Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.242
Filter
1.
Front Immunol ; 15: 1428653, 2024.
Article in English | MEDLINE | ID: mdl-39185404

ABSTRACT

The p53 protein, encoded by TP53, is a tumor suppressor that plays a critical role in regulating apoptosis, cell cycle regulation, and angiogenesis in tumor cells via controlling various downstream signals. Natural killer (NK) cell-mediated immune surveillance is a vital self-defense mechanism against cancer and other diseases, with NK cell activity regulated by various mechanisms. Among these, p53 plays a significant role in immune regulation by maintaining the homeostasis and functionality of NK cells. It enhances the transcriptional activity of NK cell-activating ligands and downregulates inhibitory ligands to boost NK cell activation and tumor-killing efficacy. Additionally, p53 influences NK cell cytotoxicity by promoting apoptosis, autophagy, and ferroptosis in different tumor cells. p53 is involved in the regulation of NK cell activity and effector functions through multiple pathways. p53 also plays a pivotal role in the tumor microenvironment (TME), regulating the activity of NK cells. NK cells are critical components of the TME and are capable of directly killing tumor cells. And p53 mutates in numerous cancers, with the most common alteration being a missense mutation. These mutations are commonly associated with poor survival rates in patients with cancer. This review details p53's role in NK cell tumor immunosurveillance, summarizing how p53 enhances NK cell recognition and tumor destruction. We also explore the potential applications of p53 in tumor immunotherapy, discussing strategies for modulating p53 to enhance NK cell function and improve the efficacy of tumor immunotherapy, along with the associated challenges. Understanding the interaction between p53 and NK cells within the TME is crucial for advancing NK cell-based immunotherapy and developing p53-related novel therapeutics.


Subject(s)
Killer Cells, Natural , Neoplasms , Tumor Microenvironment , Tumor Suppressor Protein p53 , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/immunology , Neoplasms/immunology , Tumor Microenvironment/immunology , Animals , Immunotherapy/methods , Cytotoxicity, Immunologic
2.
J Clin Invest ; 134(17)2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39024561

ABSTRACT

Notch signaling can have either an oncogenic or tumor-suppressive function in cancer depending on the cancer type and cellular context. While Notch can be oncogenic in early prostate cancer, we identified significant downregulation of the Notch pathway during prostate cancer progression from adenocarcinoma to neuroendocrine (NE) prostate cancer, where it functions as a tumor suppressor. Activation of Notch in NE and Rb1/Trp53-deficient prostate cancer models led to phenotypic conversion toward a more indolent, non-NE state with glandular features and expression of luminal lineage markers. This was accompanied by upregulation of MHC and type I IFN and immune cell infiltration. Overall, these data support Notch signaling as a suppressor of NE differentiation in advanced prostate cancer and provide insights into how Notch signaling influences lineage plasticity and the tumor microenvironment (TME).


Subject(s)
Cell Differentiation , Prostatic Neoplasms , Signal Transduction , Tumor Microenvironment , Male , Prostatic Neoplasms/immunology , Prostatic Neoplasms/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Tumor Microenvironment/immunology , Animals , Humans , Signal Transduction/immunology , Mice , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/immunology , Adenocarcinoma/immunology , Adenocarcinoma/pathology , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Neuroendocrine Cells/pathology , Neuroendocrine Cells/metabolism , Neuroendocrine Cells/immunology , Receptors, Notch/metabolism , Receptors, Notch/genetics , Receptors, Notch/immunology , Neuroendocrine Tumors/immunology , Neuroendocrine Tumors/pathology , Neuroendocrine Tumors/genetics , Neuroendocrine Tumors/metabolism , Cell Line, Tumor
3.
Sci Rep ; 14(1): 16721, 2024 07 19.
Article in English | MEDLINE | ID: mdl-39030304

ABSTRACT

Antigen-specific cytotoxic CD8 T cells are extremely effective in controlling tumor growth and have been the focus of immunotherapy approaches. We leverage in silico tools to investigate whether the occurrence of mutations in proteins previously described as immunogenic and highly expressed by glioblastoma multiforme (GBM), such as Epidermal Growth Factor Receptor (EGFR), Isocitrate Dehydrogenase 1 (IDH1), Phosphatase and Tensin homolog (PTEN) and Tumor Protein 53 (TP53), may be contributing to the differential presentation of immunogenic epitopes. We recovered Class I MHC binding information from wild-type and mutated proteins using the Immune Epitope Database (IEDB). After that, we built peptide-MHC (pMHC-I) models in HLA-arena, followed by hierarchical clustering analysis based on electrostatic surface features from each complex. We identified point mutations that are determinants for the presentation of a set of peptides from TP53 protein. We point to structural features in the pMHC-I complexes of wild-type and mutated peptides, which may play a role in the recognition of CD8 T cells. To further explore these features, we performed 100 ns molecular dynamics simulations for the peptide pairs (wt/mut) selected. In pursuit of novel therapeutic targets for GBM treatment, we selected peptides where our predictive results indicated that mutations would not disrupt epitope presentation, thereby maintaining a specific CD8 T cell immune response. These peptides hold potential for future GBM interventions, including peptide-based or mRNA vaccine development applications.


Subject(s)
Antigen Presentation , CD8-Positive T-Lymphocytes , Glioblastoma , Isocitrate Dehydrogenase , Tumor Suppressor Protein p53 , Glioblastoma/immunology , Glioblastoma/genetics , Glioblastoma/therapy , Humans , CD8-Positive T-Lymphocytes/immunology , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/immunology , Isocitrate Dehydrogenase/chemistry , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/immunology , Antigen Presentation/immunology , Mutation , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/immunology , PTEN Phosphohydrolase/chemistry , ErbB Receptors/immunology , ErbB Receptors/genetics , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/genetics , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/genetics , Brain Neoplasms/immunology , Brain Neoplasms/genetics , Brain Neoplasms/therapy
4.
Nat Cancer ; 5(7): 983-995, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39009816

ABSTRACT

The p53 tumor suppressor protein has a plethora of cell-intrinsic functions and consequences that impact diverse cell types and tissues. Recent studies are beginning to unravel how wild-type and mutant p53 work in distinct ways to modulate tumor immunity. This sets up a disequilibrium between tumor immunosurveillance and escape therefrom. The ability to exploit this emerging knowledge for translational approaches may shape immunotherapy and targeted therapeutics in the future, especially in combinatorial settings.


Subject(s)
Neoplasms , Tumor Suppressor Protein p53 , Humans , Neoplasms/immunology , Neoplasms/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/immunology , Animals , Immunotherapy/methods , Mutation , Tumor Escape
5.
Anticancer Res ; 44(7): 2921-2931, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38925841

ABSTRACT

BACKGROUND/AIM: Human papillomavirus (HPV)-positive head and neck squamous cell carcinoma (HNSCC) is clinically and immunologically distinct from HPV-negative HNSCC. Herein, we investigated the presence of tumor antigens HPV E6/E7 and wild-type p53-specific T-cell responses, and the impact of immune checkpoint blockade in patients with HPV-positive HNSCC. MATERIALS AND METHODS: Peripheral blood mononuclear cells (PBMCs) from patients with HPV-positive HNSCC were stimulated with HPV E6/E7 or wild-type p53-derived peptide mixture and evaluated using the interferon-γ enzyme-linked immunosorbent spot assay. Flow cytometry was performed to analyze the proportion of T-cell subsets and T cells expressing immune checkpoint molecules. RESULTS: HPV E6/E7-specific T cells were detected in 22 (95.7%) of 23 patients, whereas wild-type p53-specific T cells were detected in 3 (15.0%) of 20 patients. Seven (43.8%) of 16 patients exhibited wild-type p53-specific T-cell responses, as determined using whole proteins instead of peptides. Immune checkpoint blockade enhanced wild-type p53-specific T-cell responses in 9 (45.0%) of 20 patients. Flow cytometric analysis of PBMCs revealed that responders exhibiting enhanced wild-type p53-specific T-cell responses following immune checkpoint blockade had a significantly higher proportion of Ki-67+CD4+ T cells, Ki-67+CD8+ T cells, regulatory T cells, PD-1+CD4+ T cells, and TIM-3+CD4+ T cells than non-responders. CONCLUSION: Our findings indicate that tumor antigen-specific T cells are present in the peripheral blood of patients with HPV-positive HNSCC. Blockade of checkpoint pathways can enhance T-cell responses in certain patients, probably via activated T cells, Tregs, and/or exhausted CD4+ T cells.


Subject(s)
Head and Neck Neoplasms , Immune Checkpoint Inhibitors , Papillomavirus Infections , Squamous Cell Carcinoma of Head and Neck , Humans , Immune Checkpoint Inhibitors/therapeutic use , Immune Checkpoint Inhibitors/pharmacology , Squamous Cell Carcinoma of Head and Neck/immunology , Squamous Cell Carcinoma of Head and Neck/virology , Squamous Cell Carcinoma of Head and Neck/drug therapy , Male , Female , Middle Aged , Aged , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/virology , Papillomavirus Infections/immunology , Papillomavirus Infections/virology , Antigens, Neoplasm/immunology , Oncogene Proteins, Viral/immunology , Tumor Suppressor Protein p53/immunology , Adult , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Papillomaviridae/immunology , T-Lymphocytes/immunology , Human Papillomavirus Viruses
6.
J Hematol Oncol ; 17(1): 45, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38886748

ABSTRACT

BACKGROUND: p53, the most frequently mutated gene in cancer, lacks effective targeted drugs. METHODS: We developed monoclonal antibodies (mAbs) that target a p53 hotspot mutation E285K without cross-reactivity with wild-type p53. They were delivered using lipid nanoparticles (LNPs) that encapsulate DNA plasmids. Western blot, BLI, flow cytometry, single-cell sequencing (scRNA-seq), and other methods were employed to assess the function of mAbs in vitro and in vivo. RESULTS: These LNP-pE285K-mAbs in the IgG1 format exhibited a robust anti-tumor effect, facilitating the infiltration of immune cells, including CD8+ T, B, and NK cells. scRNA-seq revealed that IgG1 reduces immune inhibitory signaling, increases MHC signaling from B cells to CD8+ T cells, and enriches anti-tumor T cell and B cell receptor profiles. The E285K-mAbs were also produced in the dimeric IgA (dIgA) format, whose anti-tumor activity depended on the polymeric immunoglobulin receptor (PIGR), a membrane Ig receptor, whereas that of IgG1 relied on TRIM21, an intracellular IgG receptor. CONCLUSIONS: Targeting specific mutant epitopes using DNA-encoded and LNP-delivered mAbs represents a potential precision medicine strategy against p53 mutants in TRIM21- or PIGR-positive cancers.


Subject(s)
Antibodies, Monoclonal , Mutation , Tumor Suppressor Protein p53 , Tumor Suppressor Protein p53/immunology , Tumor Suppressor Protein p53/genetics , Animals , Humans , Mice , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Neoplasms/immunology , Neoplasms/genetics , Neoplasms/drug therapy , Neoplasms/therapy , CD8-Positive T-Lymphocytes/immunology , Nanoparticles/chemistry , Cell Line, Tumor
7.
BMC Pulm Med ; 24(1): 250, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38773432

ABSTRACT

BACKGROUND: This study assessed the diagnosis, staging and treatment guidance of lung cancer (LC) based on seven tumor-associated autoantibodies (TAAbs) -p53, PGP9.5, SOX2, GBU4-5, MAGE A1, CAGE, and GAGE7. METHODS: ELISA was used to determine the TAAb serum levels in 433 patients diagnosed with LC (161 surgical patients) and 76 patients with benign lung disease (16 surgical patients). The statistical characteristic of the TAAbs was compared among patients with different clinicopathological features. Pre- to postoperative changes in TAAb levels were analyzed to determine their value of LC. RESULTS: Among all patients, the positive rate of the seven TAAbs was 23.4%, sensitivity was 26.3%, accuracy was 36.3%, specificity was 93.4%, positive predictive value was 95.8%, and negative predictive value was 18.2%; the positive rate for the LC group (26.3%) was significantly higher than that for the benign group (6.6%; P < 0.001). Significant differences in the positive rate of the seven autoantibodies according to age (P < 0.001), smoking history (P = 0.009) and clinical LC stage (P < 0.001) were found. Smoking was positively associated with the positive of TAAbs (Τ = 0.118, P = 0.008). The positive rates of the seven TAAbs for squamous carcinoma (54.5%), other pathological types (44.4%) and poorly differentiated LC (57.1%) were significantly higher than those for the other types. The positive rate of GBU4-5 was highest among all TAAbs, and the SOX2 level in stage III-IV patients was much higher than that in other stages. For patients undergoing surgery, compared with the preoperative levels, the postoperative levels of the 7 markers, particularly p53 (P = 0.027), PGP9.5 (P = 0.007), GAGE7 (P = 0.014), and GBU4-5 (P = 0.002), were significantly different in the malignant group, especially in stage I-II patients, while no clear pre- to postoperative difference was observed in the benign group. CONCLUSIONS: When the seven TAAbs was positive, it was very helpful for the diagnosis of LC. The 7 TAAbs was valuable for staging and guiding treatment of LC in surgical patients.


Subject(s)
Autoantibodies , Biomarkers, Tumor , Lung Neoplasms , Neoplasm Staging , Humans , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Lung Neoplasms/diagnosis , Lung Neoplasms/blood , Autoantibodies/blood , Male , Female , Middle Aged , Aged , Biomarkers, Tumor/blood , Adult , SOXB1 Transcription Factors/immunology , Sensitivity and Specificity , Tumor Suppressor Protein p53/immunology , Enzyme-Linked Immunosorbent Assay , Aged, 80 and over , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/blood , Carcinoma, Squamous Cell/diagnosis , Carcinoma, Squamous Cell/pathology
8.
ACS Sens ; 9(3): 1475-1481, 2024 03 22.
Article in English | MEDLINE | ID: mdl-38441485

ABSTRACT

As a tumor-suppressing protein, p53 plays a crucial role in preventing cancer development. Its utility as an early cancer detection tool is significant, potentially enabling clinicians to forestall disease advancement and improve patient prognosis. In response to the pathological overexpression of this antigen in tumors, the prevalence of anti-p53 antibodies increases in serum, in a manner quantitatively indicative of cancer progression. This spike can be detected through techniques, such as Western blotting, immunohistochemistry, and immunoprecipitation. In this study, we present an electrochemical approach that supports ultrasensitive and highly selective anti-p53 autoantibody quantification without the use of an immuno-modified electrode. We specifically employ antigen-mimicking and antibody-capturing peptide-coated magnetic nanoparticles, along with an AC magnetic field-promoted sample mixing, prior to the presentation of Fab-captured targets to simple lectin-modified sensors. The subfemtomolar assays are highly selective and support quantification from serum-spiked samples within minutes.


Subject(s)
Antigens, Neoplasm , Autoantibodies , Magnetite Nanoparticles , Molecular Mimicry , Neoplasms , Tumor Suppressor Protein p53 , Humans , Neoplasms/diagnosis , Tumor Suppressor Protein p53/immunology , Autoantibodies/blood , Antigens, Neoplasm/immunology , Biosensing Techniques , Early Detection of Cancer
9.
Viruses ; 14(10)2022 10 21.
Article in English | MEDLINE | ID: mdl-36298868

ABSTRACT

HBx, a multifunctional regulatory protein, plays an essential role in the replication and pathogenesis of the hepatitis B virus (HBV). In this study, we found that in human hepatoma cells, the tumor suppressor p53 downregulates HBx via ubiquitin-dependent proteasomal degradation. p53 transcriptional activity that results from HBV infection was not essential for this effect. This was shown by treatment with a potent p53 inhibitor, pifithrin-α. Instead, we found that p53 facilitated the binding of E6-associated protein (E6AP), which is an E3 ligase, to HBx and induced E6AP-mediated HBx ubiquitination in a ternary complex of p53, E6AP, and HBx. The ability of p53 to induce E6AP-mediated downregulation of HBx and inhibit HBV replication was demonstrated in an in vitro HBV infection system. This study may provide insights into the regulation of HBx and HBV replication, especially with respect to p53 status, which may also help in understanding HBV-associated tumorigenesis in patients.


Subject(s)
Carcinoma, Hepatocellular , Hepatitis B , Liver Neoplasms , Tumor Suppressor Protein p53 , Humans , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/immunology , Cell Line , Hepatitis B/complications , Hepatitis B/genetics , Hepatitis B/immunology , Hepatitis B virus/physiology , Liver Neoplasms/genetics , Liver Neoplasms/immunology , Proteasome Endopeptidase Complex/metabolism , Trans-Activators/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/immunology , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/metabolism , Viral Regulatory and Accessory Proteins/metabolism , Virus Replication/physiology
10.
Cancer Immunol Res ; 10(8): 932-946, 2022 08 03.
Article in English | MEDLINE | ID: mdl-35749374

ABSTRACT

Adoptive cellular therapy (ACT) targeting neoantigens can achieve durable clinical responses in patients with cancer. Most neoantigens arise from patient-specific mutations, requiring highly individualized treatments. To broaden the applicability of ACT targeting neoantigens, we focused on TP53 mutations commonly shared across different cancer types. We performed whole-exome sequencing on 163 patients with metastatic solid cancers, identified 78 who had TP53 missense mutations, and through immunologic screening, identified 21 unique T-cell reactivities. Here, we report a library of 39 T-cell receptors (TCR) targeting TP53 mutations shared among 7.3% of patients with solid tumors. These TCRs recognized tumor cells in a TP53 mutation- and human leucocyte antigen (HLA)-specific manner in vitro and in vivo. Twelve patients with chemorefractory epithelial cancers were treated with ex vivo-expanded autologous tumor-infiltrating lymphocytes (TIL) that were naturally reactive against TP53 mutations. However, limited clinical responses (2 partial responses among 12 patients) were seen. These infusions contained low frequencies of mutant p53-reactive TILs that had exhausted phenotypes and showed poor persistence. We also treated one patient who had chemorefractory breast cancer with ACT comprising autologous peripheral blood lymphocytes transduced with an allogeneic HLA-A*02-restricted TCR specific for p53R175H. The infused cells exhibited an improved immunophenotype and prolonged persistence compared with TIL ACT and the patient experienced an objective tumor regression (-55%) that lasted 6 months. Collectively, these proof-of-concept data suggest that the library of TCRs targeting shared p53 neoantigens should be further evaluated for the treatment of patients with advanced human cancers. See related Spotlight by Klebanoff, p. 919.


Subject(s)
Hematopoietic Stem Cell Transplantation , Neoplasms , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Genes, T-Cell Receptor , Humans , Lymphocytes, Tumor-Infiltrating/immunology , Neoplasms/genetics , Neoplasms/therapy , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/immunology
11.
Nat Commun ; 13(1): 758, 2022 02 09.
Article in English | MEDLINE | ID: mdl-35140208

ABSTRACT

Immunotherapy with immune checkpoint blockade (ICB) has shown limited benefits in hepatocellular carcinoma (HCC) and other cancers, mediated in part by the immunosuppressive tumor microenvironment (TME). As p53 loss of function may play a role in immunosuppression, we herein examine the effects of restoring p53 expression on the immune TME and ICB efficacy. We develop and optimize a CXCR4-targeted mRNA nanoparticle platform to effectively induce p53 expression in HCC models. Using p53-null orthotopic and ectopic models of murine HCC, we find that combining CXCR4-targeted p53 mRNA nanoparticles with anti-PD-1 therapy effectively induces global reprogramming of cellular and molecular components of the immune TME. This effect results in improved anti-tumor effects compared to anti-PD-1 therapy or therapeutic p53 expression alone. Thus, our findings demonstrate the reversal of immunosuppression in HCC by a p53 mRNA nanomedicine when combined with ICB and support the implementation of this strategy for cancer treatment.


Subject(s)
Immune Checkpoint Inhibitors , RNA, Messenger/pharmacology , Tumor Microenvironment/immunology , Tumor Suppressor Protein p53 , Animals , Carcinoma, Hepatocellular/immunology , Cell Line, Tumor , Female , Humans , Immune Checkpoint Inhibitors/immunology , Immunosuppression Therapy , Immunotherapy/methods , Liver Neoplasms/immunology , Male , Mice , Mice, Inbred C57BL , Nanomedicine , Receptors, CXCR4/genetics , Tumor Suppressor Protein p53/immunology
12.
Mol Cell Biochem ; 477(4): 1281-1293, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35129780

ABSTRACT

Triple-Negative Breast Cancer (TNBC) is one of the most aggressive and hot BC subtypes. Our research group has recently shed the light on the utility of natural compounds as effective immunotherapeutic agents. The aim of this study is to investigate the role of a methoxylated quercetin glycoside (MQG) isolated from Cleome droserifolia in harnessing TNBC progression and tuning the tumor microenvironment and natural killer cells cytotoxicity. Results showed that MQG showed the highest potency (IC50 = 12 µM) in repressing cellular proliferation, colony-forming ability, migration, and invasion capacities. Mechanistically, MQG was found to modulate a circuit of competing endogenous RNAs where it was found to reduce the oncogenic MALAT-1 lncRNA and induce TP53 and its downstream miRNAs; miR-155 and miR-146a. Accordingly, this leads to alteration in several downstream signaling pathways such as nitric oxide synthesizing machinery, natural killer cells' cytotoxicity through inducing the expression of its activating ligands such as MICA/B, ULBP2, CD155, and ICAM-1 and trimming of the immune-suppressive cytokines such as TNF-α and IL-10. In conclusion, this study shows that MQG act as a compelling anti-cancer agent repressing TNBC hallmarks, activating immune cell recognition, and alleviating the immune-suppressive tumor microenvironment experienced by TNBC patients.


Subject(s)
Glycosides/pharmacology , MicroRNAs/immunology , RNA, Long Noncoding/immunology , RNA, Neoplasm/immunology , Triple Negative Breast Neoplasms/immunology , Tumor Suppressor Protein p53/immunology , Female , Humans , MCF-7 Cells , MicroRNAs/genetics , RNA, Long Noncoding/genetics , RNA, Neoplasm/genetics , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics , Tumor Suppressor Protein p53/genetics
13.
J Biol Chem ; 298(3): 101684, 2022 03.
Article in English | MEDLINE | ID: mdl-35124005

ABSTRACT

Adoptive cell therapy with tumor-specific T cells can mediate durable cancer regression. The prime target of tumor-specific T cells are neoantigens arising from mutations in self-proteins during malignant transformation. To understand T cell recognition of cancer neoantigens at the atomic level, we studied oligoclonal T cell receptors (TCRs) that recognize a neoepitope arising from a driver mutation in the p53 oncogene (p53R175H) presented by the major histocompatibility complex class I molecule HLA-A2. We previously reported the structures of three p53R175H-specific TCRs (38-10, 12-6, and 1a2) bound to p53R175H and HLA-A2. The structures showed that these TCRs discriminate between WT and mutant p53 by forming extensive interactions with the R175H mutation. Here, we report the structure of a fourth p53R175H-specific TCR (6-11) in complex with p53R175H and HLA-A2. In contrast to 38-10, 12-6, and 1a2, TCR 6-11 makes no direct contacts with the R175H mutation, yet is still able to distinguish mutant from WT p53. Structure-based in silico mutagenesis revealed that the 60-fold loss in 6-11 binding affinity for WT p53 compared to p53R175H is mainly due to the higher energetic cost of desolvating R175 in the WT p53 peptide during complex formation than H175 in the mutant. This indirect strategy for preferential neoantigen recognition by 6-11 is fundamentally different from the direct strategies employed by other TCRs and highlights the multiplicity of solutions to recognizing p53R175H with sufficient selectivity to mediate T cell killing of tumor but not normal cells.


Subject(s)
HLA-A2 Antigen , Immunotherapy, Adoptive , Neoplasms , Receptors, Antigen, T-Cell , Tumor Suppressor Protein p53 , Antigens, Neoplasm/chemistry , Antigens, Neoplasm/immunology , Epitopes/immunology , HLA-A2 Antigen/chemistry , HLA-A2 Antigen/immunology , Humans , Neoplasms/immunology , Neoplasms/therapy , Receptors, Antigen, T-Cell/chemistry , Receptors, Antigen, T-Cell/immunology , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/immunology
14.
Sci Rep ; 12(1): 3234, 2022 02 25.
Article in English | MEDLINE | ID: mdl-35217717

ABSTRACT

Leukemic cells proliferate faster than non-transformed counterparts. This requires them to change their metabolism to adapt to their high growth. This change can stress cells and facilitate recognition by immune cells such as cytotoxic lymphocytes, which express the activating receptor Natural Killer G2-D (NKG2D). The tumor suppressor gene p53 regulates cell metabolism, but its role in the expression of metabolism-induced ligands, and subsequent recognition by cytotoxic lymphocytes, is unknown. We show here that dichloroacetate (DCA), which induces oxidative phosphorylation (OXPHOS) in tumor cells, induces the expression of such ligands, e.g. MICA/B, ULBP1 and ICAM-I, by a wtp53-dependent mechanism. Mutant or null p53 have the opposite effect. Conversely, DCA sensitizes only wtp53-expressing cells to cytotoxic lymphocytes, i.e. cytotoxic T lymphocytes and NK cells. In xenograft in vivo models, DCA slows down the growth of tumors with low proliferation. Treatment with DCA, monoclonal antibodies and NK cells also decreased tumors with high proliferation. Treatment of patients with DCA, or a biosimilar drug, could be a clinical option to increase the effectiveness of CAR T cell or allogeneic NK cell therapies.


Subject(s)
Antineoplastic Agents , Leukemia , Tumor Suppressor Protein p53 , Antineoplastic Agents/metabolism , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Leukemia/immunology , Leukemia/metabolism , Ligands , NK Cell Lectin-Like Receptor Subfamily K/immunology , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Tumor Suppressor Protein p53/immunology , Tumor Suppressor Protein p53/metabolism
15.
ACS Appl Mater Interfaces ; 14(2): 2464-2477, 2022 Jan 19.
Article in English | MEDLINE | ID: mdl-35045602

ABSTRACT

Nanomedicine, constructed from therapeutics, presents an advantage in drug delivery for cancer therapies. However, nanocarrier-based treatment systems have problems such as interbatch variability, multicomponent complexity, poor drug delivery, and carrier-related toxicity. To solve these issues, the natural molecule honokiol (HK), an anticancer agent in a phase I clinical trial (CTR20170822), was used to form a self-assembly nanoparticle (SA) through hydrogen bonding and hydrophobicity. The preparation of SA needs no molecular precursors or excipients in aqueous solution, and 100% drug-loaded SA exhibited superior tumor-targeting ability due to the enhanced permeability and retention (EPR) effect. Moreover, SA significantly enhanced the antitumor immunity relative to free HK, and the mechanism has notable selectivity to the p53 pathway. Furthermore, SA exhibited excellent physiological stability and inappreciable toxicity. Taken together, this supramolecular self-assembly strategy provides a safe and "molecular economy" model for rational design of clinical therapies and is expected to promote targeted therapy of HK, especially in colorectal cancer patients with obvious p53 status.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Biocompatible Materials/pharmacology , Biphenyl Compounds/pharmacology , Colorectal Neoplasms/therapy , Immunotherapy , Lignans/pharmacology , Small Molecule Libraries/pharmacology , Tumor Suppressor Protein p53/antagonists & inhibitors , Animals , Antineoplastic Agents, Phytogenic/chemical synthesis , Antineoplastic Agents, Phytogenic/chemistry , Biocompatible Materials/chemical synthesis , Biocompatible Materials/chemistry , Biphenyl Compounds/chemical synthesis , Biphenyl Compounds/chemistry , Colorectal Neoplasms/immunology , Female , Humans , Lignans/chemical synthesis , Lignans/chemistry , Macromolecular Substances/chemical synthesis , Macromolecular Substances/chemistry , Materials Testing , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Structure , Neoplasms, Experimental/immunology , Neoplasms, Experimental/therapy , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry , Tumor Cells, Cultured , Tumor Suppressor Protein p53/immunology
16.
Acta Haematol ; 145(1): 9-17, 2022.
Article in English | MEDLINE | ID: mdl-34515042

ABSTRACT

Immune thrombocytopenia (ITP) is an acquired autoimmune disorder characterized by persistent thrombocytopenia resulting from increased platelet destruction and a loss of autoimmune tolerance. The pathogenesis of ITP is highly complex. Although ITP may be effectively controlled with currently available medications in some patients, a subset of cases remain refractory. The application of mesenchymal stem cells (MSCs) for human hematopoietic stem cell transplantation has increasingly demonstrated that MSCs modulate innate or adaptive immunity, thus resulting in a tolerant microenvironment. Functional defects and immunomodulatory disorders have been observed after the use of bone marrow mesenchymal stem cells (BM-MSCs) from patients with ITP. Here, we summarize the underlying mechanisms and clinical applications of various derived MSCs for ITP treatment, focusing on the main mechanisms underlying the functional defects and immune dysfunction of BM-MSCs from patients with ITP. Functional effects associated with the activation of the p53 pathway include decreased activity of the phosphatidylinositol 3 kinase/Akt pathway and activation of the TNFAIP3/NF-κB/SMAD7 pathway. Immune dysfunction appears to be associated with an impaired ability of BM-MSCs to induce various types of immune cells in ITP. At present, research focusing on MSCs in ITP remains in preliminary stages. The application of autologous or exogenous MSCs in the clinical treatment of ITP has been attempted in only a small case study and must be validated in larger-scale clinical trials.


Subject(s)
Hematopoietic Stem Cell Transplantation , Mesenchymal Stem Cells/immunology , Purpura, Thrombocytopenic, Idiopathic/immunology , Purpura, Thrombocytopenic, Idiopathic/therapy , Signal Transduction/immunology , Humans , NF-kappa B/immunology , Phosphatidylinositol 3-Kinases/immunology , Proto-Oncogene Proteins c-akt/immunology , Tumor Necrosis Factor alpha-Induced Protein 3/immunology , Tumor Suppressor Protein p53/immunology
18.
Molecules ; 26(20)2021 Oct 14.
Article in English | MEDLINE | ID: mdl-34684792

ABSTRACT

Importance: The protein p53 is an unequivocal tumor suppressor that is altered in half of all cancers. The immune system produces systemic p53 autoantibodies (p53 Abs) in many cancer patients. Objective: This systemic review and meta-analysis focuses on the prognostic value of p53 Abs expressed in the serum of patients with solid tumors. Data Sources: All the clinical investigations were searched on PubMed from the first study dated 1993 until May 2021 (date of submission of the manuscript). Study Selection: Studies were included that met the following criteria: (1) participants with cancer; (2) outcome results expressed in relation to the presence of a p53 antibody; (3) a primary outcome (disease-free survival, overall survival or progression-free survival) expressed as hazard ratio (HR). The following exclusion criteria were used: (1) insufficient data available to evaluate outcomes; (2) animal studies; (3) studies with less than 10 participants. As a result, 12 studies were included in the analysis. Data Extraction and Synthesis: PRISMA guidelines were used for abstracting and assessing data quality and validity by three independent observers. The summary estimates were generated using a fixed-effect model (Mantel-Haenszel method) or a random-effect model (DerSimonian-Laird method), depending on the absence or presence of heterogeneity (I2). Main Outcome(s) and Measure(s): The primary study outcome was to determine the prognostic value of p53 Abs from a large population of patients with solid tumors, as determined before data collection. Results: In total, 12 clinical studies involving 2094 patients were included in the meta-analysis, and it was determined that p53 Abs expression in the serum significantly correlated with poorer survival outcomes of cancer patients (95% CI 1.48 [1.24, 1.77]; p < 0.00001). Conclusions and Relevance: This is the first meta-analysis proving the diagnostic utility of p53-Abs for cancer patients in predicting poorer outcomes. The serum-p53 value (s-p53-value) may be useful for future theranostics.


Subject(s)
Autoantibodies/blood , Neoplasms/diagnosis , Neoplasms/immunology , Tumor Suppressor Protein p53/immunology , Biomarkers, Tumor/blood , Biomarkers, Tumor/genetics , Biomarkers, Tumor/immunology , Female , Humans , Male , Mutation , Neoplasms/genetics , Prognosis , Retrospective Studies , Survival Analysis , Tumor Suppressor Protein p53/genetics
19.
Front Immunol ; 12: 738435, 2021.
Article in English | MEDLINE | ID: mdl-34603319

ABSTRACT

Background: Although mRNA vaccines have been efficient for combating a variety of tumors, their effectiveness against glioma remains unclear. There is growing evidence that immunophenotyping can reflect the comprehensive immune status and microenvironment of the tumor, which correlates closely with treatment response and vaccination potency. The purpose of this research was to screen for effective antigens in glioma that could be used for developing mRNA vaccines and to further differentiate the immune subtypes of glioma to create an selection criteria for suitable patients for vaccination. Methods: Gene expression profiles and clinical data of 698 glioma samples were extracted from The Cancer Genome Atlas, and RNA_seq data of 1018 glioma samples was gathered from Chinese Glioma Genome Atlas. Gene Expression Profiling Interactive Analysis was used to determine differential expression genes and prognostic markers, cBioPortal software was used to verify gene alterations, and Tumor Immune Estimation Resource was used to investigate the relationships among genes and immune infiltrating cells. Consistency clustering was applied for consistent matrix construction and data aggregation, Gene oncology enrichment was performed for functional annotation, and a graph learning-based dimensionality reduction method was applied to describe the subtypes of immunity. Results: Four overexpressed and mutated tumor antigens associated with poor prognosis and infiltration of antigen presenting cells were identified in glioma, including TP53, IDH1, C3, and TCF12. Besides, four immune subtypes of glioma (IS1-IS4) and 10 immune gene modules were identified consistently in the TCGA data. The immune subtypes had diverse molecular, cellular, and clinical features. IS1 and IS4 displayed an immune-activating phenotype and were associated with worse survival than the other two subtypes, while IS2 and IS3 had lower levels of tumor immune infiltration. Immunogenic cell death regulators and immune checkpoints were also diversely expressed in the four immune subtypes. Conclusion: TP53, IDH1, C3, and TCF12 are effective antigens for the development of anti-glioma mRNA vaccines. We found four stable and repeatable immune subtypes of human glioma, the classification of the immune subtypes of glioma may play a crucial role in the predicting mRNA vaccine outcome.


Subject(s)
Antigens, Neoplasm/genetics , Brain Neoplasms/drug therapy , Cancer Vaccines/therapeutic use , Glioma/drug therapy , Vaccine Development , mRNA Vaccines/therapeutic use , Antigens, Neoplasm/immunology , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/immunology , Brain Neoplasms/genetics , Brain Neoplasms/immunology , Cancer Vaccines/immunology , Clinical Decision-Making , Complement C3/genetics , Complement C3/immunology , Databases, Genetic , Gene Expression Profiling , Glioma/genetics , Glioma/immunology , Humans , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/metabolism , Mutation , Precision Medicine , Transcriptome , Tumor Microenvironment , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/immunology , mRNA Vaccines/immunology
20.
J Immunol Methods ; 498: 113144, 2021 11.
Article in English | MEDLINE | ID: mdl-34481824

ABSTRACT

Bivalent VHHs have been shown to display better functional affinity compared with their monovalent counterparts. Bivalency can be achieved either by inserting a hinge region between both VHHs units or by using modules that lead to dimerization. In this report, a small self-associating peptide originating from the tetramerization domain of p53 was developed as a tool for devicing nanobody dimerization. This E3 peptide was evaluated for the dimerization of an anti-eGFP nanobody (nano-eGFP-E3) whose activity was compared to a bivalent anti-eGFP constructed in tandem using GS rich linker. The benefit of bivalency in terms of avidity and specificity was assessed in different in vitro and in cellulo assays. In ELISA and SPR, the dimeric and tandem formats were nearly equivalent in terms of gain of avidity compared to the monovalent counterpart. However, in cellulo, the nano-eGFP-E3 construct showed its superiority over the tandem format in terms of specificity with a highest and better ratio signal-to-noise. All together, the E3 peptide provides a universal suitable tool for the construction of dimeric biomolecules, in particular antibody fragments with improved functional affinity.


Subject(s)
Epitopes , Green Fluorescent Proteins/immunology , Peptide Fragments/immunology , Single-Domain Antibodies/immunology , Tumor Suppressor Protein p53/immunology , Animals , Antibody Affinity , Antibody Specificity , Green Fluorescent Proteins/genetics , HeLa Cells , Humans , Mutation , Peptide Fragments/genetics , Protein Multimerization , Tumor Suppressor Protein p53/genetics
SELECTION OF CITATIONS
SEARCH DETAIL