Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Int Wound J ; 21(2): e14756, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38339818

ABSTRACT

The primary objective of this meta-analysis was to provide the comprehensive understanding of the intricate correlation that existed between immune senescence and its effects on the advancement of lung cancer as well as recovery of cutaneous wounds. By conducting this systematic review of six rigorous studies utilizing databases such as PubMed and Web of Science, this research examined the multitude of facets pertaining to immune aging and consequences it bear on the health outcomes. The incorporated studies encompassed wide range of geographical and methodological viewpoints, with the specific emphasis on non-small-cell lung cancer and diverse scenarios related to wound recovery. This analysis synthesized discoveries regarding therapeutic responses, cellular and molecular mechanisms and impact of lifestyle factors on immune senescence. The findings suggested that immune senescence has substantial impact on the effectiveness of treatments for lung cancer and cutaneous wounds healing process; therefore, targeted therapies and holistic approaches may be able to mitigate these effects. By following the revised PRISMA guidelines, this meta-analysis guarantee thorough and ethically sound methodology for amalgamating pre-existing literature. The study concluded by emphasizing the critical nature of comprehending immune senescence in the context of clinical practice and proposed avenues for further investigation to enhance health results among the elderly.


Subject(s)
Lung Neoplasms , Wound Healing , Humans , Lung Neoplasms/immunology , Aged , Male , Female , Disease Progression , Carcinoma, Non-Small-Cell Lung/immunology , Middle Aged , Immunosenescence , Aged, 80 and over , Adult
2.
Int Immunopharmacol ; 104: 108468, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35066343

ABSTRACT

BACKGROUND: Previous studies have substantiated that M2-activated tumor-associated macrophages (M2-TAMs) are involved in multiple malignancies. Presently, we probe the impact and related mechanisms of 13-methyl-palmatrubine (13MP), the Corydalis yanhusuo extract, on M2-TAM-mediated non-small cell lung cancer (NSCLC) development. METHODS: IL-4 and IL-13 were adopted to induce M2-TAMs. The polarization state of TAMs was evaluated by quantitative reverse transcription PCR (qRT-PCR), Western blot (WB) and cellular immunofluorescence. NSCLC cells (A549 and NCL-H1975) were co-cultured with the conditioned medium (CM) of M2-TAMs. Followed by 13MP treatment, cell viability, proliferation, invasion, epithelial-mesenchymal transition (EMT), and in-vivo growth of NSCLC cells were determined. Additionally, human umbilical vein endothelial cells (HUVECs) were co-cultured with the CM of M2-TAMs. The tube formation assay was made to test the tube formation capacity of HUVECs, and the expression of MMP3, MMP9, and VEGF was assessed by WB in the co-culture model. Mechanistically, WB was performed to validate the expression of the PI3K/AKT and JAK/STAT3 pathways in NSCLC cells (A549 and NCL-H1975) as well as in endothelial cell lines co-cultured with M2-TAMs. RESULTS: 13MP inhibited the proliferation, invasion, EMT, growth and enhanced apoptosis of NSCLC cells. 13MP dose-dependently boosted the polarization of TAM from M2 to M1 state. M2-TAMs enhanced the malignant behaviors of NSCLC cells, whereas 13MP hindered M2-TAM-mediated NSCLC cell proliferation and invasion. Meanwhile, 13MP weakened the M2-TAM-mediated angiogenesis. Moreover, 13MP inactivated the PI3K/AKT and JAK/STAT3 signaling in A549 cells, NCL-H1975 cells and HUVECs. CONCLUSION: 13MP suppresses TAM-mediated NSCLC progression via transforming the polarization of TAM from M2 to M1.


Subject(s)
Antineoplastic Agents/therapeutic use , Berberine Alkaloids/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacology , Berberine Alkaloids/pharmacology , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line , Cell Movement/drug effects , Cell Proliferation/drug effects , Coculture Techniques , Cytokines/genetics , Epithelial-Mesenchymal Transition/drug effects , Humans , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Lung Neoplasms/metabolism , Mice, Inbred BALB C , Mice, Nude , Tumor-Associated Macrophages/drug effects , Tumor-Associated Macrophages/immunology
3.
Biomed Pharmacother ; 143: 112105, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34560533

ABSTRACT

Although the main focus of immuno-oncology has been manipulating the adaptive immune system, tumor associated macrophages (TAMs) are the main infiltrating component in the tumor microenvironment (TME) and play a critical role in cancer progression. TAMs are mainly divided into two different subtypes: macrophages with antitumor or killing activity are called M1 while tumor-promoting or healing macrophages are named M2. Therefore, controlling the polarization of TAMs is an important strategy for cancer treatment, but there is no particularly effective means to regulate the polarization process. Here, combined systems pharmacology targets and pathways analysis strategy, we uncovered Scutellariae Radix (SR) has the potential to regulate TAMs polarization to inhibit the growth of non-small cell lung cancer (NSCLC). Firstly, systems pharmacology approach was used to reveal the active components of SR targeting macrophages in TME through compound target prediction and target-microenvironment phenotypic association analysis. Secondly, in vitro experiment verified that WBB (wogonin, baicalein and baicalin), major active ingredients of SR are significantly related to macrophages and survival, initiated macrophages programming to M1-like macrophages to promoted the apoptosis of tumor cells. Finally, we evidenced that WBB effectively inhibited tumor growth in LLC (Lewis lung carcinoma) tumor-bearing mice and increased the infiltration of M1-type macrophages in TME. Overall, the systems pharmacology strategy offers a paradigm to understand the mechanism of polypharmacology of natural products targeting TME.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Carcinoma, Lewis Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/drug therapy , Immunity, Innate/drug effects , Lung Neoplasms/drug therapy , Network Pharmacology , Tumor-Associated Macrophages/drug effects , Animals , Apoptosis/drug effects , Carcinoma, Lewis Lung/immunology , Carcinoma, Lewis Lung/metabolism , Carcinoma, Lewis Lung/pathology , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Coculture Techniques , Databases, Genetic , Female , Humans , Lung Neoplasms/immunology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Mice, Inbred C57BL , Phenotype , RAW 264.7 Cells , Tumor Burden/drug effects , Tumor Microenvironment , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism
4.
J Ethnopharmacol ; 281: 114514, 2021 Dec 05.
Article in English | MEDLINE | ID: mdl-34384846

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Taraxacum mongolicum Hand.-Mazz. has been used in lung cancer treatment in Chinese medicine. However, its specific mechanism of action has not yet been reported, and developing pharmaceutical anti-cancer resources is important. Here, we aimed to elucidate the anti-tumor effects of dandelion in vitro and in vivo and assess its effects on immune function in lung cancer patients. AIM OF THE STUDY: In the present study, we mainly observed the therapeutic effects of total flavonoids from Taraxacum mongolicum Hand.-Mazz. (TFTM) on non-small cell lung cancer and its influence on the body's immune function. MATERIALS AND METHODS: In vitro experiments on A549 and H1299 cells were performed using the CCK8 method; the proliferation and migration of cells were observed to investigate the wound healing effects of TFTM, and flow cytometry was used to detect the apoptotic rate of TFTM on lung cancer cells. In vivo experiments were preformed to establish a non-small cell lung cancer mouse model using subcutaneously transplanted Lewis cells, and the body weight and tumor growth of the mice were recorded. Hematoxylin and eosin staining was performed for tumor tissue to assess pathological changes. The thymus, spleen, and lungs were isolated for to calculate organ index. The CD4+, CD8+, and CD4+/CD8+ levels were detected in mouse spleen using flow cytometry, and IL-2, IL-3, IFN-γ, and TNF-α levels were determined in serum using enzyme-linked immunosorbent assay. Expressions of IL-2, IL-3, IFN-γ, and TNF-α were detected using quantitative real-time PCR in tumor tissues, and Ki67 expression was observed by immunofluorescence. RESULTS: At 24 h, TFTM (100 and 200 µg/mL) had the best inhibitory effect on the proliferation of A549 and H1299 cells. The cell migration rate significantly reduced (P < 0.01), and the tumor inhibition rate increased (P < 0.01) and promoted apoptosis (P < 0.01). The mouse thymus index significantly increased (P < 0.05) and mouse spleen index reduced (P < 0.05). The CD4+, CD8+, and CD4+/CD8+ levels in Lewis lung cancer mouse model increased, as did the levels of IL-2, IL-3, IFN-γ, and TNF-α in the serum and tumor of mice; Ki67 expression in tumor tissues significantly reduced (P < 0.01). CONCLUSION: TFTM has an inhibitory effect on lung cancer. The mechanism may be that it improves the host's protective immune response by having a milder tumor growth inhibitory effect than cyclophosphamide.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Flavonoids/therapeutic use , Lung Neoplasms/drug therapy , Taraxacum , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cytokines/genetics , Cytokines/immunology , Flavonoids/pharmacology , Humans , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Male , Mice, Inbred C57BL , Spleen/drug effects , Thymus Gland/drug effects , Tumor Burden/drug effects
5.
Cancer Immunol Immunother ; 70(12): 3435-3449, 2021 Dec.
Article in English | MEDLINE | ID: mdl-33877384

ABSTRACT

Specific extracts of selected vegetables (SV) have been shown to benefit the survival of stage IIIb/IV non-small cell lung cancer patients in phase I/II studies and is currently in a phase III trial. However, the underlying mechanism of SV-mediated antitumor immune responses has not been elucidated. Our results indicate that SV modulated the NK and adoptive T cell immune responses in antitumor efficacy. Furthermore, antitumor effects of SV were also mediated by innate myeloid cell function, which requires both TLR and ß-glucan signaling in a MyD88/TRIF and Dectin-1-dependent manner, respectively. Additionally, SV treatment reduced granulocytic myeloid-derived suppressor cell (MDSC) infiltration into the tumor and limited monocytic MDSC toward the M2-like functional phenotype. Importantly, SV treatment enhanced antigen-specific immune responses by augmenting the activation of antigen-specific TH1/TH17 cells in secondary lymphoid organs and proliferative response, as well as by reducing the Treg population in the tumor microenvironment, which was driven by SV-primed activated M-MDSC. Our results support the idea that SV can subvert immune-tolerance state in the tumor microenvironment and inhibit tumor growth. The present study suggests that features, such as easy accessibility, favorable clinical efficacy, no detectable side effects and satisfactory safety make SV a feasible, appealing and convincing adjuvant therapy for the treatment of cancer patients and prevent tumor recurrence and/or metastases.


Subject(s)
Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/therapy , Lung Neoplasms/immunology , Lung Neoplasms/therapy , Nutrients/immunology , Plant Extracts/immunology , Tumor Microenvironment/immunology , Animals , Dietary Supplements , Disease Models, Animal , Immune Tolerance/immunology , Immunity/immunology , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Mice, Transgenic , Monocytes/immunology , Myeloid Cells/immunology , Myeloid-Derived Suppressor Cells/immunology , Neoplasm Recurrence, Local/immunology , Th1 Cells/immunology , Th17 Cells/immunology
6.
Aging (Albany NY) ; 13(2): 2912-2940, 2021 01 17.
Article in English | MEDLINE | ID: mdl-33460401

ABSTRACT

The clinical notably success of immunotherapy fosters an enthusiasm in developing drugs by enhancing antitumor immunity in the tumor microenvironment (TME). Epimedium, is a promising herbal medicine for tumor immunotherapy due to the pharmacological actions in immunological function modulation and antitumor. Here, we developed a novel systems pharmacology strategy to explore the polypharmacology mechanism of Epimedium involving in targeting TME of non-small cell lung cancer (NSCLC). This strategy integrates the active compounds screening, target predicting, network pharmacology analysis and onco-immune interacting to predict the potential active compounds that trigger the antitumor immunity. Icaritin (ICT), a major active ingredient of Epimedium, was predicted to have good drug-like properties and target immune microenvironment in NSCLC via regulating multiple targets and pathways. Then, we evidenced that the ICT effectively inhibited tumor growth in LLC tumor-bearing mice and increases the infiltration of CD8+ T cells in TME. In addition, we demonstrated that ICT promotes infiltration of CD8+ T cells in TME by downregulating the immunosuppressive cytokine (TNF-α, IL10, IL6) and upregulating chemotaxis (CXCL9 and CXCL10). Overall, the systems pharmacology strategy offers an important paradigm to understand the mechanism of polypharmacology of natural products targeting TME.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Epimedium , Lung Neoplasms/drug therapy , Plant Extracts/pharmacology , T-Lymphocytes, Cytotoxic/drug effects , Tumor Microenvironment/drug effects , Animals , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cytokines/metabolism , Humans , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Mice , Plant Extracts/therapeutic use , T-Lymphocytes, Cytotoxic/immunology , Tumor Microenvironment/immunology
7.
J Cell Mol Med ; 25(6): 2900-2908, 2021 03.
Article in English | MEDLINE | ID: mdl-33506637

ABSTRACT

Lung cancer is the leading cause of cancer-related death worldwide, and non-small cell lung cancer (NSCLC) accounts for 85% of lung cancer diagnoses. As an ancient therapy, moxibustion has been used to treat cancer-related symptoms in clinical practice. However, its antitumour effect on NSCLC remains largely unexplored. In the present study, a Lewis lung cancer (LLC) xenograft tumour model was established, and grain-sized moxibustion (gMoxi) was performed at the acupoint of Zusanli (ST36). Flow cytometry and RNA sequencing (RNA-Seq) were used to access the immune cell phenotype, cytotoxicity and gene expression. PK136, propranolol and epinephrine were used for natural killer (NK) cell depletion, ß-adrenoceptor blockade and activation, respectively. Results showed that gMoxi significantly inhibited LLC tumour growth. Moreover, gMoxi significantly increased the proportion, infiltration and activation of NK cells, whereas it did not affect CD4+ and CD8+ T cells. NK cell depletion reversed gMoxi-mediated tumour regression. LLC tumour RNA-Seq indicated that these effects might be related to the inhibition of adrenergic signalling. Surely, ß-blocker propranolol clearly inhibited LLC tumour growth and promoted NK cells, and gMoxi no longer increased tumour regression and promoted NK cells after propranolol treatment. Epinephrine could inhibit NK cell activity, and gMoxi significantly inhibited tumour growth and promoted NK cells after epinephrine treatment. These results demonstrated that gMoxi could promote NK cell antitumour immunity by inhibiting adrenergic signalling, suggesting that gMoxi could be used as a promising therapeutic regimen for the treatment of NSCLC, and it had a great potential in NK cell-based cancer immunotherapy.


Subject(s)
Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/metabolism , Immunomodulation , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Lung Neoplasms/immunology , Lung Neoplasms/metabolism , Moxibustion , Signal Transduction , Animals , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/therapy , Cytotoxicity, Immunologic , Disease Models, Animal , Humans , Immunophenotyping , Lung Neoplasms/pathology , Lung Neoplasms/therapy , Lymphocyte Activation , Male , Mice , Moxibustion/methods , Receptors, Adrenergic/metabolism , Xenograft Model Antitumor Assays
8.
J Cancer Res Ther ; 16(4): 745-751, 2020.
Article in English | MEDLINE | ID: mdl-32930113

ABSTRACT

OBJECTIVE: The objective of the study was to evaluate the clinical efficacy of kanglaite (KLT) injection combined with gefitinib versus gefitinib alone in the treatment of nonsmall cell lung cancer (NSCLC). METHODS: The randomized controlled trials involving NSCLC treatment with KLT injection combined with gefitinib versus gefitinib alone were searched on seven medical databases up to October 2016. Two reviewers independently assessed the methodological quality of the included studies. The RevMan 5.3 software was employed for data analysis. RESULTS: Seven randomized trials involving 554 patients met our criteria. Compared with gefitinib alone, KLT injection combined with gefitinib showed significant effects in increasing objective response rate (relative risk [RR] =1.38; 95% confidence interval [CI], 1.09-1.75), improving the performance status (RR = 1.80; 95% CI: 1.34-2.42), raising the percentages of CD4+ cells (weighted mean difference [WMD] = 4.45; 95% CI: 2.61-6.28), natural killer cells (WMD = 4.43; 95% CI: 3.85-5.01), and ratio of CD4+/CD8+ (WMD = 0.08; 95% CI: 0.02-0.14), whereas the difference was not significant in gefitinib toxicity including rash (RR 0.90; 95% CI: 0.58-1.40, P = 0.65), diarrhea (RR 1.04; 95% CI: 0.66-1.64, P = 0.88), and liver injury (RR 1.00; 95% CI: 0.58-1.73, P = 1.00), CD3+ cells (WMD = 1.16; 95% CI: -2.64-4.97) and CD8+ cells (WMD = 6.78; 95% CI: -1.68-15.23). CONCLUSION: Co-use of KLT injection and gefitinib may benefit the patients with NSCLC through enhancing the therapeutic effectiveness compared with gefitinib alone. To confirm these results, further rigorously designed trials are warranted.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Antineoplastic Combined Chemotherapy Protocols/adverse effects , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/pathology , Drugs, Chinese Herbal/administration & dosage , Drugs, Chinese Herbal/adverse effects , Gefitinib/administration & dosage , Gefitinib/adverse effects , Humans , Killer Cells, Natural/immunology , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/pharmacology , Randomized Controlled Trials as Topic
9.
Pharmacol Res ; 160: 105086, 2020 10.
Article in English | MEDLINE | ID: mdl-32687951

ABSTRACT

Traditional Chinese medicine (TCM) plays a vital part in cancer treatment due to its unique superiority. Huoxue Yiqi Recipe-2 (HYR-2) was supposed to have therapeutic effect on lung cancer, which came from Ze Qi Decoction in one of the four great classics of TCM called "Synopsis of Prescriptions of the Golden Chamber". Network pharmacology demonstrated that the targets of active components from HYR-2 were significantly enriched in the signaling pathways, which were closely associated with non-small cell lung cancer (NSCLC) and programmed death ligand 1 (PD-L1). Then, data about NSCLC was downloaded from Gene Expression Omnibus database (GEO). The Cancer Genome Atlas (TCGA) and DisGeNET was analyzed by bioinformatics, and 214 biomarkers for NSCLC were obtained, containing 14 targets of active components from HYR-2 (which were significantly enriched in the PD-L1 related signaling pathway). In vivo and in vitro experiments showed that HYR and HYR-2 could inhibit the growth of lung cancer and down-regulate the expression of PD-L1, which might be related to the blocking effect of HYR-2 on the PI3K/Akt signaling pathway. Furthermore, HYR-2 promoted the transformation of M2 macrophages into M1 macrophages as well. It is deserved to be mentioned that the level of Akkermansia muciniphila was also significantly elevated by HYR-2, which was believed to enhance the therapeutic effect of PD-L1 antibodies. To sum up, HYR-2 might play an anti-lung cancer effect by down-regulating PD-L1 together with up-regulating Akkermansia muciniphila.


Subject(s)
B7-H1 Antigen/antagonists & inhibitors , Carcinoma, Non-Small-Cell Lung/drug therapy , Drugs, Chinese Herbal/pharmacology , Immune Checkpoint Inhibitors/pharmacology , Lung Neoplasms/drug therapy , Medicine, Chinese Traditional , A549 Cells , Akkermansia/drug effects , Akkermansia/growth & development , Animals , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , Carcinoma, Lewis Lung/drug therapy , Carcinoma, Lewis Lung/genetics , Carcinoma, Lewis Lung/immunology , Carcinoma, Lewis Lung/metabolism , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Proliferation/drug effects , Gene Regulatory Networks , Hep G2 Cells , Humans , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Lung Neoplasms/metabolism , MCF-7 Cells , Macrophages/drug effects , Macrophages/metabolism , Male , Mice, Inbred C57BL , Phenotype , Protein Interaction Maps , Signal Transduction , Tumor Burden/drug effects
10.
Expert Rev Anticancer Ther ; 20(3): 221-228, 2020 03.
Article in English | MEDLINE | ID: mdl-32141356

ABSTRACT

Introduction: The recent approvals of checkpoint inhibitors as single agents or in combination with chemotherapy with programmed death ligand 1 expression of < or ≥1% have challenged clinicians when it is time to begin a metastatic lung cancer patient in second-line therapy. The advantages given by immunotherapy over conventional chemotherapy such as improved overall survival and a better toxicity profile make the second-line clinical scenario more difficult for a patient who faces a likely inferior regimen as well as toxicity which may significantly impact the quality of life.Areas covered: Options given today by the National Comprehensive Cancer Network are very limited, and essentially, we go back to conventional cytotoxic agents alone or in combination with biological agents if possible. In this article, we discuss the actual treatment available for this difficult scenario and some of the ongoing trials which aim to address this dilemma.Expert commentary: This is an unmet need in lung cancer management; we need a better understanding of the mechanism of resistance to immunotherapy so we can target them once the patient moves to second-line treatment.


Subject(s)
Carcinoma, Non-Small-Cell Lung/therapy , Immunotherapy/methods , Lung Neoplasms/therapy , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/pathology , Humans , Immune Checkpoint Inhibitors/administration & dosage , Immune Checkpoint Inhibitors/adverse effects , Immunotherapy/adverse effects , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Quality of Life , Survival Rate
11.
J Ethnopharmacol ; 247: 112283, 2020 Jan 30.
Article in English | MEDLINE | ID: mdl-31605736

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Bulbus Fritillaria cirrhosa D. Don (BFC) is a Chinese traditional herbal medicine that has long been used as an indispensable component in herbal prescriptions for bronchopulmonary diseases due to its well-established strong anti-inflammation and pulmonary harmonizing effects. Interestingly, there are few case reports in traditional Chinese medicine available where they found it to contribute in anti-tumor therapies. Imperialine is one of the most favored active substances extracted from BFC and has been widely recognized as an anti-inflammatory agent. AIM OF THE STUDY: The aim of the current work is to provide first-hand evidences both in vitro and in vivo showing that imperialine exerts anti-cancer effects against non-small cell lung cancer (NSCLC), and to explore the molecular mechanism of this anti-tumor activity. It is also necessary to examine its systemic toxicity, and to investigate how to develop strategies for feasible clinical translation of imperialine. MATERIALS AND METHODS: To investigate anti-NSCLC efficacy of imperialine using both in vitro and in vivo methods where A549 cell line were chosen as in vitro model NSCLC cells and A549 tumor-bearing mouse model was constructed for in vivo study. The detailed underlying anti-cancer mechanism has been systematically explored for the first time through a comprehensive set of molecular biology methods mainly including immunohistochemistry, western blot and enzyme-linked immunosorbent assays. The toxicity profile of imperialine treatments were evaluated using healthy nude mice by examining hemogram and histopathology. An imperialine-loaded liposomal drug delivery system was developed using thin film hydration method to evaluate target specific delivery. RESULTS: The results showed that imperialine could suppress both NSCLC tumor and associated inflammation through an inflammation-cancer feedback loop in which NF-κB activity was dramatically inhibited by imperialine. The NSCLC-targeting liposomal system was successfully developed for targeted drug delivery. The developed platform could favorably enhance imperialine cellular uptake and in vivo accumulation at tumor sites, thus improving overall anti-tumor effect. The toxicity assays revealed imperialine treatments did not significantly disturb blood cell counts in mice or exert any significant damage to the main organs. CONCLUSIONS: Imperialine exerts anti-cancer effects against NSCLC both in vitro and in vivo, and this previously unknown function is related to NF-κB centered inflammation-cancer feedback loop. Imperialine mediated anti-cancer activity is not through cytotoxicity and exhibit robust systemic safety. Furthermore, the liposome-based system we commenced would dramatically enhance therapeutic effects of imperialine while exhibiting extremely low side effects both on cellular and in NSCLC model. This work has identified imperialine as a promising novel anti-cancer compound and offered an efficient target-delivery solution that greatly facilitate practical use of imperialine.


Subject(s)
Alkaloids/administration & dosage , Carcinoma, Non-Small-Cell Lung/drug therapy , Cevanes/administration & dosage , Drugs, Chinese Herbal/administration & dosage , Fritillaria/chemistry , Lung Neoplasms/drug therapy , A549 Cells , Alkaloids/adverse effects , Alkaloids/chemistry , Alkaloids/isolation & purification , Animals , Blood Cell Count , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/pathology , Cevanes/adverse effects , Cevanes/chemistry , Cevanes/isolation & purification , Drugs, Chinese Herbal/adverse effects , Drugs, Chinese Herbal/chemistry , Drugs, Chinese Herbal/isolation & purification , Feedback, Physiological/drug effects , Humans , Liposomes , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Male , Mice , NF-kappa B/antagonists & inhibitors , NF-kappa B/immunology , Toxicity Tests , Xenograft Model Antitumor Assays
12.
J Ethnopharmacol ; 261: 112338, 2020 Oct 28.
Article in English | MEDLINE | ID: mdl-31669666

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Fritillariae cirrhosae (FC), referred to'Chuan beimu'in China. As an important edible and medicinal plant, the bulbs of F.cirrhosae is used traditionally in the treatment of pulmonary diseases associated with lung heat, inflammation and tumors. In the study, we investigated the effect of aqueous extract of FC (FC-AE) and elucidated its mechanism in non-small cell lung cancer A549 cells and a xenograft model of nude mice. MATERIALS AND METHODS: CCK-8 and plate colony formation assay were used to evaluate the effect of FC-AE in A549 cells in vitro, and the gene expression profile of FC-AE on A549 cells was assessed by RNA sequencing system. Then, the effects of FC-AE on cell cycle and apoptosis of A549 cells were analyzed by flow cytometry. In combination with RNA-seq data, RT-PCR and western blot were used to evaluate the expression of proteins related to apoptosis and immune regulation. A xenograft model of nude mice was used to assess the effect of FC-AE in vivo. RESULTS: CCK-8 and plate cloning assays showed that FC-AE inhibited the proliferation and colony formation of A549 cells. A549 cells treated with FC-AE can triggered apoptosis. GO and KEGG pathway enrichment analysis of RNA-seq data showed that most of the differentially expressed genes (DEGs) were related to immune response, apoptosis and cell cycle process. Several immune and apoptotic DEGs were identified by qRT-PCR which were consistented with RNA-seq data. In nude mice, FC-AE reduced the tumor size and promoted the secretion of cytokines IL12 and IFNγ. FC-AE up-regulated the two members (STAT1 and STAT4) of STATs and their target genes (IFNγ and IL-12, respectively) protein expressions, and actively regulates Bcl-2/Bax family proteins which resulted in cellular apoptosis in A549 cells. CONCLUSION: Our finding suggests that FC-AE mediates apoptosis through a STAT1 and STAT4-mediated co-regulatory network, which may be the key novel mechanism for its antitumor activity. The F. cirrhosa may be a promising antitumor drug for modulating immune responses to improve cancer therapy.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/drug therapy , Fritillaria , Lung Neoplasms/drug therapy , Plant Extracts/pharmacology , STAT1 Transcription Factor/metabolism , STAT4 Transcription Factor/metabolism , A549 Cells , Animals , Antineoplastic Agents, Phytogenic/isolation & purification , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Proliferation/drug effects , Cytokines/genetics , Cytokines/metabolism , Female , Fritillaria/chemistry , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Humans , Lung Neoplasms/immunology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice, Nude , Plant Extracts/isolation & purification , STAT1 Transcription Factor/genetics , STAT4 Transcription Factor/genetics , Signal Transduction , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
13.
Pathol Oncol Res ; 26(1): 499-505, 2020 Jan.
Article in English | MEDLINE | ID: mdl-30460541

ABSTRACT

As a purified active component from traditional Chinese medicine, lentinan administration can be applied as beneficial chemo-immunotherapy for anti-tumor. In this study, the immunomodulatory effects of lentinan on aberrant T subsets and cytokines profile were evaluated for non-small cell lung cancer (NSCLC). Of all NSCLC patients treated with NP chemotherapeutic protocol (combination of vinorelbin and cisplatin), 73 cases were recruited in this retrospective cohort trial study, of which 38 cases received additional lentinan. The changes of aberrant T subsets and cytokines profile were compared between two groups (chemotherapy in combination with lentinan vs. conserved single chemotherapy) by flow cytometry and molecular biology. Higher subset ratio of CD3+CD8+ cytotoxic T cells was confirmed in the peripheral blood of NSCLC patients. Chemo-immunotherapy of lentinan resulted in a significant increase of CD3 + CD56+ NKT cells (15.7 ± 3.1%), compared with 8.6 ± 1.4% of NKT cells in single chemotherapy group, and up-regulated CD3+CD8+ and CD3+CD4+ subsets as well, but caused the decrease of CD4+CD25+ Tregs induction, accompanied by significant alleviation of IL-10 and TGF-ß1, and elevation of IFN-γ, TNF-α, and IL-12 (P < 0.05). It could be confirmed that lentinan could not only enhance the cellular immunity and promote the beneficial of anti-tumor by associated immunotherapy, but also had the ability to inhibit the expansion of immune suppressive Tregs in the NSCLC patients, in whom there was a raised Tregs induction compared to health control. Lentinan-based chemo-immunotherapy is a promising strategy for anti-tumor via enhancing the proliferation of cytotoxic T cells, followed by the elevation of inflammatory chemokines/cytokines. Meanwhile, the percentage of CD4+ CD25+ Tregs is down-regulated, leading to a shift in the inflammatory status from Th2 to Th1 in NSCLC patients treated with lentinan.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/immunology , Lentinan/administration & dosage , Lung Neoplasms/drug therapy , T-Lymphocyte Subsets/drug effects , Adult , Aged , Cisplatin/administration & dosage , Cytokines/drug effects , Cytokines/immunology , Female , Humans , Immunomodulation/immunology , Immunotherapy/methods , Lung Neoplasms/immunology , Male , Middle Aged , Retrospective Studies , Vinorelbine/administration & dosage
14.
Aging (Albany NY) ; 11(24): 11814-11828, 2019 12 23.
Article in English | MEDLINE | ID: mdl-31881010

ABSTRACT

The application of cancer chronotherapy is to treat cancers based on at specific times during circadian rhythms. Previous studies have characterized the impact of circadian clock on tumorigenesis and specific immune cells. Here, by using multi-omics computation techniques, we systematically characterized the distinct roles of core circadian clock genes in thoracic cancers including lung adenocarcinoma, lung squamous cell carcinoma, and esophageal carcinoma. Strikingly, a wide range of core clock genes are epigenetically altered in lung adenocarcinomas and lung squamous cell carcinomas but not esophageal carcinomas. Further cancer hallmark analysis reveals that several core clock genes highly correlate with apoptosis and cell cycle such as RORA and PER2. Interestingly, our results reveal that CD4 and CD8 T cells are correlated with core clock molecules especially in lung adenocarcinomas and lung squamous cell carcinomas, indicating that chrono-immunotherapy may serve as a candidate option for future cancer management.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Circadian Clocks/genetics , Esophageal Neoplasms/genetics , Lung Neoplasms/genetics , Tumor Microenvironment/physiology , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/pathology , DNA Methylation , Esophageal Neoplasms/immunology , Esophageal Neoplasms/pathology , Humans , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Transcriptome/genetics
15.
Integr Cancer Ther ; 18: 1534735419890020, 2019.
Article in English | MEDLINE | ID: mdl-31838881

ABSTRACT

Conventional methods in treating non-small cell lung cancer contain surgery, chemotherapy, radiotherapy, and targeted therapy, which have various defects. Recently, with the deeper research on tumor immunity, immunotherapy has made the breakthrough in the treatment of cancers. Especially developments of programmed cell death-1/programmed cell death ligand-1 (PD-1/PD-L1) inhibitors bring the therapy into a new stage. This review mainly focuses on introducing existing monoclonal antibodies containing nivolumab, pembrolizumab, atezolizumab, avelumab, and durvalumab, along with 3 ordinary biomarkers such as PD-L1 expression, tumor mutation burden, and microsatellite instability. By understanding the resistance mechanism of anti-PD-1/L1 blockade, research is further improving the survival benefit and expanding the benefit population. So, PD-1/PD-L1 inhibitors begin to be combined with various therapeutic strategies clinically. Discussion and comparison of their effectiveness and safety are also comprehensively reviewed. Meanwhile, we explore the potential, the impact, and mechanisms of combining traditional Chinese medicine with immunotherapy.


Subject(s)
Antineoplastic Agents/therapeutic use , B7-H1 Antigen/metabolism , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/therapy , Lung Neoplasms/immunology , Lung Neoplasms/therapy , Programmed Cell Death 1 Receptor/metabolism , Humans , Immunotherapy/methods , Medicine, Chinese Traditional/methods
16.
Chin J Nat Med ; 17(10): 729-737, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31703753

ABSTRACT

Non-small cell lung cancer (NSCLC) accounts for 80%-85% of all lung malignancies and good diagnosis and prognosis of NSCLC are critical to the increase of its survival rate. Tumor-associated macrophages (TAM) abundantly present in numerous cancer types, and the role of TAMs in tumor biology and their prognostic value in cancer become major topics of interest. After various stimulations in the tumor microenvironment, TAMs develop into a M1 (tumor-inhibitory) phenotype or M2 (tumor-promoting) phenotype. Recent studies show that traditional Chinese medicine (TCM) monomers have markedly inhibitory actions for NSCLC through M1/M2 modulation. Due to the TCM monomers mainly covered five categories, i.e. terpenoids, flavonoids, polysaccharides, natural polyphenols, and alkaloids. Thus, we will discuss the regulation of TCM monomers on TAM involve in these five parts in this review. In addition, the potential role of TAMs as therapeutic targets will be discussed.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Drugs, Chinese Herbal/administration & dosage , Lung Neoplasms/drug therapy , Animals , Carcinoma, Non-Small-Cell Lung/immunology , Humans , Lung Neoplasms/immunology , Macrophages/drug effects , Macrophages/immunology , Medicine, Chinese Traditional , Prognosis , Tumor Microenvironment/drug effects
17.
Eur J Cancer ; 122: 22-41, 2019 11.
Article in English | MEDLINE | ID: mdl-31606656

ABSTRACT

The Food and Drug Administration has lately approved atezolizumab, anti-programmed death ligand 1 (PD-L1), to be used together with nanoparticle albumin-bound (nab) paclitaxel in treating patients with triple negative breast cancer (BC) expressing PD-L1. Nonetheless, immune checkpoint inhibitors (ICIs) are still challenged by the resistance and immune-related adverse effects evident in a considerable subset of treated patients without conclusive comprehension of the underlying molecular basis, biomarkers and tolerable therapeutic regimens capable of unleashing the anti-tumour immune responses. Stepping back to preclinical models is thus inevitable to address these inquiries. Herein, we comprehensively review diverse preclinical models of BC exploited in investigating ICIs underscoring their pros and cons as well as the learnt and awaited lessons to allow full exploitation of ICIs in BC therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Cell Cycle Checkpoints/immunology , Drug Evaluation, Preclinical , Immunologic Factors/therapeutic use , Immunotherapy/methods , Triple Negative Breast Neoplasms/drug therapy , Antibodies, Monoclonal, Humanized/therapeutic use , CTLA-4 Antigen/immunology , Carcinoma, Non-Small-Cell Lung/immunology , Humans , Programmed Cell Death 1 Receptor/immunology , Triple Negative Breast Neoplasms/immunology
18.
Zhongguo Fei Ai Za Zhi ; 22(9): 583-589, 2019 Sep 20.
Article in Chinese | MEDLINE | ID: mdl-31526463

ABSTRACT

BRAF gene mutation is found in about 2%-4% of the patients with non-small cell lung cancer (NSCLC). This type of NSCLC is characterized by high malignancy, low efficacy of chemotherapy and poor prognosis. Although the combination treatment of BRAF inhibitor and MEK inhibitor has achieved remarkable results in advanced NSCLC patients with BRAF V600E mutation, which has been written into the National Comprehensive Cancer Network (NCCN) guidelines, severe side effects of the combination therapy are frequently observed. There isn't effective treatment strategy after drug resistance, and targeted therapy for non-V600E mutation patients is still lacking. In this paper, we summarized the researches on expression of immune markers in NSCLC patients with mutant BRAF and analyzed the studies on efficacy of immune checkpoint inhibitor (ICI), so as to provide more options for prolonging survival of the patients.
.


Subject(s)
Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/therapy , Lung Neoplasms/genetics , Lung Neoplasms/therapy , Mutation , Proto-Oncogene Proteins B-raf/genetics , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Humans , Immunity , Lung Neoplasms/drug therapy , Lung Neoplasms/immunology , Molecular Targeted Therapy
19.
Immunotherapy ; 11(13): 1161-1176, 2019 09.
Article in English | MEDLINE | ID: mdl-31361169

ABSTRACT

Lung cancer is the leading cause of cancer deaths worldwide, with a 5-year survival rate of about 18%. Thus, there is a great need for novel therapeutic approaches to treat non-small-cell lung cancer (NSCLC). Immune checkpoint inhibitors (ICIs) have improved outcomes for a subset of patients, especially those with high programmed death-ligand 1 expression and/or high tumor mutational burden, but have failed in the majority of patients. Increasing evidence suggests that the estrogen signaling pathway may be a therapeutic target in metastatic NSCLC and that the estrogen pathway may play a role in sex-based responses to ICIs. This report will review the epidemiologic, preclinical and clinical data on the estrogen pathway in NSCLC, its implications in sex-based responses to ICIs and the potential use of antiestrogen therapy in combination with ICIs.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Carcinoma, Non-Small-Cell Lung/therapy , Estrogen Receptor Modulators/therapeutic use , Estrogens/metabolism , Immunotherapy/methods , Lung Neoplasms/therapy , Sex Factors , Animals , Carcinoma, Non-Small-Cell Lung/epidemiology , Carcinoma, Non-Small-Cell Lung/immunology , Clinical Trials as Topic , Drug Evaluation, Preclinical , Drug Therapy, Combination , Humans , Lung Neoplasms/epidemiology , Lung Neoplasms/immunology , Neoplasm Metastasis , Signal Transduction , Treatment Outcome
20.
Clin Cancer Res ; 25(12): 3630-3642, 2019 06 15.
Article in English | MEDLINE | ID: mdl-30796032

ABSTRACT

PURPOSE: We aimed to investigate efficacy and mechanism of MTI-31 (LXI-15029), a novel mTORC1/mTORC2 inhibitor currently in human trial (NCT03125746), in non-small cell lung cancer (NSCLC) models of multiple driver mutations and tyrosine kinase inhibitor (TKI)-resistance. EXPERIMENTAL DESIGN: Gene depletion, inhibitor treatment, immunological, flow cytometry, cellular, and animal studies were performed to determine in vitro and in vivo efficacy in NSCLC models of driver mutations and elucidate roles by mTOR complexes in regulating migration, epithelial-mesenchymal transition (EMT), metastasis, intracranial tumor growth, and immune-escape. RESULTS: MTI-31 potently inhibited cell proliferation (IC50 <1 µmol/L) and in vivo tumor growth in multiple NSCLC models of EGFR/T790M, EML4-ALK, c-Met, or KRAS (MED <10 mg/kg). In EGFR-mutant and/or EML4-ALK-driven NSCLC, MTI-31 or disruption of mTORC2 reduced cell migration, hematogenous metastasis to the lung, and abrogated morphological and functional traits of EMT. Disruption of mTORC2 inhibited EGFR/T790M-positive tumor growth in mouse brain and prolonged animal survival correlating a diminished tumor angiogenesis and recruitment of IBA1+ microglia/macrophages in tumor microenvironment. MTI-31 also suppressed programmed death ligand 1 (PD-L1) in EGFR- and ALK-driven NSCLC, mediated in part by mTORC2/AKT/GSK3ß-dependent proteasomal degradation. Depletion of mTOR protein or disruption of mTOR complexes profoundly downregulated PD-L1 and alleviated apoptosis in Jurkat T and primary human T cells in a tumor-T cell coculture system. CONCLUSIONS: Our results highlight mTOR as a multifaceted regulator of tumor growth, metastasis, and immune-escape in EGFR/ALK-mutant and TKI-resistant NSCLC cells. The newly characterized mechanisms mediated by the rapamycin-resistant mTORC2 warrant clinical investigation of mTORC1/mTORC2 inhibitors in patients with lung cancer.


Subject(s)
Brain Neoplasms/drug therapy , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Mechanistic Target of Rapamycin Complex 2/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Animals , Brain Neoplasms/immunology , Brain Neoplasms/metabolism , Brain Neoplasms/secondary , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Evaluation, Preclinical/methods , Epithelial-Mesenchymal Transition , Female , Humans , Immunity, Cellular/drug effects , Lung Neoplasms/immunology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mechanistic Target of Rapamycin Complex 1/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL