One of the most important treatment modalities for non-small cell lung cancer (NSCLC) is immune checkpoint inhibitor. Nevertheless, a small percentage of patients do not respond well to these therapies, highlighting the significance of identifying important CD8+ T cell subsets for immunotherapy and creating trustworthy biomarkers. The purpose of this study is to assess the potential utility of TIM3+CD8+ T cells as new biomarkers by examining their expressions in various areas of the NSCLC tumor microenvironment. Based on biopsy techniques, tumor tissue samples were obtained from patients with NSCLC and categorized into tumor central and non-central regions. Using flow cytometry, the infiltration of TIM3+CD8+ T cells and the surface expression of programmed cell death 1 (PD-1) on these cells were examined, and their correlations with the effectiveness of immunotherapy were assessed. The non-central region of tumor tissues had considerably larger infiltration of TIM3+CD8+ T lymphocytes compared to the non-central region (P<0.0001). This pattern was found in both subgroups with tumor diameters ≥3 cm or <3 cm (P<0.01). In comparison to TIM3-CD8+ T cells, TIM3+CD8+ T cells showed higher levels of PD-1 (P<0.001), with more PD-1+TIM3+CD8+ T cells invading the non-central region (P<0.01). Clinical responders to immunotherapy had considerably lower infiltration levels of TIM3+CD8+ T cells in the tumor non-central region compared to non-responders, with lower levels correlated with better clinical outcomes (P<0.01), while no correlation was identified in the tumor central region (P>0.05). According to reciever operating characteristic (ROC) curve analysis, TIM3+CD8+ T cells in the tumor non-central region had an area under the curve (AUC) of 0.9375 for predicting the effectiveness of immunotherapy, which was considerably higher than that of TIM3+CD8+ T cells in the tumor central region and programmed cell death ligand 1 (PD-L1) [tumor proportion score (TPS)]. In the tumor microenvironment of NSCLC, TIM3+CD8+ T cells show regional distribution patterns. The expression of this cell population in the non-central region of the tumor microenvironment may be a biomarker for predicting the effectiveness of immunotherapy.
Tumor-associated macrophages (TAMs) have been characterized as a critical population of immunosuppressive cells in a variety of tumor types. PD-L1 (also termed B7-H1) has been described to exert co-inhibitory and immune regulatory functions. Here, in ovarian cancer, PD-L1 is selectively overexpressed on some TAM compared that of benign ovarian disease. When expanding the data in peripheral blood, the proportion of PD-L1+CD68+ cell among CD68+ cells and the intensity of PD-L1 staining on CD68+ cell in healthy group were similar to that observed in ovarian cyst group; instead, these two measures were significantly higher in ovarian cancer group, thereafter related to TNM stage. Interestingly, intracellular levels of IL-10, IL-6, TNF-α, and IFN-γ in PD-L1+CD68+ macrophage were higher than those in PD-L1−CD68+ macrophage, especially IL-6 expression. Based on the PD-L1 receptor PD-1 expression on tumor-infiltrating cytotoxic cells, our data supported that expression of PD-L1 on TAM promoted apoptosis of T cells via interaction with PD-1 on CD8+T cells. Taken together, these results suggested that PD-L1-expressing macrophage represents a novel suppressor cell population in ovarian cancer, which contributes immune escape of ovarian cancer.
AIM: To investigate the stable expression of a chimeric antibody against CD40 molecule(ch-5C11)in CHO and its biological activity. METHODS: Human-mouse chimeric antibody against CD40 recombinant plasmid and mock plasmid were transfected into CHO cell line through lipofectamine mediation. Human κ chain and Fc fragment of ch-5C11 were characterized by FACS and Western blot. The concentration of ch-5C11 in cell supernatants was detected by Lowry assay and the inhibitory effect of ch-5C11 on the proliferation of Daudi cells was detected by MTT assay. RESULTS: RT-PCR showed that target CHO cells integrated chimeric heavy chain and chimeric light chain gene. FACS and Western blot showed that ch-5C11 in cell supernatants maintained the binding activity and specificity to human CD40 molecule, and contained human κ chain and Fc fragment. Cell supernatants were purified using protein G affinity chromatography. The concentration of human-mouse chimeric antibody against CD40 in cell supernatants was 0.535 mg/L. When co-cultured with B lymphoma cell line Daudi, ch-5C11 induced proliferation arrest of Daudi cells. CONCLUSION: The human-mouse chimeric antibody against CD40 can be expressed in CHO stably and effectively, which inhibits proliferation of Daudi.
While studies have demonstrated that certain COVID-19 vaccines administered during pregnancy did not affect neonatal or maternal outcomes significantly, the safety of inactivated SARS-CoV-2 vaccines in China, given during the first trimester, remains to be fully elucidated.A retrospective cohort study was conducted involving female participants who gave birth from January to October 2021. The study compared pregnancy, delivery, and neonatal outcomes between subjects who received one or two doses of the inactivated COVID-19 vaccines during their first trimester and unvaccinated control subjects.A total of 2658 pregnant women was recruited. Among them, 2358 (88.7%) reported ongoing pregnancies; 326 (13.8%) of these were vaccinated. Additionally, 277 (10.4%) experienced spontaneous miscarriages between 6 to 20 gestational weeks; 40 (14.4%) of these were vaccinated, yielding an odds ratio of 0.67-1.36 (95% confidence interval) for COVID-19 vaccination. The comparison of neonatal complications, including an Apgar score less than 7, preterm birth, low birth weight, and newborn respiratory complications, between unvaccinated and vaccinated participants revealed no statistical significance.The administration of COVID-19 inactivated vaccines during the first trimester of pregnancy is not associated with adverse pregnancy or neonatal outcomes, providing a substantial ground for pertinent health education.
To develop a magnetic nanoparticles based magnetic resonance (MR) probe targeting CD40 mutant in the imaging of breast cancer cells in vitro.For preparing an immunologically competent probe, monoclonal antibody was conjugated with ultrasmall superparamagnetic iron oxide (USPIO) particles basing on chemical cross-linking method.Its bioactivity was analyzed with flow cytometry, confocal microscopy and Prussian blue staining. The probe's cell MR imaging in vitro was conducted on breast cancer cells (M231) high expressing CD40 mutant. The signal data from different groups were collected and analyzed with one-way variance and least significant difference-t test.The molecular probe carrying nanoparticles and CD40 mutant antibody was constructed and separated successfully. The probe had similar magnetic property compared with original USPIO particles.It could recognize CD40 mutant on breast cancer cells (M231) with high specificity. MR cell imaging in vitro shows that T2 and T2(*) obviously shortened after probe binding with M231 cells and T2 weighted imaging become darker than control groups, the time of T2 is 5H6-USPIO (51.66 ± 5.31) , 5C11-USPIO (92.89 ± 4.72), USPIO (64.56 ± 3.85) ms. The T2 and T2(*) relaxation time of experiment group was shorter than control groups with statistical significance (P < 0.01) .MR molecular probe targeting CD40 mutant may bind with breast cancer cells (M231) to provide further in vivo animal MR imaging. And CD40 mutant is expected to provide a new target for MR molecular imaging of breast cancer.
DCs infiltrated tumors appears to be phenotypically and functionally defective. B7-H4 was highlighted for its inhibitory role in T cell responses. In this study, we showed that B7-H4 was moderately expressed in imDCs, and up-regulated by IL-10, and TNF-α could counteract the up-regulatory effects of IL-10 on expression of B7-H4 in DCs in vitro. Furthermore, tumor infiltrated DCs expressed B7-H4 at high levels. Blockade of B7-H4 expressed in DCs highly resulted in enhanced T cell proliferation and IFN-γ production significantly. Otherwise, the high level of IL-10 and TNF-α was both detected in the tumor, which suggested that TNF-α can not antagonize the effects of IL-10 on expression of B7-H4 in DCs in vivo. These data indicate that tumor environment may condition local DCs to become dysfunctional in the phenotype, and that the high expression of B7-H4 may contribute to the tumor infiltrated DCs to mediate immune invasion.
Hospital-acquired pneumonia (HAP) is one of the most common diseases in the intensive care unit, where the development of disease is closely related with the host immune response. Monocytes play an important role in both innate and adaptive immune system. We aimed to investigate the changes of circulating monocyte subsets in subjects with HAP to explore its value in monitoring HAP.In total, 60 HAP patients and 18 healthy individuals were enrolled in this study. Human monocyte subsets are classified into 3 groups: nonclassical (NC), intermediate (ITM), and classical (CL). Also, programmed death ligand 1 (PD-L1) expression on circulating monocyte subsets was measured by flow cytometry.Data showed that the ratio of NC, ITM, and CL among monocytes was comparable between HAP patients and healthy controls (P > .05). There was a remarkable imbalance of NC and CL in newly emerged HAP compared to healthy controls (P < .05), subsequently reaching normalization in recurrent HAP (P > .05). Furthermore, although PD-L1 was seemly constitutively expressed by NC, ITM, and CL groups regardless of disease status, it was noted that PD-L1 was dominantly expressed in the CL group (P < .05).Given distinct PD-L1 expression, a shift of CL/NC in newly emerged HAP would constitute an inhibitory anti-pathogen immune response. Normalization of circulating monocyte subsets on recurrence of HAP might be the consequence of immune memory of bacterial infection.