1
|
Eser M, Hekimoglu G, Yarar MH, Canbek S, Ozcelik M. KRAS G12C mutation in NSCLC in a small genetic center: insights into sotorasib therapy response potential. Sci Rep 2024; 14:26581. [PMID: 39496639 PMCID: PMC11535051 DOI: 10.1038/s41598-024-75208-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 10/03/2024] [Indexed: 11/06/2024] Open
Abstract
Lung cancer remains a significant health challenge, characterized by aberrant tissue growth within the pulmonary system. Early carcinogenic events often involve genomic instability and the emergence of a mutator phenotype. In this study, we aimed to explore the mutator phenotype in 89 patients diagnosed with non-small-cell lung cancer (NSCLC). RNA isolation from formalin-fixed paraffin-embedded (FFPE) tissue samples was performed using the Promega ReliaPrep RNA Miniprep System, facilitating gene amplification relevant to cancer through the Archer® FusionPlexComprehensiveThyroid and Lung (CTL) kit. Next-generation sequencing (NGS) on the Illumina NextSeq platform enabled comprehensive analysis of target areas. Utilizing Archer Analysis software, secondary analyses involving data cleansing, alignment, and variant/fusion identification were executed against the human reference genome hg19 (GRCh37). Expression patterns were visualized using HeatMap graphics. Our findings revealed a notable presence of KRAS gene mutations in approximately 20% of NSCLC patients. Among these mutations, the G12C variant was predominant at 50%, followed by G12V and G12D variants at 11.2% each. Notably, patients harboring the G12C variant responded favorably to sotorasib medication. These results underscore the importance of mutational profiling and targeted therapeutic approaches in managing NSCLC, particularly highlighting the promising efficacy of sotorasib in G12C-mutated cases.
Collapse
Affiliation(s)
- Metin Eser
- Department of Medical Genetics, Umraniye Education and Research Hospital, University of Health Sciences, Istanbul, Turkey
| | - Gulam Hekimoglu
- Department of Histology and Embryology, Hamidiye International Faculty of Medicine, University of Health Sciences, Istanbul, Turkey.
| | - Murat Hakki Yarar
- Department of Medical Genetics, Umraniye Education and Research Hospital, University of Health Sciences, Istanbul, Turkey
| | - Sezin Canbek
- Department of Medical Genetics, Umraniye Education and Research Hospital, University of Health Sciences, Istanbul, Turkey
| | - Melike Ozcelik
- Department of Medical Oncology, Umraniye Education and Research Hospital, University of Health Sciences, Istanbul, Turkey
| |
Collapse
|
2
|
Tang X, Zhu Y, Cao Z, Wang X, Cai X, Tang Y, Zhou J, Wu M, Zhen X, Ding L, Yan G, Wang H, Sun H, Jiang R. CDC42 deficiency leads to endometrial stromal cell senescence in recurrent implantation failure. Hum Reprod 2024:deae246. [PMID: 39487595 DOI: 10.1093/humrep/deae246] [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: 05/20/2024] [Revised: 09/22/2024] [Indexed: 11/04/2024] Open
Abstract
STUDY QUESTION Does the downregulation of cell division cycle 42 (CDC42) protein in endometrial stroma lead to endometrial senescence in patients with recurrent implantation failure (RIF), and what is the potential mechanism? SUMMARY ANSWER CDC42 deficiency causes endometrial stromal senescence and decidualization defects, impairing uterine receptivity of RIF patients, via activation of Wnt signaling pathway. WHAT IS KNOWN ALREADY Uterine aging is unique due to the cyclic remodeling and decidualization of endometrial tissue. Several transcriptomic studies have reported increased senescence in the endometrium in young patients with RIF. Our previous transcriptomic sequencing study discovered that endometrium from women with RIF showed downregulation of CDC42, which is an essential molecule affected by various senescence-related diseases. STUDY DESIGN, SIZE, DURATION The endometrial samples of a total of 71 fertile control patients and 37 RIF patients were collected to verify the association between CDC42 expression and endometrial senescence of RIF patients. Primary endometrial stromal cells (EnSCs) were isolated from endometrial biopsies taken from patients without any endometrial complications and planning to undergo IVF, then subjected to adenovirus-mediated CDC42 knockdown and decidualization induction to explore the detailed mechanism by which CDC42 governs stromal senescence and decidualization. Wnt inhibitor XAV-939 was used to correct the endometrial senescence and decidualization defect. PARTICIPANTS/MATERIALS, SETTING, METHODS Senescence was determined by cell cycle arrest markers (e.g. P16, P21, and P53), SASP molecules (e.g. IL6 and CXCL8), and SA-β-gal staining. Masson's staining and Sirius Red staining were used to detect the endometrial fibrosis. Decidualization was evaluated by the mRNA expression and protein secretion of PRL and IGFBP1, F-actin immunostaining, and the BeWo spheroids 'in vitro implantation' model. Methods used to assess cell function included adenovirus transduction, RNA-sequencing, bioinformatic analysis, western blotting, RT-qPCR, ELISA, and immunofluorescence. MAIN RESULTS AND THE ROLE OF CHANCE Here, we observed remarkably increased levels of stromal senescence and fibrosis, along with stromal CDC42 deficiency, in the endometrium of patients with RIF (P < 0.001). Knockdown of CDC42 effectively induced premature senescence in EnSCs, leading to aberrant accumulation of senescent EnSCs and collagen deposition during decidualization. CDC42 deficiency in EnSCs restrained the decidualization differentiation and receptivity to trophoblast cells. Transcriptomic analysis revealed Wnt signaling activation as a critical downstream alteration in CDC42-deficient EnSCs. Mechanistically, CDC42 interacted with AKT competitively to impede the binding of GSK3β to AKT. Knockdown of CDC42 increased AKT-mediated phosphorylation of GSK3β to inactivate the Axin-GSK3β destruction complex, leading to accumulation and nuclear translocation of β-catenin. Importantly, Wnt signaling inhibitors partially corrected the endometrial senescence caused by CDC42 deficiency, and improved both decidualization and trophoblast invasion. LARGE SCALE DATA RNA-seq data sets generated in this study have been deposited at the NCBI database with BioProject accession number PRJNA1102745. LIMITATIONS, REASONS FOR CAUTION The present study was based on in vitro cell cultures. Further studies involving CDC42-regulated endometrial senescence are needed in knockout mice model and human endometrial assembloids. WIDER IMPLICATIONS OF THE FINDINGS In addition to uncovering endometrial senescence in RIF, our findings underscore the significance of CDC42 in modulating EnSC senescence to maintain the decidualization function, and suggest Wnt signaling inhibitors as potential therapeutic agents for alleviating endometrial senescence. STUDY FUNDING/COMPETING INTEREST(S) This work was supported by the National Natural Science Foundation of China [82271698 (R.J.), 82030040 (H.S.), 82288102 (H.W.), and 82371680 (G.Y.)]; the Natural Science Foundation of Jiangsu Province [BK20231117 (R.J.)]; and the Medical Science and Technology Development Foundation of Nanjing Department of Health [YKK23097 (Y.Z.)]. The authors declare no potential conflicts of interest.
Collapse
Affiliation(s)
- Xinyi Tang
- Center for Reproductive Medicine and Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Center for Molecular Reproductive Medicine, Nanjing University, Nanjing, China
| | - Yingchun Zhu
- Center for Reproductive Medicine and Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Center for Molecular Reproductive Medicine, Nanjing University, Nanjing, China
| | - Zhiwen Cao
- Center for Reproductive Medicine and Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Center for Molecular Reproductive Medicine, Nanjing University, Nanjing, China
| | - Xiaoying Wang
- Center for Reproductive Medicine and Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Center for Molecular Reproductive Medicine, Nanjing University, Nanjing, China
| | - Xinyu Cai
- Center for Reproductive Medicine and Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Center for Molecular Reproductive Medicine, Nanjing University, Nanjing, China
| | - Yurun Tang
- Center for Reproductive Medicine and Obstetrics and Gynecology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| | - Jidong Zhou
- Center for Reproductive Medicine and Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Center for Molecular Reproductive Medicine, Nanjing University, Nanjing, China
| | - Min Wu
- Center for Reproductive Medicine and Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Center for Molecular Reproductive Medicine, Nanjing University, Nanjing, China
| | - Xin Zhen
- Center for Reproductive Medicine and Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Center for Molecular Reproductive Medicine, Nanjing University, Nanjing, China
| | - Lijun Ding
- Center for Reproductive Medicine and Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Center for Molecular Reproductive Medicine, Nanjing University, Nanjing, China
| | - Guijun Yan
- Center for Reproductive Medicine and Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Center for Molecular Reproductive Medicine, Nanjing University, Nanjing, China
| | - Haibin Wang
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Haixiang Sun
- Center for Reproductive Medicine and Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Center for Molecular Reproductive Medicine, Nanjing University, Nanjing, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, China
| | - Ruiwei Jiang
- Center for Reproductive Medicine and Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Center for Molecular Reproductive Medicine, Nanjing University, Nanjing, China
| |
Collapse
|
3
|
Zheng S, Chen R, Zhang L, Tan L, Li L, Long F, Wang T. Unraveling the future: Innovative design strategies and emerging challenges in HER2-targeted tyrosine kinase inhibitors for cancer therapy. Eur J Med Chem 2024; 276:116702. [PMID: 39059182 DOI: 10.1016/j.ejmech.2024.116702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 07/12/2024] [Accepted: 07/19/2024] [Indexed: 07/28/2024]
Abstract
Human epidermal growth factor receptor 2 (HER2) is a transmembrane receptor-like protein with tyrosine kinase activity that plays a vital role in processes such as cell proliferation, differentiation, and angiogenesis. The degree of malignancy of different cancers, notably breast cancer, is strongly associated with HER2 amplification, overexpression, and mutation. Currently, widely used clinical HER2 tyrosine kinase inhibitors (TKIs), such as lapatinib and neratinib, have several drawbacks, including susceptibility to drug resistance caused by HER2 mutations and adverse effects from insufficient HER2 selectivity. To address these issues, it is essential to create innovative HER2 TKIs with enhanced safety, effectiveness against mutations, and high selectivity. Typically, SPH5030 has advanced to phase I clinical trials for its strong suppression of four HER2 mutations. This review discusses the latest research progress in HER2 TKIs, with a focus on the structural optimization process and structure-activity relationship analysis. In particular, this study highlights promising design strategies to address these challenges, providing insightful information and inspiration for future development in this field.
Collapse
Affiliation(s)
- Sixiang Zheng
- Department of Clinical Research, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, 610041, China
| | - Ruixian Chen
- Department of Breast Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Lele Zhang
- Department of Respiratory and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Lun Tan
- Department of Respiratory and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Lintao Li
- Department of Radiotherapy, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, 610041, China.
| | - Fangyi Long
- Laboratory Medicine Center, Sichuan Provincial Maternity and Child Health Care Hospital, Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu Medical College, Chengdu, 610032, China.
| | - Ting Wang
- Department of Clinical Research, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, 610041, China.
| |
Collapse
|
4
|
Ding Z, Wei Y, Dai J, Pan C, Yang L, Li Q, Zhang Y, Yan Q, Wu C, Li A, Lan Z, Liu S, Wang X. Deficiency of SDHC promotes metastasis by reprogramming fatty acid metabolism in colorectal cancer. J Transl Med 2024; 22:544. [PMID: 38844980 PMCID: PMC11157952 DOI: 10.1186/s12967-024-05361-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 05/31/2024] [Indexed: 06/09/2024] Open
Abstract
BACKGROUND Several studies have demonstrated a strong correlation between impaired Succinate dehydrogenase (SDH) function and the advancement of tumors. As a subunit of SDH, succinate dehydrogenase complex subunit C (SDHC) has been revealed to play tumor suppressive roles in several cancers, while its specific role in colorectal cancer (CRC) still needs further investigation. METHODS Online database were utilized to investigate the expression of SDHC in colorectal cancer and to assess its correlation with patient prognosis. Cell metastasis was assessed using transwell and wound healing assays, while tumor metastasis was studied in a nude mice model in vivo. Drug screening and RNA sequencing were carried out to reveal the tumor suppressor mechanism of SDHC. Triglycerides, neutral lipids and fatty acid oxidation were measured using the Triglyceride Assay Kit, BODIPY 493/503 and Colorimetric Fatty Acid Oxidation Rate Assay Kit, respectively. The expression levels of enzymes involved in fatty acid metabolism and the PI3K/AKT signaling pathway were determined by quantitative real-time PCR and western blot. RESULTS Downregulation of SDHC was found to be closely associated with a poor prognosis in CRC. SDHC knockdown promoted CRC metastasis both in vitro and in vivo. Through drug screening and Gene set enrichment analysis, it was discovered that SDHC downregulation was positively associated with the fatty acid metabolism pathways significantly. The effects of SDHC silencing on metastasis were reversed when fatty acid synthesis was blocked. Subsequent experiments revealed that SDHC silencing activated the PI3K/AKT signaling axis, leading to lipid accumulation by upregulating the expression of aldehyde dehydrogenase 3 family member A2 (ALDH3A2) and reduction of fatty acid oxidation rate by suppressing the expression of acyl-coenzyme A oxidase 1 (ACOX1) and carnitine palmitoyltransferase 1A (CPT1A). CONCLUSIONS SDHC deficiency could potentially enhance CRC metastasis by modulating the PI3K/AKT pathways and reprogramming lipid metabolism.
Collapse
Affiliation(s)
- Zhuoyu Ding
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yiyi Wei
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jingping Dai
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Chaomin Pan
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Li Yang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qingyuan Li
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yue Zhang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qun Yan
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Changjie Wu
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Aimin Li
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhixian Lan
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Side Liu
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
- Pazhou Lab, Guangzhou, Guangdong, China.
| | - Xinke Wang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
5
|
Xu J, Tian L, Qi W, Lv Q, Wang T. Advancements in NSCLC: From Pathophysiological Insights to Targeted Treatments. Am J Clin Oncol 2024; 47:291-303. [PMID: 38375734 PMCID: PMC11107893 DOI: 10.1097/coc.0000000000001088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2024]
Abstract
With the global incidence of non-small cell lung cancer (NSCLC) on the rise, the development of innovative treatment strategies is increasingly vital. This review underscores the pivotal role of precision medicine in transforming NSCLC management, particularly through the integration of genomic and epigenomic insights to enhance treatment outcomes for patients. We focus on the identification of key gene mutations and examine the evolution and impact of targeted therapies. These therapies have shown encouraging results in improving survival rates and quality of life. Despite numerous gene mutations being identified in association with NSCLC, targeted treatments are available for only a select few. This paper offers an exhaustive analysis of the pathogenesis of NSCLC and reviews the latest advancements in targeted therapeutic approaches. It emphasizes the ongoing necessity for research and development in this domain. In addition, we discuss the current challenges faced in the clinical application of these therapies and the potential directions for future research, including the identification of novel targets and the development of new treatment modalities.
Collapse
Affiliation(s)
- Jianan Xu
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine
| | - Lin Tian
- Pulmonology Department, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, P.R. China
| | - Wenlong Qi
- Pulmonology Department, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, P.R. China
| | - Qingguo Lv
- Pulmonology Department, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, P.R. China
| | - Tan Wang
- Pulmonology Department, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, P.R. China
| |
Collapse
|
6
|
Ye H, Yu W, Ni Y, Bao X, Zhang X, Li Y, Chen A, Li J, Zheng L. Apatinib plus chemotherapy is associated with an improved tumor response, survival and tolerance compared with chemotherapy alone for advanced lung adenocarcinoma treatment. Oncol Lett 2024; 27:194. [PMID: 38495832 PMCID: PMC10941069 DOI: 10.3892/ol.2024.14327] [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: 05/06/2023] [Accepted: 11/13/2023] [Indexed: 03/19/2024] Open
Abstract
Apatinib plus chemotherapy demonstrates good efficacy in multiple advanced carcinomas; however, its use in patients with advanced lung adenocarcinoma (LUAD) has not yet been assessed. The present study evaluated the potential benefits of apatinib plus chemotherapy in patients with advanced LUAD. A total of 145 patients with advanced LUAD and negative driver genes who received apatinib plus chemotherapy (n=65) or chemotherapy alone (n=80) were analyzed. The overall response rate was significantly improved by apatinib plus chemotherapy vs. chemotherapy alone (53.8 vs. 36.3%; P=0.034). Moreover, progression-free survival (PFS) was significantly longer in patients who received apatinib plus chemotherapy, compared with those who received chemotherapy alone [median (95% CI), 13.4 months (11.5-15.3) vs. 8.2 months (6.9-9.5); P<0.001], as was overall survival (OS) [median (95% CI), 23.1 months (not reached) vs. 17.0 months (14.6-19.4; P=0.001). Following adjustment by multivariate Cox regression analysis, apatinib plus chemotherapy was associated with a significantly longer PFS [hazard ratio (HR), 0.444; P<0.001] and OS (HR, 0.347; P<0.001), compared with chemotherapy alone. Subgroup analyses revealed that PFS and OS were significantly improved following apatinib plus chemotherapy vs. chemotherapy alone (all P<0.05) in patients receiving first- or second-line treatment. Notably, the incidence of hypertension was significantly increased following apatinib plus chemotherapy vs. chemotherapy alone (43.1 vs. 25.0%; P=0.021), whereas the incidence of other adverse events was not significantly different between the two treatment groups (all P>0.05). In conclusion, apatinib plus chemotherapy is associated with an improved treatment response and survival compared with chemotherapy alone, with a tolerable safety profile in patients with advanced LUAD.
Collapse
Affiliation(s)
- Hua Ye
- Department of Pulmonary and Critical Care Medicine, Yueqing People's Hospital, Yueqing, Zhejiang 325600, P.R. China
| | - Wenwen Yu
- Department of Pulmonary and Critical Care Medicine, Yueqing People's Hospital, Yueqing, Zhejiang 325600, P.R. China
| | - Yangyang Ni
- Department of Pulmonary and Critical Care Medicine, Yueqing People's Hospital, Yueqing, Zhejiang 325600, P.R. China
| | - Xiaoqiong Bao
- Department of Pulmonary and Critical Care Medicine, Yueqing People's Hospital, Yueqing, Zhejiang 325600, P.R. China
| | - Xie Zhang
- Department of Pulmonary and Critical Care Medicine, Yueqing People's Hospital, Yueqing, Zhejiang 325600, P.R. China
| | - Yunlei Li
- Department of Pulmonary and Critical Care Medicine, Yueqing People's Hospital, Yueqing, Zhejiang 325600, P.R. China
| | - Ali Chen
- Department of Pulmonary and Critical Care Medicine, Yueqing People's Hospital, Yueqing, Zhejiang 325600, P.R. China
| | - Jifa Li
- Department of Pulmonary and Critical Care Medicine, Yueqing People's Hospital, Yueqing, Zhejiang 325600, P.R. China
| | - Long Zheng
- Department of Pulmonary and Critical Care Medicine, Yueqing People's Hospital, Yueqing, Zhejiang 325600, P.R. China
| |
Collapse
|
7
|
Wang X, Wei W. rBMA: A robust Bayesian Model Averaging Method for phase II basket trials based on informative mixture priors. Contemp Clin Trials 2024; 140:107505. [PMID: 38521384 DOI: 10.1016/j.cct.2024.107505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 01/21/2024] [Accepted: 03/13/2024] [Indexed: 03/25/2024]
Abstract
Oncology drug research in the last few decades has been driven by the development of targeted agents. In the era of targeted therapies, basket trials are often used to test the antitumor activity of a novel treatment in multiple indications sharing the same genomic alteration. As patient population are further fragmented into biomarker-defined subgroups in basket trials, novel statistical methods are needed to facilitate cross-indication learning to improve the statistical power in basket trial design. Here we propose a robust Bayesian model averaging (rBMA) technique for the design and analysis of phase II basket trials. We consider the posterior distribution of each indication (basket) as the weighted average of three different models which only differ in their priors (enthusiastic, pessimistic and non-informative). The posterior weights of these models are determined based on the effect of the experimental treatment in all the indications tested. In early phase oncology trials, different binary endpoints might be chosen for different indications (objective response, disease control or PFS at landmark times), which makes it even more challenging to borrow information across indications. Compared to previous approaches, the proposed method has the flexibility to support cross-indication learning in the presence of mixed endpoints. We evaluate and compare the performance of the proposed rBMA approach to competing approaches in simulation studies. R scripts to implement the proposed method are available at https://github.com/xwang317/rBMA.
Collapse
Affiliation(s)
- Xueting Wang
- Department of Biostatistics, Yale School of Public Health, New Haven, CT 06520, United States of America
| | - Wei Wei
- Department of Biostatistics, Yale School of Public Health, New Haven, CT 06520, United States of America.
| |
Collapse
|
8
|
Wang Y, Qiu H, Lin R, Hong W, Lu J, Ling H, Sun X, Yang C. Advancements in the Understanding of Small-Cell Neuroendocrine Cervical Cancer: Where We Stand and What Lies Ahead. J Pers Med 2024; 14:462. [PMID: 38793044 PMCID: PMC11122604 DOI: 10.3390/jpm14050462] [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: 03/22/2024] [Revised: 04/04/2024] [Accepted: 04/26/2024] [Indexed: 05/26/2024] Open
Abstract
Small-cell neuroendocrine cervical carcinoma (SCNCC) is a rare yet aggressive gynecological malignancy associated with dismal clinical outcomes. Its rarity has led to a limited number of retrospective studies and an absence of prospective research, posing significant challenges for evidence-based treatment approaches. As a result, most gynecologic oncology centers have limited experience with this tumor, emphasizing the urgent need for a comprehensive review and summary. This article systematically reviews the pathogenesis, immunohistochemical and molecular characteristics, prognostic factors, and clinical management of gynecologic SCNCC. We specifically focused on reviewing the distinct genomic characteristics of SCNCC identified via next-generation sequencing technologies, including loss of heterozygosity (LOH), somatic mutations, structural variations (SVs), and microRNA alterations. The identification of these actionable genomic events offers promise for discovering new molecular targets for drug development and enhancing therapeutic outcomes. Additionally, we delve deeper into key clinical challenges, such as determining the optimal treatment modality between chemoradiation and surgery for International Federation of Gynecology and Obstetrics (FIGO) stage I phase patients within a precision stratification framework, as well as the role of targeted therapy within the homologous recombination (HR) pathway, immune checkpoint inhibitors (ICIs), and prophylactic cranial irradiation (PCI) in the management of SCNCC. Finally, we anticipate the utilization of multiple SCNCC models, including cancer tissue-originated spheroid (CTOS) lines and patient-derived xenografts (PDXs), to decipher driver events and develop individualized therapeutic strategies for clinical application.
Collapse
Affiliation(s)
- Yan Wang
- Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Hangzhou First People’s Hospital, Hangzhou 310006, China
- Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Hui Qiu
- Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Rongjie Lin
- Department of Radiotherapy, West China Xiamen Hospital of Sichuan University, Xiamen 361021, China
| | - Weiwei Hong
- Department of Medical Oncology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Jiahao Lu
- Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Huan Ling
- Department of Ultrasound in Gynecology and Obstetrics, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Xiaoge Sun
- Department of Radiation Oncology, Affiliated Hospital of Inner Mongolia Medical University, Hohhot 750306, China
| | - Chunxu Yang
- Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| |
Collapse
|
9
|
Adamopoulos C, Papavassiliou KA, Poulikakos PI, Papavassiliou AG. RAF and MEK Inhibitors in Non-Small Cell Lung Cancer. Int J Mol Sci 2024; 25:4633. [PMID: 38731852 PMCID: PMC11083651 DOI: 10.3390/ijms25094633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 04/17/2024] [Accepted: 04/22/2024] [Indexed: 05/13/2024] Open
Abstract
Lung cancer, despite recent advancements in survival rates, represents a significant global health burden. Non-small cell lung cancer (NSCLC), the most prevalent type, is driven largely by activating mutations in Kirsten rat sarcoma viral oncogene homologue (KRAS) and receptor tyrosine kinases (RTKs), and less in v-RAF murine sarcoma viral oncogene homolog B (BRAF) and mitogen-activated protein-kinase kinase (MEK), all key components of the RTK-RAS-mitogen-activated protein kinase (MAPK) pathway. Learning from melanoma, the identification of BRAFV600E substitution in NSCLC provided the rationale for the investigation of RAF and MEK inhibition as a therapeutic strategy. The regulatory approval of two RAF-MEK inhibitor combinations, dabrafenib-trametinib, in 2017, and encorafenib-binimetinib, in 2023, signifies a breakthrough for the management of BRAFV600E-mutant NSCLC patients. However, the almost universal emergence of acquired resistance limits their clinical benefit. New RAF and MEK inhibitors, with distinct biochemical characteristics, are in preclinical and clinical development. In this review, we aim to provide valuable insights into the current state of RAF and MEK inhibition in the management of NSCLC, fostering a deeper understanding of the potential impact on patient outcomes.
Collapse
Affiliation(s)
- Christos Adamopoulos
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
- Department of Oncological Sciences, Precision Immunology Institute, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
| | - Kostas A. Papavassiliou
- First University Department of Respiratory Medicine, ‘Sotiria’ Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Poulikos I. Poulikakos
- Department of Oncological Sciences, Precision Immunology Institute, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
| | - Athanasios G. Papavassiliou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|
10
|
Liu X, Mei W, Zhang P, Zeng C. PIK3CA mutation as an acquired resistance driver to EGFR-TKIs in non-small cell lung cancer: Clinical challenges and opportunities. Pharmacol Res 2024; 202:107123. [PMID: 38432445 DOI: 10.1016/j.phrs.2024.107123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 02/20/2024] [Accepted: 02/27/2024] [Indexed: 03/05/2024]
Abstract
Epithelial growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) have significantly enhanced the treatment outcomes in non-small cell lung cancer (NSCLC) patients harboring EGFR mutations. However, the occurrence of acquired resistance to EGFR-TKIs is an unavoidable outcome observed in these patients. Disruption of the PI3K/AKT/mTOR signaling pathway can contribute to the emergence of resistance to EGFR TKIs in lung cancer. The emergence of PIK3CA mutations following treatment with EGFR-TKIs can lead to resistance against EGFR-TKIs. This review provides an overview of the current perspectives regarding the involvement of PI3K/AKT/mTOR signaling in the development of lung cancer. Furthermore, we outline the state-of-the-art therapeutic strategies targeting the PI3K/AKT/mTOR signaling pathway in lung cancer. We highlight the role of PIK3CA mutation as an acquired resistance mechanism against EGFR-TKIs in EGFR-mutant NSCLC. Crucially, we explore therapeutic strategies targeting PIK3CA-mediated resistance to EGFR TKIs in lung cancer, aiming to optimize the effectiveness of treatment.
Collapse
Affiliation(s)
- Xiaohong Liu
- Department of Medical Oncology, Shenzhen Longhua District Central Hospital, Shenzhen 518110, China
| | - Wuxuan Mei
- Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, China
| | - Pengfei Zhang
- Department of Medical Laboratory, Shenzhen Longhua District Central Hospital, Shenzhen 518110, China
| | - Changchun Zeng
- Department of Medical Laboratory, Shenzhen Longhua District Central Hospital, Shenzhen 518110, China.
| |
Collapse
|
11
|
Chi X, Yuan Y, Yu Z, Lin R. A generalized calibrated Bayesian hierarchical modeling approach to basket trials with multiple endpoints. Biom J 2024; 66:e2300122. [PMID: 38368277 PMCID: PMC11323483 DOI: 10.1002/bimj.202300122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 11/05/2023] [Accepted: 12/29/2023] [Indexed: 02/19/2024]
Abstract
A basket trial simultaneously evaluates a treatment in multiple cancer subtypes, offering an effective way to accelerate drug development in multiple indications. Many basket trials are designed and monitored based on a single efficacy endpoint, primarily the tumor response. For molecular targeted or immunotherapy agents, however, a single efficacy endpoint cannot adequately characterize the treatment effect. It is increasingly important to use more complex endpoints to comprehensively assess the risk-benefit profile of such targeted therapies. We extend the calibrated Bayesian hierarchical modeling approach to monitor phase II basket trials with multiple endpoints. We propose two generalizations, one based on the latent variable approach and the other based on the multinomial-normal hierarchical model, to accommodate different types of endpoints and dependence assumptions regarding information sharing. We introduce shrinkage parameters as functions of statistics measuring homogeneity among subgroups and propose a general calibration approach to determine the functional forms. Theoretical properties of the generalized hierarchical models are investigated. Simulation studies demonstrate that the monitoring procedure based on the generalized approach yields desirable operating characteristics.
Collapse
Affiliation(s)
- Xiaohan Chi
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, U.S.A
| | - Ying Yuan
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, U.S.A
| | - Zhangsheng Yu
- Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
- SJTU-Yale Joint Center for Biostatistics and Data Science, Shanghai Jiao Tong University, Shanghai, China
| | - Ruitao Lin
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, U.S.A
| |
Collapse
|
12
|
Gulati P, Singh CV. The Crucial Role of Molecular Biology in Cancer Therapy: A Comprehensive Review. Cureus 2024; 16:e52246. [PMID: 38352075 PMCID: PMC10863367 DOI: 10.7759/cureus.52246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 01/14/2024] [Indexed: 02/16/2024] Open
Abstract
Molecular biology shines a light of hope amid the complex terrain of cancer, bringing revolutionary approaches to cancer treatment. Instead of providing a synopsis, this review presents an engaging story that sheds light on the genetic nuances controlling the course of cancer. This review goes beyond just listing genetic alterations to examine the complex interactions that lead to oncogene activation, exploring particular triggers such as viral infections or proto-oncogene mutations. A comprehensive grasp of the significant influence of oncogenes is possible through the classification and clarification of their function in various types of cancer. Furthermore, the role of tumor suppressor genes in controlling cell division and preventing tumor growth is fully explained, providing concrete examples and case studies to ground the conversation and create a stronger story. This study highlights the practical applications of molecular biology and provides a comprehensive overview of various detection and treatment modalities. It emphasizes the effectiveness of RNA analysis, immunohistochemistry, and next-generation sequencing (NGS) in cancer diagnosis and prognosis prediction. Examples include the individualized classification of breast cancers through RNA profiling, the use of NGS to identify actionable mutations such as epidermal growth factor receptor and anaplastic lymphoma kinase in lung cancer, and the use of immunohistochemical staining for proteins such as Kirsten rat sarcoma viral oncogene to guide treatment decisions in colorectal cancer. This paper carefully examines how molecular biology is essential to creating new strategies to fight this difficult and widespread illness. It highlights the exciting array of available therapeutic approaches, offering concrete instances of how clustered regularly interspaced short palindromic repeats (CRISPR) and CRISPR-associated protein 9 (CRISPR-Cas9), targeted pharmaceuticals, immunotherapy, and treatments that induce apoptosis are driving a paradigm shift in cancer care. The revolutionary CRISPR-Cas9 system takes center stage, showcasing how precise gene editing could transform cancer therapy. This study concludes by fervently highlighting the critical role that molecular biology plays in reducing the complexity of cancer and changing the treatment landscape. It lists accomplishments but also thoughtfully examines cases and findings that progress our search for more precisely customized and effective cancer therapies.
Collapse
Affiliation(s)
- Prisha Gulati
- Medicine and Surgery, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Chandra Veer Singh
- Otolaryngology - Head and Neck Surgery, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
13
|
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: 0] [Impact Index Per Article: 0] [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
|
14
|
Schultz CW, Zhang Y, Elmeskini R, Zimmermann A, Fu H, Murai Y, Wangsa D, Kumar S, Takahashi N, Atkinson D, Saha LK, Lee C, Elenbaas B, Desai P, Sebastian R, Sharma AK, Abel M, Schroeder B, Krishnamurthy M, Kumar R, Roper N, Aladjem M, Zenke FT, Ohler ZW, Pommier Y, Thomas A. ATR inhibition augments the efficacy of lurbinectedin in small-cell lung cancer. EMBO Mol Med 2023; 15:e17313. [PMID: 37491889 PMCID: PMC10405061 DOI: 10.15252/emmm.202217313] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 06/12/2023] [Accepted: 06/19/2023] [Indexed: 07/27/2023] Open
Abstract
Small-cell lung cancer (SCLC) is the most lethal type of lung cancer. Specifically, MYC-driven non-neuroendocrine SCLC is particularly resistant to standard therapies. Lurbinectedin was recently approved for the treatment of relapsed SCLC, but combinatorial approaches are needed to increase the depth and duration of responses to lurbinectedin. Using high-throughput screens, we found inhibitors of ataxia telangiectasia mutated and rad3 related (ATR) as the most effective agents for augmenting lurbinectedin efficacy. First-in-class ATR inhibitor berzosertib synergized with lurbinectedin in multiple SCLC cell lines, organoid, and in vivo models. Mechanistically, ATR inhibition abrogated S-phase arrest induced by lurbinectedin and forced cell cycle progression causing mitotic catastrophe and cell death. High CDKN1A/p21 expression was associated with decreased synergy due to G1 arrest, while increased levels of ERCC5/XPG were predictive of increased combination efficacy. Importantly, MYC-driven non-neuroendocrine tumors which are resistant to first-line therapies show reduced CDKN1A/p21 expression and increased ERCC5/XPG indicating they are primed for response to lurbinectedin-berzosertib combination. The combination is being assessed in a clinical trial NCT04802174.
Collapse
Affiliation(s)
- Christopher W Schultz
- Developmental Therapeutics Branch, Center for Cancer ResearchNational Cancer Institute, National Institutes of HealthBethesdaMDUSA
| | - Yang Zhang
- Developmental Therapeutics Branch, Center for Cancer ResearchNational Cancer Institute, National Institutes of HealthBethesdaMDUSA
| | - Rajaa Elmeskini
- Center for Advanced Preclinical Research, Leidos Biomedical Research, IncFrederick National Laboratory for Cancer ResearchFrederickMDUSA
| | - Astrid Zimmermann
- Translational Innovation Platform OncologyMerck KGaA, Biopharma R&DDarmstadtGermany
| | - Haiqing Fu
- Developmental Therapeutics Branch, Center for Cancer ResearchNational Cancer Institute, National Institutes of HealthBethesdaMDUSA
| | - Yasuhisa Murai
- Developmental Therapeutics Branch, Center for Cancer ResearchNational Cancer Institute, National Institutes of HealthBethesdaMDUSA
| | - Darawalee Wangsa
- Genetics Branch, Center for Cancer ResearchNational Cancer Institute, National Institutes of HealthBethesdaMDUSA
| | - Suresh Kumar
- Developmental Therapeutics Branch, Center for Cancer ResearchNational Cancer Institute, National Institutes of HealthBethesdaMDUSA
| | - Nobuyuki Takahashi
- Developmental Therapeutics Branch, Center for Cancer ResearchNational Cancer Institute, National Institutes of HealthBethesdaMDUSA
- Medical Oncology BranchNational Center for Global Health and MedicineTokyoJapan
| | - Devon Atkinson
- Center for Advanced Preclinical Research, Leidos Biomedical Research, IncFrederick National Laboratory for Cancer ResearchFrederickMDUSA
| | - Liton Kumar Saha
- Developmental Therapeutics Branch, Center for Cancer ResearchNational Cancer Institute, National Institutes of HealthBethesdaMDUSA
| | - Chien‐Fei Lee
- Translational Innovation Platform OncologyEMD Serono Research and Development Institute Inc., Biopharma R&DBillericaMAUSA
| | - Brian Elenbaas
- Translational Innovation Platform OncologyEMD Serono Research and Development Institute Inc., Biopharma R&DBillericaMAUSA
| | - Parth Desai
- Developmental Therapeutics Branch, Center for Cancer ResearchNational Cancer Institute, National Institutes of HealthBethesdaMDUSA
| | - Robin Sebastian
- Developmental Therapeutics Branch, Center for Cancer ResearchNational Cancer Institute, National Institutes of HealthBethesdaMDUSA
| | - Ajit Kumar Sharma
- Developmental Therapeutics Branch, Center for Cancer ResearchNational Cancer Institute, National Institutes of HealthBethesdaMDUSA
| | - Melissa Abel
- Developmental Therapeutics Branch, Center for Cancer ResearchNational Cancer Institute, National Institutes of HealthBethesdaMDUSA
| | - Brett Schroeder
- Developmental Therapeutics Branch, Center for Cancer ResearchNational Cancer Institute, National Institutes of HealthBethesdaMDUSA
| | - Manan Krishnamurthy
- Developmental Therapeutics Branch, Center for Cancer ResearchNational Cancer Institute, National Institutes of HealthBethesdaMDUSA
| | - Rajesh Kumar
- Developmental Therapeutics Branch, Center for Cancer ResearchNational Cancer Institute, National Institutes of HealthBethesdaMDUSA
| | - Nitin Roper
- Developmental Therapeutics Branch, Center for Cancer ResearchNational Cancer Institute, National Institutes of HealthBethesdaMDUSA
| | - Mirit Aladjem
- Developmental Therapeutics Branch, Center for Cancer ResearchNational Cancer Institute, National Institutes of HealthBethesdaMDUSA
| | - Frank T Zenke
- Translational Innovation Platform OncologyMerck KGaA, Biopharma R&DDarmstadtGermany
| | - Zoe Weaver Ohler
- Center for Advanced Preclinical Research, Leidos Biomedical Research, IncFrederick National Laboratory for Cancer ResearchFrederickMDUSA
| | - Yves Pommier
- Developmental Therapeutics Branch, Center for Cancer ResearchNational Cancer Institute, National Institutes of HealthBethesdaMDUSA
| | - Anish Thomas
- Developmental Therapeutics Branch, Center for Cancer ResearchNational Cancer Institute, National Institutes of HealthBethesdaMDUSA
| |
Collapse
|
15
|
Leone GM, Candido S, Lavoro A, Vivarelli S, Gattuso G, Calina D, Libra M, Falzone L. Clinical Relevance of Targeted Therapy and Immune-Checkpoint Inhibition in Lung Cancer. Pharmaceutics 2023; 15:1252. [PMID: 37111737 PMCID: PMC10142433 DOI: 10.3390/pharmaceutics15041252] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/12/2023] [Accepted: 04/14/2023] [Indexed: 04/29/2023] Open
Abstract
Lung cancer (LC) represents the second most diagnosed tumor and the malignancy with the highest mortality rate. In recent years, tremendous progress has been made in the treatment of this tumor thanks to the discovery, testing, and clinical approval of novel therapeutic approaches. Firstly, targeted therapies aimed at inhibiting specific mutated tyrosine kinases or downstream factors were approved in clinical practice. Secondly, immunotherapy inducing the reactivation of the immune system to efficiently eliminate LC cells has been approved. This review describes in depth both current and ongoing clinical studies, which allowed the approval of targeted therapies and immune-checkpoint inhibitors as standard of care for LC. Moreover, the present advantages and pitfalls of new therapeutic approaches will be discussed. Finally, the acquired importance of human microbiota as a novel source of LC biomarkers, as well as therapeutic targets to improve the efficacy of available therapies, was analyzed. Therapy against LC is increasingly becoming holistic, taking into consideration not only the genetic landscape of the tumor, but also the immune background and other individual variables, such as patient-specific gut microbial composition. On these bases, in the future, the research milestones reached will allow clinicians to treat LC patients with tailored approaches.
Collapse
Affiliation(s)
- Gian Marco Leone
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Saverio Candido
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
- Research Center for Prevention, Diagnosis and Treatment of Cancer, University of Catania, 95123 Catania, Italy
| | - Alessandro Lavoro
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Silvia Vivarelli
- Department of Biomedical and Dental Sciences, Morphological and Functional Imaging, Section of Occupational Medicine, University of Messina, 98125 Messina, Italy
| | - Giuseppe Gattuso
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Massimo Libra
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
- Research Center for Prevention, Diagnosis and Treatment of Cancer, University of Catania, 95123 Catania, Italy
| | - Luca Falzone
- Epidemiology and Biostatistics Unit, Istituto Nazionale Tumori IRCCS “Fondazione G. Pascale”, 80131 Naples, Italy;
| |
Collapse
|
16
|
Philip AK, Samuel BA, Bhatia S, Khalifa SAM, El-Seedi HR. Artificial Intelligence and Precision Medicine: A New Frontier for the Treatment of Brain Tumors. Life (Basel) 2022; 13:24. [PMID: 36675973 PMCID: PMC9866715 DOI: 10.3390/life13010024] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/08/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
Brain tumors are a widespread and serious neurological phenomenon that can be life- threatening. The computing field has allowed for the development of artificial intelligence (AI), which can mimic the neural network of the human brain. One use of this technology has been to help researchers capture hidden, high-dimensional images of brain tumors. These images can provide new insights into the nature of brain tumors and help to improve treatment options. AI and precision medicine (PM) are converging to revolutionize healthcare. AI has the potential to improve cancer imaging interpretation in several ways, including more accurate tumor genotyping, more precise delineation of tumor volume, and better prediction of clinical outcomes. AI-assisted brain surgery can be an effective and safe option for treating brain tumors. This review discusses various AI and PM techniques that can be used in brain tumor treatment. These new techniques for the treatment of brain tumors, i.e., genomic profiling, microRNA panels, quantitative imaging, and radiomics, hold great promise for the future. However, there are challenges that must be overcome for these technologies to reach their full potential and improve healthcare.
Collapse
Affiliation(s)
- Anil K. Philip
- School of Pharmacy, University of Nizwa, Birkat Al Mouz, Nizwa 616, Oman
| | - Betty Annie Samuel
- School of Pharmacy, University of Nizwa, Birkat Al Mouz, Nizwa 616, Oman
| | - Saurabh Bhatia
- Natural and Medical Science Research Center, University of Nizwa, Birkat Al Mouz, Nizwa 616, Oman
| | - Shaden A. M. Khalifa
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, S-106 91 Stockholm, Sweden
| | - Hesham R. El-Seedi
- International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang 212013, China
- Pharmacognosy Group, Department of Pharmaceutical Biosciences, BMC, Uppsala University, SE-751 24 Uppsala, Sweden
- International Joint Research Laboratory of Intelligent Agriculture and Agri-Products Processing, Jiangsu Education Department, Jiangsu University, Nanjing 210024, China
| |
Collapse
|
17
|
Klein H, Mazor T, Siegel E, Trukhanov P, Ovalle A, Vecchio Fitz CD, Zwiesler Z, Kumari P, Van Der Veen B, Marriott E, Hansel J, Yu J, Albayrak A, Barry S, Keller RB, MacConaill LE, Lindeman N, Johnson BE, Rollins BJ, Do KT, Beardslee B, Shapiro G, Hector-Barry S, Methot J, Sholl L, Lindsay J, Hassett MJ, Cerami E. MatchMiner: an open-source platform for cancer precision medicine. NPJ Precis Oncol 2022; 6:69. [PMID: 36202909 PMCID: PMC9537311 DOI: 10.1038/s41698-022-00312-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 09/15/2022] [Indexed: 11/17/2022] Open
Abstract
Widespread, comprehensive sequencing of patient tumors has facilitated the usage of precision medicine (PM) drugs to target specific genomic alterations. Therapeutic clinical trials are necessary to test new PM drugs to advance precision medicine, however, the abundance of patient sequencing data coupled with complex clinical trial eligibility has made it challenging to match patients to PM trials. To facilitate enrollment onto PM trials, we developed MatchMiner, an open-source platform to computationally match genomically profiled cancer patients to PM trials. Here, we describe MatchMiner’s capabilities, outline its deployment at Dana-Farber Cancer Institute (DFCI), and characterize its impact on PM trial enrollment. MatchMiner’s primary goals are to facilitate PM trial options for all patients and accelerate trial enrollment onto PM trials. MatchMiner can help clinicians find trial options for an individual patient or provide trial teams with candidate patients matching their trial’s eligibility criteria. From March 2016 through March 2021, we curated 354 PM trials containing a broad range of genomic and clinical eligibility criteria and MatchMiner facilitated 166 trial consents (MatchMiner consents, MMC) for 159 patients. To quantify MatchMiner’s impact on trial consent, we measured time from genomic sequencing report date to trial consent date for the 166 MMC compared to trial consents not facilitated by MatchMiner (non-MMC). We found MMC consented to trials 55 days (22%) earlier than non-MMC. MatchMiner has enabled our clinicians to match patients to PM trials and accelerated the trial enrollment process.
Collapse
Affiliation(s)
- Harry Klein
- Department of Data Science, Dana-Farber Cancer Institute (DFCI), Boston, MA, USA.
| | - Tali Mazor
- Department of Data Science, Dana-Farber Cancer Institute (DFCI), Boston, MA, USA.
| | - Ethan Siegel
- Department of Data Science, Dana-Farber Cancer Institute (DFCI), Boston, MA, USA
| | - Pavel Trukhanov
- Department of Data Science, Dana-Farber Cancer Institute (DFCI), Boston, MA, USA
| | - Andrea Ovalle
- Department of Data Science, Dana-Farber Cancer Institute (DFCI), Boston, MA, USA
| | | | - Zachary Zwiesler
- Department of Data Science, Dana-Farber Cancer Institute (DFCI), Boston, MA, USA
| | - Priti Kumari
- Department of Data Science, Dana-Farber Cancer Institute (DFCI), Boston, MA, USA
| | | | - Eric Marriott
- Department of Data Science, Dana-Farber Cancer Institute (DFCI), Boston, MA, USA
| | - Jason Hansel
- Department of Data Science, Dana-Farber Cancer Institute (DFCI), Boston, MA, USA
| | - Joyce Yu
- Department of Data Science, Dana-Farber Cancer Institute (DFCI), Boston, MA, USA
| | - Adem Albayrak
- Informatics and Analytics, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Susan Barry
- Dana-Farber Cancer Institute, Boston, MA, USA
| | - Rachel B Keller
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | - Neal Lindeman
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Bruce E Johnson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Barrett J Rollins
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.,Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Khanh T Do
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.,Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Brian Beardslee
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Geoffrey Shapiro
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.,Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | | | - John Methot
- Informatics and Analytics, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Lynette Sholl
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - James Lindsay
- Department of Data Science, Dana-Farber Cancer Institute (DFCI), Boston, MA, USA
| | - Michael J Hassett
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Ethan Cerami
- Department of Data Science, Dana-Farber Cancer Institute (DFCI), Boston, MA, USA
| |
Collapse
|
18
|
Yan LD, Yang L, Li N, Wang M, Zhang YH, Zhou W, Yu ZQ, Peng XC, Cai J. Prognostic role of multiple abnormal genes in non-small-cell lung cancer. World J Clin Cases 2022; 10:7772-7784. [PMID: 36158484 PMCID: PMC9372825 DOI: 10.12998/wjcc.v10.i22.7772] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 05/19/2022] [Accepted: 06/17/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Non-small-cell lung cancer (NSCLC) has the highest morbidity and mortality rates among all malignant tumor types. Although therapies targeting the mutated genes such as KRAS have been used in the clinic for many years, the prognosis remains poor. Therefore, it is necessary to further study the aberrant expression or mutation of non-target genes affecting the survival and prognosis.
AIM To explore the impact of simultaneous abnormalities of multiple genes on the prognosis and survival of patients.
METHODS We used R packages to analyze gene expression data and clinical data downloaded from The Cancer Genome Atlas (TCGA) database. We also collected samples from 85 NSCLC patients from the First People’s Hospital of Jingzhou City and retrospectively followed the patients. Multivariate Cox regression analysis and survival analysis were performed.
RESULTS Analysis of gene expression data from TCGA revealed that the overexpression of the following single genes affected overall survival: TP53 (P = 0.79), PTEN (P = 0.94), RB1 (P = 0.49), CTNNB1 (P = 0.24), STK11 (P = 0.32), and PIK3CA (P = 0.013). However, the probability of multiple genes (TP53, PTEN, RB1, and STK11) affecting survival was 0.025. Retrospective analysis of clinical data revealed that sex (hazard ratio [HR] = 1.29; [95%CI: 0.64-2.62]), age (HR = 1.05; [95%CI: 1.02-1.07]), smoking status (HR = 2.26; [95%CI: 1.16-4.39]), tumor histology (HR = 0.58; [95%CI: 0.30-1.11]), cancer stage (HR = 16.63; [95%CI: 4.8-57.63]), epidermal growth factor receptor (EGFR) mutation (HR = 1.82; [95%CI: 1.05-3.16]), abundance (HR = 4.95; [95%CI: 0.78-31.36]), and treatment with tyrosine kinase inhibitors (TKIs) (HR = 0.58; [95%CI: 0.43-0.78]) affected patient survival. Co-occurring mutations of TP53, PTEN, RB1, and STK11 did not significantly affect the overall survival of patients receiving chemotherapy (P = 0.96) but significantly affected the overall survival of patients receiving TKIs (P = 0.045).
CONCLUSION Co-occurring mutation or overexpression of different genes has different effects on the overall survival and prognosis of NSCLC patients. Combined with TKI treatment, the co-occurring mutation of some genes may have a synergistic effect on the survival and prognosis of NSCLC patients.
Collapse
Affiliation(s)
- Lu-Da Yan
- Department of Oncology, The First Affiliated Hospital of Yangtze University, Jingzhou 434023, Hubei Province, China
| | - Liu Yang
- Department of Oncology, The First Affiliated Hospital of Yangtze University, Jingzhou 434023, Hubei Province, China
| | - Na Li
- Department of Oncology, The First Affiliated Hospital of Yangtze University, Jingzhou 434023, Hubei Province, China
| | - Meng Wang
- Department of Oncology, The First Affiliated Hospital of Yangtze University, Jingzhou 434023, Hubei Province, China
| | - Yan-Hua Zhang
- Department of Oncology, The First Affiliated Hospital of Yangtze University, Jingzhou 434023, Hubei Province, China
| | - Wen Zhou
- Department of Oncology, The First Affiliated Hospital of Yangtze University, Jingzhou 434023, Hubei Province, China
| | - Zhi-Qiong Yu
- Department of Oncology, The First Affiliated Hospital of Yangtze University, Jingzhou 434023, Hubei Province, China
| | - Xiao-Chun Peng
- Department of Pathophysiology, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou 434023, Hubei Province, China
| | - Jun Cai
- Department of Oncology, The First Affiliated Hospital of Yangtze University, Jingzhou 434023, Hubei Province, China
| |
Collapse
|
19
|
Girard N, Basse C, Schrock A, Ramkissoon S, Killian K, Ross JS. Comprehensive Genomic Profiling of 274 Thymic Epithelial Tumors Unveils Oncogenic Pathways and Predictive Biomarkers. Oncologist 2022; 27:919-929. [PMID: 35749302 PMCID: PMC9632319 DOI: 10.1093/oncolo/oyac115] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 04/13/2022] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Thymic malignancies represent a heterogeneous group of rare thoracic cancers, which are classified according to the World Health Organization histopathologic classification, that distinguishes thymomas from thymic carcinomas. Data regarding the biology of those tumors are limited in the literature, and the vast majority have been obtained using surgical specimens from early-stage disease. Meanwhile, treatment of advanced, refractory thymic tumors currently relies on chemotherapy, with limited efficacy. Comprehensive genomic profiling (CGP) of advanced, refractory tumors would open some opportunities for innovative treatments. PATIENTS AND METHODS A total of 90 and 174 consecutive patients with thymoma or thymic carcinoma, respectively, for whom formalin-fixed, paraffin-embedded specimens from recurrent, refractory tumor were sequenced, were included. Sequencing was performed using hybridization-captured, adaptor ligation-based libraries to a mean coverage depth of >500× for up to 315 cancer-related genes plus 37 introns from 28 genes frequently rearranged in cancer. RESULTS Thymomas featured a low frequency of genomic alterations (average of 1.8/tumor), and low levels of TMB. The genomic alterations identified in more than 10% of cases were in the CDKN2A/B and TP53 genes. Amplification in the NTRK1 gene was found in an unresectable, stage III, type B3 thymoma. Thymic carcinomas featured a significantly higher frequency of alterations at 4.0/tumor (P < .0001). Clinically relevant genomic alterations were observed in the CDKN2A, KIT, and PTEN/PI3K/MTOR pathways. Elevated TMB in thymic carcinomas was uncommon with only 6% of cases featuring ≥10 mutations/Mb. CONCLUSIONS Our cohort is the largest available so far, reporting on CGP of thymic epithelial tumors in the setting of advanced disease. The identification of clinically relevant genomic alterations in the KIT, PI3K, CDKN2A/B, or NTRK genes provides a strong rationale for potential precision medicine approaches using targeted agents. A subset of thymic carcinomas show high tumor mutation burden, what may be a predictor of efficacy of immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Nicolas Girard
- Corresponding author: Nicolas Girard, Institut Curie, 26 rue d’Ulm, 75005 Paris, France. Tel: +33 144324677; Fax: +33 153104017;
| | - Clémence Basse
- Institut Curie, Institut du Thorax Curie Montsouris, Paris, France,Faculté de Médecine Simonbe Veil, UVSQ, Paris Saclay Campus, Versailles, France
| | | | | | | | - Jeffrey S Ross
- Foundation Medicine, Cambridge, MA, USA,SUNY Upstate Medical University, Syracuse, NY, USA
| |
Collapse
|
20
|
Structural and Functional Thymic Biomarkers Are Involved in the Pathogenesis of Thymic Epithelial Tumors: An Overview. IMMUNO 2022. [DOI: 10.3390/immuno2020025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The normal human thymus originates from the third branchial cleft as two paired anlages that descend into the thorax and fuse on the midline of the anterior–superior mediastinum. Alongside the epithelial and lymphoid components, different types of lymphoid accessory cells, stromal mesenchymal and endothelial cells migrate to, or develop in, the thymus. After reaching maximum development during early postnatal life, the human thymus decreases in size and lymphocyte output drops with age. However, thymic immunological functions persist, although they deteriorate progressively. Several major techniques were fundamental to increasing the knowledge of thymic development and function during embryogenesis, postnatal and adult life; these include immunohistochemistry, immunofluorescence, flow cytometry, in vitro colony assays, transplantation in mice models, fetal organ cultures (FTOC), re-aggregated thymic organ cultures (RTOC), and whole-organ thymic scaffolds. The thymic morphological and functional characterization, first performed in the mouse, was then extended to humans. The purpose of this overview is to provide a report on selected structural and functional biomarkers of thymic epithelial cells (TEC) involved in thymus development and lymphoid cell maturation, and on the historical aspects of their characterization, with particular attention being paid to biomarkers also involved in Thymic Epithelial Tumor (TET) pathogenesis. Moreover, a short overview of targeted therapies in TET, based on currently available experimental and clinical data and on potential future advances will be proposed.
Collapse
|
21
|
Szklener K, Michalski A, Żak K, Piwoński M, Mańdziuk S. Ibrutinib in the Treatment of Solid Tumors: Current State of Knowledge and Future Directions. Cells 2022; 11:1338. [PMID: 35456016 PMCID: PMC9032968 DOI: 10.3390/cells11081338] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 04/10/2022] [Accepted: 04/12/2022] [Indexed: 12/24/2022] Open
Abstract
Bruton's Tyrosine Kinase (BTK) is considered crucial in the activation and survival of both physiological and malignant B-cells. In recent years, ibrutinib, an oral BTK inhibitor, became a breakthrough therapy for hematological malignancies, such as chronic lymphocytic. However, ibrutinib's feasibility might not end there. Several other kinases with established involvement with solid malignancies (i.e., EGFR, HER2) have been found to be inhibited by this agent. Recent discoveries indicate that BTK is a potential anti-solid tumor therapy target. Consequently, ibrutinib, a BTK-inhibitor, has been studied as a therapeutic option in solid malignancies. While most preclinical studies indicate ibrutinib to be an effective therapeutic option in some specific indications, such as NSCLC and breast cancer, clinical trials contradict these observations. Nevertheless, while ibrutinib failed as a monotherapy, it might become an interesting part of a multidrug regime: not only has a synergism between ibrutinib and other compounds, such as trametinib or dactolisib, been observed in vitro, but this BTK inhibitor has also been established as a radio- and chemosensitizer. This review aims to describe the milestones in translating BTK inhibitors to solid tumors in order to understand the future potential of this agent better.
Collapse
Affiliation(s)
- Katarzyna Szklener
- Department of Clinical Oncology and Chemotherapy, Medical University of Lublin, 20-090 Lublin, Poland; (A.M.); (K.Ż.); (M.P.); (S.M.)
| | | | | | | | | |
Collapse
|
22
|
MK2206 attenuates atherosclerosis by inhibiting lipid accumulation, cell migration, proliferation, and inflammation. Acta Pharmacol Sin 2022; 43:897-907. [PMID: 34316032 PMCID: PMC8976090 DOI: 10.1038/s41401-021-00729-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 06/27/2021] [Indexed: 02/07/2023] Open
Abstract
Cardiovascular disease is a common comorbidity in patients with cancer, and the main leading cause of noncancer-related deaths in cancer survivors. Considering that current antitumor drugs usually induce cardiovascular injury, the quest for developing new antitumor drugs, especially those with cardiovascular protection, is crucial for improving cancer prognosis. MK2206 is a phase II clinical anticancer drug and the role of this drug in cardiovascular disease is still unclear. Here, we revealed that MK2206 significantly reduced vascular inflammation, atherosclerotic lesions, and inhibited proliferation of vascular smooth muscle cell in ApoE-/- mice in vivo. We demonstrated that MK2206 reduced lipid accumulation by promoting cholesterol efflux but did not affect lipid uptake and decreased inflammatory response by modulating inflammation-related mRNA stability in macrophages. In addition, we revealed that MK2206 suppressed migration, proliferation, and inflammation in vascular smooth muscle cells. Moreover, MK2206 inhibited proliferation and inflammation of endothelial cells. The present results suggest that MK2206, as a promising drug in clinical antitumor therapy, exhibits anti-inflammatory and antiatherosclerotic potential. This report provides a novel strategy for the prevention of cardiovascular comorbidities in cancer survivors.
Collapse
|
23
|
Cai HY, Yang HS, Shan SC, Lei YY, Zou JY, Zhu Y, Luo HH. A novel signature based on immune-related gene pairs and clinical features to predict prognosis and treatment effect in "driver gene negative" lung adenocarcinoma. Cancer Med 2022; 11:2259-2270. [PMID: 35246970 PMCID: PMC9160806 DOI: 10.1002/cam4.4577] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 01/06/2022] [Accepted: 01/07/2022] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVE Examining the role of immune-related genes (IRGs) in "driver gene negative" lung adenocarcinoma (LUAD) may provide new ideas for the treatment and study for this LUAD subgroup. We aimed to find the hub immune-related gene pairs can stratify the risk of "driver-gene-negative" LUAD. MATERIALS AND METHODS IRGs were identified according to ImmPort database based on RNA sequencing results of tumors and normal tissues from 46 patients with "driver gene negative" LUAD at The First Affiliated Hospital of Sun Yat-sen University and cyclically singly paired as immune-related gene pairs (IRGPs). Multivariate Cox analysis was used to construct an immune risk model and a prognostic nomogram combining was also been developed. Immune microenvironment landscape described by CIBERSORT and drug sensitivity calculated by pRRophetic algorithm were used to explore possible treatment improvements. RESULTS A novel immune risk model with 5-IRGPs (CD1A|CXCL135, CD1A|S100A7L2, IFNA7|CMTM2, IFNA7|CSF3, CAMP|TFR2) can accurately distinguish patients in the high- and low-risk groups. Risk score act as an independent prognostic factor and is related to clinical stage. There are significant differences in tumor immune microenvironment and PD-1/PD-L1/CTLA-4 expression between groups. The low-risk patient may benefit more from the commonly used chemotherapy regimens such as gemcitabine and paclitaxel. CONCLUSION This study constructed 5-IRGPs as a reliable prognostic tool and may represent genes pairs that are potential rationale for choice of treatment for "driver gene negative" LUAD.
Collapse
Affiliation(s)
- He-Yuan Cai
- Department of Thoracic Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hao-Shuai Yang
- Department of Thoracic Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shi-Chao Shan
- Department of Thoracic Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yi-Yan Lei
- Department of Thoracic Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jian-Yong Zou
- Department of Thoracic Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ying Zhu
- Department of Radiology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Hong-He Luo
- Department of Thoracic Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
24
|
Tabbò F, Pisano C, Mazieres J, Mezquita L, Nadal E, Planchard D, Pradines A, Santamaria D, Swalduz A, Ambrogio C, Novello S, Ortiz-Cuaran S. How far we have come targeting BRAF-mutant non-small cell lung cancer (NSCLC). Cancer Treat Rev 2022; 103:102335. [DOI: 10.1016/j.ctrv.2021.102335] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 12/19/2021] [Accepted: 12/27/2021] [Indexed: 12/27/2022]
|
25
|
Mazieres J, Lafitte C, Ricordel C, Greillier L, Negre E, Zalcman G, Domblides C, Madelaine J, Bennouna J, Mascaux C, Moro-Sibilot D, Pinquie F, Cortot AB, Otto J, Cadranel J, Langlais A, Morin F, Westeel V, Besse B. Combination of Trastuzumab, Pertuzumab, and Docetaxel in Patients With Advanced Non-Small-Cell Lung Cancer Harboring HER2 Mutations: Results From the IFCT-1703 R2D2 Trial. J Clin Oncol 2022; 40:719-728. [PMID: 35073148 DOI: 10.1200/jco.21.01455] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
PURPOSE HER2 exon 20 insertions and point mutations are oncogenic drivers found in 1%-2% of patients with non-small-cell lung cancer (NSCLC). No targeted therapy is approved for this subset of patients. We prospectively evaluated the effectiveness of the combination of two antibodies against human epidermal growth factor 2 (HER2 [HER2] trastuzumab and pertuzumab with docetaxel; trastuzumab and pertuzumab) and docetaxel. METHODS The IFCT 1703-R2D2 trial is a multicenter, nonrandomized phase II study. Patients with HER2-mutated, advanced NSCLC who progressed after ≥ 1 platinum-based treatment were enrolled. Patients received pertuzumab at a loading dose of 840 mg and 420 mg thereafter; trastuzumab at an 8 mg/kg loading dose and 6 mg/kg thereafter; and docetaxel at a dose of 75 mg/m2 every 3 weeks. The primary outcome was the objective response rate (ORR). Other end points included the duration of response, progression-free survival, and safety (NCT03845270). RESULTS Forty-five patients were enrolled and treated. The median age was 64.5 years (range, 31-84 years), 35% were smokers, 72% were females, 15% had an Eastern Cooperative Oncology Group performance status of 2, and 30% had brain metastases. The objective response rate was 29% (n = 13), and 58% had stable disease (n = 26). The median progression-free survival was 6.8 months (95% CI, 4.0 to 8.5). The median duration of response in patients with a confirmed response (n = 13) was 11 months (95% CI, 2.9 to 14.9). Grade 3/4 treatment-related adverse events were observed in 64% of the patients. No patient discontinued treatment because of toxicity. The most frequent grade ≥ 3 treatment-related adverse events were neutropenia (33%), diarrhea (13%), and anemia (9%). CONCLUSION Triple therapy with trastuzumab, pertuzumab, and docetaxel is feasible and effective for HER2-mutated pretreated advanced NSCLC. These results highlight the effectiveness of the HER2 antibody-based strategy, which should be considered for these patients.
Collapse
Affiliation(s)
- Julien Mazieres
- Pneumology, CHU Toulouse-Hôpital Larrey, Université Paul Sabatier, Toulouse, France
| | - Claire Lafitte
- Pneumology, Hôpital Cardio-Vasculaire & Pneumologique Louis Pradel, Bron, France
| | | | - Laurent Greillier
- Aix Marseille Univ, APHM, INSERM, CNRS, CRCM, Hôpital Nord, Multidisciplinary Oncology and Therapeutic Innovations, Marseille, France
| | - Elodie Negre
- Thoracic Oncology, Hôpital Arnaud de Villeneuve, Montpellier, France
| | - Gérard Zalcman
- Thoracic Oncology, CIC INSERM 1425, Université de Paris, Hôpital Bichat, Paris, France
| | | | | | - Jaafar Bennouna
- Medical Oncology-CHU de Nantes, CRCINA INSERM, Nantes, France
| | - Céline Mascaux
- Pneumology, Nouvel Hôpital Civil-Hôpitaux Universitaires de Strasbourg, Inserm UMR_S 1113, IRFAC, Strasbourg University, Strasbourg, France
| | | | | | - Alexis B Cortot
- Univ. Lille, CHU Lille, Thoracic Oncology Department, CNRS, Inserm, Institut Pasteur de Lille, UMR9020-UMR-S 1277-Canther, F-59000, Lille, France
| | - Josiane Otto
- Oncology, Centre Antoine Lacassagne, Nice, France
| | | | - Alexandra Langlais
- Biostatistics, Intergroupe Francophone de Cancérologie Thoracique, Paris, France
| | - Franck Morin
- Clinical Research Unit, Intergroupe Francophone de Cancérologie Thoracique, Paris, France
| | | | - Benjamin Besse
- Medicine and Thoracic Pathology Committee, Gustave Roussy, Villejuif, France; Paris-Saclay University, Orsay, France
| |
Collapse
|
26
|
Murphy P, Glynn D, Dias S, Hodgson R, Claxton L, Beresford L, Cooper K, Tappenden P, Ennis K, Grosso A, Wright K, Cantrell A, Stevenson M, Palmer S. Modelling approaches for histology-independent cancer drugs to inform NICE appraisals: a systematic review and decision-framework. Health Technol Assess 2022; 25:1-228. [PMID: 34990339 DOI: 10.3310/hta25760] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND The first histology-independent marketing authorisation in Europe was granted in 2019. This was the first time that a cancer treatment was approved based on a common biomarker rather than the location in the body at which the tumour originated. This research aims to explore the implications for National Institute for Health and Care Excellence appraisals. METHODS Targeted reviews were undertaken to determine the type of evidence that is likely to be available at the point of marketing authorisation and the analyses required to support National Institute for Health and Care Excellence appraisals. Several challenges were identified concerning the design and conduct of trials for histology-independent products, the greater levels of heterogeneity within the licensed population and the use of surrogate end points. We identified approaches to address these challenges by reviewing key statistical literature that focuses on the design and analysis of histology-independent trials and by undertaking a systematic review to evaluate the use of response end points as surrogate outcomes for survival end points. We developed a decision framework to help to inform approval and research policies for histology-independent products. The framework explored the uncertainties and risks associated with different approval policies, including the role of further data collection, pricing schemes and stratified decision-making. RESULTS We found that the potential for heterogeneity in treatment effects, across tumour types or other characteristics, is likely to be a central issue for National Institute for Health and Care Excellence appraisals. Bayesian hierarchical methods may serve as a useful vehicle to assess the level of heterogeneity across tumours and to estimate the pooled treatment effects for each tumour, which can inform whether or not the assumption of homogeneity is reasonable. Our review suggests that response end points may not be reliable surrogates for survival end points. However, a surrogate-based modelling approach, which captures all relevant uncertainty, may be preferable to the use of immature survival data. Several additional sources of heterogeneity were identified as presenting potential challenges to National Institute for Health and Care Excellence appraisal, including the cost of testing, baseline risk, quality of life and routine management costs. We concluded that a range of alternative approaches will be required to address different sources of heterogeneity to support National Institute for Health and Care Excellence appraisals. An exemplar case study was developed to illustrate the nature of the assessments that may be required. CONCLUSIONS Adequately designed and analysed basket studies that assess the homogeneity of outcomes and allow borrowing of information across baskets, where appropriate, are recommended. Where there is evidence of heterogeneity in treatment effects and estimates of cost-effectiveness, consideration should be given to optimised recommendations. Routine presentation of the scale of the consequences of heterogeneity and decision uncertainty may provide an important additional approach to the assessments specified in the current National Institute for Health and Care Excellence methods guide. FURTHER RESEARCH Further exploration of Bayesian hierarchical methods could help to inform decision-makers on whether or not there is sufficient evidence of homogeneity to support pooled analyses. Further research is also required to determine the appropriate basis for apportioning genomic testing costs where there are multiple targets and to address the challenges of uncontrolled Phase II studies, including the role and use of surrogate end points. FUNDING This project was funded by the National Institute for Health Research (NIHR) Evidence Synthesis programme and will be published in full in Health Technology Assessment; Vol. 25, No. 76. See the NIHR Journals Library website for further project information.
Collapse
Affiliation(s)
- Peter Murphy
- Centre for Reviews and Dissemination, University of York, York, UK
| | - David Glynn
- Centre for Health Economics, University of York, York, UK
| | - Sofia Dias
- Centre for Reviews and Dissemination, University of York, York, UK
| | - Robert Hodgson
- Centre for Reviews and Dissemination, University of York, York, UK
| | - Lindsay Claxton
- Centre for Reviews and Dissemination, University of York, York, UK
| | - Lucy Beresford
- Centre for Reviews and Dissemination, University of York, York, UK
| | - Katy Cooper
- School of Health and Related Research (ScHARR) Technology Assessment Group, University of Sheffield, Sheffield, UK
| | - Paul Tappenden
- School of Health and Related Research (ScHARR) Technology Assessment Group, University of Sheffield, Sheffield, UK
| | - Kate Ennis
- School of Health and Related Research (ScHARR) Technology Assessment Group, University of Sheffield, Sheffield, UK
| | | | - Kath Wright
- Centre for Reviews and Dissemination, University of York, York, UK
| | - Anna Cantrell
- School of Health and Related Research (ScHARR) Technology Assessment Group, University of Sheffield, Sheffield, UK
| | - Matt Stevenson
- School of Health and Related Research (ScHARR) Technology Assessment Group, University of Sheffield, Sheffield, UK
| | - Stephen Palmer
- Centre for Health Economics, University of York, York, UK
| |
Collapse
|
27
|
Judd J, Abdel Karim N, Khan H, Naqash AR, Baca Y, Xiu J, VanderWalde AM, Mamdani H, Raez LE, Nagasaka M, Pai SG, Socinski MA, Nieva JJ, Kim C, Wozniak AJ, Ikpeazu C, de Lima Lopes G, Spira AI, Korn WM, Kim ES, Liu SV, Borghaei H. Characterization of KRAS Mutation Subtypes in Non-small Cell Lung Cancer. Mol Cancer Ther 2021; 20:2577-2584. [PMID: 34518295 PMCID: PMC9662933 DOI: 10.1158/1535-7163.mct-21-0201] [Citation(s) in RCA: 86] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 05/25/2021] [Accepted: 09/07/2021] [Indexed: 01/07/2023]
Abstract
KRAS is the most commonly mutated oncogene in NSCLC and development of direct KRAS inhibitors has renewed interest in this molecular variant. Different KRAS mutations may represent a unique biologic context with different prognostic and therapeutic impact. We sought to characterize genomic landscapes of advanced, KRAS-mutated non-small cell lung cancer (NSCLC) in a large national cohort to help guide future therapeutic development.Molecular profiles of 17,095 NSCLC specimens were obtained using DNA next-generation sequencing of 592 genes (Caris Life Sciences) and classified on the basis of presence and subtype of KRAS mutations. Co-occurring genomic alterations, tumor mutational burden (TMB), and PD-L1 expression [22C3, tumor proportion score (TPS) score] were analyzed by KRAS mutation type.Across the cohort, 4,706 (27.5%) samples harbored a KRAS mutation. The most common subtype was G12C (40%), followed by G12V (19%) and G12D (15%). The prevalence of KRAS mutations was 37.2% among adenocarcinomas and 4.4% in squamous cell carcinomas. Rates of high TMB (≥10 mutations/Mb) and PD-L1 expression varied across KRAS mutation subtypes. KRAS G12C was the most likely to be PD-L1 positive (65.5% TPS ≥ 1%) and PD-L1 high (41.3% TPS ≥ 50%). STK11 was mutated in 8.6% of KRAS wild-type NSCLC but more frequent in KRAS-mutant NSCLC, with the highest rate in G13 (36.2%). TP53 mutations were more frequent in KRAS wild-type NSCLC (73.6%).KRAS mutation subtypes have different co-occurring mutations and a distinct genomic landscape. The clinical relevance of these differences in the context of specific therapeutic interventions warrants investigation.
Collapse
Affiliation(s)
- Julia Judd
- Department of Hematology-Oncology, Fox Chase Cancer Center, Temple University Health System, Philadelphia, Pennsylvania
| | - Nagla Abdel Karim
- Department of Hematology-Oncology, Augusta University-Medical College of Georgia, Georgia Cancer Center, Augusta, Georgia
| | - Hina Khan
- Department of Hematology-Oncology, The Warren Alpert Medical School, Brown University, Providence, Rhode Isand
| | - Abdul Rafeh Naqash
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, Maryland.,Medical Oncology/TSET Phase 1 Program, Stephenson Cancer Center, University of Oklahoma, Oklahoma City, Oklahoma
| | | | | | - Ari M. VanderWalde
- Department of Medical Oncology, West Cancer Center and Research Institute, Memphis, Tennessee
| | - Hirva Mamdani
- Department of Oncology, Karmanos Cancer Institute/Wayne State University, Detroit, Michigan
| | - Luis E. Raez
- Department of Hematology-Oncology, Memorial Cancer Institute/Memorial Health Care System/Florida International University, Hollywood, Florida
| | - Misako Nagasaka
- Department of Oncology, Karmanos Cancer Institute/Wayne State University, Detroit, Michigan
| | - Sachin Gopalkrishna Pai
- Department of Medical Oncology, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama
| | - Mark A. Socinski
- Department of Medical Oncology, AdventHealth Cancer Institute, Orlando, Florida
| | - Jorge J. Nieva
- Department of Medical Oncology, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California
| | - Chul Kim
- Department of Hematology-Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC
| | - Antoinette J. Wozniak
- Department of Medical Oncology, University of Pittsburgh Medical Center, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Chukwuemeka Ikpeazu
- Department of Medical Oncology, Sylvester Comprehensive Cancer Center, University of Miami and the Miller School of Medicine, Miami, Florida
| | - Gilberto de Lima Lopes
- Department of Medical Oncology, Sylvester Comprehensive Cancer Center, University of Miami and the Miller School of Medicine, Miami, Florida
| | - Alexander I. Spira
- Department of Medical Oncology, Virginia Cancer Specialists, US Oncology Research, Fairfax, Virginia
| | | | - Edward S. Kim
- Department of Medical Oncology, City of Hope, Los Angeles, California
| | - Stephen V. Liu
- Department of Hematology-Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC
| | - Hossein Borghaei
- Department of Hematology-Oncology, Fox Chase Cancer Center, Temple University Health System, Philadelphia, Pennsylvania.,Corresponding Author: Hossein Borghaei, Medical Oncology, Fox Chase Cancer Center, 333 Cottman Ave, Philadelphia, PA 19111. Phone: 215-214-4297; Fax: 215-728-3639; E-mail:
| |
Collapse
|
28
|
Huang WC, Yadav VK, Cheng WH, Wang CH, Hsieh MS, Huang TY, Lin SF, Yeh CT, Kuo KT. The MEK/ERK/miR-21 Signaling Is Critical in Osimertinib Resistance in EGFR-Mutant Non-Small Cell Lung Cancer Cells. Cancers (Basel) 2021; 13:cancers13236005. [PMID: 34885115 PMCID: PMC8657072 DOI: 10.3390/cancers13236005] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/22/2021] [Accepted: 11/25/2021] [Indexed: 12/24/2022] Open
Abstract
Simple Summary Our study provided data that the inhibition of MEK/ERK signaling could overcome Osimertinib resistance both in vitro and in vivo. Mechanistically, MEK inhibitor Trametinib suppressed the tumorigenic properties of NSCLC cells by reducing the generation of CAFs. The trametinib-mediated anti-cancer function was also associated with the significantly suppressed level of miR-21, of which primary targets included PDCD4, as shown in this study and MEK inhibitor Trametinib significantly suppressed Osimertinib-resistant NSCLC tumor growth by abolishing both processes. Abstract Background: The third-generation epidermal growth factor receptor (EGFR) inhibitor, Osimertinib, is used to treat non-small cell lung cancer (NSCLC) patients with tyrosine kinase inhibitor (TKI) resistance caused by acquired EGFR T790M mutation. However, patients eventually develop resistance against Osimertinib with mechanisms not yet fully clarified. Activated alternative survival pathways within the tumor cells and cancer-associated fibroblasts (CAFs) have been proposed to contribute to Osimertinib resistance. MET and MEK inhibitors may overcome EGFR-independent resistance. Another acquired resistance mechanism of EGFR-TKI is the up-regulation of the RAS/RAF/MEK/ERK signaling pathway, which is the key to cell survival and proliferation; this may occur downstream of various other signaling pathways. In this report, we reveal the possible regulatory mechanism and inhibitory effect of the MEK inhibitor trametinib applied to MEK/ERK/miR-21 axis and PDCD4 in Osimertinib resistance. We found a possible regulatory role of PDCD4 in ERK signaling. PDCD4 is a new type of tumor suppressor that has multiple functions of inhibiting cell growth, tumor invasion, metastasis, and inducing apoptosis. Previous bioinformatics analysis has confirmed that PDCD4 contains the binding site of miR-21 and acts as a tumor suppressor in the regulation of various processes associated with the development of cancer, including cell proliferation, invasion, metastasis, and neoplastic transformation. Based on the above analysis, we hypothesized that the tumor suppressor PDCD4 is one of the effective inhibitory targets of miR-21-5p. Methods: The expression between EGFR and ERK2 in lung adenocarcinoma was evaluated from the TCGA database. Osimertinib-sensitive and resistant NSCLC cells obtained from patients were used to co-culture with human lung fibroblasts (HLFs) to generate CAF cells (termed CAF_R1 and CAF_S1), and the functional roles of these CAF cells plus the regulatory mechanisms were further explored. Then, MEK inhibitor Trametinib with or without Osimertinib was applied in xenograft model derived from patients to validate the effects on growth inhibition of Osimertinib-resistant NSCLC tumors. Result: ERK2 expression correlated with EGFR expression and higher ERK2 level was associated with worse prognosis of patients and Osimertinib resistance. CAFs derived from Osimertinib-resistant cells secreted more IL-6, IL-8, and hepatocyte growth factor (HGF), expressed stronger CAF markers including α-smooth muscle actin (α-SMA), fibroblast activation protein (FAP) plus platelet-derived growth factor receptor (PDGFR), and enhanced stemness and Osimertinib resistance in NSCLC cells. Meanwhile, increased MEK/ERK/miR-21 expressions were found in both CAFs and NSCLC cells. MEK inhibitor Trametinib significantly abrogated the abovementioned effects by modulating β-catenin, STAT3, and ERK. The xenograft model showed combining Osimertinib and Trametinib resulted in the most prominent growth inhibition of Osimertinib-resistant NSCLC tumors. Conclusions: Our results suggested that MEK/ERK/miR-21 signaling is critical in Osimertinib resistance and CAF transformation of NSCLC cells, and MEK inhibitor Trametinib significantly suppressed Osimertinib-resistant NSCLC tumor growth by abolishing both processes.
Collapse
Affiliation(s)
- Wen-Chien Huang
- Department of Medicine, MacKay Medical College, New Taipei City 252, Taiwan
- Division of Thoracic Surgery, Department of Surgery, MacKay Memorial Hospital, Taipei 104, Taiwan
| | - Vijesh Kumar Yadav
- Department of Medical Research & Education, Taipei Medical University-Shuang Ho Hospital, New Taipei City 235, Taiwan
| | - Wei-Hong Cheng
- Division of Hematology and Oncology, Department of Internal Medicine, Taipei Medical University-Shuang Ho Hospital, New Taipei City 235, Taiwan
| | - Chun-Hua Wang
- Department of Dermatology, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, 231, Taiwan
- School of Medicine, Buddhist Tzu Chi University, Hualien 970, Taiwan
| | - Ming-Shou Hsieh
- Department of Medical Research & Education, Taipei Medical University-Shuang Ho Hospital, New Taipei City 235, Taiwan
| | - Ting-Yi Huang
- Department of Medical Research & Education, Taipei Medical University-Shuang Ho Hospital, New Taipei City 235, Taiwan
| | - Shiou-Fu Lin
- Department of Pathology, Taipei Medical University-Shuang Ho Hospital, New Taipei City 235, Taiwan
| | - Chi-Tai Yeh
- Department of Medical Research & Education, Taipei Medical University-Shuang Ho Hospital, New Taipei City 235, Taiwan
- Division of Hematology and Oncology, Department of Internal Medicine, Taipei Medical University-Shuang Ho Hospital, New Taipei City 235, Taiwan
- Department of Medical Laboratory Science and Biotechnology, Yuanpei University of Medical Technology, Hsinchu 300, Taiwan
| | - Kuang-Tai Kuo
- Division of Thoracic Surgery, Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
- Division of Thoracic Surgery, Department of Surgery, Taipei Medical University-Shuang Ho Hospital, New Taipei City 235, Taiwan
| |
Collapse
|
29
|
Personalization of medical treatments in oncology: time for rethinking the disease concept to improve individual outcomes. EPMA J 2021; 12:545-558. [PMID: 34642594 PMCID: PMC8495186 DOI: 10.1007/s13167-021-00254-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 09/01/2021] [Indexed: 12/12/2022]
Abstract
The agenda of pharmacology discovery in the field of personalized oncology was dictated by the search of molecular targets assumed to deterministically drive tumor development. In this perspective, genes play a fundamental "causal" role while cells simply act as causal proxies, i.e., an intermediate between the molecular input and the organismal output. However, the ceaseless genomic change occurring across time within the same primary and metastatic tumor has broken the hope of a personalized treatment based only upon genomic fingerprint. Indeed, current models are unable in capturing the unfathomable complexity behind the outbreak of a disease, as they discard the contribution of non-genetic factors, environment constraints, and the interplay among different tiers of organization. Herein, we posit that a comprehensive personalized model should view at the disease as a "historical" process, in which different spatially and timely distributed factors interact with each other across multiple levels of organization, which collectively interact with a dynamic gene-expression pattern. Given that a disease is a dynamic, non-linear process - and not a static-stable condition - treatments should be tailored according to the "timing-frame" of each condition. This approach can help in detecting those critical transitions through which the system can access different attractors leading ultimately to diverse outcomes - from a pre-disease state to an overt illness or, alternatively, to recovery. Identification of such tipping points can substantiate the predictive and the preventive ambition of the Predictive, Preventive and Personalized Medicine (PPPM/3PM). However, an unusual effort is required to conjugate multi-omics approaches, data collection, and network analysis reconstruction (eventually involving innovative Artificial Intelligent tools) to recognize the critical phases and the relevant targets, which could help in patient stratification and therapy personalization.
Collapse
|
30
|
Jo U, Murai Y, Takebe N, Thomas A, Pommier Y. Precision Oncology with Drugs Targeting the Replication Stress, ATR, and Schlafen 11. Cancers (Basel) 2021; 13:4601. [PMID: 34572827 PMCID: PMC8465591 DOI: 10.3390/cancers13184601] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 09/08/2021] [Accepted: 09/09/2021] [Indexed: 12/14/2022] Open
Abstract
Precision medicine aims to implement strategies based on the molecular features of tumors and optimized drug delivery to improve cancer diagnosis and treatment. DNA replication is a logical approach because it can be targeted by a broad range of anticancer drugs that are both clinically approved and in development. These drugs increase deleterious replication stress (RepStress); however, how to selectively target and identify the tumors with specific molecular characteristics are unmet clinical needs. Here, we provide background information on the molecular processes of DNA replication and its checkpoints, and discuss how to target replication, checkpoint, and repair pathways with ATR inhibitors and exploit Schlafen 11 (SLFN11) as a predictive biomarker.
Collapse
Affiliation(s)
- Ukhyun Jo
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology, Center for Cancer Research, NCI, NIH, Bethesda, MD 20892-4264, USA; (Y.M.); (A.T.)
| | - Yasuhisa Murai
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology, Center for Cancer Research, NCI, NIH, Bethesda, MD 20892-4264, USA; (Y.M.); (A.T.)
- Department of Gastroenterology and Hematology, Hirosaki University Graduate School of Medicine, Hirosaki 036-8562, Japan
| | - Naoko Takebe
- Developmental Therapeutics Branch and Division of Cancer Treatment and Diagnosis, NCI, NIH, Bethesda, MD 20892-4264, USA;
| | - Anish Thomas
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology, Center for Cancer Research, NCI, NIH, Bethesda, MD 20892-4264, USA; (Y.M.); (A.T.)
| | - Yves Pommier
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology, Center for Cancer Research, NCI, NIH, Bethesda, MD 20892-4264, USA; (Y.M.); (A.T.)
| |
Collapse
|
31
|
Zhao W, Ma W, Wang F, Hu F. Incorporating covariates information in adaptive clinical trials for precision medicine. Pharm Stat 2021; 21:176-195. [PMID: 34369053 DOI: 10.1002/pst.2160] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 06/02/2021] [Accepted: 07/20/2021] [Indexed: 11/05/2022]
Abstract
Precision medicine is the systematic use of information that pertains to an individual patient to select or optimize that patient's preventative and therapeutic care. Recent studies have classified biomarkers into predictive and prognostic biomarkers based on their roles in clinical studies. To design a clinical trial for precision medicine, predictive biomarkers and prognostic biomarkers should both be included. In statistical analysis, biomarkers are mathematically treated as covariates. We first classify covariates into predictive and prognostic covariates according to their roles. We then provide a brief review of recent advances in adaptive designs that incorporate covariates. However, the literature includes no designs that incorporate both prognostic covariates and predictive covariates simultaneously. In this paper, we propose a new family of covariate-adjusted response-adaptive (CARA) designs that incorporate both prognostic and predictive covariates and the responses. It is important to note that the predictive biomarkers and prognostic biomarkers play different roles in the new designs. The advantages of the proposed methods are demonstrated via numerical studies, and some further statistical issues are also discussed.
Collapse
Affiliation(s)
- Wanying Zhao
- Department of Biostatistics, Incyte Corporation, Wilmington, Delaware, USA
| | - Wei Ma
- Institute of Statistics and Big Data, Renmin University of China, Beijing, China
| | - Fan Wang
- Department of Statistics, The George Washington University, Washington, District of Columbia, USA
| | - Feifang Hu
- Department of Statistics, The George Washington University, Washington, District of Columbia, USA
| |
Collapse
|
32
|
Krajewska M, Rauch G. A new basket trial design based on clustering of homogeneous subpopulations. J Biopharm Stat 2021; 31:425-447. [PMID: 34236938 DOI: 10.1080/10543406.2021.1897993] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
As new findings in oncology suggest a focus on individualized and targeted therapies, the demand for adequate clinical trial designs rises, whereby the focus is mainly on early development phases (phase I and II). Phase II oncology trials are often planned and analysed by Simon two-stage design, which corresponds to a one-armed trial design with the option to stop early for futility. Whereas a classical phase II study focuses on one tumour type and location, the relatively new basket trial design allows testing the efficacy of a single drug simultaneously in a number of patient subsets, which correspond to different tumour types. Such trials can be analysed in various ways, including separate analyses of all baskets or by pooling across all baskets. The work presented here tries to find an adequate compromise between these two extremes by implying rules for clustering some baskets, which are reasonably homogeneous. By means of Monte-Carlo simulations, we compare the efficiency of our proposed cluster-based basket trial design with a standard approach proposed recently which only allows for complete pooling or separate analyses. The results suggest that our new design offers a considerable advantage in power, sensitivity and specificity as well as in average sample size compared to the standard approach. The proposed clustering design is an attractive option to conduct basket trials in oncology with higher efficiency and better performance.
Collapse
Affiliation(s)
- Maja Krajewska
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, and Humboldt-Universität zu Berlin, Institute of Biometry and Clinical Epidemiology, Berlin, Germany
| | - Geraldine Rauch
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, and Humboldt-Universität zu Berlin, Institute of Biometry and Clinical Epidemiology, Berlin, Germany
| |
Collapse
|
33
|
Yu L, Wei J, Liu P. Attacking the PI3K/Akt/mTOR signaling pathway for targeted therapeutic treatment in human cancer. Semin Cancer Biol 2021; 85:69-94. [PMID: 34175443 DOI: 10.1016/j.semcancer.2021.06.019] [Citation(s) in RCA: 205] [Impact Index Per Article: 68.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 06/10/2021] [Accepted: 06/22/2021] [Indexed: 02/08/2023]
Abstract
Cancer is the second leading cause of human death globally. PI3K/Akt/mTOR signaling is one of the most frequently dysregulated signaling pathways observed in cancer patients that plays crucial roles in promoting tumor initiation, progression and therapy responses. This is largely due to that PI3K/Akt/mTOR signaling is indispensable for many cellular biological processes, including cell growth, metastasis, survival, metabolism, and others. As such, small molecule inhibitors targeting major kinase components of the PI3K/Akt/mTOR signaling pathway have drawn extensive attention and been developed and evaluated in preclinical models and clinical trials. Targeting a single kinase component within this signaling usually causes growth arrest rather than apoptosis associated with toxicity-induced adverse effects in patients. Combination therapies including PI3K/Akt/mTOR inhibitors show improved patient response and clinical outcome, albeit developed resistance has been reported. In this review, we focus on revealing the mechanisms leading to the hyperactivation of PI3K/Akt/mTOR signaling in cancer and summarizing efforts for developing PI3K/Akt/mTOR inhibitors as either mono-therapy or combination therapy in different cancer settings. We hope that this review will facilitate further understanding of the regulatory mechanisms governing dysregulation of PI3K/Akt/mTOR oncogenic signaling in cancer and provide insights into possible future directions for targeted therapeutic regimen for cancer treatment, by developing new agents, drug delivery systems, or combination regimen to target the PI3K/Akt/mTOR signaling pathway. This information will also provide effective patient stratification strategy to improve the patient response and clinical outcome for cancer patients with deregulated PI3K/Akt/mTOR signaling.
Collapse
Affiliation(s)
- Le Yu
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Biochemistry and Biophysics, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | | | - Pengda Liu
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Biochemistry and Biophysics, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
34
|
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: 11.3] [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
|
35
|
Masciale V, Banchelli F, Grisendi G, D’Amico R, Maiorana A, Stefani A, Morandi U, Dominici M, Aramini B. New Perspectives in Different Gene Expression Profiles for Early and Locally Advanced Non-Small Cell Lung Cancer Stem Cells. Front Oncol 2021; 11:613198. [PMID: 33868998 PMCID: PMC8047623 DOI: 10.3389/fonc.2021.613198] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 03/15/2021] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION Lung cancer is one of the most common cancers in the world, causing over 1.7 million deaths in 2018. Thus far, no effective treatments against lung cancer for advanced stages have been found. For early stages, although surgery is considered the gold standard treatment, 30-55% of patients develop recurrence within the first 5 years of surgery. Our aim is to assess whether cancer stem cells (CSC) display overexpression of a pool of genes that were previously identified for adenocarcinoma recurrence in patients with early and locally advanced stages of non-small cell lung cancer (NSCLC). METHODS This cross-sectional study was carried out by harvesting surgical tumor specimens obtained from patients harboring early (I-II) and locally advanced (IIIA) stages of NSCLC. For each patient, cell sorting was performed to identify and isolate the ALDHhigh (CSC) and ALDHlow (cancer cells) populations. The mRNA expressions of 31 recurrence-related genes (target genes) in both ALDHhigh and ALDHlow populations were then assessed and compared. RESULTS Surgical specimens were obtained from 22 patients harboring NSCLC. Sixteen (51.6%) out of 31 recurrence-related genes were significantly overexpressed in ALDHhigh cells in the early stages and 9 (29.0%) were overexpressed in the locally advanced stages of NSCLC. Overall, the relative mRNA expressions for these recurrence-related genes were higher in early-stage patients. The average fold change, considering all 31 recurrence-related genes together, was 4.5 (95% CI = 3.1-6.3) in early-stage patients and 1.6 (95% CI = 1.2-2.2) in locally advanced-stage patients. CONCLUSIONS Our study represents the first attempt toward identifying genes associated with recurrence that are overexpressed in cancer stem cells in patients with early and locally advanced stages of NSCLC. This finding may contribute to the identification of new target therapies tailored for NSCLC stages.
Collapse
Affiliation(s)
- Valentina Masciale
- Division of Thoracic Surgery, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Federico Banchelli
- Center of Statistic, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Giulia Grisendi
- Division of Oncology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Roberto D’Amico
- Center of Statistic, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Antonino Maiorana
- Department of Medical and Surgical Sciences, Institute of Pathology, University of Modena and Reggio Emilia, Modena, Italy
| | - Alessandro Stefani
- Division of Thoracic Surgery, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Uliano Morandi
- Division of Thoracic Surgery, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Massimo Dominici
- Division of Oncology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Beatrice Aramini
- Division of Thoracic Surgery, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
36
|
Conlon NT, Kooijman JJ, van Gerwen SJC, Mulder WR, Zaman GJR, Diala I, Eli LD, Lalani AS, Crown J, Collins DM. Comparative analysis of drug response and gene profiling of HER2-targeted tyrosine kinase inhibitors. Br J Cancer 2021; 124:1249-1259. [PMID: 33473169 PMCID: PMC8007737 DOI: 10.1038/s41416-020-01257-x] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 12/09/2020] [Accepted: 12/17/2020] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Human epidermal growth factor 2 (HER2/ERBB2) is frequently amplified/mutated in cancer. The tyrosine kinase inhibitors (TKIs) lapatinib, neratinib, and tucatinib are FDA-approved for the treatment of HER2-positive breast cancer. Direct comparisons of the preclinical efficacy of the TKIs have been limited to small-scale studies. Novel biomarkers are required to define beneficial patient populations. METHODS In this study, the anti-proliferative effects of the three TKIs were directly compared using a 115 cancer cell line panel. Novel TKI response/resistance markers were identified through cross-analysis of drug response profiles with mutation, gene copy number and expression data. RESULTS All three TKIs were effective against HER2-amplified breast cancer models; neratinib showing the most potent activity, followed by tucatinib then lapatinib. Neratinib displayed the greatest activity in HER2-mutant and EGFR-mutant cells. High expression of HER2, VTCN1, CDK12, and RAC1 correlated with response to all three TKIs. DNA damage repair genes were associated with TKI resistance. BRCA2 mutations were correlated with neratinib and tucatinib response, and high expression of ATM, BRCA2, and BRCA1 were associated with neratinib resistance. CONCLUSIONS Neratinib was the most effective HER2-targeted TKI against HER2-amplified, -mutant, and EGFR-mutant cell lines. This analysis revealed novel resistance mechanisms that may be exploited using combinatorial strategies.
Collapse
Affiliation(s)
- Neil T Conlon
- National Institute of Cellular Biotechnology, Dublin City University, Glasnevin, Dublin, Ireland.
| | - Jeffrey J Kooijman
- Netherlands Translational Research Center B.V., Kloosterstraat 9, 5349 AB, Oss, The Netherlands
| | - Suzanne J C van Gerwen
- Netherlands Translational Research Center B.V., Kloosterstraat 9, 5349 AB, Oss, The Netherlands
| | - Winfried R Mulder
- Netherlands Translational Research Center B.V., Kloosterstraat 9, 5349 AB, Oss, The Netherlands
| | - Guido J R Zaman
- Netherlands Translational Research Center B.V., Kloosterstraat 9, 5349 AB, Oss, The Netherlands
| | - Irmina Diala
- Puma Biotechnology, Inc., 10880 Wilshire Boulevard, Suite 2150, Los Angeles, CA, 90024, USA
| | - Lisa D Eli
- Puma Biotechnology, Inc., 10880 Wilshire Boulevard, Suite 2150, Los Angeles, CA, 90024, USA
| | - Alshad S Lalani
- Puma Biotechnology, Inc., 10880 Wilshire Boulevard, Suite 2150, Los Angeles, CA, 90024, USA
| | - John Crown
- National Institute of Cellular Biotechnology, Dublin City University, Glasnevin, Dublin, Ireland
- Department of Medical Oncology, St Vincent's University Hospital, Dublin, Ireland
| | - Denis M Collins
- National Institute of Cellular Biotechnology, Dublin City University, Glasnevin, Dublin, Ireland
| |
Collapse
|
37
|
Han J, Liu Y, Yang S, Wu X, Li H, Wang Q. MEK inhibitors for the treatment of non-small cell lung cancer. J Hematol Oncol 2021; 14:1. [PMID: 33402199 PMCID: PMC7786519 DOI: 10.1186/s13045-020-01025-7] [Citation(s) in RCA: 155] [Impact Index Per Article: 51.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 12/14/2020] [Indexed: 02/07/2023] Open
Abstract
BRAF and KRAS are two key oncogenes in the RAS/RAF/MEK/MAPK signaling pathway. Concomitant mutations in both KRAS and BRAF genes have been identified in non-small cell lung cancer (NSCLC). They lead to the proliferation, differentiation, and apoptosis of tumor cells by activating the RAS/RAF/MEK/ERK signaling pathway. To date, agents that target RAS/RAF/MEK/ERK signaling pathway have been investigated in NSCLC patients harboring BRAF mutations. BRAF and MEK inhibitors have gained approval for the treatment of patients with NSCLC. According to the reported findings, the combination of MEK inhibitors with chemotherapy, immune checkpoint inhibitors, epidermal growth factor receptor-tyrosine kinase inhibitors or BRAF inhibitors is highly significant for improving clinical efficacy and causing delay in the occurrence of drug resistance. This review summarized the existing experimental results and presented ongoing clinical studies as well. However, further researches need to be conducted to indicate how we can combine other drugs with MEK inhibitors to significantly increase therapeutic effects on patients with lung cancer.
Collapse
Affiliation(s)
- Jing Han
- Department of Internal Medicine, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, 127 Dong Ming Road, Zhengzhou, 450008, China
| | - Yang Liu
- Department of Radiotherapy, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, 127 Dong Ming Road, Zhengzhou, 450008, China
| | - Sen Yang
- Department of Internal Medicine, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, 127 Dong Ming Road, Zhengzhou, 450008, China
| | - Xuan Wu
- Department of Internal Medicine, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, 127 Dong Ming Road, Zhengzhou, 450008, China
| | - Hongle Li
- Department of Molecular Pathology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, 127 Dong Ming Road, Zhengzhou, 450008, China.
| | - Qiming Wang
- Department of Internal Medicine, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, 127 Dong Ming Road, Zhengzhou, 450008, China.
| |
Collapse
|
38
|
Mitani Y, Kanai M, Kou T, Kataoka S, Doi K, Matsubara J, Ohashi S, Matsumoto S, Muto M. Cancer of unknown primary with EGFR mutation successfully treated with targeted therapy directed by clinical next-generation sequencing: a case report. BMC Cancer 2020; 20:1177. [PMID: 33267781 PMCID: PMC7709432 DOI: 10.1186/s12885-020-07640-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 11/16/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Cancer of unknown primary (CUP) is usually treated with nonselective and empirical chemotherapy; however, its prognosis is generally poor, with a median survival of less than a year. Thus, clinicians eagerly await the development of more effective treatment strategies. In recent years, advances in next-generation sequencing (NGS) have made it possible to analyze comprehensively the genome of individual cancers. NGS has identified many genomic alterations, some of which are potential molecular targets of specific agents. We report a case of CUP that was successfully treated with targeted therapy directed by the genomic data obtained from an NGS-based multiplex assay. CASE PRESENTATION A 52-year-old Asian woman with right hip joint pain underwent fluorodeoxyglucose-positron emission tomography/computed tomography, which showed multiple metastatic tumors in her right hip joint, thyroid gland, lung, and vertebrae. Brain magnetic resonance imaging showed multiple cerebral metastases. Additional tests, including pathology examination and conventional epidermal growth factor receptor (EGFR) gene mutation analysis (single-strand conformation polymorphism assay), could not identify the primary origin of the tumors, so the patient was diagnosed with CUP. After empirical chemotherapy for CUP, an NGS-based multiplex assay performed using a resected specimen of thyroid tumor detected the EGFR mutation c.2573 T > G p.Leu858Arg (L858R). Her treatment was changed to erlotinib, an EGFR tyrosine-kinase inhibiter, which dramatically shrank the tumors and decreased her serum carcinoembryonic antigen level. She achieved long-term disease control and survived for 2 years and 9 months from the first diagnosis. CONCLUSION This case might support the strategy that NGS-based multiplex assays could identify actionable molecular targets for individual patients with CUP.
Collapse
Affiliation(s)
- Yosuke Mitani
- Department of Therapeutic Oncology, Kyoto University Graduate School of Medicine, 54 Shogoin Kawahara-cho, Kyoto, Sakyo-ku, 606-8507, Japan
| | - Masashi Kanai
- Department of Therapeutic Oncology, Kyoto University Graduate School of Medicine, 54 Shogoin Kawahara-cho, Kyoto, Sakyo-ku, 606-8507, Japan
| | - Tadayuki Kou
- Department of Therapeutic Oncology, Kyoto University Graduate School of Medicine, 54 Shogoin Kawahara-cho, Kyoto, Sakyo-ku, 606-8507, Japan
| | - Shigeki Kataoka
- Department of Therapeutic Oncology, Kyoto University Graduate School of Medicine, 54 Shogoin Kawahara-cho, Kyoto, Sakyo-ku, 606-8507, Japan
| | - Keitaro Doi
- Department of Therapeutic Oncology, Kyoto University Graduate School of Medicine, 54 Shogoin Kawahara-cho, Kyoto, Sakyo-ku, 606-8507, Japan
| | - Junichi Matsubara
- Department of Therapeutic Oncology, Kyoto University Graduate School of Medicine, 54 Shogoin Kawahara-cho, Kyoto, Sakyo-ku, 606-8507, Japan
| | - Shinya Ohashi
- Department of Therapeutic Oncology, Kyoto University Graduate School of Medicine, 54 Shogoin Kawahara-cho, Kyoto, Sakyo-ku, 606-8507, Japan
| | - Shigemi Matsumoto
- Department of Therapeutic Oncology, Kyoto University Graduate School of Medicine, 54 Shogoin Kawahara-cho, Kyoto, Sakyo-ku, 606-8507, Japan
| | - Manabu Muto
- Department of Therapeutic Oncology, Kyoto University Graduate School of Medicine, 54 Shogoin Kawahara-cho, Kyoto, Sakyo-ku, 606-8507, Japan.
| |
Collapse
|
39
|
Catenacci DVT, Moya S, Lomnicki S, Chase LM, Peterson BF, Reizine N, Alpert L, Setia N, Xiao SY, Hart J, Siddiqui UD, Hogarth DK, Eng OS, Turaga K, Roggin K, Posner MC, Chang P, Narula S, Rampurwala M, Ji Y, Karrison T, Liao CY, Polite BN, Kindler HL. Personalized Antibodies for Gastroesophageal Adenocarcinoma (PANGEA): A Phase II Study Evaluating an Individualized Treatment Strategy for Metastatic Disease. Cancer Discov 2020; 11:308-325. [PMID: 33234578 DOI: 10.1158/2159-8290.cd-20-1408] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 11/01/2020] [Accepted: 11/18/2020] [Indexed: 02/07/2023]
Abstract
The one-year and median overall survival (mOS) rates of advanced gastroesophageal adenocarcinomas (GEA) are ∼50% and <12 months, respectively. Baseline spatial and temporal molecular heterogeneity of targetable alterations may be a cause of failure of targeted/immunooncologic therapies. This heterogeneity, coupled with infrequent incidence of some biomarkers, has resulted in stalled therapeutic progress. We hypothesized that a personalized treatment strategy, applied at first diagnosis then serially over up to three treatment lines using monoclonal antibodies combined with optimally sequenced chemotherapy, could contend with these hurdles. This was tested using a novel clinical expansion-platform type II design with a survival primary endpoint. Of 68 patients by intention-to-treat, the one-year survival rate was 66% and mOS was 15.7 months, meeting the primary efficacy endpoint (one-sided P = 0.0024). First-line response rate (74%), disease control rate (99%), and median progression-free survival (8.2 months) were superior to historical controls. The PANGEA strategy led to improved outcomes warranting a larger randomized study. SIGNIFICANCE: This study highlights excellent outcomes achieved by individually optimizing chemotherapy, biomarker profiling, and matching of targeted therapies at baseline and over time for GEA. Testing a predefined treatment strategy resulted in improved outcomes versus historical controls. Therapeutic resistance observed in correlative analyses suggests that dual targeted inhibition may be beneficial.This article is highlighted in the In This Issue feature, p. 211.
Collapse
Affiliation(s)
- Daniel V T Catenacci
- The University of Chicago, Section of Hematology/Oncology, Department of Medicine, Chicago, Illinois.
| | - Stephanie Moya
- The University of Chicago, Section of Hematology/Oncology, Department of Medicine, Chicago, Illinois
| | - Samantha Lomnicki
- The University of Chicago, Section of Hematology/Oncology, Department of Medicine, Chicago, Illinois
| | - Leah M Chase
- The University of Chicago, Section of Hematology/Oncology, Department of Medicine, Chicago, Illinois
| | - Bryan F Peterson
- The University of Chicago, Section of Hematology/Oncology, Department of Medicine, Chicago, Illinois
| | - Natalie Reizine
- The University of Chicago, Section of Hematology/Oncology, Department of Medicine, Chicago, Illinois
| | - Lindsay Alpert
- The University of Chicago, Department of Pathology, Chicago, Illinois
| | - Namrata Setia
- The University of Chicago, Department of Pathology, Chicago, Illinois
| | - Shu-Yuan Xiao
- The University of Chicago, Department of Pathology, Chicago, Illinois
| | - John Hart
- The University of Chicago, Department of Pathology, Chicago, Illinois
| | - Uzma D Siddiqui
- The University of Chicago, Department of Medicine, Center for Endoscopic Research and Therapeutics (CERT), Chicago, Illinois
| | - D Kyle Hogarth
- The University of Chicago, Department of Medicine, Section of Pulmonology, Chicago, Illinois
| | - Oliver S Eng
- The University of Chicago, Department of Surgery, Chicago, Illinois
| | - Kiran Turaga
- The University of Chicago, Department of Surgery, Chicago, Illinois
| | - Kevin Roggin
- The University of Chicago, Department of Surgery, Chicago, Illinois
| | | | - Paul Chang
- The University of Chicago, Department of Radiology, Chicago, Illinois
| | | | | | - Yuan Ji
- The University of Chicago, Department of Public Health Sciences, Chicago, Illinois
| | - Theodore Karrison
- The University of Chicago, Department of Public Health Sciences, Chicago, Illinois
| | - Chih-Yi Liao
- The University of Chicago, Section of Hematology/Oncology, Department of Medicine, Chicago, Illinois
| | - Blase N Polite
- The University of Chicago, Section of Hematology/Oncology, Department of Medicine, Chicago, Illinois
| | - Hedy L Kindler
- The University of Chicago, Section of Hematology/Oncology, Department of Medicine, Chicago, Illinois
| |
Collapse
|
40
|
Gittings LM, Sattler R. Recent advances in understanding amyotrophic lateral sclerosis and emerging therapies. Fac Rev 2020; 9:12. [PMID: 33659944 PMCID: PMC7886072 DOI: 10.12703/b/9-12] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease that is characterized by degeneration of both upper and lower motor neurons and subsequent progressive loss of muscle function. Within the last decade, significant progress has been made in the understanding of the etiology and pathobiology of the disease; however, treatment options remain limited and only two drugs, which exert a modest effect on survival, are approved for ALS treatment in the US. Therefore, the search for effective ALS therapies continues, and over 60 clinical trials are in progress for patients with ALS and other therapeutics are at the pre-clinical stage of development. Recent advances in understanding the genetics, pathology, and molecular mechanisms of ALS have led to the identification of novel targets and strategies that are being used in emerging ALS therapeutic interventions. Here, we review the current status and mechanisms of action of a selection of emerging ALS therapies in pre-clinical or early clinical development, including gene therapy, immunotherapy, and strategies that target neuroinflammation, phase separation, and protein clearance.
Collapse
Affiliation(s)
- Lauren M Gittings
- Department of Neurobiology, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Rita Sattler
- Department of Neurobiology, Barrow Neurological Institute, Phoenix, AZ, USA
| |
Collapse
|
41
|
Basket trials: From tumour gnostic to tumour agnostic drug development. Cancer Treat Rev 2020; 90:102082. [DOI: 10.1016/j.ctrv.2020.102082] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 07/07/2020] [Accepted: 07/10/2020] [Indexed: 12/14/2022]
|
42
|
Nimmagadda S. Quantifying PD-L1 Expression to Monitor Immune Checkpoint Therapy: Opportunities and Challenges. Cancers (Basel) 2020; 12:cancers12113173. [PMID: 33137949 PMCID: PMC7692040 DOI: 10.3390/cancers12113173] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 10/19/2020] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Malignant cells hijack the regulatory roles of immune checkpoint proteins for immune evasion and survival. Therapeutics blocking those proteins can restore the balance of the immune system and lead to durable responses in cancer patients. Although a subset of patients derive benefit, there are few non-invasive technologies to guide and monitor those therapies to improve success rates. This is a review of the advancements in non-invasive methods for quantification of immune checkpoint protein programmed death ligand 1 expression, a biomarker detected by immunohistochemistry and widely used for guiding immune checkpoint therapy. Abstract Therapeutics targeting programmed death ligand 1 (PD-L1) protein and its receptor PD-1 are now dominant players in restoring anti-tumor immune responses. PD-L1 detection by immunohistochemistry (IHC) is emerging as a reproducible biomarker for guiding patient stratification for those therapies in some cancers. However, PD-L1 expression in the tumor microenvironment is highly complex. It is upregulated by aberrant genetic alterations, and is highly regulated at the transcriptional, posttranscriptional, and protein levels. Thus, PD-L1 IHC is inadequate to fully understand the relevance of PD-L1 levels in the whole body and their dynamics to improve therapeutic outcomes. Imaging technologies could potentially assist in meeting that need. Early clinical investigations show promising results in quantifying PD-L1 expression in the whole body by positron emission tomography (PET). Within this context, this review summarizes advancements in regulation of PD-L1 expression and imaging agents, and in PD-L1 PET for drug development, and discusses opportunities and challenges presented by these innovations for guiding immune checkpoint therapy (ICT).
Collapse
Affiliation(s)
- Sridhar Nimmagadda
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; ; Tel.: +1-410-502-6244; Fax: +1-410-614-3147
- Division of Clinical Pharmacology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Pharmacology and Molecular Science, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Bloomberg–Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
43
|
Xuan Y, Bateman NW, Gallien S, Goetze S, Zhou Y, Navarro P, Hu M, Parikh N, Hood BL, Conrads KA, Loosse C, Kitata RB, Piersma SR, Chiasserini D, Zhu H, Hou G, Tahir M, Macklin A, Khoo A, Sun X, Crossett B, Sickmann A, Chen YJ, Jimenez CR, Zhou H, Liu S, Larsen MR, Kislinger T, Chen Z, Parker BL, Cordwell SJ, Wollscheid B, Conrads TP. Standardization and harmonization of distributed multi-center proteotype analysis supporting precision medicine studies. Nat Commun 2020; 11:5248. [PMID: 33067419 PMCID: PMC7568553 DOI: 10.1038/s41467-020-18904-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 09/16/2020] [Indexed: 02/02/2023] Open
Abstract
Cancer has no borders: Generation and analysis of molecular data across multiple centers worldwide is necessary to gain statistically significant clinical insights for the benefit of patients. Here we conceived and standardized a proteotype data generation and analysis workflow enabling distributed data generation and evaluated the quantitative data generated across laboratories of the international Cancer Moonshot consortium. Using harmonized mass spectrometry (MS) instrument platforms and standardized data acquisition procedures, we demonstrate robust, sensitive, and reproducible data generation across eleven international sites on seven consecutive days in a 24/7 operation mode. The data presented from the high-resolution MS1-based quantitative data-independent acquisition (HRMS1-DIA) workflow shows that coordinated proteotype data acquisition is feasible from clinical specimens using such standardized strategies. This work paves the way for the distributed multi-omic digitization of large clinical specimen cohorts across multiple sites as a prerequisite for turning molecular precision medicine into reality.
Collapse
Affiliation(s)
- Yue Xuan
- Thermo Fisher Scientific GmbH, Hanna-Kunath Str. 11, Bremen, 28199, Germany.
| | - Nicholas W Bateman
- Gynecologic Cancer Center of Excellence, Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Uniformed Services University and Walter Reed National Military Medical Center, 8901 Wisconsin Avenue, Bethesda, 20889, MD, USA
| | - Sebastien Gallien
- Thermo Fisher Scientific, Paris, France.,Thermo Fisher Scientific, Precision Medicine Science Center, Cambridge, MA, USA
| | - Sandra Goetze
- Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zurich, Switzerland.,Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Yue Zhou
- Thermo Fisher Scientific Co. Ltd, Shanghai, China
| | - Pedro Navarro
- Thermo Fisher Scientific GmbH, Hanna-Kunath Str. 11, Bremen, 28199, Germany
| | - Mo Hu
- Thermo Fisher Scientific Co. Ltd, Shanghai, China
| | - Niyati Parikh
- Gynecologic Cancer Center of Excellence, Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Uniformed Services University and Walter Reed National Military Medical Center, 8901 Wisconsin Avenue, Bethesda, 20889, MD, USA
| | - Brian L Hood
- Gynecologic Cancer Center of Excellence, Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Uniformed Services University and Walter Reed National Military Medical Center, 8901 Wisconsin Avenue, Bethesda, 20889, MD, USA
| | - Kelly A Conrads
- Gynecologic Cancer Center of Excellence, Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Uniformed Services University and Walter Reed National Military Medical Center, 8901 Wisconsin Avenue, Bethesda, 20889, MD, USA
| | - Christina Loosse
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., Bunsen-Kirchhoff-Straße 11, 44139, Dortmund, Germany
| | - Reta Birhanu Kitata
- Institute of Chemistry, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei, 11529, Taiwan
| | - Sander R Piersma
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, De Boelelaan 1117, 1081 HV, Amsterdam, the Netherlands
| | - Davide Chiasserini
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, De Boelelaan 1117, 1081 HV, Amsterdam, the Netherlands.,Stoller Biomarker Discovery Centre, Institute of Cancer Sciences, Faculty of Medical and Human Sciences, University of Manchester, Manchester, M13 9PL, United Kingdom
| | - Hongwen Zhu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, China
| | - Guixue Hou
- BGI-SHENZHEN, Beishan Road, Yantian District, Shenzhen, 518083, Guangdong, China
| | - Muhammad Tahir
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, Odense M, DK-5230, Denmark
| | - Andrew Macklin
- Princess Margaret Cancer Centre, 101 College Street PMCRT 9-807, Toronto, ON, M5G 1L7, Canada
| | - Amanda Khoo
- Princess Margaret Cancer Centre, 101 College Street PMCRT 9-807, Toronto, ON, M5G 1L7, Canada
| | - Xiuxuan Sun
- National Translational Science Center for Molecular Medicine, Xi'an, 710032, China.,Department of Cell Biology, School of Basic Medicine, Air Force Medical University, Xi'an, 710032, China
| | - Ben Crossett
- Sydney Mass Spectrometry, The University of Sydney, NSW, 2006, Sydney, Australia
| | - Albert Sickmann
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., Bunsen-Kirchhoff-Straße 11, 44139, Dortmund, Germany.,Medizinische Fakultät, Medizinisches Proteom-Center (MPC), Ruhr-Universität Bochum, 44801, Bochum, Germany.,Department of Chemistry, College of Physical Sciences, University of Aberdeen, Aberdeen, AB243FX, Scotland, UK
| | - Yu-Ju Chen
- Institute of Chemistry, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei, 11529, Taiwan
| | - Connie R Jimenez
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, De Boelelaan 1117, 1081 HV, Amsterdam, the Netherlands
| | - Hu Zhou
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, China
| | - Siqi Liu
- BGI-SHENZHEN, Beishan Road, Yantian District, Shenzhen, 518083, Guangdong, China
| | - Martin R Larsen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, Odense M, DK-5230, Denmark
| | - Thomas Kislinger
- Princess Margaret Cancer Centre, 101 College Street PMCRT 9-807, Toronto, ON, M5G 1L7, Canada
| | - Zhinan Chen
- National Translational Science Center for Molecular Medicine, Xi'an, 710032, China.,Department of Cell Biology, School of Basic Medicine, Air Force Medical University, Xi'an, 710032, China
| | - Benjamin L Parker
- School of Life and Environmental Science, The University of Sydney, NSW, 2006, Sydney, Australia
| | - Stuart J Cordwell
- School of Life and Environmental Science, The University of Sydney, NSW, 2006, Sydney, Australia
| | - Bernd Wollscheid
- Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zurich, Switzerland. .,Swiss Institute of Bioinformatics, Lausanne, Switzerland.
| | - Thomas P Conrads
- Women's Health Integrated Research Center, Women's Service Line, Inova Health System, 3289 Woodburn Bldg, Annandale, VA, 22003, USA.
| |
Collapse
|
44
|
Imyanitov EN, Levchenko EV, Kuligina ES, Orlov SV. Treating non-small cell lung cancer with selumetinib: an up-to-date drug evaluation. Expert Opin Pharmacother 2020; 21:1943-1953. [DOI: 10.1080/14656566.2020.1798930] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Evgeny N. Imyanitov
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, St.-Petersburg, 197758, Russia
- Department of Clinical Genetics, St.-Petersburg Pediatric Medical University, St.-Petersburg, 194100, Russia
- Department of Oncology, I.I. Mechnikov North-Western Medical University, St.-Petersburg, 191015, Russia
- Department of Oncology, I.P. Pavlov St.-Petersburg State Medical University, St.-Petersburg, 197022, Russia
- Institute of Medical Primatology, Sochi, 354376, Russia
| | - Evgeny V. Levchenko
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, St.-Petersburg, 197758, Russia
- Department of Oncology, I.I. Mechnikov North-Western Medical University, St.-Petersburg, 191015, Russia
| | - Ekatherina S. Kuligina
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, St.-Petersburg, 197758, Russia
| | - Sergey V. Orlov
- Department of Oncology, I.P. Pavlov St.-Petersburg State Medical University, St.-Petersburg, 197022, Russia
- Institute of Medical Primatology, Sochi, 354376, Russia
| |
Collapse
|
45
|
Yu L, Lai Q, Gou L, Feng J, Yang J. Opportunities and obstacles of targeted therapy and immunotherapy in small cell lung cancer. J Drug Target 2020; 29:1-11. [PMID: 32700566 DOI: 10.1080/1061186x.2020.1797050] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Small cell lung cancer (SCLC) is an aggressive malignant tumour which accounts for approximately 13-15% of all newly diagnosed lung cancer cases. To date, platinum-based chemotherapy are still the first-line treatments for SCLC. However, chemotherapy resistance and systemic toxicity limit the long-term clinical outcome of first-line treatment in SCLC. Recent years, targeted therapy and immunotherapy have made great breakthrough in cancer therapy, and researchers aim to exploit both as a single agent or in combination with chemotherapy to improve the survival of SCLC patients, but limited effectiveness and the adverse events remain the major obstacles in the treatment of SCLC. To overcome these challenges for SCLC therapies, prevention and early diagnosis for this refractory disease is very important. At the same time, we should reveal more information about the pathogenesis of SCLC and the mechanism of drug resistance. Finally, new treatment strategies should also be taken into considerations, such as repurposing drug, optimising of targets, combination therapy strategies or prognostic biomarkers to enhance therapeutic effects and decrease the adverse events rates in SCLC patients. This article will review the molecular biology characteristics of SCLC and discuss the opportunities and obstacles of the current therapy for SCLC patients.
Collapse
Affiliation(s)
- Lin Yu
- The Clinical Laboratory of Mianyang Central Hospital, Mianyang, China.,Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Qinhuai Lai
- Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Lantu Gou
- Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Jiafu Feng
- The Clinical Laboratory of Mianyang Central Hospital, Mianyang, China
| | - Jinliang Yang
- Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| |
Collapse
|
46
|
The Evolution of Master Protocol Clinical Trial Designs: A Systematic Literature Review. Clin Ther 2020; 42:1330-1360. [DOI: 10.1016/j.clinthera.2020.05.010] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 04/10/2020] [Accepted: 05/11/2020] [Indexed: 02/07/2023]
|
47
|
Nogova L, Mattonet C, Scheffler M, Taubert M, Gardizi M, Sos ML, Michels S, Fischer RN, Limburg M, Abdulla DSY, Persigehl T, Kobe C, Merkelbach-Bruse S, Franklin J, Backes H, Schnell R, Behringer D, Kaminsky B, Eichstaedt M, Stelzer C, Kinzig M, Sörgel F, Tian Y, Junge L, Suleiman AA, Frechen S, Rokitta D, Ouyang D, Fuhr U, Buettner R, Wolf J. Sorafenib and everolimus in patients with advanced solid tumors and KRAS-mutated NSCLC: A phase I trial with early pharmacodynamic FDG-PET assessment. Cancer Med 2020; 9:4991-5007. [PMID: 32436621 PMCID: PMC7367645 DOI: 10.1002/cam4.3131] [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: 12/17/2019] [Revised: 04/14/2020] [Accepted: 04/22/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Treatment of patients with solid tumors and KRAS mutations remains disappointing. One option is the combined inhibition of pathways involved in RAF-MEK-ERK and PI3K-AKT-mTOR. METHODS Patients with relapsed solid tumors were treated with escalating doses of everolimus (E) 2.5-10.0 mg/d in a 14-day run-in phase followed by combination therapy with sorafenib (S) 800 mg/d from day 15. KRAS mutational status was assessed retrospectively in the escalation phase. Extension phase included KRAS-mutated non-small-cell lung cancer (NSCLC) only. Pharmacokinetic analyses were accompanied by pharmacodynamics assessment of E by FDG-PET. Efficacy was assessed by CT scans every 6 weeks of combination. RESULTS Of 31 evaluable patients, 15 had KRAS mutation, 4 patients were negative for KRAS mutation, and the KRAS status remained unknown in 12 patients. Dose-limiting toxicity (DLT) was not reached. The maximum tolerated dose (MTD) was defined as 7.5 mg/d E + 800 mg/d S due to toxicities at previous dose level (10 mg/d E + 800 mg/d S) including leucopenia/thrombopenia III° and pneumonia III° occurring after the DLT interval. The metabolic response rate in FDG-PET was 17% on day 5 and 20% on day 14. No patient reached partial response in CT scan. Median progression free survival (PFS) and overall survival (OS) were 3.25 and 5.85 months, respectively. CONCLUSIONS Treatment of patients with relapsed solid tumors with 7.5 mg/d E and 800 mg/d S is safe and feasible. Early metabolic response in FDG-PET was not confirmed in CT scan several weeks later. The combination of S and E is obviously not sufficient to induce durable responses in patients with KRAS-mutant solid tumors.
Collapse
Affiliation(s)
- Lucia Nogova
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Lung Cancer Group, University of Cologne, Cologne, Germany
| | - Christian Mattonet
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Lung Cancer Group, University of Cologne, Cologne, Germany.,Onkologische Praxis Moers, Moers, Germany
| | - Matthias Scheffler
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Lung Cancer Group, University of Cologne, Cologne, Germany
| | - Max Taubert
- Faculty of Medicine and University Hospital Cologne, Center for Pharmacology, Department I of Pharmacology, University of Cologne, Cologne, Germany
| | - Masyar Gardizi
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Lung Cancer Group, University of Cologne, Cologne, Germany
| | - Martin L Sos
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Lung Cancer Group, University of Cologne, Cologne, Germany
| | - Sebastian Michels
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Lung Cancer Group, University of Cologne, Cologne, Germany
| | - Rieke N Fischer
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Lung Cancer Group, University of Cologne, Cologne, Germany
| | - Meike Limburg
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Lung Cancer Group, University of Cologne, Cologne, Germany
| | - Diana S Y Abdulla
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Lung Cancer Group, University of Cologne, Cologne, Germany
| | - Thorsten Persigehl
- Faculty of Medicine and University Hospital Cologne, Institute for Diagnostics und Intervention Radiology, University of Cologne, Cologne, Germany
| | - Carsten Kobe
- Faculty of Medicine and University Hospital Cologne, Department for Nuclear Medicine, University of Cologne, Cologne, Germany
| | - Sabine Merkelbach-Bruse
- Faculty of Medicine and University Hospital Cologne, Institute for Pathology, University of Cologne, Cologne, Germany
| | - Jeremy Franklin
- Faculty of Medicine, Institute for Medical Statistics and Bioinformatics, University of Cologne, Cologne, Germany
| | - Heiko Backes
- Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Roland Schnell
- Praxis for Medical Oncology and Haematology (PIOH), Frechen, Germany
| | - Dirk Behringer
- Heamatology and Oncology, Augusta Hospital, Bochum, Germany
| | | | | | - Christoph Stelzer
- Institute for Biomedical and Pharmaceutical Research (IBMP), Nürnberg, Germany
| | - Martina Kinzig
- Institute for Biomedical and Pharmaceutical Research (IBMP), Nürnberg, Germany
| | - Fritz Sörgel
- Institute for Biomedical and Pharmaceutical Research (IBMP), Nürnberg, Germany
| | - Yingying Tian
- Faculty of Medicine and University Hospital Cologne, Center for Pharmacology, Department I of Pharmacology, University of Cologne, Cologne, Germany.,Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
| | - Lisa Junge
- Faculty of Medicine and University Hospital Cologne, Center for Pharmacology, Department I of Pharmacology, University of Cologne, Cologne, Germany
| | - Ahmed A Suleiman
- Faculty of Medicine and University Hospital Cologne, Center for Pharmacology, Department I of Pharmacology, University of Cologne, Cologne, Germany
| | - Sebastian Frechen
- Faculty of Medicine and University Hospital Cologne, Center for Pharmacology, Department I of Pharmacology, University of Cologne, Cologne, Germany
| | - Dennis Rokitta
- Faculty of Medicine and University Hospital Cologne, Center for Pharmacology, Department I of Pharmacology, University of Cologne, Cologne, Germany
| | - Dongsheng Ouyang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha, China
| | - Uwe Fuhr
- Faculty of Medicine and University Hospital Cologne, Center for Pharmacology, Department I of Pharmacology, University of Cologne, Cologne, Germany
| | - Reinhard Buettner
- Faculty of Medicine and University Hospital Cologne, Institute for Pathology, University of Cologne, Cologne, Germany
| | - Jürgen Wolf
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Lung Cancer Group, University of Cologne, Cologne, Germany
| |
Collapse
|
48
|
Zhou N, Zhao J, Huang X, Shen H, Li W, Xu Z, Xia Y. The efficacy of afatinib in patients with HER2 mutant non-small cell lung cancer: a meta-analysis. Transl Cancer Res 2020; 9:3634-3642. [PMID: 35117726 PMCID: PMC8797916 DOI: 10.21037/tcr.2020.04.09] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 03/24/2020] [Indexed: 12/30/2022]
Abstract
BACKGROUND Erb-b2 receptor tyrosine kinase 2 (ErbB2/HER2) mutation has been found in approximately 2-4% of non-small cell lung cancer (NSCLC) patients and has been identified as one of carcinogenic mutations. Afatinib, a member of irreversible HER family inhibitor, has been investigated by a number of literatures, yet whose therapeutic efficiency remains uncertain in NSCLC with HER2 mutation. To elucidate the clinical efficacy and safety of afatinib in treating HER2 mutant NSCLC, we integrated and reanalyzed the data from current available studies. METHODS We conducted a systematic literature search for published articles regarding afatinib treating HER2-mutant lung cancer. Eight studies met the inclusion and exclusion criteria. The main outcomes were the objective response rate (ORR) and disease control rate (DCR). RESULTS Ninety-five patients with HER2 mutations were identified from eight studies. The pooled ORR was 21% (95% CI: 11-34%) and the pooled DCR was 66% (95% CI: 57-76%). The patients harboring A775-G776ins YVMA mutation, the most common subtype of HER2 exon 20 mutation, derived greater clinical benefit. Most adverse events were grade 1-2, except a case of fatal acute renal injury, possibly related to afatinib. CONCLUSIONS Afatinib monotherapy demonstrated frustrating anti-tumor activity with tolerable toxicity in HER2 mutant NSCLC. Based on current available data, we do not recommend the regular application of afatinib in NSCLC with HER2 mutations unless the response heterogeneity with specific genomic variant of HER2 mutation was further clarified.
Collapse
Affiliation(s)
- Niya Zhou
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Jie Zhao
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Xiu Huang
- Department of Respiratory and Critical Care Medicine, Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu 322000, China
| | - Hui Shen
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China.,Department of Respiratory and Critical Care Medicine, Huzhou Central Hospital, Huzhou 313000, China
| | - Wen Li
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Zhihao Xu
- Department of Respiratory and Critical Care Medicine, Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu 322000, China
| | - Yang Xia
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China
| |
Collapse
|
49
|
Amaral T, Schulze M, Sinnberg T, Nieser M, Martus P, Battke F, Garbe C, Biskup S, Forschner A. Are Pathogenic Germline Variants in Metastatic Melanoma Associated with Resistance to Combined Immunotherapy? Cancers (Basel) 2020; 12:cancers12051101. [PMID: 32354124 PMCID: PMC7281129 DOI: 10.3390/cancers12051101] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 04/24/2020] [Accepted: 04/27/2020] [Indexed: 01/10/2023] Open
Abstract
Background: Combined immunotherapy has significantly improved survival of patients with advanced melanoma, but there are still patients that do not benefit from it. Early biomarkers that indicate potential resistance would be highly relevant for these patients. Methods: We comprehensively analyzed tumor and blood samples from patients with advanced melanoma, treated with combined immunotherapy and performed descriptive and survival analysis. Results: Fifty-nine patients with a median follow-up of 13 months (inter quartile range (IQR) 11–15) were included. Interestingly, nine patients were found to have pathogenic or likely pathogenic (P/LP) germline variants in one of these genes: BRCA2, POLE, WRN, FANCI, CDKN2A, BAP1, PALB2 and RAD54B. Most of them are involved in DNA repair mechanisms. Patients with P/LP germline variants had a significantly shorter progression-free survival (PFS) and melanoma specific survival (MSS) compared to patients without P/LP germline variants (HR = 2.16; 95% CI: 1.01–4.64; p = 0.048 and HR = 3.21; 95% CI: 1.31–7.87; p = 0.011, respectively). None of the patients with a P/LP germline variant responded to combined immunotherapy. In the multivariate Cox-regression analysis, presence of a P/LP germline variant, S100B and lactate dehydrogenase (LDH) remained independently significant factors for MSS (p = 0.036; p = 0.044 and p = 0.001, respectively). Conclusions: The presence of P/LP germline variants was associated with resistance to combined immunotherapy in our cohort. As genes involved in DNA repair mechanisms are also involved in lymphocyte development and T-cell differentiation, a P/LP germline variant in these genes may preclude an antitumor immune response.
Collapse
Affiliation(s)
- Teresa Amaral
- Center for Dermatooncology, Department of Dermatology, University Hospital Tuebingen, Eberhard Karls University, 72076 Tuebingen, Germany; (T.A.); (T.S.); (C.G.)
- Portuguese Air Force, Health Care Direction, 1649-020 Lisbon, Portugal
| | - Martin Schulze
- Practice for Human Genetics, 72076 Tuebingen, Germany; (M.S.); (M.N.); (S.B.)
| | - Tobias Sinnberg
- Center for Dermatooncology, Department of Dermatology, University Hospital Tuebingen, Eberhard Karls University, 72076 Tuebingen, Germany; (T.A.); (T.S.); (C.G.)
| | - Maike Nieser
- Practice for Human Genetics, 72076 Tuebingen, Germany; (M.S.); (M.N.); (S.B.)
| | - Peter Martus
- Institute for Clinical Epidemiology and applied Biostatistics (IKEaB), Eberhard Karls University, 72076 Tuebingen, Germany;
| | - Florian Battke
- Center for Genomics and Transcriptomics (CeGaT) GmbH, 72076 Tuebingen, Germany;
| | - Claus Garbe
- Center for Dermatooncology, Department of Dermatology, University Hospital Tuebingen, Eberhard Karls University, 72076 Tuebingen, Germany; (T.A.); (T.S.); (C.G.)
| | - Saskia Biskup
- Practice for Human Genetics, 72076 Tuebingen, Germany; (M.S.); (M.N.); (S.B.)
- Center for Genomics and Transcriptomics (CeGaT) GmbH, 72076 Tuebingen, Germany;
| | - Andrea Forschner
- Center for Dermatooncology, Department of Dermatology, University Hospital Tuebingen, Eberhard Karls University, 72076 Tuebingen, Germany; (T.A.); (T.S.); (C.G.)
- Correspondence: ; Tel.: +49-(0)-7071-29 84555; Fax: +49-(0)-7071-29-4599
| |
Collapse
|
50
|
Scheffler M, Holzem A, Kron A, Nogova L, Ihle MA, von Levetzow C, Fassunke J, Wömpner C, Bitter E, Koleczko S, Abdulla DSY, Michels S, Fischer R, Riedel R, Weber JP, Westphal T, Gerigk U, Kern J, Kaminsky B, Randerath W, Kambartel KO, Merkelbach-Bruse S, Büttner R, Wolf J. Co-occurrence of targetable mutations in Non-small cell lung cancer (NSCLC) patients harboring MAP2K1 mutations. Lung Cancer 2020; 144:40-48. [PMID: 32361034 DOI: 10.1016/j.lungcan.2020.04.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 04/16/2020] [Accepted: 04/18/2020] [Indexed: 02/04/2023]
Abstract
BACKGROUND MAP2K1 mutations are rare in non-small cell lung cancer (NSCLC) and considered to be mutually exclusive from known driver mutations. Activation of the MEK1-cascade is considered pivotal in resistance to targeted therapy approaches, and MAP2K1 K57 N mutation could be linked to resistance in preclinical models. We set out this study to detect MAP2K1 mutations and potentially targetable co-mutations using a molecular multiplex approach. METHODS Between 2012 and 2018, we routinely analyzed 14.512 NSCLC patients with two next-generation sequencing (NGS) panels. In a subset of patients, fluorescence in-situ hybridization was performed to detect rearrangements or amplifications. We assessed clinical parameters and co-occurring mutations and compared treatment outcomes of different forms of systemic therapy. RESULTS We identified 66 (0.5%) patients with MAP2K1 mutations. Both adenocarcinoma (n = 62) and squamous cell carcinoma (n = 4) histology. The presence of the mutations was linked to smoking, and transversions were more common than transitions. K57 N was the most frequent MAP2K1 mutation (n = 25). Additional mutations were found in 57 patients (86.4%). Mutations of TP53 were detected in 33 patients, followed by KEAP1 mutations in 28.1%. 24 patients (36.4%) had either MAP2K1-only or a co-occurring aberration considered targetable, including EGFR mutations, a BRAF V600E mutation and ROS1 rearrangements. Outcome analyses revealed a trend toward benefit from pemetrexed treatment. CONCLUSION Our analysis shows that MAP2K1-mutated NSCLC patients might frequently present with potentially targetable aberrations. Their role in providing resistance in these subtypes and the possible therapeutic opportunities justify further analyses of this rare NSCLC subgroup.
Collapse
Affiliation(s)
- Matthias Scheffler
- University Hospital of Cologne, Lung Cancer Group Cologne, Department I of Internal Medicine, Cologne, Germany
| | - Alessandra Holzem
- University Hospital of Cologne, Lung Cancer Group Cologne, Department I of Internal Medicine, Cologne, Germany
| | - Anna Kron
- University Hospital of Cologne, Lung Cancer Group Cologne, Department I of Internal Medicine, Cologne, Germany
| | - Lucia Nogova
- University Hospital of Cologne, Lung Cancer Group Cologne, Department I of Internal Medicine, Cologne, Germany
| | - Michaela A Ihle
- University of Cologne, Cologne Institute of Pathology, Cologne, Germany
| | - Cornelia von Levetzow
- University Hospital of Cologne, Lung Cancer Group Cologne, Department I of Internal Medicine, Cologne, Germany
| | - Jana Fassunke
- University of Cologne, Cologne Institute of Pathology, Cologne, Germany
| | - Claudia Wömpner
- University Hospital of Cologne, Lung Cancer Group Cologne, Department I of Internal Medicine, Cologne, Germany
| | - Elisabeth Bitter
- University Hospital of Cologne, Lung Cancer Group Cologne, Department I of Internal Medicine, Cologne, Germany
| | - Sophia Koleczko
- University Hospital of Cologne, Lung Cancer Group Cologne, Department I of Internal Medicine, Cologne, Germany
| | - Diana S Y Abdulla
- University Hospital of Cologne, Lung Cancer Group Cologne, Department I of Internal Medicine, Cologne, Germany
| | - Sebastian Michels
- University Hospital of Cologne, Lung Cancer Group Cologne, Department I of Internal Medicine, Cologne, Germany
| | - Rieke Fischer
- University Hospital of Cologne, Lung Cancer Group Cologne, Department I of Internal Medicine, Cologne, Germany
| | - Richard Riedel
- University Hospital of Cologne, Lung Cancer Group Cologne, Department I of Internal Medicine, Cologne, Germany
| | - Jan-Philipp Weber
- University Hospital of Cologne, Lung Cancer Group Cologne, Department I of Internal Medicine, Cologne, Germany
| | - Theresa Westphal
- University Hospital of Cologne, Lung Cancer Group Cologne, Department I of Internal Medicine, Cologne, Germany
| | - Ulrich Gerigk
- GFO Clinics Bonn, Marien-Hospital Bonn, Bonn, Germany
| | - Jens Kern
- KWM Missio Clinic, Würzburg, Germany
| | - Britta Kaminsky
- Bethanien Hospital Solingen, Clinic for Pulmonology and Allergology, Solingen, Germany
| | - Winfried Randerath
- Bethanien Hospital Solingen, Clinic for Pulmonology and Allergology, Solingen, Germany
| | | | | | - Reinhard Büttner
- University of Cologne, Cologne Institute of Pathology, Cologne, Germany
| | - Jürgen Wolf
- University Hospital of Cologne, Lung Cancer Group Cologne, Department I of Internal Medicine, Cologne, Germany.
| |
Collapse
|