Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 218
Filtrer
1.
Medicine (Baltimore) ; 103(29): e38874, 2024 Jul 19.
Article de Anglais | MEDLINE | ID: mdl-39029067

RÉSUMÉ

RATIONALE: Bone metastasis is a common metastatic mode of advanced lung cancer and poses a great threat to the survival and quality of life of patients with this disease. However, the available literature has limited treatment options for advanced lung cancer with bone metastases. PATIENTS CONCERNS: A 76-year-old married male patient was underwent CT due to cough and sputum for 1 month. On CT, space-occupying lesions were found in the left inferior lobe of the lung, as well as multiple bone metastases in the vertebral body and ilium. DIAGNOSES: Pathologic sectioning of the lung lesion after puncture revealed invasive lung adenocarcinoma, and a genetic test revealed EGFR exon 21: L858R (64.60%). INTERVENTIONS: Considering that the disease was not suitable for radiotherapy (extensive metastasis) and could not be treated with chemotherapy (poor underlying condition), the patient was given molecularly targeted therapy with osimertinib. OUTCOMES: After 10 months of standard treatment (80 mg orally, once a day), the lung lesions of the patients became significantly smaller, and the bone metastases distinctly improved. And the patient's condition has not shown any signs of rebound with the one-year follow-up. LESSONS SUBSECTIONS: In the present case, the bone metastases from lung adenocarcinoma almost completely disappeared after treatment with a single molecular targeted therapy agent, increasing the confidence in the treatment of advanced lung cancer.


Sujet(s)
Acrylamides , Adénocarcinome pulmonaire , Tumeurs osseuses , Tumeurs du poumon , Humains , Mâle , Sujet âgé , Tumeurs du poumon/secondaire , Tumeurs du poumon/anatomopathologie , Tumeurs osseuses/secondaire , Acrylamides/usage thérapeutique , Adénocarcinome pulmonaire/secondaire , Adénocarcinome pulmonaire/traitement médicamenteux , Adénocarcinome pulmonaire/anatomopathologie , Dérivés de l'aniline/usage thérapeutique , Antinéoplasiques/usage thérapeutique , Thérapie moléculaire ciblée/méthodes , Récepteurs ErbB/antagonistes et inhibiteurs , Indoles , Pyrimidines
2.
J Med Case Rep ; 18(1): 243, 2024 May 11.
Article de Anglais | MEDLINE | ID: mdl-38730499

RÉSUMÉ

BACKGROUND: An ascending aortic thrombus is exceedingly rare. Two instances have been reported in the setting of lung cancer, but only after cisplatin use, which is associated with hypercoagulability. We present the first case of a patient with lung cancer who developed an ascending aortic thrombus without structural risk factors or chemotherapy use. CASE: A 60-year-old white female with significant smoking history presented with several weeks of malaise. A chest computed tomography scan revealed a 2.2-cm right upper lobe mass. As an outpatient, right hilar lymph node immunohistochemistry (IHC) samples via endobronchial ultrasound confirmed thyroid transcription factor-1 adenocarcinoma. After the procedure, the patient endorsed dyspnea and was advised to go to the emergency department. A chest computed tomography angiography identified a new 2.4 × 1.1 × 1.1 cm thrombus within the proximal aortic arch. No pulmonary emboli or intrapulmonary shunts were identified. A hypercoagulable workup was negative. Transthoracic echocardiogram was without left ventricular thrombus, akinesis or hypokinesis, left atrial dilation, or intracardiac shunts. A lower extremity ultrasound was negative for deep vein thrombosis. Given the procedural risk, thrombectomy was deferred. The patient was transitioned to enoxaparin, and a repeat computed tomography for resolution is in process. CONCLUSION: To our knowledge, this is the only case detailing an in situ ascending aortic thrombus in the setting of lung cancer, without structural risk factors, chemotherapy use, or other hypercoagulable comorbidities. Optimal management for an aortic thrombus and malignant disease is less clear. Clinicians should be vigilant for unusual arterial thromboses in patients with high metastatic burden.


Sujet(s)
Adénocarcinome pulmonaire , Cisplatine , Tumeurs du poumon , Thrombose , Humains , Femelle , Adulte d'âge moyen , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/anatomopathologie , Cisplatine/usage thérapeutique , Thrombose/imagerie diagnostique , Adénocarcinome pulmonaire/traitement médicamenteux , Adénocarcinome pulmonaire/secondaire , Adénocarcinome pulmonaire/complications , Maladies de l'aorte/imagerie diagnostique , Anticoagulants/usage thérapeutique , Adénocarcinome/traitement médicamenteux , Adénocarcinome/complications , Énoxaparine/usage thérapeutique , Antinéoplasiques/usage thérapeutique , Antinéoplasiques/effets indésirables , Angiographie par tomodensitométrie , Aorte/imagerie diagnostique , Aorte/anatomopathologie
3.
Oncogene ; 43(28): 2215-2227, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38802647

RÉSUMÉ

Approximately 40% of patients with lung adenocarcinoma (LUAD) often develop bone metastases during the course of their disease. However, scarcely any in vivo model of LUAD bone metastasis has been established, leading to a poor understanding of the mechanisms underlying LUAD bone metastasis. Here, we established a multiorgan metastasis model via the left ventricular injection of luciferase-labeled LUAD cells into nude mice and then screened out lung metastasis (LuM) and bone metastasis (BoM) cell subpopulations. BoM cells exhibited greater stemness and epithelial-mesenchymal transition (EMT) plasticity than LuM cells and initially colonized the bone and subsequently disseminated to distant organs after being reinjected into mice. Moreover, a CD74-ROS1 fusion mutation (C6; R34) was detected in BoM cells but not in LuM cells. Mechanistically, BoM cells bearing the CD74-ROS1 fusion highly secrete the C-C motif chemokine ligand 5 (CCL5) protein by activating STAT3 signaling, recruiting macrophages in tumor microenvironment and strongly inducing M2 polarization of macrophages. BoM cell-activated macrophages produce a high level of TGF-ß1, thereby facilitating EMT and invasion of LUAD cells via TGF-ß/SMAD2/3 signaling. Targeting the CD74-ROS1/CCL5 axis with Crizotinib (a ROS1 inhibitor) and Maraviroc (a CCL5 receptor inhibitor) in vivo strongly impeded bone metastasis and secondary metastasis of BoM cells. Our findings reveal the critical role of the CD74-ROS1/STAT3/CCL5 axis in the interaction between LUAD bone metastasis cells and macrophages for controlling LUAD cell dissemination, highlighting the significance of the bone microenvironment in LUAD bone metastasis and multiorgan secondary metastasis, and suggesting that targeting CD74-ROS1 and CCL5 is a promising therapeutic strategy for LUAD bone metastasis.


Sujet(s)
Adénocarcinome pulmonaire , Tumeurs osseuses , Transition épithélio-mésenchymateuse , Tumeurs du poumon , Macrophages , Protein-tyrosine kinases , Protéines proto-oncogènes , Animaux , Humains , Souris , Tumeurs osseuses/secondaire , Tumeurs osseuses/génétique , Tumeurs osseuses/métabolisme , Tumeurs osseuses/anatomopathologie , Macrophages/métabolisme , Macrophages/anatomopathologie , Tumeurs du poumon/secondaire , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/génétique , Tumeurs du poumon/métabolisme , Adénocarcinome pulmonaire/génétique , Adénocarcinome pulmonaire/anatomopathologie , Adénocarcinome pulmonaire/secondaire , Adénocarcinome pulmonaire/métabolisme , Transition épithélio-mésenchymateuse/génétique , Protein-tyrosine kinases/métabolisme , Protein-tyrosine kinases/génétique , Protéines proto-oncogènes/génétique , Protéines proto-oncogènes/métabolisme , Antigènes de différenciation des lymphocytes B/métabolisme , Antigènes de différenciation des lymphocytes B/génétique , Chimiokine CCL5/métabolisme , Chimiokine CCL5/génétique , Antigènes d'histocompatibilité de classe II/génétique , Antigènes d'histocompatibilité de classe II/métabolisme , Souris nude , Lignée cellulaire tumorale , Microenvironnement tumoral , Protéines de fusion oncogènes/génétique , Protéines de fusion oncogènes/métabolisme , Facteur de transcription STAT-3/métabolisme , Facteur de transcription STAT-3/génétique , Transduction du signal
4.
BMC Cancer ; 24(1): 505, 2024 Apr 22.
Article de Anglais | MEDLINE | ID: mdl-38649856

RÉSUMÉ

Lung adenocarcinoma is the main type of lung cancer in women. Our previous findings have evidenced that 25-hydroxycholesterol (25-HC) promotes migration and invasion of lung adenocarcinoma cells (LAC), during which LXR as a 25-HC receptor plays an important role. Estrogen receptor beta (ERß) is a receptor of 27-hydroxycholesterol that is structurally analogous to 25-HC, but its role in the functional actions of 25-HC remained largely unknown. In this study, we demonstrated that 25-HC treatment triggered ERß expression in LAC. Knockdown of ERß inhibited 25-HC-mediated proliferation, migration and invasion, and reduced 25-HC-induced LAC metastasis in vivo. Further investigation revealed that ERß knockdown restrained the expression of TNFRSF17 (BCMA). In vivo experiments also confirmed that ERß knockdown blocked 25-HC-induced TNFRSF17 expression. TNFRSF17 knockdown also restrained 25-HC-induced proliferation, migration and invasion. Bioinformatic analysis showed that the levels of ERß and TNFRSF17 were elevated in lung adenocarcinoma, and were closely related to tumor stages and nodal metastasis status. These results suggested that 25-HC promoted the proliferation and metastasis of LAC by regulating ERß/TNFRSF17 axis.


Sujet(s)
Adénocarcinome pulmonaire , Mouvement cellulaire , Prolifération cellulaire , Récepteur bêta des oestrogènes , Hydroxycholestérols , Tumeurs du poumon , Animaux , Femelle , Humains , Mâle , Souris , Adénocarcinome pulmonaire/anatomopathologie , Adénocarcinome pulmonaire/métabolisme , Adénocarcinome pulmonaire/génétique , Adénocarcinome pulmonaire/secondaire , Lignée cellulaire tumorale , Récepteur bêta des oestrogènes/métabolisme , Récepteur bêta des oestrogènes/génétique , Régulation de l'expression des gènes tumoraux , Hydroxycholestérols/pharmacologie , Hydroxycholestérols/métabolisme , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/métabolisme , Tumeurs du poumon/secondaire , Tumeurs du poumon/génétique , Souris de lignée BALB C , Souris nude , Métastase tumorale , Transduction du signal
5.
J Cancer Res Ther ; 20(2): 658-664, 2024 Apr 01.
Article de Anglais | MEDLINE | ID: mdl-38687937

RÉSUMÉ

BACKGROUND: Leptomeningeal metastasis (LM) is a severe lung cancer complication, with potentially fatal consequences. The use of intrathecal therapy (IT) combined with systemic therapy has shown promise as a treatment approach for LM. Thus, this study aimed to evaluate the features and responses to IT combined therapy and identify determinants affecting patients with leptomeningeal metastasis resulting from lung adenocarcinoma (LM-LA). METHODS: A retrospective analysis of medical records from our hospital database was performed, covering from April 2018 to August 2022, for 37 patients diagnosed with LM-LA and treated with IT combined therapy. Patients who received IT combined therapy for LM-LA were evaluated for demographic characteristics, treatment efficacy, survival, and variables that impacted them. RESULTS: The median overall survival (mOS) of 37 patients was 16.0 months, and the survival rates at 6 and 12 months were 75.7% and 35.1%, respectively. Among the 21 patients with LM-LA who received IT combined with tyrosine kinase inhibitors (TKIs), the mOS was 17.0 months, which was significantly longer than that of patients treated with IT combined with chemotherapy (7.0 months, P = 0.010) and the best supportive care (6.0 months, P = 0.001). However, no significant survival benefit was observed in patients treated with IT combined with TKIs when compared with those treated with IT combined with PD-1 (5.0 months, P = 0.249). Multivariate analysis indicated that the combination of TKIs was an independent favorable prognostic factor for patients with LM-LA. CONCLUSION: Combination treatment is regarded as an additional option for patients with LM-LA. Compared with other combination therapies in our study, IT combined with TKI therapy provided a better survival outcome for patients with LM-LA.


Sujet(s)
Adénocarcinome pulmonaire , Protocoles de polychimiothérapie antinéoplasique , Injections rachidiennes , Tumeurs du poumon , Humains , Mâle , Femelle , Adulte d'âge moyen , Adénocarcinome pulmonaire/traitement médicamenteux , Adénocarcinome pulmonaire/anatomopathologie , Adénocarcinome pulmonaire/secondaire , Adénocarcinome pulmonaire/mortalité , Études rétrospectives , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/mortalité , Pronostic , Sujet âgé , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Adulte , Taux de survie , Tumeurs des méninges/secondaire , Tumeurs des méninges/traitement médicamenteux , Tumeurs des méninges/mortalité , Résultat thérapeutique , Inhibiteurs de protéines kinases/usage thérapeutique , Inhibiteurs de protéines kinases/administration et posologie , Méningite carcinomateuse/secondaire , Méningite carcinomateuse/traitement médicamenteux , Méningite carcinomateuse/mortalité , Association thérapeutique , Sujet âgé de 80 ans ou plus
6.
In Vivo ; 38(3): 1498-1502, 2024.
Article de Anglais | MEDLINE | ID: mdl-38688648

RÉSUMÉ

BACKGROUND/AIM: Spontaneous regression (SR) of cancer, which indicates the natural disappearance of malignant tumors, is rare. Little is known about the mechanisms underlying SR; however, immunological reactions, infections, injuries, and medications have been presumed. Among previously reported cases of SR, lung cancer cases have been extremely limited. CASE REPORT: Here, we present a case of lymph node metastasis exacerbation after SR of a primary adenocarcinoma following a biopsy. After complete disappearance of the primary site tumor, metastatic lymph nodes in the mediastinum gradually increased in size as a single lesion. Local treatment with resection and radiotherapy was effective for this metastasis, without recurrence for >3 years. CONCLUSION: This is an interesting case of SR of pulmonary adenocarcinoma with inconsistent features in the primary and metastatic lesions. When physicians encounter exacerbation of metastatic sites with SR of the primary site in lung cancer, local intervention may be considered as a curative treatment.


Sujet(s)
Adénocarcinome pulmonaire , Évolution de la maladie , Tumeurs du poumon , Métastase lymphatique , Humains , Adénocarcinome/anatomopathologie , Adénocarcinome pulmonaire/anatomopathologie , Adénocarcinome pulmonaire/secondaire , Biopsie , Tumeurs du poumon/anatomopathologie , Noeuds lymphatiques/anatomopathologie , Métastase lymphatique/anatomopathologie , Régression tumorale spontanée , Tomodensitométrie
7.
Anticancer Res ; 44(5): 2159-2170, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38677751

RÉSUMÉ

BACKGROUND/AIM: Both mesonephric adenocarcinoma (MA) and mesonephric-like adenocarcinoma (MLA) express thyroid transcription factor 1 (TTF1). TTF1 is also considered a highly sensitive and specific diagnostic marker for primary lung adenocarcinoma (PLA). However, distinguishing PLA from pulmonary metastatic MA/MLA (PMM) based on the expression of TTF1 alone can be difficult. This study aimed to investigate the expression of TTF1 and paired box 8 (PAX8) and assess their value in distinguishing PMM from PLA. PATIENTS AND METHODS: We reviewed the electronic medical records and pathology slides of eight PMM cases. We conducted immunostaining for TTF1 and PAX8 in 6, 8, and 21 cases of primary MA/MLA, PMM, and PLA, respectively. RESULTS: Two patients with stage IB uterine MLA developed lung metastases at 5 and 57 months after hysterectomy. Solitary pulmonary nodules were suspected to be primary lung cancer in two patients. Compared to primary tumors, all matched PMMs exhibited reduced TTF1 immunoreactivity. In contrast, the majority of PLAs showed uniform and intense TTF1 expression. All except one PMM exhibited diffuse and strong PAX8 expression, while only one PLA showed focal and weak PAX8 expression. CONCLUSION: Immunostaining for TTF1 and PAX8 can help in distinguishing PMM from PLA in the diagnosis of pulmonary lesions detected in patients with a history of MA/MLA.


Sujet(s)
Adénocarcinome pulmonaire , Adénocarcinome , Marqueurs biologiques tumoraux , Protéines de liaison à l'ADN , Immunohistochimie , Tumeurs du poumon , Facteur de transcription PAX-8 , Femelle , Humains , Mâle , Adénocarcinome/métabolisme , Adénocarcinome/diagnostic , Adénocarcinome/secondaire , Adénocarcinome/anatomopathologie , Adénocarcinome pulmonaire/métabolisme , Adénocarcinome pulmonaire/diagnostic , Adénocarcinome pulmonaire/anatomopathologie , Adénocarcinome pulmonaire/secondaire , Marqueurs biologiques tumoraux/métabolisme , Diagnostic différentiel , Tumeurs du poumon/métabolisme , Tumeurs du poumon/diagnostic , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/secondaire , Facteur de transcription PAX-8/métabolisme , Facteur-1 de transcription de la thyroïde/métabolisme , Facteurs de transcription/métabolisme
9.
Folia Neuropathol ; 62(1): 108-112, 2024.
Article de Anglais | MEDLINE | ID: mdl-38174674

RÉSUMÉ

Lung adenocarcinoma remains one of the most frequent and deadly tumour entities. Early-stage lung adenocarcinoma is extremely difficult to detect and is also easy to recur or metastasize after treatment. Since the new adenocarcinoma classification was presented in 2011, several studies have shown that patients with solid and/or micropapillary (S/MP) predominant patterns showed a worse prognosis. Here we report the case of a 54-year-old woman who was diagnosed with stage Ib lung adenocarcinoma with S/MP components and developed an isolated brain oligometastasis after resection and adjuvant therapy. A craniocerebral operation was performed, combined with radiotherapy and targeted therapy, and the patient eventually achieved a good quality of life. Our work reviews the clinical features of lung cancer complicated with S/MP components, the relationship between MP and epidermal growth factor receptor (EGFR) mutation, as well as treatment strategies for such a patient with postoperative brain oligometastasis of lung adenocarcinoma complicated with EGFR Exon19del mutation.


Sujet(s)
Adénocarcinome pulmonaire , Tumeurs du cerveau , Tumeurs du poumon , Humains , Adulte d'âge moyen , Femelle , Tumeurs du cerveau/secondaire , Adénocarcinome pulmonaire/anatomopathologie , Adénocarcinome pulmonaire/secondaire , Tumeurs du poumon/anatomopathologie
10.
J Cell Physiol ; 238(11): 2710-2723, 2023 11.
Article de Anglais | MEDLINE | ID: mdl-37733753

RÉSUMÉ

Dysregulation of polycomb group (PcG) proteins that mediate epigenetic gene silencing contributes to tumorigenesis. As core components of the polycomb repressive complex 1 (PRC1), chromobox (CBX) proteins recognize H3K27me3 to recruit PRC1 to maintain a repressive transcriptional state. However, the individual biological functions of these CBX proteins in tumorigenesis warrant in-depth investigation. In this study, we analyzed the mRNA expression of CBX family genes across multiple cancers using The Cancer Genome Atlas data and found different expression patterns of the five CBX genes in different types of cancer. This analyses together with the result of immunohistochemistry indicated that CBX8 expression was significantly higher in lung adenocarcinoma (LUAD) tissues compared to adjacent nontumor tissues. Overexpression approaches demonstrated that CBX8 facilitated LUAD cell proliferation and migration in vitro. Consistently, CBX8 knockdown reduced LUAD cell proliferation and migration in both cell culture and mouse models. RNA sequencing combined with real-time RT-PCR assays revealed CDKN2C and SCEL as target genes of CBX8. Furthermore, chromatin immunoprecipitation assays indicated that CBX8 directly bound to the promoters of CDKN2C and SCEL to establish H2AK119ub. CBX8 depletion reduced the enrichment of H2AK119ub on CDKN2C and SCEL promoters. Moreover, depletion of CDKN2C and SCEL restored the repressed growth and invasion ability of LUAD cells caused by CBX8 knockdown. These findings demonstrate that CBX8 promotes LUAD growth and metastasis through the transcriptional repression of CDKN2C and SCEL. Our study uncovers the oncogenic role of CBX8 in LUAD progression and provides a new target for the diagnosis and therapy of LUAD.


Sujet(s)
Adénocarcinome pulmonaire , Protéines de transport , Inhibiteur p18 de kinase cycline-dépendante , Régulation de l'expression des gènes tumoraux , Tumeurs du poumon , Complexe répresseur Polycomb-1 , Animaux , Humains , Souris , Cellules A549 , Adénocarcinome pulmonaire/génétique , Adénocarcinome pulmonaire/secondaire , Carcinogenèse/génétique , Lignée cellulaire tumorale , Prolifération cellulaire/génétique , Régulation de l'expression des gènes tumoraux/génétique , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Complexe répresseur Polycomb-1/génétique , Complexe répresseur Polycomb-1/métabolisme , Protéines du groupe Polycomb/génétique , Protéines du groupe Polycomb/métabolisme , Transcription génétique , Inhibiteur p18 de kinase cycline-dépendante/génétique , Protéines de transport/génétique
11.
Cancer Cell ; 41(9): 1621-1636.e8, 2023 09 11.
Article de Anglais | MEDLINE | ID: mdl-37595587

RÉSUMÉ

Brain metastasis of lung cancer causes high mortality, but the exact mechanisms underlying the metastasis remain unclear. Here we report that vascular pericytes derived from CD44+ lung cancer stem cells (CSCs) in lung adenocarcinoma (ADC) potently cause brain metastases through the G-protein-coupled receptor 124 (GPR124)-enhanced trans-endothelial migration (TEM). CD44+ CSCs in perivascular niches generate the majority of vascular pericytes in lung ADC. CSC-derived pericyte-like cells (Cd-pericytes) exhibit remarkable TEM capacity to effectively intravasate into the vessel lumina, survive in the circulation, extravasate into the brain parenchyma, and then de-differentiate into tumorigenic CSCs to form metastases. Cd-pericytes uniquely express GPR124 that activates Wnt7-ß-catenin signaling to enhance TEM capacity of Cd-pericytes for intravasation and extravasation, two critical steps during tumor metastasis. Furthermore, selective disruption of Cd-pericytes, GPR124, or the Wnt7-ß-catenin signaling markedly reduces brain and liver metastases of lung ADC. Our findings uncover an unappreciated cellular and molecular paradigm driving tumor metastasis.


Sujet(s)
Adénocarcinome pulmonaire , Tumeurs du cerveau , Tumeurs du poumon , Humains , Adénocarcinome pulmonaire/secondaire , bêta-Caténine , Tumeurs du cerveau/secondaire , Cadmium , Antigènes CD44 , Poumon , Tumeurs du poumon/anatomopathologie , Péricytes , Récepteurs couplés aux protéines G
12.
Rev Mal Respir ; 40(4): 359-365, 2023 Apr.
Article de Français | MEDLINE | ID: mdl-36868976

RÉSUMÉ

INTRODUCTION: Gastrointestinal (GI) metastases in lung cancer rarely occur. CASE REPORT: We report here the case of a 43-year-old male active smoker who was admitted to our hospital for cough, abdominal pain and melena. Initial investigations revealed poorly differentiated adenocarcinoma of the superior-right lobe of the lung: positive for thyroid transcription factor-1 and negative for protein p40 and for antigen CD56, with peritoneal, adrenal and cerebral metastasis, as well as anemia requiring major transfusion support. Over 50% of cells were positive for PDL-1, and ALK gene rearrangement was detected. GI endoscopy showed a large ulcerated nodular lesion of the genu superius with active intermittent bleeding, as well as an undifferentiated carcinoma with positivity for CK AE1/AE3 and TTF-1, and negativity for CD117, corresponding to metastatic invasion originating from lung carcinoma. Palliative immunotherapy with pembrolizumab was proposed, followed by targeted therapy with brigatinib. Gastrointestinal bleeding was controlled with a single 8Gy dose of haemostatic radiotherapy. CONCLUSION: GI metastases are rare in lung cancer and present nonspecific symptoms and signs but no characteristic endoscopic features. GI bleeding is a common revelatory complication. Pathological and immunohistological findings are critical to diagnosis. Local treatment is usually guided by the occurrence of complications. In addition to surgery and systemic therapies, palliative radiotherapy may contribute to bleeding control. However, it must be used cautiously, given a present-day lack of evidence and the pronounced radiosensitivity of certain gastrointestinal tract segments.


Sujet(s)
Adénocarcinome pulmonaire , Tumeurs du duodénum , Hémorragie gastro-intestinale , Tumeurs du poumon , Adénocarcinome pulmonaire/anatomopathologie , Adénocarcinome pulmonaire/secondaire , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/thérapie , Métastase tumorale , Hémorragie gastro-intestinale/étiologie , Hémorragie gastro-intestinale/radiothérapie , Tumeurs du duodénum/complications , Tumeurs du duodénum/secondaire , Tumeurs du duodénum/chirurgie , Humains , Adulte , Mâle , Toux/étiologie , Douleur abdominale/étiologie , Méléna/étiologie , Résultat thérapeutique
13.
Cancer Discov ; 12(2): 562-585, 2022 02.
Article de Anglais | MEDLINE | ID: mdl-34561242

RÉSUMÉ

SMARCA4/BRG1 encodes for one of two mutually exclusive ATPases present in mammalian SWI/SNF chromatin remodeling complexes and is frequently mutated in human lung adenocarcinoma. However, the functional consequences of SMARCA4 mutation on tumor initiation, progression, and chromatin regulation in lung cancer remain poorly understood. Here, we demonstrate that loss of Smarca4 sensitizes club cell secretory protein-positive cells within the lung in a cell type-dependent fashion to malignant transformation and tumor progression, resulting in highly advanced dedifferentiated tumors and increased metastatic incidence. Consistent with these phenotypes, Smarca4-deficient primary tumors lack lung lineage transcription factor activities and resemble a metastatic cell state. Mechanistically, we show that Smarca4 loss impairs the function of all three classes of SWI/SNF complexes, resulting in decreased chromatin accessibility at lung lineage motifs and ultimately accelerating tumor progression. Thus, we propose that the SWI/SNF complex via Smarca4 acts as a gatekeeper for lineage-specific cellular transformation and metastasis during lung cancer evolution. SIGNIFICANCE: We demonstrate cell-type specificity in the tumor-suppressive functions of SMARCA4 in the lung, pointing toward a critical role of the cell-of-origin in driving SWI/SNF-mutant lung adenocarcinoma. We further show the direct effects of SMARCA4 loss on SWI/SNF function and chromatin regulation that cause aggressive malignancy during lung cancer evolution.This article is highlighted in the In This Issue feature, p. 275.


Sujet(s)
Adénocarcinome pulmonaire/génétique , Transformation cellulaire néoplasique , Helicase/génétique , Tumeurs du poumon/génétique , Métastase tumorale , Protéines nucléaires/génétique , Facteurs de transcription/génétique , Adénocarcinome pulmonaire/secondaire , Animaux , Modèles animaux de maladie humaine , Humains , Tumeurs du poumon/anatomopathologie , Souris
14.
Thorac Cancer ; 13(2): 202-209, 2022 01.
Article de Anglais | MEDLINE | ID: mdl-34812577

RÉSUMÉ

BACKGROUND: Rapid intraoperative diagnosis for unconfirmed pulmonary tumor is extremely important for determining the optimal surgical procedure (lobectomy or sublobar resection). Attempts to diagnose malignant tumors using mass spectrometry (MS) have recently been described. This study evaluated the usefulness of MS and artificial intelligence (AI) for differentiating primary lung adenocarcinoma (PLAC) and colorectal metastatic pulmonary tumor. METHODS: Pulmonary samples from 40 patients who underwent pulmonary resection for PLAC (20 tumors, 20 normal lungs) or pulmonary metastases originating from colorectal metastatic pulmonary tumor (CRMPT) (20 tumors, 20 normal lungs) were collected and analyzed retrospectively by probe electrospray ionization-MS. AI using random forest (RF) algorithms was employed to evaluate the accuracy of each combination. RESULTS: The accuracy of the machine learning algorithm applied using RF to distinguish malignant tumor (PLAC or CRMPT) from normal lung was 100%. The algorithms offered 97.2% accuracy in differentiating PLAC and CRMPT. CONCLUSIONS: MS combined with an AI system demonstrated high accuracy not only for differentiating cancer from normal tissue, but also for differentiating between PLAC and CRMPT with a short working time. This method shows potential for application as a support tool facilitating rapid intraoperative diagnosis to determine the surgical procedure for pulmonary resection.


Sujet(s)
Adénocarcinome pulmonaire/diagnostic , Adénocarcinome pulmonaire/chirurgie , Intelligence artificielle , Tumeurs colorectales/diagnostic , Tumeurs du poumon/diagnostic , Tumeurs du poumon/chirurgie , Spectrométrie de masse/méthodes , Adénocarcinome pulmonaire/secondaire , Sujet âgé , Tumeurs colorectales/anatomopathologie , Femelle , Humains , Tumeurs du poumon/secondaire , Mâle
15.
Medicine (Baltimore) ; 100(39): e27289, 2021 Oct 01.
Article de Anglais | MEDLINE | ID: mdl-34596125

RÉSUMÉ

RATIONALE: Transformation to small cell lung cancer (SCLC) is one of the mechanisms of resistance to epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs). However, no standard treatment is available after the transformation. In addition, gastric metastasis of primary lung cancer is rarely observed; thus, little is known about its metastatic characteristics. PATIENT CONCERNS: A 58-year-old male patient was treated with gefitinib (0.25 g /day) as the 1st line treatment due of recurrence after surgical resection for EGFR exon 19 mutation pulmonary adenocarcinoma. However, he experienced recurrence with positive T790 M, and osimertinib (80 mg/day) was administered as the 2nd line therapy. DIAGNOSIS: One year and 6 months after osimertinib initiation, he complained of stomachache, and a diagnostic gastroscopy biopsy confirmed small cell lung cancer in the gastric body, indicating osimertinib-induced phenotypic transformation. INTERVENTIONS AND OUTCOMES: The patient was treated with etoposide and platinum chemotherapy and maintenance therapy with osimertinib. Finally, the patient achieved a partial response after 4 cycles. LESSONS: Timely second biopsies should be considered in the diagnosis of phenotypic transformation. After transformation, chemotherapeutic treatment with etoposide and platinum and maintenance therapy with osimertinib inhibited the progression of the disease.


Sujet(s)
Adénocarcinome pulmonaire/anatomopathologie , Transformation cellulaire néoplasique , Tumeurs du poumon/anatomopathologie , Carcinome pulmonaire à petites cellules/anatomopathologie , Tumeurs de l'estomac/secondaire , Adénocarcinome pulmonaire/traitement médicamenteux , Adénocarcinome pulmonaire/secondaire , Résistance aux médicaments antinéoplasiques , Géfitinib/usage thérapeutique , Humains , Tumeurs du poumon/traitement médicamenteux , Mâle , Adulte d'âge moyen , Inhibiteurs de protéines kinases/usage thérapeutique
16.
BMC Cancer ; 21(1): 1168, 2021 Oct 30.
Article de Anglais | MEDLINE | ID: mdl-34717570

RÉSUMÉ

BACKGROUND: Intramedullary metastasis (IMM) is a rare disease with poor prognosis. The incidence of IMMs has increased, which has been linked to improved systemic treatment in many cancers. Surgery and/or radiotherapy are the most commonly used treatments; only small-sample retrospective studies and case reports on stereotactic body radiotherapy (SBRT) have reported acceptable results in terms of local control and clinical improvement, with no reported toxicity. Thus, we performed this monocentric retrospective study on five cases treated with SBRT for IMMs, which we supplemented with a systematic review of the literature. METHODS: We included all patients treated for IMM with SBRT. The target tumor volume, progression-free survival, prescription patterns in SBRT, survival without neurological deficit, neurological functional improvement after treatment, and overall survival were determined. RESULTS: Five patients treated with a median dose of 30 Gy in a median number of fractions of 5 (prescribed at a median isodose of 86%) included. The median follow-up duration was 23 months. Two patients showed clinical improvement. Three patients remained stable. Radiologically, 25% of patients had complete response and 50% had stable disease. No significant treatment-related toxicity was observed. CONCLUSION: SBRT appears to be a safe, effective, and rapid treatment option for palliative patients.


Sujet(s)
Radiochirurgie , Tumeurs de la moelle épinière/radiothérapie , Tumeurs de la moelle épinière/secondaire , Adénocarcinome/radiothérapie , Adénocarcinome/secondaire , Adénocarcinome pulmonaire/radiothérapie , Adénocarcinome pulmonaire/secondaire , Adulte , Sujet âgé , Tumeurs du sein/anatomopathologie , Établissements de cancérologie , Néphrocarcinome/radiothérapie , Néphrocarcinome/secondaire , Fractionnement de la dose d'irradiation , Femelle , Études de suivi , France , Humains , Tumeurs du rein/anatomopathologie , Tumeurs du poumon/anatomopathologie , Mâle , Mélanome/radiothérapie , Mélanome/secondaire , Adulte d'âge moyen , Survie sans progression , Études rétrospectives , Tumeurs cutanées/anatomopathologie , Tumeurs de la moelle épinière/imagerie diagnostique , Tumeurs de la moelle épinière/mortalité , Charge tumorale
17.
Cancer Res ; 81(23): 5904-5918, 2021 12 01.
Article de Anglais | MEDLINE | ID: mdl-34654723

RÉSUMÉ

Invasive mucinous lung adenocarcinoma (IMA) is a subtype of lung adenocarcinoma with a strong invasive ability. IMA frequently carries "undruggable" KRAS mutations, highlighting the need for new molecular targets and therapies. Nuclear receptor HNF4α is abnormally enriched in IMA, but the potential of HNF4α to be a therapeutic target for IMA remains unknown. Here, we report that P2 promoter-driven HNF4α expression promotes IMA growth and metastasis. Mechanistically, HNF4α transactivated lncRNA BC200, which acted as a scaffold for mRNA binding protein FMR1. BC200 promoted the ability of FMR1 to bind and regulate stability of cancer-related mRNAs and HNF4α mRNA, forming a positive feedback circuit. Mycophenolic acid, the active metabolite of FDA-approved drug mycophenolate mofetil, was identified as an HNF4α antagonist exhibiting anti-IMA activities in vitro and in vivo. This study reveals the role of a HNF4α-BC200-FMR1-positive feedback loop in promoting mRNA stability during IMA progression and metastasis, providing a targeted therapeutic strategy for IMA. SIGNIFICANCE: Growth and metastatic progression of invasive mucinous lung adenocarcinoma can be restricted by targeting HNF4α, a critical regulator of a BC200-FMR1-mRNA stability axis.


Sujet(s)
Adénocarcinome pulmonaire/secondaire , Adénocarcinome mucineux/secondaire , Protéine du syndrome X fragile/métabolisme , Régulation de l'expression des gènes tumoraux , Facteur nucléaire hépatocytaire HNF-4/métabolisme , Tumeurs du poumon/anatomopathologie , ARN long non codant/génétique , Adénocarcinome pulmonaire/génétique , Adénocarcinome pulmonaire/métabolisme , Adénocarcinome mucineux/génétique , Adénocarcinome mucineux/métabolisme , Animaux , Apoptose , Marqueurs biologiques tumoraux/génétique , Marqueurs biologiques tumoraux/métabolisme , Prolifération cellulaire , Rétrocontrôle physiologique , Femelle , Protéine du syndrome X fragile/génétique , Facteur nucléaire hépatocytaire HNF-4/génétique , Humains , Tumeurs du poumon/génétique , Tumeurs du poumon/métabolisme , Souris , Souris de lignée BALB C , Souris nude , Invasion tumorale , Pronostic , Taux de survie , Cellules cancéreuses en culture , Tests d'activité antitumorale sur modèle de xénogreffe
18.
Zhongguo Fei Ai Za Zhi ; 24(8): 598-604, 2021 Aug 20.
Article de Chinois | MEDLINE | ID: mdl-34344501

RÉSUMÉ

BACKGROUND: Anaplastic lymphoma kinase (ALK) is an important therapeutic target for advanced non-small cell lung cancer (NSCLC). In recent years, with the emergence of several ALK tyrosine kinase inhibitors (TKI), the overall survival (OS) of ALK fusion positive patients is gradually extended. This paper reports the treatment of a late stage non-small cell lung cancer (NSCLC) patient with ALK fusion positive for more than 5 years, and analyzes the treatment process and effect evaluation, so as to provide experience for the follow-up treatment of patients. METHODS: The diagnosis and treatment process of a patient with advanced ALK fusion mutation positive lung cancer admitted to the third ward of Department of oncology, Chifeng hospital, Inner Mongolia on July 3, 2015 was retrospectively analyzed. RESULTS: A 42 years old male patient was admitted to our department on July 3, 2015 for "intermittent cough, chest tightness for 2 months, diagnosed with lung adenocarcinoma for 1 day". Imaging examination showed a space occupying lesion in the left lower lobe of the lung, accompanied by mediastinal lymphadenopathy and left encapsulated pleural effusion. Bronchoscopic pathology showed non-small cell carcinoma, and adenocarcinoma was tentatively suggested. DIAGNOSIS: left lower lobe adenocarcinoma T1bN2M1a stage IV. Fluorescence in situ hybridization (FISH) indicated the translocation of ALK (2p23) chromosome. After 2 cycles of docetaxel+cisplatin (DP) regimens chemotherapy, disease progression occurred, so we used 6 cycles of pemetrexed+carboplatin to apply combination chemotherapy, 4 cycles of pemetrexed monotherapy were used after that. The efficacy evaluation: PR. On April 9, 2016, the patient was treated with crizotinib. In August 2019, multiple intracranial metastases were found and whole brain radiotherapy was given. Since September 4, 2019, oral administration of nsatinib has been carried out. As of March 1, 2021, the patients were followed up well. CONCLUSIONS: The advanced ALK fusion positive lung adenocarcinoma patients, though the first-line and the second-line chemotherapy, and the follwing application of ALK-TKI treatment, has procured a total OS has reached 68 months, and the current follow-up is good.


Sujet(s)
Adénocarcinome pulmonaire , Kinase du lymphome anaplasique , Tumeurs du cerveau , Tumeurs du poumon , Adénocarcinome pulmonaire/traitement médicamenteux , Adénocarcinome pulmonaire/génétique , Adénocarcinome pulmonaire/radiothérapie , Adénocarcinome pulmonaire/secondaire , Adulte , Kinase du lymphome anaplasique/génétique , Antinéoplasiques/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Tumeurs du cerveau/traitement médicamenteux , Tumeurs du cerveau/génétique , Tumeurs du cerveau/radiothérapie , Tumeurs du cerveau/secondaire , Carboplatine/administration et posologie , Cisplatine/administration et posologie , Crizotinib/administration et posologie , Évolution de la maladie , Docetaxel/administration et posologie , Humains , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Mâle , Mutation , Protéines de fusion oncogènes/génétique , Pémétrexed/administration et posologie , Inhibiteurs de protéines kinases/usage thérapeutique , Analyse de survie , Résultat thérapeutique
19.
Cell Death Dis ; 12(7): 680, 2021 07 05.
Article de Anglais | MEDLINE | ID: mdl-34226519

RÉSUMÉ

It has been recently reported that CD38 expressed on tumor cells of multiple murine and human origins could be upregulated in response to PD-L1 antibody therapy, which led to dysfunction of tumor-infiltrating CD8+ T immune cells due to increasing the production of adenosine. However, the role of tumor expressed-CD38 on neoplastic formation and progression remains elusive. In the present study, we aimed to delineate the molecular and biochemical function of the tumor-associated CD38 in lung adenocarcinoma progression. Our clinical data showed that the upregulation of tumor-originated CD38 was correlated with poor survival of lung cancer patients. Using multiple in vitro assays we found that the enzymatic activity of tumor expressed-CD38 facilitated lung cancer cell migration, proliferation, colony formation, and tumor development. Consistently, our in vivo results showed that inhibition of the enzymatic activity or antagonizing the enzymatic product of CD38 resulted in the similar inhibition of tumor proliferation and metastasis as CD38 gene knock-out or mutation. At biochemical level, we further identified that cADPR, the mainly hydrolytic product of CD38, was responsible for inducing the opening of TRPM2 iron channel leading to the influx of intracellular Ca2+ and then led to increasing levels of NRF2 while decreasing expression of KEAP1 in lung cancer cells. These findings suggested that malignant lung cancer cells were capable of using cADPR catalyzed by CD38 to facilitate tumor progression, and blocking the enzymatic activity of CD38 could be represented as an important strategy for preventing tumor progression.


Sujet(s)
Antigènes CD38/métabolisme , Adénocarcinome pulmonaire/enzymologie , ADP-ribose cyclique/métabolisme , Tumeurs du poumon/enzymologie , Glycoprotéines membranaires/métabolisme , Cellules A549 , Antigènes CD38/génétique , Adénocarcinome pulmonaire/génétique , Adénocarcinome pulmonaire/secondaire , Animaux , Signalisation calcique , Carcinome pulmonaire de Lewis/enzymologie , Carcinome pulmonaire de Lewis/génétique , Carcinome pulmonaire de Lewis/anatomopathologie , Mouvement cellulaire , Prolifération cellulaire , Bases de données génétiques , Évolution de la maladie , Femelle , Régulation de l'expression des gènes tumoraux , Humains , Protéine-1 de type kelch associée à ECH/métabolisme , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Mâle , Glycoprotéines membranaires/génétique , Souris de lignée C57BL , Adulte d'âge moyen , Facteur-2 apparenté à NF-E2/métabolisme , Invasion tumorale , Canaux cationiques TRPM/métabolisme
20.
Proc Natl Acad Sci U S A ; 118(30)2021 07 27.
Article de Anglais | MEDLINE | ID: mdl-34301865

RÉSUMÉ

In mammals, the KRAS locus encodes two protein isoforms, KRAS4A and KRAS4B, which differ only in their C terminus via alternative splicing of distinct fourth exons. Previous studies have shown that whereas KRAS expression is essential for mouse development, the KRAS4A isoform is expendable. Here, we have generated a mouse strain that carries a terminator codon in exon 4B that leads to the expression of an unstable KRAS4B154 truncated polypeptide, hence resulting in a bona fide Kras4B-null allele. In contrast, this terminator codon leaves expression of the KRAS4A isoform unaffected. Mice selectively lacking KRAS4B expression developed to term but died perinatally because of hypertrabeculation of the ventricular wall, a defect reminiscent of that observed in embryos lacking the Kras locus. Mouse embryonic fibroblasts (MEFs) obtained from Kras4B-/- embryos proliferated less than did wild-type MEFs, because of limited expression of KRAS4A, a defect that can be compensated for by ectopic expression of this isoform. Introduction of the same terminator codon into a KrasFSFG12V allele allowed expression of an endogenous KRAS4AG12V oncogenic isoform in the absence of KRAS4B. Exposure of Kras+/FSF4AG12V4B- mice to Adeno-FLPo particles induced lung tumors with complete penetrance, albeit with increased latencies as compared with control Kras+/FSFG12V animals. Moreover, a significant percentage of these mice developed proximal metastasis, a feature seldom observed in mice expressing both mutant isoforms. These results illustrate that expression of the KRAS4AG12V mutant isoform is sufficient to induce lung tumors, thus suggesting that selective targeting of the KRAS4BG12V oncoprotein may not have significant therapeutic consequences.


Sujet(s)
Adénocarcinome pulmonaire/secondaire , Tumeurs du poumon/anatomopathologie , Protéines proto-oncogènes p21(ras)/physiologie , Adénocarcinome pulmonaire/génétique , Adénocarcinome pulmonaire/métabolisme , Animaux , Apoptose , Prolifération cellulaire , Femelle , Humains , Tumeurs du poumon/génétique , Tumeurs du poumon/métabolisme , Mâle , Souris , Souris de lignée C57BL , Souris knockout , Mutation , Isoformes de protéines , Cellules cancéreuses en culture , Tests d'activité antitumorale sur modèle de xénogreffe
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE