1
|
Young RJ, Kirkham A, Savage J, Gaskell C, Johnson S, Dockrell DH, Bower M, Westwell S, Bowman C, Leahy M, Woll P, Billingham L. Selumetinib in Combination with Anti Retroviral Therapy in HIV-associated Kaposi sarcoma (SCART): an open-label, multicentre, phase I/II trial. BMC Cancer 2025; 25:505. [PMID: 40108492 PMCID: PMC11921695 DOI: 10.1186/s12885-025-13890-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 03/10/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND Kaposi sarcoma (KS) is the commonest HIV-associated malignancy. It is caused by co-infection with Kaposi sarcoma herpesvirus (KSHV), which upregulates the MAPK pathway. The aim of the SCART trial was to identify a safe dose for the MEK inhibitor selumetinib in combination with antiretroviral therapy (ART) and to establish evidence of the combination's efficacy. METHODS SCART was a prospective, single arm, open-label, multi-centre, phase I/II trial, recruiting from four UK centres. Eligible patients were HIV positive, established on an ART regimen ≥ 3 months, had HIV viral load ≤ 200/ml, and had histologically confirmed KS with progressive disease. Phase I primary outcomes were occurrence of dose limiting toxicity (DLT) to determine the maximum tolerated dose/recommended phase II dose (RP2D), and pharmacokinetic assessments of selumetinib and N-desmethyl metabolite. Phase II primary outcome was occurrence of objective response (OR) as defined by AIDS Clinical Trials Group (ACTG) criteria. RESULTS Between 15-Jun-2012 and 25-Sep-2018, 19 patients were recruited; three did not start treatment and were not included in the final analysis. Ten eligible patients were treated in phase I and an additional six in phase II. There was one DLT at the 75 mg bd dose, which was deemed to be the RP2D. Of those patients receiving the RP2D (six within phase I, six within phase II), one achieved a partial response (OR 8.3%, 90% confidence interval: 0.4, 33.9). Further to the DLT, two serious adverse reactions, one unrelated serious adverse event (AE), and six non-serious grade 3 AEs were reported, together with 360 AEs graded 1 or 2. No detrimental impact on ART drug levels or HIV viral load were observed, with improvements in CD4 count and evidence of response in Angiopoietin-2 demonstrated. CONCLUSIONS SCART was closed early due to slow recruitment, partly due to the rarity of KS because of improvements in HIV care, but also due to patients' concerns about experiencing non-serious toxicity additional to those from ART. Although we cannot recommend the use of 75 mg bd selumetinib with ART in patients with HIV-associated KS, studies exploring selumetinib in combination with other agents including anti-angiogenic agents and/or immune checkpoint inhibitors are warranted. TRIAL REGISTRATION ISRCTN24921472.
Collapse
Affiliation(s)
- Robin J Young
- University of Sheffield, Sheffield, UK.
- Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK.
| | - Amanda Kirkham
- Cancer Research UK Clinical Trials Unit (CRCTU), University of Birmingham, Birmingham, UK
| | - Joshua Savage
- Cancer Research UK Clinical Trials Unit (CRCTU), University of Birmingham, Birmingham, UK
| | - Charlotte Gaskell
- Cancer Research UK Clinical Trials Unit (CRCTU), University of Birmingham, Birmingham, UK
| | - Sarah Johnson
- Cancer Research UK Clinical Trials Unit (CRCTU), University of Birmingham, Birmingham, UK
| | - David H Dockrell
- Institute for Regeneration and Repair, Centre for Inflammation Research, University of Edinburgh, Edinburgh, UK
| | - Mark Bower
- National Centre for HIV Malignancy, Chelsea & Westminster Hospital, London, UK
| | - Sarah Westwell
- Brighton and Sussex University Hospitals NHS Trust, Brighton, UK
| | - Christine Bowman
- Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | | | - Penella Woll
- University of Sheffield, Sheffield, UK
- Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Lucinda Billingham
- Cancer Research UK Clinical Trials Unit (CRCTU), University of Birmingham, Birmingham, UK
| |
Collapse
|
2
|
Majernikova SM. Risk and safety profile in checkpoint inhibitors on non-small-cel lung cancer: A systematic review. Hum Vaccin Immunother 2024; 20:2365771. [PMID: 38932682 PMCID: PMC11212564 DOI: 10.1080/21645515.2024.2365771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 06/05/2024] [Indexed: 06/28/2024] Open
Abstract
Treating non-small-cell lung cancer (NSCLC) has gained increased importance in recent years due to the high mortality rate and dismal five-year survival rate. Immune checkpoint inhibitors (ICI) are a promising approach with exceptional outcomes in NSCLC thanks to the antigenic nature of cells. Conversely, immune system over-stimulation with ICI is a double-edged sword that can lead to various negative effects ranging from mild to life-threatening. This review explores current breakthroughs in nanoparticle-based ICI and their limitations. The PubMed, Scopus and Web of Science were examined for relevant publications. Thirty-eight trials (N = 16,781) were included in the analyses. The mixed effects analyses on quantifying the treatment effect contributed significantly to the subgroups within studies for ICI treatment effect. Models confirmed ICI's higher impact on treatment effectivity and the decrease in respondents' mortality compared to conventional treatment regiments. ICI might be used as first-line therapy due to their proven effectiveness and safety profile.
Collapse
Affiliation(s)
- Sara Maria Majernikova
- Department for Continuing Education, The University of Oxford, Oxford, UK
- Department of Neuroscience, Physiology & Pharmacology, Division of Biosciences, Faculty of Life Sciences, University College London, London, UK
| |
Collapse
|
3
|
Duan XP, Qin BD, Jiao XD, Liu K, Wang Z, Zang YS. New clinical trial design in precision medicine: discovery, development and direction. Signal Transduct Target Ther 2024; 9:57. [PMID: 38438349 PMCID: PMC10912713 DOI: 10.1038/s41392-024-01760-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/25/2024] [Accepted: 01/29/2024] [Indexed: 03/06/2024] Open
Abstract
In the era of precision medicine, it has been increasingly recognized that individuals with a certain disease are complex and different from each other. Due to the underestimation of the significant heterogeneity across participants in traditional "one-size-fits-all" trials, patient-centered trials that could provide optimal therapy customization to individuals with specific biomarkers were developed including the basket, umbrella, and platform trial designs under the master protocol framework. In recent years, the successive FDA approval of indications based on biomarker-guided master protocol designs has demonstrated that these new clinical trials are ushering in tremendous opportunities. Despite the rapid increase in the number of basket, umbrella, and platform trials, the current clinical and research understanding of these new trial designs, as compared with traditional trial designs, remains limited. The majority of the research focuses on methodologies, and there is a lack of in-depth insight concerning the underlying biological logic of these new clinical trial designs. Therefore, we provide this comprehensive review of the discovery and development of basket, umbrella, and platform trials and their underlying logic from the perspective of precision medicine. Meanwhile, we discuss future directions on the potential development of these new clinical design in view of the "Precision Pro", "Dynamic Precision", and "Intelligent Precision". This review would assist trial-related researchers to enhance the innovation and feasibility of clinical trial designs by expounding the underlying logic, which be essential to accelerate the progression of precision medicine.
Collapse
Affiliation(s)
- Xiao-Peng Duan
- Department of Medical Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Bao-Dong Qin
- Department of Medical Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Xiao-Dong Jiao
- Department of Medical Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Ke Liu
- Department of Medical Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Zhan Wang
- Department of Medical Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Yuan-Sheng Zang
- Department of Medical Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China.
| |
Collapse
|
4
|
Meira DD, de Castro e Caetano MC, Casotti MC, Zetum ASS, Gonçalves AFM, Moreira AR, de Oliveira AH, Pesente F, Santana GM, de Almeida Duque D, Pereira GSC, de Castro GDSC, Pavan IP, Chagas JPS, Bourguignon JHB, de Oliveira JR, Barbosa KRM, Altoé LSC, Louro LS, Merigueti LP, Alves LNR, Machado MRR, Roque MLRO, Prates PS, de Paula Segáua SH, dos Santos Uchiya T, Louro TES, Daleprane VE, Guaitolini YM, Vicente CR, dos Reis Trabach RS, de Araújo BC, dos Santos EDVW, de Paula F, Lopes TJS, de Carvalho EF, Louro ID. Prognostic Factors and Markers in Non-Small Cell Lung Cancer: Recent Progress and Future Challenges. Genes (Basel) 2023; 14:1906. [PMID: 37895255 PMCID: PMC10606762 DOI: 10.3390/genes14101906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 09/29/2023] [Accepted: 10/01/2023] [Indexed: 10/29/2023] Open
Abstract
Lung cancer is a highly aggressive neoplasm and, despite the development of recent therapies, tumor progression and recurrence following the initial response remains unsolved. Several questions remain unanswered about non-small cell lung cancer (NSCLC): (1) Which patients will actually benefit from therapy? (2) What are the predictive factors of response to MAbs and TKIs? (3) What are the best combination strategies with conventional treatments or new antineoplastic drugs? To answer these questions, an integrative literature review was carried out, searching articles in PUBMED, NCBI-PMC, Google Academic, and others. Here, we will examine the molecular genetics of lung cancer, emphasizing NSCLC, and delineate the primary categories of inhibitors based on their molecular targets, alongside the main treatment alternatives depending on the type of acquired resistance. We highlighted new therapies based on epigenetic information and a single-cell approach as a potential source of new biomarkers. The current and future of NSCLC management hinges upon genotyping correct prognostic markers, as well as on the evolution of precision medicine, which guarantees a tailored drug combination with precise targeting.
Collapse
Affiliation(s)
- Débora Dummer Meira
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Maria Clara de Castro e Caetano
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Matheus Correia Casotti
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Aléxia Stefani Siqueira Zetum
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - André Felipe Monteiro Gonçalves
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - André Rodrigues Moreira
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Augusto Henrique de Oliveira
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Fellipe Pesente
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Gabriel Mendonça Santana
- Centro de Ciências da Saúde, Curso de Medicina, Universidade Federal do Espírito Santo (UFES), Vitória 29090-040, Brazil
| | - Daniel de Almeida Duque
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Gierleson Santos Cangussu Pereira
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Giulia de Souza Cupertino de Castro
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Isabele Pagani Pavan
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - João Pedro Sarcinelli Chagas
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - José Henrique Borges Bourguignon
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Juliana Ribeiro de Oliveira
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Karen Ruth Michio Barbosa
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Lorena Souza Castro Altoé
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Luana Santos Louro
- Centro de Ciências da Saúde, Curso de Medicina, Universidade Federal do Espírito Santo (UFES), Vitória 29090-040, Brazil
| | - Luiza Poppe Merigueti
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Lyvia Neves Rebello Alves
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Marlon Ramos Rosado Machado
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Maria Luísa Rodrigues Oliveira Roque
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Pedro Santana Prates
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Sayuri Honorio de Paula Segáua
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Taissa dos Santos Uchiya
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Thomas Erik Santos Louro
- Escola Superior de Ciências da Santa Casa de Misericórdia de Vitória (EMESCAM), Curso de Medicina, Vitória 29027-502, Brazil
| | - Vinicius Eduardo Daleprane
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Yasmin Moreto Guaitolini
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Creuza Rachel Vicente
- Departamento de Medicina Social, Universidade Federal do Espírito Santo, Vitória 29090-040, Brazil
| | - Raquel Silva dos Reis Trabach
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Bruno Cancian de Araújo
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Eldamária de Vargas Wolfgramm dos Santos
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Flávia de Paula
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| | - Tiago José S. Lopes
- Department of Reproductive Biology, National Center for Child Health and Development Research Institute, Tokyo 157-8535, Japan
| | - Elizeu Fagundes de Carvalho
- Instituto de Biologia Roberto Alcântara Gomes (IBRAG), Universidade do Estado do Rio de Janeiro (UERJ), Rio de Janeiro 20551-030, Brazil
| | - Iúri Drumond Louro
- Núcleo de Genética Humana e Molecular, Centro de Ciências Humanas e Naturais, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, Brazil (M.C.C.)
| |
Collapse
|
5
|
Daniel Humberto Pozza, Ramon Bezerra Andrade de Mello. Treatment Sequencing Strategies in Lung Cancer. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2022; 25:323-336. [PMID: 35599008 PMCID: PMC9127753 DOI: 10.3779/j.issn.1009-3419.2022.104.01] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 04/22/2022] [Accepted: 04/23/2022] [Indexed: 12/04/2022]
Abstract
BACKGROUND The advances in the lung cancer screening methods and therapeutics, together with awareness towards deleterious habits, such as smoking, is increasing the overall survival with better quality of life for the patients. However, lung cancer is still one of the most common and fatal neoplasm with a high incidence and consequently burden to public health worldwide. Thus, based on guidelines and recent phases II and III clinical trials studies, this manuscript summarizes the current treatment sequencing strategies in lung cancer. METHODS A comprehensive search of related articles was performed focused on phases II and III clinical trials studies. RESULTS The lung cancer management should take into consideration the tumor characteristics, histology, molecular pathology and be discussed in a multidisciplinary team. Lung cancer treatment options comprises surgery whenever possible, radiotherapy associate with/or chemotherapy and immunotherapy as monotherapy, or combined with chemotherapy and best palliative care. CONCLUSIONS The screening predictability in more patients, smoking reduction, early diagnosis, better disease understanding and individualized, more effective and tolerable therapeutics are related to an increasing in overall survival and quality of life. In the near future improvement of personalized therapy in precision medicine is expected, enhancing new predictive biomarkers, optimal doses and optimal treatment sequencing as well as anti-cancer vaccines development.
Collapse
Affiliation(s)
- Daniel Humberto Pozza
- Department of Biomedicine, Faculty of Medicine and i3s, University of Porto, 4200-319 Porto, Portugal
| | - Ramon Bezerra Andrade de Mello
- Discipline of Medical Oncology, Post-graduation Program in Medicine, Nine of July University (UNINOVE), São Paulo, Brazil./Nine of July Hospital, São Paulo, Brazil
| |
Collapse
|
6
|
Watanabe M, Boku S, Kobayashi K, Kurumida Y, Sukeno M, Masuda M, Mizushima K, Kato C, Iizumi Y, Hirota K, Naito Y, Mutoh M, Kameda T, Sakai T. A chemoproteoinformatics approach demonstrates that aspirin increases sensitivity to MEK inhibition by directly binding to RPS5. PNAS NEXUS 2022; 1:pgac059. [PMID: 36713317 PMCID: PMC9802315 DOI: 10.1093/pnasnexus/pgac059] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 05/11/2022] [Indexed: 02/01/2023]
Abstract
MEK inhibitors are among the most successful molecularly targeted agents used as cancer therapeutics. However, to treat cancer more efficiently, resistance to MEK inhibitor-induced cell death must be overcome. Although previous genetic approaches based on comprehensive gene expression analysis or RNAi libraries led to the discovery of factors involved in intrinsic resistance to MEK inhibitors, a feasible combined treatment with the MEK inhibitor has not yet been developed. Here, we show that a chemoproteoinformatics approach identifies ligands overcoming the resistance to cell death induced by MEK inhibition as well as the target molecule conferring this resistance. First, we used natural products, perillyl alcohol and sesaminol, which induced cell death in combination with the MEK inhibitor trametinib, as chemical probes, and identified ribosomal protein S5 (RPS5) as their common target protein. Consistently, trametinib induced cell death in RPS5-depleted cancer cells via upregulation of the apoptotic proteins BIM and PUMA. Using molecular docking and molecular dynamics (MD) simulations, we then screened FDA- and EMA-approved drugs for RPS5-binding ligands and found that acetylsalicylic acid (ASA, also known as aspirin) directly bound to RPS5, resulting in upregulation of BIM and PUMA and induction of cell death in combination with trametinib. Our chemoproteoinformatics approach demonstrates that RPS5 confers resistance to MEK inhibitor-induced cell death, and that aspirin could be repurposed to sensitize cells to MEK inhibition by binding to RPS5.
Collapse
Affiliation(s)
| | - Shogen Boku
- Department of Molecular-Targeting Prevention, Kyoto Prefectural University of Medicine, 602-8566 Kyoto, Japan,Cancer Treatment Center, Kansai Medical University Hospital, 573-1010 Osaka, Japan
| | - Kaito Kobayashi
- Artificial Intelligence Research Center, National Institute of Advanced Industrial Science and Technology (AIST), 135-0064 Tokyo, Japan
| | - Yoichi Kurumida
- Artificial Intelligence Research Center, National Institute of Advanced Industrial Science and Technology (AIST), 135-0064 Tokyo, Japan
| | - Mamiko Sukeno
- Department of Drug Discovery Medicine, Kyoto Prefectural University of Medicine, 602-8566 Kyoto, Japan
| | - Mitsuharu Masuda
- Department of Molecular-Targeting Prevention, Kyoto Prefectural University of Medicine, 602-8566 Kyoto, Japan
| | - Katsura Mizushima
- Department of Human Immunology and Nutrition Science, Kyoto Prefectural University of Medicine, 602-8566 Kyoto, Japan
| | - Chikage Kato
- Department of Endocrine and Breast Surgery, Kyoto Prefectural University of Medicine, 602-8566 Kyoto, Japan
| | - Yosuke Iizumi
- Department of Molecular-Targeting Prevention, Kyoto Prefectural University of Medicine, 602-8566 Kyoto, Japan
| | - Kiichi Hirota
- Department of Human Stress Response Science, Institute of Biomedical Science, Kansai Medical University, 573-1010 Osaka, Japan
| | - Yuji Naito
- Department of Human Immunology and Nutrition Science, Kyoto Prefectural University of Medicine, 602-8566 Kyoto, Japan
| | - Michihiro Mutoh
- Department of Molecular-Targeting Prevention, Kyoto Prefectural University of Medicine, 602-8566 Kyoto, Japan
| | - Tomoshi Kameda
- Artificial Intelligence Research Center, National Institute of Advanced Industrial Science and Technology (AIST), 135-0064 Tokyo, Japan
| | - Toshiyuki Sakai
- Department of Drug Discovery Medicine, Kyoto Prefectural University of Medicine, 602-8566 Kyoto, Japan
| |
Collapse
|
7
|
Selumetinib: a selective MEK1 inhibitor for solid tumor treatment. Clin Exp Med 2022; 23:229-244. [PMID: 35171389 DOI: 10.1007/s10238-021-00783-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 12/07/2021] [Indexed: 12/24/2022]
Abstract
Cancer incidence is rapidly growing. Solid tumors are responsible for a majority of cancers. Recently, molecular-targeted agents have played a significant role in cancer treatment. Ras-Raf-MEK-ERK signaling pathway, is a substantial element in the survival, propagation, and drug resistance of human cancers. MEK is a specific part of the so-called cascade, and ERK proteins are its sole target. Furthermore, their downstream position in the Ras-ERK cascade, is noteworthy to direct their function in patients with upstream mutated genes. MEK1 mutations are responsible for initiating several solid tumors. Selumetinib (AZD6244) is a second-generation, selective, potent, and non-ATP competitive allosteric MEK1 inhibitor. The efficacy of selumetinib in various solid tumors such as colorectal cancer, lung cancer, neurofibroma, and melanoma is investigated. The present paper provides an overview of the MAPK cascade, the role of selumetinib as a MEK1/2 inhibitor, and the related findings of clinical trials for solid tumor treatment.
Collapse
|
8
|
Prognostic Gene Signature for Squamous Cell Carcinoma with a Higher Risk for Treatment Failure and Accelerated MEK-ERK Pathway Activity. Cancers (Basel) 2021; 13:cancers13205182. [PMID: 34680330 PMCID: PMC8534038 DOI: 10.3390/cancers13205182] [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: 08/30/2021] [Revised: 10/07/2021] [Accepted: 10/09/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Squamous cell carcinoma (SCC) is the most prevalent type of human cancer worldwide and represents the majority of head and neck tumors. As SCC from aerodigestive or genitourinary tracts share not only common etiology and histological features but also molecular patterns, the major objectives of this study were the establishment of a pan-SCC-related prognostic gene signature by an integrative analysis of multi-omics data and the elucidation of underlying oncogenic pathway activities as potential vulnerabilities for a more efficient and less toxic therapy. Our approach delivers a reliable molecular classifier to identify HNSCC and other SCC patients at higher risk for treatment failure with tumors characterized by a more prominent MAPK activity, who might benefit from a targeted treatment with MEK inhibitors. Abstract Squamous cell carcinoma (SCC) is the most prevalent histological type of human cancer, including head and neck squamous cell carcinoma (HNSCC). However, reliable prognostic gene signatures for SCC and underlying genetic and/or epigenetic principles are still unclear. We identified 37 prognostic candidate genes by best cutoff computation based on survival in a pan-SCC cohort (n = 1334) of The Cancer Genome Atlas (TCGA), whose expression stratified not only the pan-SCC cohort but also independent HNSCC validation cohorts into three distinct prognostic subgroups. The most relevant prognostic genes were prioritized by a Least Absolute Shrinkage and Selection Operator Cox regression model and were used to identify subgroups with high or low risks for unfavorable survival. An integrative analysis of multi-omics data identified FN1, SEMA3A, CDH2, FBN1, COL5A1, and ADAM12 as key nodes in a regulatory network related to the prognostic phenotype. An in-silico drug screen predicted two MEK inhibitors (Trametinib and Selumetinib) as effective compounds for high-risk SCC based on the Cancer Cell Line Encyclopedia, which is supported by a higher p-MEK1/2 immunohistochemical staining of high-risk HNSCC. In conclusion, our data identified a molecular classifier for high-risk HNSCC as well as other SCC patients, who might benefit from treatment with MEK inhibitors.
Collapse
|
9
|
Baldelli E, El Gazzah E, Moran JC, Hodge KA, Manojlovic Z, Bassiouni R, Carpten JD, Ludovini V, Baglivo S, Crinò L, Bianconi F, Dong T, Loffredo J, Petricoin EF, Pierobon M. Wild-Type KRAS Allele Effects on Druggable Targets in KRAS Mutant Lung Adenocarcinomas. Genes (Basel) 2021; 12:genes12091402. [PMID: 34573384 PMCID: PMC8467269 DOI: 10.3390/genes12091402] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/04/2021] [Accepted: 09/08/2021] [Indexed: 11/16/2022] Open
Abstract
KRAS mutations are one of the most common oncogenic drivers in non-small cell lung cancer (NSCLC) and in lung adenocarcinomas in particular. Development of therapeutics targeting KRAS has been incredibly challenging, prompting indirect inhibition of downstream targets such as MEK and ERK. Such inhibitors, unfortunately, come with limited clinical efficacy, and therefore the demand for developing novel therapeutic strategies remains an urgent need for these patients. Exploring the influence of wild-type (WT) KRAS on druggable targets can uncover new vulnerabilities for the treatment of KRAS mutant lung adenocarcinomas. Using commercially available KRAS mutant lung adenocarcinoma cell lines, we explored the influence of WT KRAS on signaling networks and druggable targets. Expression and/or activation of 183 signaling proteins, most of which are targets of FDA-approved drugs, were captured by reverse-phase protein microarray (RPPA). Selected findings were validated on a cohort of 23 surgical biospecimens using the RPPA. Kinase-driven signatures associated with the presence of the KRAS WT allele were detected along the MAPK and AKT/mTOR signaling pathway and alterations of cell cycle regulators. FoxM1 emerged as a potential vulnerability of tumors retaining the KRAS WT allele both in cell lines and in the clinical samples. Our findings suggest that loss of WT KRAS impacts on signaling events and druggable targets in KRAS mutant lung adenocarcinomas.
Collapse
Affiliation(s)
- Elisa Baldelli
- Center for Applied Proteomics & Molecular Medicine, George Mason University, Manassas, VA 20110, USA; (E.B.); (E.E.G.); (J.C.M.); (K.A.H.); (T.D.); (J.L.); (E.F.P.)
| | - Emna El Gazzah
- Center for Applied Proteomics & Molecular Medicine, George Mason University, Manassas, VA 20110, USA; (E.B.); (E.E.G.); (J.C.M.); (K.A.H.); (T.D.); (J.L.); (E.F.P.)
- School of Systems Biology, George Mason University, Manassas, VA 20110, USA
| | - John Conor Moran
- Center for Applied Proteomics & Molecular Medicine, George Mason University, Manassas, VA 20110, USA; (E.B.); (E.E.G.); (J.C.M.); (K.A.H.); (T.D.); (J.L.); (E.F.P.)
| | - Kimberley A. Hodge
- Center for Applied Proteomics & Molecular Medicine, George Mason University, Manassas, VA 20110, USA; (E.B.); (E.E.G.); (J.C.M.); (K.A.H.); (T.D.); (J.L.); (E.F.P.)
| | - Zarko Manojlovic
- Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; (Z.M.); (R.B.); (J.D.C.)
| | - Rania Bassiouni
- Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; (Z.M.); (R.B.); (J.D.C.)
| | - John D. Carpten
- Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; (Z.M.); (R.B.); (J.D.C.)
| | - Vienna Ludovini
- Division of Medical Oncology, S. Maria della Misericordia Hospital, 06156 Perugia, Italy; (V.L.); (S.B.)
| | - Sara Baglivo
- Division of Medical Oncology, S. Maria della Misericordia Hospital, 06156 Perugia, Italy; (V.L.); (S.B.)
| | - Lucio Crinò
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy;
| | | | - Ting Dong
- Center for Applied Proteomics & Molecular Medicine, George Mason University, Manassas, VA 20110, USA; (E.B.); (E.E.G.); (J.C.M.); (K.A.H.); (T.D.); (J.L.); (E.F.P.)
| | - Jeremy Loffredo
- Center for Applied Proteomics & Molecular Medicine, George Mason University, Manassas, VA 20110, USA; (E.B.); (E.E.G.); (J.C.M.); (K.A.H.); (T.D.); (J.L.); (E.F.P.)
| | - Emanuel F. Petricoin
- Center for Applied Proteomics & Molecular Medicine, George Mason University, Manassas, VA 20110, USA; (E.B.); (E.E.G.); (J.C.M.); (K.A.H.); (T.D.); (J.L.); (E.F.P.)
| | - Mariaelena Pierobon
- Center for Applied Proteomics & Molecular Medicine, George Mason University, Manassas, VA 20110, USA; (E.B.); (E.E.G.); (J.C.M.); (K.A.H.); (T.D.); (J.L.); (E.F.P.)
- School of Systems Biology, George Mason University, Manassas, VA 20110, USA
- Correspondence: ; Tel.: +1-703-993-9839
| |
Collapse
|
10
|
Coley AB, Ward A, Keeton AB, Chen X, Maxuitenko Y, Prakash A, Li F, Foote JB, Buchsbaum DJ, Piazza GA. Pan-RAS inhibitors: Hitting multiple RAS isozymes with one stone. Adv Cancer Res 2021; 153:131-168. [PMID: 35101229 DOI: 10.1016/bs.acr.2021.07.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Mutations in the three RAS oncogenes are present in approximately 30% of all human cancers that drive tumor growth and metastasis by aberrant activation of RAS-mediated signaling. Despite the well-established role of RAS in tumorigenesis, past efforts to develop small molecule inhibitors have failed for various reasons leading many to consider RAS as "undruggable." Advances over the past decade with KRAS(G12C) mutation-specific inhibitors have culminated in the first FDA-approved RAS drug, sotorasib. However, the patient population that stands to benefit from KRAS(G12C) inhibitors is inherently limited to those patients harboring KRAS(G12C) mutations. Additionally, both intrinsic and acquired mechanisms of resistance have been reported that indicate allele-specificity may afford disadvantages. For example, the compensatory activation of uninhibited wild-type (WT) NRAS and HRAS isozymes can rescue cancer cells harboring KRAS(G12C) mutations from allele-specific inhibition or the occurrence of other mutations in KRAS. It is therefore prudent to consider alternative drug discovery strategies that may overcome these potential limitations. One such approach is pan-RAS inhibition, whereby all RAS isozymes co-expressed in the tumor cell population are targeted by a single inhibitor to block constitutively activated RAS regardless of the underlying mutation. This chapter provides a review of past and ongoing strategies to develop pan-RAS inhibitors in detail and seeks to outline the trajectory of this promising strategy of RAS inhibition.
Collapse
Affiliation(s)
- Alexander B Coley
- Department of Pharmacology, University of South Alabama, Mobile, AL, United States; Mitchell Cancer Institute, Mobile, AL, United States
| | - Antonio Ward
- Department of Pharmacology, University of South Alabama, Mobile, AL, United States; Mitchell Cancer Institute, Mobile, AL, United States
| | - Adam B Keeton
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, United States
| | - Xi Chen
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, United States
| | - Yulia Maxuitenko
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, United States
| | - Aishwarya Prakash
- Mitchell Cancer Institute, Mobile, AL, United States; Department of Biochemistry & Molecular Biology, University of South Alabama, Mobile, AL, United States
| | - Feng Li
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, United States
| | - Jeremy B Foote
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Donald J Buchsbaum
- Department of Radiation Oncology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Gary A Piazza
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, United States.
| |
Collapse
|
11
|
Solares I, Viñal D, Morales-Conejo M, Rodriguez-Salas N, Feliu J. Novel molecular targeted therapies for patients with neurofibromatosis type 1 with inoperable plexiform neurofibromas: a comprehensive review. ESMO Open 2021; 6:100223. [PMID: 34388689 PMCID: PMC8363824 DOI: 10.1016/j.esmoop.2021.100223] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 06/29/2021] [Accepted: 06/30/2021] [Indexed: 11/30/2022] Open
Abstract
Neurofibromatosis type 1 (NF1) is a genetic disorder that carries a higher risk of tumor development. Plexiform neurofibromas (PNs) are present in 50% of NF1 and cause significant morbidity when surgery is not feasible. Systemic therapies had not succeeded to reduce PN tumor volume until 2016 when the first trial with an MAPK/extracellular-signal-regulated kinase (MEK) inhibitor was published. We performed a systematic research on novel targeted therapies for patients with NF1 and PNs in PubMed, EMBASE, and conference abstracts with the last update in February 2021. Since 2016, seven trials have reported positive results with MEK inhibitors and other molecular targeted therapies (cabozantinib). Selumetinib has shown an overall response rate of 68% in children with NF1 and symptomatic inoperable PNs, and was associated with pain improvement and a manageable adverse events profile. This led to Food and Drug Administration (FDA) approval of selumetinib in May 2020. Recently, cabozantinib and mirdametinib have also proven their efficacy in adult population. Other MEK inhibitors such as trametinib and binimetinib have also communicated promising preliminary results. Ongoing trials in different populations and with intermittent dosing strategies are underway.
Collapse
Affiliation(s)
- I Solares
- Department of Internal Medicine, Reference Center for Inherited Metabolic Disease - MetabERN, University Hospital 12 de Octubre, UCM Madrid, Madrid, Spain
| | - D Viñal
- Department of Medical Oncology, Hospital Universitario La Paz, Madrid, Spain.
| | - M Morales-Conejo
- Department of Internal Medicine, Reference Center for Inherited Metabolic Disease - MetabERN, University Hospital 12 de Octubre, UCM Madrid, Madrid, Spain; Grupo de Enfermedades Mitocondriales y Neuromusculares, Instituto de Investigación Hospital 12 de Octubre (i+12), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | - N Rodriguez-Salas
- Department of Medical Oncology, Hospital Universitario La Paz, Madrid, Spain; Translational Oncology Group, IdiPAZ, Madrid, Spain; Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain; CIBERONC, Madrid, Spain
| | - J Feliu
- Department of Medical Oncology, Hospital Universitario La Paz, Madrid, Spain; Translational Oncology Group, IdiPAZ, Madrid, Spain; Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain; CIBERONC, Madrid, Spain
| |
Collapse
|
12
|
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: 197] [Impact Index Per Article: 49.3] [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
|
13
|
Marin E, Reyes R, Arcocha A, Viñolas N, Mezquita L, Gonzalvo E, Saez de Gordoa K, Jares P, Reguart N, Teixido C. Prospective Evaluation of Single Nucleotide Variants by Two Different Technologies in Paraffin Samples of Advanced Non-Small Cell Lung Cancer Patients. Diagnostics (Basel) 2020; 10:E902. [PMID: 33153192 PMCID: PMC7693424 DOI: 10.3390/diagnostics10110902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 10/30/2020] [Accepted: 11/02/2020] [Indexed: 11/17/2022] Open
Abstract
Targeted therapies are a new paradigm in lung cancer management. Next-generation sequencing (NGS) techniques have allowed for simultaneous testing of several genes in a rapid and efficient manner; however, there are other molecular diagnostic tools such as the nCounter® Vantage 3D single nucleotide variants (SNVs) solid tumour panel which also offer important benefits regarding sample input and time-to-response, making them very attractive for daily clinical use. This study aimed to test the performance of the Vantage panel in the routine workup of advanced non-squamous non-small cell lung cancer (NSCLC) patients and to validate and compare its outputs with the Oncomine Solid Tumor (OST) panel DNA kit, the standard technique in our institution. Two parallel multiplexed approaches were performed based on DNA NGS and direct digital detection of DNA with nCounter® technology to evaluate SNVs. A total of 42 advanced non-squamous NSCLC patients were prospectively included in the study. Overall, 95% of samples were successfully characterized by both technologies. The Vantage panel accounted for a sensitivity of 95% and a specificity of 82%. In terms of predictive values, the probability of truly presenting the SNV variant when it is detected by the nCounter panel was 82%, whereas the probability of not presenting the SNV variant when it is not detected by the platform was 95%. Finally, Cohen's Kappa coefficient was 0.76, indicating a substantial correlation grade between OST and Vantage panels. Our results make nCounter an analytically sensitive, practical and cost-effective tool.
Collapse
Affiliation(s)
- Elba Marin
- Division of Medical Oncology, Hospital Clínic, 08036 Barcelona, Spain; (E.M.); (R.R.); (A.A.); (N.V.); (L.M.); (N.R.)
- Translational Genomics and Targeted Therapeutics in Solid Tumors, Institut d’Investigacions Biomèdiques August Pi I Sunyer, 08036 Barcelona, Spain
- Unitat funcional de Tumors Toràcics, Hospital Clínic, 08036 Barcelona, Spain;
| | - Roxana Reyes
- Division of Medical Oncology, Hospital Clínic, 08036 Barcelona, Spain; (E.M.); (R.R.); (A.A.); (N.V.); (L.M.); (N.R.)
- Unitat funcional de Tumors Toràcics, Hospital Clínic, 08036 Barcelona, Spain;
| | - Ainara Arcocha
- Division of Medical Oncology, Hospital Clínic, 08036 Barcelona, Spain; (E.M.); (R.R.); (A.A.); (N.V.); (L.M.); (N.R.)
- Unitat funcional de Tumors Toràcics, Hospital Clínic, 08036 Barcelona, Spain;
| | - Nuria Viñolas
- Division of Medical Oncology, Hospital Clínic, 08036 Barcelona, Spain; (E.M.); (R.R.); (A.A.); (N.V.); (L.M.); (N.R.)
- Translational Genomics and Targeted Therapeutics in Solid Tumors, Institut d’Investigacions Biomèdiques August Pi I Sunyer, 08036 Barcelona, Spain
- Unitat funcional de Tumors Toràcics, Hospital Clínic, 08036 Barcelona, Spain;
| | - Laura Mezquita
- Division of Medical Oncology, Hospital Clínic, 08036 Barcelona, Spain; (E.M.); (R.R.); (A.A.); (N.V.); (L.M.); (N.R.)
- Translational Genomics and Targeted Therapeutics in Solid Tumors, Institut d’Investigacions Biomèdiques August Pi I Sunyer, 08036 Barcelona, Spain
- Unitat funcional de Tumors Toràcics, Hospital Clínic, 08036 Barcelona, Spain;
| | - Elena Gonzalvo
- Division of Pathology, Hospital Clínic, 08036 Barcelona, Spain; (E.G.); (K.S.d.G.)
| | | | - Pedro Jares
- Unitat funcional de Tumors Toràcics, Hospital Clínic, 08036 Barcelona, Spain;
- Division of Pathology, Hospital Clínic, 08036 Barcelona, Spain; (E.G.); (K.S.d.G.)
- Molecular Biology Core Facility, Hospital Clínic, 08036 Barcelona, Spain
| | - Noemi Reguart
- Division of Medical Oncology, Hospital Clínic, 08036 Barcelona, Spain; (E.M.); (R.R.); (A.A.); (N.V.); (L.M.); (N.R.)
- Translational Genomics and Targeted Therapeutics in Solid Tumors, Institut d’Investigacions Biomèdiques August Pi I Sunyer, 08036 Barcelona, Spain
- Unitat funcional de Tumors Toràcics, Hospital Clínic, 08036 Barcelona, Spain;
| | - Cristina Teixido
- Translational Genomics and Targeted Therapeutics in Solid Tumors, Institut d’Investigacions Biomèdiques August Pi I Sunyer, 08036 Barcelona, Spain
- Unitat funcional de Tumors Toràcics, Hospital Clínic, 08036 Barcelona, Spain;
- Division of Pathology, Hospital Clínic, 08036 Barcelona, Spain; (E.G.); (K.S.d.G.)
| |
Collapse
|
14
|
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: 0.6] [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
|
15
|
Choi H, Deng J, Li S, Silk T, Dong L, Brea EJ, Houghton S, Redmond D, Zhong H, Boiarsky J, Akbay EA, Smith PD, Merghoub T, Wong KK, Wolchok JD. Pulsatile MEK Inhibition Improves Anti-tumor Immunity and T Cell Function in Murine Kras Mutant Lung Cancer. Cell Rep 2020; 27:806-819.e5. [PMID: 30995478 PMCID: PMC6719696 DOI: 10.1016/j.celrep.2019.03.066] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 02/01/2019] [Accepted: 03/18/2019] [Indexed: 01/01/2023] Open
Abstract
KRAS is one of the driver oncogenes in non-small-cell lung cancer (NSCLC) but remains refractory to current modalities of targeted pathway inhibition, which include inhibiting downstream kinase MEK to circumvent KRAS activation. Here, we show that pulsatile, rather than continuous, treatment with MEK inhibitors (MEKis) maintains T cell activation and enables their proliferation. Two MEKis, selumetinib and trametinib, induce T cell activation with increased CTLA-4 expression and, to a lesser extent, PD-1 expression on T cells in vivo after cyclical pulsatile MEKi treatment. In addition, the pulsatile dosing schedule alone shows superior anti-tumor effects and delays the emergence of drug resistance. Furthermore, pulsatile MEKi treatment combined with CTLA-4 blockade prolongs survival in mice bearing tumors with mutant Kras. Our results set the foundation and show the importance of a combinatorial therapeutic strategy using pulsatile targeted therapy together with immunotherapy to optimally enhance tumor delay and promote long-term anti-tumor immunity.
Collapse
Affiliation(s)
- Hyejin Choi
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Jiehui Deng
- Division of Hematology & Medical Oncology, Laura and Isaac Perlmutter Cancer Center, New York University Langone Medical Center, New York, NY 10016, USA
| | - Shuai Li
- Division of Hematology & Medical Oncology, Laura and Isaac Perlmutter Cancer Center, New York University Langone Medical Center, New York, NY 10016, USA
| | - Tarik Silk
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Lauren Dong
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Elliott J Brea
- Molecular Pharmacology and Chemistry Program, Sloan Kettering Institute, New York, NY 10065, USA; Weill Cornell Medicine, New York, NY 10065, USA
| | - Sean Houghton
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - David Redmond
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Hong Zhong
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Jonathan Boiarsky
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Esra A Akbay
- Department of Pathology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA; Simmons Comprehensive Cancer Center, Dallas, TX 75390, USA
| | - Paul D Smith
- Bioscience, iMed Oncology, AstraZeneca, CRUK Cambridge Institute, Cambridge CB2 0RE, UK
| | - Taha Merghoub
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Weill Cornell Medicine, New York, NY 10065, USA.
| | - Kwok-Kin Wong
- Division of Hematology & Medical Oncology, Laura and Isaac Perlmutter Cancer Center, New York University Langone Medical Center, New York, NY 10016, USA.
| | - Jedd D Wolchok
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Weill Cornell Medicine, New York, NY 10065, USA.
| |
Collapse
|
16
|
Cardiovascular safety of rapidly accelerated fibrosarcoma B-type and/or mitogen-activated extracellular signal-regulated kinase inhibitors: A mixed approach combining a meta-analysis and a pharmacovigilance disproportionality analysis. Arch Cardiovasc Dis 2020; 113:420-432. [DOI: 10.1016/j.acvd.2020.03.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 02/10/2020] [Accepted: 03/09/2020] [Indexed: 12/11/2022]
|
17
|
Zheng C, Li X, Ren Y, Yin Z, Zhou B. Coexisting EGFR and TP53 Mutations in Lung Adenocarcinoma Patients Are Associated With COMP and ITGB8 Upregulation and Poor Prognosis. Front Mol Biosci 2020; 7:30. [PMID: 32175330 PMCID: PMC7056714 DOI: 10.3389/fmolb.2020.00030] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 02/11/2020] [Indexed: 12/24/2022] Open
Abstract
The heterogeneity of lung adenocarcinoma is driven by key mutations in oncogenes. To determine the gene expression, single nucleotide polymorphisms, and co-mutations participating in the initiation and progression of lung adenocarcinoma, we comprehensively analyzed the data of 491 patients from The Cancer Genome Atlas. Using log-rank and Kruskal-Wallis analysis, Oncoprint, Kaplan-Meier survival plots, and a nomogram, we found that EGFRL858R with co-mutation TP53 was significant prognostic determinant versus that with co-wild TP53 (hazard ratio, 2.77, P = 0.012). Further gene co-expression network and functional enrichment analysis indicated that co-mutation of EGFRL858R/TP53 increases the expression of COMP and ITGB8, which are involved in extracellular matrix organization and cell surface receptor signaling pathways, thus contributing to poor prognosis in lung adenocarcinoma. Validation was performed using three GEO profiles along with colony formation and CCK-8 assays for proliferation, transwell and wound-healing for migration in transfected H1299 and A549 cell lines. To the best of our knowledge, these results are the first to indicate that patients harboring the co-mutation of EGFRL858R/TP53 show increased expression of COMP and ITGB8, which participate in extracellular matrix dysfunction and can be used as prognostic biomarkers in patients with lung adenocarcinoma.
Collapse
Affiliation(s)
- Chang Zheng
- Department of Clinical Epidemiology, First Affiliated Hospital of China Medical University, Shenyang, China.,Department of Epidemiology, School of Public Health, China Medical University, Shenyang, China
| | - Xuelian Li
- Department of Epidemiology, School of Public Health, China Medical University, Shenyang, China
| | - Yangwu Ren
- Department of Epidemiology, School of Public Health, China Medical University, Shenyang, China
| | - Zhihua Yin
- Department of Epidemiology, School of Public Health, China Medical University, Shenyang, China
| | - Baosen Zhou
- Department of Clinical Epidemiology, First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
18
|
Lamping M, Benary M, Leyvraz S, Messerschmidt C, Blanc E, Kessler T, Schütte M, Lenze D, Jöhrens K, Burock S, Klinghammer K, Ochsenreither S, Sers C, Schäfer R, Tinhofer I, Beule D, Klauschen F, Yaspo ML, Keilholz U, Rieke DT. Support of a molecular tumour board by an evidence-based decision management system for precision oncology. Eur J Cancer 2020; 127:41-51. [PMID: 31982633 DOI: 10.1016/j.ejca.2019.12.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 12/06/2019] [Accepted: 12/10/2019] [Indexed: 12/19/2022]
Abstract
BACKGROUND Reliable and reproducible interpretation of molecular aberrations constitutes a bottleneck of precision medicine. Evidence-based decision management systems may improve rational therapy recommendations. To cope with an increasing amount of complex molecular data in the clinical care of patients with cancer, we established a workflow for the interpretation of molecular analyses. METHODS A specialized physician screened results from molecular analyses for potential biomarkers, irrespective of the diagnostic modality. Best available evidence was retrieved and categorized through establishment of an in-house database and interrogation of publicly available databases. Annotated biomarkers were ranked using predefined evidence levels and subsequently discussed at a molecular tumour board (MTB), which generated treatment recommendations. Subsequent translation into patient treatment and clinical outcomes were followed up. RESULTS One hundred patients were discussed in the MTB between January 2016 and May 2017. Molecular data were obtained for 70 of 100 patients (50 whole exome/RNA sequencing, 18 panel sequencing, 2 immunohistochemistry (IHC)/microsatellite instability analysis). The MTB generated a median of two treatment recommendations each for 63 patients. Thirty-nine patients were treated: 6 partial responses and 12 stable diseases were achieved as best responses. Genetic counselling for germline events was recommended for seven patients. CONCLUSION The development of an evidence-based workflow allowed for the clinical interpretation of complex molecular data and facilitated the translation of personalized treatment strategies into routine clinical care. The high number of treatment recommendations in patients with comprehensive genomic data and promising responses in patients treated with combination therapy warrant larger clinical studies.
Collapse
Affiliation(s)
- Mario Lamping
- Charité Comprehensive Cancer Center, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany.
| | - Manuela Benary
- Department of Pathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany; IRI Life Sciences, Humboldt-Universität zu Berlin, Philippstraße 13, 10115, Berlin, Germany
| | - Serge Leyvraz
- Charité Comprehensive Cancer Center, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
| | - Clemens Messerschmidt
- Core Unit Bioinformatics, Berlin Institute of Health (BIH), Anna-Louisa-Karsch-Straße 2, 10178, Berlin, Germany
| | - Eric Blanc
- Core Unit Bioinformatics, Berlin Institute of Health (BIH), Anna-Louisa-Karsch-Straße 2, 10178, Berlin, Germany
| | - Thomas Kessler
- Alacris Theranostics GmbH, Max Planck Straße 3, 12489, Berlin, Germany
| | - Moritz Schütte
- Alacris Theranostics GmbH, Max Planck Straße 3, 12489, Berlin, Germany
| | - Dido Lenze
- Department of Pathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
| | - Korinna Jöhrens
- Department of Pathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
| | - Susen Burock
- Charité Comprehensive Cancer Center, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
| | - Konrad Klinghammer
- Department of Hematology and Oncology, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Sebastian Ochsenreither
- Charité Comprehensive Cancer Center, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany; Department of Hematology and Oncology, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Christine Sers
- Department of Pathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
| | - Reinhold Schäfer
- Charité Comprehensive Cancer Center, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany; German Cancer Consortium (DKTK) and German Cancer Research Centre (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Ingeborg Tinhofer
- German Cancer Consortium (DKTK) and German Cancer Research Centre (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany; Department of Radiooncology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
| | - Dieter Beule
- Core Unit Bioinformatics, Berlin Institute of Health (BIH), Anna-Louisa-Karsch-Straße 2, 10178, Berlin, Germany
| | - Frederick Klauschen
- Department of Pathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
| | - Marie-Laure Yaspo
- Alacris Theranostics GmbH, Max Planck Straße 3, 12489, Berlin, Germany; Max Planck Institute for Molecular Genetics, Otto Warburg Laboratory Gene Regulation and Systems Biology of Cancer, Ihnestraße 63, 14195, Berlin, Germany
| | - Ulrich Keilholz
- Charité Comprehensive Cancer Center, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany; German Cancer Consortium (DKTK) and German Cancer Research Centre (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Damian T Rieke
- Charité Comprehensive Cancer Center, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany; Department of Hematology and Oncology, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Hindenburgdamm 30, 12203 Berlin, Germany; Berlin Institute of Health (BIH), Anna-Louisa-Karsch-Straße 2, 10178, Berlin, Germany.
| |
Collapse
|
19
|
Giopanou I, Pintzas A. RAS and BRAF in the foreground for non-small cell lung cancer and colorectal cancer: Similarities and main differences for prognosis and therapies. Crit Rev Oncol Hematol 2019; 146:102859. [PMID: 31927392 DOI: 10.1016/j.critrevonc.2019.102859] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 12/16/2019] [Accepted: 12/16/2019] [Indexed: 02/06/2023] Open
Abstract
Lung and colorectal cancer are included in the most tremendously threatening diseases in terms of incidence and death. Although they are located in completely different organs and differ in various characteristics they do share some common features, especially regarding their molecular mutational profile. Among several commonly mutated genes KRAS and BRAF are spotted to be highly associated with patient's poor disease outcome and resistance to targeted therapies mostly in liaison with other mutant activated genes. Many studies have shed light in these mechanisms for disease progression and numerous preclinical models, clinical trials and meta-analysis reports investigate the impact of specific treatments or combination of therapies. The present review is an effort to compare the mutational imprint of these genes between the two diseases and their impact in prognosis, current therapy, mechanisms of therapy resistance and future therapeutic plans and provide a spherical perspective regarding the systemic molecular profile of cancer.
Collapse
Affiliation(s)
- Ioanna Giopanou
- Institute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece.
| | - Alexandros Pintzas
- Institute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece.
| |
Collapse
|
20
|
Melosky B, Bradbury P, Tu D, Florescu M, Reiman A, Nicholas G, Basappa N, Rothenstein J, Goffin JR, Laurie SA, Wheatley-Price P, Leighl N, Goss G, Reaume MN, Butts C, Murray N, Card C, Ko J, Blais N, Gray S, Lui H, Brown-Walker P, Kaurah P, Prentice LM, Seymour L. Selumetinib in patients receiving standard pemetrexed and platinum-based chemotherapy for advanced or metastatic KRAS wildtype or unknown non-squamous non-small cell lung cancer: A randomized, multicenter, phase II study. Canadian Cancer Trials Group (CCTG) IND.219. Lung Cancer 2019; 133:48-55. [PMID: 31200828 DOI: 10.1016/j.lungcan.2019.04.027] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 04/27/2019] [Accepted: 04/30/2019] [Indexed: 01/09/2023]
Abstract
INTRODUCTION Activation of the RAS/RAF/MEK/ERK pathway may confer resistance to chemotherapy in non-small cell lung cancer (NSCLC). Selumetinib (AZD6244, ARRY142886), a MEK1/2 inhibitor combined with chemotherapy in patients with NSCLC was evaluated in two schedules to evaluate efficacy and toxicity. METHODS IND.219 was a three-arm study of first line pemetrexed/platinum chemotherapy with two schedules of selumetinib (Arm A: intermittent given on days 2-19; Arm B: continuous given on days 1-21) versus chemotherapy alone (Arm C). The primary endpoint was objective response rate (ORR); secondary objectives were tolerability, progression-free survival (PFS), overall survival (OS). The trial was stopped at the planned interim analysis. RESULTS Arms A/B/C enrolled 20/21/21 patients, ORR was 35% (95% CI 15-59% median duration 3.8 months), 62% (95% CI 38-82%; median duration 6.3 months), 24% (95% CI 8-47%; median duration 11.6 months) respectively. The PFS (months Arm A, B, C) was 7.5, 6.7, 4.0 respectively (hazard ratio (HR) PFS Arm A over Arm C: 0.76 [95% CI, 0.38-1.51, 2-sided p = 0.42]; Arm B over Arm C 0.75 [95% CI 0.37-1.54, p = 0.43]. Skin and gastrointestinal adverse events were more common with the addition of selumetinib. A high incidence of venous thromboembolism was seen in all arms. CONCLUSIONS Selumetinib combined with chemotherapy was associated with a higher response rate. Continuous selumetinib appeared to be superior to an intermittent schedule. PFS was prolonged with the addition of selumetinib, however this was not statistically significant.
Collapse
Affiliation(s)
| | - Penelope Bradbury
- Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Dongsheng Tu
- Canadian Cancer Trials Group, Kingston, ON, Canada
| | | | - Anthony Reiman
- Atlantic Health Sciences Corporation, Saint John, NB, Canada
| | | | | | | | - John R Goffin
- Juravinski Cancer Centre at Hamilton Health Sciences, Hamilton, ON, Canada
| | | | | | - Natasha Leighl
- Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Glenwood Goss
- Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - M Neil Reaume
- Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | | | - Nevin Murray
- BCCA - Vancouver Cancer Centre, Vancouver, BC, Canada
| | | | - Jenny Ko
- BCCA - Abbotsford Centre, Abbotsford, BC, Canada
| | | | - Samantha Gray
- Atlantic Health Sciences Corporation, Saint John, NB, Canada
| | - Hongbo Lui
- Canadian Cancer Trials Group, Kingston, ON, Canada
| | | | | | | | | |
Collapse
|
21
|
Rieke DT, Lamping M, Schuh M, Le Tourneau C, Basté N, Burkard ME, Metzeler KH, Leyvraz S, Keilholz U. Comparison of Treatment Recommendations by Molecular Tumor Boards Worldwide. JCO Precis Oncol 2018; 2:1-14. [PMID: 35135153 DOI: 10.1200/po.18.00098] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025] Open
Abstract
PURPOSE Precision oncology holds the promise of improving patient outcome. It is based on the idea that the testing of genomic biomarkers can lead to the recommendation of a treatment option tailored to the specific patient. To derive treatment recommendations from molecular profiles, interdisciplinary molecular tumor boards (MTBs) have been established recently in many academic institutions. The recommendation process in MTBs, however, has not been well defined, which limits applicability to larger clinical trials and patient populations. METHODS We created four fictional patients on the basis of recent real cases with genomic information on mutations, fusions, copy numbers, and gene expression. We identified 29 tumor boards from nine countries worldwide and asked them to provide treatment recommendations for the sample patients. In addition, a questionnaire regarding the setup and methods used by MTBs was circulated. RESULTS Five MTBs from four countries provided treatment recommendations and answered the questionnaire. For one patient, three tumor board treatment recommendations were identical, and two tumor boards had identical treatment strategies for the other three patients. There was heterogeneity in the interpretation of tumor and germline aberrations as well as in standards of prioritization. CONCLUSION Differences in the interpretation and recommendation process contribute to heterogeneity in MTB recommendations. Additional comparative analyses of recommendations could help improve rational decision making and lead to standardization.
Collapse
Affiliation(s)
- Damian T Rieke
- Damian T. Rieke, Mario Lamping, Serge Leyvraz, and Ulrich Keilholz, Charité Comprehensive Cancer Center, Charité-Universitätsmedizin Berlin; Damian T. Rieke, Berlin Institute of Health, Berlin; Klaus H. Metzeler, University Hospital, LMU Munich, Munich, Germany; Marissa Schuh, Markey Cancer Center, University of Kentucky, Lexington, KY; Christophe Le Tourneau, Institut Curie and INSERM U900 Research Unit, Saint-Cloud; Christophe Le Tourneau, Institut Curie, Paris; Christophe Le Tourneau, Versailles-Saint-Quentin-en-Yvelines University, Montigny-le-Bretonneux, France; Neus Basté, Vall d'Hebron Institute of Oncology, Barcelona, Spain; and Mark E. Burkard, UW Carbone Cancer Center, University of Wisconsin, Madison, WI
| | - Mario Lamping
- Damian T. Rieke, Mario Lamping, Serge Leyvraz, and Ulrich Keilholz, Charité Comprehensive Cancer Center, Charité-Universitätsmedizin Berlin; Damian T. Rieke, Berlin Institute of Health, Berlin; Klaus H. Metzeler, University Hospital, LMU Munich, Munich, Germany; Marissa Schuh, Markey Cancer Center, University of Kentucky, Lexington, KY; Christophe Le Tourneau, Institut Curie and INSERM U900 Research Unit, Saint-Cloud; Christophe Le Tourneau, Institut Curie, Paris; Christophe Le Tourneau, Versailles-Saint-Quentin-en-Yvelines University, Montigny-le-Bretonneux, France; Neus Basté, Vall d'Hebron Institute of Oncology, Barcelona, Spain; and Mark E. Burkard, UW Carbone Cancer Center, University of Wisconsin, Madison, WI
| | - Marissa Schuh
- Damian T. Rieke, Mario Lamping, Serge Leyvraz, and Ulrich Keilholz, Charité Comprehensive Cancer Center, Charité-Universitätsmedizin Berlin; Damian T. Rieke, Berlin Institute of Health, Berlin; Klaus H. Metzeler, University Hospital, LMU Munich, Munich, Germany; Marissa Schuh, Markey Cancer Center, University of Kentucky, Lexington, KY; Christophe Le Tourneau, Institut Curie and INSERM U900 Research Unit, Saint-Cloud; Christophe Le Tourneau, Institut Curie, Paris; Christophe Le Tourneau, Versailles-Saint-Quentin-en-Yvelines University, Montigny-le-Bretonneux, France; Neus Basté, Vall d'Hebron Institute of Oncology, Barcelona, Spain; and Mark E. Burkard, UW Carbone Cancer Center, University of Wisconsin, Madison, WI
| | - Christophe Le Tourneau
- Damian T. Rieke, Mario Lamping, Serge Leyvraz, and Ulrich Keilholz, Charité Comprehensive Cancer Center, Charité-Universitätsmedizin Berlin; Damian T. Rieke, Berlin Institute of Health, Berlin; Klaus H. Metzeler, University Hospital, LMU Munich, Munich, Germany; Marissa Schuh, Markey Cancer Center, University of Kentucky, Lexington, KY; Christophe Le Tourneau, Institut Curie and INSERM U900 Research Unit, Saint-Cloud; Christophe Le Tourneau, Institut Curie, Paris; Christophe Le Tourneau, Versailles-Saint-Quentin-en-Yvelines University, Montigny-le-Bretonneux, France; Neus Basté, Vall d'Hebron Institute of Oncology, Barcelona, Spain; and Mark E. Burkard, UW Carbone Cancer Center, University of Wisconsin, Madison, WI
| | - Neus Basté
- Damian T. Rieke, Mario Lamping, Serge Leyvraz, and Ulrich Keilholz, Charité Comprehensive Cancer Center, Charité-Universitätsmedizin Berlin; Damian T. Rieke, Berlin Institute of Health, Berlin; Klaus H. Metzeler, University Hospital, LMU Munich, Munich, Germany; Marissa Schuh, Markey Cancer Center, University of Kentucky, Lexington, KY; Christophe Le Tourneau, Institut Curie and INSERM U900 Research Unit, Saint-Cloud; Christophe Le Tourneau, Institut Curie, Paris; Christophe Le Tourneau, Versailles-Saint-Quentin-en-Yvelines University, Montigny-le-Bretonneux, France; Neus Basté, Vall d'Hebron Institute of Oncology, Barcelona, Spain; and Mark E. Burkard, UW Carbone Cancer Center, University of Wisconsin, Madison, WI
| | - Mark E Burkard
- Damian T. Rieke, Mario Lamping, Serge Leyvraz, and Ulrich Keilholz, Charité Comprehensive Cancer Center, Charité-Universitätsmedizin Berlin; Damian T. Rieke, Berlin Institute of Health, Berlin; Klaus H. Metzeler, University Hospital, LMU Munich, Munich, Germany; Marissa Schuh, Markey Cancer Center, University of Kentucky, Lexington, KY; Christophe Le Tourneau, Institut Curie and INSERM U900 Research Unit, Saint-Cloud; Christophe Le Tourneau, Institut Curie, Paris; Christophe Le Tourneau, Versailles-Saint-Quentin-en-Yvelines University, Montigny-le-Bretonneux, France; Neus Basté, Vall d'Hebron Institute of Oncology, Barcelona, Spain; and Mark E. Burkard, UW Carbone Cancer Center, University of Wisconsin, Madison, WI
| | - Klaus H Metzeler
- Damian T. Rieke, Mario Lamping, Serge Leyvraz, and Ulrich Keilholz, Charité Comprehensive Cancer Center, Charité-Universitätsmedizin Berlin; Damian T. Rieke, Berlin Institute of Health, Berlin; Klaus H. Metzeler, University Hospital, LMU Munich, Munich, Germany; Marissa Schuh, Markey Cancer Center, University of Kentucky, Lexington, KY; Christophe Le Tourneau, Institut Curie and INSERM U900 Research Unit, Saint-Cloud; Christophe Le Tourneau, Institut Curie, Paris; Christophe Le Tourneau, Versailles-Saint-Quentin-en-Yvelines University, Montigny-le-Bretonneux, France; Neus Basté, Vall d'Hebron Institute of Oncology, Barcelona, Spain; and Mark E. Burkard, UW Carbone Cancer Center, University of Wisconsin, Madison, WI
| | - Serge Leyvraz
- Damian T. Rieke, Mario Lamping, Serge Leyvraz, and Ulrich Keilholz, Charité Comprehensive Cancer Center, Charité-Universitätsmedizin Berlin; Damian T. Rieke, Berlin Institute of Health, Berlin; Klaus H. Metzeler, University Hospital, LMU Munich, Munich, Germany; Marissa Schuh, Markey Cancer Center, University of Kentucky, Lexington, KY; Christophe Le Tourneau, Institut Curie and INSERM U900 Research Unit, Saint-Cloud; Christophe Le Tourneau, Institut Curie, Paris; Christophe Le Tourneau, Versailles-Saint-Quentin-en-Yvelines University, Montigny-le-Bretonneux, France; Neus Basté, Vall d'Hebron Institute of Oncology, Barcelona, Spain; and Mark E. Burkard, UW Carbone Cancer Center, University of Wisconsin, Madison, WI
| | - Ulrich Keilholz
- Damian T. Rieke, Mario Lamping, Serge Leyvraz, and Ulrich Keilholz, Charité Comprehensive Cancer Center, Charité-Universitätsmedizin Berlin; Damian T. Rieke, Berlin Institute of Health, Berlin; Klaus H. Metzeler, University Hospital, LMU Munich, Munich, Germany; Marissa Schuh, Markey Cancer Center, University of Kentucky, Lexington, KY; Christophe Le Tourneau, Institut Curie and INSERM U900 Research Unit, Saint-Cloud; Christophe Le Tourneau, Institut Curie, Paris; Christophe Le Tourneau, Versailles-Saint-Quentin-en-Yvelines University, Montigny-le-Bretonneux, France; Neus Basté, Vall d'Hebron Institute of Oncology, Barcelona, Spain; and Mark E. Burkard, UW Carbone Cancer Center, University of Wisconsin, Madison, WI
| |
Collapse
|
22
|
Gambardella V, Martin-Martorell P, Cervantes A. In the literature: August 2018. ESMO Open 2018; 3:e000427. [PMID: 30233823 PMCID: PMC6135457 DOI: 10.1136/esmoopen-2018-000427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Affiliation(s)
- Valentina Gambardella
- Department of Medical Oncology, Biomedical Research Institute INCLIVA, CiberOnc, University of Valencia, Valencia, Spain
| | - Paloma Martin-Martorell
- Department of Medical Oncology, Biomedical Research Institute INCLIVA, CiberOnc, University of Valencia, Valencia, Spain
| | - Andrés Cervantes
- Department of Medical Oncology, Biomedical Research Institute INCLIVA, CiberOnc, University of Valencia, Valencia, Spain
| |
Collapse
|
23
|
Tong X, Xu H, Carlile DJ, Tomkinson H, Al‐Huniti N, Zhou D. Population Pharmacokinetic and Exposure‐Response Analysis of Selumetinib and Its N‐desmethyl Metabolite in Patients With Non‐Small Cell Lung Cancer. J Clin Pharmacol 2018; 59:112-122. [DOI: 10.1002/jcph.1295] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 07/06/2018] [Indexed: 11/05/2022]
Affiliation(s)
- Xiao Tong
- Quantitative Clinical PharmacologyEarly Clinical DevelopmentIMED Biotech UnitAstraZeneca Boston MA USA
| | - Hongmei Xu
- Quantitative Clinical PharmacologyEarly Clinical DevelopmentIMED Biotech UnitAstraZeneca Boston MA USA
| | - David J. Carlile
- Quantitative Clinical PharmacologyEarly Clinical DevelopmentIMED Biotech UnitAstraZeneca Cambridge UK
| | - Helen Tomkinson
- Quantitative Clinical PharmacologyEarly Clinical DevelopmentIMED Biotech UnitAstraZeneca Cambridge UK
| | - Nidal Al‐Huniti
- Quantitative Clinical PharmacologyEarly Clinical DevelopmentIMED Biotech UnitAstraZeneca Boston MA USA
| | - Diansong Zhou
- Quantitative Clinical PharmacologyEarly Clinical DevelopmentIMED Biotech UnitAstraZeneca Boston MA USA
| |
Collapse
|