1
|
Melosky B, Juergens RA, Banerji S, Sacher A, Wheatley-Price P, Snow S, Tsao MS, Leighl NB, Martins I, Cheema P, Liu G, Chu QSC. The continually evolving landscape of novel therapies in oncogene-driven advanced non-small-cell lung cancer. Ther Adv Med Oncol 2025; 17:17588359241308784. [PMID: 39776537 PMCID: PMC11705342 DOI: 10.1177/17588359241308784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025] Open
Abstract
Non-small-cell lung cancer (NSCLC) is a highly heterogeneous disease that is frequently associated with a host of known oncogenic alterations. Advances in molecular diagnostics and drug development have facilitated the targeting of novel alterations such that the majority of NSCLC patients have driver mutations that are now clinically actionable. The goal of this review is to gain insights into clinical research and development principles by summary, analysis, and discussion of data on agents targeting known alterations in oncogene-driven, advanced NSCLC beyond those in the epidermal growth factor receptor (EGFR) and the anaplastic lymphoma kinase (ALK). A search of published and presented literature was conducted to identify prospective trials and integrated analyses reporting outcomes for agents targeting driver gene alterations (except those in EGFR and ALK) in molecularly selected, advanced NSCLC. Clinical efficacy data were extracted from eligible reports and summarized in text and tables. Findings show that research into alteration-directed therapies in oncogene-driven, advanced NSCLC is an extremely active research field. Ongoing research focuses on the expansion of new agents targeting both previously identified targets (particularly hepatocyte growth factor receptor (MET), human epidermal growth factor receptor 2 (HER2), and Kirsten rat sarcoma viral oncogene homolog (KRAS)) as well as novel, potentially actionable targets (such as neuregulin-1 (NRG1) and phosphatidylinositol 3-kinase (PI3K)). The refinement of biomarker selection criteria and the development of more selective and potent agents are allowing for increasingly specific and effective therapies and the expansion of clinically actionable alterations. Clinical advances in this field have resulted in a large number of regulatory approvals over the last 3 years. Future developments should focus on the continued application of alteration therapy matching principles and the exploration of novel ways to target oncogene-driven NSCLC.
Collapse
Affiliation(s)
- Barbara Melosky
- Medical Oncology, BC Cancer Agency—Vancouver, University of British Columbia, 600 West 10th Avenue, Vancouver, BC V5Z 4E6, Canada
| | | | - Shantanu Banerji
- Paul Albrechtsen Research Institute, CancerCare Manitoba, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Adrian Sacher
- Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Paul Wheatley-Price
- Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Stephanie Snow
- QEII Health Sciences Centre, Dalhousie University, Halifax, NS, Canada
| | - Ming-Sound Tsao
- University Health Network and Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Natasha B. Leighl
- Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | | | - Parneet Cheema
- William Osler Health System, University of Toronto, Brampton, ON, Canada
| | - Geoffrey Liu
- Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Quincy S. C. Chu
- Cross Cancer Institute, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
2
|
Deng R, Zhang W, Lv J, Wang F, Chen Y, Jiang C, Guan Y, Zhang C. Afatinib as first-line treatment for advanced lung squamous cell carcinoma harboring uncommon EGFR G719C and S768I co-mutation: A case report and literature review. Heliyon 2024; 10:e35304. [PMID: 39166093 PMCID: PMC11334663 DOI: 10.1016/j.heliyon.2024.e35304] [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: 03/22/2024] [Revised: 07/25/2024] [Accepted: 07/25/2024] [Indexed: 08/22/2024] Open
Abstract
Ten percent of non-small cell lung cancer patients with epidermal growth factor receptor (EGFR) mutations harbor uncommon variants. These mutations are mainly involved in lung adenocarcinomas but are rare in lung squamous cell carcinoma (LSCC). In 2018, the Food and Drug Administration-approved afatinib for this specific patient population. However, there is limited information regarding the effectiveness of afatinib for LSCC with EGFR mutations. This case report documented a unique case of a patient with LSCC, which had a rare compound EGFR mutation (G719C and S768I) and showed significant response to afatinib treatment, with 10 months of progression-free survival. New NTRK1 and RET gene mutations may play a potential role in the development of acquired resistance to afatinib following clinical progression. This case highlights the importance of genetic profiling in patients with LSCC. Although these patients have a low positive rate of EGFR mutations, searching for EGFR mutations in these patients might broaden their treatment options.
Collapse
Affiliation(s)
- Ruoyu Deng
- Department of Oncology, Qujing First People's Hospital/The Qujing Affiliated Hospital of Kunming Medical University, Qujing, 655000, China
| | - Wen Zhang
- Department of Oncology, Qujing First People's Hospital/The Qujing Affiliated Hospital of Kunming Medical University, Qujing, 655000, China
| | - Jialing Lv
- Department of Oncology, Qujing First People's Hospital/The Qujing Affiliated Hospital of Kunming Medical University, Qujing, 655000, China
| | - Fang Wang
- Department of Pathology, Second People's Hospital of Qujing City, Qujing, 655000, China
| | - Yanqiong Chen
- Department of Oncology, Qujing First People's Hospital/The Qujing Affiliated Hospital of Kunming Medical University, Qujing, 655000, China
| | - Chengqi Jiang
- Department of Oncology, Qujing First People's Hospital/The Qujing Affiliated Hospital of Kunming Medical University, Qujing, 655000, China
| | - Yaling Guan
- Department of Oncology, Qujing First People's Hospital/The Qujing Affiliated Hospital of Kunming Medical University, Qujing, 655000, China
| | - Chao Zhang
- Department of Oncology, Qujing First People's Hospital/The Qujing Affiliated Hospital of Kunming Medical University, Qujing, 655000, China
| |
Collapse
|
3
|
Shen L, Zhao J, Yang Y, Mu S, Yu Y, Han Y, Lu S. Prominent response to savolitinib monotherapy in high-grade fetal adenocarcinoma with MET amplification and concurrent brain metastasis: a case report. Transl Lung Cancer Res 2024; 13:1407-1413. [PMID: 38973955 PMCID: PMC11225042 DOI: 10.21037/tlcr-24-124] [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: 02/02/2024] [Accepted: 04/29/2024] [Indexed: 07/09/2024]
Abstract
Background Mesenchymal-epithelial transition (MET) represents a potential therapeutic target in various cancers, with amplification of the MET gene identified in a subset of patients with pulmonary adenocarcinomas. However, MET gene amplification is rarely observed in high-grade fetal adenocarcinoma (H-FLAC). Case Description Here we present a novel case of a patient diagnosed with stage IV H-FLAC harboring MET amplifications and treated with savolitinib. The 69-year-old male patient, who presented with a primary complaint of cough and white sputum, had a history of hypertension for over 10 years and a 45-year smoking history. The patient received savolitinib monotherapy treatment due to brain metastases. Despite the omission of radiotherapy for asymptomatic brain metastases, a notable response to savolitinib therapy was observed, with a partial response (PR) achieved after 4 weeks and a reduction in the brain tumor. At the time of the submission of this report, the patient received over 24 weeks of savolitinib treatment, and was maintained PR. The patient was still undergoing treatment. This highlights the potential clinical benefits of targeted therapy against MET amplification in H-FLAC. Conclusions H-FLAC harboring MET amplification and brain metastasis is rare. Treatment with savolitinib monotherapy resulted in a PR, providing preliminary insights to the efficacy of savolitinib for H-FLAC with MET amplification.
Collapse
Affiliation(s)
- Lan Shen
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jikai Zhao
- Department of Pathology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ying Yang
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shuya Mu
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yongfeng Yu
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuchen Han
- Department of Pathology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shun Lu
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
4
|
Kim HH, Lee JC, Oh IJ, Kim EY, Yoon SH, Lee SY, Lee MK, Lee JE, Park CK, Lee KY, Lee SY, Kim SJ, Lim JH, Choi CM. Real-World Outcomes of Crizotinib in ROS1-Rearranged Advanced Non-Small-Cell Lung Cancer. Cancers (Basel) 2024; 16:528. [PMID: 38339278 PMCID: PMC10854608 DOI: 10.3390/cancers16030528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 01/21/2024] [Accepted: 01/23/2024] [Indexed: 02/12/2024] Open
Abstract
Real-world data on the use and outcomes of crizotinib in ROS1-rearranged non-small-cell lung cancer (NSCLC) are limited. This study aims to analyze the real-world efficacy of crizotinib in South Korea and explore the utilization of liquid biopsies that implement next-generation sequencing (NGS) using cell-free total nucleic acids. In this prospective multicenter cohort study, 40 patients with ROS1-rearranged NSCLC, either starting or already on crizotinib, were enrolled. Patients had a median age of 61 years, with 32.5% presenting brain/central nervous system (CNS) metastases at treatment initiation. At the data cutoff, 48.0% were still in treatment; four continued with it even after disease progression due to the clinical benefits. The objective response rate was 70.0%, with a median duration of response of 27.8 months. The median progression-free survival was 24.1 months, while the median overall survival was not reached. Adverse events occurred in 90.0% of patients, primarily with elevated transaminases, yet these were mostly manageable. The NGS assay detected a CD74-ROS1 fusion in 2 of the 14 patients at treatment initiation and identified emerging mutations, such as ROS1 G2032R, ROS1 D2033N, and KRAS G12D, during disease progression. These findings confirm crizotinib's sustained clinical efficacy and safety in a real-world context, which was characterized by a higher elderly population and higher rates of brain/CNS metastases. The study highlights the clinical relevance of liquid biopsy for detecting resistance mechanisms, suggesting its value in personalized treatment strategies.
Collapse
Affiliation(s)
- Hyeon Hwa Kim
- Division of Pulmonology and Critical Care Medicine, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 05505, Republic of Korea;
| | - Jae Cheol Lee
- Department of Oncology, Asan Medical Centre, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea;
| | - In-Jae Oh
- Department of Internal Medicine, Chonnam National University Medical School and Hwasun Hospital, Gwangju 58128, Republic of Korea;
| | - Eun Young Kim
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Seong Hoon Yoon
- Department of Internal Medicine, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea;
| | - Shin Yup Lee
- Department of Internal Medicine, Kyungpook National University School of Medicine, Daegu 41404, Republic of Korea
| | - Min Ki Lee
- Division of Pulmonology, Allergy and Critical Care Medicine, Department of Internal Medicine, Pusan National University Hospital, Busan 49241, Republic of Korea;
| | - Jeong Eun Lee
- Division of Pulmonology, Department of Internal Medicine, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Chan Kwon Park
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 16247, Republic of Korea;
| | - Kye Young Lee
- Departments of Internal Medicine, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul 05030, Republic of Korea;
| | - Sung Yong Lee
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Internal Medicine, Korea University Guro Hospital, Korea University College of Medicine, Seoul 08308, Republic of Korea
| | - Seung Joon Kim
- Division of Pulmonology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul 16247, Republic of Korea;
| | - Jun Hyeok Lim
- Department of Internal Medicine, Inha University Hospital, Incheon 22332, Republic of Korea
| | - Chang-min Choi
- Division of Pulmonology and Critical Care Medicine, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 05505, Republic of Korea;
- Department of Oncology, Asan Medical Centre, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea;
| |
Collapse
|
5
|
Jha AK, Sherkhane UB, Mthun S, Jaiswar V, Purandare N, Prabhash K, Wee L, Rangarajan V, Dekker A. External Validation of Robust Radiomic Signature to Predict 2-Year Overall Survival in Non-Small-Cell Lung Cancer. J Digit Imaging 2023; 36:2519-2531. [PMID: 37735307 PMCID: PMC10584779 DOI: 10.1007/s10278-023-00835-8] [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: 02/06/2023] [Revised: 03/16/2023] [Accepted: 04/13/2023] [Indexed: 09/23/2023] Open
Abstract
Lung cancer is the second most fatal disease worldwide. In the last few years, radiomics is being explored to develop prediction models for various clinical endpoints in lung cancer. However, the robustness of radiomic features is under question and has been identified as one of the roadblocks in the implementation of a radiomic-based prediction model in the clinic. Many past studies have suggested identifying the robust radiomic feature to develop a prediction model. In our earlier study, we identified robust radiomic features for prediction model development. The objective of this study was to develop and validate the robust radiomic signatures for predicting 2-year overall survival in non-small cell lung cancer (NSCLC). This retrospective study included a cohort of 300 stage I-IV NSCLC patients. Institutional 200 patients' data were included for training and internal validation and 100 patients' data from The Cancer Image Archive (TCIA) open-source image repository for external validation. Radiomic features were extracted from the CT images of both cohorts. The feature selection was performed using hierarchical clustering, a Chi-squared test, and recursive feature elimination (RFE). In total, six prediction models were developed using random forest (RF-Model-O, RF-Model-B), gradient boosting (GB-Model-O, GB-Model-B), and support vector(SV-Model-O, SV-Model-B) classifiers to predict 2-year overall survival (OS) on original data as well as balanced data. Model validation was performed using 10-fold cross-validation, internal validation, and external validation. Using a multistep feature selection method, the overall top 10 features were chosen. On internal validation, the two random forest models (RF-Model-O, RF-Model-B) displayed the highest accuracy; their scores on the original and balanced datasets were 0.81 and 0.77 respectively. During external validation, both the random forest models' accuracy was 0.68. In our study, robust radiomic features showed promising predictive performance to predict 2-year overall survival in NSCLC.
Collapse
Affiliation(s)
- Ashish Kumar Jha
- Department of Radiation Oncology (MAASTRO), GROW School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, The Netherlands.
- Department of Nuclear Medicine, Tata Memorial Hospital, Mumbai, Maharashtra, India.
- Homi Bhabha National Institute, Mumbai, Maharashtra, India.
| | - Umeshkumar B Sherkhane
- Department of Radiation Oncology (MAASTRO), GROW School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, The Netherlands
- Department of Nuclear Medicine, Tata Memorial Hospital, Mumbai, Maharashtra, India
| | - Sneha Mthun
- Department of Radiation Oncology (MAASTRO), GROW School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, The Netherlands
- Department of Nuclear Medicine, Tata Memorial Hospital, Mumbai, Maharashtra, India
- Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Vinay Jaiswar
- Department of Nuclear Medicine, Tata Memorial Hospital, Mumbai, Maharashtra, India
| | - Nilendu Purandare
- Department of Nuclear Medicine, Tata Memorial Hospital, Mumbai, Maharashtra, India
- Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Kumar Prabhash
- Homi Bhabha National Institute, Mumbai, Maharashtra, India
- Department of Medical Oncology, Tata Memorial Hospital, Mumbai, Maharashtra, India
| | - Leonard Wee
- Department of Radiation Oncology (MAASTRO), GROW School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Venkatesh Rangarajan
- Department of Nuclear Medicine, Tata Memorial Hospital, Mumbai, Maharashtra, India
- Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Andre Dekker
- Department of Radiation Oncology (MAASTRO), GROW School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, The Netherlands
| |
Collapse
|
6
|
Lefebvre AM, Adam J, Nicolazzi C, Larois C, Attenot F, Falda-Buscaiot F, Dib C, Masson N, Ternès N, Bauchet AL, Demers B, Chadjaa M, Sidhu S, Combeau C, Soria JC, Scoazec JY, Naimi S, Angevin E, Chiron M, Henry C. The search for therapeutic targets in lung cancer: Preclinical and human studies of carcinoembryonic antigen-related cell adhesion molecule 5 expression and its associated molecular landscape. Lung Cancer 2023; 184:107356. [PMID: 37660479 DOI: 10.1016/j.lungcan.2023.107356] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 08/11/2023] [Accepted: 08/25/2023] [Indexed: 09/05/2023]
Abstract
OBJECTIVES CEACAM5 is a cell-surface glycoprotein expressed on epithelial cells of some solid tumors. Tusamitamab ravtansine (SAR408701), a humanized antibody-drug conjugate targeting CEACAM5, is in clinical development for nonsquamous non-small cell lung cancer (NSQ-NSCLC) with CEACAM5 high expression (HE), defined as membranous CEACAM5 immunohistochemistry staining at ≥ 2+ intensity in ≥ 50% of tumor cells. MATERIALS AND METHODS We investigated correlations between CEACAM5 expression by immunohistochemistry, CEACAM5 protein expression by ELISA, and CEACAM5 RNA expression by RNA-seq in NSQ-NSCLC patient-derived xenograft (PDX) models, and tumor responses to tusamitamab ravtansine in these models. We assessed prevalence of CEACAM5 HE, clinicopathologic characteristics and molecular markers in patients with NSQ-NSCLC in clinical cohorts. RESULTS In a lung PDX set of 10 NSQ-NSCLC specimens, correlations between CEACAM5 by IHC, ELISA and RNA-seq ranged from 0.72 to 0.88. In a larger lung PDX set, higher H-scores were present in NSQ- (n = 93) vs SQ-NSCLC (n = 128) models, and in 12 of these NSQ-NSCLC models, more tumor responses to tusamitamab ravtansine occurred in CEACAM5 HE (5/8; 62.5%) versus moderate or negative expression (1/4; 25%), including 3 with KRAS mutations among the 6 responders. In clinical NSQ-NSCLC samples, CEACAM5 HE prevalence was (52/214; 24.3%) in primary tumors and (6/17; 35.3%) in metastases. In NSQ-NSCLC primary tumors, CEACAM5 HE prevalence was significantly higher in KRAS-altered versus wild-type (35.0% vs 19.5%; P = 0.028) and in programmed cell death ligand 1 (PD-L1) negative (tumor cells 0%)/low (1-49%) versus high (≥50%) (33.3%, 26.1%, 5.0%; P = 0.031), but not significantly different in EGFR-mutated versus wild-type (20.0% vs 25.7%, P = 0.626). CONCLUSIONS In NSQ-NSCLC tumors, CEACAM5 HE prevalence was 24.3% overall and was higher with KRAS altered and with PD-L1 negative/low tumors but similar regardless of EGFR mutation status. These findings support targeting CEACAM5 and the clinical development of tusamitamab ravtansine for patients with NSQ-NSCLC with CEACAM5 HE.
Collapse
Affiliation(s)
| | - Julien Adam
- International Thoracic Cancer Center, Inserm U1186, Gustave Roussy, Villejuif, France
| | - Céline Nicolazzi
- Sanofi Research and Development, Sanofi, Vitry-sur-Seine, France
| | | | - Florence Attenot
- Sanofi Research and Development, Sanofi, Vitry-sur-Seine, France
| | | | - Colette Dib
- Sanofi Research and Development, Sanofi, Vitry-sur-Seine, France
| | - Nina Masson
- IT&M Stats on behalf of Sanofi, Neuilly-sur-Seine, France
| | - Nils Ternès
- Sanofi Research and Development, Sanofi, Chilly-Mazarin, France
| | | | - Brigitte Demers
- Sanofi Research and Development, Sanofi, Vitry-sur-Seine, France
| | - Mustapha Chadjaa
- Sanofi Research and Development, Sanofi, Vitry-sur-Seine, France
| | - Sukhvinder Sidhu
- Sanofi Research and Development, Sanofi, Vitry-sur-Seine, France
| | - Cécile Combeau
- Sanofi Research and Development, Sanofi, Chilly-Mazarin, France
| | | | - Jean-Yves Scoazec
- Department of Pathology and Laboratory Medicine, Gustave Roussy, Villejuif , France; Faculté de Médecine de Bicêtre, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Souad Naimi
- Sanofi Research and Development, Sanofi, Chilly-Mazarin, France
| | - Eric Angevin
- Faculté de Médecine de Bicêtre, Université Paris-Saclay, Le Kremlin-Bicêtre, France; Drug Development Department (DITEP) and Clinical Research Division, Gustave Roussy, Villejuif, France
| | - Marielle Chiron
- Sanofi Research and Development, Sanofi, Vitry-sur-Seine, France
| | - Christophe Henry
- Sanofi Research and Development, Sanofi, Vitry-sur-Seine, France.
| |
Collapse
|
7
|
McLaughlin J, Berkman J, Nana-Sinkam P. Targeted therapies in non-small cell lung cancer: present and future. Fac Rev 2023; 12:22. [PMID: 37675274 PMCID: PMC10477963 DOI: 10.12703/r/12-22] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/08/2023] Open
Abstract
Lung cancer is the leading cause of malignancy-related death in the United States and the second most common cancer diagnosis worldwide. In the last two decades, lung cancer treatment has evolved to include advances in the development of mutation-based targeting, immunotherapy, radiation therapy, and minimally invasive surgical techniques. The discovery of lung cancer as a molecularly heterogeneous disease has driven investigation into the development of targeted therapies resulting in improved patient outcomes. Despite these advances, there remain opportunities, through further investigation of mechanisms of resistance, to develop novel therapeutics that better direct the personalization of lung cancer therapy. In this review, we highlight developments in the evolution of targeted therapies in non-small cell lung cancer, as well as future directions shaped by emerging patterns of resistance.
Collapse
Affiliation(s)
- Jessica McLaughlin
- Department of Internal Medicine, Division of Pulmonary Disease and Critical Care Medicine, Virginia Commonwealth University Health System, Richmond, VA 23298
| | - Jonathan Berkman
- Department of Internal Medicine, Division of Hematology, Oncology and Palliative Care, Virginia Commonwealth University Health System, Richmond, VA 23298
| | - Patrick Nana-Sinkam
- Department of Internal Medicine, Division of Pulmonary Disease and Critical Care Medicine, Virginia Commonwealth University Health System, Richmond, VA 23298
| |
Collapse
|
8
|
Zhang J, Yang T, Han M, Wang X, Yang W, Guo N, Ren Y, Cui W, Li S, Zhao Y, Zhai X, Jia L, Yang J, Wu C, Wang L. Gain-of-function mutations in the catalytic domain of DOT1L promote lung cancer malignant phenotypes via the MAPK/ERK signaling pathway. SCIENCE ADVANCES 2023; 9:eadc9273. [PMID: 37256945 PMCID: PMC10413674 DOI: 10.1126/sciadv.adc9273] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 04/25/2023] [Indexed: 06/02/2023]
Abstract
Lung cancer is a lethal malignancy lacking effective therapies. Emerging evidence suggests that epigenetic enzyme mutations are closely related to the malignant phenotype of lung cancer. Here, we identified a series of gain-of-function mutations in the histone methyltransferase DOT1L. The strongest of them is R231Q, located in the catalytic DOT domain. R231Q can enhance the substrate binding ability of DOT1L. Moreover, R231Q promotes cell growth and drug resistance of lung cancer cells in vitro and in vivo. Mechanistic studies also revealed that the R231Q mutant specifically activates the MAPK/ERK signaling pathway by enriching H3K79me2 on the RAF1 promoter and epigenetically regulating the expression of downstream targets. The combination of a DOT1L inhibitor (SGC0946) and a MAPK/ERK axis inhibitor (binimetinib) can effectively reverse the R231Q-induced phenomena. Our results reveal gain-of-function mutations in an epigenetic enzyme and provide promising insights for the precise treatment of lung cancer patients.
Collapse
Affiliation(s)
- Jiayu Zhang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, China
- Benxi Institute of Pharmaceutical Research, Shenyang Pharmaceutical University, Benxi 117004, China
| | - Ting Yang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, China
- Benxi Institute of Pharmaceutical Research, Shenyang Pharmaceutical University, Benxi 117004, China
| | - Mei Han
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, China
- Benxi Institute of Pharmaceutical Research, Shenyang Pharmaceutical University, Benxi 117004, China
| | - Xiaoxuan Wang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, China
- Benxi Institute of Pharmaceutical Research, Shenyang Pharmaceutical University, Benxi 117004, China
| | - Weiming Yang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, China
- Benxi Institute of Pharmaceutical Research, Shenyang Pharmaceutical University, Benxi 117004, China
| | - Ning Guo
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, China
- Benxi Institute of Pharmaceutical Research, Shenyang Pharmaceutical University, Benxi 117004, China
| | - Yong Ren
- Department of Pathology, General Hospital of Central Theater Command of People's Liberation Army, Wuhan 430070, China
| | - Wei Cui
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Shangxiao Li
- Department of Biochemistry and Molecular Biology, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yongshan Zhao
- Department of Biochemistry and Molecular Biology, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xin Zhai
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Lina Jia
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jingyu Yang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Chunfu Wu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Lihui Wang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, China
- Benxi Institute of Pharmaceutical Research, Shenyang Pharmaceutical University, Benxi 117004, China
| |
Collapse
|
9
|
Julian C, Pal N, Gershon A, Evangelista M, Purkey H, Lambert P, Shi Z, Zhang Q. Overall survival in patients with advanced non-small cell lung cancer with KRAS G12C mutation with or without STK11 and/or KEAP1 mutations in a real-world setting. BMC Cancer 2023; 23:352. [PMID: 37069542 PMCID: PMC10108521 DOI: 10.1186/s12885-023-10778-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 03/27/2023] [Indexed: 04/19/2023] Open
Abstract
BACKGROUND KRAS mutations occur frequently in advanced non-small cell lung cancer (aNSCLC); the G12C mutation is the most prevalent. Alterations in STK11 or KEAP1 commonly co-occur with KRAS mutations in aNSCLC. Using real-world data, we assessed the effect of KRAS G12C mutation with or without STK11 and/or KEAP1 mutations on overall survival (OS) in patients with aNSCLC receiving cancer immunotherapy (CIT), chemotherapy, or both in first line (1L) and second line (2L). METHODS Patients diagnosed with aNSCLC between January 2011 and March 2020 in a clinico-genomic database were included. Cox proportional hazards models adjusted for left truncation, baseline demographics and clinical characteristics were used to analyze the effect of STK11 and/or KEAP1 co-mutational status on OS in patients with KRAS wild-type (WT) or G12C mutation. RESULTS Of 2715 patients with aNSCLC without other actionable driver mutations, 1344 (49.5%) had KRAS WT cancer, and 454 (16.7%) had KRAS G12C-positive cancer. At 1L treatment start, significantly more patients with KRAS G12C-positive cancer were female, smokers, and had non-squamous histology, a higher prevalence of metastasis and programmed death-ligand 1 positivity than those with KRAS WT cancer. Median OS was comparable between patients with KRAS G12C-positive and KRAS WT cancer when receiving chemotherapy or combination CIT and chemotherapy in the 1L or 2L. Median OS was numerically longer in patients with KRAS G12C vs KRAS WT cancer treated with 1L CIT (30.2 vs 10.6 months, respectively) or 2L CIT (11.3 vs 7.6 months, respectively). Co-mutation of STK11 and KEAP1 was associated with significantly shorter OS in patients receiving any type of 1L therapy, regardless of KRAS G12C mutational status. CONCLUSIONS This real-world study showed that patients with KRAS G12C-positive or KRAS WT cancer have similar OS in the 1L or 2L when treated with chemotherapy or combination CIT and chemotherapy. In contrast to aNSCLC patients with EGFR or ALK driver mutations, patients with KRAS G12C-positive cancer may benefit from CIT monotherapy. Co-mutation of STK11 and KEAP1 was associated with significantly shorter survival, independent of KRAS G12C mutational status, reflecting the poor prognosis and high unmet need in this patient population.
Collapse
Affiliation(s)
- Cristina Julian
- 1 DNA Way, Genentech, Inc, South San Francisco, CA, 94080, USA
| | - Navdeep Pal
- 1 DNA Way, Genentech, Inc, South San Francisco, CA, 94080, USA
| | - Anda Gershon
- 1 DNA Way, Genentech, Inc, South San Francisco, CA, 94080, USA
| | | | - Hans Purkey
- 1 DNA Way, Genentech, Inc, South San Francisco, CA, 94080, USA
| | - Peter Lambert
- 1 DNA Way, Genentech, Inc, South San Francisco, CA, 94080, USA
| | - Zhen Shi
- 1 DNA Way, Genentech, Inc, South San Francisco, CA, 94080, USA.
| | - Qing Zhang
- 1 DNA Way, Genentech, Inc, South San Francisco, CA, 94080, USA.
| |
Collapse
|
10
|
Cho BC, Simi A, Sabari J, Vijayaraghavan S, Moores S, Spira A. Amivantamab, an Epidermal Growth Factor Receptor (EGFR) and Mesenchymal-epithelial Transition Factor (MET) Bispecific Antibody, Designed to Enable Multiple Mechanisms of Action and Broad Clinical Applications. Clin Lung Cancer 2023; 24:89-97. [PMID: 36481319 DOI: 10.1016/j.cllc.2022.11.004] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 11/02/2022] [Accepted: 11/02/2022] [Indexed: 11/13/2022]
Abstract
Substantial therapeutic advancements have been made in identifying and treating activating mutations in advanced non-small cell lung cancer (NSCLC); however, resistance to epidermal growth factor receptor (EGFR) and mesenchymal-epithelial transition factor (MET) inhibitors remains common with current targeted therapies. Amivantamab, a fully human bispecific antibody targeting EGFR and MET, is approved in the United States and other countries for the treatment of patients with advanced NSCLC with EGFR exon 20 insertion mutations, for whom disease has progressed on or after platinum-based chemotherapy. Preliminary efficacy and safety have also been demonstrated in patients with common EGFR- or MET-mutated NSCLC. Amivantamab employs 3 distinct potential mechanisms of action (MOAs) including ligand blocking, receptor degradation, and immune cell-directing activity, such as antibody-dependent cellular cytotoxicity and trogocytosis. Notably, efficacy with amivantamab does not require all 3 MOAs to occur simultaneously, broadening applicability by using diverse antitumor mechanisms. This review focuses on the molecular characteristics of amivantamab and its unique MOAs leading to in vitro and in vivo efficacy and safety in preclinical and clinical studies.
Collapse
Affiliation(s)
- Byoung Chul Cho
- Division of Medical Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, 50-1 Yonsei-ro Sinchon-dong, Seodaemun-gu, Seoul, South Korea.
| | - Allison Simi
- Janssen Scientific Affairs, LLC, 800 Ridgeview Drive, Horsham, PA
| | - Joshua Sabari
- NYU Langone Health, 160 E 34th St 8th floor, New York, NY
| | | | - Sheri Moores
- Janssen Research & Development, LLC, 1400 McKean Road, Spring House, PA
| | | |
Collapse
|
11
|
Bates M, Mohamed BM, Ward MP, Kelly TE, O'Connor R, Malone V, Brooks R, Brooks D, Selemidis S, Martin C, O'Toole S, O'Leary JJ. Circulating tumour cells: The Good, the Bad and the Ugly. Biochim Biophys Acta Rev Cancer 2023; 1878:188863. [PMID: 36796527 DOI: 10.1016/j.bbcan.2023.188863] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 01/06/2023] [Accepted: 01/21/2023] [Indexed: 02/17/2023]
Abstract
This review is an overview of the current knowledge regarding circulating tumour cells (CTCs), which are potentially the most lethal type of cancer cell, and may be a key component of the metastatic cascade. The clinical utility of CTCs (the "Good"), includes their diagnostic, prognostic, and therapeutic potential. Conversely, their complex biology (the "Bad"), including the existence of CD45+/EpCAM+ CTCs, adds insult to injury regarding their isolation and identification, which in turn hampers their clinical translation. CTCs are capable of forming microemboli composed of both non-discrete phenotypic populations such as mesenchymal CTCs and homotypic and heterotypic clusters which are poised to interact with other cells in the circulation, including immune cells and platelets, which may increase their malignant potential. These microemboli (the "Ugly") represent a prognostically important CTC subset, however, phenotypic EMT/MET gradients bring additional complexities to an already challenging situation.
Collapse
Affiliation(s)
- Mark Bates
- Department of Histopathology, Trinity College Dublin, Dublin 2, Ireland; Emer Casey Molecular Pathology Research Laboratory, Coombe Women & Infants University Hospital, Dublin 8, Ireland; Trinity St James's Cancer Institute, Dublin 8, Ireland.
| | - Bashir M Mohamed
- Department of Histopathology, Trinity College Dublin, Dublin 2, Ireland; Emer Casey Molecular Pathology Research Laboratory, Coombe Women & Infants University Hospital, Dublin 8, Ireland; Trinity St James's Cancer Institute, Dublin 8, Ireland
| | - Mark P Ward
- Department of Histopathology, Trinity College Dublin, Dublin 2, Ireland; Emer Casey Molecular Pathology Research Laboratory, Coombe Women & Infants University Hospital, Dublin 8, Ireland; Trinity St James's Cancer Institute, Dublin 8, Ireland
| | - Tanya E Kelly
- Department of Histopathology, Trinity College Dublin, Dublin 2, Ireland; Emer Casey Molecular Pathology Research Laboratory, Coombe Women & Infants University Hospital, Dublin 8, Ireland; Trinity St James's Cancer Institute, Dublin 8, Ireland
| | - Roisin O'Connor
- Department of Histopathology, Trinity College Dublin, Dublin 2, Ireland; Emer Casey Molecular Pathology Research Laboratory, Coombe Women & Infants University Hospital, Dublin 8, Ireland; Trinity St James's Cancer Institute, Dublin 8, Ireland; Department of Pathology, Coombe Women & Infants University Hospital, Dublin 8, Ireland
| | - Victoria Malone
- Department of Histopathology, Trinity College Dublin, Dublin 2, Ireland; Emer Casey Molecular Pathology Research Laboratory, Coombe Women & Infants University Hospital, Dublin 8, Ireland; Trinity St James's Cancer Institute, Dublin 8, Ireland; Department of Pathology, Coombe Women & Infants University Hospital, Dublin 8, Ireland
| | - Robert Brooks
- Cancer Research Institute, University of South Australia, Adelaide, SA 5001, Australia
| | - Doug Brooks
- Department of Histopathology, Trinity College Dublin, Dublin 2, Ireland; Trinity St James's Cancer Institute, Dublin 8, Ireland; Cancer Research Institute, University of South Australia, Adelaide, SA 5001, Australia
| | - Stavros Selemidis
- School of Health and Biomedical Sciences, Royal Melbourne Institute of Technology, Bundoora, VIC 3083, Australia
| | - Cara Martin
- Department of Histopathology, Trinity College Dublin, Dublin 2, Ireland; Emer Casey Molecular Pathology Research Laboratory, Coombe Women & Infants University Hospital, Dublin 8, Ireland; Trinity St James's Cancer Institute, Dublin 8, Ireland; Department of Pathology, Coombe Women & Infants University Hospital, Dublin 8, Ireland
| | - Sharon O'Toole
- Department of Histopathology, Trinity College Dublin, Dublin 2, Ireland; Emer Casey Molecular Pathology Research Laboratory, Coombe Women & Infants University Hospital, Dublin 8, Ireland; Trinity St James's Cancer Institute, Dublin 8, Ireland; Department of Obstetrics and Gynaecology, Trinity College Dublin, Dublin 2, Ireland
| | - John J O'Leary
- Department of Histopathology, Trinity College Dublin, Dublin 2, Ireland; Emer Casey Molecular Pathology Research Laboratory, Coombe Women & Infants University Hospital, Dublin 8, Ireland; Trinity St James's Cancer Institute, Dublin 8, Ireland; Department of Pathology, Coombe Women & Infants University Hospital, Dublin 8, Ireland
| |
Collapse
|
12
|
Wang Z, Liu X, Wang W, Xu J, Sun H, Wei J, Yu Y, Zhao Y, Wang X, Liao Z, Sun W, Jia L, Zhang Y. UPLC-MS based integrated plasma proteomic and metabolomic profiling of TSC-RAML and its relationship with everolimus treatment. Front Mol Biosci 2023; 10:1000248. [PMID: 36891236 PMCID: PMC9986496 DOI: 10.3389/fmolb.2023.1000248] [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: 07/21/2022] [Accepted: 02/10/2023] [Indexed: 02/22/2023] Open
Abstract
Aim: To profile the plasma proteomics and metabolomics of patients with renal cysts, sporadic angiomyolipoma (S-AML) and tuberous sclerosis complex related angiomyolipoma (TSC-RAML) before and after everolimus treatment, and to find potential diagnostic and prognostic biomarkers as well as reveal the underlying mechanism of TSC tumorigenesis. Materials and Methods: We retrospectively measured the plasma proteins and metabolites from November 2016 to November 2017 in a cohort of pre-treatment and post-treatment TSC-RAML patients and compared them with renal cyst and S-AML patients by ultra-performance liquid chromatography-mass spectrometer (UPLC-MS). The tumor reduction rates of TSC-RAML were assessed and correlated with the plasma protein and metabolite levels. In addition, functional analysis based on differentially expressed molecules was performed to reveal the underlying mechanisms. Results: Eighty-five patients with one hundred and ten plasma samples were enrolled in our study. Multiple proteins and metabolites, such as pre-melanosome protein (PMEL) and S-adenosylmethionine (SAM), demonstrated both diagnostic and prognostic effects. Functional analysis revealed many dysregulated pathways, including angiogenesis synthesis, smooth muscle proliferation and migration, amino acid metabolism and glycerophospholipid metabolism. Conclusion: The plasma proteomics and metabolomics pattern of TSC-RAML was clearly different from that of other renal tumors, and the differentially expressed plasma molecules could be used as prognostic and diagnostic biomarkers. The dysregulated pathways, such as angiogenesis and amino acid metabolism, may shed new light on the treatment of TSC-RAML.
Collapse
Affiliation(s)
- Zhan Wang
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Xiaoyan Liu
- School of Basic Medical College, Core facility of instrument, Institution of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Wenda Wang
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Jiyu Xu
- School of Basic Medical College, Core facility of instrument, Institution of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Haidan Sun
- School of Basic Medical College, Core facility of instrument, Institution of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Jing Wei
- Clinical Research Center, National Center for Children's Health, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Yuncui Yu
- Clinical Research Center, National Center for Children's Health, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Yang Zhao
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Xu Wang
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Zhangcheng Liao
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Wei Sun
- School of Basic Medical College, Core facility of instrument, Institution of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Lulu Jia
- Clinical Research Center, National Center for Children's Health, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Yushi Zhang
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| |
Collapse
|
13
|
Al Achkar M, Basu Roy U, Manley E, Standifer M, Baik C, Walsh CA. A qualitative study of interactions with oncologists among patients with advanced lung cancer. Support Care Cancer 2022; 30:9049-9055. [PMID: 35948849 PMCID: PMC9365681 DOI: 10.1007/s00520-022-07309-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 07/27/2022] [Indexed: 11/04/2022]
Abstract
INTRODUCTION To support the care of lung cancer patients, oncologists have needed to stay current on treatment advancements and build relationships with a new group of survivors in an era where lung cancer survivorship has been re-defined. The objectives of the study were to (1) understand the perspectives of advanced lung cancer patients whose tumors have oncogenic alterations about their care experiences with their oncologist(s) and (2) describe the perceptions of advanced lung cancer patients about seeking second opinions and navigating care decisions. METHODS In this qualitative study, patients with advanced lung cancer (n = 25) on targeted therapies were interviewed to discuss their ongoing experience with their oncologists. We used deductive and inductive qualitative approaches in the coding of the data. We organized the data using the self-determination framework. RESULTS Patients described both positive and negative aspects of their care as related to autonomy, provider competency, and connectedness. Patients sought second opinions for three primary reasons: expertise, authoritative advice, and access to clinical trial opportunities. When there is disagreement in the treatment plan between the primary oncologist and the specialist, there can be confusion and tension, and patients have to make difficult choices about their path forward. CONCLUSIONS Patients value interactions that support their autonomy, demonstrate the competency of their providers, and foster connectedness. To ensure that patients receive quality and goal-concordant care, developing decision aids and education materials that help patients negotiate recommendations from two providers is an area that deserves further attention.
Collapse
Affiliation(s)
- Morhaf Al Achkar
- Department of Family Medicine, University of Washington, School of Medicine, Box 356390, Seattle, WA, 98195, USA.
| | | | | | - Maisha Standifer
- LUNGevity Foundation, Chicago, IL, USA
- Satcher Health Leadership Institute, Morehouse School of Medicine, Atlanta, GA, USA
| | - Christina Baik
- Department of Family Medicine, University of Washington, School of Medicine, Box 356390, Seattle, WA, 98195, USA
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Casey A Walsh
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Orthopaedics & Sports Medicine, University of Washington School of Medicine, Seattle, WA, USA
| |
Collapse
|
14
|
Chow YP, Zainul Abidin N, Kow KS, Tho LM, Wong CL. Analytical and clinical validation of a custom 15-gene next-generation sequencing panel for the evaluation of circulating tumor DNA mutations in patients with advanced non-small-cell lung cancer. PLoS One 2022; 17:e0276161. [PMID: 36256645 PMCID: PMC9578623 DOI: 10.1371/journal.pone.0276161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 09/30/2022] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND This is a pilot proof-of-concept study to evaluate the utility of a custom 15-gene circulating tumor DNA (ctDNA) panel as a potential companion molecular next-generation sequencing (NGS) assay for identifying somatic single nucleotide variants and indels in non-small-cell lung cancer (NSCLC) patients. The custom panel covers the hotspot mutations in EGFR, KRAS, NRAS, BRAF, PIK3CA, ERBB2, MET, KIT, PDGFRA, ALK, ROS1, RET, NTRK1, NTRK2 and NTRK3 genes which serve as biomarkers for guiding treatment decisions in NSCLC patients. METHOD The custom 15-gene ctDNA NGS panel was designed using ArcherDX Assay Designer. A total of 20 ng or 50 ng input ctDNA was used to construct the libraries. The analytical performance was evaluated using reference standards at different allellic frequencies (0.1%, 1%, 5% and parental). The clinical performance was evaluated using plasma samples collected from 10 treatment naïve advanced stage III or IV NSCLC patients who were tested for tissue EGFR mutations. The bioinformatics analysis was performed using the proprietary Archer Analysis Software. RESULTS For the analytical validation, we achieved 100% sensitivity and specificity for the detection of known mutations in the reference standards. The limit of detection was 1% allelic frequency. Clinical validation showed that the clinical sensitivity and specificity of the assay for detecting EGFR mutation were 83.3% and 100% respectively. In addition, the NGS panel also detected other mutations of uncertain significance in 6 out of 10 patients. CONCLUSION This preliminary analysis showed that the custom 15-gene ctDNA NGS panel demonstrated good analytical and clinical performances for the EGFR mutation. Further studies incorporating the validation of other candidate gene mutations are warranted.
Collapse
Affiliation(s)
- Yock Ping Chow
- Clinical Research Centre, Sunway Medical Centre, Petaling Jaya, Selangor Darul Ehsan, Malaysia
| | - Norziha Zainul Abidin
- Molecular Diagnostics Laboratory, Sunway Medical Centre, Petaling Jaya, Selangor Darul Ehsan, Malaysia
| | - Ken Siong Kow
- Department of Medicine, Sunway Medical Centre, Petaling Jaya, Selangor Darul Ehsan, Malaysia
| | - Lye Mun Tho
- Department of Medicine, Sunway Medical Centre, Petaling Jaya, Selangor Darul Ehsan, Malaysia
| | - Chieh Lee Wong
- Clinical Research Centre, Sunway Medical Centre, Petaling Jaya, Selangor Darul Ehsan, Malaysia
- Molecular Diagnostics Laboratory, Sunway Medical Centre, Petaling Jaya, Selangor Darul Ehsan, Malaysia
- Department of Medicine, Sunway Medical Centre, Petaling Jaya, Selangor Darul Ehsan, Malaysia
- Haematology Unit, Department of Medicine, Sunway Medical Centre, Petaling Jaya, Selangor Darul Ehsan, Malaysia
- Centre for Haematology, Hammersmith Hospital, London, United Kingdom
- Faculty of Medicine, Imperial College London, London, United Kingdom
- * E-mail:
| |
Collapse
|
15
|
Qiu Y, Liu L, Yang H, Chen H, Deng Q, Xiao D, Lin Y, Zhu C, Li W, Shao D, Jiang W, Wu K, He J. Intratumor heterogeneity of driver mutations and TMB distribution in 30 early-stage LUAD patients with multiple lesions. Front Oncol 2022; 12:952572. [PMID: 36110964 PMCID: PMC9469651 DOI: 10.3389/fonc.2022.952572] [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: 05/25/2022] [Accepted: 08/05/2022] [Indexed: 11/13/2022] Open
Abstract
Background Differentiating multiple pulmonary lesions as multiple primary lung cancer (MLC) or intra-pulmonary metastasis (IPM) is critical. Lung cancer also has a high genetic heterogeneity, which influenced the treatment strategy. Genetic information may aid in tracing lineage information on multiple lung lesions. This study applied comprehensive genomic profiling to decipher the intrinsic genetics of multiple lung lesions. Methods Sixty-six lung adenocarcinomas (LUAD) tumor lesions (FFEP) archived from 30 patients were included in this study. The 508 cancer-related genes were evaluated by targeted next-generation sequencing (MGI-seq 2000). Results The study included a total of 30 LUADs (66 samples). The majority of tumors demonstrated intra-tumoral heterogeneity. Two hundred twenty-four mutations were detected by sequencing the 66 samples. We investigated the driver gene mutations of NSCLC patients with multiple lesions. EGFR was the most frequently (48/198) mutated driver gene. The codons in EGFR mainly affected by mutations were p.L858R (18/66 [27.3%]) and exon 19del (8/66 [12.1%]). In addition, additional driver genes were found, including TP53, BRAF, ERBB2, MET, and PIK3CA. We also found that the inter-component heterogeneity of different lesions and more than two different mutation types of EGFR were detected in seven patients with two lesions (P3, P10, P24, P25, P28, P29, and P30). The TMB values of different lesions in each patient were different in 26 patients (except P4, P5, P14, and P30). Conclusions Comprehensive genomic profiling should be applied to distinguishing the nature of multiple lung lesions irrespective of radiologic and histologic diagnoses.
Collapse
Affiliation(s)
- Yuan Qiu
- National Clinical Research Center of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Liping Liu
- National Clinical Research Center of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Haihong Yang
- National Clinical Research Center of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Hanzhang Chen
- National Clinical Research Center of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qiuhua Deng
- National Clinical Research Center of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Dakai Xiao
- National Clinical Research Center of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yongping Lin
- National Clinical Research Center of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | | | - Weiwei Li
- BGI Genomics, BGI-Shenzhen, Shenzhen, China
| | - Di Shao
- BGI Genomics, BGI-Shenzhen, Shenzhen, China
| | | | - Kui Wu
- BGI-Shenzhen, Shenzhen, China
| | - Jianxing He
- National Clinical Research Center of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- *Correspondence: Jianxing He,
| |
Collapse
|
16
|
Vikas, Sahu HK, Mehata AK, Viswanadh MK, Priya V, Muthu MS. Dual-receptor-targeted nanomedicines: emerging trends and advances in lung cancer therapeutics. Nanomedicine (Lond) 2022; 17:1375-1395. [PMID: 36317852 DOI: 10.2217/nnm-2021-0470] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Cancer is the leading cause of mortality worldwide. Among all cancer types, lung cancer is recognized as the most lethal and highly metastatic. The application of targeted nanomedicine loaded with anticancer drugs is highly desirable for successful lung cancer treatment. However, due to the heterogenicity and complexity of lung cancer, the therapeutic effectiveness of a single receptor targeting nanomedicine is unfortunately limited. Therefore, the concept of dual-receptor-targeted nanomedicine is an emerging trend for the advancement in lung cancer therapeutics. In this review, the authors discuss various single- and dual-receptor-targeted nanomedicines that have been developed for lung cancer treatment. Furthermore, the authors also discussed all the types of receptors that can be utilized in combination for the development of dual-receptor-targeted nanomedicines.
Collapse
Affiliation(s)
- Vikas
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh, 221005, India
| | - Hemendra Kumar Sahu
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh, 221005, India
| | - Abhishesh Kumar Mehata
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh, 221005, India
| | - Matte Kasi Viswanadh
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh, 221005, India
| | - Vishnu Priya
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh, 221005, India
| | - Madaswamy S Muthu
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh, 221005, India
| |
Collapse
|
17
|
Viswanathan VS, Toro P, Corredor G, Mukhopadhyay S, Madabhushi A. The state of the art for artificial intelligence in lung digital pathology. J Pathol 2022; 257:413-429. [PMID: 35579955 PMCID: PMC9254900 DOI: 10.1002/path.5966] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 04/26/2022] [Accepted: 05/15/2022] [Indexed: 12/03/2022]
Abstract
Lung diseases carry a significant burden of morbidity and mortality worldwide. The advent of digital pathology (DP) and an increase in computational power have led to the development of artificial intelligence (AI)-based tools that can assist pathologists and pulmonologists in improving clinical workflow and patient management. While previous works have explored the advances in computational approaches for breast, prostate, and head and neck cancers, there has been a growing interest in applying these technologies to lung diseases as well. The application of AI tools on radiology images for better characterization of indeterminate lung nodules, fibrotic lung disease, and lung cancer risk stratification has been well documented. In this article, we discuss methodologies used to build AI tools in lung DP, describing the various hand-crafted and deep learning-based unsupervised feature approaches. Next, we review AI tools across a wide spectrum of lung diseases including cancer, tuberculosis, idiopathic pulmonary fibrosis, and COVID-19. We discuss the utility of novel imaging biomarkers for different types of clinical problems including quantification of biomarkers like PD-L1, lung disease diagnosis, risk stratification, and prediction of response to treatments such as immune checkpoint inhibitors. We also look briefly at some emerging applications of AI tools in lung DP such as multimodal data analysis, 3D pathology, and transplant rejection. Lastly, we discuss the future of DP-based AI tools, describing the challenges with regulatory approval, developing reimbursement models, planning clinical deployment, and addressing AI biases. © 2022 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
| | - Paula Toro
- Department of PathologyCleveland ClinicClevelandOHUSA
| | - Germán Corredor
- Department of Biomedical EngineeringCase Western Reserve UniversityClevelandOHUSA
- Louis Stokes Cleveland VA Medical CenterClevelandOHUSA
| | | | - Anant Madabhushi
- Department of Biomedical EngineeringCase Western Reserve UniversityClevelandOHUSA
- Louis Stokes Cleveland VA Medical CenterClevelandOHUSA
| |
Collapse
|
18
|
TCRP1 activated by mutant p53 promotes NSCLC proliferation via inhibiting FOXO3a. Oncogenesis 2022; 11:19. [PMID: 35459265 PMCID: PMC9033812 DOI: 10.1038/s41389-022-00392-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 02/17/2022] [Accepted: 03/24/2022] [Indexed: 11/08/2022] Open
Abstract
Previously, our lab explored that tongue cancer resistance-associated protein (TCRP1) plays a central role in cancer chemo-resistance and progression. Absolutely, TCRP1 was significantly increased in lung cancer. But the mechanism is far from elucidated. Here, we found that TCRP1 was increased in p53-mutant non-small-cell lung cancer (NSCLC), comparing to that in NSCLC with wild type p53. Further study showed that mutant p53 couldn't bind to the promoter of TCRP1 to inhibit its expression. While the wild type p53 did so. Next, loss-and gain-of-function assays demonstrated that TCRP1 promoted cell proliferation and tumor growth in NSCLC. Regarding the mechanism, TCRP1 encouraged AKT phosphorylation and blocked FOXO3a nuclear localization through favoring FOXO3a ubiquitination in cytoplasm, thus, promoted cell cycle progression. Conclusionly, TCRP1 was upregulated in NSCLC cells with mutant p53. TCRP1 promoted NSCLC progression via regulating cell cycle.
Collapse
|
19
|
Xu J, Pu Y, Lin R, Xiao S, Fu Y, Wang T. PEAC: An Ultrasensitive and Cost-Effective MRD Detection System in Non-small Cell Lung Cancer Using Plasma Specimen. Front Med (Lausanne) 2022; 9:822200. [PMID: 35308511 PMCID: PMC8928926 DOI: 10.3389/fmed.2022.822200] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 01/14/2022] [Indexed: 12/25/2022] Open
Abstract
Circulating tumor DNA (ctDNA), a tumor-derived fraction of cell-free DNA (cfDNA), has emerged as a promising marker in targeted therapy, immunotherapy, and minimal residual disease (MRD) monitoring in postsurgical patients. However, ctDNA level in early-stage cancers and postsurgical patients is very low, which posed many technical challenges to improve the detection rate and sensitivity, especially in the clinical practice of MRD detection. These challenges usually include insufficient DNA input amount, limit of detection (LOD), and high experimental costs. To resolve these challenges, we developed an ultrasensitive ctDNA MRD detection system in this study, namely PErsonalized Analysis of Cancer (PEAC), to simultaneously detect up to 37 mutations, which account for 70–80% non-small cell lung cancer (NSCLC) driver mutations from low plasma sample volume and enables LOD of 0.01% at a single-site level. We demonstrated the high performance achieved by PEAC on both cfDNA reference standards and clinical plasma samples from three NSCLC patient cohorts. For cfDNA reference standards, PEAC achieved a specificity of 99% and a sensitivity of 87% for the mutations at 0.01% allele fraction. In the second cohort, PEAC showed 100% concordance rate between ddPCR and Next-generation sequencing (NGS) among 29 samples. In the third cohort, 22 of 59 patients received EGFR TKI treatment. Among them, three in four patients identified low level actionable gene mutations only by PEAC had partial responses after targeted therapy, demonstrating high ctDNA detection ability of PEAC. Overall, the developed PEAC system can detect the majority of NSCLC driver mutations using 8–10 ml plasma samples, and has the advantages of high detection sensitivity and lower costs compared with the existing technologies such as ddPCR and NGS. These advantages make the PEAC system quite appropriate for ctDNA and MRD detection in early-stage NSCLC and postsurgical recurrence monitoring.
Collapse
Affiliation(s)
- Jianping Xu
- Department of Medical Oncology, Cancer Hospital, Chinese Academy of Medical Sciences, Beijing, China
- Peking Union Medical College, Beijing, China
- *Correspondence: Jianping Xu
| | - Yue Pu
- Department of Research and Development, Hangzhou Repugene Technology Co., Ltd., Hangzhou, China
| | - Rui Lin
- Department of Research and Development, Hangzhou Repugene Technology Co., Ltd., Hangzhou, China
| | - Shanshan Xiao
- Department of Research and Development, Hangzhou Repugene Technology Co., Ltd., Hangzhou, China
| | - Yingxue Fu
- Department of Research and Development, Hangzhou Repugene Technology Co., Ltd., Hangzhou, China
| | - Tao Wang
- Department of Research and Development, Hangzhou Repugene Technology Co., Ltd., Hangzhou, China
- Tao Wang
| |
Collapse
|
20
|
In-silico studies for the development of novel RET inhibitors for cancer treatment. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2021.132040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
21
|
Review of Therapeutic Strategies for Anaplastic Lymphoma Kinase-Rearranged Non-Small Cell Lung Cancer. Cancers (Basel) 2022; 14:cancers14051184. [PMID: 35267492 PMCID: PMC8909087 DOI: 10.3390/cancers14051184] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 02/18/2022] [Accepted: 02/23/2022] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Anaplastic lymphoma kinase (ALK)-rearranged non-small cell lung cancer (NSCLC) was first reported in 2007. Following the development of crizotinib as a tyrosine kinase inhibitor (TKI) targeting ALK, the treatment of advanced NSCLC with ALK-rearrangements has made remarkable progress. Currently, there are five ALK-TKIs approved by the FDA, and the development of new agents, including fourth-generation TKI, is ongoing. Clinical trials with angiogenesis inhibitors and immune checkpoint inhibitors are also underway, and further progress in the treatment of ALK-rearranged advanced NSCLC is expected. The purpose of this manuscript is to provide information on the recent clinical trials of ALK-TKIs, angiogenesis inhibitors, immune checkpoint inhibitors, and chemotherapy, to describe tissue and liquid biopsy as a method to investigate the mechanisms of resistance against ALK-TKIs and suggest a proposed treatment algorithm. Abstract Non-small cell lung cancer (NSCLC) with anaplastic lymphoma kinase rearrangement (ALK) was first reported in 2007. ALK-rearranged NSCLC accounts for about 3–8% of NSCLC. The first-line therapy for ALK-rearranged advanced NSCLC is tyrosine kinase inhibitors (TKI) targeting ALK. Following the development of crizotinib, the first ALK-TKI, patient prognosis has been greatly improved. Currently, five TKIs are approved by the FDA. In addition, clinical trials of the novel TKI, ensartinib, and fourth-generation ALK-TKI for compound ALK mutation are ongoing. Treatment with angiogenesis inhibitors and immune checkpoint inhibitors is also being studied. However, as the disease progresses, cancers tend to develop resistance mechanisms. In addition to ALK mutations, other mechanisms, including the activation of bypass signaling pathways and histological transformation, cause resistance, and the identification of these mechanisms is important in selecting subsequent therapy. Studies on tissue and liquid biopsy have been reported and are expected to be useful tools for identifying resistance mechanisms. The purpose of this manuscript is to provide information on the recent clinical trials of ALK-TKIs, angiogenesis inhibitors, immune checkpoint inhibitors, and chemotherapy to describe tissue and liquid biopsy as a method to investigate the mechanisms of resistance against ALK-TKIs and suggest a proposed treatment algorithm.
Collapse
|
22
|
Molecular Testing and Treatment Strategies in RET-Rearranged NSCLC Patients: Stay on Target to Look Forward. JOURNAL OF MOLECULAR PATHOLOGY 2022. [DOI: 10.3390/jmp3010003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RET alterations are recognized as key oncogenic drivers in different cancer types, including non-small cell lung cancer (NSCLC). Multikinase inhibitors (MKIs) with anti-RET activities resulted in variable efficacy with significant toxicities because of low target specificity. Selective RET kinase inhibitors, such as pralsetinib and selepercatinib, demonstrated high efficacy and favorable tolerability in advanced RET-rearranged NSCLC patients, leading to their introduction in the clinical setting. Among the different approaches available for the identification of RET rearrangements, next-generation sequencing (NGS) assays present substantial advantages in terms of turnaround time and diagnostic accuracy, even if potentially limited by accessibility issues. The recent advent of novel effective targeted therapies raises several questions regarding the emergence of resistance mechanisms and the potential ways to prevent/overcome them. In this review, we discuss molecular testing and treatment strategies to manage RET fusion positive NSCLC patients with a focus on resistance mechanisms and future perspectives in this rapidly evolving scenario.
Collapse
|
23
|
Dogan I, Khanmammadov N, Ahmed MA, Yıldız A, Saip P, Aydiner A, Vatansever S. Crizotinib in Metastatic ALK mutant Non-small Cell Lung Cancer Patients: A Single Centre Experience. CLINICAL CANCER INVESTIGATION JOURNAL 2022. [DOI: 10.51847/87n2fddtb1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
24
|
Zhang SS, Nagasaka M. Spotlight on Sotorasib (AMG 510) for KRAS G12C Positive Non-Small Cell Lung Cancer. LUNG CANCER-TARGETS AND THERAPY 2021; 12:115-122. [PMID: 34675734 PMCID: PMC8504654 DOI: 10.2147/lctt.s334623] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 09/24/2021] [Indexed: 01/07/2023]
Abstract
Mutations in codon 12 of KRAS have been identified in 13% of non-small cell lung cancer patients. Developing targeted therapies against KRASG12C mutation has proven to be challenging due to the abundance of GTP in the cytoplasm, rapid hydrolysis of GTP, and difficulty designing small molecules to achieve sufficient concentration for KRAS inhibition. Based on promising results in both preclinical and clinical trials, sotorasib, a novel KRASG12C inhibitor, was given conditional approval by the FDA in May 2021. The Phase I portion of the clinical trial produced 32% confirmed response with 56% of patients with stable disease. About 91.2% of patients who received the highest dose of 960mg daily achieved disease control. The Phase II portion, which used 960mg daily dosing resulted in 37.1% of patients with confirmed response and 80.6% of patients with disease control. Both phase I and phase II had similar progression-free survival, in 6.3 months and 6.8 months, respectively. In both phases, grade 4 adverse events occurred in only one patient. The most common adverse events were elevations in LFTs, which down-trended upon dose reduction and steroid treatment. While the conditional approval of sotorasib was a major breakthrough for those patients harboring KRASG12C mutations, resistance mutations to sotorasib are increasingly common. Many proposals have been made to address this, such as the use of combination therapy for synthetic lethality, which are producing encouraging results. Here, we explore in further detail the development of sotorasib, its efficacy, mechanism of resistance, and strategies to overcome these resistances.
Collapse
Affiliation(s)
- Shannon S Zhang
- Department of Medicine, University of California Irvine School of Medicine, Orange, CA, USA
| | - Misako Nagasaka
- Department of Medicine, University of California Irvine School of Medicine, Orange, CA, USA.,Chao Family Comprehensive Cancer Center, Orange, CA, USA.,St. Marianna University School of Medicine, Kawasaki, Japan
| |
Collapse
|
25
|
da Cunha IW, de Almeida Coudry R, de Macedo MP, de Assis EACP, Stefani S, Soares FA. A call to action: molecular pathology in Brazil. SURGICAL AND EXPERIMENTAL PATHOLOGY 2021. [DOI: 10.1186/s42047-021-00096-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Abstract
Background
Adoption of molecular pathology in Brazil is currently very limited. Of note, there are no programs for training new molecular pathologists in the country; thus, documents compiling nationally applicable information on molecular pathology are few.
Methods
A selected panel of Brazilian experts in fields related to molecular pathology were provided with a series of relevant questions to address prior to the multi-day conference. Within this conference, each narrative was discussed and edited by the entire group, through numerous drafts and rounds of discussion until a consensus was achieved.
Results
The panel proposes specific and realistic recommendations for implementing molecular pathology in cancer care in Brazil. In creating these recommendations, the authors strived to address all barriers to the widespread use and impediments to access mentioned previously within this manuscript.
Conclusion
This manuscript provides a review of molecular pathology principles as well as the current state of molecular pathology in Brazil. Additionally, the panel proposes practical and actionable recommendations for the implementation of molecular pathology throughout the country in order to increase awareness of the importance molecular pathology in Brazil.
Collapse
|
26
|
Chang CY, Wu KL, Chang YY, Liu YW, Huang YC, Jian SF, Lin YS, Tsai PH, Hung JY, Tsai YM, Hsu YL. The Downregulation of LSAMP Expression Promotes Lung Cancer Progression and Is Associated with Poor Survival Prognosis. J Pers Med 2021; 11:jpm11060578. [PMID: 34202934 PMCID: PMC8234324 DOI: 10.3390/jpm11060578] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 06/15/2021] [Accepted: 06/16/2021] [Indexed: 12/12/2022] Open
Abstract
Lung cancer has been a leading cause of cancer-related death for decades and therapeutic strategies for non-driver mutation lung cancer are still lacking. A novel approach for this type of lung cancer is an emergent requirement. Here we find that loss of LSAMP (Limbic System Associated Membrane Protein), compared to other IgLON family of proteins NTM (Neurotrimin) and OPCML (OPioid-binding Cell adhesion MoLecule), exhibits the strongest prognostic and therapeutic significance in predicting lung adenocarcinoma (LUAD) progression. Lower expression of LSAMP and NTM, but not OPCML, were found in tumor parts compared with normal parts in six LUAD patients, and this was validated by public datasets, Oncomine® and TCGA. The lower expression of LSAMP, but not NTM, was correlated to shorter overall survival. Two epigenetic regulations, including hypermethylation and miR-143-3p upregulation but not copy number variation, were associated with downregulation of LSAMP in LUAD patients. Pathway network analysis showed that NEGR1 (Neuronal Growth Regulator 1) was involved in the regulatory loop of LSAMP. The biologic functions by LSMAP knockdown in lung cancer cells revealed LSMAP was linked to cancer cell migration via epithelial-mesenchymal transition (EMT) but not proliferation nor stemness of LUAD. Our result showed for the first time that LSAMP acts as a potential tumor suppressor in regulating lung cancer. A further deep investigation into the role of LSAMP in lung cancer tumorigenesis would provide therapeutic hope for such affected patients.
Collapse
Affiliation(s)
- Chao-Yuan Chang
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (C.-Y.C.); (K.-L.W.); (Y.-C.H.); (S.-F.J.); (Y.-S.L.); (P.-H.T.); (J.-Y.H.); (Y.-L.H.)
- Department of Anatomy, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Kuan-Li Wu
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (C.-Y.C.); (K.-L.W.); (Y.-C.H.); (S.-F.J.); (Y.-S.L.); (P.-H.T.); (J.-Y.H.); (Y.-L.H.)
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Yung-Yun Chang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Division of General Medicine, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Yu-Wei Liu
- Division of Thoracic Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Yung-Chi Huang
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (C.-Y.C.); (K.-L.W.); (Y.-C.H.); (S.-F.J.); (Y.-S.L.); (P.-H.T.); (J.-Y.H.); (Y.-L.H.)
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Shu-Fang Jian
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (C.-Y.C.); (K.-L.W.); (Y.-C.H.); (S.-F.J.); (Y.-S.L.); (P.-H.T.); (J.-Y.H.); (Y.-L.H.)
| | - Yi-Shiuan Lin
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (C.-Y.C.); (K.-L.W.); (Y.-C.H.); (S.-F.J.); (Y.-S.L.); (P.-H.T.); (J.-Y.H.); (Y.-L.H.)
| | - Pei-Hsun Tsai
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (C.-Y.C.); (K.-L.W.); (Y.-C.H.); (S.-F.J.); (Y.-S.L.); (P.-H.T.); (J.-Y.H.); (Y.-L.H.)
| | - Jen-Yu Hung
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (C.-Y.C.); (K.-L.W.); (Y.-C.H.); (S.-F.J.); (Y.-S.L.); (P.-H.T.); (J.-Y.H.); (Y.-L.H.)
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Ying-Ming Tsai
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (C.-Y.C.); (K.-L.W.); (Y.-C.H.); (S.-F.J.); (Y.-S.L.); (P.-H.T.); (J.-Y.H.); (Y.-L.H.)
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Correspondence:
| | - Ya-Ling Hsu
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (C.-Y.C.); (K.-L.W.); (Y.-C.H.); (S.-F.J.); (Y.-S.L.); (P.-H.T.); (J.-Y.H.); (Y.-L.H.)
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| |
Collapse
|
27
|
Melosky B, Wheatley-Price P, Juergens RA, Sacher A, Leighl NB, Tsao MS, Cheema P, Snow S, Liu G, Card PB, Chu Q. The rapidly evolving landscape of novel targeted therapies in advanced non-small cell lung cancer. Lung Cancer 2021; 160:136-151. [PMID: 34353680 DOI: 10.1016/j.lungcan.2021.06.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/25/2021] [Accepted: 06/02/2021] [Indexed: 01/15/2023]
Abstract
Lung cancer is a highly heterogeneous disease often driven by well-characterized driver mutations. Although the best studied are common alterations in the epidermal growth factor receptor (EGFR) and anaplastic lymphoma kinase (ALK) oncogenes, rapid advances in molecular characterization has led to the development of novel therapeutics that inhibit additional oncogenic alterations in advanced NSCLC. The literature search identified 62 eligible phase I/II clinical trials or integrated analyses of assessing novel targeted agents against the following molecular alterations: ROS1-rearranged, BRAF V600E-mutant, NTRK-rearranged, MET-altered, uncommon EGFR-mutant, RET-rearranged, HER2-positive, KRAS G12C-mutant and NRG1-rearranged. This rapidly evolving field has produced many new targeted treatment options and promising outcomes have led to the FDA approval of seven novel agents for use in ROS1-rearranged, BRAF V600E-mutant, NTRK-rearranged, MET exon 14 skipping-mutant or RET-rearranged advanced NSCLC. Research continues at a rapid pace, with a number of phase III trials underway to fully evaluate new promising agents under development for improving outcomes in patients with NSCLC harboring distinct molecular subtypes. This review will provide a comprehensive summary of existing data as well as a user-friendly guide on the current status of novel targeted therapy in oncogene-driven advanced NSCLC.
Collapse
Affiliation(s)
- Barbara Melosky
- Medical Oncology, BCCA - 600 W 10th Ave, Vancouver, BC, V5Z 4E6, Canada.
| | - Paul Wheatley-Price
- Ottawa Hospital Research Institute, University of Ottawa, 501 Smyth Box 511, Ottawa, ON, K1H 8L6, Canada
| | - Rosalyn A Juergens
- Juravinski Cancer Centre, McMaster University, 699 Concession Street, Hamilton, ON, L8V5C2, Canada
| | - Adrian Sacher
- Princess Margaret Cancer Centre, University of Toronto, 101 College Street, Toronto, ON, M5G1L7, Canada
| | - Natasha B Leighl
- Princess Margaret Cancer Centre, University of Toronto, 7-913 700 University Avenue, Toronto, ON, M5G1Z5, Canada
| | - Ming-Sound Tsao
- Princess Margaret Cancer Centre, University of Toronto, 101 College Street, Toronto, ON, M5G1L7, Canada
| | - Parneet Cheema
- William Osler Health System, University of Toronto, 101 Humber College Blvd, Etobicoke, ON, M9V 1R8, Canada
| | - Stephanie Snow
- QEII Health Sciences Centre, Dalhousie University, 1276 South Park Street Halifax, NS, B3H 2Y9, Canada
| | - Geoffrey Liu
- Princess Margaret Cancer Centre, University of Toronto, 101 College Street, Toronto, ON, M5G1L7, Canada
| | - Paul B Card
- Kaleidoscope Strategic Inc., 146 Marion St., Toronto, ON, M6R 1E7, Canada
| | - Quincy Chu
- Cross Cancer Institute, University of Alberta, 11560 University Ave, 2nd Floor, Edmonton, AB, T6G 1Z2, Canada
| |
Collapse
|
28
|
El Husseini K, Wislez M. The rising challenge of oncogene addiction in lung cancer. Bull Cancer 2021; 108:559-561. [PMID: 34144763 DOI: 10.1016/j.bulcan.2021.04.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 04/23/2021] [Indexed: 12/24/2022]
Affiliation(s)
- Kinan El Husseini
- Hôpital Cochin, Groupe Hospitalier HUPC, Department of Pneumology, Thoracic Oncology Unit, AP-HP, 27, rue du Faubourg Saint-Jacques, Paris, France
| | - Marie Wislez
- Hôpital Cochin, Groupe Hospitalier HUPC, Department of Pneumology, Thoracic Oncology Unit, AP-HP, 27, rue du Faubourg Saint-Jacques, Paris, France.
| |
Collapse
|
29
|
Wu R, Yuan B, Li C, Wang Z, Song Y, Liu H. A narrative review of advances in treatment and survival prognosis of HER2-positive malignant lung cancers. J Thorac Dis 2021; 13:3708-3720. [PMID: 34277062 PMCID: PMC8264687 DOI: 10.21037/jtd-20-3265] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 04/14/2021] [Indexed: 12/15/2022]
Abstract
Human epidermal growth factor receptor 2 (HER2), as a receptor tyrosine kinase of EGF receptor family, whose mutation is often associated with even if less frequency but poor prognosis and shorter survival in pulmonary malignant tumor. HER2 status include mutation, overexpression, amplification and also some rare genotypes, detected by next generation sequencing (NGS), immunohistochemistry (IHC), and also fluorescence in situ hybridization (FISH). Different genotypes represent different therapeutic targets and indicate different clinical prognosis concluded by previous studies. Unfortunately, no standard guidelines for first-line treatment are widely recognized, and current therapeutic schedules include chemotherapy, radiotherapy, targeted therapy, and immunotherapy. Especially for patients with advanced metastasis, chemotherapy is based as a systemic therapy using studies of breast cancer or EGFR-positive lung adenocarcinoma as a template. Studies already explored treatment including EGFR tyrosine kinase inhibitors (TKIs) such as gefitinib and afatinib, and also trastuzumab and its conjugation like HER2-targeted antibody-drug conjugate trastuzumab emtansine (T-DM1) and conjugate trastuzumab deruxtecan (T-DXd). Also, he researches explored combination therapy with chemotherapy and TKIs or monoclonal antibodies. This review describes commonly used therapies for HER2-positive/HER2-overexpression patients and general relationship between genotypes of HER2, drug selection and final prognosis in order to provide suggestions for future diagnosis and treatment.
Collapse
Affiliation(s)
- Ranpu Wu
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Southeast University of Medicine, Nanjing, China
| | - Bingxiao Yuan
- Department of Respiratory and Critical Care Medicine, Affiliated Jinling Hospital, Medical School of Nanjing Medical University, Nanjing, China
| | - Chuling Li
- Department of Respiratory and Critical Care Medicine, Affiliated Jinling Hospital, Medical School of Nanjing Medical University, Nanjing, China
| | - Zimu Wang
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Yong Song
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Southeast University of Medicine, Nanjing, China.,Department of Respiratory and Critical Care Medicine, Affiliated Jinling Hospital, Medical School of Nanjing Medical University, Nanjing, China
| | - Hongbing Liu
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Southeast University of Medicine, Nanjing, China.,Department of Respiratory and Critical Care Medicine, Affiliated Jinling Hospital, Medical School of Nanjing Medical University, Nanjing, China
| |
Collapse
|
30
|
Cucchiara F, Petrini I, Romei C, Crucitta S, Lucchesi M, Valleggi S, Scavone C, Capuano A, De Liperi A, Chella A, Danesi R, Del Re M. Combining liquid biopsy and radiomics for personalized treatment of lung cancer patients. State of the art and new perspectives. Pharmacol Res 2021; 169:105643. [PMID: 33940185 DOI: 10.1016/j.phrs.2021.105643] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 04/22/2021] [Accepted: 04/22/2021] [Indexed: 12/11/2022]
Abstract
Lung cancer has become a paradigm for precision medicine in oncology, and liquid biopsy (LB) together with radiomics may have a great potential in this scenario. They are both minimally invasive, easy to perform, and can be repeated during patient's follow-up. Also, increasing evidence suggest that LB and radiomics may provide an efficient way to screen and diagnose tumors at an early stage, including the monitoring of any change in the tumor molecular profile. This could allow treatment optimization, improvement of patients' quality of life, and healthcare-related costs reduction. Latest reports on lung cancer patients suggest a combination of these two strategies, along with cutting-edge data analysis, to decode valuable information regarding tumor type, aggressiveness, progression, and response to treatment. The approach seems more compatible with clinical practice than the current standard, and provides new diagnostic companions being able to suggest the best treatment strategy compared to conventional methods. To implement radiomics and liquid biopsy directly into clinical practice, an artificial intelligence (AI)-based system could help to link patients' clinical data together with tumor molecular profiles and imaging characteristics. AI could also solve problems and limitations related to LB and radiomics methodologies. Further work is needed, including new health policies and the access to large amounts of high-quality and well-organized data, allowing a complementary and synergistic combination of LB and imaging, to provide an attractive choice e in the personalized treatment of lung cancer.
Collapse
Affiliation(s)
- Federico Cucchiara
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Pisa, Italy
| | - Iacopo Petrini
- Unit of Pneumology, Department of Translational Research and New Technologies in Medicine, University Hospital of Pisa, Pisa, Italy
| | - Chiara Romei
- Unit II of Radio-diagnostics, Department of Diagnostic and Imaging, University Hospital of Pisa, Pisa, Italy
| | - Stefania Crucitta
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Pisa, Italy
| | - Maurizio Lucchesi
- Unit of Pneumology, Department of Translational Research and New Technologies in Medicine, University Hospital of Pisa, Pisa, Italy
| | - Simona Valleggi
- Unit of Pneumology, Department of Translational Research and New Technologies in Medicine, University Hospital of Pisa, Pisa, Italy
| | - Cristina Scavone
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Annalisa Capuano
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Annalisa De Liperi
- Unit II of Radio-diagnostics, Department of Diagnostic and Imaging, University Hospital of Pisa, Pisa, Italy
| | - Antonio Chella
- Unit of Pneumology, Department of Translational Research and New Technologies in Medicine, University Hospital of Pisa, Pisa, Italy
| | - Romano Danesi
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Pisa, Italy.
| | - Marzia Del Re
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Pisa, Italy
| |
Collapse
|
31
|
Liu S, Polsdofer EV, Zhou L, Ruan S, Lyu H, Hou D, Liu H, Thor AD, He Z, Liu B. Upregulation of endogenous TRAIL-elicited apoptosis is essential for metformin-mediated antitumor activity against TNBC and NSCLC. MOLECULAR THERAPY-ONCOLYTICS 2021; 21:303-314. [PMID: 34141868 PMCID: PMC8167201 DOI: 10.1016/j.omto.2021.04.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 04/24/2021] [Indexed: 12/24/2022]
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) shows promising antitumor activity in preclinical studies. However, the efficacy of recombinant TRAIL in clinical trials is compromised by its short serum half-life and low in vivo stability. Induction of endogenous TRAIL may overcome the limitations and become a new strategy for cancer treatment. Here, we discovered that metformin increased TRAIL expression and induced apoptosis in triple-negative breast cancer (TNBC) and non-small cell lung cancer (NSCLC) cells. Metformin did not alter the expression of TRAIL receptors (TRAIL-R1/DR4 and TRAIL-R2/DR5). Metformin-upregulated TRAIL was secreted into conditioned medium (CM) and found to be functional, since the CM promoted TNBC cells undergoing apoptosis, which was abrogated by a recombinant TRAIL-R2-Fc chimera. Moreover, blockade of TRAIL binding to DR4/DR5 or specific knockdown of TRAIL expression significantly attenuated metformin-induced apoptosis. Studies with a tumor xenograft model revealed that metformin not only significantly inhibited tumor growth but also elicited apoptosis and enhanced TRAIL expression in vivo. Collectively, we have demonstrated that upregulation of TRAIL and activation of death receptor signaling are pivotal for metformin-induced apoptosis in TNBC and NSCLC cells. Our studies identify a novel mechanism of action of metformin exhibiting potent antitumor activity via induction of endogenous TRAIL.
Collapse
Affiliation(s)
- Shuang Liu
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, Guangdong 510095, China.,Department of Interdisciplinary Oncology, Stanley S. Scott Cancer Center, School of Medicine, Louisiana State University (LSU) Health Sciences Center, New Orleans, LA 70112, USA
| | - Erik V Polsdofer
- Department of Pathology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Lukun Zhou
- Department of Interdisciplinary Oncology, Stanley S. Scott Cancer Center, School of Medicine, Louisiana State University (LSU) Health Sciences Center, New Orleans, LA 70112, USA
| | - Sanbao Ruan
- Department of Interdisciplinary Oncology, Stanley S. Scott Cancer Center, School of Medicine, Louisiana State University (LSU) Health Sciences Center, New Orleans, LA 70112, USA
| | - Hui Lyu
- Department of Interdisciplinary Oncology, Stanley S. Scott Cancer Center, School of Medicine, Louisiana State University (LSU) Health Sciences Center, New Orleans, LA 70112, USA
| | - Defu Hou
- Department of Interdisciplinary Oncology, Stanley S. Scott Cancer Center, School of Medicine, Louisiana State University (LSU) Health Sciences Center, New Orleans, LA 70112, USA
| | - Hao Liu
- Department of Interdisciplinary Oncology, Stanley S. Scott Cancer Center, School of Medicine, Louisiana State University (LSU) Health Sciences Center, New Orleans, LA 70112, USA
| | - Ann D Thor
- Department of Pathology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Zhimin He
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, Guangdong 510095, China
| | - Bolin Liu
- Department of Interdisciplinary Oncology, Stanley S. Scott Cancer Center, School of Medicine, Louisiana State University (LSU) Health Sciences Center, New Orleans, LA 70112, USA
| |
Collapse
|
32
|
Camidge DR, Otterson GA, Clark JW, Ignatius Ou SH, Weiss J, Ades S, Shapiro GI, Socinski MA, Murphy DA, Conte U, Tang Y, Wang SC, Wilner KD, Villaruz LC. Crizotinib in Patients With MET-Amplified NSCLC. J Thorac Oncol 2021; 16:1017-1029. [PMID: 33676017 DOI: 10.1016/j.jtho.2021.02.010] [Citation(s) in RCA: 105] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 02/02/2021] [Accepted: 02/09/2021] [Indexed: 12/23/2022]
Abstract
INTRODUCTION MET amplification is a rare, potentially actionable, primary oncogenic driver in patients with NSCLC. METHODS The influence of MET amplification on the clinical activity of the ALK, ROS1, and MET inhibitor, crizotinib (250 mg twice daily), was examined in patients with NSCLC (NCT00585195) who were enrolled into high (≥4 MET-to-CEP7 ratio), medium (>2.2 to <4 MET-to-CEP7 ratio), or low (≥1.8 to ≤2.2 MET-to-CEP7 ratio) amplification categories. Retrospective next-generation sequencing profiling was performed on archival tumor tissue. End points included objective response rate (ORR), duration of response, and progression-free survival. RESULTS A total of 38 patients with a MET-to-CEP7 ratio greater than or equal to 1.8 by local fluorescence in situ hybridization testing received crizotinib. All patients were response-assessable, among whom 21, 14, and 3 had high, medium, and low MET amplification, respectively. ORRs of 8 of 21 (38.1%), 2 of 14 (14.3%), and 1 of 3 (33.3%), median duration of response of 5.2, 3.8, and 12.2 months, and median progression-free survival values of 6.7, 1.9, and 1.8 months were observed for those with high, medium, and low MET amplification, respectively. MET amplification gene copy number greater than or equal to 6 was detected by next-generation sequencing in 15 of 19 (78.9%) analyzable patients. Of these 15 patients, objective responses were observed in six (40%), two of whom had concurrent MET exon 14 alterations. No responses were observed among five patients with concurrent KRAS, BRAF, or EGFR mutations. CONCLUSIONS Patients with high-level, MET-amplified NSCLC responded to crizotinib with the highest ORR. Use of combined diagnostics for MET and other oncogenes may potentially identify patients most likely to respond to crizotinib.
Collapse
Affiliation(s)
| | | | | | | | - Jared Weiss
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Steven Ades
- The University of Vermont Medical Center, Burlington, Vermont
| | - Geoffrey I Shapiro
- Dana-Farber Cancer Institute, Boston, Massachusetts; Brigham and Women's Hospital, Boston, Massachusetts
| | | | | | | | | | | | | | - Liza C Villaruz
- University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center, Pittsburgh, Pennsylvania
| |
Collapse
|
33
|
Verusingam ND, Chen YC, Lin HF, Liu CY, Lee MC, Lu KH, Cheong SK, Han-Kiat Ong A, Chiou SH, Wang ML. Generation of osimertinib-resistant cells from epidermal growth factor receptor L858R/T790M mutant non-small cell lung carcinoma cell line. J Chin Med Assoc 2021; 84:248-254. [PMID: 33009209 DOI: 10.1097/jcma.0000000000000438] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Lung cancer contributes to high cancer mortality worldwide with 80% of total cases diagnosed as non-small cell lung cancer (NSCLC). Epidermal growth factor receptor (EGFR) tyrosine kinase (TK) domain serves as a druggable target in NSCLC patients with exon 19 deletion and L858R mutation. However, patients eventually succumbed to resistance to first- and second-generation EGFR-TK inhibitors through activation of T790M mutation. Third-generation EGFR-TKI, Osimertinib exhibits high efficacy in patients with exon 19 deletion/L858R/T790M mutation but they experienced acquired resistance thereafter. Available treatment options in NSCLC patients remains a challenge due to unknown molecular heterogeneity responsible for acquired resistance to EGFR-TKI. In this study, we aim to generate Osimertinib-resistant (OR) cells from H1975 carrying L858R/T790M double mutation which can be used as a model to elucidate mechanism of resistance. METHODS OR cells were established via stepwise-dose escalation and limiting single-cell dilution method. We then evaluated Osimertinib resistance potential via cell viability assay. Proteins expression related to EGFR-signalling, epithelial to mesenchymal transition (EMT), and autophagy were analyzed via western blot. RESULTS OR cell lines exhibited increased drug resistance potential compared to H1975. Distinguishable mesenchymal-like features were observed in OR cells. Protein expression analysis revealed EGFR-independent signaling involved in the derived OR cells as well as EMT and autophagy activity. CONCLUSION We generated OR cell lines in-vitro as evidenced by increased drug resistance potential, increased mesenchymal features, and enhanced autophagy activity. Development of Osimertinib resistance cells may serve as in-vitro model facilitating discovery of molecular aberration present during acquired mechanism of resistance.
Collapse
Affiliation(s)
- Nalini Devi Verusingam
- Centre for Stem Cell Research, Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Selangor, Malaysia
- Institute of Pharmacology, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
- National Cancer Council (MAKNA), Kuala Lumpur, Malaysia
| | - Yi-Chen Chen
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Heng-Fu Lin
- Division of Thoracic Surgery, Department of Surgery, Far Eastern Memorial Hospital, New Taipei City, Taiwan, ROC
| | - Chao-Yu Liu
- Division of Traumatology, Department of Surgery, Far Eastern Memorial Hospital, New Taipei City, Taiwan, ROC
| | - Ming-Cheng Lee
- Division of Infectious Diseases, Department of Internal Medicine, Cheng-Hsin General Hospital, Taipei, Taiwan, ROC
| | - Kai-Hsi Lu
- Department of Medical Research and Education, Cheng-Hsin General Hospital, Taipei, Taiwan, ROC
| | - Soon-Keng Cheong
- Centre for Stem Cell Research, Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Selangor, Malaysia
- National Cancer Council (MAKNA), Kuala Lumpur, Malaysia
| | - Alan Han-Kiat Ong
- Centre for Stem Cell Research, Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Selangor, Malaysia
| | - Shih-Hwa Chiou
- Institute of Pharmacology, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Stem Cell & Genomic Center, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
- Genomic Research Center, Academia Sinica, Taipei, Taiwan, ROC
| | - Mong-Lien Wang
- Institute of Pharmacology, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Institute of Food Safety and Health Risk Assessment, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| |
Collapse
|
34
|
Li WQ, Li LY, Chai J, Cui JW. Cost-effectiveness analysis of first-line treatments for advanced epidermal growth factor receptor-mutant non-small cell lung cancer patients. Cancer Med 2021; 10:1964-1974. [PMID: 33626238 PMCID: PMC7957173 DOI: 10.1002/cam4.3733] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/01/2020] [Accepted: 01/04/2021] [Indexed: 12/14/2022] Open
Abstract
Objectives Recent studies showed prolonged survival for advanced epidermal growth factor receptor (EGFR)‐mutant non‐small cell lung cancer (NSCLC) patients treated with both monotherapies and combined therapies. However, high costs limit clinical applications. Thus, we conducted this cost‐effectiveness analysis to explore an optimal first‐line treatment for advanced EGFR‐mutant NSCLC patients. Materials and Methods Survival data were extracted from six clinical trials, including ARCHER1050 (dacomitinib vs. gefitinib); FLAURA (osimertinib vs. gefitinib/erlotinib); JO25567 and NEJ026 (bevacizumab +erlotinib vs. erlotinib); NEJ009 (gefitinib +chemotherapy vs. gefitinib); and NCT02148380 (gefitinib +chemotherapy vs. gefitinib vs. chemotherapy) trials. Cost‐related data were obtained from hospitals and published literature. The effect parameter (quality‐adjusted life year [QALY]) was the reflection of both survival and utility. Incremental cost‐effectiveness ratio (ICER), average cost‐effectiveness ratio (ACER), and net benefit were calculated, and the willingness‐to‐pay (WTP) threshold was set at $30828/QALY from the perspective of the Chinese healthcare system. Sensitivity analysis was performed to explore the stability of results. Results We compared treatment groups with control groups in each trial. ICERs were $1897750.74/QALY (ARCHER1050), $416560.02/QALY (FLAURA), ‐$477607.48/QALY (JO25567), ‐$464326.66/QALY (NEJ026), ‐$277121.22/QALY (NEJ009), ‐$399360.94/QALY (gefitinib as comparison, NCT02148380), and ‐$170733.05/QALY (chemotherapy as comparison, NCT02148380). Moreover, ACER and net benefit showed that the combination of EGFR‐TKI with chemotherapy and osimertinib was of more economic benefit following first‐generation EGFR‐TKIs. Sensitivity analyses showed that the impact of utilities and monotherapy could be cost‐effective with a 50% cost reduction. Conclusion First‐generation EGFR‐TKI therapy remained the most cost‐effective treatment option for advanced EGFR‐mutant NSCLC patients. Our results could serve as both a reference for both clinical practice and the formulation of medical insurance reimbursement.
Collapse
Affiliation(s)
- Wen-Qian Li
- Department of Cancer center, the First Hospital of Jilin University, Changchun, China
| | - Ling-Yu Li
- Department of Cancer center, the First Hospital of Jilin University, Changchun, China
| | - Jin Chai
- Department of pharmacy, the Second Hospital of Jilin University, Changchun, China
| | - Jiu-Wei Cui
- Department of Cancer center, the First Hospital of Jilin University, Changchun, China
| |
Collapse
|
35
|
Bebb DG, Banerji S, Blais N, Desmeules P, Gill S, Grin A, Feilotter H, Hansen AR, Hyrcza M, Krzyzanowska M, Melosky B, Noujaim J, Purgina B, Ruether D, Simmons CE, Soulieres D, Torlakovic EE, Tsao MS. Canadian Consensus for Biomarker Testing and Treatment of TRK Fusion Cancer in Adults. Curr Oncol 2021; 28:523-548. [PMID: 33467570 PMCID: PMC7903287 DOI: 10.3390/curroncol28010053] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 12/31/2020] [Accepted: 01/08/2021] [Indexed: 12/13/2022] Open
Abstract
The tyrosine receptor kinase (TRK) inhibitors larotrectinib and entrectinib were recently approved in Canada for the treatment of solid tumours harbouring neurotrophic tyrosine receptor kinase (NTRK) gene fusions. These NTRK gene fusions are oncogenic drivers found in most tumour types at a low frequency (<5%), and at a higher frequency (>80%) in a small number of rare tumours (e.g., secretory carcinoma of the salivary gland and of the breast). They are generally mutually exclusive of other common oncogenic drivers. Larotrectinib and entrectinib have demonstrated impressive overall response rates and tolerability in Phase I/II trials in patients with TRK fusion cancer with no other effective treatment options. Given the low frequency of TRK fusion cancer and the heterogeneous molecular testing landscape in Canada, identifying and optimally managing such patients represents a new challenge. We provide a Canadian consensus on when and how to test for NTRK gene fusions and when to consider treatment with a TRK inhibitor. We focus on five tumour types: thyroid carcinoma, colorectal carcinoma, non-small cell lung carcinoma, soft tissue sarcoma, and salivary gland carcinoma. Based on the probability of the tumour harbouring an NTRK gene fusion, we also suggest a tumour-agnostic consensus for NTRK gene fusion testing and treatment. We recommend considering a TRK inhibitor in all patients with TRK fusion cancer with no other effective treatment options.
Collapse
Affiliation(s)
- D. Gwyn Bebb
- Tom Baker Cancer Centre and University of Calgary, Calgary, AB T2N 4N2, Canada
| | - Shantanu Banerji
- Research Institute in Oncology and Hematology, CancerCare Manitoba, University of Manitoba, Winnipeg, MB R3E 0V9, Canada;
| | - Normand Blais
- Centre Hospitalier Universitaire de Montreal, Department of Medicine, University of Montreal, Montreal, QC H2X 3E4, Canada; (N.B.); (D.S.)
| | - Patrice Desmeules
- Service D’Anatomopathologie et de Cytologie, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Quebec City, QC G1V 0A6, Canada;
| | - Sharlene Gill
- BC Cancer, Vancouver, BC V5Z 4E6, Canada; (S.G.); (B.M.); (C.E.S.)
| | - Andrea Grin
- Department of Pathology and Molecular Medicine, Queen’s University, Kingston, ON K7L 3N6, Canada; (A.G.); (H.F.)
| | - Harriet Feilotter
- Department of Pathology and Molecular Medicine, Queen’s University, Kingston, ON K7L 3N6, Canada; (A.G.); (H.F.)
| | - Aaron R. Hansen
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada; (A.R.H.); (M.K.)
| | - Martin Hyrcza
- Department of Pathology and Laboratory Medicine, Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB T2N 4Z6, Canada;
| | - Monika Krzyzanowska
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada; (A.R.H.); (M.K.)
| | - Barbara Melosky
- BC Cancer, Vancouver, BC V5Z 4E6, Canada; (S.G.); (B.M.); (C.E.S.)
| | | | - Bibiana Purgina
- The Ottawa Hospital, Department of Pathology and Laboratory Medicine, University of Ottawa, Ottawa, ON K1N 6N5, Canada;
| | - Dean Ruether
- Department of Oncology, Tom Baker Cancer Centre, Calgary, AB T2N 4N2, Canada;
| | | | - Denis Soulieres
- Centre Hospitalier Universitaire de Montreal, Department of Medicine, University of Montreal, Montreal, QC H2X 3E4, Canada; (N.B.); (D.S.)
| | - Emina Emilia Torlakovic
- Department of Pathology and Laboratory Medicine, Saskatchewan Health Authority and University of Saskatchewan, Saskatoon, SK S7N 5B5, Canada;
| | - Ming-Sound Tsao
- Department of Pathology, Laboratory Medicine Program, University Health Network, Toronto, ON M5G 2C4, Canada
| |
Collapse
|
36
|
Li J, Gao A, Zhang F, Wang S, Wang J, Wang J, Han S, Yang Z, Chen X, Fang Y, Jiang G, Sun Y. ILT3 promotes tumor cell motility and angiogenesis in non-small cell lung cancer. Cancer Lett 2020; 501:263-276. [PMID: 33152402 DOI: 10.1016/j.canlet.2020.10.048] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 10/24/2020] [Accepted: 10/29/2020] [Indexed: 12/13/2022]
Abstract
Immunoglobulin-like transcript (ILT) 3 is an immunosuppressive molecule that negatively regulates myeloid cell activation. ILT3 overexpression in tumor cells induces immune escape of solid tumors and facilitates invasion of monocytic acute myeloid leukemia cells. However, the expression and function of ILT3 in non-small cell lung cancer (NSCLC) cells remain elusive. Herein, we found that ILT3 was enriched in human NSCLC cells, and predicted advanced disease and poor overall survival. ILT3 overexpression enhanced the migration and invasion of NSCLC cells and tubule formation of human umbilical vein endothelial cells by upregulating and interacting with its ligand apolipoprotein E (ApoE) in vitro. Mechanistically, ILT3 recruited SHP2 and SHIP1, and subsequently activated ERK1/2 signaling mediating epithelial-mesenchymal transition (EMT) and increasing vascular endothelial growth factor (VEGF)-A expression in NSCLC cells, which are responsible for tumor cell motility and angiogenesis, respectively. Using murine metastasis models, we further confirmed ILT3 promoted NSCLC metastasis and explored the exact correlation of ILT3 with ApoE, EMT, and VEGF-A in vivo. These results unraveled novel mechanisms for ILT3-induced tumor progression and proposed ILT3 as a potential therapeutic target and prognostic biomarker for NSCLC patients.
Collapse
Affiliation(s)
- Juan Li
- Department of Oncology, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250013, PR China
| | - Aiqin Gao
- Department of Oncology, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250013, PR China
| | - Fang Zhang
- Department of Oncology, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250013, PR China
| | - Shuyun Wang
- Department of Oncology, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250013, PR China
| | - Jingnan Wang
- Department of Oncology, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250013, PR China
| | - Jing Wang
- Department of Oncology, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250013, PR China
| | - Shuyi Han
- Medical Research and Laboratory Diagnostic Center, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250013, PR China
| | - Zijiang Yang
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China
| | - Xiaozheng Chen
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China
| | - Yuying Fang
- Department of Oncology, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250013, PR China
| | - Guosheng Jiang
- Department of Immunology, College of Basic Medical, Binzhou Medical University, Yantai, Shandong, 256600, PR China
| | - Yuping Sun
- Department of Oncology, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250013, PR China; Department of Oncology, Jinan Central Hospital affiliated to Shandong First Medical University, Jinan, Shandong, 250013, PR China.
| |
Collapse
|