1
|
Thomas QD, Vendrell JA, Khellaf L, Cavaillon S, Quantin X, Solassol J, Cabello‐Aguilar S. Artificial intelligence-driven microsatellite instability profiling reveals distinctive genetic features in patients with lung cancer. Cancer 2025; 131:e35882. [PMID: 40297960 PMCID: PMC12038786 DOI: 10.1002/cncr.35882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 02/10/2025] [Accepted: 03/24/2025] [Indexed: 04/30/2025]
Abstract
BACKGROUND Microsatellite instability (MSI) has emerged as a predictive biomarker for immunotherapy response in various cancers, but its role in non-small cell lung cancer (NSCLC) is not fully understood. METHODS The authors used the bioinformatics tool MIAmS to assess microsatellite status from next-generation sequencing (NGS) data using a tailored microsatellite score. Immunohistochemistry (IHC) assays were also performed to evaluate the correspondence between MSI and deficient mismatch repair (dMMR) status. A retrospective analysis of 1547 lung cancer patients was conducted, focusing on those with an MSI phenotype. Clinical characteristics, co-occurring molecular alterations, tumor mutation burden (TMB), and homologous recombination deficiency (HRD) status were evaluated in this subset. RESULTS Of the 1547 patients analyzed, eight (0.52%) were identified as having MSI through MIAmS, with six (0.39%) of these cases also being dMMR on IHC. All patients with dMMR had an MS score ≥2 and a history of smoking. Most patients showed loss of MLH1 and PMS2 staining on IHC. No correlation was found between MSI status and programmed death-ligand 1 expression, although all MSI patients exhibited high TMB, averaging 21.4 ± 5.6 mutations per megabase. DISCUSSION MSI/dMMR in lung cancer is exceedingly rare, affecting less than 1% of cases. NGS-based analysis combined with bioinformatics tools provides a robust method to identify MSI/dMMR patients, potentially guiding immunotherapy decisions. This comprehensive approach integrates molecular genotyping and MSI detection, offering personalized treatment options for lung cancer patients. NGS-based MSI testing is emerging as the preferred method for detecting microsatellite instability in various tumor types, including rare cancers.
Collapse
Affiliation(s)
- Quentin Dominique Thomas
- Department of Medical OncologyInstitute du Cancer de MontpellierMontpellier UniversityMontpellierFrance
- Oncogenic Pathways in Lung CancerMontpellier Cancer Research InstituteUniversity of MontpellierMontpellierFrance
| | - Julie Adèle Vendrell
- Solid Tumor LaboratoryDepartment of Pathology and OncobiologyMontpellier University Hospital MontpellierArnaud de Villeneuve HospitalMontpellierFrance
| | - Lakhdar Khellaf
- Department of PathologyInstitute du Cancer de MontpellierMontpellier UniversityMontpellierFrance
| | - Sarah Cavaillon
- Department of Medical OncologyInstitute du Cancer de MontpellierMontpellier UniversityMontpellierFrance
| | - Xavier Quantin
- Department of Medical OncologyInstitute du Cancer de MontpellierMontpellier UniversityMontpellierFrance
- Oncogenic Pathways in Lung CancerMontpellier Cancer Research InstituteUniversity of MontpellierMontpellierFrance
| | - Jérôme Solassol
- Solid Tumor LaboratoryDepartment of Pathology and OncobiologyMontpellier University Hospital MontpellierArnaud de Villeneuve HospitalMontpellierFrance
| | - Simon Cabello‐Aguilar
- Solid Tumor LaboratoryDepartment of Pathology and OncobiologyMontpellier University Hospital MontpellierArnaud de Villeneuve HospitalMontpellierFrance
- Montpellier BioInformatics for Clinical DiagnosisMolecular Medicine and Genomics PlatformMontpellier University Hospital MontpellierMontpellierFrance
| |
Collapse
|
2
|
Andrade DAP, Bonatelli M, de Paula FE, Berardinelli GN, Teixeira GR, dos Reis MT, Barbin FF, Andrade CEMDC, Aguiar VP, Hermoza AD, Hirai WY, Schmidt RL, Reis RM, dos Reis R. Implementation of the ProMisE classifier and validation of its prognostic impact in Brazilian endometrial carcinomas. Front Oncol 2024; 14:1503901. [PMID: 39735598 PMCID: PMC11671357 DOI: 10.3389/fonc.2024.1503901] [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: 09/29/2024] [Accepted: 11/25/2024] [Indexed: 12/31/2024] Open
Abstract
Purpose Molecular classification of endometrial cancer (EC) has emerged as a key approach to individualize therapy and define prognostic outcomes. This study aimed to implement the traditional ProMisE classification in a Brazilian population, compared with a molecular setting of ProMisE biomarkers, and evaluate its impact on patients' prognosis. Patient and methods A prospective cohort of 114 patients with primary EC treated at Barretos Cancer Hospital (BCH) between October 2020 and December 2022 was conducted. Pathology diagnosis, staging, treatment, and follow-up data were collected. The traditional ProMisE methodology was carried out by POLE hotspot sequencing and immunohistochemistry (IHC) for p53 and mismatch repair (MMR) proteins. We further evaluate the MMR and TP53 status by molecular approach, namely microsatellite instability (MSI) by PCR-based and TP53 mutation analysis by next-generation sequencing (NGS). The results of the 4 molecular groups in both methodologies were compared regarding agreement accuracy and survival outcomes. Results Among the 114 cases, the traditional ProMisE groups were: POLEmut 15.8%, MMRd 28.1%, p53abn 27.2%, and no specific molecular profile (NSMP) 28.9%. Considering the molecular classification approach, we observed a POLEmut group of 15.8%, MSI group of 23.7%, TP53 mutation of 27.2%, and NSMP of 33.3%. The concordance rate of both approaches was 86.8% (99/114 cases) with an overall accuracy of 0.87. Importantly, both traditional and molecular ProMisE approaches were associated with significant distinct overall survival (OS) and progression-free survival (PFS) outcomes, with POLEmut patients exhibiting a better prognosis (93.8% OS, at 24 months), whereas the p53abn having a worse survival time (68.9% of OS, at 24 months). Conclusion We reported for the first time the Brazilian profile of the ProMisE classification of endometrial cancer and demonstrated the prognostic impact of the traditional and molecular ProMisE classification on patient outcomes.
Collapse
Affiliation(s)
| | - Murilo Bonatelli
- Molecular Diagnostic Laboratory, Barretos Cancer Hospital, São Paulo, Brazil
| | | | | | - Gustavo Ramos Teixeira
- Molecular Diagnostic Laboratory, Barretos Cancer Hospital, São Paulo, Brazil
- Pathology Laboratory, Barretos Cancer Hospital, São Paulo, Brazil
- Barretos School of Health Sciences Dr. Paulo Prata, FACISB, São Paulo, Brazil
| | | | | | - Carlos Eduardo Mattos da Cunha Andrade
- Barretos School of Health Sciences Dr. Paulo Prata, FACISB, São Paulo, Brazil
- Gynecologic Oncology Department, Barretos Cancer Hospital, São Paulo, Brazil
| | | | | | - Welinton Yoshio Hirai
- Department of Epidemiology and Biostatistics, Barretos Cancer Hospital, São Paulo, Brazil
| | - Ronaldo Luís Schmidt
- Department of Surgical Oncology, Lagarto Unit, Barretos Cancer Hospital, Sergipe, Brazil
| | - Rui Manuel Reis
- Molecular Diagnostic Laboratory, Barretos Cancer Hospital, São Paulo, Brazil
- Molecular Oncology Research Center, Barretos Cancer Hospital, São Paulo, Brazil
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
| | - Ricardos dos Reis
- Gynecologic Oncology Department, Barretos Cancer Hospital, São Paulo, Brazil
| |
Collapse
|
3
|
De Marchi P, Leal LF, da Silva LS, Cavagna RDO, da Silva FAF, da Silva VD, da Silva EC, Saito AO, de Lima VCC, Reis RM. Gene expression profiles (GEPs) of immuno-oncologic pathways as predictors of response to checkpoint inhibitors in advanced NSCLC. Transl Oncol 2024; 39:101818. [PMID: 37918166 PMCID: PMC10638041 DOI: 10.1016/j.tranon.2023.101818] [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: 07/18/2023] [Revised: 10/12/2023] [Accepted: 10/27/2023] [Indexed: 11/04/2023] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) revolutionized non-small-cell lung cancer (NSCLC) treatment. However, improving patients' selection for this therapy is needed. Gene expression profile (GEP) is a promising biomarker tool. We assessed the predictive value of 48 onco-immune GEPs in an NSCLC real-world scenario. METHODS Retrospective cohort of Brazilian NSCLC patients treated with ICIs in any line. GEP was assessed in FFPE tumor tissue using the nCounter PanCancer IO360 panel, comprising 770 cancer immune genes. RESULTS The median age of the 135 patients was 61 years old, most male (57.8 %), history of smoking (83.6 %), ECOG-PS 0-1 (88.7 %), clinical stage IV (91.9 %) and adenocarcinoma (65.1 %). First-line ICI in 40 % of cases, alone or in combination with chemotherapy. The median follow-up was 28 months, overall survival after starting immunotherapy (post-immunotherapy survival - PIS) was 17.8 months, and real-world progression-free survival was 5.5 months. The GEP analysis was possible in 66 patients. We found that 14 different GEPs associated with PIS, namely IDO1, PD-L2, Cytotoxicity, Cytotoxic Cells, IFN Downstream, CTLA4, PD-L1, TIGIT, Lymphoid, Immunoproteasome, Exhausted CD8, IFN Gamma, TIS and APM. TIS and IFN-γ were the most significant GEPs associated with favorable outcomes. The median PIS for patients with high TIS expression was 29.2 versus 15.5 months (HR 0.42; 95 %CI; 0.17-0.67; p<0.05) for those with low expression. Similar results were observed for IFN-γ. CONCLUSIONS The TIS (tumor inflammation signature) and IFN-γ signatures constitute predictive biomarkers to identify patients with NSCLC patients who would possibly benefit from ICI therapies.
Collapse
Affiliation(s)
- Pedro De Marchi
- Oncoclinicas, Rio De Janeiro, Brazil; Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil.
| | - Leticia Ferro Leal
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil; Barretos School of Health Sciences, Dr. Paulo Prata - FACISB, Barretos, SP, Brazil
| | | | | | | | | | - Eduardo Ca da Silva
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil
| | | | | | - Rui Manuel Reis
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil; Life and Health Sciences Research Institute (ICVS), Medical School, University of Minho, Braga, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| |
Collapse
|
4
|
Argyropoulos K, Basu A, Park K, Zhou F, Moreira AL, Narula N. Correlation of Programmed Death-Ligand 1 Expression With Lung Adenocarcinoma Histologic and Molecular Subgroups in Primary and Metastatic Sites. Mod Pathol 2023; 36:100245. [PMID: 37307880 DOI: 10.1016/j.modpat.2023.100245] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 05/09/2023] [Accepted: 05/31/2023] [Indexed: 06/14/2023]
Abstract
Programmed death-ligand 1 (PD-L1) expression in terms of the tumor proportion score (TPS) is the main predictive biomarker approved for immunotherapy against lung nonsmall cell carcinoma. Although some studies have explored the associations between histology and PD-L1 expression in pulmonary adenocarcinoma, they have been limited in sample size and/or extent of examined histologic variables, which may have resulted in conflicting information. In this observational retrospective study, we identified primary and metastatic lung adenocarcinoma cases in the span of 5 years and tabulated the detailed histopathologic features, including pathological stage, tumor growth pattern, tumor grade, lymphovascular and pleural invasion, molecular alterations, and the associated PD-L1 expression for each case. Statistical analyses were performed to detect associations between PD-L1 and these features. Among 1658 cases, 643 were primary tumor resections, 751 were primary tumor biopsies, and 264 were metastatic site biopsies or resections. Higher TPS significantly correlated with high-grade growth patterns, grade 3 tumors, higher T and N stage, presence of lymphovascular invasion, and presence of MET and TP53 alterations, whereas lower TPS correlated with lower-grade tumors and presence of EGFR alterations. There was no difference in PD-L1 expression in matched primary and metastases, although higher TPS was observed in metastatic tumors due to the presence of high-grade patterns in these specimens. TPS showed a strong association with a histologic pattern. Higher-grade tumors had higher TPS, which is also associated with more aggressive histologic features. Tumor grade should be kept in mind when selecting cases and blocks for PD-L1 testing.
Collapse
Affiliation(s)
- Kimon Argyropoulos
- Department of Pathology, New York University Langone Health, New York, New York; Now with Memorial Sloan Kettering Cancer Center, New York, New York
| | - Atreyee Basu
- Department of Pathology, New York University Langone Health, New York, New York; Now with Tufts Medical Center, Boston, Massachusetts
| | - Kyung Park
- Department of Pathology, New York University Langone Health, New York, New York
| | - Fang Zhou
- Department of Pathology, New York University Langone Health, New York, New York
| | - Andre L Moreira
- Department of Pathology, New York University Langone Health, New York, New York.
| | - Navneet Narula
- Department of Pathology, New York University Langone Health, New York, New York
| |
Collapse
|
5
|
Tostes K, Siqueira AP, Reis RM, Leal LF, Arantes LMRB. Biomarkers for Immune Checkpoint Inhibitor Response in NSCLC: Current Developments and Applicability. Int J Mol Sci 2023; 24:11887. [PMID: 37569262 PMCID: PMC10418476 DOI: 10.3390/ijms241511887] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/20/2023] [Accepted: 07/21/2023] [Indexed: 08/13/2023] Open
Abstract
Lung cancer has the highest mortality rate among all cancer types, resulting in over 1.8 million deaths annually. Immunotherapy utilizing immune checkpoint inhibitors (ICIs) has revolutionized the treatment of non-small cell lung cancer (NSCLC). ICIs, predominantly monoclonal antibodies, modulate co-stimulatory and co-inhibitory signals crucial for maintaining immune tolerance. Despite significant therapeutic advancements in NSCLC, patients still face challenges such as disease progression, recurrence, and high mortality rates. Therefore, there is a need for predictive biomarkers that can guide lung cancer treatment strategies. Currently, programmed death-ligand 1 (PD-L1) expression is the only established biomarker for predicting ICI response. However, its accuracy and robustness are not consistently reliable. This review provides an overview of potential biomarkers currently under development or in the validation stage that hold promise in improving the classification of responders and non-responders to ICI therapy in the near future.
Collapse
Affiliation(s)
- Katiane Tostes
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-400, São Paulo, Brazil; (K.T.)
| | - Aléxia Polo Siqueira
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-400, São Paulo, Brazil; (K.T.)
| | - Rui Manuel Reis
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-400, São Paulo, Brazil; (K.T.)
- Life and Health Sciences Research Institute (ICVS), Medical School, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B’s-PT Government Associate Laboratory, 4806-909 Guimarães, Portugal
| | - Leticia Ferro Leal
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-400, São Paulo, Brazil; (K.T.)
- Barretos School of Health Sciences, Dr. Paulo Prata-FACISB, Barretos 14785-002, São Paulo, Brazil
| | | |
Collapse
|
6
|
Li N, Wan Z, Lu D, Chen R, Ye X. Long-term benefit of immunotherapy in a patient with squamous lung cancer exhibiting mismatch repair deficient/high microsatellite instability/high tumor mutational burden: A case report and literature review. Front Immunol 2023; 13:1088683. [PMID: 36703977 PMCID: PMC9871463 DOI: 10.3389/fimmu.2022.1088683] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 12/20/2022] [Indexed: 01/12/2023] Open
Abstract
Genetic mutations that render mismatch repair defective may result in microsatellite instability, which is common in colorectal carcinomas and gastric cancers as well as Lynch syndrome. Mismatch repair deficiency/high microsatellite instability (dMMR/MSI-H) predicts the tumor response to immune checkpoint inhibitors. However, few studies have evaluated the efficacy of immune checkpoint inhibitors in non-small cell lung cancer (NSCLC) patients with dMMR/MSI-H. In this work, we present a patient with advanced squamous lung cancer with dMMR/MSI-H and a high tumor mutational burden (TMB-H) who obtained a long-term benefit from immunotherapy. NSCLC patients with dMMR/MSI-H/TMB-H may thus benefit from immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Na Li
- First Clinical Medical College, Guangzhou University of Traditional Chinese, Guangzhou, China
| | - Zixuan Wan
- First Clinical Medical College, Guangzhou University of Traditional Chinese, Guangzhou, China
| | - Dongyan Lu
- Department of Oncology, The First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
| | - Ruilian Chen
- Department of Oncology, The First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, China,*Correspondence: Ruilian Chen, ; Xiaowei Ye,
| | - Xiaowei Ye
- Department of Oncology, The First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, China,*Correspondence: Ruilian Chen, ; Xiaowei Ye,
| |
Collapse
|
7
|
Shigeta N, Murakami S, Yokose T, Miyagi Y, Saito H. Case report: anaplastic lymphoma kinase ( ALK) rearranged adenocarcinoma with high level of microsatellite instability response to pembrolizumab. Front Oncol 2023; 13:1110638. [PMID: 37114121 PMCID: PMC10126330 DOI: 10.3389/fonc.2023.1110638] [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: 11/29/2022] [Accepted: 03/27/2023] [Indexed: 04/29/2023] Open
Abstract
The presence of anaplastic lymphoma kinase (ALK) rearrangement is reported to be related to the lack of efficacy of immune checkpoint inhibitors (ICIs). High levels of microsatellite instability (MSI-high) are important biomarkers of ICIs, particularly in colorectal cancer. The therapeutic effect of ICIs for MSI-high NSCLC is uncertain because of the rarity of these tumors. Here we report a case of ALK rearranged NSCLC with MSI-high. A 48-year-old male was diagnosed with lung adenocarcinoma, cT4N3M1a, stage IVA with ALK rearrangement, high PD-L1 expression with a tumor proportion score (TPS) of 100%, and MSI-high. The patient was treated with alectinib as the first-line therapy but progressed at five months with left atrial invasion re-expansion. The patient discontinued alectinib and was switched to pembrolizumab monotherapy. After two months, left atrial invasion significantly decreased. The patient continued pembrolizumab for a year without noticeable adverse events, and tumor shrinkage persisted. This case supports the efficacy of ICIs for MSI-high NSCLC, even in the presence of ALK rearrangement.
Collapse
Affiliation(s)
- Naoko Shigeta
- Department of Thoracic Oncology, Kanagawa Cancer Center, Yokohama, Japan
| | - Shuji Murakami
- Department of Thoracic Oncology, Kanagawa Cancer Center, Yokohama, Japan
- *Correspondence: Shuji Murakami,
| | - Tomoyuki Yokose
- Department of Pathology, Kanagawa Cancer Center, Yokohama, Japan
| | - Yohei Miyagi
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama, Japan
| | - Haruhiro Saito
- Department of Thoracic Oncology, Kanagawa Cancer Center, Yokohama, Japan
| |
Collapse
|
8
|
Hu Z, Liu Z, Zheng J, Peng Y, Lu X, Li J, Tan K, Cui H. Microsatellite instability-related prognostic risk score (MSI-pRS) defines a subset of lung squamous cell carcinoma (LUSC) patients with genomic instability and poor clinical outcome. Front Genet 2023; 14:1061002. [PMID: 36873930 PMCID: PMC9981642 DOI: 10.3389/fgene.2023.1061002] [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: 10/04/2022] [Accepted: 01/30/2023] [Indexed: 02/19/2023] Open
Abstract
Background: Lung squamous cell carcinoma (LUSC) shares less typical onco-drivers and target resistance, but a high overall mutation rate and marked genomic complexity. Mismatch repair (MMR) deficiency leads to microsatellite instability (MSI) and genomic instability. MSI is not an ideal option for prognosis of LUSC, whereas its function deserves exploration. Method: MSI status was classified by MMR proteins using unsupervised clustering in the TCGA-LUSC dataset. The MSI score of each sample was determined by gene set variation analysis. Intersections of the differential expression genes and differential methylation probes were classified into functional modules by weighted gene co-expression network analysis. Least absolute shrinkage and selection operator regression and stepwise gene selection were performed for model downscaling. Results: Compared with the MSI-low (MSI-L) phenotype, MSI-high (MSI-H) displayed higher genomic instability. The MSI score was decreased from MSI-H to normal samples (MSI-H > MSI-L > normal). A total of 843 genes activated by hypomethylation and 430 genes silenced by hypermethylation in MSI-H tumors were classified into six functional modules. CCDC68, LYSMD1, RPS7, and CDK20 were used to construct MSI-related prognostic risk score (MSI-pRS). Low MSI-pRS was a protective prognostic factor in all cohorts (HR = 0.46, 0.47, 0.37; p-value = 7.57e-06, 0.009, 0.021). The model contains tumor stage, age, and MSI-pRS that showed good discrimination and calibration. Decision curve analyses indicated that microsatellite instability-related prognostic risk score added extra value to the prognosis. A low MSI-pRS was negatively correlated with genomic instability. LUSC with low MSI-pRS was associated with increased genomic instability and cold immunophenotype. Conclusion: MSI-pRS is a promising prognostic biomarker in LUSC as the substitute of MSI. Moreover, we first declared that LYSMD1 contributed to genomic instability of LUSC. Our findings provided new insights in the biomarker finder of LUSC.
Collapse
Affiliation(s)
- Zixin Hu
- Beijing University of Chinese Medicine, Beijing, China.,Department of Oncology, China-Japan Friendship Hospital, Beijing, China
| | - Zhening Liu
- Beijing University of Chinese Medicine, Beijing, China.,Department of Oncology, China-Japan Friendship Hospital, Beijing, China
| | - Jiabin Zheng
- Department of Oncology, China-Japan Friendship Hospital, Beijing, China
| | - Yanmei Peng
- Department of Oncology, Fangshan Hospital, Beijing, China
| | - Xingyu Lu
- Beijing University of Chinese Medicine, Beijing, China.,Department of Oncology, China-Japan Friendship Hospital, Beijing, China
| | - Jia Li
- Beijing University of Chinese Medicine, Beijing, China.,Department of Oncology, China-Japan Friendship Hospital, Beijing, China
| | - Kexin Tan
- Beijing University of Chinese Medicine, Beijing, China.,Department of Oncology, China-Japan Friendship Hospital, Beijing, China
| | - Huijuan Cui
- Department of Oncology, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
9
|
Lesbon JCC, Garnica TK, Xavier PLP, Rochetti AL, Reis RM, Müller S, Fukumasu H. A Screening of Epigenetic Therapeutic Targets for Non-Small Cell Lung Cancer Reveals PADI4 and KDM6B as Promising Candidates. Int J Mol Sci 2022; 23:ijms231911911. [PMID: 36233212 PMCID: PMC9570250 DOI: 10.3390/ijms231911911] [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: 08/09/2022] [Revised: 09/28/2022] [Accepted: 10/04/2022] [Indexed: 11/23/2022] Open
Abstract
Despite advances in diagnostic and therapeutic approaches for lung cancer, new therapies targeting metastasis by the specific regulation of cancer genes are needed. In this study, we screened a small library of epigenetic inhibitors in non-small-cell lung cancer (NSCLC) cell lines and evaluated 38 epigenetic targets for their potential role in metastatic NSCLC. The potential candidates were ranked by a streamlined approach using in silico and in vitro experiments based on publicly available databases and evaluated by real-time qPCR target gene expression, cell viability and invasion assays, and transcriptomic analysis. The survival rate of patients with lung adenocarcinoma is inversely correlated with the gene expression of eight epigenetic targets, and a systematic review of the literature confirmed that four of them have already been identified as targets for the treatment of NSCLC. Using nontoxic doses of the remaining inhibitors, KDM6B and PADI4 were identified as potential targets affecting the invasion and migration of metastatic lung cancer cell lines. Transcriptomic analysis of KDM6B and PADI4 treated cells showed altered expression of important genes related to the metastatic process. In conclusion, we showed that KDM6B and PADI4 are promising targets for inhibiting the metastasis of lung adenocarcinoma cancer cells.
Collapse
Affiliation(s)
- Jéssika Cristina Chagas Lesbon
- Laboratory of Comparative and Translational Oncology, Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering, University of São Paulo, Duque de Caxias, 225-Jardim Elite, Pirassununga 13635-900, SP, Brazil
| | - Taismara Kustro Garnica
- Laboratory of Comparative and Translational Oncology, Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering, University of São Paulo, Duque de Caxias, 225-Jardim Elite, Pirassununga 13635-900, SP, Brazil
| | - Pedro Luiz Porfírio Xavier
- Laboratory of Comparative and Translational Oncology, Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering, University of São Paulo, Duque de Caxias, 225-Jardim Elite, Pirassununga 13635-900, SP, Brazil
| | - Arina Lázaro Rochetti
- Laboratory of Comparative and Translational Oncology, Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering, University of São Paulo, Duque de Caxias, 225-Jardim Elite, Pirassununga 13635-900, SP, Brazil
| | - Rui Manuel Reis
- Molecular Oncology Research Center, Hospital de Amor, Antenor Duarte Viléla, 1331-Dr. Paulo Prata, Barretos 14784-400, SP, Brazil
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, 4710-057 Braga, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, 4805-017 Guimarães, Portugal
| | - Susanne Müller
- Structural Genomics Consortium, Buchmann Institute for Molecular Life Sciences, Johann Wolfgang Goethe University, Max-von-Laue-Str 15-60438, 60438 Frankfurt am Main, Germany
| | - Heidge Fukumasu
- Laboratory of Comparative and Translational Oncology, Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering, University of São Paulo, Duque de Caxias, 225-Jardim Elite, Pirassununga 13635-900, SP, Brazil
- Correspondence:
| |
Collapse
|
10
|
Sato Y. Clinical utility of liquid biopsy-based companion diagnostics in the non-small-cell lung cancer treatment. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2022; 3:630-642. [PMID: 36338524 PMCID: PMC9630093 DOI: 10.37349/etat.2022.00104] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 07/12/2022] [Indexed: 12/04/2022] Open
Abstract
Recently, technological advances in the detection and biological characterization of circulating tumor DNA (ctDNA) have enabled the implementation of liquid biopsy testing into clinical practice. Methods for analysis of liquid biopsies have rapidly evolved over the past few years and have continued to advance, thus providing details about tumor biological characteristics such as tumor progression, metastasis, tumor heterogeneity, genomic mutation profile, clonal evolution, etc. In tandem with technological advances, the implementation of liquid biopsy in routine clinical settings has proceeded. In 2016, the Food and Drug Administration (FDA) approved the first ctDNA liquid biopsy test to detect epidermal growth factor receptor (EGFR) gene mutations in patients with non-small-cell lung cancer (NSCLC) as a companion diagnostic for molecular targeted drug of EGFR-tyrosine kinase inhibitor (TKI, EGFR-TKI). More recently, multigene panel assays of liquid biopsy have been approved as companion diagnostics and have been used in routine clinical settings. The estimation of blood tumor mutation burden (bTMB) to predict the efficacy of immune checkpoint inhibitor (ICI) treatment can be one of the promising approaches to liquid biopsy. The next stage of implementation of liquid biopsy for routine clinical settings is for monitoring of ctDNA after surgical treatment to predict prognosis and to detect disease relapse earlier than conventional imaging diagnosis. Its clinical utility is under assessment in several clinical trials. This review introduces recent advances in liquid biopsy methodology, the development of biomarkers, and its clinical utility in the treatment of NSCLC patients.
Collapse
Affiliation(s)
- Yoshiharu Sato
- DNA Chip Research Inc., Tokyo 105-0022, Japan,Correspondence: Yoshiharu Sato, DNA Chip Research Inc., 1-15-1 Kaigan, Minato-ku, Tokyo 105-0022, Japan.
| |
Collapse
|