401
|
Belić M, Sopić M, Roksandić-Milenković M, Ćeriman V, Guzonijić A, Vukašinović A, Ostanek B, Dimić N, Jovanović D, Kotur-Stevuljević J. Correlation of Short Leukocyte Telomeres and Oxidative Stress with the Presence and Severity of Lung Cancer Explored by Principal Component Analysis. Folia Biol (Praha) 2023; 69:59-68. [PMID: 38063002 DOI: 10.14712/fb2023069020059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
Lung cancer (LC) is the second most common malignancy and leading cause of cancer death. The potential "culprit" for local and systemic telomere shortening in LC patients is oxidative stress. We investigated the correlation between the peripheral blood leukocyte (PBL) telomere length (TL) and the presence/severity of LC and oxidative stress, and its usefulness as LC diagnostic marker. PBL TL was measured in 89 LC patients and 83 healthy subjects using the modified Cawthon RTq-PCR method. The relative PBL TL, found to be a potential diagnostic marker for LC with very good accuracy (P < 0.001), was significantly shorter in patients compared to the control group (CG) (P < 0.001). Significantly shorter telomeres were found in patients with LC TNM stage IV than in patients with stages I-III (P = 0.014), in patients without therapy compared to those on therapy (P = 0.008), and in patients with partial response and stable/progressive disease compared to those with complete response (P = 0.039). The total oxidant status (TOS), advanced oxidation protein products (AOPP), prooxidant-antioxidant balance (PAB) and C-reactive protein (CRP) were significantly higher in patients compared to CG (P < 0.001) and correlated negatively with TL in both patients and CG (P < 0.001). PCA showed a relation between PAB and TL, and between the EGFR status and TL. Oxidative stress and PBL telomere shortening are probably associated with LC development and progression.
Collapse
Affiliation(s)
| | - Miron Sopić
- Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, Serbia.
| | | | - Vesna Ćeriman
- Institute for Lung Diseases, Thoracic Surgery and Tuberculosis, Clinical Center of Serbia, Belgrade, Serbia
| | - Azra Guzonijić
- Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, Serbia
| | - Aleksandra Vukašinović
- Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Italy
| | - Barbara Ostanek
- Department of Clinical Biochemistry, Faculty of Pharmacy, University of Ljubljana, Slovenia
| | - Nemanja Dimić
- University Clinical-Hospital Center Dr. Dragisa Misovic, Belgrade, Serbia
| | | | | |
Collapse
|
402
|
Machado-Rugolo J, Baldavira CM, Prieto TG, Olivieri EHR, Fabro AT, Rainho CA, Castelli EC, Ribolla PEM, Ab'Saber AM, Takagaki T, Nagai MA, Capelozzi VL. Concomitant TP53 mutation in early-stage resected EGFR-mutated non-small cell lung cancer: a narrative approach in a genetically admixed Brazilian cohort. Braz J Med Biol Res 2023; 56:e12488. [PMID: 37042869 PMCID: PMC10085757 DOI: 10.1590/1414-431x2023e12488] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 02/27/2023] [Indexed: 04/13/2023] Open
Abstract
TP53 mutations are frequent in non-small cell lung cancer (NSCLC) and have been associated with poor outcome. The prognostic and predictive relevance of EGFR/TP53 co-mutations in NSCLC is controversial. We analyzed lung tissue specimens from 70 patients with NSCLC using next-generation sequencing to determine EGFR and TP53 status and the association between these status with baseline patient and tumor characteristics, adjuvant treatments, relapse, and progression-free (PFS) and overall survival (OS) after surgical resection. We found the EGFR mutation in 32.9% of patients (20% classical mutations and 12.9% uncommon mutations). TP53 missense mutations occurred in 25.7% and TP53/EGFR co-mutations occurred in 43.5% of patients. Stage after surgical resection was significantly associated with OS (P=0.028). We identified an association between progression-free survival and poor outcome in patients with distant metastases (P=0.007). We found a marginally significant difference in OS between genders (P=0.057) and between mutant and wild type TP53 (P=0.079). In univariate analysis, distant metastases (P=0.027), pathological stage (IIIA-IIIB vs I-II; P=0.028), and TP53 status (borderline significance between wild type and mutant; P=0.079) influenced OS. In multivariable analysis, a significant model for high risk of death and poor OS (P=0.029) selected patients in stage IIIA-IIIB, with relapse and distant metastases, non-responsive to platin-based chemotherapy and erlotinib, with tumors harboring EGFR uncommon mutations, with TP53 mutant, and with EGFR/TP53 co-mutations. Our study suggested that TP53 mutation tends to confer poor survival and a potentially negative predictive effect associated with a non-response to platinum-based chemotherapy and erlotinib in early-stage resected EGFR-mutated NSCLC.
Collapse
Affiliation(s)
- J Machado-Rugolo
- Laboratório de Histomorfometria e Genômica Pulmonar, Departamento de Patologia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
- Centro de Avaliação de Tecnologias em Saúde, Hospital das Clínicas da Faculdade de Medicina de Botucatu, Universidade Estadual Paulista, Botucatu, SP, Brasil
| | - C M Baldavira
- Laboratório de Histomorfometria e Genômica Pulmonar, Departamento de Patologia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
| | - T G Prieto
- Laboratório de Histomorfometria e Genômica Pulmonar, Departamento de Patologia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
| | - E H R Olivieri
- Centro Internacional de Pesquisa/CIPE, AC Camargo Cancer Center, São Paulo, São Paulo, SP, Brasil
| | - A T Fabro
- Laboratório de Histomorfometria e Genômica Pulmonar, Departamento de Patologia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
- Departamento de Patologia e Medicina Legal, Laboratório de Medicina Respiratória, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brasil
| | - C A Rainho
- Instituto de Biociências, Departamento de Ciências Químicas e Biológicas, Universidade Estadual Paulista, Botucatu, SP, Brasil
| | - E C Castelli
- Laboratório de Genética Molecular e Bioinformática, Unidade de Pesquisa Experimental, Faculdade de Medicina, Universidade Estadual Paulista, Botucatu, SP, Brasil
- Departamento de Patologia, Faculdade de Medicina, Universidade Estadual Paulista, Botucatu, SP, Brasil
| | - P E M Ribolla
- Instituto de Biotecnologia, Universidade Estadual Paulista, Botucatu, SP, Brasil
- Instituto de Biociências, Departamento de Bioestatística, Biologia Vegetal, Parasitologia e Zoologia, Universidade Estadual Paulista, Botucatu, SP, Brasil
| | - A M Ab'Saber
- Laboratório de Histomorfometria e Genômica Pulmonar, Departamento de Patologia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
| | - T Takagaki
- Divisão de Pneumologia, Instituto do Coração, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
| | - M A Nagai
- Departamento de Radiologia e Oncologia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
- Laboratório de Genética Molecular, Centro de Pesquisa Translacional em Oncologia, Instituto do Câncer de São Paulo, São Paulo, SP, Brasil
| | - V L Capelozzi
- Laboratório de Histomorfometria e Genômica Pulmonar, Departamento de Patologia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
| |
Collapse
|
403
|
Fabrizio FP, Sparaneo A, Muscarella LA. Monitoring EGFR-lung cancer evolution: a possible beginning of a "methylation era" in TKI resistance prediction. Front Oncol 2023; 13:1137384. [PMID: 37152062 PMCID: PMC10157092 DOI: 10.3389/fonc.2023.1137384] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 04/06/2023] [Indexed: 05/09/2023] Open
Abstract
The advances in scientific knowledge on biological therapies of the last two decades have impressively oriented the clinical management of non-small-cell lung cancer (NSCLC) patients. The treatment with tyrosine kinase inhibitors (TKIs) in patients harboring Epidermal Growth Factor Receptor (EGFR)-activating mutations is dramatically associated with an improvement in disease control. Anyhow, the prognosis for this selected group of patients remains unfavorable, due to the innate and/or acquired resistance to biological therapies. The methylome analysis of many tumors revealed multiple patterns of methylation at single/multiple cytosine-phosphate-guanine (CpG) sites that are linked to the modulation of several cellular pathways involved in cancer onset and progression. In lung cancer patients, ever increasing evidences also suggest that the association between DNA methylation changes at promoter/intergenic regions and the consequent alteration of gene-expression signatures could be related to the acquisition of resistance to biological therapies. Despite this intriguing hypothesis, large confirmatory studies are demanded to consolidate and finalize many preliminary observations made in this field. In this review, we will summarize the available knowledge about the dynamic role of DNA methylation in EGFR-mutated NSCLC patients.
Collapse
|
404
|
Fernández Aceñero MJ, Díaz Del Arco C, Dinarés C, Labiano T, Tejerina E, Bernabé MJ, Forcen E, Saiz-Pardo M, Pérez P, Lozano MD. Overview and update on molecular testing in non-small cell lung carcinoma utilizing endobronchial ultrasound-guided transbronchial needle aspiration (EBUS-TBNA) samples. Diagn Cytopathol 2023; 51:26-35. [PMID: 35899869 DOI: 10.1002/dc.25019] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/12/2022] [Accepted: 07/18/2022] [Indexed: 12/13/2022]
Abstract
Lung carcinoma remains one of the most frequent and aggressive human neoplasms. Fortunately, in the last decades, the increasing knowledge of the molecular mechanisms leading to cancer development has allowed the use of targeted therapies with improvement of prognosis in many patients. Clinical management has also changed after the introduction of endobronchialultrasonographic bronchoscopy that allows a conservative staging of lung tumors, avoiding the need of mediastinoscopy for lymph node staging. Lung pathologists and cytopathologists are facing the challenge of giving the more comprehensive prognostic and predictive information with ever smaller tissue or cytological samples. The aim of this review is to summarize the molecular testing for non-small cell lung carcinoma and how pathologists can contribute to the patient's outcome with a conscious management of biological samples.
Collapse
Affiliation(s)
| | | | - Carme Dinarés
- Department of Surgical Pathology, Valld'Hebron, Barcelona, Spain
| | - Tania Labiano
- Department of Surgical Pathology, Clínica Universitaria of Navarra, Pamplona, Spain
| | - Eva Tejerina
- Department of Surgical Pathology, Clínica Puerta de Hierro, Madrid, Spain
| | - Mª José Bernabé
- Department of Pneumology, Hospital Clínico San Carlos, Madrid, Spain
| | - Elena Forcen
- Department of Pneumology, Hospital Clínico San Carlos, Madrid, Spain
| | - Melchor Saiz-Pardo
- Departments of Surgical Pathology, Hospital Clínico San Carlos, Madrid, Spain
| | - Pablo Pérez
- Departments of Surgical Pathology, Hospital Clínico San Carlos, Madrid, Spain
| | - Maria D Lozano
- Department of Surgical Pathology, Clínica Universitaria of Navarra, Pamplona, Spain
| |
Collapse
|
405
|
Skopelidou V, Strakoš J, Škarda J, Raška M, Kafková-Rašková L. Potential predictors of immunotherapy in small cell lung cancer. Pathol Oncol Res 2023; 29:1611086. [PMID: 37206058 PMCID: PMC10191143 DOI: 10.3389/pore.2023.1611086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Accepted: 04/18/2023] [Indexed: 05/21/2023]
Abstract
Lung cancer is one of the leading causes of cancer-related deaths worldwide, with small cell lung cancer (SCLC) having the worst prognosis. SCLC is diagnosed late in the disease's progression, limiting treatment options. The most common treatment for SCLC is chemotherapy. As the disease progresses, immunotherapy, most commonly checkpoint inhibitor medication, becomes more important. Efforts should be made in the development of immunotherapy to map specific biomarkers, which play a role in properly assigning a type of immunotherapy to the right cohort of patients, where the benefits outweigh any risks or adverse effects. The objective of this review was to provide a thorough assessment of current knowledge about the nature of the tumor process and treatment options for small cell lung cancer, with a focus on predictive biomarkers. According to the information obtained, the greatest potential, which has already been directly demonstrated in some studies, has characteristics such as tumor microenvironment composition, tumor mutation burden, and molecular subtyping of SCLC. Several other aspects appear to be promising, but more research, particularly prospective studies on a larger number of probands, is required. However, it is clear that this field of study will continue to expand, as developing a reliable method to predict immunotherapy response is a very appealing goal of current medicine and research in the field of targeted cancer therapy.
Collapse
Affiliation(s)
- Valeria Skopelidou
- Institute of Molecular and Clinical Pathology and Medical Genetics, University Hospital Ostrava, Ostrava, Czechia
- Institute of Molecular and Clinical Pathology and Medical Genetics, Faculty of Medicine, University of Ostrava, Ostrava, Czechia
- *Correspondence: Valeria Skopelidou,
| | - Jan Strakoš
- Institute of Molecular and Clinical Pathology and Medical Genetics, University Hospital Ostrava, Ostrava, Czechia
- Institute of Molecular and Clinical Pathology and Medical Genetics, Faculty of Medicine, University of Ostrava, Ostrava, Czechia
| | - Jozef Škarda
- Institute of Molecular and Clinical Pathology and Medical Genetics, University Hospital Ostrava, Ostrava, Czechia
- Institute of Molecular and Clinical Pathology and Medical Genetics, Faculty of Medicine, University of Ostrava, Ostrava, Czechia
- Department of Clinical and Molecular Pathology, Faculty of Medicine and Dentistry, Palacký University Olomouc, Olomouc, Czechia
| | - Milan Raška
- Department of Immunology, Faculty of Medicine and Dentistry, Palacký University Olomouc, Olomouc, Czechia
- Department of Immunology, University Hospital Olomouc, Olomouc, Czechia
| | - Leona Kafková-Rašková
- Department of Immunology, Faculty of Medicine and Dentistry, Palacký University Olomouc, Olomouc, Czechia
- Department of Immunology, University Hospital Olomouc, Olomouc, Czechia
| |
Collapse
|
406
|
Abstract
Treatment of non-small-cell lung cancer depends heavily on the cancer stage, and immunotherapy can play a major role at any stage. For locally advanced stages, the addition of an immune checkpoint inhibitor (ICI) to neoadjuvant chemotherapy improves pathological response and event-free survival. In the adjuvant setting, adding ICI, after adjuvant chemotherapy for resectable cancer, increases the disease-free survival. In unresectable stage III treated with concomitant chemotherapy and radiotherapy, adding ICI as a maintenance therapy increases progression-free survival and overall survival. In the metastatic setting, the addition of ICI to chemotherapy improves overall survival, progression-free survival, and response rates irrespective of the PD-L1 expression. ICI on its own may be considered in cases of PD-L1 expression equal or greater than at least 50%.
Collapse
Affiliation(s)
- Geoffrey Merle
- Oncology Department, University Hospital, Geneva, Switzerland
| | - Alfredo Addeo
- Oncology Department, University Hospital, Geneva, Switzerland
| |
Collapse
|
407
|
Zhang H, Chen L, Gu X, Zhang M, Qin Y, Yao F, Wang Z, Gu Y, Yang GZ. Trustworthy learning with (un)sure annotation for lung nodule diagnosis with CT. Med Image Anal 2023; 83:102627. [PMID: 36283199 DOI: 10.1016/j.media.2022.102627] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 07/22/2022] [Accepted: 09/10/2022] [Indexed: 02/04/2023]
Abstract
Recent evolution in deep learning has proven its value for CT-based lung nodule classification. Most current techniques are intrinsically black-box systems, suffering from two generalizability issues in clinical practice. First, benign-malignant discrimination is often assessed by human observers without pathologic diagnoses at the nodule level. We termed these data as "unsure-annotation data". Second, a classifier does not necessarily acquire reliable nodule features for stable learning and robust prediction with patch-level labels during learning. In this study, we construct a sure-annotation dataset with pathologically-confirmed labels and propose a collaborative learning framework to facilitate sure nodule classification by integrating unsure-annotation data knowledge through nodule segmentation and malignancy score regression. A loss function is designed to learn reliable features by introducing interpretability constraints regulated with nodule segmentation maps. Furthermore, based on model inference results that reflect the understanding from both machine and experts, we explore a new nodule analysis method for similar historical nodule retrieval and interpretable diagnosis. Detailed experimental results demonstrate that our approach is beneficial for achieving improved performance coupled with trustworthy model reasoning for lung cancer prediction with limited data. Extensive cross-evaluation results further illustrate the effect of unsure-annotation data for deep-learning based methods in lung nodule classification.
Collapse
Affiliation(s)
- Hanxiao Zhang
- Institute of Medical Robotics, Shanghai Jiao Tong University, Shanghai, China
| | - Liang Chen
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xiao Gu
- Imperial College London, London, UK
| | - Minghui Zhang
- Institute of Medical Robotics, Shanghai Jiao Tong University, Shanghai, China
| | | | - Feng Yao
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Zhexin Wang
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China.
| | - Yun Gu
- Institute of Medical Robotics, Shanghai Jiao Tong University, Shanghai, China; Shanghai Center for Brain Science and Brain-Inspired Technology, Shanghai, China.
| | - Guang-Zhong Yang
- Institute of Medical Robotics, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
408
|
Wei X, Wei Z, Zheng G, Xie T, Huo Z, Huang Y, Chen X, Bai J. Prognostic significance of circulating Epstein-Barr virus DNA in pulmonary lymphoepithelioma-like carcinoma: A meta-analysis and validation study. J Med Virol 2023; 95:e28349. [PMID: 36428250 DOI: 10.1002/jmv.28349] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 11/05/2022] [Accepted: 11/20/2022] [Indexed: 11/27/2022]
Abstract
In small-scale studies, circulating Epstein-Barr virus (EBV) DNA levels have prognostic value in patients with pulmonary lymphoepithelioma-like carcinoma (LELC). Therefore, we performed a comprehensive meta-analysis to evaluate the prognostic significance of circulating EBV DNA levels in patients with pulmonary LELC. Studies that discussed the prognostic significance of circulating EBV DNA detection in pulmonary LELC were eligible for inclusion in this study. The overall survival (OS) and progression-free survival (PFS) were the primary outcomes. Pooled hazard ratio (HR), 95% confidence intervals (CIs), and p value were calculated to estimate the prognostic significance of EBV DNA levels. Additionally, we conducted a further observation using an independent cohort. The pooled HR and 95% CI of pretreatment EBV DNA levels for OS and PFS were 3.63 (95% CI: 2.90-4.55) and 2.88 (95% CI: 1.90-4.38), respectively. The pooled HR and 95% CI for Posttreatment EBV DNA levels for OS and PFS were 3.77 (95% CI: 2.96-4.80) and 3.52 (95% CI: 1.91-6.51, p < 0.001), respectively. The independent cohort showed similar results that patients with high pretreatment EBV DNA or positive posttreatment EBV DNA had significantly inferior PFS. Circulating EBV DNA levels provide prognostic values of survival and treatment response in pulmonary LELC patients.
Collapse
Affiliation(s)
- Xinyan Wei
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Zuyou Wei
- Department of Respiratory and Critical Care Medicine, Guangxi Academy of Medical Sciences, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Guixian Zheng
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Ting Xie
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Zengyu Huo
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Yanbing Huang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Xiaoli Chen
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Jing Bai
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
409
|
Yasin AKA, Mohamed A, Mohamed A, Elamin N, Al-Tikrity MA, Wazwaz B. Pulmonary Enteric Adenocarcinoma: A Very Rare Case Report from Qatar. Case Rep Oncol 2023; 16:759-764. [PMID: 37933317 PMCID: PMC10625813 DOI: 10.1159/000533220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 07/20/2023] [Indexed: 11/08/2023] Open
Abstract
A 78-year-old male patient presented with dyspnea, loss of appetite, and weight loss. Workup and imaging showed suspected malignant lung lesion. Biopsy was done and showed features of pulmonary enteric adenocarcinoma (PEAC). This is a very rare disease and its diagnosis is challenging.
Collapse
Affiliation(s)
- Ahmed K A Yasin
- Department of Internal Medicine, Hamad General Hospital, Hamad Medical Corporation, Doha, Qatar
| | - Abdelaziz Mohamed
- Department of Internal Medicine, Hamad General Hospital, Hamad Medical Corporation, Doha, Qatar
| | - Anas Mohamed
- Department of Internal Medicine, Hamad General Hospital, Hamad Medical Corporation, Doha, Qatar
| | - Nusiba Elamin
- Department of Internal Medicine, Hamad General Hospital, Hamad Medical Corporation, Doha, Qatar
| | - Mustafa A Al-Tikrity
- Department of Pulmonary Medicine, Hamad General Hospital, Hamad Medical Corporation, Doha, Qatar
| | - Bara Wazwaz
- Department of Pathology, Hamad General Hospital, Hamad Medical Corporation, Doha, Qatar
| |
Collapse
|
410
|
Kishikawa S, Hayashi T, Takamochi K, Takekawa S, Sasahara N, Handa T, Saito T, Suzuki K, Yao T. Comprehensive clinicopathological characteristics and mucin core protein expression profiles of bronchiolar adenoma. Histopathology 2023; 82:264-275. [PMID: 36180978 DOI: 10.1111/his.14806] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 09/07/2022] [Accepted: 09/25/2022] [Indexed: 12/13/2022]
Abstract
AIMS Bronchiolar adenoma (BA) is a novel entity in the 2021 WHO classification of lung tumours. The expression profile of mucin core proteins in BAs is not clear. The aim of this study was to clarify the expression profiles of mucins and to validate the clinicopathologic and molecular features of BAs. METHODS AND RESULTS We examined the clinicopathological, immunohistochemical, and molecular features of 20 BAs. Our cohort comprised 10 proximal and 10 distal BAs. Only seven of 18 patients (39%) were accurately diagnosed with BA at the time of intraoperative consultation. The frequent genetic alterations were BRAF V600E (35%) and KRAS (30%) mutations, which were mutually exclusive. The expression of MUC1, MUC4, and MUC5B was observed in all cases and that of MUC5AC and MUC6 was observed in nine (45%) and five (25%) cases, respectively. MUC4 was diffusely expressed in 18 cases. In contrast, MUC1, MUC5AC, MUC5B, and MUC6 displayed a patchy expression pattern. These expression patterns were similar to that of bronchiolar epithelium in normal lung tissue. In addition, overexpression of MUC1 and MUC4 on the entire cell surface was not observed in any of the BAs, suggesting their benign nature. CONCLUSION BA commonly exhibits diffuse MUC4 and patchy MUC1 and MUC5B expression. Its unique expression pattern is probably attributed to mucin expression specific to the bronchiolar epithelium. These results confirm the clinicopathologic and molecular characteristics of BA, including the difficulty in intraoperative frozen section diagnosis and the broad morphologic spectrum of BA derived from the bronchiolar epithelium.
Collapse
Affiliation(s)
- Satsuki Kishikawa
- Department of Human Pathology, Graduate School of Medicine, Juntendo University, Bunkyo-ku, Tokyo, Japan
| | - Takuo Hayashi
- Department of Human Pathology, Graduate School of Medicine, Juntendo University, Bunkyo-ku, Tokyo, Japan
| | - Kazuya Takamochi
- Department of General Thor, acic Surgery, Graduate School of Medicine, Juntendo University, Bunkyo-ku, Tokyo, Japan
| | - Shiori Takekawa
- Department of General Thor, acic Surgery, Graduate School of Medicine, Juntendo University, Bunkyo-ku, Tokyo, Japan
| | - Noriko Sasahara
- Department of Human Pathology, Graduate School of Medicine, Juntendo University, Bunkyo-ku, Tokyo, Japan
| | - Takafumi Handa
- Department of Human Pathology, Graduate School of Medicine, Juntendo University, Bunkyo-ku, Tokyo, Japan
| | - Tsuyoshi Saito
- Department of Human Pathology, Graduate School of Medicine, Juntendo University, Bunkyo-ku, Tokyo, Japan
| | - Kenji Suzuki
- Department of General Thor, acic Surgery, Graduate School of Medicine, Juntendo University, Bunkyo-ku, Tokyo, Japan
| | - Takashi Yao
- Department of Human Pathology, Graduate School of Medicine, Juntendo University, Bunkyo-ku, Tokyo, Japan
| | | |
Collapse
|
411
|
Prognostic significance of examined lymph node count in older patients with stage T1N0M0 pulmonary carcinoid tumours: a population-based propensity score-matched analysis. Aging Clin Exp Res 2023; 35:203-212. [PMID: 36319940 DOI: 10.1007/s40520-022-02293-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 10/20/2022] [Indexed: 01/07/2023]
Abstract
BACKGROUND The optimal number of examined lymph nodes (ELNs) for older early-stage pulmonary carcinoid tumour (PC) patients is unknown. AIMS To explore the prognostic effect of the ELN count in older patients with stage T1N0M0 PC resection. METHODS Clinical data from the Surveillance, Epidemiology, and End Results database on stage T1N0M0 PC older patients (age ≥ 65 years) who underwent sublobar resection and lobectomy between 2000 and 2018 were retrospectively analysed for two ELNs-stratified (≥ 7 vs. < 7 ELNs) propensity score-matched (PSM) groups. Overall survival (OS) was calculated and compared with Kaplan-Meier analysis and log-rank test, respectively, and the independent prognostic factors were estimated using a Cox proportional hazard model. RESULTS Among 1077 participants (median dissected LN 4; mean ELNs 6.19 ± 7.04), 393 (36.49%) in the ≥ 7 ELNs group had better 5- and 10-year OS than the < 7 ELNs group (before PSM: 5-year OS = 93.01 vs. 85.22%, 10-year OS = 72.38 vs. 58.99%, p < 0.001; after PSM: 5-year OS = 93.12 vs. 85.97%, 10-year OS = 75.25 vs. 60.03%, p = 0.001). Subgroup analysis stratified by histologic type and surgical method showed a similar survival trend. Age-stratified analysis showed that, compared with the < 7 ELNs group, the ≥ 7 ELNs group had better 5- and 10-year OS but only better 5-year OS in participants > 75 years. Compared with the < 7 ELNs group, subgroup analysis by tumour size showed superior OS with 1.1-3.0 cm tumours in the ≥ 7 ELNs group, but no significant intergroup difference with tumours < 1.0 cm. Multivariate Cox analysis showed ≥ 7 ELNs was associated with improved OS. CONCLUSION Higher ELNs correlated with increased long-term survival in older early‑stage PC patients, and a minimum of 7 ELNs are recommended for prognostication in these patients (especially those aged 65-75 years, with tumour size 1.1-3.0 cm).
Collapse
|
412
|
Zhang Y, Zhang Y, Hu Y, Zhang S, Zhu M, Hu B, Guo X, Lu J, Zhang Y. Validation of the novel International Association for the Study of Lung Cancer grading system and prognostic value of filigree micropapillary and discohesive growth pattern in invasive pulmonary adenocarcinoma. Lung Cancer 2023; 175:79-87. [PMID: 36481678 DOI: 10.1016/j.lungcan.2022.11.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 11/18/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022]
Abstract
INTRODUCTION The Pathology Committee of the International Association for the Study of Lung Cancer (IASLC) proposed a new histological grading system based on the combination of predominant and high-grade patterns in 2020. MATERIALS AND METHODS Pathological sections from 631 patients with stage I-III invasive lung adenocarcinoma were reviewed. We then determined the histological grade according to the new grading system and confirmed the pathological features that included the filigree micropapillary and discohesive growth pattern. Applying of the novel IASLC grading system in prognosis stratification was verified and the clinical significance of the pathological characteristics was explored. RESULTS Cox multivariable analysis revealed that in the stage I-III invasive lung adenocarcinoma, the IASLC grading system was significantly associated with disease-free survival (DFS) [hazard ratio (HR) = 1.419; 95 % confidence interval (CI): 1.040-1.937; P = 0.027] and overall survival (OS) (HR = 1.899; 95 % CI: 1.168-3.087; P = 0.010). In patients with IASLC Grades 1 and 2, the simultaneous presence of filigree micropapillary and discohesive growth pattern was significantly correlated with DFS (HR = 1.899; 95 % CI:1.168-3.087; P = 0.010). However, the filigree micropapillary and discohesive growth pattern did not affect the OS (HR = 2.786; P = 0.317). The competitive risk model revealed that in the stage I cohort, the simultaneous presence of filigree micropapillary and discohesive growth pattern was a risk factor for recurrence and metastasis [sub- distribution HR (SHR) = 1.987; 95 %CI: 1.122-3.518; P = 0.019]. CONCLUSION Our study verified that the new prognostic stratification system was an effective stratification tool. Filigree micropapillary and discohesive growth pattern may also be risk factors for DFS, postoperative recurrence and metastasis.
Collapse
Affiliation(s)
- Yuan Zhang
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao Yang Hospital, Capital Medical University, Beijing, China
| | - Yanjun Zhang
- Department of Pathology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Yi Hu
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao Yang Hospital, Capital Medical University, Beijing, China
| | - Shu Zhang
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao Yang Hospital, Capital Medical University, Beijing, China
| | - Min Zhu
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao Yang Hospital, Capital Medical University, Beijing, China
| | - Bin Hu
- Department of Thoracic Surgery, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Xiaojuan Guo
- Department of Radiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Jun Lu
- Department of Pathology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China.
| | - Yuhui Zhang
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao Yang Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
413
|
Zheng Y, Han X, Wu Y, Jia X, Zhang K, Fan J, Shi H. Prognostic Factors for Survival in Multiple Primary Lung Adenocarcinomas: A Retrospective Analysis of 283 Patients. Technol Cancer Res Treat 2023; 22:15330338231185278. [PMID: 37365877 DOI: 10.1177/15330338231185278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2023] Open
Abstract
Purpose: In recent years, a rising number of multiple primary lung cancers have been detected with the advancement of imaging technology. No detailed study has assessed the prognosis of multiple primary lung adenocarcinomas based on computed tomography characteristics. The present study aimed to analyze outcomes and determine valuable factors for predicting the prognosis of multiple primary lung adenocarcinoma. Methods: This single-center retrospective study was performed from January 2013 to October 2021. All patients were divided into 3 groups based on tumor density as follows: multi-pure ground-glass nodules, at least one part-solid nodule without solid nodules, and at least one solid nodule. Clinicopathologic features, computed tomography signs, and survival outcomes were compared between these groups. The Kaplan-Meier method was used for survival analysis. The multivariable Cox proportional hazards regression model was used to identify independent predictors for recurrence-free survival and overall survival. Results: The sample included 283 patients with 623 lesions who met the inclusion criteria for multiple primary lung adenocarcinoma. Of these patients, 71 (25.1%) presented with multi-pure ground-glass nodules, 100 (35.3%) with at least one part-solid nodule without solid nodule, and 112 (39.6%) with at least one solid nodule. The 3 groups had distinguished clinicopathologic and radiological features of age, adjuvant therapy, types of tumor resection, TNM stage, pathological subtypes, pleural indentation, spicule, and vacuole (all P < .001). Multivariate analysis found that lesion number was an independent predictor for both recurrence-free survival (hazard ratio 2.41; 95% confidence interval 1.12-5.19; P = .025) and overall survival (hazard ratio 4.78; 95% confidence interval 1.88-12.18; P = .001), and the at least one solid nodule was an independent predictor for overall survival (hazard ratio 5.307; 95% confidence interval 1.16-24.31; P = .032). Stage III (hazard ratio 5.71; 95% confidence interval 1.94-16.81; P = .002) and adjuvant therapy (hazard ratio 2.52; 95% confidence interval 1.24-5.13; P = .011) influenced the recurrence-free survival. Conclusions: Survival of multiple primary lung adenocarcinoma patients is strongly correlated with the lesion number and the at least one solid nodule tumors in radiological. This information may be useful for predicting survival and making clinical decisions in future studies.
Collapse
Affiliation(s)
- Yuting Zheng
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, People's Republic of China
| | - Xiaoyu Han
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, People's Republic of China
| | - Ying Wu
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Xi Jia
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, People's Republic of China
| | - Kailu Zhang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, People's Republic of China
| | - Jun Fan
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Heshui Shi
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, People's Republic of China
| |
Collapse
|
414
|
Chang CY, Huang YC, Chiang HH, Wu YY, Wu KL, Chang YY, Liu LX, Tsai YM, Hsu YL. Ladinin 1 Shortens Survival via Promoting Proliferation and Enhancing Invasiveness in Lung Adenocarcinoma. Int J Mol Sci 2022; 24:431. [PMID: 36613882 PMCID: PMC9820746 DOI: 10.3390/ijms24010431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/13/2022] [Accepted: 12/13/2022] [Indexed: 12/29/2022] Open
Abstract
Lung cancer is one of the deadliest cancers worldwide, including in Taiwan. The poor prognosis of the advanced lung cancer lies in delayed diagnosis and non-druggable targets. It is worth paying more attention to these ongoing issues. Public databases and an in-house cohort were used for validation. The KM plotter was utilized to discover the clinical significance. GSEA and GSVA were adopted for a functional pathway survey. Molecular biological methods, including proliferation, migration, and the EMT methods, were used for verification. Based on public databases, the increased expression of Ladinin 1 (LAD1) was presented in tumor and metastatic sites. Furthermore, an in-house cohort revealed a higher intensity of LAD1 in tumor rather than in normal parts. The greater the expression of LAD1 was, the shorter the duration of lung adenocarcinoma (LUAD) patient survival. Moreover, the association of B3GNT3 with LAD1 affected the survival of LUAD patients. Functional analyses using GSEA and GSVA revealed the associations with survival, migration, invasion, and EMT. Biologic functions supported the roles of LAD1 in proliferation via the cell cycle and migration in EMT. This study reveals that LAD1 plays a major role in regulating proliferation and migration in lung cancer and impacts survival in LUAD. It is worth investing in further studies and in the development of drugs targeting LAD1.
Collapse
Affiliation(s)
- Chao-Yuan Chang
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Anatomy, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Yung-Chi Huang
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Hung-Hsing Chiang
- Division of Thoracic Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Yu-Yuan Wu
- School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Kuan-Li Wu
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Division of Pulmonary and Critical Care Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Yung-Yun Chang
- Division of Pulmonary and Critical Care Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Division of General Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Lian-Xiu Liu
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Ying-Ming Tsai
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Division of Pulmonary and Critical Care Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Ya-Ling Hsu
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| |
Collapse
|
415
|
Prieto TG, Baldavira CM, Machado-Rugolo J, Olivieri EHR, da Silva ECA, Ab’ Saber AM, Takagaki TY, Capelozzi VL. Proposing Specific Neuronal Epithelial-to-Mesenchymal Transition Genes as an Ancillary Tool for Differential Diagnosis among Pulmonary Neuroendocrine Neoplasms. Genes (Basel) 2022; 13:genes13122309. [PMID: 36553576 PMCID: PMC9777553 DOI: 10.3390/genes13122309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 11/25/2022] [Accepted: 11/29/2022] [Indexed: 12/13/2022] Open
Abstract
Pulmonary neuroendocrine neoplasms (PNENs) are currently classified into four major histotypes, including typical carcinoid (TC), atypical carcinoid (AC), large cell neuroendocrine carcinoma (LCNEC), and small cell lung carcinoma (SCLC). This classification was designed to be applied to surgical specimens mostly anchored in morphological parameters, resulting in considerable overlapping among PNENs, which may result in important challenges for clinicians' decisions in the case of small biopsies. Since PNENs originate from the neuroectodermic cells, epithelial-to-mesenchymal transition (EMT) gene expression shows promise as biomarkers involved in the genotypic transformation of neuroectodermic cells, including mutation burden with the involvement of chromatin remodeling genes, apoptosis, and mitosis rate, leading to modification in final cellular phenotype. In this situation, additional markers also applicable to biopsy specimens, which correlate PNENs subtypes with systemic treatment response, are much needed, and current potential candidates are neurogenic EMT genes. This study investigated EMT genes expression and its association with PNENs histotypes in tumor tissues from 24 patients with PNENs. PCR Array System for 84 EMT-related genes selected 15 differentially expressed genes among the PNENs, allowing to discriminate TC from AC, LCNEC from AC, and SCLC from AC. Functional enrichment analysis of the EMT genes differentially expressed among PNENs subtypes showed that they are involved in cellular proliferation, extracellular matrix degradation, regulation of cell apoptosis, oncogenesis, and tumor cell invasion. Interestingly, four EMT genes (MAP1B, SNAI2, MMP2, WNT5A) are also involved in neurological diseases, in brain metastasis, and interact with platinum-based chemotherapy and tyrosine-kinase inhibitors. Collectively, these findings emerge as an important ancillary tool to improve the strategies of histologic diagnosis in PNENs and unveil the four EMT genes that can play an important role in driving chemical response in PNENs.
Collapse
Affiliation(s)
- Tabatha Gutierrez Prieto
- Laboratory of Genomics and Histomorphometry, Department of Pathology, University of São Paulo Medical School (USP), São Paulo 01246-903, SP, Brazil
| | - Camila Machado Baldavira
- Laboratory of Genomics and Histomorphometry, Department of Pathology, University of São Paulo Medical School (USP), São Paulo 01246-903, SP, Brazil
| | - Juliana Machado-Rugolo
- Laboratory of Genomics and Histomorphometry, Department of Pathology, University of São Paulo Medical School (USP), São Paulo 01246-903, SP, Brazil
- Health Technology Assessment Center (NATS), Clinical Hospital (HCFMB), Medical School of São Paulo State University (UNESP), Botucatu 18618-970, SP, Brazil
| | | | | | - Alexandre Muxfeldt Ab’ Saber
- Laboratory of Genomics and Histomorphometry, Department of Pathology, University of São Paulo Medical School (USP), São Paulo 01246-903, SP, Brazil
- Fundação Oncocentro do Estado de São Paulo (FOSP), São Paulo 05409-012, SP, Brazil
| | - Teresa Yae Takagaki
- Division of Pneumology, Instituto do Coração (Incor), Medical School of University of São Paulo, São Paulo 01246-903, SP, Brazil
| | - Vera Luiza Capelozzi
- Laboratory of Genomics and Histomorphometry, Department of Pathology, University of São Paulo Medical School (USP), São Paulo 01246-903, SP, Brazil
- Correspondence:
| |
Collapse
|
416
|
Wu Y, Yan Z, Pan J, Chang X, Huang B, Luo D, Meng R, Shi H, Fan J, Nie X. Partial response to pralsetinib in an advanced pulmonary sarcomatoid carcinoma patient harboring a KIF5B-RET rearrangement: a case report. World J Surg Oncol 2022; 20:386. [PMID: 36471407 PMCID: PMC9724403 DOI: 10.1186/s12957-022-02848-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 11/20/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Pulmonary sarcomatoid carcinoma (PSC) is a rare and unconventional non-small-cell lung cancer (NSCLC) that appears to be aggressive, with a poor prognosis and response to conventional treatment. Approximately 30% of PSCs have potentially targetable genomic alterations, but few studies have involved RET gene fusions, and corresponding targeted therapies are lacking. CASE PRESENTATION In this report, we describe a patient with PSC harboring a KIF5B-RET gene fusion who was initially diagnosed with stage IVb lung cancer. Due to the poor performance status, the patient was unable to tolerate any radiotherapy or chemotherapy. Based on the next-generation sequencing (NGS) result of RET gene fusion, the patient was treated with pralsetinib. Two months after the treatment, the patient achieved a partial response. CONCLUSIONS Our case indicates that RET is one of the main driver oncogenes of PSC and provides useful information for precise RET inhibitor administration in the future. Thus, the use of comprehensive genomic profiling may provide important treatment options for PSC.
Collapse
Affiliation(s)
- Ying Wu
- grid.33199.310000 0004 0368 7223Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 Hubei China
| | - Zhecheng Yan
- grid.33199.310000 0004 0368 7223Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 Hubei China
| | - Juan Pan
- grid.33199.310000 0004 0368 7223Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 Hubei China
| | - Xiaona Chang
- grid.33199.310000 0004 0368 7223Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 Hubei China
| | - Bo Huang
- grid.33199.310000 0004 0368 7223Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 Hubei China
| | - Danju Luo
- grid.33199.310000 0004 0368 7223Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 Hubei China
| | - Rui Meng
- grid.33199.310000 0004 0368 7223Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Heshui Shi
- grid.33199.310000 0004 0368 7223Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 Hubei China
| | - Jun Fan
- grid.33199.310000 0004 0368 7223Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 Hubei China
| | - Xiu Nie
- grid.33199.310000 0004 0368 7223Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 Hubei China
| |
Collapse
|
417
|
Lung Cancer Clinical Trials with a Seamless Phase II/III Design: Systematic Review. J Clin Med 2022; 11:jcm11237176. [PMID: 36498749 PMCID: PMC9739886 DOI: 10.3390/jcm11237176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 11/29/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022] Open
Abstract
Current lung cancer clinical research focuses on biomarkers and personalized treatment strategies. Adaptive clinical trial designs have gained significant ground due to their increased flexibility, compared to the conventional model of drug development from phase I to phase IV trials. One such adaptive approach is the seamless phase II/III design, which has been used to reduce the total sample size and drug development time. In this context, an algorithmic systematic search was conducted in MEDLINE (PUBMED), SCOPUS, EMBASE and Cochrane Central Register of Controlled Trials until 31 June 2022 in order to identify lung cancer trials of systematic treatments that have employed the seamless phase II/III methodology and to describe their characteristics. The search strategy yielded a total of 1420 records that were screened through their title and abstract; 28 eligible trials were included in the systematic review. Based on the study endpoints, the most common subtype included phase II/III trials with inefficacy/futility analyses (61%; 17/28), followed by dose escalation phase II/III trials (18%; 5/28), one multi-arm multi stage trial and 5 trials with other design (18%). Most eligible trials were open-label (71%; 20/27), included patients with non-small cell lung cancer (82%; 23/28), evaluated targeted therapies and/or immunotherapies (82%; 23/28) and recruited patients with advanced disease (89.3%; 25/28). In conclusion, the seamless phase II/III design is a feasible and suitable approach in lung cancer research, with distinct design subcategories according to study endpoints.
Collapse
|
418
|
Qin Y, Li F, Tan Y, Duan Q, Zhang Q. Case report: Dramatic response to alectinib in a lung adenosquamous carcinoma patient harbouring a novel CPE-ALK fusion. Front Oncol 2022; 12:998545. [PMID: 37082099 PMCID: PMC10111186 DOI: 10.3389/fonc.2022.998545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 11/18/2022] [Indexed: 12/03/2022] Open
Abstract
Lung Adenosquamous carcinoma (ASC) is a rare histological subtype of lung cancer accounting for 0.4%–4% of all lung cancers. ASC is generally considered to be an aggressive cancer with poor prognosis. There is no specific standard treatment for ASC, and current treatment of ASC is relied on the guideline for non-small cell lung cancer (NSCLC). To date, only sporadic canonical EML4-ALK fusions have been reported in ASC patients, and the efficiency of ALK-TKI is still unclear in non-canonical ALK fusion positive ASC patients. Here we describe the case of a stage IV ASC patient harboring a novel CPE-ALK fusion detected via 74 genes panel analysis. Interestingly, the TP53 was wild-type and no another somatic mutation was found within 74 genes. In addition, immunohistochemical staining (IHC) also supports an oncogenic role for the CPE-ALK fusion. Based on these findings, the patient received alectinib 600 mg twice daily. After 4 months on treatment the patients achieved a radiological partial response (PR) and his symptoms were significantly relieved. Imaging showed that lesions of the patient were reduced, and the clinical evaluation was partial response (PR). To the best of our knowledge, this is the first report of a dramatic tumor response to alectinib in a patient with ASC harboring a CPE-ALK fusion. In addition, targeted NGS analysis may improve detection of ALK fusion in routine practice.
Collapse
Affiliation(s)
- Yanyan Qin
- Department of Respiratory and Critical Care Medicine, Shanxi Provincial People’s Hospital, Shanxi, China
- *Correspondence: Yanyan Qin,
| | - Fei Li
- Department of Respiratory and Critical Care Medicine, Shanxi Provincial People’s Hospital, Shanxi, China
| | - Yuan Tan
- The Medical Department, Jiangsu Simcere Diagnostics Co., Ltd, Nanjing, China
- Nanjing Simcere Medical Laboratory Science Co., Ltd, Nanjing, China
- The State Key Lab of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Diagnostics Co., Ltd, Nanjing, China
| | - Qianqian Duan
- The Medical Department, Jiangsu Simcere Diagnostics Co., Ltd, Nanjing, China
- Nanjing Simcere Medical Laboratory Science Co., Ltd, Nanjing, China
- The State Key Lab of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Diagnostics Co., Ltd, Nanjing, China
| | - Qin Zhang
- The Medical Department, Jiangsu Simcere Diagnostics Co., Ltd, Nanjing, China
- Nanjing Simcere Medical Laboratory Science Co., Ltd, Nanjing, China
- The State Key Lab of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Diagnostics Co., Ltd, Nanjing, China
| |
Collapse
|
419
|
Park H, Tseng SC, Sholl LM, Hatabu H, Awad MM, Nishino M. Molecular Characterization and Therapeutic Approaches to Small Cell Lung Cancer: Imaging Implications. Radiology 2022; 305:512-525. [PMID: 36283111 PMCID: PMC9713457 DOI: 10.1148/radiol.220585] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 06/10/2022] [Accepted: 06/14/2022] [Indexed: 01/16/2023]
Abstract
Small cell lung cancer (SCLC) is a highly aggressive malignancy with exceptionally poor prognosis, comprising approximately 15% of lung cancers. Emerging knowledge of the molecular and genomic landscape of SCLC and recent successful clinical applications of new systemic agents have allowed for precision oncology treatment approaches. Imaging is essential for the diagnosis, staging, and treatment monitoring of patients with SCLC. The role of imaging is increasing with the approval of new treatment agents, including immune checkpoint inhibitors, which lead to novel imaging manifestations of response and toxicities. The purpose of this state-of-the-art review is to provide the reader with the latest information about SCLC, focusing on the subtyping of this malignancy (molecular characterization) and the emerging systemic therapeutic approaches and their implications for imaging. The review will also discuss the future directions of SCLC imaging, radiomics and machine learning.
Collapse
Affiliation(s)
- Hyesun Park
- From the Departments of Radiology (H.P., S.C.T., H.H., M.N.),
Pathology (L.M.S.), Medical Oncology (M.M.A.), and Medicine (M.M.A.),
Dana-Farber Cancer Institute and Brigham and Women's Hospital, 450
Brookline Ave, Boston, MA 02215
| | | | - Lynette M. Sholl
- From the Departments of Radiology (H.P., S.C.T., H.H., M.N.),
Pathology (L.M.S.), Medical Oncology (M.M.A.), and Medicine (M.M.A.),
Dana-Farber Cancer Institute and Brigham and Women's Hospital, 450
Brookline Ave, Boston, MA 02215
| | - Hiroto Hatabu
- From the Departments of Radiology (H.P., S.C.T., H.H., M.N.),
Pathology (L.M.S.), Medical Oncology (M.M.A.), and Medicine (M.M.A.),
Dana-Farber Cancer Institute and Brigham and Women's Hospital, 450
Brookline Ave, Boston, MA 02215
| | - Mark M. Awad
- From the Departments of Radiology (H.P., S.C.T., H.H., M.N.),
Pathology (L.M.S.), Medical Oncology (M.M.A.), and Medicine (M.M.A.),
Dana-Farber Cancer Institute and Brigham and Women's Hospital, 450
Brookline Ave, Boston, MA 02215
| | - Mizuki Nishino
- From the Departments of Radiology (H.P., S.C.T., H.H., M.N.),
Pathology (L.M.S.), Medical Oncology (M.M.A.), and Medicine (M.M.A.),
Dana-Farber Cancer Institute and Brigham and Women's Hospital, 450
Brookline Ave, Boston, MA 02215
| |
Collapse
|
420
|
Chen K, Liu S, Lu C, Gu X. A prognostic and therapeutic hallmark developed by the integrated profile of basement membrane and immune infiltrative landscape in lung adenocarcinoma. Front Immunol 2022; 13:1058493. [PMID: 36532024 PMCID: PMC9748099 DOI: 10.3389/fimmu.2022.1058493] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 11/16/2022] [Indexed: 12/05/2022] Open
Abstract
Basement membranes (BMs) are specialised extracellular matrices that maintain cellular integrity and resist the breaching of carcinoma cells for metastases while regulating tumour immunity. The tumour immune microenvironment (TME) is essential for tumour growth and the response to and benefits from immunotherapy. In this study, the BM score and TME score were constructed based on the expression signatures of BM-related genes and the presence of immune cells in lung adenocarcinoma (LUAD), respectively. Subsequently, the BM-TME classifier was developed with the combination of BM score and TME score for accurate prognostic prediction. Further, Kaplan-Meier survival estimation, univariate Cox regression analysis and receiver operating characteristic curves were used to cross-validate and elucidate the prognostic prediction value of the BM-TME classifier in several cohorts. Findings from functional annotation analysis suggested that the potential molecular regulatory mechanisms of the BM-TME classifier were closely related to the cell cycle, mitosis and DNA replication pathways. Additionally, the guiding value of the treatment strategy of the BM-TME classifier for LUAD was determined. Future clinical disease management may benefit from the findings of our research.
Collapse
Affiliation(s)
- Kaijie Chen
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China,Shanghai Key Laboratory of Molecular Imaging, Zhoupu Hospital, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Shuang Liu
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China,Shanghai Key Laboratory of Molecular Imaging, Zhoupu Hospital, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Changlian Lu
- School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai, China,*Correspondence: Xuefeng Gu, ; Changlian Lu,
| | - Xuefeng Gu
- Shanghai Key Laboratory of Molecular Imaging, Zhoupu Hospital, Shanghai University of Medicine and Health Sciences, Shanghai, China,School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai, China,*Correspondence: Xuefeng Gu, ; Changlian Lu,
| |
Collapse
|
421
|
Delineating the dynamic evolution from preneoplasia to invasive lung adenocarcinoma by integrating single-cell RNA sequencing and spatial transcriptomics. Exp Mol Med 2022; 54:2060-2076. [PMID: 36434043 PMCID: PMC9722784 DOI: 10.1038/s12276-022-00896-9] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 09/18/2022] [Accepted: 10/05/2022] [Indexed: 11/27/2022] Open
Abstract
The cell ecology and spatial niche implicated in the dynamic and sequential process of lung adenocarcinoma (LUAD) from adenocarcinoma in situ (AIS) to minimally invasive adenocarcinoma (MIA) and subsequent invasive adenocarcinoma (IAC) have not yet been elucidated. Here, we performed an integrative analysis of single-cell RNA sequencing (scRNA-seq) and spatial transcriptomics (ST) to characterize the cell atlas of the invasion trajectory of LUAD. We found that the UBE2C + cancer cell subpopulation constantly increased during the invasive process of LUAD with remarkable elevation in IAC, and its spatial distribution was in the peripheral cancer region of the IAC, representing a more malignant phenotype. Furthermore, analysis of the TME cell type subpopulation showed a constant decrease in mast cells, monocytes, and lymphatic endothelial cells, which were implicated in the whole process of invasive LUAD, accompanied by an increase in NK cells and MALT B cells from AIS to MIA and an increase in Tregs and secretory B cells from MIA to IAC. Notably, for AIS, cancer cells, NK cells, and mast cells were colocalized in the cancer region; however, for IAC, Tregs colocalized with cancer cells. Finally, communication and interaction between cancer cells and TME cell-induced constitutive activation of TGF-β signaling were involved in the invasion of IAC. Therefore, our results reveal the specific cellular information and spatial architecture of cancer cells and TME subpopulations, as well as the cellular interaction between them, which will facilitate the identification and development of precision medicine in the invasive process of LUAD from AIS to IAC.
Collapse
|
422
|
Luo J, Ji C, Campisi A, Chen T, Weder W, Fang W. Surgical Outcomes of Video-Assisted versus Open Pneumonectomy for Lung Cancer: A Real-World Study. Cancers (Basel) 2022; 14:5683. [PMID: 36428775 PMCID: PMC9688192 DOI: 10.3390/cancers14225683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/06/2022] [Accepted: 11/15/2022] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND The safety, feasibility and potential benefits of Video-assisted thoracoscopic surgery (VATS) pneumonectomy remain to be investigated. METHODS Patients receiving VATS or Open pneumonectomy during the study period were included to compare surgical outcomes. Propensity-score matched (PSM) analysis was performed to eliminate potential biases. RESULTS From 2013 to 2020, 583 consecutive patients receiving either VATS (105, 18%) or Open (478, 82%) pneumonectomy were included. Conversion from VATS to open was found in 20 patients (19.0%). The conversion patients had similar rates of major complications and perioperative mortality compared with the Open group. After PSM, 203 patients were included. No significant differences were observed in major complications and perioperative mortality between the two groups. For patients with stage pT2 tumors, the major complication rate in the VATS group was significantly lower than in the Open group (7.6% vs. 20.6%, p = 0.042). Compared with left pneumonectomy, the incidence of bronchopleural fistula (BPF) was significantly higher in right pneumonectomy for both VATS (0 vs. 16.7%, p = 0.005) and Open (0.7% vs. 6.5%, p = 0.002) approaches. CONCLUSIONS Perioperative results of VATS pneumonectomy are non-inferior to those of the Open approach. Conversion to open surgery does not compromise perioperative outcomes. Patients with lower pT stage tumors who need pneumonectomy may benefit from VATS.
Collapse
Affiliation(s)
- Jizhuang Luo
- Department of Thoracic Surgery, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, No. 241 West Huaihai Road, Shanghai 200032, China
| | - Chunyu Ji
- Department of Thoracic Surgery, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, No. 241 West Huaihai Road, Shanghai 200032, China
| | - Alessio Campisi
- Department of Thoracic Surgery, University and Hospital of Trust-Ospedale Borgo Trento, Piazzale Aristide Stefani 1, 37126 Verona, Italy
| | - Tangbing Chen
- Department of Thoracic Surgery, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, No. 241 West Huaihai Road, Shanghai 200032, China
| | - Walter Weder
- Department of Thoracic Surgery, University Hospital Zurich, CH-8091 Zurich, Switzerland
| | - Wentao Fang
- Department of Thoracic Surgery, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, No. 241 West Huaihai Road, Shanghai 200032, China
| |
Collapse
|
423
|
Pulmonary cancers across different histotypes share hybrid tuft cell/ionocyte-like molecular features and potentially druggable vulnerabilities. Cell Death Dis 2022; 13:979. [PMID: 36402755 PMCID: PMC9675833 DOI: 10.1038/s41419-022-05428-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 11/08/2022] [Accepted: 11/10/2022] [Indexed: 11/21/2022]
Abstract
Tuft cells are chemosensory epithelial cells in the respiratory tract and several other organs. Recent studies revealed tuft cell-like gene expression signatures in some pulmonary adenocarcinomas, squamous cell carcinomas (SQCC), small cell carcinomas (SCLC), and large cell neuroendocrine carcinomas (LCNEC). Identification of their similarities could inform shared druggable vulnerabilities. Clinicopathological features of tuft cell-like (tcl) subsets in various lung cancer histotypes were studied in two independent tumor cohorts using immunohistochemistry (n = 674 and 70). Findings were confirmed, and additional characteristics were explored using public datasets (RNA seq and immunohistochemical data) (n = 555). Drug susceptibilities of tuft cell-like SCLC cell lines were also investigated. By immunohistochemistry, 10-20% of SCLC and LCNEC, and approximately 2% of SQCC expressed POU2F3, the master regulator of tuft cells. These tuft cell-like tumors exhibited "lineage ambiguity" as they co-expressed NCAM1, a marker for neuroendocrine differentiation, and KRT5, a marker for squamous differentiation. In addition, tuft cell-like tumors co-expressed BCL2 and KIT, and tuft cell-like SCLC and LCNEC, but not SQCC, also highly expressed MYC. Data from public datasets confirmed these features and revealed that tuft cell-like SCLC and LCNEC co-clustered on hierarchical clustering. Furthermore, only tuft cell-like subsets among pulmonary cancers significantly expressed FOXI1, the master regulator of ionocytes, suggesting their bidirectional but immature differentiation status. Clinically, tuft cell-like SCLC and LCNEC had a similar prognosis. Experimentally, tuft cell-like SCLC cell lines were susceptible to PARP and BCL2 co-inhibition, indicating synergistic effects. Taken together, pulmonary tuft cell-like cancers maintain histotype-related clinicopathologic characteristics despite overlapping unique molecular features. From a therapeutic perspective, identification of tuft cell-like LCNECs might be crucial given their close kinship with tuft cell-like SCLC.
Collapse
|
424
|
Zhou F, Guo H, Zhou X, Xie H, Tian T, Zhao W, Gao G, Xiong A, Wang L, Li W, Chen X, Zhang Y, Fan J, Wu F, Zhang Y, Zhou C. Immune checkpoint inhibitors plus chemotherapy in patients with locally advanced or metastatic pulmonary sarcomatoid carcinoma: a multicentric real-world study. Ther Adv Med Oncol 2022; 14:17588359221136759. [PMID: 36419724 PMCID: PMC9676284 DOI: 10.1177/17588359221136759] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Accepted: 10/14/2022] [Indexed: 08/08/2023] Open
Abstract
INTRODUCTION Immune checkpoint inhibitors (ICIs) have demonstrated promising efficacy as monotherapy in patients with pulmonary sarcomatoid carcinoma (PSC). We performed the current multi-institutional, real-world study to assess the efficacy of ICIs plus chemotherapy in patients with PSC. METHODS All consecutive patients with locally advanced or metastatic PSC from three centers treated with ICIs between January 2018 and July 2021 were enrolled. Programmed death ligand 1 (PD-L1) expression was stained and evaluated using immunohistochemical with 22C3. Single-cell RNA sequencing (scRNA-seq) was performed in two patients with PSC and two patients with adenocarcinoma to understand the cell-type-specific transcriptome landscape of cancer cells and tumor microenvironment (TME) of PSC. RESULTS A cohort of 42 PSC patients was identified. In the overall population, the objective response rate (ORR) was 73.8%, median progression-free survival (mPFS) was 10.3 months and median overall survival was not reached and 2-year survival rate was 51.2%. For 34 treatment-naïve patients who received first-line ICIs plus chemotherapy, the ORR was 70.6%, mPFS was 10.3 months and 2-year survival rate was 57.8%. In patients with PD-L1 tumor proportion score (TPS) < 1%, 1-49%, and ⩾50%, the ORR was 33.3%, 72.7%, and 85.7% and mPFS was 6.0, 6.7, and 10.3 months, respectively. Notably, two patients with transformed PSC from lung adenocarcinoma after epidermal growth factor receptor-tyrosine kinase inhibitor treatment also responded well to ICIs plus chemotherapy. scRNA-seq revealed immune-cell-inflamed TME, lower intratumoral heterogeneity, and activated immune response pathway in PSC. CONCLUSIONS Our study demonstrated remarkable efficacy of ICIs plus chemotherapy as first-line therapy for patient with locally advanced or metastatic PSC.
Collapse
Affiliation(s)
| | | | | | | | | | - Wencheng Zhao
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Guanghui Gao
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Anwen Xiong
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Lei Wang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Wei Li
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Xiaoxia Chen
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Yan Zhang
- Singleron Biotechnologies, Nanjing, Jiangsu, China
| | - Jue Fan
- Singleron Biotechnologies, Nanjing, Jiangsu, China
| | | | | | | |
Collapse
|
425
|
Variant Enrichment Analysis to Explore Pathways Disruption in a Necropsy Series of Asbestos-Exposed Shipyard Workers. Int J Mol Sci 2022; 23:ijms232113628. [DOI: 10.3390/ijms232113628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 10/28/2022] [Accepted: 11/02/2022] [Indexed: 11/09/2022] Open
Abstract
The variant enrichment analysis (VEA), a recently developed bioinformatic workflow, has been shown to be a valuable tool for whole-exome sequencing data analysis, allowing finding differences between the number of genetic variants in a given pathway compared to a reference dataset. In a previous study, using VEA, we identified different pathway signatures associated with the development of pulmonary toxicities in mesothelioma patients treated with radical hemithoracic radiation therapy. Here, we used VEA to discover novel pathways altered in individuals exposed to asbestos who developed or not asbestos-related diseases (lung cancer or mesothelioma). A population-based autopsy study was designed in which asbestos exposure was evaluated and quantitated by investigating objective signs of exposure. We selected patients with similar exposure to asbestos. Formalin-fixed paraffin-embedded (FFPE) tissues were used as a source of DNA and whole-exome sequencing analysis was performed, running VEA to identify potentially disrupted pathways in individuals who developed thoracic cancers induced by asbestos exposure. By using VEA analysis, we confirmed the involvement of pathways considered as the main culprits for asbestos-induced carcinogenesis: oxidative stress and chromosome instability. Furthermore, we identified protective genetic assets preserving genome stability and susceptibility assets predisposing to a worst outcome.
Collapse
|
426
|
Factors Associated With Lymph Node Yield and Effects of Lymph Node Density on Survival of Patients With Pulmonary Sarcomatoid Carcinoma. Am J Clin Oncol 2022; 45:458-464. [PMID: 36256867 PMCID: PMC9624378 DOI: 10.1097/coc.0000000000000946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
OBJECTIVE The objective of this study was to identify factors associated with lymph node yield (LNY) during surgeries for pulmonary sarcomatoid carcinoma (PSC) and to determine effects of lymph node density (LND) on the overall survival (OS) of patients with PSC. MATERIALS AND METHODS The SEER Research Plus database was searched for data on patients with PSC from 1988 to 2018. Poisson regression was used of all patients with PSC to identify relevant factors associated with LNY. Univariate and multivariate Cox regression analyses were adopted for lymph node (LN)-positive patients to evaluate the impact of LND on OS. The 5-year OS rates of patients with PSC were compared based on their LN status and LND. RESULTS There were 545 eligible patients in the study sample, 175 of which were LN-positive. These patients had significantly lower 5-year OS than those with no positive LNs ( P <0.001). Poisson regression analysis indicated relevant factors increasing LNY included higher diagnosis age, non-Hispanic American Indian or Alaska Native races, larger tumor, pleomorphic carcinoma histology, and more advanced disease stages. The Cox regression analysis indicated higher LND ( P =0.022) was probably associated with a worse prognosis for LN-positive patients. The group with LND ≥0.12 had a higher risk of death than the group with LND <0.12 ( P <0.001) among LN-positive patients with PSC. CONCLUSIONS Patients with PSC with high LND experienced worse outcomes than those with low LND. Further risk stratification of patients with PSC may help to improve survival benefits based on prognostic indicators of LND.
Collapse
|
427
|
Chen J, Xu J, Xiang J, Wan T, Deng H, Li D. A multivalent activatable aptamer probe with ultralow background signal and high sensitivity for diagnosis of lung adenocarcinoma. Talanta 2022. [DOI: 10.1016/j.talanta.2022.124056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
428
|
Chang CL, Hsieh MS, Shih JY, Lee YH, Liao WY, Hsu CL, Yang CY, Chen KY, Lee JH, Ho CC, Tsai TH, Yang JCH, Yu CJ. Real-world treatment patterns and outcomes among patients with advanced non-small-cell lung cancer with spindle cell and/or giant cell carcinoma. Ther Adv Med Oncol 2022; 14:17588359221133889. [PMID: 36324732 PMCID: PMC9618761 DOI: 10.1177/17588359221133889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 10/03/2022] [Indexed: 11/06/2022] Open
Abstract
Objectives A definitive diagnosis of pulmonary sarcomatoid carcinoma cannot be made with small biopsies. In clinical practice, a diagnosis of advanced non-small-cell lung cancer with spindle cell and/or giant cell carcinoma (NSCLCsg), or possible sarcomatoid carcinoma, is acceptable. Therefore, we aimed to investigate the treatment patterns and outcomes of advanced NSCLCsg. Materials and methods Between 01 January 2012 and 01 April 2021, patients with pathologically proven advanced NSCLCsg were enrolled. The choice of treatment was based on clinician discretion. Results In all, 101 patients with advanced NSCLCsg were enrolled. In total, 77 (76.2%) patients received at least one line of systemic therapy; 44 patients (43.1%) had received platinum doublet chemotherapy; 27 (26.7%) patients had been treated with targeted therapies; and 23 patients (22.8%) had been given an immune checkpoint inhibitor (ICI). The median overall survival (OS) was 6.3 months [95% confidence interval (CI): 3.6-9.0 months]. Excluding patients without systemic therapy, patients who had received an ICI had better OS (median: 18.2 months) than those who had not (median 3.8 months, log-rank test p = 0.002). No significant difference in OS was detected between patients who had or had not received platinum doublet chemotherapy (log-rank test p = 0.279), or targeted therapy (log-rank test p = 0.416). Having received any systemic therapy [hazard ratio (HR): 0.33, 95% CI: 0.18-0.61, p < 0.0001) and ICI (HR: 0.38, 95% CI: 0.19-0.78, p = 0.008) were independent factors for better OS. Patients with programmed death ligand-1 (PD-L1) expression ⩾50% had better OS than those with PD-L1 expression <50% (HR: 0.51, 95%: 0.30-0.86, p = 0.012). Conclusion Although advanced NSCLCsg has a poor survival outcome, our results showed that ICI may prolong OS in patients with advanced NSCLCsg. Further prospective studies are warranted to gain more understanding of the role of ICI in this specific patient population.
Collapse
Affiliation(s)
- Chia-Ling Chang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, National Taiwan University Hospital, Hsin-Chu Branch, and National Taiwan University College of Medicine
| | - Min-Shu Hsieh
- Department of Pathology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei,Graduate Institute of Pathology, National Taiwan University College of Medicine, Taipei
| | - Jin-Yuan Shih
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei
| | - Yi-Hsuan Lee
- Department of Pathology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei,Graduate Institute of Pathology, National Taiwan University College of Medicine, Taipei
| | | | - Chia-Lin Hsu
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei
| | - Ching-Yao Yang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei
| | - Kuan-Yu Chen
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei
| | - Jih-Hsiang Lee
- Department of Oncology, National Taiwan University Hospital, Hsin-Chu Branch, and National Taiwan University College of Medicine
| | - Chao-Chi Ho
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei
| | - Tzu-Hsiu Tsai
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei
| | - James Chih-Hsin Yang
- Department of Oncology, National Taiwan University Cancer Center and National Taiwan University College of Medicine, Taipei
| | - Chong-Jen Yu
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, National Taiwan University Hospital, Hsin-Chu Branch, and National Taiwan University College of Medicine
| |
Collapse
|
429
|
Souza da Silva R, Pinto R, Cirnes L, Schmitt F. Tissue management in precision medicine: What the pathologist needs to know in the molecular era. Front Mol Biosci 2022; 9:983102. [DOI: 10.3389/fmolb.2022.983102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 10/13/2022] [Indexed: 11/13/2022] Open
Abstract
Precision medicine is “an emerging approach for disease treatment and prevention that takes into account individual variability in genes, environment, and lifestyle for each person.” Among many medical specialists involved in precision medicine, the pathologists play an important and key role in the implementation and development of molecular tests that are in the center of decision of many therapeutic choices. Besides many laboratory procedures directly involved in the molecular tests, is fundamental to guarantee that tissues and cells collected for analysis be managed correctly before the DNA/RNA extraction. In this paper we explore the pivotal and interconnected points that can influence molecular studies, such as pre-analytical issues (fixation and decalcification); diagnosis and material selection, including the calculation of nuclei neoplastic fraction. The standardization of sample processing and morphological control ensures the accuracy of the diagnosis. Tissue or cytological samples constitutes the main foundation for the determination of biomarkers and development of druggable targets. Pathology and precision oncology still have a long way to go in terms of research and clinical practice: improving the accuracy and dissemination of molecular tests, learning in molecular tumor boards for advanced disease, and knowledge about early disease. Precision medicine needs pathology to be precise.
Collapse
|
430
|
Chauhan A, Del Rivero J, Ramirez RA, Soares HP, Li D. Treatment Sequencing Strategies in Advanced Neuroendocrine Tumors: A Review. Cancers (Basel) 2022; 14:5248. [PMID: 36358667 PMCID: PMC9656186 DOI: 10.3390/cancers14215248] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 10/04/2022] [Accepted: 10/14/2022] [Indexed: 10/15/2023] Open
Abstract
Neuroendocrine tumor (NET) incidence has grown. The treatment landscape for advanced NETs is rapidly evolving, but there are limited head-to-head data to guide treatment sequencing decisions. We assessed the available clinical data to aid practicing clinicians in their routine clinical decision-making. Clinical trials have demonstrated efficacy benefits for new therapies in advanced NETs. Emerging long-term data from these trials have enabled clinicians to make more accurate risk-benefit assessments, particularly for patients receiving multiple lines of therapy. However, clinical data specifically regarding treatment sequencing are limited. In lieu of definitive data, treatment sequencing should be based on disease-related factors (e.g., site of tumor origin, volume of disease) and patient-related characteristics (e.g., comorbidities, patient preferences). Clinical decision-making in advanced NETs remains highly individualized and complex; important evidence gaps regarding treatment sequencing remain. Given this, advanced NET management should be a joint effort of multidisciplinary teams at referring and high-volume centers. Additional clinical trial and real-world evidence are needed to meet the challenge of understanding how to sequence available NET therapies. Until these trials are conducted, the best practices provided in this review may serve as a guide for clinicians making treatment sequencing decisions based on the available data.
Collapse
Affiliation(s)
- Aman Chauhan
- Division of Medical Oncology, Department of Internal Medicine, Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA
| | - Jaydira Del Rivero
- Developmental Therapeutics Branch, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Robert A. Ramirez
- Division of Hematology Oncology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Heloisa P. Soares
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute at University of Utah, Salt Lake City, UT 84112, USA
| | - Daneng Li
- Department of Medical Oncology & Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| |
Collapse
|
431
|
The Clinical Characteristics and Treatments for Large Cell Carcinoma Patients Older than 65 Years Old: A Population-Based Study. Cancers (Basel) 2022; 14:cancers14215231. [PMID: 36358648 PMCID: PMC9658669 DOI: 10.3390/cancers14215231] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 10/09/2022] [Accepted: 10/18/2022] [Indexed: 11/17/2022] Open
Abstract
Background: Pulmonary large cell carcinoma, a type of non-small cell lung cancer (NSCLC), is a rare neoplasm with poor prognosis. In this study, our aim was to investigate the impact of radiation sequences with surgery for stage III/IV LCC patients between different age groups, especially in the elderly patients. Patients and Methods: The patients with LCC and other types of NSCLC in the Surveillance, Epidemiology and End Results (SEER) database from 2004 to 2015 were retrospectively analyzed. Then we divided the LCC patients into two age groups: <65 years old group and ≥65 years old group. Propensity score method (PSM) was used to control potential differences between different groups. The overall survival (OS) of LCC patients and other types of NSCLC patients were evaluated by Kaplan−Meier analysis. Univariate and multivariate Cox regression analysis were employed to explore the independent risk factors of OS. The forest plots of HRs for OS were generated to show the above outcomes more visually. Results: In total, 11,349 LCC patients and 129,118 other types of NSCLC patients were enrolled in this study. We divided LCC patients into <65 years old group (4300) and ≥65 years old group (7049). LCC patients was more common in whites (81.4%), males (58.3%), elderly (≥65 years old: 62.1%), east regions (52.7%), upper lobe (51.6%), right-origin of primary (55.4%), with advanced grade (54.2%) or stage (76.7%). After PSM, Kaplan−Meier analysis and multivariate Cox analysis showed significantly worse survival prognosis for LCC patients compared to other types of NSCLC, especially in the group ≥65 years old (HR: 1.230; 95% CI: 1.171−1.291; p < 0.001). For LCC patients, there were some risk survival factors including whites, males, not upper lobe, advanced stage, elder age at diagnosis, bone metastasis, liver metastasis, singled status, no lymphadenectomy, no surgery, and no chemotherapy (p < 0.05). In LCC patients ≥65 years old, radiation after surgery had significantly better impact on overall survival outcomes (HR: 0.863, 95% CI: 0.765−0.973, p = 0.016), whereas radiation prior to surgery (HR: 1.425, 95% CI: 1.059−1.916, p = 0.019) had significantly worse impact on prognosis of patients. In LCC patients <65 years old, radiation sequences with surgery had no significant impact on the OS of patients (p = 0.580), but ≥4 LNRs had significantly survival benefits to prognosis (HR:0.707, 95% CI: 0.584−0.855). Elderly LCC patients had worse malignant tumors than young patients, of which the majority were diagnosed as stage III/IV tumors. Conclusions: Postoperative radiotherapy may achieve a better prognosis for stage III/IV LCC patients older than 65 years old compared to other radiation sequences with surgery.
Collapse
|
432
|
Ma Y, Li W, Li Z, Chen J, Wang H, Jiang T, Zhu J. Immunophenotyping of pulmonary sarcomatoid carcinoma. Front Immunol 2022; 13:976739. [PMID: 36341325 PMCID: PMC9633134 DOI: 10.3389/fimmu.2022.976739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 10/10/2022] [Indexed: 11/29/2022] Open
Abstract
Background Previous studies have suggested that patients with pulmonary sarcomatoid carcinoma (PSC)may benefit from immune checkpoint inhibitors (ICIs); however, relevant data are lacking. This study aimed to establish the immunophenotype of PSC by assessing PD-L1 and CD8+ T-cell infiltration. Methods A retrospective analysis of pathologically confirmed PSC cases from two centers was performed from January 2009 to May 2021. According to the infiltration of CD8+ T cells in different spatial regions, patients were classified into three types: immune-inflamed, immune-excluded, and immune desert. PD-L1 staining was also performed on the intratumoral region and the tumor proportion score (TPS) was used for scoring. Combined with CD8+ T-cell infiltration and PD-L1 expression in the intratumoral region, immunophenotyping can be divided into four types: type I (PD-L1+/CD8+, adaptive immune resistance), type II (PD-L1-/CD8-, immunologic ignorance), type III (PD-L1+/CD8-, intrinsic induction), and type IV (PD-L1-/CD8+, tolerance). Finally, correlation analysis was performed on the immunophenotype, clinicopathological characteristics, and outcomes of the patients. Results A total of 32 patients with PSC were included in the final analysis. Of these patients, 65.6% (21/32), 15.6% (5/32), and 18.8% (6/32) were classified as immune-inflamed, immune-excluded, and immune-desert, respectively. Notably, the immune-inflamed type is predominantly observed in pleomorphic carcinomas (PC, 66.7%). Moreover, among these participants, 19 (59.4%) were classified as PD-L1 positive according to the TPS score. In particular, 11 (34.4%) patients had PD-L1 TPS scores >50%. Next, we immunophenotyped patients with PSC based on CD8+ T cell infiltration and tumor cell PD-L1 expression (types I–IV). Type I (PD-L1+/CD8+, adaptive immune resistance) was the most prevalent subtype, accounting for 46.9% (15/32), followed by type II (PD-L1-/CD8-, immunological ignorance) (21.9%), type IV (PD-L1-/CD8+, tolerance) (18.7%), and type III (PD-L1+/CD8-, intrinsic induction) (12.5%). Finally, we performed a survival analysis and found that neither immunophenotype was a predictor of prognosis in patients with PSC. Multivariate analysis showed that pneumonectomy increased the risk of death by four times compared with lobectomy (RR: 4.1; 95% CI:1.3-12.4, P=0.014). Conclusion Patients with PSC are characterized by immune-inflamed type and type I (PD-L1+/CD8+, adaptive immune resistance), explaining the intrinsic reasons for their high response rate to immunotherapy.
Collapse
Affiliation(s)
- Yu Ma
- Department of Pathology, Shaanxi Provincial People’s Hospital, Xi’an, China
| | - Wensheng Li
- Department of Pathology, Shaanxi Provincial People’s Hospital, Xi’an, China
| | - Zhenzhen Li
- Department of Pathology, Shaanxi Provincial People’s Hospital, Xi’an, China
| | - Jie Chen
- Department of Pathology, Shaanxi Provincial People’s Hospital, Xi’an, China
| | - Hongtao Wang
- Department of Thoracic Surgery, Shaanxi Provincial People’s Hospital, Xi’an, China
| | - Tao Jiang
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi’an, China
- *Correspondence: Jianfei Zhu, ; Tao Jiang,
| | - Jianfei Zhu
- Department of Thoracic Surgery, Shaanxi Provincial People’s Hospital, Xi’an, China
- *Correspondence: Jianfei Zhu, ; Tao Jiang,
| |
Collapse
|
433
|
Zhang MY, Tang LS, Qin ZJ, Hao YT, Cheng K, Zheng A. Clinical features and prognostic factors of pulmonary carcinosarcoma: A nomogram development and validation based on surveillance epidemiology and end results database. Front Med (Lausanne) 2022; 9:988830. [PMID: 36330063 PMCID: PMC9622765 DOI: 10.3389/fmed.2022.988830] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 10/03/2022] [Indexed: 12/20/2023] Open
Abstract
BACKGROUND Pulmonary carcinosarcoma (PCS) is a rare but aggressive malignant disease in the lung. It is characterized by coexisting histologic elements of carcinomatous and sarcomatous components. This study aimed to comprehensively understand the clinical features of PCS and develop a nomogram for prognostic prediction of PCS patients. METHODS Data were collected from the Surveillance Epidemiology and End Results (SEER) database between 1975 and 2018. Propensity-score matching (PSM) was used to match the demographic characteristic of the PCS vs. pulmonary sarcoma (PS). Cancer-specific survival (CSS) and overall survival (OS) were the main endpoints of the survival of patients and were evaluated using the Kaplan Meier curves and Cox proportional hazards regression. We further randomly split enrolled PCS patients from SEER into the training and validation sets. All independent predictors for OS of the training set were integrated to create a predictive nomogram. The performance of the nomogram was determined by discrimination, calibration ability, clinical usefulness, and risk stratification ability both in the training and validation cohorts. In addition, the clinical data of PCS patients from the West China Hospital were also retrospectively analyzed by this model. RESULTS A total of 428 PCS patients and 249 PS patients were enrolled from SEER. Compared to pure PS, PCS was associated with significantly better survival in the unmatched cohorts, whereas non-significantly better survival after PSM. In subgroup analysis, PCS showed significantly worse survival than pure PS in subgroups among the race, marital status, and radiation treatment. A nomogram was constructed for PCS patients' survival prediction by combining the independent risk factors, including gender, stage, surgery, radiation, and chemotherapy. The nomogram showed good discrimination, calibration, and predictive power in the training and validation sets. Risk stratification analysis indicated that the nomogram scores efficiently divided PCS patients into low and high-risk groups. CONCLUSION PCS is a rare malignant disease of the lung with distinct clinical features. It had a comparable survival compared with pure PS in the matched cohorts. In addition, a nomogram was developed and validated for predicting the OS in PCS patients.
Collapse
Affiliation(s)
- Ming-Yi Zhang
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lian-Sha Tang
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhao-Juan Qin
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ya-Ting Hao
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Ke Cheng
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ai Zheng
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
434
|
Liang Y, Wang N, Zhang Y, Jiang W, Fang C, Feng Y, Ma H, Jiang F, Dong G. Self-restricted circular RNA circSOX2 suppressed the malignant progression in SOX2-amplified LUSC. Cell Death Dis 2022; 13:873. [PMID: 36243874 PMCID: PMC9568965 DOI: 10.1038/s41419-022-05288-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 09/20/2022] [Accepted: 09/21/2022] [Indexed: 11/08/2022]
Abstract
Lung squamous cell carcinoma (LUSC) is a histological subtype of non-small cell lung cancer with the worse progression. SRY-Box Transcription Factor 2 (SOX2) copy number amplification (CNA) is the oncogenic driver in ~60% of patients diagnosed with LUSC. Thus, SOX2 represents an effective therapeutic target in SOX2-amplified LUSC. However, SOX2 protein was considered undruggable. Here, we report the expression of a circular RNA, cicSOX2 in SOX2-amplified LUSC. Patients with SOX2-CAN LUSC expressing circSOX2 manifested increased survival outcomes. CircSOX2 suppressed the proliferation, metastasis, and sphere formation in SOX2-amplified LUSC in vitro and in vivo. CircSOX2 originates in the reverse strand of the SOX2 gene and its sequence was reverse complement to partial 3'UTR of SOX2-coding transcript (mSOX2). CircSOX2 bound to AUF1 and occupied in the 3'UTR of mSOX2, inducing the degradation of mSOX2. In general, circSOX2 is an endogenous self-restricted circRNA in SOX2-amplified LUSC. CircSOX2 might be an effective and stable nucleic acid drug candidate in SOX2-amplified LUSC with low immunogenicity.
Collapse
Affiliation(s)
- Yingkuan Liang
- grid.429222.d0000 0004 1798 0228Department of Thoracic Surgery, the First Affiliated Hospital of Soochow University, 215006 Suzhou, China ,grid.263761.70000 0001 0198 0694Department of Thoracic Surgery, Dushu Lake Hospital Affiliated to Soochow University, Medical Center of Soochow University, Suzhou Dushu Lake Hospital, 215000 Suzhou, China
| | - Nan Wang
- grid.263761.70000 0001 0198 0694Department of Thoracic Surgery, Dushu Lake Hospital Affiliated to Soochow University, Medical Center of Soochow University, Suzhou Dushu Lake Hospital, 215000 Suzhou, China
| | - Yijian Zhang
- grid.452509.f0000 0004 1764 4566Department of Thoracic Surgery, Nanjing Medical University Affiliated Cancer Hospital, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Cancer Institute of Jiangsu Province, 210009 Nanjing, China
| | - Wei Jiang
- grid.263761.70000 0001 0198 0694Department of Thoracic Surgery, Dushu Lake Hospital Affiliated to Soochow University, Medical Center of Soochow University, Suzhou Dushu Lake Hospital, 215000 Suzhou, China
| | - Chen Fang
- grid.429222.d0000 0004 1798 0228Department of Thoracic Surgery, the First Affiliated Hospital of Soochow University, 215006 Suzhou, China
| | - Yu Feng
- grid.429222.d0000 0004 1798 0228Department of Thoracic Surgery, the First Affiliated Hospital of Soochow University, 215006 Suzhou, China
| | - Haitao Ma
- grid.429222.d0000 0004 1798 0228Department of Thoracic Surgery, the First Affiliated Hospital of Soochow University, 215006 Suzhou, China ,grid.263761.70000 0001 0198 0694Department of Thoracic Surgery, Dushu Lake Hospital Affiliated to Soochow University, Medical Center of Soochow University, Suzhou Dushu Lake Hospital, 215000 Suzhou, China
| | - Feng Jiang
- grid.452509.f0000 0004 1764 4566Department of Thoracic Surgery, Nanjing Medical University Affiliated Cancer Hospital, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Cancer Institute of Jiangsu Province, 210009 Nanjing, China
| | - Gaochao Dong
- grid.452509.f0000 0004 1764 4566Department of Thoracic Surgery, Nanjing Medical University Affiliated Cancer Hospital, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Cancer Institute of Jiangsu Province, 210009 Nanjing, China
| |
Collapse
|
435
|
Definition of a Novel Cuproptosis-Relevant lncRNA Signature for Uncovering Distinct Survival, Genomic Alterations, and Treatment Implications in Lung Adenocarcinoma. J Immunol Res 2022; 2022:2756611. [PMID: 36281357 PMCID: PMC9587678 DOI: 10.1155/2022/2756611] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/04/2022] [Accepted: 09/27/2022] [Indexed: 11/18/2022] Open
Abstract
Objective Cuproptosis is a newly discovered copper-independent cell death modality, and limited evidence suggests the critical implications in human cancers. Nonetheless, the clinical impacts of cuproptosis-relevant lncRNAs in lung adenocarcinoma (LUAD) remain largely ill-defined. The present study was aimed at defining a cuproptosis-relevant lncRNA signature for LUAD and discuss the clinical utility. Methods We collected transcriptome expression profiling, clinical information, somatic mutation, and copy number variations from TCGA-LUAD cohort retrospectively. The genetic alterations of cuproptosis genes were systematically assessed across LUAD, and cuproptosis-relevant lncRNAs were screened for defining a LASSO prognostic model. Genomic alterations, immunological and stemness features, and therapeutic sensitivity were studied with a series of computational approaches. Results Cuproptosis genes displayed aberrant expression and widespread genomic alterations across LUAD, potentially modulated by m6A/m5C/m1A RNA modification mechanisms. We defined a cuproptosis-relevant lncRNA signature, with a reliable efficacy in predicting clinical outcomes. High-risk subset displayed higher somatic mutations, CNVs, TMB, SNV neoantigens, aneuploidy score, CTA score, homologous recombination defects, and intratumor heterogeneity, cytolytic activity, CD8+ T effector, and antigen processing machinery, proving that this subset might benefit from immunotherapy. Increased stemness indexes and activity of oncogenic pathways might contribute to undesirable prognostic outcomes for high-risk subset. Additionally, high-risk patients generally exhibited higher response to chemotherapeutic agents (cisplatin, etc.). We also predicted several small molecule compounds (GSK461364, KX2-391, etc.) for treating this subset. Conclusion Accordingly, this cuproptosis-relevant lncRNA signature offers an efficient approach to identify and characterize diverse prognosis, genomic alterations, and treatment outcomes in LUAD, thus potentially assisting personalized therapy.
Collapse
|
436
|
Yang L, Fan Y, Lu H. Pulmonary Large Cell Neuroendocrine Carcinoma. PATHOLOGY AND ONCOLOGY RESEARCH 2022; 28:1610730. [PMID: 36304941 PMCID: PMC9592721 DOI: 10.3389/pore.2022.1610730] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 09/29/2022] [Indexed: 11/30/2022]
Abstract
Pulmonary large cell neuroendocrine carcinoma (LCNEC) is a rare subtype of malignant pulmonary tumor. The incidence rate of LCNEC was reported to be 0.3%–3% in lung cancers. Although LCNEC is classified as non-small cell lung cancer (NSCLC), it is more aggressive and malignant than other NSCLC, and its biological behavior is similar to that of small cell lung cancer (SCLC). Most of the LCNEC patients are elderly smoking male and the clinical manifestations are not specific. The imaging manifestations of the tumors are often located in the periphery and the upper lobes, and the enlargement of mediastinal or hilar lymph nodes is common. The diagnosis is mainly based on pathology by the histological features and immunohistochemistry (IHC). Specific neuroendocrine markers such as chromogranin A (CgA), synaptophysin (Syn) and CD56 are usually diffusely positive in LCNEC, and found that insulinoma-associated protein (INSM1) and high rate of Ki-67 are helpful for diagnosis. More differential diagnoses also increase the difficulty of correctly diagnosing LCNEC. The rise of LCNEC molecular typing in recent years may be helpful for diagnosis and subsequent treatment. This review focuses on the epidemiological features, imaging studies, pathology, diagnosis, treatment, and prognosis of LCNEC.
Collapse
Affiliation(s)
- Lan Yang
- Zhejiang Key Laboratory of Diagnosis and Treatment Technology on Thoracic Oncology (Lung and Esophagus), Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
- Department of Thoracic Medical Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
- The Second Clinical Medical College, Wenzhou Medical University, Wenzhou, China
| | - Ying Fan
- Zhejiang Key Laboratory of Diagnosis and Treatment Technology on Thoracic Oncology (Lung and Esophagus), Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
- Department of Thoracic Medical Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
- The Second Clinical Medical College, Wenzhou Medical University, Wenzhou, China
| | - Hongyang Lu
- Zhejiang Key Laboratory of Diagnosis and Treatment Technology on Thoracic Oncology (Lung and Esophagus), Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
- Department of Thoracic Medical Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
- The Second Clinical Medical College, Wenzhou Medical University, Wenzhou, China
- *Correspondence: Hongyang Lu,
| |
Collapse
|
437
|
Zografos E, Dimitrakopoulos FI, Koutras A. Prognostic Value of Circulating Tumor DNA (ctDNA) in Oncogene-Driven NSCLC: Current Knowledge and Future Perspectives. Cancers (Basel) 2022; 14:4954. [PMID: 36230877 PMCID: PMC9563444 DOI: 10.3390/cancers14194954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 10/03/2022] [Accepted: 10/05/2022] [Indexed: 11/16/2022] Open
Abstract
As we enter an unprecedented era of personalized medicine, molecular targeted therapies have the potential to induce improved survival outcome in patients with non-small cell lung cancer (NSCLC). However, a significant percentage of oncogene-driven NSCLC patients will relapse even after definitive treatment, whereas chronic and durable response to targeted therapies is a less common event in advanced-stage lung cancer. This phenomenon could be attributed to minimal residual disease (MRD), defined as a population of disseminated tumor cells that survive during the course or after treatment, eventually leading to recurrence and limiting patient survival. Circulating tumor DNA (ctDNA) is a powerful biomarker for MRD detection and monitoring and is a non-invasive approach of treating cancer, and especially NSCLC, based on a real-time assessment of the tumor genomic landscape. In this review, we present the key findings of studies that have used ctDNA with regard to its prognostic value and in respect to the most common druggable driver mutations of genes in NSCLC, such as epidermal growth factor receptor (EGFR), anaplastic lymphoma kinase (ALK), c-ros oncogene 1 (ROS1), rearranged during transfection (RET), Kirsten rat sarcoma virus (KRAS), B-Raf proto-oncogene (BRAF), and mesenchymal epithelial transition factor receptor (MET).
Collapse
Affiliation(s)
- Eleni Zografos
- Division of Oncology, University Hospital of Patras, University of Patras, 26504 Patras, Greece
- Molecular Oncology Laboratory, Division of Oncology, Department of Medicine, University of Patras, 26504 Patras, Greece
| | - Foteinos-Ioannis Dimitrakopoulos
- Division of Oncology, University Hospital of Patras, University of Patras, 26504 Patras, Greece
- Molecular Oncology Laboratory, Division of Oncology, Department of Medicine, University of Patras, 26504 Patras, Greece
| | - Angelos Koutras
- Division of Oncology, University Hospital of Patras, University of Patras, 26504 Patras, Greece
- Molecular Oncology Laboratory, Division of Oncology, Department of Medicine, University of Patras, 26504 Patras, Greece
| |
Collapse
|
438
|
Zhong YM, Yin K, Chen Y, Xie Z, Lv ZY, Yang JJ, Yang XN, Zhou Q, Wang BC, Zhong WZ, Gao LL, Zhou WB, Chen J, Tu HY, Liao RQ, Zhang DK, Zhang SL, Lu DX, Zheng HB, Zhang HH, Wu YL, Zhang XC. PD-1/PD-L1 combined with LAG3 is associated with clinical activity of immune checkpoint inhibitors in metastatic primary pulmonary lymphoepithelioma-like carcinoma. Front Immunol 2022; 13:951817. [PMID: 36263036 PMCID: PMC9574915 DOI: 10.3389/fimmu.2022.951817] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 08/16/2022] [Indexed: 11/13/2022] Open
Abstract
Primary pulmonary lymphoepithelioma-like carcinoma (PLELC) is an Epstein–Barr virus (EBV)-related, rare subtype of non-small-cell lung cancer (NSCLC). Immune checkpoint inhibitors (ICI) show durable responses in advanced NSCLC. However, their effects and predictive biomarkers in PLELC remain poorly understood. We retrospectively analyzed the data of 48 metastatic PLELC patients treated with ICI. Pretreated paraffin-embedded specimens (n = 19) were stained for PD-1, PD-L1, LAG3, TIM3, CD3, CD4, CD8, CD68, FOXP3, and cytokeratin (CK) by multiple immunohistochemistry (mIHC). Next-generation sequencing was performed for 33 PLELC samples. Among patients treated with ICI monotherapy (n = 30), the objective response rate (ORR), disease control rate (DCR), median progression-free survival (mPFS), and overall survival (mOS) were 13.3%, 80.0%, 7.7 months, and 24.9 months, respectively. Patients with PD-L1 ≥1% showed a longer PFS (8.4 vs. 2.1 months, p = 0.015) relative to those with PD-L1 <1%. Among patients treated with ICI combination therapy (n = 18), ORR, DCR, mPFS, and mOS were 27.8%, 100.0%, 10.1 months, and 19.7 months, respectively. Patients with PD-L1 ≥1% showed a significantly superior OS than those with PD-L1 <1% (NA versus 11.7 months, p = 0.001). Among the 19 mIHC patients, those with high PD-1/PD-L1 and LAG3 expression showed a longer PFS (19.0 vs. 3.9 months, p = 0.003). ICI also showed promising efficacy for treating metastatic PLELC. PD-L1 may be both predictive of ICI treatment efficacy and prognostic for survival in PLELC. PD-1/PD-L1 combined with LAG3 may serve as a predictor of ICI treatment effectiveness in PLELC. Larger and prospective trials are warranted to validate both ICI activity and predictive biomarkers in PLELC.This study was partly presented as a poster at the IASLC 20th World Conference on Lung Cancer 2019, 7–10 September 2019, Barcelona, Spain.
Collapse
Affiliation(s)
- Yu-Min Zhong
- Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Guangdong Lung Cancer Institute, Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Cancer Center, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Medical Research Center, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Kai Yin
- Guangdong Lung Cancer Institute, Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Cancer Center, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Yu Chen
- Guangdong Lung Cancer Institute, Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Cancer Center, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Medical Research Center, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Zhi Xie
- Guangdong Lung Cancer Institute, Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Cancer Center, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Medical Research Center, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Zhi-Yi Lv
- Guangdong Lung Cancer Institute, Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Cancer Center, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Medical Research Center, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Jin-Ji Yang
- Guangdong Lung Cancer Institute, Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Cancer Center, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Xue-Ning Yang
- Guangdong Lung Cancer Institute, Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Cancer Center, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Qing Zhou
- Guangdong Lung Cancer Institute, Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Cancer Center, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Bin-Chao Wang
- Guangdong Lung Cancer Institute, Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Cancer Center, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Wen-Zhao Zhong
- Guangdong Lung Cancer Institute, Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Cancer Center, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Ling-Ling Gao
- Guangdong Lung Cancer Institute, Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Cancer Center, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Wen-Bin Zhou
- Guangdong Lung Cancer Institute, Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Cancer Center, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Ji Chen
- Guangdong Lung Cancer Institute, Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Cancer Center, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Hai-Yan Tu
- Guangdong Lung Cancer Institute, Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Cancer Center, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Ri-Qiang Liao
- Guangdong Lung Cancer Institute, Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Cancer Center, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Dong-Kun Zhang
- Department of Thoracic Surgery, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Shui-Lian Zhang
- Guangdong Lung Cancer Institute, Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Cancer Center, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Medical Research Center, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Dan-Xia Lu
- Guangdong Lung Cancer Institute, Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Cancer Center, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Medical Research Center, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Hong-Bo Zheng
- Department of Medical Affairs, Genecast Biotechnology, Wuxi, China
| | - Heng-Hui Zhang
- Department of Medical Affairs, Genecast Biotechnology, Wuxi, China
| | - Yi-Long Wu
- Guangdong Lung Cancer Institute, Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Cancer Center, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Xu-Chao Zhang
- Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Guangdong Lung Cancer Institute, Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Cancer Center, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Medical Research Center, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
- *Correspondence: Xu-Chao Zhang,
| |
Collapse
|
439
|
Malapelle U, Pisapia P, Pepe F, Russo G, Buono M, Russo A, Gomez J, Khorshid O, Mack PC, Rolfo C, Troncone G. The evolving role of liquid biopsy in lung cancer. Lung Cancer 2022; 172:53-64. [PMID: 35998482 DOI: 10.1016/j.lungcan.2022.08.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 07/22/2022] [Accepted: 08/05/2022] [Indexed: 12/20/2022]
Abstract
Liquid biopsy has revolutionized the management of cancer patients. In particular, liquid biopsy-based testing has proven to be highly beneficial for identifying actionable cancer markers, especially when solid tissue biopsies are insufficient or unattainable. Beyond the predictive role, liquid biopsy may be a useful tool for comprehensive tumor genotyping, identification of emergent resistance mechanisms, monitoring of minimal residual disease, early detection, and cancer interception. The application of next generation sequencing to liquid biopsy has led to the "quantum leap" of predictive molecular pathology. Here, we review the evolving role of liquid biopsy in lung cancer.
Collapse
Affiliation(s)
- Umberto Malapelle
- Department of Public Health, University of Naples Federico II, Naples, Italy.
| | - Pasquale Pisapia
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Francesco Pepe
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Gianluca Russo
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Mauro Buono
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | | | - Jorge Gomez
- Center for Thoracic Oncology, Tisch Cancer Institute, Mount Sinai Medical System & Icahn School of Medicine, New York, NY, USA
| | - Ola Khorshid
- National Cancer Institute, Cairo University, Cairo, Egypt
| | - Philip C Mack
- Center for Thoracic Oncology, Tisch Cancer Institute, Mount Sinai Medical System & Icahn School of Medicine, New York, NY, USA
| | - Christian Rolfo
- Center for Thoracic Oncology, Tisch Cancer Institute, Mount Sinai Medical System & Icahn School of Medicine, New York, NY, USA
| | - Giancarlo Troncone
- Department of Public Health, University of Naples Federico II, Naples, Italy
| |
Collapse
|
440
|
Zhao C, Gao S, Xue Q, Tan F, Gao Y, Mao Y, Wang D, Zhao J, Yang D, Hong Q, Mu J. Clinical characteristics and prognostic factors of pulmonary sarcomatoid carcinoma. J Thorac Dis 2022; 14:3773-3781. [PMID: 36389311 PMCID: PMC9641323 DOI: 10.21037/jtd-22-393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 09/09/2022] [Indexed: 11/24/2022]
Abstract
BACKGROUND Pulmonary sarcomatoid carcinoma (PSC) is a kind of rare lung cancer. We aim to analyze the clinical characteristics and prognostic factors of patients with PSC. METHODS From January 1, 2006 to December 31, 2015, 119 patients in the Cancer Hospital Chinese Academy of Medical Sciences were diagnosed with PSC, and they received treatment. We retrospectively collected information on gender, age, body mass index (BMI), symptoms, family history, smoking history, tumor size, tumor location, tumor diameter, tumor-node-metastasis (TNM), pathological type, and other factors to analyze the relationship between these factors and 1-, 3-, 5-year, and overall survival (OS). RESULTS Male patients who had a smoking history (n=76) comprised the main group of PSC. Median patient age was 60.67±10.50 years (range, 26-89 years). The majority of these patients (n=82) presented with respiratory symptoms. The median survival of patients who died of PSC was 11.87 months (6.38-21.48 months). The 1-, 3-, and 5-year survival rates were 61.3%, 34.5%, and 31.9%, respectively. Patients with a lower T stage and without lymph node metastasis and distant metastasis had a better OS (P<0.05). Other clinical characteristics and the difference in treatments did not influence the prognosis significantly (P>0.05). CONCLUSIONS PSC is a rare malignant neoplasm of the lung with poor prognosis. Surgery is a major therapeutic method for this disease entity. TNM-stage is the main factors affecting prognosis.
Collapse
Affiliation(s)
- Chenguang Zhao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shugeng Gao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qi Xue
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fengwei Tan
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yushun Gao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yousheng Mao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Dali Wang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jun Zhao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ding Yang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qian Hong
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Juwei Mu
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
441
|
Yu F, Tan W, Chen Z, Shen X, Mo X, Mo X, He J, Deng Z, Wang J, Luo Z, Yang J. Nitidine chloride induces caspase 3/GSDME-dependent pyroptosis by inhibting PI3K/Akt pathway in lung cancer. Chin Med 2022; 17:115. [PMID: 36175965 PMCID: PMC9524076 DOI: 10.1186/s13020-022-00671-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 09/12/2022] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND As the increasing mortality and incidence of lung cancer (LC), there is an urgent need to discover novel treatment agent. In this study, we aimed to investigate the anti-LC effects of nitidine chloride (NC), a small molecular compound extracted from Chinese herbal medicine, while detailing its underlying mechanisms. METHODS Cell viability was detected by MTT assays and five cell death inhibitors, including ferrostatin-1 (Fer-1), Z-VAD-FMK, necrostatin-1 (Nec-1), disulfiram (DSF) and IM-54 were used to explore the type of cell death induced by NC. The microscopic features of NC-induced pyroptosis were assessed by transmission electron microscopy (TEM) and the pyroptotic-related proteins such as caspase and gasdermin family, were examined by western blot. Network pharmacology was employed to predict the potential mechanisms of NC in lung cancer treatment. CETSA and DARTs were used to determine the activity of NC binding to targeted protein. Xenograft mice model was established to further investigate the inhibitory effect and mechanism of NC against LC. RESULTS The pyroptosis inhibitor (DSF) and apoptosis inhibitor (Z-VAD-FMK) but not IM-54, necrostatin-1, or Ferrostatin-1 rescued NC-induced cell death. Morphologically, H1688 and A549 cells treated with NC showed notably pyroptotic features, such as cell swelling and large bubbles emerging from the plasma membrane. Gasdermin E (GSDME) rather than GSDMC or GSDMD was cleaved in NC-treated H1688 and A549 cells with an increased cleavage of caspase 3. Combined with network pharmacology and molecule docking, PI3K/Akt signaling axis was predicted and was further verified by CETSA and DARTs assay. In addition, the activation of PI3K is able to rescue the pyroptosis induced by NC in vitro. In xenograft model of LC, NC significantly hindered the transduction of PI3K-AKT pathway, inducing pyroptosis of tumor. CONCLUSION Our data indicated that NC is a potential therapeutic agent for the treatment of LC via triggering GSDME-dependent pyroptosis.
Collapse
Affiliation(s)
- Fei Yu
- grid.256607.00000 0004 1798 2653Department of Pharmacology, School of Pharmacy, 406 Graduate School of Guangxi Medical University, Nanning, 530021 Guangxi People’s Republic of China
| | - Weidan Tan
- grid.256607.00000 0004 1798 2653Department of Pharmacology, School of Pharmacy, 406 Graduate School of Guangxi Medical University, Nanning, 530021 Guangxi People’s Republic of China
| | - Zhiquan Chen
- grid.256607.00000 0004 1798 2653Department of Pharmacology, School of Pharmacy, 406 Graduate School of Guangxi Medical University, Nanning, 530021 Guangxi People’s Republic of China
| | - Xiaoju Shen
- grid.256607.00000 0004 1798 2653Department of Pharmacology, School of Pharmacy, 406 Graduate School of Guangxi Medical University, Nanning, 530021 Guangxi People’s Republic of China
| | - Xiaoxiang Mo
- grid.256607.00000 0004 1798 2653Department of Pharmacology, School of Pharmacy, 406 Graduate School of Guangxi Medical University, Nanning, 530021 Guangxi People’s Republic of China
| | - Xiaocheng Mo
- grid.256607.00000 0004 1798 2653Department of Pharmacology, School of Pharmacy, 406 Graduate School of Guangxi Medical University, Nanning, 530021 Guangxi People’s Republic of China
| | - Jingchuan He
- grid.256607.00000 0004 1798 2653Department of Pharmacology, School of Pharmacy, 406 Graduate School of Guangxi Medical University, Nanning, 530021 Guangxi People’s Republic of China
| | - Zhihua Deng
- grid.256607.00000 0004 1798 2653Department of Pharmacology, School of Pharmacy, 406 Graduate School of Guangxi Medical University, Nanning, 530021 Guangxi People’s Republic of China
| | - Jie Wang
- grid.256607.00000 0004 1798 2653Department of Pharmacology, School of Pharmacy, 406 Graduate School of Guangxi Medical University, Nanning, 530021 Guangxi People’s Republic of China
| | - Zhuo Luo
- grid.256607.00000 0004 1798 2653Department of Pharmacology, School of Pharmacy, 406 Graduate School of Guangxi Medical University, Nanning, 530021 Guangxi People’s Republic of China
| | - Jie Yang
- grid.256607.00000 0004 1798 2653Department of Pharmacology, School of Pharmacy, 406 Graduate School of Guangxi Medical University, Nanning, 530021 Guangxi People’s Republic of China
| |
Collapse
|
442
|
Liu Y, Zhang X, Cheng X, Luo Q, Yu M, Long K, Qu W, Tang Y, Gong M, Liang L, Ke X, Song Y. Characterization of fatty acid metabolism-related lncRNAs in lung adenocarcinoma identifying potential novel prognostic targets. Front Genet 2022; 13:990153. [PMID: 36299578 PMCID: PMC9589892 DOI: 10.3389/fgene.2022.990153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 09/01/2022] [Indexed: 11/15/2022] Open
Abstract
Lung adenocarcinoma (LUAD), a malignant respiratory tumor with an extremely poor prognosis, has troubled the medical community all over the world. According to recent studies, fatty acid metabolism (FAM) and long non-coding RNAs (lncRNAs) regulation have shown exciting results in tumor therapy. In this study, the original LUAD patient data was obtained from the TCGA database, and 12 FAM-related lncRNAs (AL390755.1, AC105020.6, TMPO-AS1, AC016737.2, AC127070.2, LINC01281, AL589986.2, GAS6-DT, AC078993.1, LINC02198, AC007032.1, and AL021026.1) that were highly related to the progression of LUAD were finally identified through bioinformatics analysis, and a risk score model for clinical reference was constructed. The window explores the immunology and molecular mechanism of LUAD, aiming to shed the hoping light on LUAD treatment.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Xixian Ke
- *Correspondence: Xixian Ke, ; Yongxiang Song,
| | | |
Collapse
|
443
|
Artificial Intelligence-Powered Whole-Slide Image Analyzer Reveals a Distinctive Distribution of Tumor-Infiltrating Lymphocytes in Neuroendocrine Neoplasms. Diagnostics (Basel) 2022; 12:diagnostics12102340. [PMID: 36292028 PMCID: PMC9600129 DOI: 10.3390/diagnostics12102340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/20/2022] [Accepted: 09/23/2022] [Indexed: 12/03/2022] Open
Abstract
Despite the importance of tumor-infiltrating lymphocytes (TIL) and PD-L1 expression to the immune checkpoint inhibitor (ICI) response, a comprehensive assessment of these biomarkers has not yet been conducted in neuroendocrine neoplasm (NEN). We collected 218 NENs from multiple organs, including 190 low/intermediate-grade NENs and 28 high-grade NENs. TIL distribution was derived from Lunit SCOPE IO, an artificial intelligence (AI)-powered hematoxylin and eosin (H&E) analyzer, as developed from 17,849 whole slide images. The proportion of intra-tumoral TIL-high cases was significantly higher in high-grade NEN (75.0% vs. 46.3%, p = 0.008). The proportion of PD-L1 combined positive score (CPS) ≥ 1 case was higher in high-grade NEN (85.7% vs. 33.2%, p < 0.001). The PD-L1 CPS ≥ 1 group showed higher intra-tumoral, stromal, and combined TIL densities, compared to the CPS < 1 group (7.13 vs. 2.95, p < 0.001; 200.9 vs. 120.5, p < 0.001; 86.7 vs. 56.1, p = 0.004). A significant correlation was observed between TIL density and PD-L1 CPS (r = 0.37, p < 0.001 for intra-tumoral TIL; r = 0.24, p = 0.002 for stromal TIL and combined TIL). AI-powered TIL analysis reveals that intra-tumoral TIL density is significantly higher in high-grade NEN, and PD-L1 CPS has a positive correlation with TIL densities, thus showing its value as predictive biomarkers for ICI response in NEN.
Collapse
|
444
|
Chen H, Zheng M, Zhang W, Long Y, Xu Y, Yuan M. Research Status of Mouse Models for Non-Small-Cell Lung Cancer (NSCLC) and Antitumor Therapy of Traditional Chinese Medicine (TCM) in Mouse Models. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:6404853. [PMID: 36185084 PMCID: PMC9519343 DOI: 10.1155/2022/6404853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 08/31/2022] [Indexed: 11/25/2022]
Abstract
Non-small-cell lung cancer (NSCLC) is known as one of the most lethal cancers, causing more than 1 million deaths annually worldwide. Therefore, the development of novel therapeutic drugs for NSCLC has become an urgent need. Herein, various mouse models provide great convenience not only for researchers but also for the development of antitumor drug. Meanwhile, TCM, as a valuable and largely untapped resource pool for modern medicine, provides research resources for the treatment of various diseases. Until now, cell-derived xenograft (CDX) model, patient-derived xenograft (PDX) model, syngeneic model, orthotopic model, humanized mouse model (HIS), and genetically engineered mouse models (GEMMs) have been reported in TCM evaluation. This review shows the role and current status of kinds of mouse models in antitumor research and summarizes the application progress of TCM including extracts, formulas, and isolated single molecules for NSCLC therapy in various mouse models; more importantly, it provides a theoretical exploration of what kind of mouse models is ideal for TCM efficacy evaluation in future. However, there are still huge challenges and limitations in the development of mouse models specifically for the TCM research, and none of the available models are perfectly matching the characteristics of TCM, which suppress the tumor growth through various mechanisms, especially by regulating immune function. Nevertheless, with fully functional immune system existing in syngeneic model and humanized mouse model (HIS), it is still suggested that these two models are more suitable for development of TCM especially for TCM extracts or formulas. Moreover, continued efforts are needed to generate more reliable mouse models to test TCM formulas in future research.
Collapse
Affiliation(s)
- Hongkui Chen
- Shanghai Lidebiotech Co. Ltd., Shanghai 201203, China
| | - Min Zheng
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Wenhui Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai 201203, China
| | - Yuan Long
- Shanghai Lidebiotech Co. Ltd., Shanghai 201203, China
| | - Yu Xu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai 201203, China
| | - Man Yuan
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai 201203, China
| |
Collapse
|
445
|
Wu N, Cao QW, Wang CN, Hu HG, Shi H, Deng K. Association between quantitative spectral CT parameters, Ki-67 expression, and invasiveness in lung adenocarcinoma manifesting as ground-glass nodules. Acta Radiol 2022; 64:1400-1409. [PMID: 36131377 DOI: 10.1177/02841851221128213] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
BACKGROUND Few studies about lung ground-glass nodules (GGNs) have been done using non-enhancement spectral computed tomography (CT) imaging. PURPOSE To examine the association between spectral CT parameters, Ki-67 expression, and invasiveness in lung adenocarcinoma manifesting as GGNs. MATERIAL AND METHODS Spectral CT parameters were analyzed in 106 patients with lung GGNs. The Ki-67 labeling index (Ki-67 LI) was measured, and patients were divided into low expression and high expression groups according to the number of positive-stained cells (low expression ≤10%; high expression >10%). Spectral CT parameters were compared between low and high expression groups. The correlation between spectral CT parameters and Ki-67 LI was estimated by Spearman correlation analysis. Cases were divided into a preinvasive and minimally invasive adenocarcinoma (MIA) group (atypical adenomatous hyperplasia, adenocarcinoma in situ, and MIA) and invasive adenocarcinoma (IA) group. Spectral CT parameters were compared between the two groups. The diagnostic performance was evaluated using receiver operating characteristic analysis. RESULTS There were significant differences in water concentration of lesions (WCL) and monochromatic CT values between the low and high expression groups. CT 40 keV had the highest correlation coefficient with Ki-67 LI. WCL and monochromatic CT values were significantly higher in the IA group than in the pre/MIA group. The value of area under the curve of CT 40 keV was 0.946 (95% confidence interval=0.905-0.988) for differentiating the two groups; the cutoff was -280.66 Hu. CONCLUSION Spectral CT is an effective non-invasive method for the prediction of proliferation and invasiveness in lung adenocarcinoma manifesting as GGNs.
Collapse
Affiliation(s)
- Nan Wu
- Shandong Provincial Qianfoshan Hospital, 159393Shandong University, Jinan, PR China
| | - Qi-Wei Cao
- Department of Pathology, 66310The First Affiliated Hospital of Shandong First Medical University, Jinan, PR China
| | - Chao-Nan Wang
- Department of Cardiology, 66310The Affiliated Hospital of Shandong University of TCM, Jinan, PR China
| | - Hong-Guang Hu
- Department of Radiology, 66310The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, PR China
| | - Hao Shi
- Shandong Provincial Qianfoshan Hospital, 159393Shandong University, Jinan, PR China
| | - Kai Deng
- Department of Radiology, 66310The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, PR China
| |
Collapse
|
446
|
Tsai HC, Huang JY, Hsieh MY, Wang BY. Survival of Lung Cancer Patients by Histopathology in Taiwan from 2010 to 2016: A Nationwide Study. J Clin Med 2022; 11:jcm11195503. [PMID: 36233370 PMCID: PMC9570537 DOI: 10.3390/jcm11195503] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 09/17/2022] [Accepted: 09/17/2022] [Indexed: 12/24/2022] Open
Abstract
Objective: Lung cancer poses a tremendous threat to the modern world. According to Taiwan’s Ministry of Health and Welfare, lung cancer took first place in total cancer deaths in 2021. This study investigated the overall lung cancer survival based on histopathology between 2010 and 2016 in Taiwan. Method: Data from 2010 to 2016 was collected from the Taiwan Cancer Registry (TCR). The characteristics and overall survival of 71,334 lung cancer patients were analyzed according to the tumor, node, metastasis (TNM) 7th staging system. Univariate and multivariate analyses were performed to identify differences in 1-year, 3-year, and 5-year survival between different histopathologies of lung cancer. Results: The 1-year overall survival rate increased from 54.07% in 2010 to 66.14% in 2016. The 3-year overall survival rate increased from 26.57% in 2010 to 41.12% in 2016 in all patients. Among the histopathologies of lung cancer, 3-year overall survival of adenocarcinoma patients increased the most and largely contributed to the increased 3-year overall survival of all lung cancer patients. Conclusions: The introduction of target therapy has led to a tremendous increase in overall survival for lung adenocarcinoma patients. However, target therapy differs by histopathology. Choosing the right target therapy and determining the correct histopathology of lung cancer is a pivotal key in increasing the overall survival of patients. Together with immune therapy, the landscape of lung cancer treatments is changing.
Collapse
Affiliation(s)
- Hsuan-Chih Tsai
- Department of Family Medicine, Taichung Armed Forces General Hospital, Taichung 41148, Taiwan
- Department of Occupational Medicine, Taichung Veterans General Hospital, Taichung 40705, Taiwan
- School of Medicine, National Defense Medical Center, Taipei 11490, Taiwan
| | - Jing-Yang Huang
- Center for Health Data Science, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
| | - Ming-Yu Hsieh
- Department of Otorhinolaryngology-Head and Neck Surgery, Changhua Christian Hospital, No. 135, Nanxium St., Changhua 50006, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung 40227, Taiwan
| | - Bing-Yen Wang
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung 40227, Taiwan
- Division of Thoracic Surgery, Department of Surgery, Changhua Christian Hospital, Changhua 50006, Taiwan
- School of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
- School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Institute of Genomics and Bioinformatics, National Chung Hsing University, Taichung 40227, Taiwan
- Correspondence: ; Tel.: +886-(2)-7238595; Fax: +886-(2)-7228289
| |
Collapse
|
447
|
Luo J, Ding B, Campisi A, Chen T, Teng H, Ji C. Molecular, clinicopathological characteristics and surgical results of resectable SMARCA4-deficient thoracic tumors. J Cancer Res Clin Oncol 2022:10.1007/s00432-022-04359-6. [PMID: 36121510 DOI: 10.1007/s00432-022-04359-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 09/13/2022] [Indexed: 11/24/2022]
Abstract
PURPOSE SMARCA4-deficient thoracic tumors are rapid aggressive malignancies, often diagnosed at an advanced and inoperable stage. The value of pulmonary resection for resectable SMARCA4-deficient thoracic tumors is largely unknown. METHODS In this observational study, we included 45 patients who received surgery for stage I-III SMARCA4-deficient tumors. We compared the molecular, clinicopathological characteristics and survival between SMARCA4-dNSCLC and SMARCA4-deficient undifferentiated tumor (SMARCA4-dUT) patients. RESULTS Thirty-four SMARCA4-dNSCLC and 11 SMARCA4-dUT patients were included in this study. Molecular profiles were available in 33 out of 45 patients. The most common mutated gene was TP53 (21, 64%), and followed by STK11 (9, 27%), KRAS (5, 15%), FGFR1 (4, 12%) and ROS1 (4, 12%). There were 3 patients that harbored ALK mutation including 1 EML4-ALK rearrangement. There were 2 patients that harbored EGFR rare site missense mutation. SMARCA4-dUT patients had significance worse TTP (HR = 4.35 95% CI 1.77-10.71, p = 0.001) and OS (HR = 4.27, 95% CI 1.12-16.35, p = 0.022) compared to SMARCA4-dNSCLC patients. SMARCA4-dUT histologic type, stage II/III, R1/2 resection and lymphovascular invasion were independent poor prognostic predictors for both TTP and OS. There were 8 patients who received immunotherapy, the objective response rate was 50%. The SMARCA4-dNSCLC patient with ALK rearrangement was treated with crizotinib as second-line therapy, and achieved stable disease for 9.7 months. CONCLUSION Patients with SMARCA4-deficient tumors have a high probability of early recurrence after surgery, except for stage I patients. Immunotherapy seems to be a valuable strategy to treat recurrence.
Collapse
Affiliation(s)
- Jizhuang Luo
- Department of Thoracic Surgery, School of Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, 241 West Huaihai Rd, Xuhui District, Shanghai, 200030, China
| | - Bowen Ding
- Department of Pathology, School of Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, 241 West Huaihai Rd, Xuhui District, Shanghai, 200030, China
| | - Alessio Campisi
- Department of Thoracic Surgery, School of Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, 241 West Huaihai Rd, Xuhui District, Shanghai, 200030, China.,Department of Thoracic Surgery, University and Hospital Trust-Ospedale Borgo Trento, piazzale aristide stefani 1, Verona, Italy
| | - Tangbing Chen
- Department of Thoracic Surgery, School of Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, 241 West Huaihai Rd, Xuhui District, Shanghai, 200030, China
| | - Haohua Teng
- Department of Pathology, School of Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, 241 West Huaihai Rd, Xuhui District, Shanghai, 200030, China.
| | - Chunyu Ji
- Department of Thoracic Surgery, School of Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, 241 West Huaihai Rd, Xuhui District, Shanghai, 200030, China.
| |
Collapse
|
448
|
Buszka K, Ntzifa A, Owecka B, Kamińska P, Kolecka-Bednarczyk A, Zabel M, Nowicki M, Lianidou E, Budna-Tukan J. Liquid Biopsy Analysis as a Tool for TKI-Based Treatment in Non-Small Cell Lung Cancer. Cells 2022; 11:2871. [PMID: 36139444 PMCID: PMC9497234 DOI: 10.3390/cells11182871] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/07/2022] [Accepted: 09/09/2022] [Indexed: 12/03/2022] Open
Abstract
The treatment of non-small cell lung cancer (NSCLC) has recently evolved with the introduction of targeted therapy based on the use of tyrosine kinase inhibitors (TKIs) in patients with certain gene alterations, including EGFR, ALK, ROS1, BRAF, and MET genes. Molecular targeted therapy based on TKIs has improved clinical outcomes in a large number of NSCLC patients with advanced disease, enabling significantly longer progression-free survival (PFS). Liquid biopsy is an increasingly popular diagnostic tool for treating TKI-based NSCLC. The studies presented in this article show that detection and analysis based on liquid biopsy elements such as circulating tumor cells (CTCs), cell-free DNA (cfDNA), exosomes, and/or tumor-educated platelets (TEPs) can contribute to the appropriate selection and monitoring of targeted therapy in NSCLC patients as complementary to invasive tissue biopsy. The detection of these elements, combined with their molecular analysis (using, e.g., digital PCR (dPCR), next generation sequencing (NGS), shallow whole genome sequencing (sWGS)), enables the detection of mutations, which are required for the TKI treatment. Despite such promising results obtained by many research teams, it is still necessary to carry out prospective studies on a larger group of patients in order to validate these methods before their application in clinical practice.
Collapse
Affiliation(s)
- Karolina Buszka
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland
- Doctoral School, Poznan University of Medical Sciences, 60-812 Poznan, Poland
| | - Aliki Ntzifa
- Analysis of Circulating Tumor Cells Lab, Lab of Analytical Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, 15771 Athens, Greece
| | - Barbara Owecka
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland
| | - Paula Kamińska
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland
- Doctoral School, Poznan University of Medical Sciences, 60-812 Poznan, Poland
| | - Agata Kolecka-Bednarczyk
- Department of Immunology, Chair of Pathomorphology and Clinical Immunology, Poznan University of Medical Sciences, 60-806 Poznan, Poland
| | - Maciej Zabel
- Division of Anatomy and Histology, University of Zielona Góra, 65-046 Zielona Góra, Poland
| | - Michał Nowicki
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland
| | - Evi Lianidou
- Analysis of Circulating Tumor Cells Lab, Lab of Analytical Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, 15771 Athens, Greece
| | - Joanna Budna-Tukan
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland
| |
Collapse
|
449
|
Watanabe M, Matsumura Y, Yamaguchi H, Mine H, Takagi H, Ozaki Y, Fukuhara M, Muto S, Okabe N, Shio Y, Suzuki H. Large cell neuroendocrine carcinoma of the lung controlled for 4 years by a single administration of pembrolizumab: A case report. Thorac Cancer 2022; 13:2817-2822. [PMID: 36064196 PMCID: PMC9527153 DOI: 10.1111/1759-7714.14615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 08/01/2022] [Accepted: 08/02/2022] [Indexed: 11/30/2022] Open
Abstract
Large cell neuroendocrine carcinoma of the lung (LCNEC) is a rare and highly progressive tumor with a poor prognosis. Although immune checkpoint inhibitors have been approved for treatment of both small cell and non–small cell lung cancers, their role in the treatment of LCNEC is unclear. We describe a patient with postoperative recurrence of LCNEC who maintained complete remission for 4 years after a single administration of pembrolizumab. A 68‐year‐old Japanese man underwent thoracoscopic right lower lobectomy for LCNEC (pathological stage pT1bN0M0, stage IA2). Epidermal growth factor receptor and anaplastic lymphoma kinase were negative, and the programmed death ligand 1 expression rate in tumor cells was 5% (clone 22C3). Eight months later, the patient developed recurrence with mediastinal lymph node metastasis and pleural dissemination. Therefore, chemotherapy with cisplatin and etoposide was administered. However, relapse occurred 6 months later. Pembrolizumab was administered as second‐line chemotherapy, which was discontinued after first dose because of interstitial pneumonia 1 month later. Thereafter, however, both the lymph node metastasis and pleural dissemination disappeared and did not relapse for 4 years. Pembrolizumab may be used as a treatment option for pulmonary LCNEC.
Collapse
Affiliation(s)
- Masayuki Watanabe
- Department of Chest Surgery, Fukushima Medical University, Fukushima, Japan
| | - Yuki Matsumura
- Department of Chest Surgery, Fukushima Medical University, Fukushima, Japan
| | - Hikaru Yamaguchi
- Department of Chest Surgery, Fukushima Medical University, Fukushima, Japan
| | - Hayato Mine
- Department of Chest Surgery, Fukushima Medical University, Fukushima, Japan
| | - Hironori Takagi
- Department of Chest Surgery, Fukushima Medical University, Fukushima, Japan
| | - Yuki Ozaki
- Department of Chest Surgery, Fukushima Medical University, Fukushima, Japan
| | - Mitsuro Fukuhara
- Department of Chest Surgery, Fukushima Medical University, Fukushima, Japan
| | - Satoshi Muto
- Department of Chest Surgery, Fukushima Medical University, Fukushima, Japan
| | - Naoyuki Okabe
- Department of Chest Surgery, Fukushima Medical University, Fukushima, Japan
| | - Yutaka Shio
- Department of Chest Surgery, Fukushima Medical University, Fukushima, Japan
| | - Hiroyuki Suzuki
- Department of Chest Surgery, Fukushima Medical University, Fukushima, Japan
| |
Collapse
|
450
|
Song Q, Song B, Li X, Wang B, Li Y, Chen W, Wang Z, Wang X, Yu Y, Min X, Ma D. A CT-based nomogram for predicting the risk of adenocarcinomas in patients with subsolid nodule according to the 2021 WHO classification. Cancer Imaging 2022; 22:46. [PMID: 36064495 PMCID: PMC9446567 DOI: 10.1186/s40644-022-00483-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 08/23/2022] [Indexed: 11/10/2022] Open
Abstract
Purpose To establish a nomogram for predicting the risk of adenocarcinomas in patients with subsolid nodules (SSNs) according to the 2021 WHO classification. Methods A total of 656 patients who underwent SSNs resection were retrospectively enrolled. Among them, 407 patients were assigned to the derivation cohort and 249 patients were assigned to the validation cohort. Univariate and multi-variate logistic regression algorithms were utilized to identity independent risk factors of adenocarcinomas. A nomogram based on the risk factors was generated to predict the risk of adenocarcinomas. The discrimination ability of the nomogram was evaluated using the concordance index (C-index), its performance was calibrated using a calibration curve, and its clinical significance was evaluated using decision curves and clinical impact curves. Results Lesion size, mean CT value, vascular change and lobulation were identified as independent risk factors for adenocarcinomas. The C-index of the nomogram was 0.867 (95% CI, 0.833-0.901) in derivation cohort and 0.877 (95% CI, 0.836-0.917) in validation cohort. The calibration curve showed good agreement between the predicted and actual risks. Analysis of the decision curves and clinical impact curves revealed that the nomogram had a high standardized net benefit. Conclusions A nomogram for predicting the risk of adenocarcinomas in patients with SSNs was established in light of the 2021 WHO classification. The developed model can be adopted as a pre-operation tool to improve the surgical management of patients. Supplementary Information The online version contains supplementary material available at 10.1186/s40644-022-00483-1.
Collapse
Affiliation(s)
- Qilong Song
- Department of Radiology, Anhui Chest Hospital, Hefei, China.,Clinical College of Chest, Anhui Medical University, Hefei, China
| | - Biao Song
- Department of Radiology, Anhui Chest Hospital, Hefei, China.,Clinical College of Chest, Anhui Medical University, Hefei, China
| | - Xiaohu Li
- Department of Radiology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Bin Wang
- Department of Radiology, Anhui Chest Hospital, Hefei, China
| | - Yuan Li
- Department of Radiology, Anhui Chest Hospital, Hefei, China
| | - Wu Chen
- Department of Radiology, Anhui Chest Hospital, Hefei, China
| | - Zhaohua Wang
- Department of Radiology, Anhui Chest Hospital, Hefei, China
| | - Xu Wang
- Department of Radiology, Anhui Chest Hospital, Hefei, China
| | - Yongqiang Yu
- Department of Radiology, the First Affiliated Hospital of Anhui Medical University, Hefei, China.
| | - Xuhong Min
- Department of Radiology, Anhui Chest Hospital, Hefei, China. .,Clinical College of Chest, Anhui Medical University, Hefei, China.
| | - Dongchun Ma
- Clinical College of Chest, Anhui Medical University, Hefei, China. .,Department of Thoracic Surgery, Anhui Chest Hospital, Hefei, China.
| |
Collapse
|