1
|
Ghazali N, Garassino MC, Leighl NB, Bestvina CM. Immunotherapy in advanced, KRAS G12C-mutant non-small-cell lung cancer: current strategies and future directions. Ther Adv Med Oncol 2025; 17:17588359251323985. [PMID: 40093982 PMCID: PMC11907553 DOI: 10.1177/17588359251323985] [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: 11/15/2024] [Accepted: 02/05/2025] [Indexed: 03/19/2025] Open
Abstract
Kirsten rat sarcoma (KRAS) mutations are present in up to 25% of non-small-cell lung cancer (NSCLC). KRAS G12C is the most common type of mutation, representing approximately half of the cases in KRAS-mutant NSCLC. Mutations in KRAS activate the RAF-MEK-ERK pathway, leading to increased cell proliferation and survival. Recent advances in drug development have led to the approval of KRAS G12C inhibitors sotorasib and adagrasib. This review explores the emerging therapeutic strategies in KRAS G12C-mutant NSCLC, including dual checkpoint blockade and combinations with checkpoint inhibitors, with a focus on the setting of advanced disease.
Collapse
Affiliation(s)
- Nadia Ghazali
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | | | - Natasha B Leighl
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Christine M Bestvina
- Department of Medicine, The University of Chicago Medicine, 5841 S. Maryland Avenue, MC 2115, Chicago, IL 60637, USA
| |
Collapse
|
2
|
Trelford CB, Shepherd TG. Insights into targeting LKB1 in tumorigenesis. Genes Dis 2025; 12:101402. [PMID: 39735555 PMCID: PMC11681833 DOI: 10.1016/j.gendis.2024.101402] [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: 02/14/2024] [Revised: 06/03/2024] [Accepted: 06/22/2024] [Indexed: 12/31/2024] Open
Abstract
Genetic alterations to serine-threonine kinase 11 (STK11) have been implicated in Peutz-Jeghers syndrome and tumorigenesis. Further exploration of the context-specific roles of liver kinase B1 (LKB1; encoded by STK11) observed that it regulates AMP-activated protein kinase (AMPK) and AMPK-related kinases. Given that both migration and proliferation are enhanced with the loss of LKB1 activity combined with the prevalence of STK11 genetic alterations in cancer biopsies, LKB1 was marked as a tumor suppressor. However, the role of LKB1 in tumorigenesis is paradoxical as LKB1 activates autophagy and reactive oxygen species scavenging while dampening anoikis, which contribute to cancer cell survival. Due to the pro-tumorigenic properties of LKB1, targeting LKB1 pathways is now relevant for cancer treatment. With the recent successes of targeting LKB1 signaling in research and clinical settings, and enhanced cytotoxicity of chemical compounds in LKB1-deficient tumors, there is now a need for LKB1 inhibitors. However, validating LKB1 inhibitors is challenging as LKB1 adaptor proteins, nucleocytoplasmic shuttling, and splice variants all manipulate LKB1 activity. Furthermore, STE-20-related kinase adaptor protein (STRAD) and mouse protein 25 dictate LKB1 cellular localization and kinase activity. For these reasons, prior to assessing the efficacy and potency of pharmacological candidates, the functional status of LKB1 needs to be defined. Therefore, to improve the understanding of LKB1 in physiology and oncology, this review highlights the role of LKB1 in tumorigenesis and addresses the therapeutic relevancy of LKB1 inhibitors.
Collapse
Affiliation(s)
- Charles B. Trelford
- The Mary & John Knight Translational Ovarian Cancer Research Unit, London Regional Cancer Program, London, ON N6A 4L6, Canada
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 3K7, Canada
| | - Trevor G. Shepherd
- The Mary & John Knight Translational Ovarian Cancer Research Unit, London Regional Cancer Program, London, ON N6A 4L6, Canada
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 3K7, Canada
- Department of Oncology, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 3K7, Canada
- Department of Obstetrics and Gynaecology, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 3K7, Canada
| |
Collapse
|
3
|
Ramos-Ramírez M, Caballe-Pérez E, Lucio-Lozada J, Romero-Nuñez E, Castillo-Ruiz C, Dorantes-Sánchez L, Flores-Estrada D, Recondo G, Barrios-Bernal P, Cabrera-Miranda L, Bravo-Dominguez H, Hernández-Pedro N, Arrieta O. Immunomodulatory role of oncogenic alterations in non-small cell lung cancer: a review of implications for immunotherapy. Cancer Metastasis Rev 2025; 44:30. [PMID: 39915358 DOI: 10.1007/s10555-025-10245-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 01/16/2025] [Indexed: 03/28/2025]
Abstract
Immune checkpoint inhibitors (ICIs) have improved clinical outcomes in patients with non-small cell lung cancer (NSCLC) lacking targetable oncogenic alterations. However, their efficacy in individuals with such genomic alterations remains heterogeneous and poorly understood. In detail, certain oncogenic alterations in TP53, EGFR (uncommon mutations), KRAS (G12C), BRAF (non-V600E), MET (amplifications), FGFR1 and FGFR4, actively modify MAPK, PI3K, and STING signaling, thus remodeling tumoral immune phenotype and are associated with high TMB counts, enriched T lymphocyte tumor infiltration, and high expression of antigen-presenting molecules, supporting their consideration as part of the eligibility criteria for ICIs treatment. Nonetheless, other oncogenic alterations are associated with an immunosuppressive TME, low TMB counts, and downregulation of targetable immune checkpoints, in which novel therapeutic approaches are currently being tested to overcome their intrinsic resistance. In this context, this review discusses the fundamental mechanisms by which frequent driver alterations affect ICIs efficacy in patients with NSCLC, and outlines their prognostic relevance in the era of immunotherapy.
Collapse
Affiliation(s)
- Maritza Ramos-Ramírez
- Thoracic Oncology Functional Unit (UFOT), Instituto Nacional de Cancerología, (INCAN), Mexico City , (CDMX), Mexico
| | - Enrique Caballe-Pérez
- Thoracic Oncology Functional Unit (UFOT), Instituto Nacional de Cancerología, (INCAN), Mexico City , (CDMX), Mexico
| | - José Lucio-Lozada
- Personalized Medicine Laboratory, Instituto Nacional de Cancerología (INCAN), Mexico City, (CDMX), Mexico
| | - Eunice Romero-Nuñez
- Personalized Medicine Laboratory, Instituto Nacional de Cancerología (INCAN), Mexico City, (CDMX), Mexico
| | - Cesar Castillo-Ruiz
- Personalized Medicine Laboratory, Instituto Nacional de Cancerología (INCAN), Mexico City, (CDMX), Mexico
| | - Lorena Dorantes-Sánchez
- Personalized Medicine Laboratory, Instituto Nacional de Cancerología (INCAN), Mexico City, (CDMX), Mexico
| | - Diana Flores-Estrada
- Thoracic Oncology Functional Unit (UFOT), Instituto Nacional de Cancerología, (INCAN), Mexico City , (CDMX), Mexico
| | - Gonzalo Recondo
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Pedro Barrios-Bernal
- Personalized Medicine Laboratory, Instituto Nacional de Cancerología (INCAN), Mexico City, (CDMX), Mexico
| | - Luis Cabrera-Miranda
- Thoracic Oncology Functional Unit (UFOT), Instituto Nacional de Cancerología, (INCAN), Mexico City , (CDMX), Mexico
| | - Heyman Bravo-Dominguez
- Thoracic Oncology Functional Unit (UFOT), Instituto Nacional de Cancerología, (INCAN), Mexico City , (CDMX), Mexico
| | - Norma Hernández-Pedro
- Personalized Medicine Laboratory, Instituto Nacional de Cancerología (INCAN), Mexico City, (CDMX), Mexico.
- Thoracic Oncology Functional Unit (UFOT), Instituto Nacional de Cancerología, (INCAN), Mexico City , (CDMX), Mexico.
| | - Oscar Arrieta
- Personalized Medicine Laboratory, Instituto Nacional de Cancerología (INCAN), Mexico City, (CDMX), Mexico.
- Thoracic Oncology Functional Unit (UFOT), Instituto Nacional de Cancerología, (INCAN), Mexico City , (CDMX), Mexico.
| |
Collapse
|
4
|
Barberis M, Rappa A, de Marinis F, Pelosi G, Guerini Rocco E, Zhan Y, Tiana G. A rationale for the poor response to alectinib in a patient with adenocarcinoma of the lung harbouring a STRN-ALK fusion by artificial intelligence and molecular modelling: a case report. Transl Lung Cancer Res 2024; 13:3807-3814. [PMID: 39830770 PMCID: PMC11736610 DOI: 10.21037/tlcr-24-667] [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: 07/31/2024] [Accepted: 11/16/2024] [Indexed: 01/22/2025]
Abstract
Background Non-small cell lung cancers (NSCLCs) with ALK fusions are effectively treated with ALK tyrosine kinase inhibitors (TKIs). The widespread use of next-generation sequencing (NGS) assays to study the molecular profile of NSCLCs, can identify rare fusion partners of ALK. Therapy decisions are made without considering which fusion partner is present and its potential oncogenic properties. However clinical and experimental studies have shown that the 5' partner of kinase fusion variants could have a biological role in the response to targeted therapies. The objective of this report was to study the impact of a rare fusion partner of ALK on the specific TKI treatment with an in silico molecular modelling evaluating the efficiency of the protein-ligand site. Case Description Here we describe a case of a stage IV lung adenocarcinoma with a rare striatin STRN-ALK fusion with a Partial Response of short duration to alectinib and no response to lorlatinib at progression. We investigated a computational molecular model of the protein translated from the translocated gene to suggest a mechanistic explanation for the clinical findings. Conclusions Our model calculations suggested that the effect of the translocation was to induce the dimerization of ALK into a complex that distorted the binding pocket, which is the same for alectinib, lorlatinib and crizotinib. The distortion of the binding pocket observed in the simulations also provides a rationale to explain the different variations of efficacy of alectinib, lorlatinib and crizotinib caused by the translocation. Our observations suggest that molecular modelling based on artificial intelligence (AI) tools may offer potential predictive information in fusions with rare partner genes. Further retrospective and prospective studies are warranted to demonstrate the predictive robustness of these tools.
Collapse
Affiliation(s)
- Massimo Barberis
- Department of Experimental Oncology, Istituto Europeo di Oncologia, IRCCS, Milano, Italy
| | - Alessandra Rappa
- Division of Pathology, Istituto Europeo di Oncologia, IRCCS, Milano, Italy
| | - Filippo de Marinis
- Division of Thoracic Oncology, Istituto Europeo di Oncologia, IRCCS, Milano, Italy
| | - Giuseppe Pelosi
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | | | - Yinxiu Zhan
- Department of Experimental Oncology, Istituto Europeo di Oncologia, IRCCS, Milano, Italy
| | - Guido Tiana
- Department of Physics and Center for Complexity and Biosystems, Università degli Studi di Milano and INFN, Milano, Italy
| |
Collapse
|
5
|
Jiang H, Sun N, Li R, Guan W, Zhu Y, Xie Z, Xie X, Liu M, Lin X, Zhou C. Evaluating Safety and Clinical Activity of Front-line Treatment with Cadonilimab plus Chemotherapy in Advanced/Metastatic Nonsmall Cell Lung Cancer Harboring STK11 Genetic Aberration: A Protocol of Phase II Study. Clin Lung Cancer 2024; 25:e393-e396. [PMID: 39095236 DOI: 10.1016/j.cllc.2024.07.006] [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: 05/18/2024] [Revised: 07/02/2024] [Accepted: 07/08/2024] [Indexed: 08/04/2024]
Abstract
BACKGROUND Cadonilimab is a first-in-class bispecific PD-1/CTLA-4 antibody. Serine/threonine kinase (STK11) mutation was shown to be related to low PD-L1 expression and objective response rate (ORR) in nonsmall cell lung cancer (NSCLC), resulting in poor progression-free survival (PFS) and overall survival (OS). Herein, we hypothesized that combining cadonilimab with chemotherapy could enhance antitumor immunity and extend survival in these patients. Consequently, we designed this study to explore the clinical activity and safety of cadonilimab combined with chemotherapy in patients with advanced/metastatic NSCLC harboring STK11 alteration. TRIAL DESIGN This single-center, open-label, single-arm phase II trial is conducted at the first affiliated hospital of Guangzhou Medical University. Treatment-naïve advanced/metastatic NSCLC patients harboring STK11 mutation will be enrolled in this study. Eligible patients will receive either cadonilimab (10mg/kg on Day 1) plus pemetrexed (500 mg/m2) and carboplatin (AUC = 5) for nonsquamous NSCLC or abraxane (100 mg/m2) and carboplatin (AUC = 5) for squamous NSCLC for 4 cycles, followed by maintenance therapy (cadonilimab plus pemetrexed or abraxane). The treatment will be discontinued when disease progression, intolerability to cadonilimab, and/or chemotherapy occurs. Measurable lesions were assessed according to the Response Evaluation Criteria in Solid Tumors (1.1). The main endpoint is ORR and safety. Subordinate endpoints include PFS, disease control rate, and duration of response. RESULTS The study commenced enrolment in September 2023, with preliminary findings regarding the primary endpoint anticipated by January 2025.
Collapse
Affiliation(s)
- Huixin Jiang
- Department of Respiratory and Critical Care Medicine, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, PR China
| | - Ni Sun
- Department of Respiratory and Critical Care Medicine, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, PR China
| | - Ru Li
- Department of Respiratory and Critical Care Medicine, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, PR China
| | - Wenhui Guan
- Department of Respiratory and Critical Care Medicine, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, PR China
| | - Yue Zhu
- Department of Respiratory and Critical Care Medicine, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, PR China
| | - Zhanhong Xie
- Department of Respiratory and Critical Care Medicine, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, PR China
| | - Xiaohong Xie
- Department of Respiratory and Critical Care Medicine, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, PR China
| | - Ming Liu
- Department of Respiratory and Critical Care Medicine, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, PR China
| | - Xinqing Lin
- Department of Respiratory and Critical Care Medicine, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, PR China.
| | - Chengzhi Zhou
- Department of Respiratory and Critical Care Medicine, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, PR China.
| |
Collapse
|
6
|
Moorthi S, Paguirigan A, Itagi P, Ko M, Pettinger M, Hoge AC, Nag A, Patel NA, Wu F, Sather C, Levine KM, Fitzgibbon MP, Thorner AR, Anderson GL, Ha G, Berger AH. The genomic landscape of lung cancer in never-smokers from the Women's Health Initiative. JCI Insight 2024; 9:e174643. [PMID: 39052387 PMCID: PMC11385083 DOI: 10.1172/jci.insight.174643] [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] [Received: 08/14/2023] [Accepted: 07/19/2024] [Indexed: 07/27/2024] Open
Abstract
Over 200,000 individuals are diagnosed with lung cancer in the United States every year, with a growing proportion of cases, especially lung adenocarcinoma, occurring in individuals who have never smoked. Women over the age of 50 comprise the largest affected demographic. To understand the genomic drivers of lung adenocarcinoma and therapeutic response in this population, we performed whole genome and/or whole exome sequencing on 73 matched lung tumor/normal pairs from postmenopausal women who participated in the Women's Health Initiative. Somatic copy number alterations showed little variation by smoking status, suggesting that aneuploidy may be a general characteristic of lung cancer regardless of smoke exposure. Similarly, clock-like and APOBEC mutation signatures were prevalent but did not differ in tumors from smokers and never-smokers. However, mutations in both EGFR and KRAS showed unique allelic differences determined by smoking status that are known to alter tumor response to targeted therapy. Mutations in the MYC-network member MGA were more prevalent in tumors from smokers. Fusion events in ALK, RET, and ROS1 were absent, likely due to age-related differences in fusion prevalence. Our work underscores the profound effect of smoking status, age, and sex on the tumor mutational landscape and identifies areas of unmet medical need.
Collapse
Affiliation(s)
| | | | - Pushpa Itagi
- Human Biology Division
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Minjeong Ko
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Mary Pettinger
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Anna Ch Hoge
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Anwesha Nag
- Center for Cancer Genomics, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Neil A Patel
- Center for Cancer Genomics, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Feinan Wu
- Genomics and Bioinformatics Shared Resource, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Cassie Sather
- Genomics and Bioinformatics Shared Resource, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Kevin M Levine
- Human Biology Division
- Division of Hematology and Oncology, Department of Medicine and
| | - Matthew P Fitzgibbon
- Genomics and Bioinformatics Shared Resource, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Aaron R Thorner
- Center for Cancer Genomics, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Garnet L Anderson
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Gavin Ha
- Human Biology Division
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Department of Genome Sciences, University of Washington, Seattle, Washington, USA
| | - Alice H Berger
- Human Biology Division
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Department of Genome Sciences, University of Washington, Seattle, Washington, USA
| |
Collapse
|
7
|
Kimbrough EO, Marin-Acevedo JA, Drusbosky LM, Mooradian A, Zhao Y, Manochakian R, Lou Y. Sex- and Age-Associated Differences in Genomic Alterations among Patients with Advanced Non-Small Cell Lung Cancer (NSCLC). Cancers (Basel) 2024; 16:2366. [PMID: 39001428 PMCID: PMC11240325 DOI: 10.3390/cancers16132366] [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: 04/11/2024] [Revised: 06/15/2024] [Accepted: 06/21/2024] [Indexed: 07/16/2024] Open
Abstract
Genomic mutations impact non-small cell lung cancer (NSCLC) biology. The influence of sex and age on the distribution of these alterations is unclear. We analyzed circulating-tumor DNA from individuals with advanced NSCLC from March 2018 to October 2020. EGFR, KRAS, ALK, ROS1, BRAF, NTRK, ERBB2, RET, MET, PIK3CA, STK11, and TP53 alterations were assessed. We evaluated the differences by sex and age (<70 and ≥70) using Fisher's exact test. Of the 34,277 samples, 30,790 (89.83%) had a detectable mutation and 19,923 (58.12%) had an alteration of interest. The median age of the ctDNA positive population was 69 (18-102), 16,756 (54.42%) were female, and 28,835 (93.65%) had adenocarcinoma. Females had more alterations in all the assessed EGFR mutations, KRAS G12C, and ERBB2 ex20 ins. Males had higher numbers of MET amp and alterations in STK11 and TP53. Patients <70 years were more likely to have alterations in EGFR exon 19 del/exon 20 ins/T790M, KRAS G12C/D, ALK, ROS1, BRAF V600E, ERBB2 Ex20ins, MET amp, STK11, and TP53. Individuals ≥70 years were more likely to have alterations in EGFR L861Q, MET exon 14 skipping, and PIK3CA. We provided evidence of sex- and age-associated differences in the distribution of genomic alterations in individuals with advanced NSCLC.
Collapse
Affiliation(s)
- ErinMarie O Kimbrough
- Division of Hematology and Oncology, Mayo Clinic, Jacksonville, FL 32224, USA
- Department of Hematology and Oncology, Division of Internal Medicine, Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN 46202, USA
| | - Julian A Marin-Acevedo
- Department of Hematology and Oncology, Division of Internal Medicine, Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN 46202, USA
| | | | - Ariana Mooradian
- Division of Hematology and Oncology, Mayo Clinic, Jacksonville, FL 32224, USA
- Division of Hematology and Medical Oncology, University of Florida, Jacksonville, FL 32209, USA
| | - Yujie Zhao
- Division of Hematology and Oncology, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Rami Manochakian
- Division of Hematology and Oncology, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Yanyan Lou
- Division of Hematology and Oncology, Mayo Clinic, Jacksonville, FL 32224, USA
| |
Collapse
|
8
|
Trelford CB, Shepherd TG. LKB1 biology: assessing the therapeutic relevancy of LKB1 inhibitors. Cell Commun Signal 2024; 22:310. [PMID: 38844908 PMCID: PMC11155146 DOI: 10.1186/s12964-024-01689-5] [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: 03/22/2024] [Accepted: 05/28/2024] [Indexed: 06/10/2024] Open
Abstract
Liver Kinase B1 (LKB1), encoded by Serine-Threonine Kinase 11 (STK11), is a master kinase that regulates cell migration, polarity, proliferation, and metabolism through downstream adenosine monophosphate-activated protein kinase (AMPK) and AMPK-related kinase signalling. Since genetic screens identified STK11 mutations in Peutz-Jeghers Syndrome, STK11 mutants have been implicated in tumourigenesis labelling it as a tumour suppressor. In support of this, several compounds reduce tumour burden through upregulating LKB1 signalling, and LKB1-AMPK agonists are cytotoxic to tumour cells. However, in certain contexts, its role in cancer is paradoxical as LKB1 promotes tumour cell survival by mediating resistance against metabolic and oxidative stressors. LKB1 deficiency has also enhanced the selectivity and cytotoxicity of several cancer therapies. Taken together, there is a need to develop LKB1-specific pharmacological compounds, but prior to developing LKB1 inhibitors, further work is needed to understand LKB1 activity and regulation. However, investigating LKB1 activity is strenuous as cell/tissue type, mutations to the LKB1 signalling pathway, STE-20-related kinase adaptor protein (STRAD) binding, Mouse protein 25-STRAD binding, splicing variants, nucleocytoplasmic shuttling, post-translational modifications, and kinase conformation impact the functional status of LKB1. For these reasons, guidelines to standardize experimental strategies to study LKB1 activity, associate proteins, spliced isoforms, post-translational modifications, and regulation are of upmost importance to the development of LKB1-specific therapies. Therefore, to assess the therapeutic relevancy of LKB1 inhibitors, this review summarizes the importance of LKB1 in cell physiology, highlights contributors to LKB1 activation, and outlines the benefits and risks associated with targeting LKB1.
Collapse
Affiliation(s)
- Charles B Trelford
- The Mary &, John Knight Translational Ovarian Cancer Research Unit, London Regional Cancer Program, 790 Commissioners Road East, Room A4‑921, London, ON, N6A 4L6, Canada.
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada.
| | - Trevor G Shepherd
- The Mary &, John Knight Translational Ovarian Cancer Research Unit, London Regional Cancer Program, 790 Commissioners Road East, Room A4‑921, London, ON, N6A 4L6, Canada
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- Department of Oncology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- Department of Obstetrics and Gynaecology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| |
Collapse
|
9
|
Ricciuti B, Garassino MC. Precision Immunotherapy for STK11/KEAP1-Mutant NSCLC. J Thorac Oncol 2024; 19:877-882. [PMID: 38849167 DOI: 10.1016/j.jtho.2024.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 01/30/2024] [Accepted: 03/04/2024] [Indexed: 06/09/2024]
Affiliation(s)
- Biagio Ricciuti
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts; Department of Medicine, Harvard Medical School, Boston, Massachusetts.
| | | |
Collapse
|
10
|
Nishio H, Matsuda R, Iwata T, Yamagami W. Gastric-type adenocarcinoma of the uterine cervix: clinical features and future directions. Jpn J Clin Oncol 2024; 54:516-520. [PMID: 38366663 PMCID: PMC11075765 DOI: 10.1093/jjco/hyae019] [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: 11/13/2023] [Accepted: 01/25/2024] [Indexed: 02/18/2024] Open
Abstract
The concept of gastric-type mucinous carcinoma of the uterine cervix (GAS) has been accepted worldwide because of its aggressive clinical behaviour and the absence of high-risk human papilloma virus infection. The World Health Organization (WHO) 2020 classification divides cervical tumours into two categories: human papilloma virus-associated and human papilloma virus-independent. Hence, GAS is now classified as an human papilloma virus-independent gastric type. Because clinical studies have reported that GAS is refractory to conventional treatments such as chemotherapy and radiotherapy, especially at an advanced stage, and has aggressive features with widespread dissemination to unusual sites, such as the omentum, peritoneum and distant organs, it is urgent to establish new treatment strategies by comparing the molecular profiles of human papilloma virus-associated adenocarcinomas. A series of genetic mutations characteristic to GAS encourage the development of future treatment strategies such as targeted therapy and immunotherapy.
Collapse
Affiliation(s)
- Hiroshi Nishio
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Risa Matsuda
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Takashi Iwata
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Wataru Yamagami
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
11
|
Xu K, Lu W, Yu A, Wu H, He J. Effect of the STK11 mutation on therapeutic efficacy and prognosis in patients with non-small cell lung cancer: a comprehensive study based on meta-analyses and bioinformatics analyses. BMC Cancer 2024; 24:491. [PMID: 38632512 PMCID: PMC11025184 DOI: 10.1186/s12885-024-12130-y] [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] [Received: 11/23/2023] [Accepted: 03/15/2024] [Indexed: 04/19/2024] Open
Abstract
BACKGROUND This study aimed to systematically analyze the effect of a serine/threonine kinase (STK11) mutation (STK11mut) on therapeutic efficacy and prognosis in patients with non-small cell lung cancer (NSCLC). METHODS Candidate articles were identified through a search of relevant literature published on or before April 1, 2023, in PubMed, Embase, Cochrane Library, CNKI and Wanfang databases. The extracted and analyzed data included the hazard ratios (HRs) of PFS and OS, the objective response rate (ORR) of immune checkpoint inhibitors (ICIs), and the positive rates of PD-L1 expression. The HR of PFS and OS and the merged ratios were calculated using a meta-analysis. The correlation between STK11mut and clinical characteristics was further analyzed in NSCLC datasets from public databases. RESULTS Fourteen retrospective studies including 4317 patients with NSCLC of whom 605 had STK11mut were included. The meta-analysis revealed that the ORR of ICIs in patients with STK11mut was 10.1% (95%CI 0.9-25.2), and the positive rate of PD-L1 expression was 41.1% (95%CI 25.3-57.0). STK11mut was associated with poor PFS (HR = 1.49, 95%CI 1.28-1.74) and poor OS (HR = 1.44, 95%CI 1.24-1.67). In the bioinformatics analysis, PFS and OS in patients with STK11 alterations were worse than those in patients without alterations (p < 0.001, p = 0.002). Nutlin-3a, 5-fluorouracil, and vinorelbine may have better sensitivity in patients with STK11mut than in those with STK11wt. CONCLUSIONS Patients with STK11-mutant NSCLC had low PD-L1 expression and ORR to ICIs, and their PFS and OS were worse than patients with STK11wt after comprehensive treatment. In the future, more reasonable systematic treatments should be explored for this subgroup of patients with STK11-mutant NSCLC.
Collapse
Affiliation(s)
- Ke Xu
- Clinical Medical College, Chengdu Medical College, Chengdu, Sichuan, China
- Department of Oncology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Weinan Lu
- Clinical Medical College, Chengdu Medical College, Chengdu, Sichuan, China
- Department of Gastroenterology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Airu Yu
- Clinical Medical College, Chengdu Medical College, Chengdu, Sichuan, China
| | - Hongwei Wu
- Clinical Medical College, Chengdu Medical College, Chengdu, Sichuan, China
| | - Jie He
- Clinical Medical College, Chengdu Medical College, Chengdu, Sichuan, China.
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China.
| |
Collapse
|
12
|
Oudart JB, Garinet S, Leger C, Barlesi F, Mazières J, Jeannin G, Audigier-Valette C, Morot-Sibilot D, Langlais A, Amour E, Mathiot N, Birsen G, Blons H, Wislez M. STK11/LKB1 alterations worsen the poor prognosis of KRAS mutated early-stage non-squamous non-small cell lung carcinoma, results based on the phase 2 IFCT TASTE trial. Lung Cancer 2024; 190:107508. [PMID: 38428265 DOI: 10.1016/j.lungcan.2024.107508] [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: 12/18/2023] [Revised: 02/14/2024] [Accepted: 02/18/2024] [Indexed: 03/03/2024]
Abstract
BACKGROUND STK11/LKB1 mutations have been associated with primary resistance to PD-1 axis inhibitors and poor prognosis in advanced KRAS-mutant lung adenocarcinoma. This study aimed to assess the prognostic significance of STK11/LKB1 alterations in localized non-squamous non-small cell lung carcinoma (non-sq NSCLC). PATIENTS AND METHODS Surgical samples from patients undergoing complete resection for stage IIa, IIb, or IIIa (N2 excluded) non-sq NSCLC in the randomized adjuvant phase II trial (NCT00775385 IFCT-1801 TASTE trial) were examined. Patients received either standard chemotherapy (Pemetrexed Cisplatin) or personalized treatment based on EGFR mutation (Erlotinib) and ERCC1 expression. Tumor molecular profiles were analyzed using targeted NGS and correlated with overall survival (OS) and disease-free survival (DFS), adjusting for relevant clinical variables. Additionally, interactions between treatment groups and molecular alterations on OS, PD-L1 expression, and tumor-circulating DNA in post-operative plasma samples were evaluated. RESULTS Among 134 patients (predominantly male smokers with adenocarcinoma), KRAS mutations were associated with shorter DFS (HR: 1.95, 95 % CI: 1.1-3.4, p = 0.02) and OS (HR: 2.32, 95 % CI: 1.2-4.6, p = 0.014). Isolated STK11/LKB1 mutations (n = 18) did not significantly impact DFS or OS. However, within KRAS-mutated samples (n = 53), patients with concurrent STK11/LKB1 mutations (n = 10) exhibited significantly shorter DFS (HR: 3.85, CI: 1.5-10.2, p = 0.006) and a trend towards shorter OS (HR: 1.80, CI: 0.6-5.3, p = 0.28). No associations were found between PD-L1 expression, other gene mutations, progression-free survival (PFS), or OS. CONCLUSION This analysis reinforces KRAS mutations as predictive factors for relapse and poor survival in localized non-sq NSCLC. Furthermore, the presence of concomitant STK11/LKB1 mutations exacerbated the prognosis within the KRAS-mutated subset. These findings emphasize the clinical relevance of these molecular markers and their potential impact on treatment strategies in non-sq NSCLC.
Collapse
Affiliation(s)
- Jean Baptiste Oudart
- Assistance publique-hôpitaux de Paris, European Georges Pompidou Hospital, Department of Biochemistry, Somatic Oncology and pharmacogenomics Unit, Paris Cancer Institute CARPEM, Paris, France
| | - Simon Garinet
- Assistance publique-hôpitaux de Paris, European Georges Pompidou Hospital, Department of Biochemistry, Somatic Oncology and pharmacogenomics Unit, Paris Cancer Institute CARPEM, Paris, France; Centre de Recherche des Cordeliers, INSERM, CNRS SNC 5096, Sorbonne Université, Université Paris Cité, Paris, France
| | - Caroline Leger
- Assistance publique-hôpitaux de Paris, European Georges Pompidou Hospital, Department of Biochemistry, Somatic Oncology and pharmacogenomics Unit, Paris Cancer Institute CARPEM, Paris, France
| | - Fabrice Barlesi
- Medical Oncology Department, Gustave Roussy, Villejuif, France
| | - Julien Mazières
- Thoracic Oncology Department, CHU Toulouse - Hôpital Larrey, Toulouse, France
| | | | | | | | | | - Elodie Amour
- French Cooperative Thoracic Intergroup (IFCT), Paris, France
| | - Nathalie Mathiot
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Team Inflammation, Complement, and Cancer, Université Paris cité, Paris, France
| | - Gary Birsen
- Oncology Thoracic Unit Pulmonology Department, AP-HP, Hôpital Cochin, F-75014 Paris, France
| | - Hélène Blons
- Assistance publique-hôpitaux de Paris, European Georges Pompidou Hospital, Department of Biochemistry, Somatic Oncology and pharmacogenomics Unit, Paris Cancer Institute CARPEM, Paris, France; Centre de Recherche des Cordeliers, INSERM, CNRS SNC 5096, Sorbonne Université, Université Paris Cité, Paris, France
| | - Marie Wislez
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Team Inflammation, Complement, and Cancer, Université Paris cité, Paris, France; Oncology Thoracic Unit Pulmonology Department, AP-HP, Hôpital Cochin, F-75014 Paris, France.
| |
Collapse
|
13
|
Stratmann JA, Althoff FC, Doebel P, Rauh J, Trummer A, Hünerlitürkoglu AN, Frost N, Yildirim H, Christopoulos P, Burkhard O, Büschenfelde CMZ, Becker von Rose A, Alt J, Aries SP, Webendörfer M, Kaldune S, Uhlenbruch M, Tritchkova G, Waller CF, Rittmeyer A, Hoffknecht P, Braess J, Kopp HG, Grohé C, Schäfer M, Schumann C, Griesinger F, Kuon J, Sebastian M, Reinmuth N. Sotorasib in KRAS G12C-mutated non-small cell lung cancer: A multicenter real-world experience from the compassionate use program in Germany. Eur J Cancer 2024; 201:113911. [PMID: 38377774 DOI: 10.1016/j.ejca.2024.113911] [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: 01/07/2024] [Accepted: 01/29/2024] [Indexed: 02/22/2024]
Abstract
BACKGROUND Sotorasib is a first-in-class KRAS p.G12C-inhibitor that has entered clinical trials in pretreated patients with non-small cell lung cancer (NSCLC) in 2018. First response rates were promising in the CodeBreaK trials. It remains unclear whether response to sotorasib and outcomes differ in a real-world setting when including patients underrepresented in clinical trials. METHODS Patients with KRAS p.G12C-mutated advanced or metastatic NSCLC received sotorasib within the German multicenter sotorasib compassionate use program between 2020 to 2022. Data on efficacy, tolerability, and survival were analyzed in the full cohort and in subgroups of special interest such as co-occurring mutations and across PD-L1 expression levels. RESULTS We analyzed 163 patients who received sotorasib after a median of two treatment lines (range, 0 to 7). Every fourth patient had a poor performance status and 38% had brain metastases (BM). The objective response rate was 38.7%. The median overall survival was 9.8 months (95% CI, 6.5 to not reached). Median real-world (rw) progression-free survival was 4.8 months (9% CI, 3.9 to 5.9). Dose reductions and permanent discontinuation were necessary in 35 (21.5%) and 7 (4.3%) patients, respectively. Efficacy seems to be influenced by PD-L1 expression and a co-occurring KEAP1 mutation. KEAP1 was associated with an inferior survival. Other factors such as BM, STK11, and TP53 mutations had no impact on response and survival. CONCLUSION First results from a real-world population confirm promising efficacy of sotorasib for the treatment of advanced KRAS p.G12C-mutated NSCLC. Patients with co-occurring KEAP1 mutations seem to derive less benefit.
Collapse
Affiliation(s)
- Jan A Stratmann
- Goethe University Frankfurt, University Hospital, Department of Internal Medicine II, Hematology/Oncology, Frankfurt am Main, Germany
| | - Friederike C Althoff
- Goethe University Frankfurt, University Hospital, Department of Internal Medicine II, Hematology/Oncology, Frankfurt am Main, Germany.
| | - Paula Doebel
- Goethe University Frankfurt, University Hospital, Department of Internal Medicine II, Hematology/Oncology, Frankfurt am Main, Germany
| | - Jacqueline Rauh
- Hospital Witten, Medical Specialist Center of Internal Medicine, Witten, Germany
| | - Arne Trummer
- Municipal Clinic Braunschweig, Medical Specialist Center of Hematology/Oncology, Braunschweig, Germany
| | | | - Nikolaj Frost
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Department of Infectious Diseases and Pulmonary Medicine, Berlin, Germany
| | | | - Petros Christopoulos
- University Hospital Heidelberg, Thoraxklinik Heidelberg, Heidelberg, Germany; Translational Lung Research Center Heidelberg, member of the German Center for Lung Research (DZL), Germany
| | - Oswald Burkhard
- Medical Specialist Center of Internal Medicine, Hematology Oncology, Palliative Medicine in Worms, Worms, Germany
| | | | - Aaron Becker von Rose
- Technical University Munich, Klinikum rechts der Isar, Medical Department for Haematology and Oncology, Munich, Germany
| | - Jürgen Alt
- University Medical Center Mainz, Department of Internal Medicine III, Mainz, Germany
| | | | - Maximilian Webendörfer
- University Hospital Essen, West German Cancer Center, Department of Medical Oncology, Essen, Germany
| | - Stefan Kaldune
- RoMed Clinic Rosenheim, Department of Hematology/Oncology, Rosenheim, Germany
| | - Mark Uhlenbruch
- Kaiserswerther Diakonie Florence-Nightingale-Hospital Düsseldorf, Department of Hematology/Oncology, Düsseldorf, Germany
| | - Guergana Tritchkova
- University Hospital Dresden, TU Dresden, Clinic for Internal Medicine I, Dresden, Germany
| | - Cornelius F Waller
- University of Freiburg, University Medical Centre Freiburg, Department of Haematology, Oncology and Stem Cell Transplantation, Freiburg, Germany
| | | | - Petra Hoffknecht
- Niels-Stensen-Kliniken Franziskus Hospital Harderberg, Department of Hematology/Oncology, Hardenberg, Germany
| | - Jan Braess
- Hospital Barmherzige Brüder Regensburg, Regensburg, Germany
| | - Hans-Georg Kopp
- Robert Bosch Center for Tumor Diseases, Robert-Bosch-Hospital, Stuttgart, Germany
| | | | - Monica Schäfer
- Helios Klinikum Emil von Behring GmbH, Lungenklinik Heckeshorn, Berlin, Germany
| | - Christian Schumann
- Klinikverbund Allgäu gGmbH, Clinic for Pneumology, Thoracic Oncology, Sleep and Respiratory Medicine, Kempten and Immenstadt, Germany
| | - Frank Griesinger
- Pius-Hospital, University Medicine Oldenburg, Department of Hematology and Oncology, University Department Internal Medicine-Oncology, Oldenburg, Germany
| | - Jonas Kuon
- Lungenklinik Löwenstein, Department of Oncology, Löwenstein, Germany
| | - Martin Sebastian
- Goethe University Frankfurt, University Hospital, Department of Internal Medicine II, Hematology/Oncology, Frankfurt am Main, Germany
| | | |
Collapse
|
14
|
Alfaro-Murillo JA, Townsend JP. Pairwise and higher-order epistatic effects among somatic cancer mutations across oncogenesis. Math Biosci 2023; 366:109091. [PMID: 37996064 PMCID: PMC10847963 DOI: 10.1016/j.mbs.2023.109091] [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] [Received: 05/02/2023] [Revised: 09/21/2023] [Accepted: 10/20/2023] [Indexed: 11/25/2023]
Abstract
Cancer occurs as a consequence of multiple somatic mutations that lead to uncontrolled cell growth. Mutual exclusivity and co-occurrence of mutations imply-but do not prove-that mutations exert synergistic or antagonistic epistatic effects on oncogenesis. Knowledge of these interactions, and the consequent trajectories of mutation and selection that lead to cancer has been a longstanding goal within the cancer research community. Recent research has revealed mutation rates and scaled selection coefficients for specific recurrent variants across many cancer types. However, there are no current methods to quantify the strength of selection incorporating pairwise and higher-order epistatic effects on selection within the trajectory of likely cancer genotoypes. Therefore, we have developed a continuous-time Markov chain model that enables the estimation of mutation origination and fixation (flux), dependent on somatic cancer genotype. Coupling this continuous-time Markov chain model with a deconvolution approach provides estimates of underlying mutation rates and selection across the trajectory of oncogenesis. We demonstrate computation of fluxes and selection coefficients in a somatic evolutionary model for the four most frequently variant driver genes (TP53, LRP1B, KRAS and STK11) from 565 cases of lung adenocarcinoma. Our analysis reveals multiple antagonistic epistatic effects that reduce the possible routes of oncogenesis, and inform cancer research regarding viable trajectories of somatic evolution whose progression could be forestalled by precision medicine. Synergistic epistatic effects are also identified, most notably in the somatic genotype TP53 LRP1B for mutations in the KRAS gene, and in somatic genotypes containing KRAS or TP53 mutations for mutations in the STK11 gene. Large positive fluxes of KRAS variants were driven by large selection coefficients, whereas the flux toward LRP1B mutations was substantially aided by a large mutation rate for this gene. The approach enables inference of the most likely routes of site-specific variant evolution and estimation of the strength of selection operating on each step along the route, a key component of what we need to know to develop and implement personalized cancer therapies.
Collapse
Affiliation(s)
- Jorge A Alfaro-Murillo
- Department of Biostatistics, Yale School of Public Health, New Haven, CT, United States of America
| | - Jeffrey P Townsend
- Department of Biostatistics, Yale School of Public Health, New Haven, CT, United States of America; Department of Ecology and Evolutionary Biology, Yale University, New Haven, CT, United States of America; Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, United States of America.
| |
Collapse
|
15
|
Adler N, Bahcheli AT, Cheng KC, Al-Zahrani KN, Slobodyanyuk M, Pellegrina D, Schramek D, Reimand J. Mutational processes of tobacco smoking and APOBEC activity generate protein-truncating mutations in cancer genomes. SCIENCE ADVANCES 2023; 9:eadh3083. [PMID: 37922356 PMCID: PMC10624356 DOI: 10.1126/sciadv.adh3083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 10/04/2023] [Indexed: 11/05/2023]
Abstract
Mutational signatures represent a genomic footprint of endogenous and exogenous mutational processes through tumor evolution. However, their functional impact on the proteome remains incompletely understood. We analyzed the protein-coding impact of single-base substitution (SBS) signatures in 12,341 cancer genomes from 18 cancer types. Stop-gain mutations (SGMs) (i.e., nonsense mutations) were strongly enriched in SBS signatures of tobacco smoking, APOBEC cytidine deaminases, and reactive oxygen species. These mutational processes alter specific trinucleotide contexts and thereby substitute serines and glutamic acids with stop codons. SGMs frequently affect cancer hallmark pathways and tumor suppressors such as TP53, FAT1, and APC. Tobacco-driven SGMs in lung cancer correlate with smoking history and highlight a preventable determinant of these harmful mutations. APOBEC-driven SGMs are enriched in YTCA motifs and associate with APOBEC3A expression. Our study exposes SGM expansion as a genetic mechanism by which endogenous and carcinogenic mutational processes directly contribute to protein loss of function, oncogenesis, and tumor heterogeneity.
Collapse
Affiliation(s)
- Nina Adler
- Computational Biology Program, Ontario Institute for Cancer Research, Toronto, ON, Canada
| | - Alexander T. Bahcheli
- Computational Biology Program, Ontario Institute for Cancer Research, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Kevin C. L. Cheng
- Computational Biology Program, Ontario Institute for Cancer Research, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | | | - Mykhaylo Slobodyanyuk
- Computational Biology Program, Ontario Institute for Cancer Research, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Diogo Pellegrina
- Computational Biology Program, Ontario Institute for Cancer Research, Toronto, ON, Canada
| | - Daniel Schramek
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- Lunenfeld-Tanenbaum Research Institute, Toronto, ON, Canada
| | - Jüri Reimand
- Computational Biology Program, Ontario Institute for Cancer Research, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
16
|
Huang Y, Pfeiffer SM, Zhang Q. Primary tumor type prediction based on US nationwide genomic profiling data in 13,522 patients. Comput Struct Biotechnol J 2023; 21:3865-3874. [PMID: 37593720 PMCID: PMC10432138 DOI: 10.1016/j.csbj.2023.07.036] [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: 07/26/2022] [Revised: 07/16/2023] [Accepted: 07/25/2023] [Indexed: 08/19/2023] Open
Abstract
Timely and accurate primary tumor diagnosis is critical, and misdiagnoses and delays may cause undue health and economic burden. To predict primary tumor types based on genomics data from a de-identified US nationwide clinico-genomic database (CGDB), the XGBoost-based Clinico-Genomic Machine Learning Model (XC-GeM) was developed to predict 13 primary tumor types based on data from 12,060 patients in the CGDB, derived from routine clinical comprehensive genomic profiling (CGP) testing and chart-confirmed electronic health records (EHRs). The SHapley Additive exPlanations method was used to interpret model predictions. XC-GeM reached an outstanding area under the curve (AUC) of 0.965 and Matthew's correlation coefficient (MCC) of 0.742 in the holdout validation dataset. In the independent validation cohort of 955 patients, XC-GeM reached 0.954 AUC and 0.733 MCC and made correct predictions in 77% of non-small cell lung cancer (NSCLC), 86% of colorectal cancer, and 84% of breast cancer patients. Top predictors for the overall model (e.g. tumor mutational burden (TMB), gender, and KRAS alteration), and for specific tumor types (e.g., TMB and EGFR alteration for NSCLC) were supported by published studies. XC-GeM also achieved an excellent AUC of 0.880 and positive MCC of 0.540 in 507 patients with missing primary diagnosis. XC-GeM is the first algorithm to predict primary tumor type using US nationwide data from routine CGP testing and chart-confirmed EHRs, showing promising performance. It may enhance the accuracy and efficiency of cancer diagnoses, enabling more timely treatment choices and potentially leading to better outcomes.
Collapse
Affiliation(s)
| | | | - Qing Zhang
- Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| |
Collapse
|
17
|
Verma S, Chitikela S, Singh V, Khurana S, Pushpam D, Jain D, Kumar S, Gupta Y, Malik PS. A phase II study of metformin plus pemetrexed and carboplatin in patients with non-squamous non-small cell lung cancer (METALUNG). Med Oncol 2023; 40:192. [PMID: 37261532 DOI: 10.1007/s12032-023-02057-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 05/16/2023] [Indexed: 06/02/2023]
Abstract
Immune checkpoint inhibitors (ICIs) ± chemotherapy is the standard treatment for driver mutation-negative non-small cell lung cancer (NSCLC). However, accessibility to ICIs in LMICs is limited due to high cost, and platinum-based chemotherapy remains the mainstay of treatment. Metformin has anticancer properties, and studies suggest synergism between metformin and pemetrexed. Based on preclinical evidence, this combination may be more beneficial for STK11-mutated NSCLC, a subgroup, inherently resistant to ICIs. In this Simon two-stage, single-arm phase 2 trial, we investigated metformin with pemetrexed-carboplatin (PC) in patients with treatment-naive stage IV non-squamous NSCLC. The primary outcome was 6-month progression-free survival (PFS) rate. Secondary outcomes were safety, overall survival (OS), overall response rate (ORR), proportion of STK11 mutation, and effect of STK11 mutation on 6-month PFS rate. The study was terminated for futility after interim analysis. The median follow-up was 34.1 months. The 6-month PFS rate was 28% (95% CI 12.4-0.46). The median PFS and OS were 4.5 (95% CI 2.2-6.1) and 7.4 months (95% CI 5.3-15.3), respectively. The ORR was 72%. Gastrointestinal toxicities were the most common. No grade 4/5 toxicities were reported. Targeted sequencing was possible in nine cases. Two patients had STK11 mutation and a poor outcome (PFS < 12 weeks). We could not demonstrate the benefit of metformin with CP in terms of improvement in 6-month PFS rate; however, the combination was safe (CTRI/2019/02/017815).
Collapse
Affiliation(s)
- S Verma
- Department of Medical Oncology, All India Institute of Medical Sciences, New Delhi, India
| | - S Chitikela
- Department of Medical Oncology, All India Institute of Medical Sciences, New Delhi, India
| | - V Singh
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - S Khurana
- Department of Medical Oncology, All India Institute of Medical Sciences, New Delhi, India
| | - D Pushpam
- Department of Medical Oncology, All India Institute of Medical Sciences, New Delhi, India
| | - D Jain
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - S Kumar
- Department of Medical Oncology, All India Institute of Medical Sciences, New Delhi, India
| | - Y Gupta
- Department of Endocrinology, All India Institute of Medical Sciences, New Delhi, India
| | - P S Malik
- Department of Medical Oncology, All India Institute of Medical Sciences, New Delhi, India.
- Department of Medical Oncology, Dr.B.R.A.I.R.C.H., All India Institute of Medical Sciences, Room 245, New Delhi, India.
| |
Collapse
|
18
|
Affiliation(s)
- Isabelle Mahé
- Université Paris Cité, Paris, France
- Assistance Publique des Hôpitaux de Paris, Hôpital Louis Mourier, Médecine Interne, Colombes, France
- Innovative Therapies in Haemostasis, INSERM UMR_S1140, Paris, France
- INNOVTE, FCRIN, France
| | - Florian Scotté
- Gustave Roussy, Patient Pathway Department, Villejuif, France
| | - Ismail Elalamy
- INNOVTE, FCRIN, France
- Hematology and Thrombosis Center, Hôpital Tenon, Hôpitaux Universitaires de l’Est Parisien, Assistance Publique Hôpitaux de Paris, Faculté de Médecine, Sorbonne Université, Paris, France
- Research Group “Cancer, Vessel Biology and Therapeutics,” INSERM U938, Centre de Recherche Saint-Antoine, Institut Universitaire de Cancérologie, Faculty of Medicine, Sorbonne University, Paris, France
| |
Collapse
|
19
|
Moorthi S, Paguirigan A, Ko M, Pettinger M, Hoge ACH, Nag A, Patel NA, Wu F, Sather C, Fitzgibbon MP, Thorner AR, Anderson GL, Ha G, Berger AH. Somatic mutation but not aneuploidy differentiates lung cancer in never-smokers and smokers. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.05.522947. [PMID: 36712079 PMCID: PMC9881937 DOI: 10.1101/2023.01.05.522947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Lung cancer in never-smokers disproportionately affects older women. To understand the mutational landscape of this cohort, we performed detailed genome characterization of 73 lung adenocarcinomas from participants of the Women’s Health Initiative (WHI). We find enrichment of EGFR mutations in never-/light-smokers and KRAS mutations in heavy smokers as expected, but we also show that the specific variants of these genes differ by smoking status, with important therapeutic implications. Mutational signature analysis revealed signatures of clock, APOBEC, and DNA repair deficiency in never-/light-smokers; however, the mutational load of these signatures did not differ significantly from those found in smokers. Last, tumors from both smokers and never-/light-smokers shared copy number subtypes, with no significant differences in aneuploidy. Thus, the genomic landscape of lung cancer in never-/light-smokers and smokers is predominantly differentiated by somatic mutations and not copy number alterations.
Collapse
|
20
|
Katipally RR, Spurr LF, Gutiontov SI, Turchan WT, Connell P, Juloori A, Malik R, Binkley MS, Jiang AL, Rouhani SJ, Chervin CS, Wanjari P, Segal JP, Ng V, Loo BW, Gomez DR, Bestvina CM, Vokes EE, Ferguson MK, Donington JS, Diehn M, Pitroda SP. STK11 Inactivation Predicts Rapid Recurrence in Inoperable Early-Stage Non-Small-Cell Lung Cancer. JCO Precis Oncol 2023; 7:e2200273. [PMID: 36603171 PMCID: PMC10530422 DOI: 10.1200/po.22.00273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 10/04/2022] [Accepted: 11/14/2022] [Indexed: 01/06/2023] Open
Abstract
PURPOSE Molecular factors predicting relapse in early-stage non-small-cell lung cancer (ES-NSCLC) are poorly understood, especially in inoperable patients receiving radiotherapy (RT). In this study, we compared the genomic profiles of inoperable and operable ES-NSCLC. MATERIALS AND METHODS This retrospective study included 53 patients with nonsquamous ES-NSCLC (stage I-II) treated at a single institution (University of Chicago) with surgery (ie, operable; n = 30) or RT (ie, inoperable; n = 23) who underwent tumor genomic profiling. A second cohort of ES-NSCLC treated with RT (Stanford, n = 39) was included to power clinical analyses. Prognostic gene alterations were identified and correlated with clinical variables. The primary clinical end point was the correlation of prognostic genes with the cumulative incidence of relapse, disease-free survival, and overall survival (OS) in a pooled RT cohort from the two institutions (N = 62). RESULTS Although the surgery cohort exhibited lower rates of relapse, the RT cohort was highly enriched for somatic STK11 mutations (43% v 6.7%). Receiving supplemental oxygen (odds ratio [OR] = 5.5), 20+ pack-years of tobacco smoking (OR = 6.1), and Black race (OR = 4.3) were associated with increased frequency of STK11 mutations. In the pooled RT cohort (N = 62), STK11 mutation was strongly associated with inferior oncologic outcomes: 2-year incidence of relapse was 62% versus 20% and 2-year OS was 52% versus 85%, remaining independently prognostic on multivariable analyses (relapse: subdistribution hazard ratio = 4.0, P = .0041; disease-free survival: hazard ratio, 6.8, P = .0002; OS: hazard ratio, 6.0, P = .022). STK11 mutations were predominantly associated with distant failure, rather than local. CONCLUSION In this cohort of ES-NSCLC, STK11 inactivation was associated with poor oncologic outcomes after RT and demonstrated a novel association with clinical hypoxia, which may underlie its correlation with medical inoperability. Further validation in larger cohorts and investigation of effective adjuvant systemic therapies may be warranted.
Collapse
Affiliation(s)
- Rohan R. Katipally
- Department of Radiation and Cellular Oncology, University of Chicago Medicine, Chicago, IL
| | - Liam F. Spurr
- Department of Radiation and Cellular Oncology, University of Chicago Medicine, Chicago, IL
- The Pritzker School of Medicine, The University of Chicago, Chicago, IL
| | - Stanley I. Gutiontov
- Department of Radiation and Cellular Oncology, University of Chicago Medicine, Chicago, IL
| | - William Tyler Turchan
- Department of Radiation and Cellular Oncology, University of Chicago Medicine, Chicago, IL
| | - Philip Connell
- Department of Radiation and Cellular Oncology, University of Chicago Medicine, Chicago, IL
| | - Aditya Juloori
- Department of Radiation and Cellular Oncology, University of Chicago Medicine, Chicago, IL
| | - Renuka Malik
- Department of Radiation and Cellular Oncology, University of Chicago Medicine, Chicago, IL
| | - Michael S. Binkley
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA
| | - Alice L. Jiang
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA
| | - Sherin J. Rouhani
- Section of Hematology/Oncology, Department of Medicine, University of Chicago Medicine, Chicago, IL
| | - Carolina Soto Chervin
- Section of Hematology/Oncology, Department of Medicine, NorthShore University HealthSystem, Evanston, IL
| | - Pankhuri Wanjari
- Department of Pathology, University of Chicago Medicine, Chicago, IL
| | - Jeremy P. Segal
- Department of Pathology, University of Chicago Medicine, Chicago, IL
| | - Victor Ng
- Department of Radiation Oncology, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Billy W. Loo
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA
| | - Daniel R. Gomez
- Department of Radiation Oncology, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Christine M. Bestvina
- Section of Hematology/Oncology, Department of Medicine, University of Chicago Medicine, Chicago, IL
| | - Everett E. Vokes
- Section of Hematology/Oncology, Department of Medicine, University of Chicago Medicine, Chicago, IL
| | - Mark K. Ferguson
- Section of Thoracic Surgery, Department of Surgery, University of Chicago Medicine, Chicago, IL
| | - Jessica S. Donington
- Section of Thoracic Surgery, Department of Surgery, University of Chicago Medicine, Chicago, IL
| | - Maximilian Diehn
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA
| | - Sean P. Pitroda
- Department of Radiation and Cellular Oncology, University of Chicago Medicine, Chicago, IL
| |
Collapse
|
21
|
Giraldo NA, Drill E, Satravada BA, Dika IE, Brannon AR, Dermawan J, Mohanty A, Ozcan K, Chakravarty D, Benayed R, Vakiani E, Abou-Alfa GK, Kundra R, Schultz N, Li BT, Berger MF, Harding JJ, Ladanyi M, O’Reilly EM, Jarnagin W, Vanderbilt C, Basturk O, Arcila ME. Comprehensive Molecular Characterization of Gallbladder Carcinoma and Potential Targets for Intervention. Clin Cancer Res 2022; 28:5359-5367. [PMID: 36228155 PMCID: PMC9772093 DOI: 10.1158/1078-0432.ccr-22-1954] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/04/2022] [Accepted: 10/11/2022] [Indexed: 01/24/2023]
Abstract
PURPOSE Gallbladder carcinoma (GBC) is an uncommon and aggressive disease, which remains poorly defined at a molecular level. Here, we aimed to characterize the molecular landscape of GBC and identify markers with potential prognostic and therapeutic implications. EXPERIMENTAL DESIGN GBC samples were analyzed using the MSK-IMPACT (Memorial Sloan Kettering-Integrated Mutation Profiling of Actionable Cancer Targets) platform (targeted NGS assay that analyzes 505 cancer-associated genes). Variants with therapeutic implications were identified using OncoKB database. The associations between recurrent genetic alterations and clinicopathologic characteristics (Fisher exact tests) or overall survival (univariate Cox regression) were evaluated. P values were adjusted for multiple testing. RESULTS Overall, 244 samples (57% primary tumors and 43% metastases) from 233 patients were studied (85% adenocarcinomas, 10% carcinomas with squamous differentiation, and 5% neuroendocrine carcinomas). The most common oncogenic molecular alterations appeared in the cell cycle (TP53 63% and CDKN2A 21%) and RTK_RAS pathways (ERBB2 15% and KRAS 11%). No recurrent structural variants were identified. There were no differences in the molecular landscape of primary and metastasis samples. Variants in SMAD4 and STK11 independently associated with reduced survival in patients with metastatic disease. Alterations considered clinically actionable in GBC or other solid tumor types (e.g., NTRK1 fusions or oncogenic variants in ERBB2, PIK3CA, or BRCA1/2) were identified in 35% of patients; 18% of patients with metastatic disease were treated off-label or enrolled in a clinical trial based on molecular findings. CONCLUSIONS GBC is a genetically diverse malignancy. This large-scale genomic analysis revealed alterations with potential prognostic and therapeutic implications and provides guidance for the development of targeted therapies.
Collapse
Affiliation(s)
- Nicolas A. Giraldo
- Department of Pathology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Esther Drill
- Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Baby A Satravada
- Marie-Josée & Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Imane El Dika
- Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
- Weill Medical College at Cornell University, 1275 York Avenue, New York, NY, 10065, USA
| | - A. Rose Brannon
- Department of Pathology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Josephine Dermawan
- Department of Pathology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Abhinita Mohanty
- Department of Pathology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Kerem Ozcan
- Department of Pathology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Debyani Chakravarty
- Marie-Josée & Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Ryma Benayed
- Department of Pathology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Efsevia Vakiani
- Department of Pathology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
- Marie-Josée & Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Ghassan K. Abou-Alfa
- Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
- Weill Medical College at Cornell University, 1275 York Avenue, New York, NY, 10065, USA
| | - Ritika Kundra
- Marie-Josée & Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Nikolaus Schultz
- Marie-Josée & Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Bob T. Li
- Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
- Weill Medical College at Cornell University, 1275 York Avenue, New York, NY, 10065, USA
| | - Michael F. Berger
- Department of Pathology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - James J. Harding
- Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
- Weill Medical College at Cornell University, 1275 York Avenue, New York, NY, 10065, USA
| | - Marc Ladanyi
- Department of Pathology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Eileen M. O’Reilly
- Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
- Weill Medical College at Cornell University, 1275 York Avenue, New York, NY, 10065, USA
| | - William Jarnagin
- Surgery, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
- Weill Medical College at Cornell University, 1275 York Avenue, New York, NY, 10065, USA
| | - Chad Vanderbilt
- Department of Pathology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Olca Basturk
- Department of Pathology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
- Weill Medical College at Cornell University, 1275 York Avenue, New York, NY, 10065, USA
| | - Maria E. Arcila
- Department of Pathology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
- Weill Medical College at Cornell University, 1275 York Avenue, New York, NY, 10065, USA
| |
Collapse
|
22
|
Yang L, Zhang Q, Xiong Y, Dang Z, Xiao H, Chen Q, Dai X, Zhang L, Zhu J, Wang D, Li M. A subset of VEGFR-TKIs activates AMPK in LKB1-mutant lung cancer. Cancer Sci 2022; 114:1651-1662. [PMID: 36459496 PMCID: PMC10067398 DOI: 10.1111/cas.15677] [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: 09/06/2022] [Revised: 11/10/2022] [Accepted: 11/24/2022] [Indexed: 12/03/2022] Open
Abstract
The mutation of tumor suppressor gene liver kinase B1 (LKB1) has a prevalence of about 20% in non-small cell lung cancer (NSCLC). LKB1-mutant lung cancer is characterized by enhanced aggressiveness and immune escape and is associated with poor prognosis. Therefore, it is urgent to develop effective therapeutic methods for LKB1-mutant NSCLC. Recently, apatinib, a VEGFR-TKI, was found to significantly improve the outcome of LKB1-mutant NSCLC, but the mechanism is not completely clear. In this study, AMP-activated protein kinase (AMPK), the crucial downstream kinase of LKB1 was excavated as the potential target of apatinib. Biochemical experiments verified that apatinib is a direct AMPK activator. Moreover, clinically available VEGFR-TKIs were found to regulate AMPK differently: Apatinib and anlotinib can directly activate AMPK, while axitinib and sunitinib can directly inhibit AMPK. Activation of AMPK by apatinib leads to the phosphorylation of acetyl-CoA carboxylase (ACC) and inhibition of de novo fatty acid synthesis (FAsyn), which is upregulated in LKB1-null cancers. Moreover, the killing effect of apatinib was obviously enhanced under delipidated condition, and the combination of exogenous FA restriction with apatinib treatment can be a promising method for treating LKB1-mutant NSCLC. This study discovered AMPK as an important off-target of apatinib and elucidated different effects of this cluster of VEGFR-TKIs on AMPK. This finding can be the basis for the accurate and combined application of these drugs in clinic and highlights that the subset of VEGFR-TKIs including apatinib and anlotinib are potentially valuable in the treatment of LKB1-mutant NSCLC.
Collapse
Affiliation(s)
- Lujie Yang
- Cancer Center, Daping Hospital, Army Medical University, Chongqing, China
| | - Qin Zhang
- Cancer Center, Daping Hospital, Army Medical University, Chongqing, China
| | - Yanli Xiong
- Cancer Center, Daping Hospital, Army Medical University, Chongqing, China
| | - Zhaoqian Dang
- Cancer Center, Daping Hospital, Army Medical University, Chongqing, China
| | - He Xiao
- Cancer Center, Daping Hospital, Army Medical University, Chongqing, China
| | - Qian Chen
- Cancer Center, Daping Hospital, Army Medical University, Chongqing, China
| | - Xiaoyan Dai
- Cancer Center, Daping Hospital, Army Medical University, Chongqing, China
| | - Lei Zhang
- Cancer Center, Daping Hospital, Army Medical University, Chongqing, China
| | - Jianwu Zhu
- Cancer Center, Daping Hospital, Army Medical University, Chongqing, China
| | - Dong Wang
- Cancer Center, Daping Hospital, Army Medical University, Chongqing, China
| | - Mengxia Li
- Cancer Center, Daping Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
23
|
Isolation of TTF-1 Positive Circulating Tumor Cells for Single-Cell Sequencing by Using an Automatic Platform Based on Microfluidic Devices. Int J Mol Sci 2022; 23:ijms232315139. [PMID: 36499466 PMCID: PMC9736518 DOI: 10.3390/ijms232315139] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/23/2022] [Accepted: 11/28/2022] [Indexed: 12/05/2022] Open
Abstract
Single-cell sequencing provides promising information in tumor evolution and heterogeneity. Even with the recent advances in circulating tumor cell (CTC) technologies, it remains a big challenge to precisely and effectively isolate CTCs for downstream analysis. The Cell RevealTM system integrates an automatic CTC enrichment and staining machine, an AI-assisted automatic CTC scanning and identification system, and an automatic cell picking machine for CTC isolation. H1975 cell line was used for the spiking test. The identification of CTCs and the isolation of target CTCs for genetic sequencing were performed from the peripheral blood of three cancer patients, including two with lung cancer and one with both lung cancer and thyroid cancer. The spiking test revealed a mean recovery rate of 81.81% even with extremely low spiking cell counts with a linear relationship between the spiked cell counts and the recovered cell counts (Y = 0.7241 × X + 19.76, R2 = 0.9984). The three cancer patients had significantly higher TTF-1+ CTCs than healthy volunteers. All target CTCs were successfully isolated by the Cell Picker machine for a subsequent genetic analysis. Six tumor-associated mutations in four genes were detected. The present study reveals the Cell RevealTM platform can precisely identify and isolate target CTCs and then successfully perform single-cell sequencing by using commercially available genetic devices.
Collapse
|
24
|
Ehmann S, Sassine D, Straubhar AM, Praiss AM, Aghajanian C, Alektiar KM, Broach V, Cadoo KA, Jewell EL, Boroujeni AM, Kyi C, Leitao MM, Mueller JJ, Murali R, Bhaloo SI, O'Cearbhaill RE, Park KJ, Sonoda Y, Weigelt B, Zamarin D, Abu-Rustum N, Friedman CF. Gastric-type adenocarcinoma of the cervix: Clinical outcomes and genomic drivers. Gynecol Oncol 2022; 167:458-466. [PMID: 36253302 PMCID: PMC10155605 DOI: 10.1016/j.ygyno.2022.10.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/30/2022] [Accepted: 10/03/2022] [Indexed: 11/04/2022]
Abstract
OBJECTIVES Gastric-type endocervical adenocarcinoma (GEA) is a rare form of cervical cancer not associated with human papilloma virus (HPV) infection. We summarize our experience with GEA at a large cancer center. METHODS Clinical and demographic information on all patients diagnosed with GEA between June 1, 2002 and July 1, 2019 was obtained retrospectively from clinical charts. Kaplan-Meier survival analysis was performed to describe progression-free survival (PFS) and overall survival (OS). Tumors from a subset of patients underwent next generation sequencing (NGS) analysis. RESULTS A total of 70 women with GEA were identified, including 43 who received initial treatment at our institution: of these 4 (9%) underwent surgery alone, 15 (35%) underwent surgery followed by adjuvant therapy, 10 (23%) were treated with definitive concurrent chemoradiation (CCRT), 7 (16%) with chemotherapy alone, and 3 (7%) with neoadjuvant CCRT and hysterectomy with or without chemotherapy. One-third (n = 14) of patients experienced disease progression, of whom 86% (n = 12) had prior CCRT. The median PFS and OS for patients with stage I GEA were 107 months (95% CI 14.8-199.2 months) and 111 months (95% CI 17-205.1 months) respectively, compared to 17 months (95% CI 5.6-28.4 months) and 33 months (95% CI 28.2-37.8 months) for patients with stages II-IV, respectively. On NGS, 4 patients (14%) had ERBB2 alterations, including 2 patients who received trastuzumab. CONCLUSIONS GEA is an aggressive form of cervical cancer with poor PFS and OS when diagnosed at stage II or later. Further investigation is needed to identify the optimal management approach for this rare subtype.
Collapse
Affiliation(s)
- Sarah Ehmann
- Gynecology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Dib Sassine
- Gynecology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Alli M Straubhar
- Gynecology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Aaron M Praiss
- Gynecology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Carol Aghajanian
- Gynecologic Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Kaled M Alektiar
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Vance Broach
- Gynecology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Karen A Cadoo
- St. James's Hospital Dublin, Trinity St. James's Cancer Institute, Dublin 8 DO8 NHY1, Ireland
| | - Elizabeth L Jewell
- Gynecology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Amir Momeni Boroujeni
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Chrisann Kyi
- Gynecologic Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Mario M Leitao
- Gynecology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Jennifer J Mueller
- Gynecology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Rajmohan Murali
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Shirin Issa Bhaloo
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Roisin E O'Cearbhaill
- Gynecologic Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Kay J Park
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Yukio Sonoda
- Gynecology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Britta Weigelt
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Dmitriy Zamarin
- Gynecologic Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Nadeem Abu-Rustum
- Gynecology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Claire F Friedman
- Gynecologic Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA.
| |
Collapse
|
25
|
Yu L, Gong C. Pancancer analysis of a potential gene mutation model in the prediction of immunotherapy outcomes. Front Genet 2022; 13:917118. [PMID: 36092890 PMCID: PMC9459043 DOI: 10.3389/fgene.2022.917118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 07/27/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Immune checkpoint blockade (ICB) represents a promising treatment for cancer, but predictive biomarkers are needed. We aimed to develop a cost-effective signature to predict immunotherapy benefits across cancers.Methods: We proposed a study framework to construct the signature. Specifically, we built a multivariate Cox proportional hazards regression model with LASSO using 80% of an ICB-treated cohort (n = 1661) from MSKCC. The desired signature named SIGP was the risk score of the model and was validated in the remaining 20% of patients and an external ICB-treated cohort (n = 249) from DFCI.Results: SIGP was based on 18 candidate genes (NOTCH3, CREBBP, RNF43, PTPRD, FAM46C, SETD2, PTPRT, TERT, TET1, ROS1, NTRK3, PAK7, BRAF, LATS1, IL7R, VHL, TP53, and STK11), and we classified patients into SIGP high (SIGP-H), SIGP low (SIGP-L) and SIGP wild type (SIGP-WT) groups according to the SIGP score. A multicohort validation demonstrated that patients in SIGP-L had significantly longer overall survival (OS) in the context of ICB therapy than those in SIGP-WT and SIGP-H (44.00 months versus 13.00 months and 14.00 months, p < 0.001 in the test set). The survival of patients grouped by SIGP in non-ICB-treated cohorts was different, and SIGP-WT performed better than the other groups. In addition, SIGP-L + TMB-L (approximately 15% of patients) had similar survivals to TMB-H, and patients with both SIGP-L and TMB-H had better survival. Further analysis on tumor-infiltrating lymphocytes demonstrated that the SIGP-L group had significantly increased abundances of CD8+ T cells.Conclusion: Our proposed model of the SIGP signature based on 18-gene mutations has good predictive value for the clinical benefit of ICB in pancancer patients. Additional patients without TMB-H were identified by SIGP as potential candidates for ICB, and the combination of both signatures showed better performance than the single signature.
Collapse
Affiliation(s)
- Lishan Yu
- Yanqi Lake Beijing Institute Mathematical Sciences and Applications, Beijing, China
- Yau Mathematical Sciences Center, Tsinghua University, Beijing, China
| | - Caifeng Gong
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- *Correspondence: Caifeng Gong,
| |
Collapse
|
26
|
Luo Y, Deng X, Que J, Li Z, Xie W, Dai G, Chen L, Wang H. Cell Trajectory-Related Genes of Lung Adenocarcinoma Predict Tumor Immune Microenvironment and Prognosis of Patients. Front Oncol 2022; 12:911401. [PMID: 35924143 PMCID: PMC9339705 DOI: 10.3389/fonc.2022.911401] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 06/23/2022] [Indexed: 01/21/2023] Open
Abstract
Background Lung adenocarcinoma (LUAD) is the most common subtype of lung cancer which typically exhibits a diverse progression trajectory. Our study sought to explore the cell differentiation trajectory of LUAD and its clinical relevance. Methods Utilizing a single-cell RNA-sequencing dataset (GSE117570), we identified LUAD cells of distinct differential status along with differentiation-related genes (DRGs). DRGs were applied to the analysis of bulk-tissue RNA-sequencing dataset (GSE72094) to classify tumors into different subtypes, whose clinical relevance was further analyzed. DRGs were also applied to gene co-expression network analysis (WGCNA) using another bulk-tissue RNA-sequencing dataset (TCGA-LUAD). Genes from modules that demonstrated a significant correlation with clinical traits and were differentially expressed between normal tissue and tumors were identified. Among these, genes with significant prognostic relevance were used for the development of a prognostic nomogram, which was tested on TCGA-LUAD dataset and validated in GSE72094. Finally, CCK-8, EdU, cell apoptosis, cell colony formation, and Transwell assays were used to verify the functions of the identified genes. Results Four clusters of cells with distinct differentiation status were characterized, whose DRGs were predominantly correlated with pathways of immune regulation. Based on DRGs, tumors could be clustered into four subtypes associated with distinct immune microenvironment and clinical outcomes. DRGs were categorized into four modules. A total of nine DRGs (SFTPB, WFDC2, HLA-DPA1, TIMP1, MS4A7, HLA-DQA1, VCAN, KRT8, and FABP5) with most significant survival-predicting power were integrated to develop a prognostic model, which outperformed the traditional parameters in predicting clinical outcomes. Finally, we verified that knockdown of WFDC2 inhibited proliferation, migration, and invasion but promoted the apoptosis of A549 cells in vitro. Conclusion The cellular composition and cellular differentiation status of tumor mass can predict the clinical outcomes of LUAD patients. It also plays an important role in shaping the tumor immune microenvironment.
Collapse
Affiliation(s)
- Yu Luo
- Department of Thoracic Surgery, The First Affiliated Hospital with Nanjing Medical University (Jiangsu Province Hospital), Nanjing, China
| | - Xiaheng Deng
- Department of Thoracic Surgery, The First Affiliated Hospital with Nanjing Medical University (Jiangsu Province Hospital), Nanjing, China
| | - Jun Que
- Department of Thoracic Surgery, The First Affiliated Hospital with Nanjing Medical University (Jiangsu Province Hospital), Nanjing, China
| | - Zhihua Li
- Department of Thoracic Surgery, The First Affiliated Hospital with Nanjing Medical University (Jiangsu Province Hospital), Nanjing, China
| | - Weiping Xie
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital with Nanjing Medical University (Jiangsu Province Hospital), Nanjing, China
| | - Guanqun Dai
- Department of General Practice, The First Affiliated Hospital with Nanjing Medical University (Jiangsu Province Hospital), Nanjing, China
| | - Liang Chen
- Department of Thoracic Surgery, The First Affiliated Hospital with Nanjing Medical University (Jiangsu Province Hospital), Nanjing, China
- *Correspondence: Liang Chen, ; Hong Wang, ;
| | - Hong Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital with Nanjing Medical University (Jiangsu Province Hospital), Nanjing, China
- *Correspondence: Liang Chen, ; Hong Wang, ;
| |
Collapse
|
27
|
Rosellini P, Amintas S, Caumont C, Veillon R, Galland-Girodet S, Cuguillière A, Nguyen L, Domblides C, Gouverneur A, Merlio JP, Bezin J, Girodet PO. Clinical impact of STK11 mutation in advanced-stage non-small cell lung cancer. Eur J Cancer 2022; 172:85-95. [PMID: 35759814 DOI: 10.1016/j.ejca.2022.05.026] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 05/10/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Mutations in STK11/LKB1 gene present a negative impact on tumour immune microenvironment, especially with concomitant activating KRAS mutation. These recent data may explain a decreased response to immunotherapy treatment in STK11 mutant non-small cell lung cancer (NSCLC). OBJECTIVE The primary objective is to evaluate, in a real-life setting, overall survival (OS) in patients with NSCLC according to the presence of STK11 mutation. The secondary objective is to assess time to treatment failure (TTF) for the first-line chemotherapy or immunotherapy. METHODS This observational multicentric study was conducted in Nouvelle-Aquitaine (France), for 24 months. Clinical, histopathological and imagery data were collected in each centre while the next-generation sequencing analysis was performed in Bordeaux Hospital University. Patient's data were longitudinally followed from NSCLC diagnosis date to the occurrence of censoring events (therapeutic failure or death, as applicable) or until the study end date. RESULTS median OS from the first drug administration was significantly longer for STK11wt patients than STK11mut patients (16.2 months [11 - nr] versus 4.7 months [2.5-9.4]; Log-rank test P < 0.001). The Presence of STK11 mutation was significantly associated with shortened OS (RR = 2.26 [1.35-3.79], P = 0.002). First-line TTF was significantly shorter in STK11mut population and the presence of the mutation was significantly associated with an increase in treatment failures (RR = 1.87 [1.21-2.89], P = 0.005). The type of treatment (chemotherapy, immunotherapy) does not influence the amplitude of reduced TTF in patients with STK11mut. CONCLUSION The presence of STK11 mutation is associated with poor prognosis in NSCLC.
Collapse
Affiliation(s)
- Pietro Rosellini
- CHU de Bordeaux, CIC1401, Service de Pharmacologie Médicale, Service de Biologie des tumeurs, Service des Maladies Respiratoires, F-33000 Pessac, France
| | - Samuel Amintas
- CHU de Bordeaux, CIC1401, Service de Pharmacologie Médicale, Service de Biologie des tumeurs, Service des Maladies Respiratoires, F-33000 Pessac, France; Univ. Bordeaux, Inserm CIC1401, Inserm CR1219, Inserm U1035, F-33000 Bordeaux, France
| | - Charline Caumont
- CHU de Bordeaux, CIC1401, Service de Pharmacologie Médicale, Service de Biologie des tumeurs, Service des Maladies Respiratoires, F-33000 Pessac, France
| | - Rémi Veillon
- CHU de Bordeaux, CIC1401, Service de Pharmacologie Médicale, Service de Biologie des tumeurs, Service des Maladies Respiratoires, F-33000 Pessac, France
| | | | - Alain Cuguillière
- Bagatelle Hôpital d'instruction des Armées, F-33000 Villenave-d'Ornon, France
| | | | - Charlotte Domblides
- Department of Medical Oncology, Hôpital Saint-André, CHU Bordeaux-University of Bordeaux, F-33000 Bordeaux, France; ImmunoConcEpt, CNRS UMR 5164, Bordeaux University, F-33000 Bordeaux, France
| | - Amandine Gouverneur
- CHU de Bordeaux, CIC1401, Service de Pharmacologie Médicale, Service de Biologie des tumeurs, Service des Maladies Respiratoires, F-33000 Pessac, France; Univ. Bordeaux, Inserm CIC1401, Inserm CR1219, Inserm U1035, F-33000 Bordeaux, France
| | - Jean-Philippe Merlio
- CHU de Bordeaux, CIC1401, Service de Pharmacologie Médicale, Service de Biologie des tumeurs, Service des Maladies Respiratoires, F-33000 Pessac, France; Univ. Bordeaux, Inserm CIC1401, Inserm CR1219, Inserm U1035, F-33000 Bordeaux, France
| | - Julien Bezin
- CHU de Bordeaux, CIC1401, Service de Pharmacologie Médicale, Service de Biologie des tumeurs, Service des Maladies Respiratoires, F-33000 Pessac, France; Univ. Bordeaux, Inserm CIC1401, Inserm CR1219, Inserm U1035, F-33000 Bordeaux, France
| | - Pierre-Olivier Girodet
- CHU de Bordeaux, CIC1401, Service de Pharmacologie Médicale, Service de Biologie des tumeurs, Service des Maladies Respiratoires, F-33000 Pessac, France; Univ. Bordeaux, Inserm CIC1401, Inserm CR1219, Inserm U1035, F-33000 Bordeaux, France.
| |
Collapse
|
28
|
Farooq H, Bien H, Chang V, Becker D, Park YH, Bates SE. Loss of function STK11 alterations and poor outcomes in non-small-cell lung cancer: Literature and case series of US Veterans. Semin Oncol 2022; 49:319-325. [PMID: 35831213 DOI: 10.1053/j.seminoncol.2022.06.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 06/13/2022] [Accepted: 06/14/2022] [Indexed: 11/11/2022]
Abstract
Emerging evidence suggests that STK11 alterations, frequently found in non-small-cell lung cancers, may be prognostic and/or predictive of response to therapy, particularly immunotherapy. STK11 affects multiple important cellular pathways, and mutations lead to tumor growth by creating an immunosuppressive and altered metabolic environment through changes in AMPK, STING, and vascular endothelial growth factor pathways. We illustrate the questions surrounding STK11 genomic alteration in NSCLC with a case series comprising six United States Veterans from a single institution. We discuss the history of STK11, review studies on its clinical impact, and describe putative mechanisms of how loss of STK11 might engender resistance to immunotherapy or other therapies. While the exact impact of STK11 alteration in non-small-cell lung cancer remain to be fully elucidated, future research and ongoing clinical trials will help us better understand its role in cancer development and devise more effective treatment strategies.
Collapse
Affiliation(s)
- Hafsa Farooq
- Section of Hematology and Oncology, VA Northport Medical Center, Northport, NY; Division of Hematology and Oncology, Renaissance School of Medicine, Stony Brook, NY.
| | - Harold Bien
- Section of Hematology and Oncology, VA Northport Medical Center, Northport, NY; Division of Hematology and Oncology, Renaissance School of Medicine, Stony Brook, NY
| | - Victor Chang
- Section of Hematology Oncology, VA New Jersey Health Care System, East Orange, NJ; Division of Hematology/Oncology, Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ
| | - Daniel Becker
- Section of Hematology Oncology, New York Harbor Health Care System, New York, NY; Division of Hematology and Medical Oncology, NYU Langone School of Medicine, New York, NY
| | - Yeun-Hee Park
- Section of Hematology Oncology, James J Peters VAMC, Bronx, NY; Division of Hematology/Oncology, Columbia Vagelos College of Physicians and Surgeons, New York, NY
| | - Susan E Bates
- Section of Hematology Oncology, James J Peters VAMC, Bronx, NY; Division of Hematology/Oncology, Columbia Vagelos College of Physicians and Surgeons, New York, NY
| |
Collapse
|
29
|
Garinet S, Wang P, Mansuet-Lupo A, Fournel L, Wislez M, Blons H. Updated Prognostic Factors in Localized NSCLC. Cancers (Basel) 2022; 14:cancers14061400. [PMID: 35326552 PMCID: PMC8945995 DOI: 10.3390/cancers14061400] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 03/06/2022] [Accepted: 03/08/2022] [Indexed: 12/25/2022] Open
Abstract
Lung cancer is the most common cause of cancer mortality worldwide, and non-small cell lung cancer (NSCLC) represents 80% of lung cancer subtypes. Patients with localized non-small cell lung cancer may be considered for upfront surgical treatment. However, the overall 5-year survival rate is 59%. To improve survival, adjuvant chemotherapy (ACT) was largely explored and showed an overall benefit of survival at 5 years < 7%. The evaluation of recurrence risk and subsequent need for ACT is only based on tumor stage (TNM classification); however, more than 25% of patients with stage IA/B tumors will relapse. Recently, adjuvant targeted therapy has been approved for EGFR-mutated resected NSCLC and trials are evaluating other targeted therapies and immunotherapies in adjuvant settings. Costs, treatment duration, emergence of resistant clones and side effects stress the need for a better selection of patients. The identification and validation of prognostic and theranostic markers to better stratify patients who could benefit from adjuvant therapies are needed. In this review, we report current validated clinical, pathological and molecular prognosis biomarkers that influence outcome in resected NSCLC, and we also describe molecular biomarkers under evaluation that could be available in daily practice to drive ACT in resected NSCLC.
Collapse
Affiliation(s)
- Simon Garinet
- Pharmacogenomics and Molecular Oncology Unit, Biochemistry Department, Assistance Publique—Hopitaux de Paris, Hôpital Européen Georges Pompidou, 75015 Paris, France;
- Centre de Recherche des Cordeliers, INSERM UMRS-1138, Sorbonne Université, Université de Paris, 75006 Paris, France
| | - Pascal Wang
- Oncology Thoracic Unit, Pulmonology Department, Assistance Publique—Hopitaux de Paris, Hôpital Cochin, 75014 Paris, France; (P.W.); (M.W.)
| | - Audrey Mansuet-Lupo
- Pathology Department, Assistance Publique—Hopitaux de Paris, Hôpital Cochin, 75014 Paris, France;
| | - Ludovic Fournel
- Thoracic Surgery Department, Assistance Publique—Hopitaux de Paris, Hôpital Cochin, 75014 Paris, France;
| | - Marie Wislez
- Oncology Thoracic Unit, Pulmonology Department, Assistance Publique—Hopitaux de Paris, Hôpital Cochin, 75014 Paris, France; (P.W.); (M.W.)
| | - Hélène Blons
- Pharmacogenomics and Molecular Oncology Unit, Biochemistry Department, Assistance Publique—Hopitaux de Paris, Hôpital Européen Georges Pompidou, 75015 Paris, France;
- Centre de Recherche des Cordeliers, INSERM UMRS-1138, Sorbonne Université, Université de Paris, 75006 Paris, France
- Correspondence:
| |
Collapse
|
30
|
Canterbury CR, Fernandes H, Crapanzano JP, Murty VV, Mansukhani MM, Shu CA, Szabolcs M, Saqi A. ALK Gene Rearrangements in Lung Adenocarcinomas: Concordance of Immunohistochemistry, Fluorescence In Situ Hybridization, RNA In Situ Hybridization, and RNA Next-Generation Sequencing Testing. JTO Clin Res Rep 2021; 2:100223. [PMID: 34746883 PMCID: PMC8552107 DOI: 10.1016/j.jtocrr.2021.100223] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 08/05/2021] [Accepted: 08/29/2021] [Indexed: 11/29/2022] Open
Abstract
Introduction The 2018 updated molecular testing guidelines for patients with advanced lung cancer incorporated ALK immunohistochemistry (IHC) analysis as an equivalent to fluorescence in situ hybridization (FISH) method recommended in 2013. Nevertheless, no specific recommendation for alternative methods was proposed owing to insufficient data. The aim of this study was to compare the results of ALK IHC, FISH, RNA next-generation sequencing (NGS), and RNA in situ hybridization (ISH) with available clinical data. Methods A search for lung carcinomas with ALK testing by greater than or equal to one modality (i.e., ALK IHC, FISH, NGS) was performed; a subset underwent RNA ISH. When available, clinical data were recorded. Results The results were concordant among all performed testing modalities in 86 of 90 cases (95.6%). Of the four discordant cases, two were ALK positive by FISH but negative by IHC, RNA NGS, and RNA ISH. The remaining two cases failed RNA NGS testing, one was IHC negative, FISH positive, RNA ISH negative and the second was IHC positive, FISH positive, RNA ISH equivocal. RNA NGS identified one rare and one novel ALK fusion. Sufficient therapy data were available in 10 cases treated with tyrosine kinase inhibitors; three had disease progression, including one with discordant results (FISH positive, RNA NGS negative, IHC negative, RNA ISH negative) and two with concordant ALK positivity among all modalities. Conclusions Our results reveal high concordance among IHC, RNA NGS, and RNA ISH. In cases of discordance with available RNA NGS, FISH result was positive whereas IHC and ISH results were negative. On the basis of our data, multimodality testing is recommended to identify discrepant results and patients (un)likely to respond to tyrosine kinase inhibitors.
Collapse
Affiliation(s)
- Carleigh R Canterbury
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York
| | - Helen Fernandes
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York
| | - John P Crapanzano
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York
| | - Vundavalli V Murty
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York
| | - Mahesh M Mansukhani
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York
| | - Catherine A Shu
- Division of Hematology/Oncology, Department of Medicine, Columbia University Medical Center, New York, New York
| | - Matthias Szabolcs
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York
| | - Anjali Saqi
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York
| |
Collapse
|
31
|
An J, Yan M, Yu N, Chennamadhavuni A, Furqan M, Mott SL, Loeffler BT, Kruser T, Sita TL, Feldman L, Nguyen R, Pasquinelli M, Hanna NH, Abu Hejleh T. Outcomes of patients with stage III non-small cell lung cancer (NSCLC) that harbor a STK11 mutation. Transl Lung Cancer Res 2021; 10:3608-3615. [PMID: 34584860 DOI: 10.21037/tlcr-21-177] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Accepted: 08/09/2021] [Indexed: 12/24/2022]
Abstract
Background STK11 mutation (STK11m ) in patients (pts) with stage IV non-small cell lung cancer (NSCLC) is associated with inferior survival and poor response to immune checkpoint inhibitors (ICI). The significance of STK11m in stage III NSCLC pts treated with concurrent chemoradiation (CCRT) with or without consolidation ICI is unknown. Methods Stage III NSCLC patients who received CCRT and had known STK11 mutational status were included in this retrospective study. The data on the STK11m pts were collected from 4 cancer institutions. A cohort of pts with wild type STK11 (STK11w ) from the University of Iowa served as a comparison group. Patient demographics and clinical characteristics were collected. Cox regression models were used to explore the effect of STK11 mutation on survival. Results 75 pts with stage III NSCLC who had known STK11 mutational status were identified. 16/75 (21%) had STK11m . 5/16 with STK11 m did not receive CCRT so they were excluded from the analysis. The clinical and demographic characteristics for the 11 STK11m and 59 STK11w pts were not statistically different (STK11m vs. STK11w ): mean age: 57 vs. 64 yrs, non-squamous histology: 8/11 (73%) vs. 37/59 (63%), KRAS mutation: 3/11 (27%) vs. 11/59 (19%), TP53 mutation: 6/11 (55%) vs. 15/59 (25%), PD-L1 ≥50%: 1/8 (13%) vs. 10/32 (31%), and consolidation ICI 6/11 (55%) vs. 17/59 (29%). Regarding the 6 STK11m pts who received ICI (4 pembrolizumab, 2 durvalumab), the median number of ICI infusions was 8 (range, 3-17) vs. 6 (range, 1-25) in the 17 pts with STK11w who received ICI (durvalumab). After adjusting for performance status and cancer stage, multivariable analysis showed that progression free survival (PFS) for the STK11m pts was significantly worse than STK11 w pts (HR =2.25; 95% CI, 1.03-4.88, P=0.04), whereas overall survival (OS) showed no significant difference for STK11m vs. STK11w patients (HR 1.47, 95% CI, 0.49-4.38, P=0.49). Conclusions In stage III NSCLC patients who received CCRT, STK11m was associated with worse PFS compared to STK11w . Larger studies are needed to further explore the prognostic implications of STK11m in stage III NSCLC and whether ICI impacts survival for this subgroup.
Collapse
Affiliation(s)
- Josiah An
- Division of Hematology, Oncology, Blood & Marrow Transplantation, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | - Melissa Yan
- Division of Hematology and Oncology, Indiana University Health - Melvin and Bren Simon Cancer Center, Indianapolis, IN, USA
| | - Nanmeng Yu
- Division of Hematology, Oncology, Blood & Marrow Transplantation, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | - Adithya Chennamadhavuni
- Division of Hematology, Oncology, Blood & Marrow Transplantation, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | - Muhammad Furqan
- Division of Hematology, Oncology, Blood & Marrow Transplantation, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | - Sarah L Mott
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA
| | - Bradley T Loeffler
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA
| | - Timothy Kruser
- Turville Bay Radiation Oncology, SSM Health, Madison, WI, USA
| | - Timothy L Sita
- Department of Radiation Oncology, Northwestern Memorial Hospital, Chicago, IL, USA
| | - Lawrence Feldman
- Division of Hematology and Oncology, The University of Illinois at Chicago, Chicago, IL, USA
| | - Ryan Nguyen
- Division of Hematology and Oncology, The University of Illinois at Chicago, Chicago, IL, USA
| | - Mary Pasquinelli
- Division of Hematology and Oncology, The University of Illinois at Chicago, Chicago, IL, USA
| | - Nasser H Hanna
- Division of Hematology and Oncology, Indiana University Health - Melvin and Bren Simon Cancer Center, Indianapolis, IN, USA
| | - Taher Abu Hejleh
- Division of Hematology, Oncology, Blood & Marrow Transplantation, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| |
Collapse
|
32
|
Mazzaschi G, Leonetti A, Minari R, Gnetti L, Quaini F, Tiseo M, Facchinetti F. Modulating Tumor Microenvironment: A Review on STK11 Immune Properties and Predictive vs Prognostic Role for Non-small-cell Lung Cancer Immunotherapy. Curr Treat Options Oncol 2021; 22:96. [PMID: 34524570 DOI: 10.1007/s11864-021-00891-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/21/2021] [Indexed: 01/07/2023]
Abstract
The quest for immunotherapy (IT) biomarkers is an element of highest clinical interest in both solid and hematologic tumors. In non-small-cell lung cancer (NSCLC) patients, besides PD-L1 expression evaluation with its intrinsic limitations, tissue and circulating parameters, likely portraying the tumor and its stromal/immune counterparts, have been proposed as potential predictors of IT responsiveness. STK11 mutations have been globally labeled as markers of IT resistance. After a thorough literature review, STK11 mutations condition the prognosis of NSCLC patients receiving ICI-containing regimens, implying a relevant biological and clinical significance. On the other hand, waiting for prospective and solid data, the putative negative predictive value of STK11 inactivation towards IT is sustained by less evidence. The physiologic regulation of multiple cellular pathways performed by STK11 likely explains the multifaceted modifications in tumor cells, stroma, and tumor immune microenvironment (TIME) observed in STK11 mutant lung cancer, particularly explored in the molecular subgroup of KRAS co-mutation. IT approaches available thus far in NSCLC, mainly represented by anti-PD-1/PD-L1 inhibitors, are not promising in the case of STK11 inactivation. Perceptive strategies aimed at modulating the TIME, regardless of STK11 status or specifically addressed to STK11-mutated cases, will hopefully provide valid therapeutic options to be adopted in the clinical practice.
Collapse
Affiliation(s)
- Giulia Mazzaschi
- Medical Oncology Unit, University Hospital of Parma, Via Gramsci 14, 43126, Parma, Italy
| | - Alessandro Leonetti
- Medical Oncology Unit, University Hospital of Parma, Via Gramsci 14, 43126, Parma, Italy
| | - Roberta Minari
- Medical Oncology Unit, University Hospital of Parma, Via Gramsci 14, 43126, Parma, Italy
| | - Letizia Gnetti
- Pathology Unit, University Hospital of Parma, Via Gramsci 14, 43126, Parma, Italy
| | - Federico Quaini
- Department of Medicine & Surgery, University of Parma, Via Gramsci 14, 43126, Parma, Italy
| | - Marcello Tiseo
- Medical Oncology Unit, University Hospital of Parma, Via Gramsci 14, 43126, Parma, Italy
- Department of Medicine & Surgery, University of Parma, Via Gramsci 14, 43126, Parma, Italy
| | - Francesco Facchinetti
- Université Paris-Saclay, Institut Gustave Roussy, Inserm, Biomarqueurs Prédictifs et Nouvelles Stratégies Thérapeutiques en Oncologie, 114 Rue Edouard Vaillant, 94800, Villejuif, France.
| |
Collapse
|
33
|
Sonkar A, Kumar P, Gautam A, Maity B, Saha S. New Scope of Targeted Therapies in Lung Carcinoma. Mini Rev Med Chem 2021; 22:629-639. [PMID: 34353252 DOI: 10.2174/1389557521666210805104714] [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: 08/29/2020] [Revised: 12/30/2020] [Accepted: 04/27/2021] [Indexed: 11/22/2022]
Abstract
Lung cancer (LC) is the leading cause of cancer deaths worldwide. Recent research has also shown LC as a genomic disease, causing somatic mutations in patients. Tests related to mutational analysis and genome profiles have lately expanded significantly in the genetics/genomics field of LC. This review summarizes the current knowledge about different signalling pathways of LC based on the clinical impact of molecular targets. It describes the main molecular pathways and changes involved in the development, progression, and cellular breakdown of LC and the molecular changes. This review focuses on approved and targeted experimental therapies such as immunotherapy and clinical trials that examine the different targeted approaches to treating LC. We aimto clarify the differences in the extent of various genetic mutations in several areas for LC patients. Targeted molecular therapies for LC can be continued with advanced racial differences in genetic changes, which have a significant impact on the choice of drug treatment and our understanding of the profile of drug susceptibility/resistance. The most relevant genes described in this review are EGFR, KRAS, MET, BRAF, PIK3CA, STK11, ERBB3, PTEN, and RB1. Combined research efforts in this field are required to understand the genetic difference in LC outcomes in the future.
Collapse
Affiliation(s)
- Archana Sonkar
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raibareli Road, Lucknow 226025. India
| | - Pranesh Kumar
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raibareli Road, Lucknow 226025. India
| | - Anurag Gautam
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raibareli Road, Lucknow 226025. India
| | - Biswanath Maity
- Centre of Biomedical Research, SGPGIMS Campus, Raebareli Road, Lucknow 226014, Uttar Pradesh. India
| | - Sudipta Saha
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raibareli Road, Lucknow 226025. India
| |
Collapse
|
34
|
Goodwin D, Rathi V, Conron M, Wright GM. Genomic and Clinical Significance of Multiple Primary Lung Cancers as Determined by Next-Generation Sequencing. J Thorac Oncol 2021; 16:1166-1175. [PMID: 33845213 DOI: 10.1016/j.jtho.2021.03.018] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 02/17/2021] [Accepted: 03/19/2021] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Marked variations in survival rates have brought into question whether standard clinicopathologic classification should be applied to patients presenting with multiple primary lung cancers (MPLCs). This study investigated the genetic profiles of MPLCs in a cohort of patients using next-generation sequencing and correlated results to clinicopathologic data and patient outcome. METHODS Patients treated surgically with curative intent for two putative primaries of similar histopathology from January 2000 to December 2019 at St Vincent's Hospital Melbourne. DNA and RNA was extracted from formalin-fixed, paraffin-embedded tumor tissue and sequenced on an Ion Torrent Personal Genome Machine system. Patient outcome was determined by overall survival and disease-free survival. RESULTS A total of 40 cases fulfilled the inclusion criteria. Mutational profiling was concordant with clinicopathologic diagnosis in most cases; however, seven cases (17.5%) revealed shared mutations suggesting metastatic disease and this was associated with a substantial reduction in overall survival (p < 0.05). CONCLUSIONS Our results suggest that gene sequencing technologies are potentially a more accurate diagnostic and prognostic tool compared with traditional histopathologic evaluation in patients presenting with suspected MPLCs, which could better guide management and predict outcomes.
Collapse
Affiliation(s)
- Daryn Goodwin
- Department of Surgery, St Vincent's Hospital Melbourne, The University of Melbourne, Fitzroy, Australia; Alfred Hospital, Melbourne, Australia
| | - Vivek Rathi
- Department of Anatomical Pathology, St Vincent's Hospital Melbourne, The University of Melbourne, Fitzroy, Australia
| | - Matthew Conron
- Department of Respiratory and Sleep Medicine, St Vincent's Hospital Melbourne, The University of Melbourne, Fitzroy, Australia
| | - Gavin M Wright
- Department of Surgery, St Vincent's Hospital Melbourne, The University of Melbourne, Fitzroy, Australia; Research and Education Lead Program, Victorian Comprehensive Cancer Centre, Parkville, Australia; Division of Cancer Surgery, Peter MacCallum Cancer Centre, Parkville, Australia.
| |
Collapse
|
35
|
Gu M, Xu T, Chang P. KRAS/LKB1 and KRAS/TP53 co-mutations create divergent immune signatures in lung adenocarcinomas. Ther Adv Med Oncol 2021; 13:17588359211006950. [PMID: 33995590 PMCID: PMC8072935 DOI: 10.1177/17588359211006950] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 03/11/2021] [Indexed: 12/11/2022] Open
Abstract
Lung adenocarcinomas exhibit various patterns of genomic alterations. During the development of this cancer, KRAS serves as a driver oncogene with a relatively high mutational frequency. Emerging data suggest that lung adenocarcinomas with KRAS mutations can show enhanced PD-L1 expression and additional somatic mutations, thus linking the prospect of applying immune checkpoint blockade therapy to this disease. However, the responses of KRAS-mutant lung adenocarcinomas to this therapy are distinct, which is largely attributed to the heterogeneity in the tumoral immune milieus. Recently, it was revealed that KRAS-mutant lung adenocarcinomas simultaneously expressing either a LKB1 or TP53 mutation typically have different immune profiles of their tumours: tumours with a KRAS/TP53 co-mutation generally present with a significant upregulation of PD-L1 expression and tumoricidal T-cell accumulation, and those with a KRAS/LKB1 co-mutation are frequently negative for PD-L1 expression and have few tumoricidal immune infiltrates. In this regard, interrogating TP53 or LKB1 mutation in addition to PD-L1 expression will be promising in guiding clinical use of immune checkpoint blockade therapy for KRAS-mutant lung adenocarcinomas.
Collapse
Affiliation(s)
- Meichen Gu
- Department of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun, P.R. China
| | - Tiankai Xu
- Department of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun, P.R. China
| | - Pengyu Chang
- Department of Radiation Oncology & Therapy, The First Hospital of Jilin University, No.71, Xinmin Str, Changchun, 130021, P.R. China
| |
Collapse
|
36
|
LKB1 Down-Modulation by miR-17 Identifies Patients With NSCLC Having Worse Prognosis Eligible for Energy-Stress-Based Treatments. J Thorac Oncol 2021; 16:1298-1311. [PMID: 33887464 DOI: 10.1016/j.jtho.2021.04.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 03/19/2021] [Accepted: 04/04/2021] [Indexed: 11/21/2022]
Abstract
INTRODUCTION Preclinical models recently unveiled the vulnerability of LKB1/KRAS comutated NSCLC to metabolic stress-based treatments. Because miR-17 is a potential epigenetic regulator of LKB1, we hypothesized that wild-type LKB1 (LKB1WT) NSCLC with high miR-17 expression may be sensitive to an energetic stress condition, and eligible for metabolic frailties-based therapeutic intervention. METHODS We took advantage of NSCLC cell lines with different combinations of KRAS mutation and LKB1 deletion and of patient-derived xenografts (PDXs) with high (LKB1WT/miR-17 high) or low (LKB1WT/miR-17 low) miR-17 expression. We evaluated LKB1 pathway impairment and apoptotic response to metformin. We retrospectively evaluated LKB1 and miR-17 expression levels in tissue specimens of patients with NSCLC and PDXs. In addition, a lung cancer series from The Cancer Genome Atlas data set was analyzed for miR-17 expression and potential correlation with clinical features. RESULTS We identified miR-17 as an epigenetic regulator of LKB1 in NSCLC and confirmed targeting of miR-17 to LKB1 3' untranslated region by luciferase reporter assay. We found that miR-17 overexpression functionally impairs the LKB1/AMPK pathway. Metformin treatment prompted apoptosis on miR-17 overexpression only in LKB1WT cell lines, and in LKB1WT/miR-17 high PDXs. A retrospective analysis in patients with NSCLC revealed an inverse correlation between miR-17 and LKB1 expression and highlighted a prognostic role of miR-17 expression in LKB1WT patients, which was further confirmed by The Cancer Genome Atlas data analysis. CONCLUSIONS We identified miR-17 as a mediator of LKB1 expression in NSCLC tumors. This study proposes a miR-17 expression score potentially exploitable to discriminate LKB1WT patients with NSCLC with impaired LKB1 expression and poor outcome, eligible for energy-stress-based treatments.
Collapse
|
37
|
Ye Q, Mohamed R, Dakhlallah D, Gencheva M, Hu G, Pearce MC, Kolluri SK, Marsh CB, Eubank TD, Ivanov AV, Guo NL. Molecular Analysis of ZNF71 KRAB in Non-Small-Cell Lung Cancer. Int J Mol Sci 2021; 22:3752. [PMID: 33916522 PMCID: PMC8038441 DOI: 10.3390/ijms22073752] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/29/2021] [Accepted: 03/30/2021] [Indexed: 02/07/2023] Open
Abstract
Our previous study found that zinc finger protein 71 (ZNF71) mRNA expression was associated with chemosensitivity and its protein expression was prognostic of non-small-cell lung cancer (NSCLC). The Krüppel associated box (KRAB) transcriptional repression domain is commonly present in human zinc finger proteins, which are linked to imprinting, silencing of repetitive elements, proliferation, apoptosis, and cancer. This study revealed that ZNF71 KRAB had a significantly higher expression than the ZNF71 KRAB-less isoform in NSCLC tumors (n = 197) and cell lines (n = 117). Patients with higher ZNF71 KRAB expression had a significantly worse survival outcome than patients with lower ZNF71 KRAB expression (log-rank p = 0.04; hazard ratio (HR): 1.686 [1.026, 2.771]), whereas ZNF71 overall and KRAB-less expression levels were not prognostic in the same patient cohort. ZNF71 KRAB expression was associated with epithelial-to-mesenchymal transition (EMT) in both patient tumors and cell lines. ZNF71 KRAB was overexpressed in NSCLC cell lines resistant to docetaxel and paclitaxel treatment compared to chemo-sensitive cell lines, consistent with its association with poor prognosis in patients. Therefore, ZNF71 KRAB isoform is a more effective prognostic factor than ZNF71 overall and KRAB-less expression for NSCLC. Functional analysis using CRISPR-Cas9 and RNA interference (RNAi) screening data indicated that a knockdown/knockout of ZNF71 did not significantly affect NSCLC cell proliferation in vitro.
Collapse
Affiliation(s)
- Qing Ye
- WVU Cancer Institute, West Virginia University, Morgantown, WV 26506, USA; (Q.Y.); (R.M.); (D.D.); (G.H.); (T.D.E.); (A.V.I.)
- Lane Department of Computer Science and Electrical Engineering, West Virginia University, Morgantown, WV 26506, USA
| | - Rehab Mohamed
- WVU Cancer Institute, West Virginia University, Morgantown, WV 26506, USA; (Q.Y.); (R.M.); (D.D.); (G.H.); (T.D.E.); (A.V.I.)
| | - Duaa Dakhlallah
- WVU Cancer Institute, West Virginia University, Morgantown, WV 26506, USA; (Q.Y.); (R.M.); (D.D.); (G.H.); (T.D.E.); (A.V.I.)
- Department of Microbiology, Immunology & Cell Biology, West Virginia University, Morgantown, WV 26506, USA;
- Institute of Global Health and Human Ecology, School of Sciences & Engineering, The American University of Cairo, New Cairo 11835, Egypt
| | - Marieta Gencheva
- Department of Microbiology, Immunology & Cell Biology, West Virginia University, Morgantown, WV 26506, USA;
| | - Gangqing Hu
- WVU Cancer Institute, West Virginia University, Morgantown, WV 26506, USA; (Q.Y.); (R.M.); (D.D.); (G.H.); (T.D.E.); (A.V.I.)
- Department of Microbiology, Immunology & Cell Biology, West Virginia University, Morgantown, WV 26506, USA;
| | - Martin C. Pearce
- Cancer Research Laboratory, Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR 97331, USA; (M.C.P.); (S.K.K.)
| | - Siva Kumar Kolluri
- Cancer Research Laboratory, Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR 97331, USA; (M.C.P.); (S.K.K.)
| | - Clay B. Marsh
- Department of Medicine, West Virginia University, Morgantown, WV 26506, USA;
| | - Timothy D. Eubank
- WVU Cancer Institute, West Virginia University, Morgantown, WV 26506, USA; (Q.Y.); (R.M.); (D.D.); (G.H.); (T.D.E.); (A.V.I.)
- Department of Microbiology, Immunology & Cell Biology, West Virginia University, Morgantown, WV 26506, USA;
| | - Alexey V. Ivanov
- WVU Cancer Institute, West Virginia University, Morgantown, WV 26506, USA; (Q.Y.); (R.M.); (D.D.); (G.H.); (T.D.E.); (A.V.I.)
- Department of Biochemistry, West Virginia University, Morgantown, WV 26506, USA
| | - Nancy Lan Guo
- WVU Cancer Institute, West Virginia University, Morgantown, WV 26506, USA; (Q.Y.); (R.M.); (D.D.); (G.H.); (T.D.E.); (A.V.I.)
- Department of Occupational and Environmental Health Sciences, West Virginia University, Morgantown, WV 26506, USA
| |
Collapse
|
38
|
Kumar R, Castillero F, Bhandari S, Malapati S, Kloecker G. The Hispanic paradox in non-small cell lung cancer. Hematol Oncol Stem Cell Ther 2021; 15:21-29. [PMID: 33775613 DOI: 10.1016/j.hemonc.2021.02.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 02/14/2021] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE/BACKGROUND According to the U.S. Census Bureau, 18% of the total population in the United States identified themselves as Hispanic in 2016 making it the largest minority group. This study aimed to evaluate the effect of Hispanic ethnicity on the overall survival of patients with non-small cell lung cancer (NSCLC) using a large national cancer database. METHODS We used the National Cancer Database to identify patients diagnosed with NSCLC between 2010 and 2015. The two comparative groups for this study were non-Hispanic Whites (NHWs) and Hispanics. The primary outcome was overall survival. RESULTS Of the 555,475 patients included in the study, 96.9% and 3.1% were NHWs and Hispanics with a median follow up of 12.6 months (interquartile range 4.1-30.6) and 12.1 months (interquartile range 3.8-29.5), respectively. Hispanics were more likely to be uninsured, and live in areas with lower median household income or education level. In the age-, sex-, and comorbidities-adjusted Cox model, the overall survival was significantly better in Hispanics compared with NHWs (hazard ratio [HR] 0.92, 95% confidence interval 0.90-0.93, p < .001). In a demographic, socioeconomic, clinical, and facility characteristics adjusted Cox model, Hispanics had further improvement in survival (HR 0.79, 95% confidence interval 0.78-0.81, p < .001). The survival advantage was seen in all cancer stages: Stage I-HR 0.76 (0.71-0.80), Stage II-HR 0.85 (0.79-0.92), Stage III-HR 0.81 (0.77-0.85), and Stage IV-HR 0.79 (0.77-0.81). CONCLUSION Hispanic ethnicity was associated with better survival in NSCLC. This survival advantage is likely the result of complex interactions amongst several physical, social, cultural, genomic, and environmental factors.
Collapse
Affiliation(s)
- Rohit Kumar
- Division of Medical Oncology and Hematology, James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA.
| | | | - Shruti Bhandari
- Division of Medical Oncology and Hematology, James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - Sindhu Malapati
- Division of Hematology and Oncology, Van Elslander Cancer Center, Ascension St. John Hospital and Medical Center, Grosse Pointe Woods, MI, USA
| | | |
Collapse
|
39
|
Biomarker Discovery and Validation: Statistical Considerations. J Thorac Oncol 2021; 16:537-545. [PMID: 33545385 DOI: 10.1016/j.jtho.2021.01.1616] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 01/04/2021] [Accepted: 01/07/2021] [Indexed: 12/23/2022]
Abstract
Biomarkers have various applications including disease detection, diagnosis, prognosis, prediction of response to intervention, and disease monitoring. In this era of precision medicine, having validated biomarkers to inform clinical decision making is more important than ever. In this article, we discuss best the practices and potential issues in biomarker discovery and validation. We encourage team science partnerships to bring cutting-edge discovery from bench to bedside, leading to improved patient care and outcomes.
Collapse
|
40
|
Karthikeyan SK, Gimbrone NT, Percy TR, Cress WD. Loss of cellular identity in common pre-clinical models of serine‑threonine kinase 11 (Liver kinase B1) loss. Cancer Treat Res Commun 2021; 26:100286. [PMID: 33338855 PMCID: PMC10022640 DOI: 10.1016/j.ctarc.2020.100286] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 12/01/2020] [Accepted: 12/10/2020] [Indexed: 11/26/2022]
Abstract
Nearly 1/3 of lung adenocarcinomas have loss of STK11 (LKB1) function. Herein, a bioinformatics approach was used to determine how accurately preclinical model systems reflect the in vivo biology of STK11 loss in human patients. Hierarchical and K-mean clustering, principle component, and gene set enrichment analyses were employed to model gene expression due to STK11 loss in patient cohorts representing nearly 1000 lung adenocarcinoma patients. K-means clustering classified STK11 loss patient tumors into three distinct sub-groups; positive (54%), neuroendocrine (NE) (35%) and negative (11%). The positive and NE groups are both defined by the expression of NKX2-1. In addition to NKX2-1, NE patients express neuroendocrine markers such as ASCL1 and CALCA. In contrast, the negative group does not express NKX2-1 (or neuroendocrine markers) and is characterized by significantly reduced survival relative to the two other groups. Two gene expression signatures were derived to explain both neuroendocrine features and differentiation (NKX2-1 loss) and were validated through two public datasets involving chemical differentiation (DCI) and NKX2-1 reconstitution. Patients results were then compared with established cell lines, transgenic mice, and patient-derived xenograft models of STK11 loss. Interestingly, all cell line and PDX models cluster and show expression patterns similar with the NKX2-1 negative subset of STK11-loss human tumors. Surprisingly, even mouse models of STK11 loss do not resemble patient tumors based on gene expression patterns. Results suggest pre-clinical models of STK11 loss are pronounced by marked elimination of type II pneumocyte identity, opposite of most in vivo human tumors.
Collapse
Affiliation(s)
| | - Nicholas T Gimbrone
- Program in Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Trent R Percy
- Program in Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - W Douglas Cress
- Program in Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA.
| |
Collapse
|
41
|
Popper H. Primary tumor and metastasis-sectioning the different steps of the metastatic cascade. Transl Lung Cancer Res 2020; 9:2277-2300. [PMID: 33209649 PMCID: PMC7653118 DOI: 10.21037/tlcr-20-175] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Patients with lung cancer in the majority die of metastases. Treatment options include surgery, chemo- and radiotherapy, targeted therapy by tyrosine kinase inhibitors (TKIs), and immuno-oncologic treatment. Despite the success with these treatment options, cure of lung cancer is achieved in only a very small proportion of patients. In most patients’ recurrence and metastasis will occur, and finally kill the patient. Metastasis is a multistep procedure. It requires a change in adhesion of tumor cells for detachment from their neighboring cells. The next step is migration either as single cells [epithelial-mesenchymal transition (EMT)], or as cell clusters (hybrid-EMT or bulk migration). A combination of genetic changes is required to facilitate migration. Then tumor cells have to orient themselves along matrix proteins, detect oxygen concentrations, prevent attacks by immune cells, and induce a tumor-friendly switch of stroma cells (macrophages, myofibroblasts, etc.). Having entered the blood stream tumor cells need to adapt to shear stress, avoid being trapped by coagulation, but also use coagulation in small veins for adherence to endothelia, and express homing molecules for extravasation. Within a metastatic site, tumor cells need a well-prepared niche to establish a metastatic focus. Tumor cells again have to establish a vascular net for maintaining nutrition and oxygen supply, communicate with stroma cells, grow out and set further metastases. In this review the different steps will be discussed with a focus on pulmonary carcinomas. The vast amount of research manuscripts published so far are not easy to analyze: in most reports’ single steps of the metastatic cascade are interpreted as evidence for the whole process; for example, migration is interpreted as evidence for metastasis. In lung cancer most often latency periods are shorter, in between 1–5 years. In other cases, despite widespread migration occurs, tumor cells die within the circulation and do not reach a metastatic site. Therefore, migration is a requisite, but does not necessarily predict metastasis. The intention of this review is to point to these different aspects and hopefully provoke research directed into a more functional analysis of the metastatic process.
Collapse
Affiliation(s)
- Helmut Popper
- Institute of Pathology, Medical University of Graz, Graz, Austria
| |
Collapse
|
42
|
Prieto M, Bobbio A, Fournel L, Icard P, Canny EH, Mansuet Lupo A, Leroy K, Wislez M, Damotte D, Alifano M. [Surgical management of resectable non-small cell lung cancer: Towards new paradigms]. Bull Cancer 2020; 107:904-911. [PMID: 32674934 DOI: 10.1016/j.bulcan.2020.05.010] [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: 12/30/2019] [Revised: 04/21/2020] [Accepted: 05/11/2020] [Indexed: 10/23/2022]
Abstract
Adapting therapies and providing personalized care for patients with resectable non-small cell lung cancer represent major challenges. This involves integrating several parameters into the patient's management, not only crude pathologic results, but also a better understanding of the mechanisms involved in tumor progression. Many studies have looked at the impact of host and tumor characteristics and their interactions through inflammatory processes or tumor immune environment. Beyond tumor stage, poor nutrition, sarcopenia and inflammatory state have been identified as independent factors that can directly impact postoperative outcome. The development of Enhanced Recovery After Surgery (ERAS), in which patient becomes the main player in their own management, seems to be an interesting answer since it seems to allow a reduction in postoperative complications, length of stay and indirectly reduction in costs. A broader and more complete vision including morphometric evaluation of the patient, physical performances, inflammatory state and nutritional state would provide additional discriminating information which can predict postoperative outcome and help in adapting therapies in a personalized way.
Collapse
Affiliation(s)
- Mathilde Prieto
- AP-HP Centre, UNIVERSITE de Paris, hôpital Cochin, service de chirurgie thoracique, 27, rue du Faubourg Saint-Jacques, 75014 Paris, France
| | - Antonio Bobbio
- AP-HP Centre, UNIVERSITE de Paris, hôpital Cochin, service de chirurgie thoracique, 27, rue du Faubourg Saint-Jacques, 75014 Paris, France
| | - Ludovic Fournel
- AP-HP Centre, UNIVERSITE de Paris, hôpital Cochin, service de chirurgie thoracique, 27, rue du Faubourg Saint-Jacques, 75014 Paris, France
| | - Philippe Icard
- AP-HP Centre, UNIVERSITE de Paris, hôpital Cochin, service de chirurgie thoracique, 27, rue du Faubourg Saint-Jacques, 75014 Paris, France
| | - Emelyne Hamelin Canny
- AP-HP Centre, UNIVERSITE de Paris, hôpital Cochin, service de chirurgie thoracique, 27, rue du Faubourg Saint-Jacques, 75014 Paris, France
| | - Audrey Mansuet Lupo
- AP-HP Centre, université de Paris, hôpital Cochin, service de pathologie, Paris, France
| | - Karen Leroy
- AP-HP Centre, université de Paris, hôpital Cochin, service de génétique et biologie moléculaire, Paris, France
| | - Marie Wislez
- AP-HP Centre, université de Paris, hôpital Cochin, service de pneumologie, Paris, France
| | - Diane Damotte
- AP-HP Centre, université de Paris, hôpital Cochin, service de pathologie, Paris, France
| | - Marco Alifano
- AP-HP Centre, UNIVERSITE de Paris, hôpital Cochin, service de chirurgie thoracique, 27, rue du Faubourg Saint-Jacques, 75014 Paris, France.
| |
Collapse
|
43
|
Brassart-Pasco S, Dalstein V, Brassart B, Dewolf M, Clavel C, Oudart JB. Immunotherapy in non-small-cell lung cancer: from targeted molecules to resistance patterns. Pharmacogenomics 2020; 21:705-720. [PMID: 32567537 DOI: 10.2217/pgs-2020-0021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Immunotherapies are now considered as a pillar of non-small-cell lung cancer treatment. The main targets of immune-checkpoint inhibitors (ICI) are programmed cell death 1/programmed cell death ligand 1 and cytotoxic T-lymphocyte antigen 4, aiming at restoring antitumor immunity. Despite durable responses observed in some patients, all patients do not benefit from the treatment and almost all responders ultimately relapse after some time. In this review, we discuss the biomarkers that could be used to predict response to ICI, the current indications of ICI in non-small-cell lung cancer, the mechanisms inducing tumor-cell intrinsic or extrinsic resistance to ICI and finally, the potential treatment response monitoring.
Collapse
Affiliation(s)
- Sylvie Brassart-Pasco
- Université de Reims Champagne-Ardenne, SFR CAP-Santé (FED 4231), Laboratoire de Biochimie Médicale et Biologie Moléculaire, 51100 Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire - MEDyC, 51100 Reims, France
| | - Véronique Dalstein
- Université de Reims Champagne Ardenne, INSERM, P3Cell UMR-S1250, SFR CAP-SANTE, 51100 Reims, France.,CHU Reims, Service de Pathologie, 51100 Reims, France
| | - Bertrand Brassart
- Université de Reims Champagne-Ardenne, SFR CAP-Santé (FED 4231), Laboratoire de Biochimie Médicale et Biologie Moléculaire, 51100 Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire - MEDyC, 51100 Reims, France
| | - Maxime Dewolf
- CHU Reims, Service des maladies respiratoires et allergiques, 51100 Reims, France
| | - Christine Clavel
- Université de Reims Champagne Ardenne, INSERM, P3Cell UMR-S1250, SFR CAP-SANTE, 51100 Reims, France.,CHU Reims, Service de Pathologie, 51100 Reims, France
| | - Jean-Baptiste Oudart
- Université de Reims Champagne-Ardenne, SFR CAP-Santé (FED 4231), Laboratoire de Biochimie Médicale et Biologie Moléculaire, 51100 Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire - MEDyC, 51100 Reims, France.,CHU Reims, Service de Biochimie-Pharmacologie-Toxicologie, 51100 Reims, France
| |
Collapse
|
44
|
Targeted next-generation sequencing of matched localized and metastatic primary high-risk SCCs identifies driver and co-occurring mutations and novel therapeutic targets. J Dermatol Sci 2020; 99:30-43. [PMID: 32595073 DOI: 10.1016/j.jdermsci.2020.05.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 05/01/2020] [Accepted: 05/12/2020] [Indexed: 12/31/2022]
Abstract
BACKGROUND Cutaneous squamous cell carcinoma (SCC) is the second most common type of skin cancer and is responsible for over one million cases annually. While only 3-5 % of SCCs metastasize, those that do are associated with significant morbidity and mortality. Using gene mutations to help predict metastasis and select therapeutics is still being explored. OBJECTIVE To present novel data from targeted sequencing of 20 case-matched localized and metastatic high-risk SCCs. METHODS A cancer-associated gene panel of 76 genes was run from formalin-fixed paraffin-embedded samples of 20 case-matched localized (10) and metastatic (10) high-risk SCCs (Vela Diagnostics). RESULTS Using spatial clustering analysis, primary driver mutations were identified asEGFR in localized SCC and CDH1 in metastatic SCC. ERBB4 and STK11 were found to be significant co-occurring mutations in localized SCC. Pathway analyses showed the RTK/RAS, TP53, TGF-b, NOTCH1, PI3K, and cell cycle pathways to be highly relevant in all high-risk SCCs with the Wnt pathway enhanced in metastatic SCC only. CONCLUSIONS This study compared gene mutations between localized and metastatic SCC with the intent of identifying key differences and new potential targeted treatment options. To our knowledge, the co-occurrence ofERBB4 and STK11 mutations has not been previously reported. Targeted inhibition of CDH1 and the Wnt pathway should be further explored in metastatic SCC.
Collapse
|
45
|
Less immune cell infiltration and worse prognosis after immunotherapy for patients with lung adenocarcinoma who harbored STK11 mutation. Int Immunopharmacol 2020; 84:106574. [PMID: 32413741 DOI: 10.1016/j.intimp.2020.106574] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 05/05/2020] [Accepted: 05/06/2020] [Indexed: 01/10/2023]
Abstract
The STK11 mutation defined a special subtype for patients with lung adenocarcinoma. The cBioPortal data platform was applied to analyze STK11 mutation frequency and the relationship between STK11 mutation and immune prognostic markers. The TIMER database was used to analyze the relationship between STK11 mutation and immune cell infiltration. The survival difference for lung adenocarcinoma patients harbored STK11 mutation who received immunotherapy also used the cBioPortal database. The results showed that STK11 mutation co-occurrence more KRAS and KEAP1 mutation and fewer TP53 and EGFR mutation (all, P < 0.05); the patients harbored STK11 mutation had a lower expression of PDL1 (P = 0.002), higher TMB score (P = 0.002), higher proportion of males and smoking history; the patients harbored STK11 mutation had fewer immune cell infiltration including B cell (P < 0.01), CD8+ T cell (P < 0.001), CD4+ T cell (P < 0.001), Macrophage (P < 0.001), Neutrophil (P < 0.001) and Dendritic cell (P < 0.001). Importantly, we found the patients harbored STK11 mutation who received immune checkpoint inhibitors have worse overall survival (OS) with median survival only 6 months. In conclusion, our study demonstrated that STK11 mutation defined a special subtype for lung adenocarcinoma patients with different co-occurrence gene mutation, lower PDL1 expression, fewer immune cell infiltration and worse OS benefit from immune checkpoint inhibitors.
Collapse
|
46
|
Gill CM, Loewenstern J, Rutland JW, Arib H, Pain M, Umphlett M, Kinoshita Y, McBride RB, Bederson J, Donovan M, Sebra R, Fowkes M, Shrivastava RK. STK11 mutation status is associated with decreased survival in meningiomas. Neurol Sci 2020; 41:2585-2589. [PMID: 32253637 DOI: 10.1007/s10072-020-04372-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Accepted: 03/24/2020] [Indexed: 01/11/2023]
Abstract
BACKGROUND Emerging evidence suggests that STK11 mutations may influence clinical outcome and response to immunotherapy in cancer. MATERIALS AND METHODS Next-generation targeted sequencing of STK11 mutation status in a large cohort of 188 meningiomas. RESULTS STK11 loss-of-function mutations were identified in 3.7% of meningiomas. STK11 mutations were found in both low- and high-grade lesions and samples from primary and recurrent disease. There was a 2.8-fold increased risk of death for patients whose meningioma harbored an STK11 mutation, after controlling for lesion grade and occurrence status. The median overall survival for patients with STK11-mutated meningiomas was 4.4 years compared with 16.8 years. CONCLUSION These data identify recurrent STK11 mutations in a subset of meningiomas. Genotyping of STK11 is encouraged for meningioma patients undergoing immunotherapy-based therapy.
Collapse
Affiliation(s)
- Corey M Gill
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA.
| | - Joshua Loewenstern
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - John W Rutland
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Hanane Arib
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Margaret Pain
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Melissa Umphlett
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yayoi Kinoshita
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Russell B McBride
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,The Institute for Translational Epidemiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Joshua Bederson
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Michael Donovan
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Robert Sebra
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Sema4, A Mount Sinai Venture, Stamford, CT, USA
| | - Mary Fowkes
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Raj K Shrivastava
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| |
Collapse
|
47
|
Papillon-Cavanagh S, Doshi P, Dobrin R, Szustakowski J, Walsh AM. STK11 and KEAP1 mutations as prognostic biomarkers in an observational real-world lung adenocarcinoma cohort. ESMO Open 2020; 5:e000706. [PMID: 32312757 PMCID: PMC7199918 DOI: 10.1136/esmoopen-2020-000706] [Citation(s) in RCA: 152] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 02/27/2020] [Accepted: 03/02/2020] [Indexed: 12/11/2022] Open
Abstract
INTRODUCTION Somatic mutations in STK11 and KEAP1, frequently comutated in non-squamous non-small cell lung cancer (NSQ NSCLC), have been associated with poor response to immune checkpoint blockade (ICB). However, previous reports lack non-ICB controls needed to properly ascertain the predictive nature of those biomarkers. The objective of this study was to evaluate the predictive versus prognostic effect of STK11 or KEAP1 mutations in NSQ NSCLC. METHODS Patients diagnosed with stage IIIB, IIIC, IVA or IVB NSQ NSCLC from a real-world data cohort from the Flatiron Health Network linked with genetic testing from Foundation Medicine were retrospectively assessed. Real-world, progression-free survival (rwPFS) and overall survival (OS) were calculated from time of initiation of first-line treatment. RESULTS We analysed clinical and mutational data for 2276 patients including patients treated with anti-programmed death-1 (PD-1)/anti-programmed death ligand 1 (PD-L1) inhibitors at first line (n=574). Mutations in STK11 or KEAP1 were associated with poor outcomes across multiple therapeutic classes and were not specifically associated with poor outcomes in ICB cohorts. There was no observable interaction between STK11 mutations and anti-PD-1/anti-PD-L1 treatment on rwPFS (HR, 1.05; 95% CI 0.76 to 1.44; p=0.785) or OS (HR, 1.13; 95% CI 0.76 to 1.67; p=0.540). Similarly, there was no observable interaction between KEAP1 mutations and treatment on rwPFS (HR, 0.93; 95% CI 0.67 to 1.28; p=0.653) or OS (HR, 0.98; 95% CI 0.66 to 1.45; p=0.913). CONCLUSION Our results show that STK11-KEAP1 mutations are prognostic, not predictive, biomarkers for anti-PD-1/anti-PD-L1 therapy.
Collapse
Affiliation(s)
| | - Parul Doshi
- Translational Medicine, Bristol-Myers Squibb Co, Princeton, New Jersey, USA
| | - Radu Dobrin
- Translational Medicine, Bristol-Myers Squibb Co, Princeton, New Jersey, USA
| | | | - Alice M Walsh
- Translational Medicine, Bristol-Myers Squibb Co, Princeton, New Jersey, USA
| |
Collapse
|
48
|
Torralvo J, Friedlaender A, Achard V, Addeo A. The Activity of Immune Checkpoint Inhibition in KRAS Mutated Non-small Cell Lung Cancer: A Single Centre Experience. Cancer Genomics Proteomics 2020; 16:577-582. [PMID: 31659111 DOI: 10.21873/cgp.20160] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 07/17/2019] [Accepted: 07/29/2019] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND/AIM KRAS mutation is the most frequent molecular alteration found in advanced non-small cell lung cancer (NSCLC). It is associated with a poor prognosis without available targeted therapy. Treatment options for NSCLC have been recently enriched by the development of immune checkpoint inhibitors (ICIs), and data about their efficacy in patients with KRAS-mutant NSCLC are discordant. This study assessed the routine efficacy of ICIs in advanced KRAS-mutant NSCLC. PATIENTS AND METHODS All stage IV NSCLC patients treated in our institution from January 2016 to December 2017 with immunotherapy were included in our analysis. We collected the status of KRAS and other mutations, as well as the type of ICI administered. We assessed four clinical outcomes: i) disease control rate (DCR), ii) partial response (PR), iii) progression-free survival (PFS) and iv) overall survival (OS). RESULTS A total of 45 patients were initially identified but 7 were excluded due to insufficient clinical data, so 38 were included in the end. In the KRAS wild-type cohort, the DCR was 59% with 49% PR, while the PFS was 8.4 months and OS 16.8 months. Among KRAS mutated patients, results were more favourable, the DCR was 81%, with 62% PR. PFS was 13.6 months and OS was 18.5 months. The median follow-up was 24 months (17 to 34 months) and 7 patients were still on treatment at the time of analysis. CONCLUSION Our data suggest that KRAS mutation is predictive of a superior response to immunotherapy. Furthermore, the lack of response of STK11 and KRAS co-mutated NSCLC patients to ICIs, is indeed negated by an additional TP53 mutation.
Collapse
Affiliation(s)
- Javier Torralvo
- Oncology Department, Geneva University Hospital, Geneva, Switzerland
| | - Alex Friedlaender
- Oncology Department, Geneva University Hospital, Geneva, Switzerland
| | - Verane Achard
- Department of Radiation Oncology, Geneva University Hospital, Geneva, Switzerland
| | - Alfredo Addeo
- Oncology Department, Geneva University Hospital, Geneva, Switzerland
| |
Collapse
|
49
|
Possibilities of Improving the Clinical Value of Immune Checkpoint Inhibitor Therapies in Cancer Care by Optimizing Patient Selection. Int J Mol Sci 2020; 21:ijms21020556. [PMID: 31952311 PMCID: PMC7014370 DOI: 10.3390/ijms21020556] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 01/13/2020] [Accepted: 01/14/2020] [Indexed: 12/12/2022] Open
Abstract
Immune checkpoint inhibitor (ICI) therapies have become the most important medical therapies in many malignancies, such as melanoma, non-small-cell lung cancer, and urogenital cancers. However, due to generally low response rates of PD-(L)1 monotherapy, both PD-(L)1 combination therapies and novel therapeutics are under large-scale clinical evaluation. Thus far, clinical trials have rather suboptimally defined the patient population most likely to benefit from ICI therapy, and there is an unmet need for negative predictive markers aiming to reduce the number of non-responding patients in clinical practice. Furthermore, there is a strong need for basic tumor immunology research and innovative clinical trials to fully unleash the potential of ICI combinations for the benefit of patients.
Collapse
|
50
|
Domingues I, Cedres S, Callejo A, Vivancos A, Martinez-Marti A, Felip E. Long duration of immunotherapy in a STK11 mutated/KRAS wild-type non-small cell lung cancer patient. Pulmonology 2020; 26:49-50. [DOI: 10.1016/j.pulmoe.2019.05.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Revised: 05/06/2019] [Accepted: 05/07/2019] [Indexed: 01/22/2023] Open
|