Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.173
Filter
1.
PLoS One ; 19(6): e0298469, 2024.
Article in English | MEDLINE | ID: mdl-38917176

ABSTRACT

Prostate stem cell antigen (PSCA) is associated with disease progression, promotion of angiogenesis, invasion, metastasis and immune evasion in cancer. However, its expression pattern and diagnostic and prognostic potential have not been thoroughly analysed from a pan-cancer perspective. This study aimed to examine the effects of PSCA on the prognosis and inflammatory cell infiltration patterns of various cancer types. We analysed the relationship between PSCA expression and immunological subtypes in tumor microenvironment (TME) and the role of molecular subtypes, potentially promising immune biomarkers and tumour-infiltrating lymphocytes (TILs) in various cancer types, especially lung adenocarcinoma (LUAD). In addition, we investigated the prognostic significance of PSCA expression in LUAD. The co-expression network of PSCA was found to be mainly involved in the regulation of immune responses and antigen processing and expression and was significantly enriched in pathological and substance metabolism-related pathways in cancer. Altogether, this study reveals that PSCA is a promising target for immunotherapy in patients with cancer.


Subject(s)
Antigens, Neoplasm , GPI-Linked Proteins , Lymphocytes, Tumor-Infiltrating , Neoplasm Proteins , Tumor Microenvironment , Humans , Antigens, Neoplasm/immunology , Antigens, Neoplasm/metabolism , Prognosis , Tumor Microenvironment/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Neoplasm Proteins/immunology , Neoplasm Proteins/metabolism , GPI-Linked Proteins/metabolism , Biomarkers, Tumor/metabolism , Neoplasms/immunology , Neoplasms/pathology , Adenocarcinoma of Lung/immunology , Adenocarcinoma of Lung/pathology , Adenocarcinoma of Lung/genetics , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Male
2.
Nat Med ; 30(6): 1636-1644, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38867077

ABSTRACT

Despite recent therapeutic advances, metastatic castration-resistant prostate cancer (mCRPC) remains lethal. Chimeric antigen receptor (CAR) T cell therapies have demonstrated durable remissions in hematological malignancies. We report results from a phase 1, first-in-human study of prostate stem cell antigen (PSCA)-directed CAR T cells in men with mCRPC. The starting dose level (DL) was 100 million (M) CAR T cells without lymphodepletion (LD), followed by incorporation of LD. The primary end points were safety and dose-limiting toxicities (DLTs). No DLTs were observed at DL1, with a DLT of grade 3 cystitis encountered at DL2, resulting in addition of a new cohort using a reduced LD regimen + 100 M CAR T cells (DL3). No DLTs were observed in DL3. Cytokine release syndrome of grade 1 or 2 occurred in 5 of 14 treated patients. Prostate-specific antigen declines (>30%) occurred in 4 of 14 patients, as well as radiographic improvements. Dynamic changes indicating activation of peripheral blood endogenous and CAR T cell subsets, TCR repertoire diversity and changes in the tumor immune microenvironment were observed in a subset of patients. Limited persistence of CAR T cells was observed beyond 28 days post-infusion. These results support future clinical studies to optimize dosing and combination strategies to improve durable therapeutic outcomes. ClinicalTrials.gov identifier NCT03873805 .


Subject(s)
Antigens, Neoplasm , GPI-Linked Proteins , Immunotherapy, Adoptive , Neoplasm Proteins , Prostatic Neoplasms, Castration-Resistant , Humans , Male , Prostatic Neoplasms, Castration-Resistant/therapy , Prostatic Neoplasms, Castration-Resistant/immunology , Prostatic Neoplasms, Castration-Resistant/pathology , Aged , Middle Aged , Antigens, Neoplasm/immunology , Immunotherapy, Adoptive/adverse effects , Immunotherapy, Adoptive/methods , GPI-Linked Proteins/immunology , Neoplasm Proteins/immunology , Receptors, Chimeric Antigen/immunology , Neoplasm Metastasis , T-Lymphocytes/immunology , T-Lymphocytes/transplantation , Prostate-Specific Antigen/blood
3.
Front Immunol ; 15: 1341745, 2024.
Article in English | MEDLINE | ID: mdl-38765012

ABSTRACT

Individuals with Kabuki syndrome present with immunodeficiency; however, how pathogenic variants in the gene encoding the histone-modifying enzyme lysine methyltransferase 2D (KMT2D) lead to immune alterations remain poorly understood. Following up on our prior report of KMT2D-altered integrin expression in B-cells, we performed targeted analyses of KMT2D's influence on integrin expression in T-cells throughout development (thymocytes through peripheral T-cells) in murine cells with constitutive- and conditional-targeted Kmt2d deletion. Using high-throughput RNA-sequencing and flow cytometry, we reveal decreased expression (both at the transcriptional and translational levels) of a cluster of leukocyte-specific integrins, which perturb aspects of T-cell activation, maturation, adhesion/localization, and effector function. H3K4me3 ChIP-PCR suggests that these evolutionary similar integrins are under direct control of KMT2D. KMT2D loss also alters multiple downstream programming/signaling pathways, including integrin-based localization, which can influence T-cell populations. We further demonstrated that KMT2D deficiency is associated with the accumulation of murine CD8+ single-positive (SP) thymocytes and shifts in both human and murine peripheral T-cell populations, including the reduction of the CD4+ recent thymic emigrant (RTE) population. Together, these data show that the targeted loss of Kmt2d in the T-cell lineage recapitulates several distinct features of Kabuki syndrome-associated immune deficiency and implicates epigenetic mechanisms in the regulation of integrin signaling.


Subject(s)
Integrins , Lymphocyte Activation , Animals , Mice , Integrins/metabolism , Integrins/genetics , Lymphocyte Activation/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Mice, Knockout , Vestibular Diseases/genetics , Vestibular Diseases/immunology , Vestibular Diseases/metabolism , Face/abnormalities , Humans , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Mice, Inbred C57BL , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Neoplasm Proteins/metabolism , Signal Transduction , Gene Expression Regulation , Abnormalities, Multiple , Hematologic Diseases , Myeloid-Lymphoid Leukemia Protein
4.
J Clin Invest ; 134(10)2024 May 15.
Article in English | MEDLINE | ID: mdl-38747288

ABSTRACT

Triple-negative breast cancer (TNBC) presents a formidable challenge in oncology due to its aggressive phenotype and the immunosuppressive nature of its tumor microenvironment (TME). In this issue of the JCI, Zhu, Banerjee, and colleagues investigated the potential of targeting the OTU domain-containing protein 4 (OTUD4)/CD73 axis to mitigate immunosuppression in TNBC. They identified elevated CD73 expression as a hallmark of immunosuppression in TNBC. Notably, the CD73 expression was regulated by OTUD4-mediated posttranslational modifications. Using ST80, a pharmacologic inhibitor of OTUD4, the authors demonstrated the restoration of cytotoxic T cell function and enhanced efficacy of anti-PD-L1 therapy in preclinical models. These findings underscore the therapeutic potential of targeting the OTUD4/CD73 axis in TNBC.


Subject(s)
5'-Nucleotidase , Protein Processing, Post-Translational , Triple Negative Breast Neoplasms , Tumor Microenvironment , Humans , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/genetics , 5'-Nucleotidase/immunology , 5'-Nucleotidase/genetics , 5'-Nucleotidase/metabolism , Tumor Microenvironment/immunology , Female , Neoplasm Proteins/immunology , Neoplasm Proteins/metabolism , Neoplasm Proteins/genetics , Animals
5.
J Clin Invest ; 134(11)2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38652550

ABSTRACT

The immune system can control cancer progression. However, even though some innate immune sensors of cellular stress are expressed intrinsically in epithelial cells, their potential role in cancer aggressiveness and subsequent overall survival in humans is mainly unknown. Here, we show that nucleotide-binding oligomerization domain-like receptor (NLR) family CARD domain-containing 4 (NLRC4) is downregulated in epithelial tumor cells of patients with colorectal cancer (CRC) by using spatial tissue imaging. Strikingly, only the loss of tumor NLRC4, but not stromal NLRC4, was associated with poor immune infiltration (mainly DCs and CD4+ and CD8+ T cells) and accurately predicted progression to metastatic stage IV and decrease in overall survival. By combining multiomics approaches, we show that restoring NLRC4 expression in human CRC cells triggered a broad inflammasome-independent immune reprogramming consisting of type I interferon (IFN) signaling genes and the release of chemokines and myeloid growth factors involved in the tumor infiltration and activation of DCs and T cells. Consistently, such reprogramming in cancer cells was sufficient to directly induce maturation of human DCs toward a Th1 antitumor immune response through IL-12 production in vitro. In multiple human carcinomas (colorectal, lung, and skin), we confirmed that NLRC4 expression in patient tumors was strongly associated with type I IFN genes, immune infiltrates, and high microsatellite instability. Thus, we shed light on the epithelial innate immune sensor NLRC4 as a therapeutic target to promote an efficient antitumor immune response against the aggressiveness of various carcinomas.


Subject(s)
Calcium-Binding Proteins , Colorectal Neoplasms , Gene Expression Regulation, Neoplastic , Interferon Type I , Signal Transduction , Humans , Calcium-Binding Proteins/genetics , Interferon Type I/metabolism , Interferon Type I/immunology , Interferon Type I/genetics , Signal Transduction/immunology , Colorectal Neoplasms/immunology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , CARD Signaling Adaptor Proteins/genetics , CARD Signaling Adaptor Proteins/metabolism , Female , Dendritic Cells/immunology , Dendritic Cells/metabolism , Male , Cell Line, Tumor , Lymphocytes, Tumor-Infiltrating/immunology , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology
6.
J Clin Invest ; 134(11)2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38652549

ABSTRACT

CD8+ T cell dysfunction impedes antitumor immunity in solid cancers, but the underlying mechanisms are diverse and poorly understood. Extracellular matrix (ECM) composition has been linked to impaired T cell migration and enhanced tumor progression; however, impacts of individual ECM molecules on T cell function in the tumor microenvironment (TME) are only beginning to be elucidated. Upstream regulators of aberrant ECM deposition and organization in solid tumors are equally ill-defined. Therefore, we investigated how ECM composition modulates CD8+ T cell function in undifferentiated pleomorphic sarcoma (UPS), an immunologically active desmoplastic tumor. Using an autochthonous murine model of UPS and data from multiple human patient cohorts, we discovered a multifaceted mechanism wherein the transcriptional coactivator YAP1 promotes collagen VI (COLVI) deposition in the UPS TME. In turn, COLVI induces CD8+ T cell dysfunction and immune evasion by remodeling fibrillar collagen and inhibiting T cell autophagic flux. Unexpectedly, collagen I (COLI) opposed COLVI in this setting, promoting CD8+ T cell function and acting as a tumor suppressor. Thus, CD8+ T cell responses in sarcoma depend on oncogene-mediated ECM composition and remodeling.


Subject(s)
CD8-Positive T-Lymphocytes , Extracellular Matrix , Sarcoma , Tumor Microenvironment , YAP-Signaling Proteins , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Animals , Tumor Microenvironment/immunology , Mice , YAP-Signaling Proteins/immunology , YAP-Signaling Proteins/genetics , Humans , Extracellular Matrix/immunology , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Sarcoma/immunology , Sarcoma/pathology , Sarcoma/genetics , Sarcoma/metabolism , Collagen Type VI/genetics , Collagen Type VI/immunology , Collagen Type VI/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/immunology , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription Factors/immunology , Oncogenes , Neoplasm Proteins/immunology , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Collagen Type I/metabolism , Collagen Type I/genetics , Collagen Type I/immunology
7.
Curr Pharm Des ; 30(11): 868-876, 2024.
Article in English | MEDLINE | ID: mdl-38482625

ABSTRACT

BACKGROUND: Interleukin 2 (IL-2) is a vital cytokine in the induction of T and NK cell responses, the proliferation of CD8+ T cells, and the effective treatment of human cancers such as melanoma and renal cell carcinoma. However, widespread use of this cytokine is limited due to its short half-life, severe toxicity, lack of specific tumor targeting, and activation of Treg cells mediated by high-affinity interleukin-2 receptors. OBJECTIVE: In this study, a tumor-targeting LIV-1 VHH-mutIL2 immunocytokine with reduced CD25 (α chain of the high-affinity IL-2 receptor) binding activity was developed to improve IL-2 half-life by decreasing its renal infiltration in comparison with wild and mutant IL-2 molecules. METHODS: The recombinant immunocytokine was designed and expressed. The biological activity of the purified fusion protein was investigated in in vitro and in vivo experiments. RESULTS: The fusion protein represented specific binding to MCF7 (the breast cancer cell line) and more efficient cytotoxicity than wild-type IL-2 and mutant IL-2. The PK parameters of the recombinant immunocytokine were also improved in comparison to the IL-2 molecules. CONCLUSION: The observed results showed that LIV1-mIL2 immunocytokine could be considered as an effective agent in the LIV-1-targeted treatment of cancers due to its longer half-life and stronger cytotoxicity.


Subject(s)
Antineoplastic Agents , Interleukin-2 , Humans , Interleukin-2/metabolism , Interleukin-2/immunology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Mice , Female , Neoplasm Proteins/immunology , Neoplasm Proteins/metabolism , Neoplasm Proteins/genetics , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , MCF-7 Cells , Single-Domain Antibodies/immunology , Single-Domain Antibodies/pharmacology , Mice, Inbred BALB C , Recombinant Fusion Proteins/pharmacology , Recombinant Fusion Proteins/genetics , Adaptor Proteins, Signal Transducing , Inhibitor of Apoptosis Proteins
8.
J Clin Invest ; 134(10)2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38530357

ABSTRACT

Despite widespread utilization of immunotherapy, treating immune-cold tumors remains a challenge. Multiomic analyses and experimental validation identified the OTUD4/CD73 proteolytic axis as a promising target in treating immune-suppressive triple negative breast cancer (TNBC). Mechanistically, deubiquitylation of CD73 by OTUD4 counteracted its ubiquitylation by TRIM21, resulting in CD73 stabilization inhibiting tumor immune responses. We further demonstrated the importance of TGF-ß signaling for orchestrating the OTUD4/CD73 proteolytic axis within tumor cells. Spatial transcriptomics profiling discovered spatially resolved features of interacting malignant and immune cells pertaining to expression levels of OTUD4 and CD73. In addition, ST80, a newly developed inhibitor, specifically disrupted proteolytic interaction between CD73 and OTUD4, leading to reinvigoration of cytotoxic CD8+ T cell activities. In preclinical models of TNBC, ST80 treatment sensitized refractory tumors to anti-PD-L1 therapy. Collectively, our findings uncover what we believe to be a novel strategy for targeting the immunosuppressive OTUD4/CD73 proteolytic axis in treating immune-suppressive breast cancers with the inhibitor ST80.


Subject(s)
5'-Nucleotidase , Proteolysis , Triple Negative Breast Neoplasms , Animals , Female , Humans , Mice , 5'-Nucleotidase/genetics , 5'-Nucleotidase/immunology , 5'-Nucleotidase/antagonists & inhibitors , Cell Line, Tumor , GPI-Linked Proteins/immunology , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , GPI-Linked Proteins/antagonists & inhibitors , Neoplasm Proteins/immunology , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasm Proteins/antagonists & inhibitors , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/pathology , Ubiquitination , Ubiquitin-Specific Proteases
9.
Life Sci ; 336: 122314, 2024 Jan 01.
Article in English | MEDLINE | ID: mdl-38030057

ABSTRACT

Nucleus accumbens-associated protein 1 (NACC1) is a member of the broad complex, tramtrack, bric-a-brac/poxvirus and zinc finger (BTB/POZ) protein families, mainly exerting its biological functions as a transcription co-regulator. NACC1 forms homo- or hetero-dimers through the BTB/POZ or BANP, E5R, and NACC1 (BEN) domain with other transcriptional regulators to regulate downstream signals. Recently, the overexpression of NACC1 has been observed in various tumors and is positively associated with tumor progression, high recurrence rate, indicating poor prognosis. NACC1 also regulates biological processes such as embryonic development, stem cell pluripotency, innate immunity, and related diseases. Our review combines recent research to summarize advancements in the structure, biological functions, and relative molecular mechanisms of NACC1. The future development of NACC1 clinical appliances is also discussed.


Subject(s)
Neoplasm Proteins , Neoplasms , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Neoplasms/genetics , Neoplasms/immunology , Nervous System Diseases/genetics , Nervous System Diseases/immunology , Gene Expression , Humans
10.
Dis Markers ; 2022: 6408879, 2022.
Article in English | MEDLINE | ID: mdl-35968498

ABSTRACT

Previous studies have confirmed long noncoding RNA LEMD1-AS1 (LEMD1-AS1) as a functional factor in several tumors. The present work is aimed at exploring the prognostic and diagnostic values of LEMD1-AS1 in patients with epithelial ovarian cancer (EOC). We examined the expressions of LEMD1-AS1 in pan-cancer from TCGA microarray datasets and GTEx Project. The expressions of LEMD1-AS1 were detected by qRT-PCR in EOC specimens and normal ovarian specimens from 30 EOC patients. The χ 2 test was applied to compare the clinicopathological characteristics of different groups. ROC curves were established to determine the diagnostic values of LEMD1-AS1 in screening EOC tissues. The association of LEMD1-AS1 expression with clinical outcome was determined by the Kaplan-Meier methods and COX assays. A decreased expression of LEMD1-AS1 was observed in EOC tissues compared to matched normal specimens (p < 0.01). Low LEMD1-AS1 expression could be used to distinguish EOC from adjacent normal specimens. A clinical study revealed that patients with low LEMD1-AS1 expression have a shorter overall survival (p = 0.035) and progress-free interval (p = 0.041) than those with high LEMD1-AS1 expression. The Spearman correlation test revealed that LEMD1-AS1 expressions were negatively associated with the expressions of neutrophil and myeloid dendritic cell. Overall, our finding suggested that LEMD1-AS1 may have potential roles as a potential biomarker and/or a therapeutic target in EOC.


Subject(s)
Carcinoma, Ovarian Epithelial , Neoplasm Proteins , Ovarian Neoplasms , RNA, Antisense , Carcinoma, Ovarian Epithelial/diagnosis , Carcinoma, Ovarian Epithelial/genetics , Carcinoma, Ovarian Epithelial/immunology , Carcinoma, Ovarian Epithelial/pathology , Down-Regulation , Female , Humans , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Ovarian Neoplasms/diagnosis , Ovarian Neoplasms/genetics , Ovarian Neoplasms/immunology , Ovarian Neoplasms/pathology , Prognosis , RNA, Antisense/genetics , RNA, Antisense/immunology , RNA, Long Noncoding/genetics , RNA, Long Noncoding/immunology
11.
Science ; 376(6588): 17, 2022 04.
Article in English | MEDLINE | ID: mdl-35357932

ABSTRACT

Small versions of antibodies bind to virtually any target protein.


Subject(s)
Antibodies/chemistry , Antibodies/immunology , Drug Design/methods , Software , Antibodies/therapeutic use , Neoplasm Proteins/immunology , Protein Folding , Viral Proteins/immunology
12.
BMC Cancer ; 22(1): 154, 2022 Feb 08.
Article in English | MEDLINE | ID: mdl-35135489

ABSTRACT

BACKGROUND: Immune checkpoint inhibitors prolong the survival of non-small cell lung cancer (NSCLC) patients. Although it has been acknowledged that there is some correlation between the efficacy of anti-programmed cell death-1 (PD-1) antibody therapy and immunohistochemical analysis, this technique is not yet considered foolproof for predicting a favorable outcome of PD-1 antibody therapy. We aimed to predict the efficacy of nivolumab based on a comprehensive analysis of RNA expression at the gene level in advanced NSCLC. METHODS: This was a retrospective study on patients with NSCLC who were administered nivolumab at the Kansai Medical University Hospital. To identify genes associated with response to anti-PD-1 antibodies, we grouped patients into responders (complete and partial response) and non-responders (stable and progressive disease) to nivolumab therapy. Significant genes were then identified for these groups using Welch's t-test. RESULTS: Among 42 analyzed cases (20 adenocarcinomas and 22 squamous cell carcinomas), enhanced expression of MAGE-A4, BBC3, and OTOA genes was observed in responders with adenocarcinoma, and enhanced expression of DAB2, HLA-DPB,1 and CDH2 genes was observed in responders with squamous cell carcinoma. CONCLUSIONS: This study predicted the efficacy of nivolumab based on a comprehensive analysis of mRNA expression at the gene level in advanced NSCLC. We also revealed different gene expression patterns as predictors of the effectiveness of anti PD-1 antibody therapy in adenocarcinoma and squamous cell carcinoma.


Subject(s)
Adenocarcinoma/drug therapy , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Squamous Cell/drug therapy , Immune Checkpoint Inhibitors/therapeutic use , Lung Neoplasms/drug therapy , Nivolumab/therapeutic use , Adaptor Proteins, Signal Transducing/immunology , Adenocarcinoma/immunology , Adult , Aged , Aged, 80 and over , Antigens, CD/immunology , Antigens, Neoplasm/immunology , Apoptosis Regulatory Proteins/immunology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/immunology , Cadherins/immunology , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Squamous Cell/immunology , Drug Resistance, Neoplasm/genetics , Drug Resistance, Neoplasm/immunology , Female , GPI-Linked Proteins/immunology , Gene Expression/drug effects , Gene Expression/immunology , HLA-DP beta-Chains/immunology , Humans , Lung Neoplasms/immunology , Male , Middle Aged , Neoplasm Proteins/immunology , Predictive Value of Tests , Programmed Cell Death 1 Receptor/drug effects , Programmed Cell Death 1 Receptor/immunology , Proto-Oncogene Proteins/immunology , RNA, Messenger/drug effects , RNA, Messenger/immunology , Retrospective Studies , Treatment Outcome
13.
Biochem Biophys Res Commun ; 598: 69-73, 2022 04 02.
Article in English | MEDLINE | ID: mdl-35151206

ABSTRACT

Two variants of Ca2+-regulated photoprotein obelin, extended from the N-terminus with small tumor markers - melanoma inhibitory activity protein (MIA) and survivin, one of the protein inhibitors of apoptosis, were designed, obtained and studied. Both domains in the obtained hybrid proteins exhibit the properties of the initial molecules: the main features of Ca2+-triggered bioluminescence are close to those of obelin, and the tumor markers' domains are recognized and bound by the corresponding antibodies. The obtained hybrids compete with the corresponding tumor markers for binding with antibodies, immobilized on the surface and their use has been shown to be promising as bioluminescent labels in a one-stage solid-phase competitive immunoassay.


Subject(s)
Extracellular Matrix Proteins/blood , Immunoassay/methods , Luminescent Proteins , Neoplasm Proteins/blood , Survivin/blood , Biomarkers, Tumor/blood , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/immunology , Humans , Limit of Detection , Luminescent Measurements , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Protein Engineering/methods , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Survivin/genetics , Survivin/immunology
15.
J Clin Invest ; 132(4)2022 02 15.
Article in English | MEDLINE | ID: mdl-35166238

ABSTRACT

Extracellular proteolysis is frequently dysregulated in disease and can generate proteoforms with unique neoepitopes not found in healthy tissue. Here, we demonstrate that Abs that selectively recognize a proteolytic neoepitope on CUB domain containing protein 1 (CDCP1) could enable more effective and safer treatments for solid tumors. CDCP1 is highly overexpressed in RAS-driven cancers, and its ectodomain is cleaved by extracellular proteases. Biochemical, biophysical, and structural characterization revealed that the 2 cleaved fragments of CDCP1 remain tightly associated with minimal proteolysis-induced conformational change. Using differential phage display, we generated recombinant Abs that are exquisitely selective to cleaved CDCP1 with no detectable binding to the uncleaved form. These Abs potently targeted cleaved CDCP1-expressing cancer cells as an Ab-drug conjugate, an Ab-radionuclide conjugate, and a bispecific T cell engager. In a syngeneic pancreatic tumor model, these cleaved-specific Abs showed tumor-specific localization and antitumor activity with superior safety profiles compared with a pan-CDCP1 approach. Targeting proteolytic neoepitopes could provide an orthogonal "AND" gate for improving the therapeutic index.


Subject(s)
Antigens, Neoplasm/immunology , Cell Adhesion Molecules/immunology , Epitopes/immunology , Neoplasm Proteins/immunology , Neoplasms, Experimental/immunology , Pancreatic Neoplasms/immunology , Proteolysis , Animals , Antigens, Neoplasm/genetics , Cell Adhesion Molecules/genetics , Cell Line, Tumor , Epitopes/genetics , Humans , Male , Mice , Mice, Nude , Neoplasm Proteins/genetics , Neoplasms, Experimental/genetics , Pancreatic Neoplasms/genetics
16.
Int J Mol Sci ; 23(3)2022 Jan 27.
Article in English | MEDLINE | ID: mdl-35163401

ABSTRACT

Conjunctival melanoma (CM) accounts for 5% of all ocular melanomas and arises from malignantly transformed melanocytes in the conjunctival epithelium. Current therapies using surgical excision in combination with chemo- or cryotherapy still have high rates for recurrences and metastatic disease. Lately, novel signal transduction-targeted and immune checkpoint inhibitors like cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) inhibitors, programmed cell death protein-1 (PD-1) receptor inhibitors, BRAF- or MEK-inhibitors for systemic treatment of melanoma have improved the outcome even for unresectable cutaneous melanoma, improving patient survival dramatically. The use of these therapies is now also recommended for CM; however, the immunological background of CM is barely known, underlining the need for research to better understand the immunological basics when treating CM patients with immunomodulatory therapies. Immune checkpoint inhibitors activate tumor defense by interrupting inhibitory interactions between tumor cells and T lymphocytes at the so-called checkpoints. The tumor cells exploit these inhibitory targets on T-cells that are usually used by dendritic cells (DCs). DCs are antigen-presenting cells at the forefront of immune response induction. They contribute to immune tolerance and immune defense but in the case of tumor development, immune tolerance is often prevalent. Enhancing the immune response via DCs, interfering with the lymphatic pathways during immune cell migration and tumor development and specifically targeting tumor cells is a major therapeutic opportunity for many tumor entities including CM. This review summarizes the current knowledge on the function of lymphatic vessels in tumor growth and immune cell transport and continues to compare DC subsets in CM with related melanomas, such as cutaneous melanoma and mucosal melanoma.


Subject(s)
Conjunctival Neoplasms , Dendritic Cells , Immune Checkpoint Inhibitors/therapeutic use , Immunotherapy , Lymphatic Vessels , Melanoma , Neoplasm Proteins/immunology , Skin Neoplasms , Animals , Conjunctival Neoplasms/immunology , Conjunctival Neoplasms/pathology , Conjunctival Neoplasms/therapy , Dendritic Cells/immunology , Dendritic Cells/pathology , Humans , Lymphatic Vessels/immunology , Lymphatic Vessels/pathology , Melanoma/immunology , Melanoma/pathology , Melanoma/therapy , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Skin Neoplasms/therapy , Melanoma, Cutaneous Malignant
17.
Bioengineered ; 13(2): 2412-2429, 2022 02.
Article in English | MEDLINE | ID: mdl-35094638

ABSTRACT

This study investigated the role of ADAM metallopeptidase domain 12 (ADAM12) in clear cell renal cell carcinoma (ccRCC). The mRNA expression of ADAM12 was analyzed using The Cancer Genome Atlas (TCGA) database and the Gene Expression Omnibus (GEO) database, and the protein expression level of ADAM12 in renal clear cell carcinoma cell lines was detected by Western blot analysis. The Wilcoxon rank-sum test, logistic regression analysis, Cox regression analysis, and Kaplan-Meier analysis were used to assess the relationship between the clinicopathological characteristics and the prognosis of ccRCC patients and ADAM12 expression. The miRNAs and lncRNAs associated with ADAM12 were predicted, and a ceRNA network was constructed using the Starbase database. Gene Set Enrichment Analysis (GSEA) and Gene Ontology (GO) analysis were used to identify relevant pathways. The relationship between ADAM12 and immune infiltration and checkpoints was analyzed using the TIMER and Gene Expression Profiling Interactive Analysis (GEPIA) databases. The results showed that ADAM12 expression was increased in ccRCC tissues and cells and significantly correlated with patient gender, Tumor stage, Metastasis stage, Node stage, and clinical grade. Survival analysis showed that ccRCC patients with high ADAM12 expression had a low overall survival rate. Univariate and multivariate Cox regression analyses showed that ADAM12 was an independent prognostic factor. Enrichment analysis showed that ADAM12 expression was associated with immune-related pathways. Immune infiltration analysis showed that ADAM12 expression was related to immune cell infiltration, PD-1, PD-L1, and CTLA4. These results suggest that ADAM12 may be a potential diagnostic and prognostic biomarker for ccRCC.


Subject(s)
ADAM12 Protein/immunology , Carcinoma, Renal Cell/immunology , Gene Expression Regulation, Enzymologic/immunology , Gene Expression Regulation, Neoplastic/immunology , Kidney Neoplasms/immunology , Neoplasm Proteins/immunology , Carcinoma, Renal Cell/diagnosis , Cell Line, Tumor , Humans , Kidney Neoplasms/diagnosis , Prognosis
18.
Signal Transduct Target Ther ; 7(1): 19, 2022 01 20.
Article in English | MEDLINE | ID: mdl-35046386

ABSTRACT

Interleukin-37b (hereafter called IL-37) was identified as fundamental inhibitor of natural and acquired immunity. The molecular mechanism and function of IL-37 in colorectal cancer (CRC) has been elusive. Here, we found that IL-37 transgenic (IL-37tg) mice were highly susceptible to colitis-associated colorectal cancer (CAC) and suffered from dramatically increased tumor burdens in colon. Nevertheless, IL-37 is dispensable for intestinal mutagenesis, and CRC cell proliferation, apoptosis, and migration. Notably, IL-37 dampened protective cytotoxic T cell-mediated immunity in CAC and B16-OVA models. CD8+ T cell dysfunction is defined by reduced retention and activation as well as failure to proliferate and produce cytotoxic cytokines in IL-37tg mice, enabling tumor evasion of immune surveillance. The dysfunction led by IL-37 antagonizes IL-18-induced proliferation and effector function of CD8+ T cells, which was dependent on SIGIRR (single immunoglobulin interleukin-1 receptor-related protein). Finally, we observed that IL-37 levels were significantly increased in CRC patients, and positively correlated with serum CRC biomarker CEA levels, but negatively correlated with the CD8+ T cell infiltration in CRC patients. Our findings highlight the role of IL-37 in harnessing antitumor immunity by inactivation of cytotoxic T cells and establish a new defined inhibitory factor IL-37/SIGIRR in cancer-immunity cycle as therapeutic targets in CRC.


Subject(s)
Carcinogenesis/immunology , Colitis/immunology , Colorectal Neoplasms/immunology , Interleukin-1/immunology , Neoplasm Proteins/immunology , Receptors, Interleukin-1/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Carcinogenesis/genetics , Colitis/genetics , Colitis/pathology , Colorectal Neoplasms/genetics , Interleukin-1/genetics , Mice , Mice, Transgenic , Neoplasm Proteins/genetics , Receptors, Interleukin-1/genetics
19.
J Clin Lab Anal ; 36(2): e24232, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34995016

ABSTRACT

BACKGROUND: Combined biomarkers can improve the sensitivity and specificity of ovarian cancer (OC) diagnosis and effectively predict patient prognosis. This study explored the diagnostic and prognostic values of serum CCL18 and CXCL1 antigens combined with C1D, FXR1, ZNF573, and TM4SF1 autoantibodies in OC. METHODS: CCL18 and CXCL1 monoclonal antibodies and C1D, FXR1, ZNF573, and TM4SF1 antigens were coated with microspheres. Logistic regression was used to construct a serum antigen-antibody combined detection model; receiver-operating characteristic curve (ROC) was used to evaluate the diagnostic efficacy of the model; and the Kaplan-Meier method and Cox regression models were used for survival analysis to evaluate the prognosis of OC. Data from The Cancer Genome Atlas (TCGA) and Genotype-Tissue Expression (GTEx) projects and online survival analysis tools were used to evaluate prognostic genes for OC. The CIBERSORT immune score was used to explore the factors influencing prognosis and their relationship with tumor-infiltrating immune cells. RESULTS: The levels of each index in the blood samples of patients with OC were higher than those of the other groups. The combined detection model has higher specificity and sensitivity in the diagnosis of OC, and its diagnostic efficiency is better than that of CA125 alone and diagnosing other malignant tumors. CCL18 and TM4SF1 may be factors affecting the prognosis of OC, and CCL18 may be related to immune-infiltrating cells. CONCLUSIONS: The serum antigen-antibody combined detection model established in this study has high sensitivity and specificity for the diagnosis of OC.


Subject(s)
Autoantibodies/blood , Biomarkers, Tumor/blood , Chemokine CXCL1/blood , Chemokines, CC/blood , Ovarian Neoplasms/diagnosis , Antigens, Surface/immunology , Co-Repressor Proteins/immunology , Female , Humans , Immunoglobulin G/blood , Neoplasm Proteins/immunology , Prognosis , RNA-Binding Proteins/immunology , Reference Values , Sensitivity and Specificity
20.
Biomed Res Int ; 2022: 5092505, 2022.
Article in English | MEDLINE | ID: mdl-35036435

ABSTRACT

BACKGROUND: Sufficient evidence indicated the crucial role of NF-κB family played in gastric cancer (GC). The novel discovery that NF-κB could regulate cancer metabolism and immune evasion greatly increased its attraction in cancer research. However, the correlation among NF-κB, metabolism, and cancer immunity in GC still requires further improvement. METHODS: TCGA, hTFtarget, and MSigDB databases were employed to identify NF-κB-related metabolic genes (NFMGs). Based on NFMGs, we used consensus clustering to divide GC patients into two subtypes. GSVA was employed to analyze the enriched pathway. ESTIMATE, CIBERSORT, ssGSEA, and MCPcounter algorithms were applied to evaluate immune infiltration in GC. The tumor immune dysfunction and exclusion (TIDE) algorithm was used to predict patients' response to immunotherapy. We also established a NFMG-related risk score by using the LASSO regression model and assessed its efficacy in TCGA and GSE62254 datasets. RESULTS: We used 27 NFMGs to conduct an unsupervised clustering on GC samples and classified them into two clusters. Cluster 1 was characterized by high active metabolism, tumor mutant burden, and microsatellite instability, while cluster 2 was featured with high immune infiltration. Compared to cluster 2, cluster 1 had a better prognosis and higher response to immunotherapy. In addition, we constructed a 12-NFMG (ADCY3, AHCY, CHDH, GUCY1A2, ITPA, MTHFD2, NRP1, POLA1, POLR1A, POLR3A, POLR3K, and SRM) risk score. Followed analysis indicated that this risk score acted as an effectively prognostic factor in GC. CONCLUSION: Our data suggested that GC subtypes classified by NFMGs may effectively guide prognosis and immunotherapy. Further study of these NFMGs will deepen our understanding of NF-κB-mediated cancer metabolism and immunity.


Subject(s)
Biomarkers, Tumor/immunology , Gene Expression Regulation, Neoplastic/immunology , NF-kappa B/metabolism , Neoplasm Proteins/immunology , Stomach Neoplasms , Tumor Microenvironment/immunology , Humans , Neoplasm Proteins/metabolism , Prognosis , Stomach Neoplasms/diagnosis , Stomach Neoplasms/genetics , Stomach Neoplasms/immunology , Stomach Neoplasms/therapy , Tumor Microenvironment/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...