Project description:p53 is the most mutated gene in cancer, yet there are no effective drugs targeting p53 mutants. Here, we report the development of monoclonal antibodies targeting a p53 hotspot mutation E285K (E285K-mAbs). These mAbs recognize the mutant E285K epitope without cross-reactivity against wild-type p53. They are delivered by lipid nanoparticles (LNPs) that encapsulate DNA plasmids. The LNP-pE285K-mAbs in the IgG1 formats exhibit a robust anti-tumor effect, facilitating the infiltration of immune cells, including CD8+ T cells, B cells, and NK cells. Single-cell sequencing reveals that the therapeutic effects of IgG1 are associated with reduced immune inhibitory signaling, increased MHC signaling from B cells to CD8+ T cells, and enriched anti-tumor T cell and B cell receptor profiles. The E285K-mAbs can also be made in the dimeric IgA (dIgA) format. The anti-tumor activity of IgA is dependent on PIGR, whereas that of IgG1 is dependent on TRIM21. These findings indicate that targeting specific mutant epitopes through DNA-encoded and LNP-delivered mAbs represents a novel approach for precision medicine against p53 mutants in PIGR- or TRIM21-positive cancers.
Project description:Relapse after allo-HCT is a major cause of death of AML patients and results from immune evasion of AML blasts. Dysfunction of the p53 signaling pathway is frequent in AML and often caused by upregulation of the central p53 negative regulator Murine Double Minute 2 (MDM2). Besides its oncogenic effects p53 also regulates immune function and immune surveillance of solid cancer. We hypothesize that p53 also controls immune-related genes in AML cells and that p53 reactivation via MDM2-inhibition may enhance the immunogenicity of AML cells to allogeneic T cells.
Project description:Therapeutic use of agonistic anti-CD40 antibodies is a potentially powerful approach for activating the immune response to eradicate tumors. However, the translation of this approach to clinical practice has been significantly restricted due to the severe dose-limiting toxicities observed in multiple clinical trials. Here, we demonstrate that conventional type-1 dendritic cells are essential for triggering antitumor immunity but not toxicity by CD40 agonists, while macrophages, platelets, and monocytes lead to the toxic events. Therefore, we designed bispecific antibodies that target CD40 activation preferentially to dendritic cells. These bispecific reagents demonstrate a superior safety profile compared to their parental CD40 monospecific antibody, while triggering potent anti-tumor activity. We suggest such cell-selective bispecific agonistic antibodies as a drug platform to bypass the dose-limiting toxicities of anti-CD40, and of additional types of agonistic antibodies used for cancer immunotherapy.
Project description:Proteomic identification and characterization of antibodies comprising the serological response to antigen can provide unique insight into the functional dynamics of adaptive immunity. We have developed a novel method to overcome the technical challenges which previously limited the direct analysis of immunoglobulin proteins in serum, as demonstrated by the identification of human anti-tetanus toxoid (TT) immunoglobulin G (IgG) proteins following booster vaccination. We analyzed the serum IgG repertoire across four time-points corresponding to pre-vaccination, 7 days, 3 months, and 9 months post vaccination. Antigen-specific antibodies were affinity purified against immobilized TT protein and sequenced by bottom-up nanoLC-MS/MS. Interpretation of mass spectra required a custom reference database of IgG heavy and light chain variable sequences determined by NextGen RNA sequencing of the donor's circulating plasmablasts and memory B cells following booster vaccination.
Project description:Background: Our former study demonstrated that anti-trophoblast antibodies (ATAB) are more prevalent in the sera of patients with unexplained recurrent miscarriages (uRM), and the detection of ATAB is restricted to binding to JEG-3 cells and the quantification by flow cytometry. We further proved that the ATAB-positive sera decrease the production of β-hCG and progesterone in the same cell model. The specific antigenic epitopes of ATAB are still unclear, therefore our aim was to identify one specific target of ATAB in uRM patients. Methods: The possible targets of ATAB were identified by western blots and mass spectrometry, and α-Enolase (ENO1) was further confirmed by a competition assay. The level of anti-ENO1 antibodies in the sera and the production of β-hCG and progesterone were evaluated with enzyme-linked immunosorbent assay (ELISA). Additionally, the expression of ENO1 in the first trimester placenta was analyzed with immunohistochemistry and triple fluorescence staining in uRM patients and healthy controls. Findings: ENO1 was identified to be a prominent target of ATAB and anti-ENO1 antibodies can decrease the secretion of β-hCG and progesterone. The titer of anti-ENO1 antibodies was increased in the sera of ATAB-positive patients compared to ATAB-negative patients. Overexpression of ENO1 and co-expression of ENO1 and β-arrestin was found in the extra villous trophoblasts of uRM patients in the first trimester placenta. Interpretation: Anti-ENO1 antibody in the sera may be a novel autoimmune biomarker of uRM. Decreasing anti-ENO1 antibodies or inhibiting ENO1 expression might be a potential therapeutic treatment for uRM patients.
Project description:One of the most common genetic backgrounds for mice used as model to investigate human diseases is the BALB/c strain. This work is aimed to characterize the pattern of natural anti-carbohydrate antibodies present in serum of BALB/c mice by printed glycan array technology. Natural antibodies from serum of BALB/c mice interacted with at least 70 glycans from a library of 419 different carbohydrate structures. However, only a restricted number of these (12, ~17%) were highly recognized by natural antibodies, and shared by more than 80% of mice under examination. This conserved pattern differed dramatically from that of humans. This finding together with not identical repertoires of antibodies between individual mice should be specially considered when mouse models are intended to be used for investigation of human natural antibodies in different biomedical research and diagnostic contexts.
Project description:Despite the clinical success of checkpoints inhibitors, a significant gap still exists in our understanding of their in vivo mechanism of action, thus limiting the rational development of next generation agents. Amongst these, anti-CTLA-4 antibodies were originally developed to block inhibitory signals into activated effector T cells. However, several recent studies have demonstrated that depletion of regulatory T cells expressing high levels of CTLA-4 is critical to anti-CTLA-4 anti-tumor activity. Whereas the mechanism of action remains controversial, the emerging data support clinical development of new antibodies with enhanced killing activity. Here, using single-cell RNA sequencing in in vivo and in vitro mouse models, we sought to dissect the impact of anti-CTLA-4 blocking, Treg depleting, and FcR engaging activity on the immune responses within tumors. We observed a rapid remodeling of the innate immune landscape as early as 24-hours post-treatment. Immune remodeling was driven mainly by FcγR-engagement, and not by Treg depletion or CTLA-4 blockade, and included reduction of suppressive macrophages and activation of type-I interferon signaling. Our findings indicate that FcγR engagement and innate immune remodeling are involved in successful anti-CTLA-4 treatment, supporting further development of optimized immunotherapy agents with these features.