the structural-functional relationships and anti-tumor activities/potency. We found that the Fc portion of T-cell-engaging bispecific antibodies can significantly impact antigen binding activity, potency, and stability in addition to eliciting different mechanisms of action that contribute the killing of cancer cells.


Subject(s)
Antibodies, Bispecific , Immunoglobulin G , T-Lymphocytes , Antibodies, Bispecific/pharmacology , Antibodies, Bispecific/immunology , Humans , Immunoglobulin G/immunology , T-Lymphocytes/immunology , CD3 Complex/immunology , Cell Line, Tumor , ErbB Receptors/immunology , Female , Breast Neoplasms/immunology , Breast Neoplasms/therapy , Ovarian Neoplasms/immunology , Ovarian Neoplasms/therapy
4.
Int Immunopharmacol ; 137: 112424, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-38878486

ABSTRACT

Colorectal cancer is a major global health burden, with limited efficacy of traditional treatment modalities in improving survival rates. However, recently advances in immunotherapy has improved treatment outcomes for patients with this cancer. To address the continuing need for improved treatment efficacy, this study introduced a novel tri-specific antibody, IMT030122, that targets EpCAM, 4-1BB, and CD3. We evaluated the pharmacological efficacy and mechanism of action of IMT030122 in vitro and in vivo. In in vitro studies, IMT030122 exhibited differential binding to antigens and cells expressing EpCAM, 4-1BB, and CD3. Moreover, IMT030122 relied on EpCAM-targeted activation of intracellular CD3 and 4-1BB signaling and mediated T cell cytotoxicity specific to HCT116 colorectal cancer cells. In vivo, IMT030122 demonstrated potent anti-tumor activity, significantly inhibiting the growth of colon cancer HCT116 and MC38-hEpCAM subcutaneous grafts. Further pharmacological analysis revealed that IMT030122 recruited lymphocytes from peripheral blood into colorectal cancer tissue and exerted durable anti-tumor activity, predominantly by promoting the activation, proliferation, and differentiation of CD8T cells. Notably, IMT030122 still exhibited anti-tumor efficacy even in the presence of significantly depleted lymphocytes in colorectal cancer tissue. The potent pharmacological activity and anti-tumor effects of IMT030122 suggest it may enhance treatment efficacy and substantially extend the survival of patients with colorectal cancer in the future.


Subject(s)
CD3 Complex , Colorectal Neoplasms , Epithelial Cell Adhesion Molecule , Animals , Humans , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/immunology , Colorectal Neoplasms/therapy , Epithelial Cell Adhesion Molecule/metabolism , CD3 Complex/immunology , Mice , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism , HCT116 Cells , Xenograft Model Antitumor Assays , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Agents, Immunological/therapeutic use , Female , Cell Line, Tumor , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Mice, Inbred BALB C , T-Lymphocytes/immunology , T-Lymphocytes/drug effects , Antibodies, Bispecific/pharmacology , Antibodies, Bispecific/therapeutic use , Immunotherapy/methods
5.
Front Immunol ; 15: 1415834, 2024.
Article in English | MEDLINE | ID: mdl-38933272

ABSTRACT

Introduction: Bispecific antibodies (BsAbs) can simultaneously target two epitopes of different antigenic targets, bringing possibilities for diversity in antibody drug design and are promising tools for the treatment of cancers and other diseases. T-cell engaging bsAb is an important application of the bispecific antibody, which could promote T cell-mediated tumor cell killing by targeting tumor-associated antigen (TAA) and CD3 at the same time. Methods: This study comprised antibodies purification, Elisa assay for antigen binding, cytotoxicity assays, T cell activation by flow cytometry in vitro and xenogenic tumor model in vivo. Results: We present a novel bsAb platform named PHE-Ig technique to promote cognate heavy chain (HC)-light chain (LC) pairing by replacing the CH1/CL regions of different monoclonal antibodies (mAbs) with the natural A and B chains of PHE1 fragment of Integrin ß2 based on the knob-in-hole (KIH) technology. We had also verified that PHE-Ig technology can be effectively used as a platform to synthesize different desired bsAbs for T-cell immunotherapy. Especially, BCMA×CD3 PHE-Ig bsAbs exhibited robust anti-multiple myeloma (MM) activity in vitro and in vivo. Discussion: Moreover, PHE1 domain was further shortened with D14G and R41S mutations, named PHE-S, and the PHE-S-based BCMA×CD3 bsAbs also showed anti BCMA+ tumor effect in vitro and in vivo, bringing more possibilities for the development and optimization of different bsAbs. To sum up, PHE1-based IgG-like antibody platform for bsAb construction provides a novel strategy for enhanced T-cell immunotherapy.


Subject(s)
Antibodies, Bispecific , T-Lymphocytes , Antibodies, Bispecific/immunology , Animals , Humans , T-Lymphocytes/immunology , Mice , Immunoglobulin G/immunology , Immunotherapy/methods , Cell Line, Tumor , Multiple Myeloma/immunology , Multiple Myeloma/therapy , Xenograft Model Antitumor Assays , Lymphocyte Activation/immunology , CD3 Complex/immunology , Antigens, Neoplasm/immunology
6.
Dev Comp Immunol ; 158: 105210, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38844187

ABSTRACT

Interleukin (IL) 20 is a multifunctional cytokine and plays a vital role in regulating autoimmune diseases, inflammation, and immune responses. IL-20 homologs have been described in fish. However, due to the lack of antibodies, cellular sources and immunological functions of fish IL-20 in response to infections have not been fully characterized. In this study, a monoclonal antibody (mAb) was generated against the recombinant grass carp (Ctenopharyngodon idella) IL-20 protein and characterized by immunoblotting, immunofluorescent microscopy and flow cytometry. It was shown that the IL-20 mAb specifically recognized recombinant IL-20 proteins expressed in the E. coli cells and HEK293 cells. Using confocal microscopy, the IL-20+ cells were identified in the head kidney, gills and intestine of grass carp, and induced after infection with Aeromonas hydrophila. Moreover, the IL-20 protein was found to be secreted mainly by CD3γδ T cells which were located predominantly in the gill filaments and intestinal mucosa. Taken together, our results suggest that IL-20 producing T cells are required for the mucosal immunity against bacterial infection in fish.


Subject(s)
Aeromonas hydrophila , Carps , Fish Diseases , Fish Proteins , Gram-Negative Bacterial Infections , Immunity, Mucosal , Interleukins , Animals , Carps/immunology , Carps/microbiology , Aeromonas hydrophila/immunology , Gram-Negative Bacterial Infections/immunology , Gram-Negative Bacterial Infections/veterinary , Fish Diseases/immunology , Fish Diseases/microbiology , Fish Proteins/immunology , Fish Proteins/metabolism , Fish Proteins/genetics , Humans , Interleukins/metabolism , Interleukins/immunology , HEK293 Cells , Gills/immunology , Gills/metabolism , CD3 Complex/immunology , CD3 Complex/metabolism , Antibodies, Monoclonal/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , T-Lymphocytes/immunology , Mucous Membrane/immunology
8.
Nat Commun ; 15(1): 4091, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38750034

ABSTRACT

Cibisatamab is a bispecific antibody-based construct targeting carcinoembryonic antigen (CEA) on tumour cells and CD3 epsilon chain as a T-cell engager. Here we evaluated cibisatamab for advanced CEA-positive solid tumours in two open-label Phase 1 dose-escalation and -expansion studies: as a single agent with or without obinutuzumab in S1 (NCT02324257) and with atezolizumab in S2 (NCT02650713). Primary endpoints were safety, dose finding, and pharmacokinetics in S1; safety and dose finding in S2. Secondary endpoints were anti-tumour activity (including overall response rate, ORR) and pharmacodynamics in S1; anti-tumour activity, pharmacodynamics and pharmacokinetics in S2. S1 and S2 enrolled a total of 149 and 228 patients, respectively. Grade ≥3 cibisatamab-related adverse events occurred in 36% of S1 and 49% of S2 patients. The ORR was 4% in S1 and 7% in S2. In S2, patients with microsatellite stable colorectal carcinoma (MSS-CRC) given flat doses of cibisatamab and atezolizumab demonstrated an ORR of 14%. In S1 and S2, 40% and 52% of patients, respectively, developed persistent anti-drug antibodies (ADAs). ADA appearance could be mitigated by obinutuzumab-pretreatment, with 8% of patients having persistent ADAs. Overall, cibisatamab warrants further exploration in immunotherapy combination strategies for MSS-CRC.


Subject(s)
Antibodies, Bispecific , Antibodies, Monoclonal, Humanized , CD3 Complex , Carcinoembryonic Antigen , Neoplasms , Humans , Antibodies, Bispecific/therapeutic use , Antibodies, Bispecific/pharmacokinetics , Antibodies, Bispecific/administration & dosage , Antibodies, Bispecific/adverse effects , Antibodies, Bispecific/pharmacology , Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal, Humanized/pharmacokinetics , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/adverse effects , Female , Male , Middle Aged , Aged , CD3 Complex/immunology , Adult , Carcinoembryonic Antigen/immunology , Neoplasms/drug therapy , Neoplasms/immunology , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics
9.
Front Immunol ; 15: 1392933, 2024.
Article in English | MEDLINE | ID: mdl-38779683

ABSTRACT

Introduction: Antigen binding to the T cell antigen receptor (TCR) leads to the phosphorylation of the immunoreceptor tyrosine-based activation motifs (ITAMs) of the CD3 complex, and thereby to T cell activation. The CD3ε subunit plays a unique role in TCR activation by recruiting the kinase LCK and the adaptor protein NCK prior to ITAM phosphorylation. Here, we aimed to investigate how phosphorylation of the individual CD3ε ITAM tyrosines impacts the CD3ε signalosome. Methods: We mimicked irreversible tyrosine phosphorylation by substituting glutamic acid for the tyrosine residues in the CD3ε ITAM. Results: Integrating CD3ε phospho-mimetic variants into the complete TCR-CD3 complex resulted in reduced TCR signal transduction, which was partially compensated by the involvement of the other TCR-CD3 ITAMs. By using novel CD3ε phospho-mimetic Chimeric Antigen Receptor (CAR) variants, we avoided any compensatory effects of other ITAMs in the TCR-CD3 complex. We demonstrated that irreversible CD3ε phosphorylation prevented signal transduction upon CAR engagement. Mechanistically, we demonstrated that glutamic acid substitution at the N-terminal tyrosine residue of the CD3ε ITAM (Y39E) significantly reduces NCK binding to the TCR. In contrast, mutation at the C-terminal tyrosine of the CD3ε ITAM (Y50E) abolished LCK recruitment to the TCR, while increasing NCK binding. Double mutation at the C- and N-terminal tyrosines (Y39/50E) allowed ZAP70 to bind, but reduced the interaction with LCK and NCK. Conclusions: The data demonstrate that the dynamic phosphorylation of the CD3ε ITAM tyrosines is essential for CD3ε to orchestrate optimal TCR and CAR signaling and highlights the key role of CD3ε signalosome to tune signal transduction.


Subject(s)
CD3 Complex , Receptors, Antigen, T-Cell , Receptors, Chimeric Antigen , Signal Transduction , CD3 Complex/metabolism , CD3 Complex/immunology , Phosphorylation , Humans , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell/immunology , Receptors, Chimeric Antigen/metabolism , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/genetics , Lymphocyte Activation/immunology , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Receptor-CD3 Complex, Antigen, T-Cell/metabolism , Receptor-CD3 Complex, Antigen, T-Cell/immunology , Receptor-CD3 Complex, Antigen, T-Cell/genetics , HEK293 Cells , ZAP-70 Protein-Tyrosine Kinase/metabolism , ZAP-70 Protein-Tyrosine Kinase/genetics , Immunoreceptor Tyrosine-Based Activation Motif , Protein Binding , Jurkat Cells , Oncogene Proteins
10.
Biomed Pharmacother ; 175: 116718, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38744221

ABSTRACT

Advanced cholangiocarcinoma (CCA) presents a clinical challenge due to limited treatment options, necessitating exploration of innovative therapeutic approaches. Bispecific T cell engager (BTE)-armed T cell therapy shows promise in hematological and solid malignancies, offering potential advantages in safety over continuous BTE infusion. In this context, we developed a novel BTE, targeting CD3 on T cells and integrin αvß6, an antigen elevated in various epithelial malignancies, on cancer cells. The novel BTE was generated by fusing an integrin αvß6-binding peptide (A20) to an anti-CD3 (OKT3) single-chain variable fragment (scFv) through a G4S peptide linker (A20/αCD3 BTE). T cells were then armed with A20/αCD3 BTE (A20/αCD3-armed T cells) and assessed for antitumor activity. Our results highlight the specific binding of A20/αCD3 BTE to CD3 on T cells and integrin αvß6 on target cells, effectively redirecting T cells towards these targets. After co-culture, A20/αCD3-armed T cells exhibited significantly heightened cytotoxicity against integrin αvß6-expressing target cells compared to unarmed T cells in both KKU-213A cells and A375.ß6 cells. Moreover, in a five-day co-culture, A20/αCD3-armed T cells demonstrated superior cytotoxicity against KKU-213A spheroids compared to unarmed T cells. Importantly, A20/αCD3-armed T cells exhibited an increased proportion of the effector memory T cell (Tem) subset, upregulation of T cell activation markers, enhanced T cell proliferation, and increased cytolytic molecule/cytokine production, when compared to unarmed T cells in an integrin αvß6-dependent manner. These findings support the potential of A20/αCD3-armed T cells as a novel therapeutic approach for integrin αvß6-expressing cancers.


Subject(s)
Antigens, Neoplasm , Bile Duct Neoplasms , Cholangiocarcinoma , Integrins , T-Lymphocytes , Humans , Cholangiocarcinoma/immunology , Cholangiocarcinoma/therapy , Cholangiocarcinoma/pathology , Antigens, Neoplasm/immunology , T-Lymphocytes/immunology , Integrins/metabolism , Cell Line, Tumor , Bile Duct Neoplasms/immunology , Bile Duct Neoplasms/pathology , Bile Duct Neoplasms/therapy , CD3 Complex/immunology , Single-Chain Antibodies/pharmacology , Coculture Techniques , Antibodies, Bispecific/pharmacology
11.
Exp Mol Pathol ; 137: 104897, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38691979

ABSTRACT

BACKGROUND: Signaling by toll-like receptors (TLRs) initiates important immune responses against viral infection. The role of TLRs in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is not well elucidated. Thus, we investigated the interaction of TLRs agonists and SARS-COV-2 antigens with immune cells in vitro. MATERIAL & METHODS: 30 coronavirus disease 2019 (COVID-19) patients (15 severe and 15 moderate) and 10 age and sex-matched healthy control (HC) were enrolled. Peripheral blood mononuclear cells (PBMCs) were isolated and activated with TLR3, 7, 8, and 9 agonists, the spike protein (SP) of SARS-CoV-2, and the receptor binding domain (RBD) of SP. Frequencies of CD3+IFN-ß+ T cells, and CD3+IFN-γ+ T cells were evaluated by flow cytometry. Interferon (IFN)-ß gene expression was assessed by qRT-PCR. RESULTS: The frequency of CD3+IFN-ß+ T cells was higher in PBMCs from moderate (p < 0.0001) and severe (p = 0.009) patients at baseline in comparison with HCs. The highest increase in the frequency of CD3+IFN-ß+ T cells in cell from moderate patients was induced by TLR8 agonist and SP (p < 0.0001 for both) when compared to HC, while, the highest increase of the frequency of CD3+IFN-ß+ T cells in sample of severe patients was seen with TLR8 and TLR7 agonists (both p = 0.002). The frequency of CD3+IFN-γ+ T cells was significantly increased upon stimulation with TLR agonists in cell from patients with moderate and severe COVID-19, compared with HC (all p < 0.01), except with TLR7 and TLR8 agonists. The TLR8 agonist did not significantly increase the frequency of CD3+IFN-γ+ T cells in PBMCs of severe patients, but did so in cells from patients with moderate disease (p = 0.01). Moreover, IFN-ß gene expression was significantly upregulated in CD3+T cells from moderate (p < 0.0001) and severe (p = 0.002) COVID-19 patients, compared to HC after stimulation with the TLR8 agonist, while, stimulation of T cells with SP, significantly up-regulated IFN-ß mRNA expression in cells from patients with moderate (p = 0.0003), but not severe disease. CONCLUSION: Stimulation of PBMCs from COVID-19 patients, especially patients with moderate disease, with TLR8 agonist and SP increased the frequency of IFN-ß-producing T cells and IFN-ß gene expression.


Subject(s)
CD3 Complex , COVID-19 , SARS-CoV-2 , T-Lymphocytes , Toll-Like Receptors , Humans , COVID-19/immunology , COVID-19/virology , SARS-CoV-2/immunology , Male , Female , Middle Aged , Toll-Like Receptors/agonists , Toll-Like Receptors/genetics , CD3 Complex/immunology , CD3 Complex/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/drug effects , Adult , Interferon-gamma/metabolism , Interferon-gamma/genetics , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/genetics , Interferon-beta/genetics , Interferon-beta/immunology , Aged , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/drug effects , Toll-Like Receptor Agonists
12.
Expert Opin Biol Ther ; 24(5): 321-326, 2024 May.
Article in English | MEDLINE | ID: mdl-38717336

ABSTRACT

INTRODUCTION: At the 65th American Society of Hematology (ASH) 2023 Annual Meeting, the latest advancements in CD20×CD3 BsAbs for B-cell lymphoma (BCL) were highlighted, particularly in relapsed/refractory (R/R) follicular lymphoma (FL) and R/R diffuse large B-cell lymphoma (DLBCL). AREAS COVERED: This summary highlights some of the major studies on CD20×CD3 BsAbs for BCL. EXPERT OPINION/COMMENTARY: CD20×CD3 is the most widely studied BsAb, with promising results in patients with R/R DLBCL and R/R FL ≥ two prior lines of systemic therapy. Trials with the first line of B-cell lymphoma also revealed promising results. Hopefully, BsAb monotherapy or BsAb-containing regimens may become the standard therapy in patients with FL and DLBCL.


Subject(s)
Antibodies, Bispecific , Antigens, CD20 , CD3 Complex , Immunotherapy , Lymphoma, Large B-Cell, Diffuse , Adult , Humans , Antibodies, Bispecific/therapeutic use , Antigens, CD20/immunology , Antineoplastic Agents, Immunological/therapeutic use , CD3 Complex/immunology , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/drug therapy , Lymphoma, B-Cell/therapy , Lymphoma, Follicular/immunology , Lymphoma, Follicular/drug therapy , Lymphoma, Follicular/therapy , Lymphoma, Large B-Cell, Diffuse/immunology , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/therapy , Congresses as Topic
14.
PLoS One ; 19(5): e0300174, 2024.
Article in English | MEDLINE | ID: mdl-38696390

ABSTRACT

Off-the-shelf immunotherapeutics that suppress tumor growth and provide durable protection against relapse could enhance cancer treatment. We report preclinical studies on a CD33 x CD3 bivalent bispecific diabody, AMV564, that not only suppresses tumor growth, but also facilitates memory responses in a mouse model of acute myelogenous leukemia (AML). Mechanistically, a single 5-day treatment with AMV564 seems to reduce tumor burden by redirection of T cells, providing a time window for allogeneic or other T cells that innately recognize tumor antigens to become activated and proliferate. When the concentration of bispecific becomes negligible, the effector: target ratio has also shifted, and these activated T cells mediate long-term tumor control. To test the efficacy of AMV564 in vivo, we generated a CD33+ MOLM13CG bioluminescent human cell line and optimized conditions needed to control these cells for 62 days in vivo in NSG mice. Of note, not only did MOLM13CG become undetectable by bioluminescence imaging in response to infusion of human T cells plus AMV564, but also NSG mice that had cleared the tumor also resisted rechallenge with MOLM13CG in spite of no additional AMV564 treatment. In these mice, we identified effector and effector memory human CD4+ and CD8+ T cells in the peripheral blood immediately prior to rechallenge that expanded significantly during the subsequent 18 days. In addition to the anti-tumor effects of AMV564 on the clearance of MOLM13CG cells in vivo, similar effects were seen when primary CD33+ human AML cells were engrafted in NSG mice even when the human T cells made up only 2% of the peripheral blood cells and AML cells made up 98%. These studies suggest that AMV564 is a novel and effective bispecific diabody for the targeting of CD33+ AML that may provide long-term survival advantages in the clinic.


Subject(s)
Antibodies, Bispecific , CD3 Complex , Immunologic Memory , Leukemia, Myeloid, Acute , Sialic Acid Binding Ig-like Lectin 3 , Animals , Humans , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/drug therapy , Antibodies, Bispecific/pharmacology , Antibodies, Bispecific/immunology , Mice , CD3 Complex/immunology , Immunologic Memory/drug effects , Cell Line, Tumor , T-Lymphocytes/immunology , T-Lymphocytes/drug effects
15.
J Immunol ; 213(1): 7-13, 2024 07 01.
Article in English | MEDLINE | ID: mdl-38775415

ABSTRACT

Lymphocyte activation gene 3 (LAG3) is an inhibitory receptor that plays a critical role in controlling T cell tolerance and autoimmunity and is a major immunotherapeutic target. LAG3 is expressed on the cell surface as a homodimer but the functional relevance of this is unknown. In this study, we show that the association between the TCR/CD3 complex and a murine LAG3 mutant that cannot dimerize is perturbed in CD8+ T cells. We also show that LAG3 dimerization is required for optimal inhibitory function in a B16-gp100 tumor model. Finally, we demonstrate that a therapeutic LAG3 Ab, C9B7W, which does not block LAG3 interaction with its cognate ligand MHC class II, disrupts LAG3 dimerization and its association with the TCR/CD3 complex. These studies highlight the functional importance of LAG3 dimerization and offer additional approaches to therapeutically target LAG3.


Subject(s)
Antigens, CD , CD8-Positive T-Lymphocytes , Lymphocyte Activation Gene 3 Protein , Protein Multimerization , Animals , Mice , Antigens, CD/immunology , Antigens, CD/metabolism , Antigens, CD/genetics , CD8-Positive T-Lymphocytes/immunology , Melanoma, Experimental/immunology , Mice, Inbred C57BL , Receptor-CD3 Complex, Antigen, T-Cell/immunology , CD3 Complex/immunology , Humans , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Lymphocyte Activation/immunology , Protein Binding
16.
Front Immunol ; 15: 1391954, 2024.
Article in English | MEDLINE | ID: mdl-38765008

ABSTRACT

Sarcomas are rare and heterogeneous malignancies that are difficult to treat. Approximately 50% of patients diagnosed with sarcoma develop metastatic disease with so far very limited treatment options. The transmembrane protein B7-H3 reportedly is expressed in various malignancies, including different sarcoma subtypes. In several cancer entities B7-H3 expression is associated with poor prognosis. In turn, B7-H3 is considered a promising target for immunotherapeutic approaches. We here report on the preclinical characterization of a B7-H3xCD3 bispecific antibody in an IgG-based format, termed CC-3, for treatment of different sarcoma subtypes. We found B7-H3 to be expressed on all sarcoma cells tested and expression on sarcoma patients correlated with decreased progression-free and overall survival. CC-3 was found to elicit robust T cell responses against multiple sarcoma subtypes, resulting in significant activation, release of cytokines and effector molecules. In addition, CC-3 promoted T cell proliferation and differentiation, resulting in the generation of memory T cell subsets. Finally, CC-3 induced potent target cell lysis in a target cell restricted manner. Based on these results, a clinical trial evaluating CC-3 in soft tissue sarcoma is currently in preparation.


Subject(s)
Antibodies, Bispecific , B7 Antigens , Sarcoma , Humans , Antibodies, Bispecific/pharmacology , Antibodies, Bispecific/therapeutic use , Sarcoma/immunology , Sarcoma/drug therapy , B7 Antigens/immunology , B7 Antigens/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Cell Line, Tumor , Bone Neoplasms/immunology , Bone Neoplasms/pathology , Female , Male , Animals , Lymphocyte Activation/immunology , Middle Aged , CD3 Complex/immunology , Aged , Cell Proliferation , Adult
17.
Eur J Cancer ; 204: 114071, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38691878

ABSTRACT

Systemic sclerosis, a severe inflammatory autoimmune disease, shares a common thread with cancer through the underlying mechanism of inflammation. This inflammatory milieu not only drives the immune dysregulation characteristic of autoimmune diseases but also plays a pivotal role in the pathogenesis of cancer. Among the cellular components involved, B cells have emerged as key players in hematologic tumor and autoimmune disease, contributing to immune dysregulation and persistent tissue fibrosis in systemic sclerosis, as well as tumor progression and immune evasion in cancer. Consequently, novel therapeutic strategies targeting B cells hold promise in both conditions. Recent exploration of CD19 CAR T cells in severe systemic sclerosis patients has shown great potential, but also introduced possible risks and drawbacks associated with viral vectors, prolonged CAR T cell persistence, lengthy production timelines, high costs, and the necessity of conditioning patients with organotoxic and fertility-damaging chemotherapy. Given these challenges, alternative CD19-depleting approaches are of high interest for managing severe systemic autoimmune diseases. Here, we present the pioneering use of blinatumomab, a bispecific anti-CD3/anti-CD19 T cell engager in a patient with progressive, severe systemic sclerosis, offering a promising alternative for such challenging cases.


Subject(s)
Antibodies, Bispecific , Antigens, CD19 , Scleroderma, Systemic , Humans , Antibodies, Bispecific/therapeutic use , Scleroderma, Systemic/drug therapy , Scleroderma, Systemic/immunology , Antigens, CD19/immunology , T-Lymphocytes/immunology , T-Lymphocytes/drug effects , Female , CD3 Complex/immunology , CD3 Complex/metabolism , Middle Aged , Immunotherapy, Adoptive/methods
18.
Nature ; 630(8015): 222-229, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38657677

ABSTRACT

Gamma delta (γδ) T cells, a unique T cell subgroup, are crucial in various immune responses and immunopathology1-3. The γδ T cell receptor (TCR), which is generated by γδ T cells, recognizes a diverse range of antigens independently of the major histocompatibility complex2. The γδ TCR associates with CD3 subunits, initiating T cell activation and holding great potential in immunotherapy4. Here we report the structures of two prototypical human Vγ9Vδ2 and Vγ5Vδ1 TCR-CD3 complexes5,6, revealing two distinct assembly mechanisms that depend on Vγ usage. The Vγ9Vδ2 TCR-CD3 complex is monomeric, with considerable conformational flexibility in the TCRγ-TCRδ extracellular domain and connecting peptides. The length of the connecting peptides regulates the ligand association and T cell activation. A cholesterol-like molecule wedges into the transmembrane region, exerting an inhibitory role in TCR signalling. The Vγ5Vδ1 TCR-CD3 complex displays a dimeric architecture, whereby two protomers nestle back to back through the Vγ5 domains of the TCR extracellular domains. Our biochemical and biophysical assays further corroborate the dimeric structure. Importantly, the dimeric form of the Vγ5Vδ1 TCR is essential for T cell activation. These findings reveal organizing principles of the γδ TCR-CD3 complex, providing insights into the unique properties of γδ TCR and facilitating immunotherapeutic interventions.


Subject(s)
CD3 Complex , Receptors, Antigen, T-Cell, gamma-delta , T-Lymphocytes , Humans , CD3 Complex/chemistry , CD3 Complex/immunology , CD3 Complex/metabolism , CD3 Complex/ultrastructure , Cholesterol/metabolism , Cholesterol/chemistry , Cryoelectron Microscopy , Ligands , Lymphocyte Activation/immunology , Models, Molecular , Protein Domains , Protein Multimerization , Receptors, Antigen, T-Cell, gamma-delta/chemistry , Receptors, Antigen, T-Cell, gamma-delta/immunology , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Receptors, Antigen, T-Cell, gamma-delta/ultrastructure , T-Lymphocytes/chemistry , T-Lymphocytes/cytology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Signal Transduction , Cell Membrane/chemistry , Cell Membrane/metabolism
19.
Int Immunopharmacol ; 133: 112115, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38652959

ABSTRACT

BACKGROUND: Human T cells play an important role in immunity against tuberculosis (TB) infection. Activating receptor HLA-DR and inhibitory receptor KLRG1 are critical regulators of T cell function during viral infection and tumorigenesis, but they have been less studied in TB infection. METHODS: In this study, we explored the relationship between CD3+ T cell expression of HLA-DR and KLRG1 receptors and function against TB infection. Flow cytometry was conducted to assess the immunomodulatory effects of HLA-DR and KLRG1 receptors on CD3+ T cells in patients with different TB infection status. RESULTS: We found activating receptors HLA-DR, NKG2C, CD57 and NKP46, and inhibitory receptors KLRG1 and KIR on CD3+ T cells in different TB infection status showed different distribution patterns; the cytotoxic potential and cytokine secretion capacity of CD3+ T cells after Mtb-specific antigen stimulation were significantly enhanced in TB infection groups. Further studies revealed HLA-DR+ T and KLRG1+ T cells expressed higher activating and inhibitory receptors than the negative population. In addition, the expression of cytotoxic potential and cytokine secretion capacity of HLA-DR+ T and KLRG1+ T cells was significantly higher than that of HLA-DR- T and KLRG1- T cells. CONCLUSIONS: Expression of HLA-DR and KLRG1 enhances the cytotoxic potential and cytokine secretion capacity of CD3+ T cells in TB patients, suggesting CD3+ T cells expressing HLA-DR and KLRG1 are important effector cell phenotypes involved in the host anti-TB infection. HLA-DR and KLRG1 expressed by CD3+ T cells may be potential predictive markers of TB disease progression and clinical immune assessment.


Subject(s)
Cytokines , HLA-DR Antigens , Lectins, C-Type , Receptors, Immunologic , Tuberculosis , Adult , Female , Humans , Male , Middle Aged , Young Adult , CD3 Complex/metabolism , CD3 Complex/immunology , Cells, Cultured , Cytokines/metabolism , Cytotoxicity, Immunologic , HLA-DR Antigens/metabolism , HLA-DR Antigens/immunology , Lectins, C-Type/metabolism , Mycobacterium tuberculosis/immunology , Receptors, Immunologic/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tuberculosis/immunology
20.
Nat Med ; 30(6): 1593-1601, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38671240

ABSTRACT

Bispecific T cell engagers (BiTEs) kill B cells by engaging T cells. BiTEs are highly effective in acute lymphoblastic leukemia. Here we treated six patients with multidrug-resistant rheumatoid arthritis (RA) with the CD19xCD3 BiTE blinatumomab under compassionate use. Low doses of blinatumomab led to B cell depletion and concomitant decrease of T cells, documenting their engager function. Treatment was safe, with brief increase in body temperature and acute phase proteins during first infusion but no signs of clinically relevant cytokine-release syndrome. Blinatumomab led to a rapid decline in RA clinical disease activity in all patients, improved synovitis in ultrasound and FAPI-PET-CT and reduced autoantibodies. High-dimensional flow cytometry analysis of B cells documented an immune reset with depletion of activated memory B cells, which were replaced by nonclass-switched IgD-positive naïve B cells. Together, these data suggest the feasibility and potential for BiTEs to treat RA. This approach warrants further exploration on other B-cell-mediated autoimmune diseases.


Subject(s)
Antibodies, Bispecific , Arthritis, Rheumatoid , B-Lymphocytes , T-Lymphocytes , Humans , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/drug therapy , Antibodies, Bispecific/therapeutic use , T-Lymphocytes/immunology , Female , B-Lymphocytes/immunology , Male , Middle Aged , Antigens, CD19/immunology , Aged , Adult , CD3 Complex/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...