Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Clin Transl Med ; 14(5): e1674, 2024 May.
Article in English | MEDLINE | ID: mdl-38685486

ABSTRACT

BACKGROUND: The current standard of care for locally advanced gastric cancer (GC) involves neoadjuvant chemotherapy followed by radical surgery. Recently, neoadjuvant treatment for this condition has involved the exploration of immunotherapy plus chemotherapy as a potential approach. However, the efficacy remains uncertain. METHODS: A single-arm, phase 2 study was conducted to evaluate the efficacy and tolerability of neoadjuvant camrelizumab combined with mFOLFOX6 and identify potential biomarkers of response through multi-omics analysis in patients with resectable locally advanced GC. The primary endpoint was the pathological complete response (pCR) rate. Secondary endpoints included the R0 rate, near pCR rate, progression-free survival (PFS), disease-free survival (DFS), and overall survival (OS). Multi-omics analysis was assessed by whole-exome sequencing, transcriptome sequencing, and multiplex immunofluorescence (mIF) using biopsies pre- and post-neoadjuvant therapy. RESULTS: This study involved 60 patients, of which 55 underwent gastrectomy. Among these, five (9.1%) attained a pathological complete response (pCR), and 11 (20.0%) reached near pCR. No unexpected treatment-emergent adverse events or perioperative mortality were observed, and the regimen presented a manageable safety profile. Molecular changes identified through multi-omics analysis correlated with treatment response, highlighting associations between HER2-positive and CTNNB1 mutations with treatment sensitivity and a favourable prognosis. This finding was further supported by immune cell infiltration analysis and mIF. Expression data uncovered a risk model with four genes (RALYL, SCGN, CCKBR, NTS) linked to poor response. Additionally, post-treatment infiltration of CD8+ T lymphocytes positively correlates with pathological response. CONCLUSION: The findings suggest the combination of PD-1-inhibitor and mFOLFOX6 showed efficacy and acceptable toxicity for locally advanced GC. Extended follow-up is required to determine the duration of the response. This study lays essential groundwork for developing precise neoadjuvant regimens.


Subject(s)
Antibodies, Monoclonal, Humanized , Antineoplastic Combined Chemotherapy Protocols , Neoadjuvant Therapy , Stomach Neoplasms , Humans , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Male , Female , Middle Aged , Neoadjuvant Therapy/methods , Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal, Humanized/pharmacology , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Adult , Leucovorin/therapeutic use , Fluorouracil/therapeutic use , Organoplatinum Compounds/therapeutic use , Organoplatinum Compounds/pharmacology , Treatment Outcome , Multiomics
2.
Am J Cancer Res ; 13(10): 4708-4720, 2023.
Article in English | MEDLINE | ID: mdl-37970356

ABSTRACT

BACKGROUND: Although sulforaphene has potential anticancer effects, little is known about its effect on oesophageal squamous cell carcinoma (ESCC) invasiveness. METHODS: To investigate whether sulforaphene inhibits the growth of oesophageal cancer cells, MTT and anchorage-independent cell growth assays were performed. Global changes in the proteome and phosphoproteome of oesophageal cancer cells after sulforaphene treatment were analysed by mass spectrometry (MS), and the underlying molecular mechanism was further verified by in vivo and in vitro experiments. RESULTS: Sulforaphene treatment markedly affected proteins that regulate several cellular processes in oesophageal cancer cells, and mitogen- and stress-activated kinase 2 (MSK2) was the main genetic target of sulforaphene in reducing the growth of oesophageal cancer cells. Sulforaphene significantly suppressed ESCC cell proliferation in vitro and reduced the tumour size in an oesophageal patient-derived xenograft (PDX) SCID mouse model. Furthermore, the binding of sulforaphane to MSK2 in vitro was verified using a cellular thermal dhift assay, and the effect of MSK2 knockdown on the ESCC phenotype was observed using a shMSK2 model. CONCLUSION: The results showed that sulforaphene suppresses ESCC growth in both human oesophageal squamous cells and PDX mouse model by inhibiting MSK2 expression, implicating sulforaphene as a promising candidate for ESCC treatment.

3.
Oncogene ; 42(32): 2456-2470, 2023 08.
Article in English | MEDLINE | ID: mdl-37400530

ABSTRACT

Colorectal cancer (CRC) is a highly aggressive cancer in which metastasis plays a key role. However, the mechanisms underlying metastasis have not been fully elucidated. Peroxisome proliferator-activated receptor gamma coactivator 1α (PGC-1α), a regulator of mitochondrial function, has been reported as a complicated factor in cancer. In this study, we found that PGC-1α was highly expressed in CRC tissues and was positively correlated with lymph node and liver metastasis. Subsequently, PGC-1α knockdown was shown to inhibit CRC growth and metastasis in both in vitro and in vivo studies. Transcriptomic analysis revealed that PGC-1α regulated ATP-binding cassette transporter 1 (ABCA1) mediated cholesterol efflux. Mechanistically, PGC-1α interacted with YY1 to promote ABCA1 transcription, resulting in cholesterol efflux, which subsequently promoted CRC metastasis through epithelial-to-mesenchymal transition (EMT). In addition, the study identified the natural compound isoliquiritigenin (ISL) as an inhibitor that targeted ABCA1 and significantly reduced CRC metastasis induced by PGC-1α. Overall, this study sheds light on how PGC-1α promotes CRC metastasis by regulating ABCA1-mediated cholesterol efflux, providing a basis for further research to inhibit CRC metastasis.


Subject(s)
Colorectal Neoplasms , Mitochondria , Humans , Mitochondria/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Cholesterol , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , ATP Binding Cassette Transporter 1/genetics
4.
Front Immunol ; 14: 1145028, 2023.
Article in English | MEDLINE | ID: mdl-37205112

ABSTRACT

Immunotherapy has emerged as an effective therapeutic approach to several cancer types. The reinvigoration of tumor-infiltrating lymphocyte-mediated immune responses via the blockade of immune checkpoint markers, such as program cell death-1 (PD-1) or its cognate ligand PD-L1, has been the basis for developing clinically effective anticancer therapies. We identified pentamidine, an FDA-approved antimicrobial agent, as a small-molecule antagonist of PD-L1. Pentamidine enhanced T-cell-mediated cytotoxicity against various cancer cells in vitro by increasing the secretion of IFN-γ, TNF-α, perforin, and granzyme B in the culture medium. Pentamidine promoted T-cell activation by blocking the PD-1/PD-L1 interaction. In vivo administration of pentamidine attenuated the tumor growth and prolonged the survival of tumor-bearing mice in PD-L1 humanized murine tumor cell allograft models. Histological analysis of tumor tissues showed an increased number of tumor-infiltrating lymphocytes in tissues derived from pentamidine-treated mice. In summary, our study suggests that pentamidine holds the potential to be repurposed as a novel PD-L1 antagonist that may overcome the limitations of monoclonal antibody therapy and can emerge as a small molecule cancer immunotherapy.


Subject(s)
Neoplasms , Pentamidine , Mice , Animals , Pentamidine/pharmacology , B7-H1 Antigen , Programmed Cell Death 1 Receptor , Immunotherapy , Neoplasms/therapy
5.
Am J Cancer Res ; 12(11): 5255-5270, 2022.
Article in English | MEDLINE | ID: mdl-36504888

ABSTRACT

Heterogeneity is a fundamental feature of human tumors and plays a major role in drug resistance and disease progression. In the present study, we selected single-cell-derived cell lines (SCDCLs) derived from Lewis lung carcinoma (LLC1) cells to investigate tumorigenesis and heterogeneity. SCDCLs were generated using limiting dilution. Five SCDCLs were subcutaneously injected into wild-type C57BL/6N mice; however, they displayed significant differences in tumor growth. Subclone SCC1 grew the fastest in vivo, whereas it grew slower in vitro. The growth pattern of SCC2 was the opposite to that of SCC1. Genetic differences in these two subclones showed marked differences in cell adhesion and proliferation. Pathway enrichment results indicate that signal transduction and immune system responses were the most significantly altered functional categories in SCC2 cells compared to those in SCC1 cells in vitro. The number and activation of CD3+ and CD8+ T cells and NK cells in the tumor tissue of tumor-bearing mice inoculated with SCC2 were significantly higher, whereas those of myeloid cells were significantly lower, than those in the SCC1 and LLC1 groups. Our results suggest that the in vivo growth of two subclones derived from LLC1 was determined by the tumor microenvironment rather than their intrinsic proliferative cell characteristics.

6.
Int Immunopharmacol ; 113(Pt A): 109286, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36252474

ABSTRACT

The high heterogeneity of tumor cells and the surrounding immune microenvironment affects the response to treatment in colorectal cancer (CRC) patients. Therefore, there is a need to identify new immune biomarkers to predict the treatment efficacy of CRC. This study aimed to explore the predictive value of tumor-infiltrating lymphocytes (TIL) for survival in CRC patients. Flow cytometry and gated analysis were performed to measure the TILs in tissue samples obtained from 536 CRC patients. The COX regression analysis showed that the CD8 + CD279+ cells had the highest impact of all evaluated TILs on postoperative disease-free survival (DFS) (P < 0.05). The optimal CD8 + CD279+ cutoff point for the prediction of survival was 12.2%. The Kaplan-Meier analysis showed significantly higher DFS in the high CD8 + CD279+ group compared with the low CD8 + CD279+ group (P < 0.05). CD8 + CD279+ cells were associated with DFS in CRC patients with the KARS mutation, MSI/MMR, perineural invasion, and those treated with neoadjuvant chemotherapy and other chemotherapeutic treatments (P < 0.05). After the multivariate adjustment, the expression of CD8 + CD279+ remained an independent risk factor for DFS. Overall, the CD8 + CD279+ cells were identified as an independent prognostic factor in CRC patients and could be used as a potential marker for postoperative DFS.


Subject(s)
Colorectal Neoplasms , Lymphocytes, Tumor-Infiltrating , Humans , Flow Cytometry , Colorectal Neoplasms/pathology , CD8-Positive T-Lymphocytes , Kaplan-Meier Estimate , Biomarkers/metabolism , Prognosis , Tumor Microenvironment
7.
Oncogene ; 41(42): 4736-4753, 2022 10.
Article in English | MEDLINE | ID: mdl-36109629

ABSTRACT

Abnormal translation of the MYC proto-oncogene is a hallmark of the initiation and maintenance of tumorigenesis. However, the molecular mechanism underlying increased MYC protein levels in certain cancer types without a corresponding increase in MYC mRNA levels is unclear. Here, we identified a novel lncRNA, MTAR1, which is critical for post-transcriptional regulation of MYC-induced tumorigenesis. MTAR1 is essential for recruiting IGF2BPs into PABP1-mediated liquid-liquid phase separation (LLPS) complexes and facilitates IGF2BPs-mediated MYC mRNA translation. MTAR1 enhanced binding between IGF2BPs and PABP1, thereby promoting MYC mRNA stability and increased MYC mRNA translation. In summary, MTAR1 is a novel MYC-related lncRNA that contributes to tumor progression by enhancing MYC translation through mediating PABP1/IGF2BPs liquid-liquid phase separation.


Subject(s)
Neoplasms , Proto-Oncogene Proteins c-myc , RNA, Long Noncoding , Carcinogenesis/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Neoplasms/genetics , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics
8.
Cell Death Dis ; 13(2): 189, 2022 02 25.
Article in English | MEDLINE | ID: mdl-35217647

ABSTRACT

Esophageal squamous cell carcinoma (ESCC), a malignant neoplasm with high incidence, is a severe global public health threat. The current modalities used for treating ESCC include surgery, chemotherapy, and radiotherapy. Although ESCC management and treatment strategies have improved over the last decade, the overall 5-year survival rate remains <20%. Therefore, the identification of novel therapeutic strategies that can increase ESCC patient survival rates is urgently needed. Oxethazaine, an amino-amide anesthetic agent, is mainly prescribed in combination with antacids to relieve esophagitis, dyspepsia, and other gastric disorders. In the present study, we found that oxethazaine inhibited the proliferation and migration of esophageal cancer cells. According to the results of in vitro screening and binding assays, oxethazaine binds directly to AURKA, suppresses AURKA activity, and inhibits the downstream effectors of AURKA. Notably, we found that oxethazaine suppressed tumor growth in three patient-derived esophageal xenograft mouse models and tumor metastasis in vivo. Our findings suggest that oxethazaine can inhibit ESCC proliferation and metastasis in vitro and in vivo by targeting AURKA.


Subject(s)
Aurora Kinase A , Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Ethanolamines , Animals , Aurora Kinase A/antagonists & inhibitors , Aurora Kinase A/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/drug therapy , Esophageal Squamous Cell Carcinoma/genetics , Ethanolamines/therapeutic use , Gene Expression Regulation, Neoplastic , Humans , Mice , Neoplasm Metastasis
9.
Front Immunol ; 13: 1056144, 2022.
Article in English | MEDLINE | ID: mdl-36685525

ABSTRACT

Background: While the tumor microenvironment (TME) affects immune checkpoint blockade (ICB) efficacy, ICB also reshapes the characteristics of TME. Thus far, studies have focused on the TME evolution during neoadjuvant or adjuvant ICB therapy in gastric cancer (GC). However, the interaction between TME characteristics and neoadjuvant immunotherapy plus chemotherapy remains to be elucidated. Methods: We performed single-cell RNA sequencing on ten GC specimens pre- and post-neoadjuvant camrelizumab plus mFOLFOX6 to determine the impact of the TME on the efficacy of the combination therapy and the remodeling of TME by the therapy. Results: A high baseline interferon gamma (IFN-γ) signature in CD8+ T cells predicts better responses to the combination therapy. We also observed that the IFN-γ signature significantly decreased in multiple cell types, and the exhausted signature of CD8+ T cells was significantly suppressed during the neoadjuvant therapy. Conclusions: Our data reveal interactions between the TME and neoadjuvant immunotherapy plus chemotherapy in GC. Importantly, it also highlights the signature of CD8+ T cells in predicting response to the combination therapy in GC.


Subject(s)
CD8-Positive T-Lymphocytes , Stomach Neoplasms , Humans , Interferon-gamma/metabolism , Neoadjuvant Therapy , Stomach Neoplasms/therapy , Immunotherapy , Tumor Microenvironment
10.
FEBS Open Bio ; 11(10): 2784-2799, 2021 10.
Article in English | MEDLINE | ID: mdl-34428354

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the most fatal cancers worldwide. In this article, we show that expression of abnormal spindle-like microcephaly-associated protein (ASPM) is up-regulated in liver cancer samples, and this up-regulation is significantly associated with tumor aggressiveness and reduced survival times of patients. Down-regulation of ASPM expression inhibits the proliferation, invasion, migration and epithelial-to-mesenchymal transition of HCC cells in vitro and inhibits tumor formation in nude mice. ASPM interacts with disheveled-2 (Dvl2) and antagonizes autophagy-mediated Dvl2 degradation by weakening the functional interaction between Dvl2 and the lipidated form of microtubule-associated proteins 1A/1B light chain 3A (LC3II), thereby increasing Dvl2 protein abundance and leading to Wnt/ß-catenin signaling activation in HCC cells. Thus, our results define ASPM as a novel oncoprotein in HCC and indicate that disruption of the Wnt-ASPM-Dvl2-ß-catenin signaling axis might have potential clinical value.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Animals , Autophagy , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Dishevelled Proteins/genetics , Dishevelled Proteins/metabolism , Humans , Liver Neoplasms/pathology , Mice , Mice, Nude , Nerve Tissue Proteins/metabolism , beta Catenin/metabolism
11.
Zhongguo Gu Shang ; 34(7): 605-11, 2021 Jul 25.
Article in Chinese | MEDLINE | ID: mdl-34318634

ABSTRACT

OBJECTIVE: To analyze the influence of preoperative serum nutritional indexes and postoperative nutritional guidance on 1-year mortality in elderly patients with hip fracture. METHODS: From January 2015 to December 2017, 396 elderly patients with hip fracture were included in the study, including 267 females and 129 males, aged 68 to 80(75.48±2.62) years; the course of disease was 2 to 10 (6.12±1.35) days;all patients were followed up for 1-year, and were divided into death group and survival group according to whether the patients died or not. Multivariate logistic regression model was used to analyze the influencing factors of 1 year mortality. RESULTS: Duringthe follow-up, 4 patients lost contact and were treated as shedding, among which 67 patients died and 325 patients survived. The age, male patients, patients with more than three basic diseases, American Society of Anesthesiologists grade Ⅲ-Ⅳ and patients with postoperative complications in the death group were significantly higher than those in the survival group (all P<0.05). There was no significant difference in body mass index(BMI), number of smokers, fracture type and operation type (all P>0.05). The serum albumin (ALB), prealbumin (PA), lymphocyte (LYM), lymphocyte percentage(LYM%), hemoglobin(HB), transferrin(TRF), total protein(TP) in the death group were significantly lower than those in the survival group (t=5.884, 5.826, 2.020, 5.665, 4.726, 4.935, 2.862;all P<0.05). The number of patients receiving nutritional guidance in the death group was significantly less than that in the survival group (χ2=12.597, P= 0.000). There were no significant difference on white blood cell(WBC) and red blood cell(RBC) between two groups. Multivariate logistic regression analysis showed that old age, male and not receiving significant nutritional guidance were independent risk factors for 1 year mortality of elderly patients with hip fracture (OR=1.309, 43.548, 6.032;all P<0.05);high serum ALB, PA, HB, LYM% levels and combined with two or less basic diseases were protective factors (OR=0.958, 0. 913, 0.985, 0.954, 0.832; all P<0.05). CONCLUSION: Advanced age, male and multiple underlying diseases were independent risk factors for 1-year mortality in elderly patients with hip fracture, while higher preoperative nutritional level and routine nutritional guidance were protective factors.


Subject(s)
Hip Fractures , Aged , Female , Hip Fractures/surgery , Humans , Logistic Models , Male , Postoperative Complications , Postoperative Period , Retrospective Studies , Risk Factors
12.
Eur J Pharmacol ; 906: 174270, 2021 Sep 05.
Article in English | MEDLINE | ID: mdl-34171392

ABSTRACT

In addition to its pivotal role in purine metabolism, xanthine oxidoreductase (XOR) is one of the key enzymes involved in superoxide radical generation. Oxidative stress has been implicated in the etiology of colorectal cancer, but the contribution of XOR remains unclear. Here we investigated the role of XOR in colitis-associated colorectal cancer (CAC) and the underlying mechanisms. Using clinical samples, we demonstrated that XOR up-regulation was an early event in colonic carcinogenesis. Pharmacological inhibition of XOR effectively delayed the progression of CAC. Moreover, XOR activity positively correlated with tumor necrosis factor-alpha (TNFα) protein levels. Mechanistically, TNFα may activate XOR transcription via activator protein-1 and, thus, promote endogenous hydrogen peroxide generation, resulting in oxidative DNA damage in colon cancer cells. On the other hand, XOR may regulate the TNFα mRNA transcripts by mediating LPS-induced macrophage M1 polarization. Collectively, XOR promotes tumor development by programming the tumor microenvironment and stimulates CAC progression via DNA damage-induced genetic instability.


Subject(s)
Colitis-Associated Neoplasms/immunology , DNA Damage/immunology , Macrophages/immunology , Oxidative Stress/immunology , Xanthine Dehydrogenase/metabolism , Animals , Carcinogenesis/chemically induced , Carcinogenesis/immunology , Cell Line, Tumor , Colitis-Associated Neoplasms/genetics , Colitis-Associated Neoplasms/pathology , Colon/immunology , Colon/pathology , Disease Models, Animal , Disease Progression , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/immunology , Humans , Macrophages/metabolism , Male , Transcriptional Activation/immunology , Tumor Microenvironment/immunology , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation , Xanthine Dehydrogenase/genetics
13.
Front Immunol ; 12: 654463, 2021.
Article in English | MEDLINE | ID: mdl-34054817

ABSTRACT

Immune checkpoint inhibitors, such as monoclonal antibodies targeting programmed death 1 (PD-1) and programmed death ligand-1 (PD-L1), have achieved enormous success in the treatment of several cancers. However, monoclonal antibodies are expensive to produce, have poor tumor penetration, and may induce autoimmune side effects, all of which limit their application. Here, we demonstrate that PDI-1 (also name PD1/PD-L1 inhibitor 1), a small molecule antagonist of PD-1/PD-L1 interactions, shows potent anti-tumor activity in vitro and in vivo and acts by relieving PD-1/PD-L1-induced T cell exhaustion. We show that PDI-1 binds with high affinity to purified human and mouse PD-1 and PD-L1 proteins and is a competitive inhibitor of human PD-1/PD-L1 binding in vitro. Incubation of ex vivo activated human T cells with PDI-1 enhanced their cytotoxicity towards human lung cancer and melanoma cells, and concomitantly increased the production of granzyme B, perforin, and inflammatory cytokines. Luciferase reporter assays showed that PDI-1 directly increases TCR-mediated activation of NFAT in a PD-1/PD-L1-dependent manner. In two syngeneic mouse tumor models, the intraperitoneal administration of PDI-1 reduced the growth of tumors derived from human PD-L1-transfected mouse lung cancer and melanoma cells; increased and decreased the abundance of tumor-infiltrating CD8+ and FoxP3+ CD4+ T cells, respectively; decreased the abundance of PD-L1-expressing tumor cells, and increased the production of inflammatory cytokines. The anti-tumor effect of PDI-1 in vivo was comparable to that of the anti-PD-L1 antibody atezolizumab. These results suggest that the small molecule inhibitors of PD-1/PD-L1 may be effective as an alternative or complementary immune checkpoint inhibitor to monoclonal antibodies.


Subject(s)
B7-H1 Antigen/metabolism , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/metabolism , Immune Checkpoint Inhibitors/pharmacology , Melanoma/immunology , Melanoma/metabolism , Programmed Cell Death 1 Receptor/metabolism , Animals , B7-H1 Antigen/chemistry , Cell Line , Cytokines/metabolism , Cytotoxicity, Immunologic , Disease Models, Animal , Dose-Response Relationship, Drug , Humans , Immune Checkpoint Inhibitors/chemistry , Immune Checkpoint Inhibitors/therapeutic use , Lung Neoplasms/drug therapy , Lung Neoplasms/immunology , Lung Neoplasms/metabolism , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Melanoma/drug therapy , Melanoma, Experimental , Mice , Models, Biological , Molecular Structure , Programmed Cell Death 1 Receptor/chemistry , Protein Binding/drug effects , Signal Transduction , Structure-Activity Relationship , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Xenograft Model Antitumor Assays
14.
Sci Rep ; 11(1): 253, 2021 01 08.
Article in English | MEDLINE | ID: mdl-33420287

ABSTRACT

Posttransplant cyclophosphamide (PTCy) as graft-versus-host disease (GVHD) prophylaxis is an effective strategie for patients receiving matched sibling donor hematopoietic stem cell transplantation (MSD-HSCT) and haploidentical HSCT (haplo-HSCT). We evaluated the effectiveness and safety of reduced-dose cyclophosphamide, 20 mg/kg for 13 patients in MSD-HSCT cohort and 25 mg/kg for 22 patients in haplo-HSCT cohort, on days + 3, + 4 combined with cotransplantation of peripheral blood stem cells (PBSCs) and human umbilical cord-derived mesenchymal stem cells (UC-MSCs) for severe aplastic anemia (SAA). In MSD-PTCy cohort, the times to neutrophil and platelet engraftment were significantly shorter than those in the MSD-control cohort (P < 0.05). The cumulative incidence of acute GVHD (aGVHD) at day + 100 (15.4%) was lower than that in the MSD-control cohort (P = 0.050). No patient developed chronic GVHD (cGVHD). The 1-year overall survival (OS) and event-free survival (EFS) rates were 100% and 92.3%. In haplo-PTCy cohort, the times to neutrophil and platelet engraftment were significantly shorter than those in the haplo-control cohort (P < 0.05). The cumulative incidences of aGVHD at day + 100 and 1-year cGVHD were 31.8% and 18.2%, and the 1-year OS and EFS rates were 81.8% and 66.9%. Reduced-dose PTCy and cotransplantation of PBSCs and UC-MSCs is an acceptable alternative to patients with SAA.


Subject(s)
Anemia, Aplastic/therapy , Cyclophosphamide/administration & dosage , Graft vs Host Disease/prevention & control , Mesenchymal Stem Cell Transplantation/methods , Peripheral Blood Stem Cell Transplantation/methods , Transplantation Conditioning/methods , Adolescent , Adult , Anemia, Aplastic/mortality , Child , Cyclophosphamide/adverse effects , Feasibility Studies , Female , Graft vs Host Disease/mortality , Humans , Kaplan-Meier Estimate , Male , Mesenchymal Stem Cell Transplantation/adverse effects , Mesenchymal Stem Cell Transplantation/mortality , Off-Label Use , Peripheral Blood Stem Cell Transplantation/adverse effects , Peripheral Blood Stem Cell Transplantation/mortality , Siblings , Tissue Donors , Transplantation, Haploidentical/methods , Treatment Outcome , Umbilical Cord/cytology , Young Adult
15.
Nat Immunol ; 22(3): 358-369, 2021 03.
Article in English | MEDLINE | ID: mdl-33432230

ABSTRACT

CD8+ T cell exhaustion dampens antitumor immunity. Although several transcription factors have been identified that regulate T cell exhaustion, the molecular mechanisms by which CD8+ T cells are triggered to enter an exhausted state remain unclear. Here, we show that interleukin-2 (IL-2) acts as an environmental cue to induce CD8+ T cell exhaustion within tumor microenvironments. We find that a continuously high level of IL-2 leads to the persistent activation of STAT5 in CD8+ T cells, which in turn induces strong expression of tryptophan hydroxylase 1, thus catalyzing the conversion to tryptophan to 5-hydroxytryptophan (5-HTP). 5-HTP subsequently activates AhR nuclear translocation, causing a coordinated upregulation of inhibitory receptors and downregulation of cytokine and effector-molecule production, thereby rendering T cells dysfunctional in the tumor microenvironment. This molecular pathway is not only present in mouse tumor models but is also observed in people with cancer, identifying IL-2 as a novel inducer of T cell exhaustion.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , CD8-Positive T-Lymphocytes/drug effects , Interleukin-2/metabolism , Lymphocytes, Tumor-Infiltrating/drug effects , Neoplasms/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Tumor Microenvironment , 5-Hydroxytryptophan/metabolism , Animals , Antibodies, Neutralizing/pharmacology , Antineoplastic Agents/pharmacology , Basic Helix-Loop-Helix Transcription Factors/deficiency , Basic Helix-Loop-Helix Transcription Factors/genetics , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Gene Expression Regulation, Neoplastic , HCT116 Cells , HEK293 Cells , Humans , Interleukin-2/antagonists & inhibitors , Interleukin-2/genetics , Jurkat Cells , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , MCF-7 Cells , Melanoma, Experimental/drug therapy , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , NIH 3T3 Cells , Neoplasms/drug therapy , Neoplasms/immunology , Neoplasms/pathology , Receptors, Aryl Hydrocarbon/deficiency , Receptors, Aryl Hydrocarbon/genetics , Signal Transduction , Tryptophan Hydroxylase/metabolism , Xenograft Model Antitumor Assays
16.
Drug Deliv Transl Res ; 11(1): 192-204, 2021 02.
Article in English | MEDLINE | ID: mdl-32394334

ABSTRACT

At present, treating of triple negative breast cancer (TNBC) mainly depends on chemotherapy with more toxic side effects, but the effect is limited and it is highly prone to drug resistance. Gene therapy using anti-microRNAs maybe one of alternative therapeutic strategies. Due to the poor cell permeability and significant in vivo decomposition rate of anti-microRNAs, which limits their clinical application, we developed a core-shell supramolecular nanovector of "chitosome" that were self-assembled from the synthetic amphiphilic chitosan derivatives. The constructed chitosomes could co-load hydrophilic anti-miR-21 and hydrophobic docetaxel (DTX) into one combo nanocarrier with entrapment efficiency of more than 80%, as well as spherical morphology and average particle size of 90 nm. In comparison with the naked ones, anti-miR-21 encapsulated with chitosomes showed significantly increased cellular transfection and stability against degradation by nuclease in serum. Compared with DTX or anti-miR-21 formulations used alone, the co-delivery of the two drugs with the combo chitosome obtained improved chemosensitivity of TNBC cells to DTX treatment through their synergistic mechanisms. Taken together, the developed chitosome could be a promising candidate for simultaneous delivery of insoluble chemotherapeutic drugs and gene agents for TNBC therapy. Graphical abstract.


Subject(s)
Antineoplastic Agents , MicroRNAs , Nanoparticles , Triple Negative Breast Neoplasms , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Docetaxel , Drug Carriers/therapeutic use , Humans , MicroRNAs/therapeutic use , Particle Size , Triple Negative Breast Neoplasms/drug therapy
17.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-877399

ABSTRACT

@#目的:分析Claudin-2蛋白在食管鳞癌(esophageal squamous cell carcinomas,ESCC)组织中的表达及其与患者临床病理特征、5年生存率的关系,探索其对ESCC细胞KYSE450的增殖、迁移和侵袭的影响。方法:选取河南省肿瘤医院2010至2013年间初治的ESCC患者手术切除肿瘤组织52例及其中20例对应的癌旁组织标本,采用免疫组化和WB法检测Claudin-2的表达并分析其与患者临床病理特征和5年生存率的关系。WB法检测ESCC细胞(KYSE450、KYSE150、KYSE510、KYSE140)和人永生化食管上皮细胞SHEE中Claudin-2的表达,构建Claudin-2 shRNA慢病毒载体并转染KYSE450细胞构建敲低Claudin-2表达的细胞系,进一步通过克隆形成实验、细胞划痕实验及Transwell实验检测敲低Claudin-2对KYSE450细胞增殖、迁移和侵袭的影响。结果:ESCC组织中Claudin-2阳性率显著高于癌旁组织(P<0.05),ESCC组织中Claudin-2的表达与淋巴结转移有关(P<0.05)。Claudin-2表达阳性患者5年生存率显著低于阴性者(P<0.05)。成功构建敲低Claudin-2表达的KYSE450细胞系。sh-Claudin-2组细胞的克隆形成数、伤口愈合率和侵袭细胞数均显著低于sh-NT组和对照组(P<0.05)。结论:ESCC组织中Claudin-2的表达高于癌旁组织,且与患者5年生存率呈负相关,Claudin-2能够增强KYSE450细胞的增殖、迁移和侵袭能力。

18.
Oncogene ; 39(43): 6733-6746, 2020 10.
Article in English | MEDLINE | ID: mdl-32963350

ABSTRACT

Colon cancer is the most aggressive tumor in both men and women globally. As many the chemotherapeutic regimens have adverse side effects and contribute to the resistance and recurrence, therefore, finding novel therapeutic targets and developing effective agents are urgent. Based on the TCGA and GTEx database analysis, RSK1 and MSK2 were found abnormal expressed in colon cancer. RSK1 and MSK2 were overexpressed in colon cancer tissues confirmed by western blot and IHC. After knocking down RSK1 or MSK2, cell proliferation and anchorage-independent cell growth were markedly inhibited. Using a computer docking model, we identified a novel dual-target inhibitor, APIO-EE-07, that could block both RSK1 and MSK2 kinase activity in a dose-dependent manner. APIO-EE-07 inhibited cell growth and induced apoptosis and also increased expression of Bax as well as cleaved caspase-3 and -PARP in colon cancer cells by downregulating RSK1 and MSK2 downstream targets, including CREB and ATF1. Furthermore, APIO-EE-07 decreased tumor volume and weight in human patient-derived xenografts tumors implanted in SCID mice. In summary, our results demonstrate that RSK1 and MSK2 are the potential targets for the treatment of colon cancer. APIO-EE-07, a novel dual-target inhibitor of RSK1 and MSK2, can suppress the growth of colon cancer by attenuating RSK1 and MSK2 signaling.


Subject(s)
Antineoplastic Agents/pharmacology , Colonic Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Ribosomal Protein S6 Kinases, 90-kDa/antagonists & inhibitors , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Apoptosis , Cell Line, Tumor , Colonic Neoplasms/pathology , Crystallography, X-Ray , Drug Discovery , Female , HEK293 Cells , Humans , Mice , Molecular Docking Simulation , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/therapeutic use , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/ultrastructure , Xenograft Model Antitumor Assays
19.
Int Immunopharmacol ; 87: 106798, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32693357

ABSTRACT

BACKGROUND: Gastric cancer has extremely high morbidity and mortality. Currently, it is lack of effective biomarkers and therapeutic targets for guiding clinical treatment. In this study, we aimed to identify novel biomarkers and therapeutic targets for gastric cancer. METHODS: Differentially expressed genes (DEGs) between gastric cancer and normal tissues were obtained from Gene Expression Omnibus (GEO). Core genes were identified by constructing protein-protein interaction network of DEGs. The expression of core genes was verified in Gene Expression Profiling Interactive Analysis (GEPIA), UALCAN and clinical samples. Further, the mutation, DNA methylation, prognostic value, and immune infiltration of core genes were validated by cBioPortal, MethSurv, Kaplan-Meier plotter, and Tumor Immune Estimation Resource (TIMER) databases. Additionally, drug response analysis was performed by Cancer Therapy Response Portal (CTRP). RESULTS: A total of seven collagen family members were identified as core genes among upregulated genes. And copy number amplification may be involved in the upregulation of COL1A1 and COL1A2. Importantly, the collagen family was associated with the poor prognosis of patients with metastasis. Among them, COL1A1 had a higher hazard ratio (HR) for overall survival than other members (HR = 2.33). The correlation between DNA methylation levels at CpG sites of collagen family members and the prognosis was verified in gastric cancer. Besides, collagen family expression was positively correlated with macrophages infiltration and the expression of M2 macrophages markers. Further, collagen expression was related to the sensitivity and resistance of gastric cancer cell lines to certain drugs. CONCLUSIONS: The collagen family, especially COL1A1, COL1A2, and COL12A1, may act as potential prognostic biomarkers and immune-associated therapeutic targets in gastric cancer.


Subject(s)
Biomarkers, Tumor/metabolism , Collagen Type I/metabolism , Collagen Type XII/metabolism , Macrophages/immunology , Stomach Neoplasms/diagnosis , Biomarkers, Tumor/genetics , Cell Line, Tumor , Collagen Type I/genetics , Collagen Type I, alpha 1 Chain , Collagen Type XII/genetics , Cytokines/metabolism , DNA Methylation , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Microarray Analysis , Neoplasm Staging , Prognosis , Stomach Neoplasms/immunology , Stomach Neoplasms/mortality , Survival Analysis , Th2 Cells/immunology
20.
Theranostics ; 10(14): 6201-6215, 2020.
Article in English | MEDLINE | ID: mdl-32483448

ABSTRACT

Background: Gastric cancer remains the second leading cause of cancer-related death, and the third in mortality due to lack of effective therapeutic targets for late stage cancer patients. This study aims to identify potential druggable target biomarkers as potential therapeutic options for patients with gastric cancer. Methods: Immunohistochemistry of human gastric tumor tissues was conducted to determine the expression level of cyclin-dependent kinase 12 (CDK12). Multiple in vitro and in vivo assays such as RNAi, mass spectrometry, computer docking models, kinase assays, cell xenograft NU/NU mouse models (CDXs) and patient-derived xenograft NOD/SCID mouse models (PDXs) were conducted to study the function and molecular interaction of CDK12 with p21 activated kinase 2 (PAK2), as well as to find CDK12 inhibitors as potential treatment options for human gastric cancer. Results: Here we identified that CDK12 is a driver gene in human gastric cancer growth. Mechanistically, CDK12 directly binds to and phosphorylates PAK2 at T134/T169 to activate MAPK signaling pathway. We further identified FDA approved clinical drug procaterol can serve as an effective CDK12 inhibitor, leading to dramatic restriction of cancer cell proliferation and tumor growth in human gastric cancer cells and PDXs. Conclusions: Our data highlight the potential of CDK12/PAK2 as therapeutic targets for patients with gastric cancer, and we propose procaterol treatment as a novel therapeutic strategy for human gastric cancer.


Subject(s)
Cyclin-Dependent Kinases/antagonists & inhibitors , Procaterol/pharmacology , Stomach Neoplasms/drug therapy , p21-Activated Kinases/antagonists & inhibitors , Adult , Aged , Aged, 80 and over , Animals , Bronchodilator Agents/pharmacology , Cell Line, Tumor , Cyclin-Dependent Kinases/metabolism , Female , Humans , Male , Mice , Mice, Nude , Middle Aged , Phosphorylation , Signal Transduction , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Xenograft Model Antitumor Assays , p21-Activated Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...