1.Chinese multidisciplinary expert consensus on the management of adverse drug reactions associated with savolitinib.
Li ZHANG ; Yong Sheng WANG ; Li Zhu LIN ; Yong Feng YU ; Shun LU
Chinese Journal of Oncology 2023;45(4):298-312
MET gene is a proto-oncogene, which encodes MET protein with tyrosine kinase activity. After binding to its ligand, hepatocyte growth factor, MET protein can induce MET dimerization and activate downstream signaling pathways, which plays a crucial role in tumor formation and metastasis. Savolitinib, as a specific tyrosine kinase inhibitor (TKI) targeting MET, selectively inhibits the phosphorylation of MET kinase with a significant inhibitory effect on tumors with MET abnormalities. Based on its significant efficacy shown in the registration studies, savolitinib was approved for marketing in China on June 22, 2021 for the treatment of advanced non-small cell lung cancer with MET 14 exon skipping mutations. In addition, many studies have shown that MET TKIs are equally effective in patients with advanced solid tumors with MET gene amplification or MET protein overexpression, and relevant registration clinical studies are ongoing. The most common adverse reactions during treatment with savolitinib include nausea, vomiting, peripheral edema, pyrexia, and hepatotoxicity. Based on two rounds of extensive nationwide investigations to guide clinicians, the consensus is compiled to use savolitinib rationally, prevent and treat various adverse reactions scientifically, and improve the clinical benefits and quality of life of patients. This consensus was prepared under the guidance of multidisciplinary experts, especially including the whole-process participation and valuable suggestions of experts in Traditional Chinese Medicine, thus reflecting the clinical treatment concept of integrated Chinese and western medicines.
Humans
;
Carcinoma, Non-Small-Cell Lung/genetics*
;
Lung Neoplasms/pathology*
;
Consensus
;
Quality of Life
;
Proto-Oncogene Proteins c-met/genetics*
;
Protein Kinase Inhibitors/adverse effects*
;
Drug-Related Side Effects and Adverse Reactions
;
Mutation
2.Advances in Predictive Research of Immune Checkpoint Inhibitors-related Adverse Events.
Jing ZHANG ; Xueqin CHEN ; Shenglin MA
Chinese Journal of Lung Cancer 2023;26(10):789-794
The era of tumor treatment has been revolutionized by the advent of immune checkpoint inhibitors. However, while immunotherapy benefits patients, it can also lead to immune-related adverse events that may affect multiple organs and systems throughout the body, potentially even posing a life-threatening risk. The diverse clinical manifestations and onset times of these adverse events further complicate their prediction and diagnosis. The purpose of this paper is to review the clinical characteristics and predicted biomarkers of adverse events related to inhibitors at immune checkpoints, in order to help clinicians evaluate drug risks and early warn adverse events.
.
Humans
;
Immune Checkpoint Inhibitors/adverse effects*
;
Lung Neoplasms/drug therapy*
;
Neoplasms/pathology*
;
Immunotherapy/adverse effects*
3.Puerarin Inhibits the Proliferation,Invasion,and Migration of Non-small Cell Lung Cancer Cells through Regulating miR-490/Denticleless E3 Ubiquitin Protein Ligase.
Yu-Xin ZHANG ; Zhen-Zhen ZHANG ; Li-Gang ZHAO ; Lin-Lin ZHAO
Acta Academiae Medicinae Sinicae 2022;44(1):91-101
Objective To explore the mechanism of puerarin inhibiting the proliferation,invasion,and migration of non-small cell lung cancer cells. Methods A549 cells were cultured and treated with different concentrations of puerarin.The inhibition rate (IR) on cell proliferation was detected by CCK-8,and qRT-PCR was performed to detect the mRNA levels of miR-490 and denticleless E3 ubiquitin protein ligase(DTL).Double luciferase reporter assay was employed to identify the targets of miR-490 and DTL based on the establishment of NC mimic group,miR-490 mimic group,NC inhibitor group,and miR-490 inhibitor group.The cells treated by 20 μmol/L puerarin were classified into six groups:DMSO,puerarin,puerarin+NC inhibitor,puerarin+miR-490 inhibitor,puerarin+miR-490 inhibitor+Si-NC,and puerarin+miR-490 inhibitor+Si-DTL.Transwell was used to detect cell migration and invasion.Western blotting was performed to detect the protein levels of epithelial-mesenchymal transition-related markers E-cadherin,N-cadherin,and Vimentin. Results With the increase in puerarin concentration,the IR gradually elevated (F=105.375,P<0.001),miR-490 expression gradually increased (F=32.919,P<0.001),and DTL expression gradually decreased (F=116.120,P<0.001).Compared with NC mimic group,miR-490 mimic group had decreased luciferase activity (t=7.762,P=0.016),raised miR-490 mRNA level (t=13.319,P<0.001),and declined DTL mRNA level (t=7.415,P=0.002).Compared with those in NC inhibitor group,miR-490 demonstrated decreased mRNA level (t=9.523,P=0.001) and DTL presented increased mRNA level (t=11.305,P<0.001) in miR-490 inhibitor group.Western blotting showed that the protein level of DTL was higher in NC mimic group (t=7.953,P=0.001) than in miR-490 mimic group and higher in miR-490 inhibitor group than in NC inhibitor group (t=10.552,P<0.001).Compared with DMSO group,puerarin group showed up-regulated mRNA level of miR-490 (t=10.255,P=0.001) while down-regulated mRNA level of DTL (t=6.682,P=0.003).Compared with those in puerarin+NC inhibitor group,the mRNA level of miR-490 declined (t=10.995,P<0.001) while that of DTL raised (t=12.478,P<0.001) in puerarin+miR-490 inhibitor group.The mRNA level of miR-490 had no significant difference between puerarin+miR-490 inhibitor+Si-NC group and puerarin+miR-490 inhibitor+Si-DTL group (t=1.081,P=0.341),and that of DTL was lower in the latter group (t=14.321,P<0.001).The protein level of DTL was higher in puerarin+miR-490 inhibitor group than in puerarin+NC inhibitor group (t=11.423,P<0.001),and lower in puerarin+miR-490 inhibitor+Si-DTL group than in puerarin+miR-490 inhibitor+Si-NC group (t=12.080,P<0.001).Compared with DMSO group,puerarin group showed inhibited cell proliferation (F=129.27,P<0.001).The activity of cell proliferation was higher in puerarin+miR-490 inhibitor group than in puerarin+NC inhibitor group (F=75.12,P<0.001),and higher in puerarin+miR-490 inhibitor+Si-NC group than in puerarin+miR-490 inhibitor+Si-DTL group (F=52.59,P<0.001).Compared with DMSO group,puerarin group had suppressed cell migration (t=8.963,P=0.001).The cell migration ability was higher in puerarin+miR-490 inhibitor group than in puerarin+NC inhibitor group (t=12.117,P<0.001) and higher in puerarin+miR-490 inhibitor+Si-NC group than in puerarin+miR-490 inhibitor+Si-DTL group (t=12.934,P<0.001).Puerarin group showed weakened cell invasion ability compared with DMSO group (t=4.710,P=0.009).The cell invasion ability was higher in puerarin+miR-490 inhibitor group than in puerarin+NC inhibitor group (t=13.264,P<0.001) and lower in puerarin+miR-490 inhibitor+Si-DTL group than in puerarin+miR-490 inhibitor+Si-NC group (t=13.476,P<0.001).Compared with DMSO group,puerarin group showed up-regulated protein level of E-cadherin (t=7.137,P=0.002) while down-regulated protein levels of N-cadherin (t=8.828,P=0.001) and vimentin (t=6.594,P=0.003).Compared with those in puerarin+NC inhibitor group,the protein level of E-cadherin (t=12.376,P<0.001) decreased while those of N-cadherin (t=13.436,P<0.001) and vimentin (t=11.467,P<0.001) increased in puerarin+miR-490 inhibitor group.Compared with puerarin+miR-490 inhibitor+Si-NC group,puerarin+miR-490 inhibitor+Si-DTL group up-regulated the protein level of E-cadherin (t=13.081,P<0.001) while down-regulated the protein levels of N-cadherin (t=10.835,P<0.001) and vimentin (t=11.862,P<0.001). Conclusion Puerarin could inhibit the proliferation,invasion,and migration of non-small cell lung cancer cells by up-regulating miR-490 and down-regulating DTL.
Carcinoma, Non-Small-Cell Lung/pathology*
;
Cell Line, Tumor
;
Cell Movement/drug effects*
;
Cell Proliferation/drug effects*
;
Humans
;
Isoflavones/pharmacology*
;
Lung Neoplasms
;
MicroRNAs/metabolism*
;
Ubiquitin-Protein Ligases/metabolism*
4.Expert consensus on ensartinib in the treatment of anaplastic lymphoma kinase-positive non-small cell lung cancer.
Chinese Journal of Oncology 2022;44(4):297-307
The mutation rate of anaplastic lymphoma kinase (ALK) in patients with non-small cell lung cancer is 3% to 7%. Due to its low mutation rate and better long-term survival compared with epidermal growth factor receptor-positive non-small cell lung cancer patients, therefore, it's called "diamond mutation". At present, there are three generations of ALK tyrosine kinase inhibitor (TKI) drugs in the world. The first-generation ALK-TKI drug approved in China is crizotinib, and the second-generation drugs are alectinib, ceritinib and ensartinib. Among them, ensartinib is an ALK-TKI domestically developed, and its efficacy is similar to that of alectinib. The main adverse event is transient rash, and compliance to ensartinib is better from the perspective of long-term survival of patients. The manifestation of rash caused by ensartinib is different from that of other ALK-TKI drugs. In order to facilitate clinical application and provide patients with more treatment options, under the guidance of the Committee of Cancer Rehabilitation and Palliative Care of China Anti-Cancer Association, this article collects and summarizes the common adverse reactions of ensartinib. Based on the clinical practice, a clear adverse classification and specific treatment plan are formulated, in order to provide a corresponding reference for clinicians to make more comprehensive clinical decisions.
Anaplastic Lymphoma Kinase
;
Carbazoles/adverse effects*
;
Carcinoma, Non-Small-Cell Lung/pathology*
;
Consensus
;
Exanthema/drug therapy*
;
Humans
;
Lung Neoplasms/pathology*
;
Piperazines
;
Protein Kinase Inhibitors/adverse effects*
;
Pyridazines
5.Preparation of purified proteins from fresh Pheretima and their inhibitory effect against pulmonary fibrosis in mice.
Shu Yu LI ; Qi Xin YANG ; An Na ZUO ; Lin Hua TIAN ; Jin Hai HUO ; Yan Li MENG ; Qing Fa TANG ; Wei Ming WANG
Journal of Southern Medical University 2022;42(4):618-624
OBJECTIVE:
To develop a convenient method for rapid purification of fresh Pheretima proteins and assess the inhibitory effect of these proteins against pulmonary fibrosis.
METHODS:
The crude extract of fresh Pheretima was obtained by freeze-drying method and then purified by size exclusion chromatography. The composition of the purified proteins was analyzed by mass spectrometry. MRC-5 cells were treated with 5 ng/mL TGF-β1 alone (model group) or in combination with SB431542 (2 μmol/L) or the purified proteins (13.125 μg/mL), and the cytotoxicity of purified proteins and their inhibitory effects on cell proliferation were detected with CCK8 assay. Flow cytometry was used to detect the changes in cell apoptosis, and the cellular expressions of α-SMA, Vimentin, E-cadherin, collagen I, Smad2/3 and P-Smad2/3 were detected using RT-PCR and Western blotting. In the animal experiment, adult male C57BL/6 mice were subjected to intratracheal instillation of bleomycin followed by treatment with the purified proteins (5 mg/mL) for 21 days, after which HE and Masson staining was used to observe the pathological changes in the lung tissue of the mice.
RESULTS:
We successfully obtained purified proteins from fresh Pheretima protein by size exclusion chromatography. Treatment with the purified proteins significantly inhibited TGF-β1-induced proliferation of MRC-5 cells (P < 0.01), reduced the cellular expressions of α-SMA, Vimentin and collagen I (P < 0.001 or P < 0.01), increased the expression of E-cadherin (P < 0.01), and inhibited the expressions of Smad2/3 and P-Smad2/3 (P < 0.001 or P < 0.01). In male C57BL/6 mice models of bleomycin-induced pulmonary fibrosis, treatment with the purified proteins obviously reduced the number of inflammatory cells and fibrotic area in the lungs.
CONCLUSION
The purified proteins from fresh Pheretima obtained by size exclusion chromatography can inhibit pulmonary fibrosis in mice by regulating the TGF-β/ Smad pathway.
Animals
;
Biological Products/pharmacology*
;
Bleomycin/adverse effects*
;
Cadherins/metabolism*
;
Collagen Type I
;
Lung/pathology*
;
Male
;
Mice
;
Mice, Inbred C57BL
;
Oligochaeta/chemistry*
;
Pulmonary Fibrosis/drug therapy*
;
Transforming Growth Factor beta1/metabolism*
;
Vimentin/metabolism*
6.A review: drug-drug interactions of epithelial growth factor receptor-tyrosine kinase inhibitors.
Chinese Journal of Oncology 2022;44(7):717-724
Mutations in the epithelial growth factor receptor (EGFR) is a driving factor that causes non-small cell lung carcinoma (NSCLC). The epithelial growth factor receptor-tyrosine kinase inhibitors (EGFR-TKIs) is a crucial discovery in the treatment of lung cancer, particularly the efficacy of EGFR-TKIs is superior to that of the standard chemotherapy for patients with EGFR mutation-positive advanced NSCLC. Patients with NSCLC use EGFR-TKIs and other medications simultaneously is commonly seen, especially among those with comorbidities, which increases the risk of drug-drug interactions (DDIs) of EGFR-TKIs. The most common mechanisms underlying the DDIs of EGFR-TKIs are modulations of cytochrome P450 (CYP) and drug transporters [including P-glycoprotein (P-gp) and breast cancer resistance protein (BCRP)], as well as gastrointestinal acid-inhibitory drugs [proton pump inhibitors (PPIs) and H(2) receptor antagonists (H(2)RA)]. Inhibitors or inducers of CYP enzymes and drug transporters can inhibit or accelerate the metabolism of EGFR-TKIs, which increase or reduce the exposure of EGFR-TKIs, thereby affect the efficacy and safety of EGFR-TKIs. In addition, PPIs or H(2)RA can decrease the solubility, bioavailability and efficacy of EGFR-TKIs. This review summarizes the mechanisms of DDIs of gefitinib, erlotinib, icotinib, afatinib, dacomitinib and osimertinib; the management recommendations for DDIs of those EGFR-TKIs from the Chinese and global guideline, as well as from the recent pre-clinical and clinical studies, which provide the reference and evidence for managing the combination therapies of EGFR-TKIs and other medications in clinics.
ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics*
;
Carcinoma, Non-Small-Cell Lung/pathology*
;
Drug Interactions
;
ErbB Receptors/genetics*
;
Humans
;
Lung Neoplasms/pathology*
;
Mutation
;
Neoplasm Proteins/metabolism*
;
Protein Kinase Inhibitors/adverse effects*
7.Chinese Expert Consensus on Management of Special Adverse Effects Associated with Lorlatinib.
Qing ZHOU ; Shun LU ; Yong LI ; Fujun JIA ; Guanjun LI ; Zhen HONG ; You LU ; Yun FAN ; Jianying ZHOU ; Zhe LIU ; Juan LI ; Yi-Long WU
Chinese Journal of Lung Cancer 2022;25(8):555-566
Anaplastic lymphoma kinase (ALK) fusions represent the second most common oncogenic driver mutation in non-small cell lung cancer (NSCLC). As the new class of 3rd generation of ALK tyrosine kinase inhibitor (TKI), lorlatinib has shown robust potency and brain-penetrant clinical activity against a wide spectrum of multiple resistance mutations within the ALK domain detected during crizotinib and 2nd generation ALK TKI treatment. Lorlatinib is generally well-tolerated with unique adverse drug reaction/adverse event, including hyperlipidemia and central nervous system effects, which are mostly mild to moderate severity and manageable through dosage modifications and/or standard medical intervention. For advanced NSCLC with ALK positivity, patients should be evaluated for baseline characteristics and pre-existing medication, informed of the potential toxicities, and periodically monitored to balance benefits and risks. Moreover, a multidisciplinary group of experts is essential to establish a comprehensive diagnostic and therapeutic strategy.
.
Aminopyridines
;
Carcinoma, Non-Small-Cell Lung/pathology*
;
China
;
Consensus
;
Drug Resistance, Neoplasm/genetics*
;
Drug-Related Side Effects and Adverse Reactions/drug therapy*
;
Humans
;
Lactams
;
Lactams, Macrocyclic/adverse effects*
;
Lung Neoplasms/pathology*
;
Protein Kinase Inhibitors/adverse effects*
;
Protein-Tyrosine Kinases/genetics*
;
Pyrazoles
8.Lowest observed adverse effect level of pulmonary pathological alterations due to nitrous acid exposure in guinea pigs.
Masayuki OHYAMA ; Hiroshi NISHIMURA ; Kenichi AZUMA ; Chika MINEJIMA ; Norimichi TAKENAKA ; Shuichi ADACHI
Environmental Health and Preventive Medicine 2020;25(1):56-56
BACKGROUND:
We previously demonstrated that continuous exposure to nitrous acid gas (HONO) for 4 weeks, at a concentration of 3.6 parts per million (ppm), induced pulmonary emphysema-like alterations in guinea pigs. In addition, we found that HONO affected asthma symptoms, based on the measurement of respiratory function in rats exposed to 5.8 ppm HONO. This study aimed to investigate the dose-response effects of HONO exposure on the histopathological alterations in the respiratory tract of guinea pigs to determine the lowest observed adverse effect level (LOAEL) of HONO.
METHODS:
We continuously exposed male Hartley guinea pigs (n = 5) to four different concentrations of HONO (0.0, 0.1, 0.4, and 1.7 ppm) for 4 weeks (24 h/day). We performed histopathological analysis by observing lung tissue samples. We examined samples from three guinea pigs in each group under a light microscope and measured the alveolar mean linear intercept (Lm) and the thickness of the bronchial smooth muscle layer. We further examined samples from two guinea pigs in each group under a scanning electron microscope (SEM) and a transmission electron microscope (TEM).
RESULTS:
We observed the following dose-dependent changes: pulmonary emphysema-like alterations in the centriacinar regions of alveolar ducts, significant increase in Lm in the 1.7 ppm HONO-exposure group, tendency for hyperplasia and pseudostratification of bronchial epithelial cells, and extension of the bronchial epithelial cells and smooth muscle cells in the alveolar duct regions.
CONCLUSIONS
These histopathological findings suggest that the LOAEL of HONO is < 0.1 ppm.
Alveolar Epithelial Cells
;
drug effects
;
Animals
;
Bronchi
;
drug effects
;
Dose-Response Relationship, Drug
;
Emphysema
;
chemically induced
;
Epithelial Cells
;
drug effects
;
Guinea Pigs
;
Hyperplasia
;
chemically induced
;
Inhalation Exposure
;
adverse effects
;
Lung
;
drug effects
;
pathology
;
ultrastructure
;
Male
;
Microscopy, Electron, Scanning
;
Microscopy, Electron, Transmission
;
Myocytes, Smooth Muscle
;
drug effects
;
Nitrous Acid
;
toxicity
9.Carfilzomib inhibits the growth of lung adenocarcinoma via upregulation of Gadd45a expression.
Fang YANG ; Wang-Wang LIU ; Hui CHEN ; Jia ZHU ; Ai-Hua HUANG ; Fei ZHOU ; Yi GAN ; Yan-Hua ZHANG ; Li MA
Journal of Zhejiang University. Science. B 2020;21(1):64-76
Proteasome inhibitors have shown remarkable success in the treatment of hematologic neoplasm. There has been a lot of attention to applying these drugs for solid tumor treatment. Recent preclinical study has signified the effectiveness on cell proliferation inhibition in lung adenocarcinoma treated by carfilzomib (CFZ), a second generation proteasome inhibitor. However, no insight has been gained regarding the mechanism. In this study, we have systematically investigated the CFZ functions in cell proliferation and growth, cell cycle arrest, and apoptosis in lung adenocarcinoma cells. Flow cytometry experiments showed that CFZ significantly induced G2/M cell cycle arrest and apoptosis in lung adenocarcinoma. MTS and colony formation assays revealed that CFZ substantially inhibited survival of lung adenocarcinoma cells. All results were consistently correlated to the upregulation expression of Gadd45a, which is an important gene in modulating cell cycle arrest and apoptosis in response to physiologic and environmental stresses. Here, upregulation of Gadd45a expression was observed after CFZ treatment. Knocking down Gadd45a expression suppressed G2/M arrest and apoptosis in CFZ-treated cells, and reduced cytotoxicity of this drug. The protein expression analysis has further identified that the AKT/FOXO3a pathway is involved in Gadd45a upregulation after CFZ treatment. These findings unveil a novel mechanism of proteasome inhibitor in anti-solid tumor activity, and shed light on novel preferable therapeutic strategy for lung adenocarcinoma. We believe that Gadd45a expression can be a highly promising candidate predictor in evaluating the efficacy of proteasome inhibitors in solid tumor therapy.
Adenocarcinoma of Lung/pathology*
;
Apoptosis/drug effects*
;
Cell Cycle Checkpoints/drug effects*
;
Cell Cycle Proteins/genetics*
;
Cell Line, Tumor
;
Forkhead Box Protein O3/physiology*
;
Gene Expression Regulation, Neoplastic/drug effects*
;
Humans
;
Lung Neoplasms/pathology*
;
Oligopeptides/pharmacology*
;
Proto-Oncogene Proteins c-akt/physiology*
;
Up-Regulation
10.Effect of methanol-ethyl acetate partitioned fractions from on proliferation and apoptosis of human non-small cell lung cancer H1975 cells.
Jiahui GUI ; Meilin ZHU ; Xiangjian BAI ; Bohan LI ; Meijia GAO ; Hui MA ; Hongmei LI ; Chengzhu WU
Journal of Southern Medical University 2019;39(2):169-174
OBJECTIVE:
To investigate the effects of methanol-ethyl acetate partitioned fractions from (MEDS) on the proliferation and apoptosis of human non-small cell lung cancer H1975 cells.
METHODS:
The systemic solvent extraction method was used to preliminary separation of the effective fractions in the methanol extract of . The cytotoxicity of each extract (5, 10, 20, 40, and 80 μg/mL) was tested using MTT assay. Colony cloning method was used to assess the effect of different concentrations of methanol-ethyl acetate partitioned fractions from MEDS (5, 10, 20, 40, and 80 μg/ mL) on the proliferation of H1975 cells. Flow cytometric analysis with Annexin V-FITC/PI staining was performed to detect the apoptosis of the cells after treatment with different concentrations of MEDS fractions (10, 20, and 40 μg/mL). Western blotting was used to evaluate the effects of MEDS fractions on the expressions of apoptosis-related proteins Akt, Bax, and Bcl-2. The anti-tumor activity of 100 mg/kg MEDS fractions was tested in a nude mouse model bearing H1975 cell xenografts.
RESULTS:
MTT assay and colony forming experiment showed that MEDS fractions significantly inhibited the proliferation of H1975 cells in a dose-and time-dependent manner ( < 0.05). The results of flow cytometry showed that MEDS fractions induced obvious apoptosis of H1975 cells in a concentration-dependent manner ( < 0.05). MEDS fractions also significantly decreased the expressions of Bcl-2 and Akt protein and increased the protein expression of Bax ( < 0.05). In the tumor-bearing nude mouse model, MEDS fractions showed potent anti-tumor effects with a low toxicity to affect the body weight and organs of the mice.
CONCLUSIONS
The methanol-ethyl acetate partitioned fractions from MEDS show potent anti-tumor activity both and , suggesting their value as promising therapeutic agents against lung cancer.
Acetates
;
Animals
;
Antineoplastic Agents, Phytogenic
;
isolation & purification
;
pharmacology
;
Apoptosis
;
drug effects
;
Carcinoma, Non-Small-Cell Lung
;
pathology
;
Cell Line, Tumor
;
Cell Proliferation
;
drug effects
;
Heterografts
;
Humans
;
Lung Neoplasms
;
pathology
;
Methanol
;
Mice
;
Mice, Nude
;
Plant Extracts
;
isolation & purification
;
pharmacology

Result Analysis
Print
Save
E-mail