251
|
Leroy L, Bonhomme B, Le Moulec S, Soubeyran I, Italiano A, Godbert Y. Remarkable Response to Ceritinib and Brigatinib in an Anaplastic Lymphoma Kinase-Rearranged Anaplastic Thyroid Carcinoma Previously Treated with Crizotinib. Thyroid 2020; 30:343-344. [PMID: 31892283 DOI: 10.1089/thy.2019.0202] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
| | | | | | | | - Antoine Italiano
- Bergonie Institute, Bordeaux, France
- University of Bordeaux, Bordeaux, France
| | | |
Collapse
|
252
|
Zhu C, Zhuang W, Chen L, Yang W, Ou WB. Frontiers of ctDNA, targeted therapies, and immunotherapy in non-small-cell lung cancer. Transl Lung Cancer Res 2020; 9:111-138. [PMID: 32206559 PMCID: PMC7082279 DOI: 10.21037/tlcr.2020.01.09] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 01/02/2020] [Indexed: 12/19/2022]
Abstract
Non-small-cell lung cancer (NSCLC), a main subtype of lung cancer, is one of the most common causes of cancer death in men and women worldwide. Circulating tumor DNA (ctDNA), tyrosine kinase inhibitors (TKIs) and immunotherapy have revolutionized both our understanding of NSCLC, from its diagnosis to targeted NSCLC therapies, and its treatment. ctDNA quantification confers convenience and precision to clinical decision making. Furthermore, the implementation of TKI-based targeted therapy and immunotherapy has significantly improved NSCLC patient quality of life. This review provides an update on the methods of ctDNA detection and its impact on therapeutic strategies; therapies that target epidermal growth factor receptor (EGFR) and anaplastic lymphoma kinase (ALK) using TKIs such as osimertinib and lorlatinib; the rise of various resistant mechanisms; and the control of programmed cell death-1 (PD-1), programmed cell death ligand-1 (PD-L1), and cytotoxic T-lymphocyte antigen-4 (CTLA-4) by immune checkpoint inhibitors (ICIs) in immunotherapy; blood tumor mutational burden (bTMB) calculated by ctDNA assay as a novel biomarker for immunotherapy. However, NSCLC patients still face many challenges. Further studies and trials are needed to develop more effective drugs or therapies to treat NSCLC.
Collapse
Affiliation(s)
- Chennianci Zhu
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Weihao Zhuang
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Limin Chen
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Wenyu Yang
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Wen-Bin Ou
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
| |
Collapse
|
253
|
Lah TT, Novak M, Breznik B. Brain malignancies: Glioblastoma and brain metastases. Semin Cancer Biol 2020; 60:262-273. [DOI: 10.1016/j.semcancer.2019.10.010] [Citation(s) in RCA: 236] [Impact Index Per Article: 47.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 10/10/2019] [Accepted: 10/16/2019] [Indexed: 02/06/2023]
|
254
|
Alexander M, Pavlakis N, John T, O'Connell R, Kao S, Hughes BGM, Lee A, Hayes SA, Howell VM, Clarke SJ, Millward M, Burbury K, Solomon B, Itchins M. A multicenter study of thromboembolic events among patients diagnosed with ROS1-rearranged non-small cell lung cancer. Lung Cancer 2020; 142:34-40. [PMID: 32087434 DOI: 10.1016/j.lungcan.2020.01.017] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 01/16/2020] [Accepted: 01/20/2020] [Indexed: 12/21/2022]
Abstract
OBJECTIVES This study aimed to describe the longitudinal thromboembolism (TE) risk relative to the natural history of disease and clinical course of ROS1 rearranged non-small cell lung cancer (NSCLC). MATERIALS AND METHODS Cases of ROS1-rearranged NSCLC from six Australian hospitals were pooled and evaluated for incidence, timing, predictors and outcomes of venous or arterial TE, as well as objective response rate (ORR) to active therapy and overall survival (OS). RESULTS Of 42 patients recruited, 20 (48%) experienced TE; one (2%) arterial, 13 (31%) a pulmonary emboli (PE), and 12 (29%) a deep vein thrombosis. Among those with TE, six (30%) experienced multiple events, three as concurrent and three as recurrent diagnoses. The cumulative incidence of TE over time, adjusted for death as a competing risk factor, approached 50%. TE occurred prior to, during and post the peri-diagnostic period and occurred irrespective of treatment strategy. A thrombophilia was identified in n = 3/10 (30%) cases screened: in two factor V Leiden and in one anti-thrombin III (ATIII) deficiency. Median OS was 21.3 months in those with TE vs. 28.8 months in those without; hazard ratio 1.16 (95%CI 0.43-3.15). Respective ORR to first-line therapy with TE was 50% vs. 44% without TE in the chemotherapy arm and 67% vs. 50% in the targeted therapy arm. CONCLUSION In the rare cancer subtype, ROS1, these real-world data demonstrate sustained TE risk beyond the diagnostic period irrespective of therapeutic strategy. High incidence of PE, concurrent TE, and recurrent TE warrant validation in larger cohorts. Consideration of primary thromboprophylaxis in ROS1 populations is recommended.
Collapse
Affiliation(s)
- Marliese Alexander
- Department of Pharmacy, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia.
| | - Nick Pavlakis
- Bill Walsh Translational Research Laboratory, Kolling Institute Medical Institute of Research, Royal North Shore Hospital, St Leonards, New South Wales, Australia; Northern Clinical School, University of Sydney, St Leonards, New South Wales, Australia; Northern Cancer Institute, St Leonards, New South Wales, Australia; Department of Medical Oncology, Royal North Shore Hospital, St Leonards, New South Wales, Australia
| | - Thomas John
- Medical Oncology Unit, Olivia Newton John Cancer and Wellness Centre, Austin Health, Melbourne, Australia; Olivia Newton-John Cancer Research Institute, Melbourne, Victoria, Australia; Department of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Victoria, Australia
| | - Rachel O'Connell
- NHMRC Clinical Trial Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Steven Kao
- Sydney Medical School, University of Sydney, Camperdown, New South Wales, Australia; Department of Medical Oncology, Chris O'Brien Lifehouse, Camperdown, New South Wales, Australia
| | - Brett G M Hughes
- Department of Medical Oncology, The Prince Charles Hospital, Chermside West, Queensland, Australia; School of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Adrian Lee
- Bill Walsh Translational Research Laboratory, Kolling Institute Medical Institute of Research, Royal North Shore Hospital, St Leonards, New South Wales, Australia; Northern Clinical School, University of Sydney, St Leonards, New South Wales, Australia; Northern Cancer Institute, St Leonards, New South Wales, Australia; Department of Medical Oncology, Royal North Shore Hospital, St Leonards, New South Wales, Australia
| | - Sarah A Hayes
- Bill Walsh Translational Research Laboratory, Kolling Institute Medical Institute of Research, Royal North Shore Hospital, St Leonards, New South Wales, Australia; Northern Clinical School, University of Sydney, St Leonards, New South Wales, Australia
| | - Viive M Howell
- Bill Walsh Translational Research Laboratory, Kolling Institute Medical Institute of Research, Royal North Shore Hospital, St Leonards, New South Wales, Australia; Northern Clinical School, University of Sydney, St Leonards, New South Wales, Australia
| | - Stephen J Clarke
- Bill Walsh Translational Research Laboratory, Kolling Institute Medical Institute of Research, Royal North Shore Hospital, St Leonards, New South Wales, Australia; Northern Clinical School, University of Sydney, St Leonards, New South Wales, Australia; Northern Cancer Institute, St Leonards, New South Wales, Australia; Department of Medical Oncology, Royal North Shore Hospital, St Leonards, New South Wales, Australia
| | - Michael Millward
- School of Medicine, University of Western Australia, Perth, Western Australia, Australia; Department of Medical Oncology, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia
| | - Kate Burbury
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia; Department of Haematology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Benjamin Solomon
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia; Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Malinda Itchins
- Bill Walsh Translational Research Laboratory, Kolling Institute Medical Institute of Research, Royal North Shore Hospital, St Leonards, New South Wales, Australia; Northern Clinical School, University of Sydney, St Leonards, New South Wales, Australia; Northern Cancer Institute, St Leonards, New South Wales, Australia; Department of Medical Oncology, Royal North Shore Hospital, St Leonards, New South Wales, Australia
| |
Collapse
|
255
|
Lee SJ, Hong JY, Kim K, Kim KM, Kang SY, Lee T, Kim ST, Park SH, Park YS, Lim HY, Kang WK, Lee J, Park JO. Detection of Fusion Genes Using a Targeted RNA Sequencing Panel in Gastrointestinal and Rare Cancers. JOURNAL OF ONCOLOGY 2020; 2020:4659062. [PMID: 32411236 PMCID: PMC7204148 DOI: 10.1155/2020/4659062] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 12/09/2019] [Accepted: 12/19/2019] [Indexed: 12/22/2022]
Abstract
Successful identification and targeting of oncogenic gene fusion is a major breakthrough in cancer treatment. Here, we investigate the therapeutic implications and feasibility of using a targeted RNA sequencing panel to identify fusion genes in gastrointestinal and rare cancers. From February through December 2017, patients with gastrointestinal, hepatobiliary, gynecologic, sarcoma, or rare cancers were recruited for a clinical sequencing project at Samsung Medical Center (NCT #02593578). The median age of the patients was 58 years (range, 31-81 years), and the male-to-female ratio was 1.3 : 1. A total of 118 patients passed the quality control process for a next-generation sequencing- (NGS-) based targeted sequencing assay. The NGS-based targeted sequencing assay was performed to detect gene fusions in 36-53 cancer-implicated genes. The following cancer types were included in this study: 28 colorectal cancers, 27 biliary tract cancers, 25 gastric cancers, 18 soft tissue sarcomas, 9 pancreatic cancers, 6 ovarian cancers, and 9 other rare cancers. Strong fusion was detected in 25 samples (21.2%). We found that 5.9% (7/118) of patients had known targetable fusion genes involving NTRK1 (n=3), FGFR (n=3), and RET (n=1), and 10.2% (12/118) of patients had potentially targetable fusion genes involving RAF1 (n=4), BRAF (n=2), ALK (n=2), ROS1 (n=1), EGFR (n=1), and CLDN18 (n=2). Thus, we successfully identified a substantial proportion of patients harboring fusion genes by RNA panel sequencing of gastrointestinal/rare cancers. Targetable and potentially targetable involved fusion genes were NTRK1, RET, FGFR3, FGFR2, BRAF, RAF1, ALK, ROS1, and CLDN18. Detection of fusion genes by RNA panel sequencing may be beneficial in refractory patients with gastrointestinal/rare cancers.
Collapse
Affiliation(s)
- Su Jin Lee
- Division of Hematology-Oncology, Department of Medicine, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Division of Hematology-Oncology, Department of Internal Medicine, Ewha Womans University College of Medicine, Seoul, Republic of Korea
| | - Jung Yong Hong
- Division of Hematology-Oncology, Department of Medicine, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Kyung Kim
- Division of Hematology-Oncology, Department of Medicine, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Kyoung-Mee Kim
- Department of Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - So Young Kang
- Department of Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Taeyang Lee
- Division of Hematology-Oncology, Department of Medicine, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Seung Tae Kim
- Division of Hematology-Oncology, Department of Medicine, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Se Hoon Park
- Division of Hematology-Oncology, Department of Medicine, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Young Suk Park
- Division of Hematology-Oncology, Department of Medicine, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Ho Yeong Lim
- Division of Hematology-Oncology, Department of Medicine, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Won Ki Kang
- Division of Hematology-Oncology, Department of Medicine, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jeeyun Lee
- Division of Hematology-Oncology, Department of Medicine, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Joon Oh Park
- Division of Hematology-Oncology, Department of Medicine, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| |
Collapse
|
256
|
Remon J, Tabbò F, Jimenez B, Collazo A, de Castro J, Novello S. Sequential blinded treatment decisions in ALK-positive non-small cell lung cancers in the era of precision medicine. Clin Transl Oncol 2020; 22:1425-1429. [PMID: 31955355 DOI: 10.1007/s12094-020-02290-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 01/03/2020] [Indexed: 11/25/2022]
Abstract
Next-generation ALK TKIs have become the new standard of care in first-line setting in advanced ALK-positive NSCLC patients. However, sequential strategies at progression are relevant, as may have an impact on patients' outcome. In this commentary we discuss whether genomic-tailored strategies at progression would be more suitable for improving outcome of ALK-positive NSCLC patients.
Collapse
Affiliation(s)
- J Remon
- Department of Medical Oncology, Centro Integral Oncológico Clara Campal (HM-CIOCC), Hospital HM Delfos, HM Hospitales, Avinguda de Vallcarca, 151, 08023, Barcelona, Spain.
| | - F Tabbò
- Thoracic Oncology Unit, Department of Oncology, University of Torino, Regione Gonzole, 10, 10043, Orbassano (TO), Turin, Italy
| | - B Jimenez
- Medical Oncology Department, Centro Integral Oncológico Clara Campal Madrid, HM Sanchinarro, Calle Oña, 10, 28050, Madrid, Spain
| | - A Collazo
- Medical Oncology Department, Centro Integral Oncológico Clara Campal Madrid, HM Sanchinarro, Calle Oña, 10, 28050, Madrid, Spain
| | - J de Castro
- Medical Oncology Department, Centro Integral Oncológico Clara Campal Madrid, HM Sanchinarro, Calle Oña, 10, 28050, Madrid, Spain
| | - S Novello
- Thoracic Oncology Unit, Department of Oncology, University of Torino, Regione Gonzole, 10, 10043, Orbassano (TO), Turin, Italy
| |
Collapse
|
257
|
Abstract
A 40-year-old Japanese man with advanced pulmonary adenocarcinoma harboring anaplastic lymphoma kinase (ALK)-rearranged was administered the selective ALK inhibitor ceritinib as a third-line treatment and continued treatment for nine months. After fourth-line treatment, we performed rechallenge with ceritinib as a fifth-line treatment. On day 54 after rechallenge, the patient developed acutely deteriorating dyspnea. Chest computed tomography showed extensive ground-glass opacities. We diagnosed him with ceritinib-induced interstitial lung disease (ILD) and initiated methylprednisolone pulse therapy. To our knowledge, this is the first report of ceritinib-induced ILD in a Japanese patient. Since it may newly emerge with rechallenge therapy, close attention is necessary.
Collapse
Affiliation(s)
- Takamasa Hotta
- Division of Medical Oncology & Respiratory Medicine, Department of Internal Medicine, Shimane University, Japan
| | - Tamio Okimoto
- Division of Medical Oncology & Respiratory Medicine, Department of Internal Medicine, Shimane University, Japan
| | - Megumi Hamaguchi
- Division of Medical Oncology & Respiratory Medicine, Department of Internal Medicine, Shimane University, Japan
| | - Yukari Tsubata
- Division of Medical Oncology & Respiratory Medicine, Department of Internal Medicine, Shimane University, Japan
| | - Takeshi Isobe
- Division of Medical Oncology & Respiratory Medicine, Department of Internal Medicine, Shimane University, Japan
| |
Collapse
|
258
|
Wang Y, Han L, Liu F, Yang F, Jiang X, Sun H, Feng F, Xue J, Liu W. Targeted degradation of anaplastic lymphoma kinase by gold nanoparticle-based multi-headed proteolysis targeting chimeras. Colloids Surf B Biointerfaces 2020; 188:110795. [PMID: 31991291 DOI: 10.1016/j.colsurfb.2020.110795] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 12/19/2019] [Accepted: 01/12/2020] [Indexed: 02/01/2023]
Abstract
Anaplastic lymphoma kinase (ALK) is a major target in treating non-small-cell lung cancer, and several ALK inhibitors have been developed to antagonize its kinase activity. However, patients treated with inhibitors ultimately develop drug resistance. Therefore, therapies with new mechanisms of action are needed. Proteolysis targeting chimeras (PROTACs) are molecules that comprise a ligand for binding a protein of interest (POI), a connecting linker and a ligand for recruiting E3 ligase, and cause degradation of the target POI. Here, the first multi-headed PROTAC, as a proof of concept, is developed as a gold nanoparticle (GNP)-based drug delivery system for delivering PROTACs to target ALK. Pegylated GNPs loaded with both ceritinib and pomalidomide molecules, termed Cer/Pom-PEG@GNPs, showed good stability in several media. The GNP conjugates potently decreased the levels of ALK fusion proteins in a dose- and time-dependent manner, and specifically inhibited the proliferation of NCI-H2228 cells. In comparison with small molecule PROTACs, the new multi-headed PROTAC promoted the formation of coacervates of POIs/multi-headed PROTAC/E3 ubiquitin ligases, and POI and E3 ubiquitin ligase interacted through multidirectional ligands and a flexible linker, thereby avoiding the need for complicated structure optimization of PROTACs. In conclusion, Cer/Pom-PEG@GNPs can degrade intracellular ALK fusion proteins with minor off-target toxicity and can be applied in patients resistant to ALK inhibitors. As a nano-based drug carrier, Cer/Pom-PEG@GNPs have the potential to enable prolonged circulation and specifically distribute drugs to tumor regions in vivo; thus, further investigation is warranted.
Collapse
Affiliation(s)
- Yingming Wang
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, Jiangsu 210009, China
| | - Lingfei Han
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, Jiangsu 210009, China
| | - Fulei Liu
- The Joint Laboratory of China Pharmaceutical University and Taian City Central Hospital, Taian City Central Hospital, Taian, Shandong, 271000, China; Pharmaceutical Department, Taian City Central Hospital, Taian, Shandong 271000, China
| | - Fubai Yang
- The Joint Laboratory of China Pharmaceutical University and Taian City Central Hospital, Taian City Central Hospital, Taian, Shandong, 271000, China
| | - Xueyang Jiang
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, Jiangsu 210009, China; The Joint Laboratory of China Pharmaceutical University and Taian City Central Hospital, Taian City Central Hospital, Taian, Shandong, 271000, China
| | - Haopeng Sun
- The Joint Laboratory of China Pharmaceutical University and Taian City Central Hospital, Taian City Central Hospital, Taian, Shandong, 271000, China; Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing, Jiangsu 210009, China
| | - Feng Feng
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, Jiangsu 210009, China; The Joint Laboratory of China Pharmaceutical University and Taian City Central Hospital, Taian City Central Hospital, Taian, Shandong, 271000, China
| | - Jingwei Xue
- The Joint Laboratory of China Pharmaceutical University and Taian City Central Hospital, Taian City Central Hospital, Taian, Shandong, 271000, China.
| | - Wenyuan Liu
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, Jiangsu 210009, China.
| |
Collapse
|
259
|
Lo Gullo R, Daimiel I, Morris EA, Pinker K. Combining molecular and imaging metrics in cancer: radiogenomics. Insights Imaging 2020; 11:1. [PMID: 31901171 PMCID: PMC6942081 DOI: 10.1186/s13244-019-0795-6] [Citation(s) in RCA: 122] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 09/25/2019] [Indexed: 02/07/2023] Open
Abstract
Background Radiogenomics is the extension of radiomics through the combination of genetic and radiomic data. Because genetic testing remains expensive, invasive, and time-consuming, and thus unavailable for all patients, radiogenomics may play an important role in providing accurate imaging surrogates which are correlated with genetic expression, thereby serving as a substitute for genetic testing. Main body In this article, we define the meaning of radiogenomics and the difference between radiomics and radiogenomics. We provide an up-to-date review of the radiomics and radiogenomics literature in oncology, focusing on breast, brain, gynecological, liver, kidney, prostate and lung malignancies. We also discuss the current challenges to radiogenomics analysis. Conclusion Radiomics and radiogenomics are promising to increase precision in diagnosis, assessment of prognosis, and prediction of treatment response, providing valuable information for patient care throughout the course of the disease, given that this information is easily obtainable with imaging. Larger prospective studies and standardization will be needed to define relevant imaging biomarkers before they can be implemented into the clinical workflow.
Collapse
Affiliation(s)
- Roberto Lo Gullo
- Department of Radiology, Breast Imaging Service, Memorial Sloan Kettering Cancer Center, 300 E 66th St, New York, NY, 10065, USA.
| | - Isaac Daimiel
- Department of Radiology, Breast Imaging Service, Memorial Sloan Kettering Cancer Center, 300 E 66th St, New York, NY, 10065, USA
| | - Elizabeth A Morris
- Department of Radiology, Breast Imaging Service, Memorial Sloan Kettering Cancer Center, 300 E 66th St, New York, NY, 10065, USA
| | - Katja Pinker
- Department of Radiology, Breast Imaging Service, Memorial Sloan Kettering Cancer Center, 300 E 66th St, New York, NY, 10065, USA.,Department of Biomedical Imaging and Image-guided Therapy, Molecular and Gender Imaging Service, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Wien, Austria
| |
Collapse
|
260
|
Gotfrit J, Shin JJ, Mallick R, Stewart DJ, Wheatley‐Price P. Potential Life-Years Lost: The Impact of the Cancer Drug Regulatory and Funding Process in Canada. Oncologist 2020; 25:e130-e137. [PMID: 31506392 PMCID: PMC6964142 DOI: 10.1634/theoncologist.2019-0314] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 08/06/2019] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Canada has an established publicly funded health care system with a complex drug approval and funding process. After proof of efficacy (POE; key publication/presentation) and before becoming publicly accessible, each drug undergoes a Health Canada approval process, a health technology assessment (HTA), a pricing negotiation, and finally individual provincial funding agreements. We quantified potential life-years lost during this process. METHODS We analyzed drugs for advanced lung, breast, and colorectal cancer that underwent the HTA process between 2011 and 2016. Life-years lost were calculated by multiplying documented improvement in progression-free and overall survival, number of eligible patients, and time from POE to first public funding. For conservative calculation, we assumed all eligible patients in Canada had access at the time of first public funding, whereas in reality provinces fund at different time points. RESULTS We analyzed 21 drugs. Of these, 15 have been funded publicly. The time from POE to first public funding ranged from 14.0 to 99.2 months (median 26.6 months). Total overall life-years lost from POE to first public funding were 39,067 (lung 32,367; breast 6,691). Progression-free life-years lost from POE to first public funding were 48,037 (lung 9,139, breast 15,827, colorectal 23,071). CONCLUSION The number of potential life-years lost during the drug regulatory and funding process in Canada is substantial, largely driven by delays to funding of colorectal cancer drugs. Recognizing that interprovincial differences exist and that eligible patients may not all receive a given drug, if even a fraction does so, the impact of delays remains substantive. Collaborative national initiatives are required to address this major barrier to treatment access. IMPLICATIONS FOR PRACTICE Patients may spend lengthy periods of time awaiting access to new and effective cancer drugs. Patients with private drug insurance or personal funds or who reside in certain Canadian provinces may obtain some drugs sooner than others, potentially creating a two-tiered access system. The cancer drug access and public funding system must be expedited to improve equity.
Collapse
Affiliation(s)
- Joanna Gotfrit
- Division of Medical Oncology, Department of Medicine, The Ottawa Hospital Cancer Centre and the University of OttawaOttawaOntarioCanada
| | | | | | - David J. Stewart
- Division of Medical Oncology, Department of Medicine, The Ottawa Hospital Cancer Centre and the University of OttawaOttawaOntarioCanada
- The Ottawa Hospital Research InstituteOttawaOntarioCanada
| | - Paul Wheatley‐Price
- Division of Medical Oncology, Department of Medicine, The Ottawa Hospital Cancer Centre and the University of OttawaOttawaOntarioCanada
- The Ottawa Hospital Research InstituteOttawaOntarioCanada
| |
Collapse
|
261
|
Bylicki O, Guisier F, Monnet I, Doubre H, Gervais R, Janicot H, Perol M, Fournel P, Lamy R, Auliac JB, Chouaid C. Efficacy and safety of programmed cell-death-protein-1 and its ligand inhibitors in pretreated patients with epidermal growth-factor receptor-mutated or anaplastic lymphoma kinase-translocated lung adenocarcinoma. Medicine (Baltimore) 2020; 99:e18726. [PMID: 32011450 PMCID: PMC7220079 DOI: 10.1097/md.0000000000018726] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Immune-checkpoint inhibitor (ICI) efficacy in patients with non-small cell lung cancer (NSCLC) harboring molecular alterations remains poorly elucidated. This study was undertaken to determine ICI efficacy against epidermal growth-factor receptor (EGFR)/anaplastic lymphoma kinase (ALK)/c-ros oncogene 1 (ROS1)-mutated NSCLC patients in the real-world setting.In this retrospective, multicenter study on adults with ICI-treated EGFR-mutated or ALK- or ROS1-translated NSCLCs, we analyzed clinical characteristics and outcomes: ICI-treatment duration, and progression-free survival (PFS), objective response rate, duration of response, and overall survival (OS) from immunotherapy initiation.Fifty-one NSCLC patients (mean age, 58.0 years) were included from 20 French centers: 61% were never-smokers and 59% were women. Among them, 82% had EGFR-activating mutations, 16% ALK translocations, or 2% ROS1 translocations. Before ICI therapy, patients had received a median of 3 treatment lines (including tyrosine-kinase inhibitor). The median PFS was 2.1 (95% confidence interval [CI], 1.5-3.2) months for the entire cohort, 2.2 (95% CI, 1.4-3.2) for EGFR-mutated patients, and 2.4 (95% CI, 2.1-not reached) months for ALK-translocated patients. The median OS was 14.7 (95% CI, 12.1-19.2) months for the entire population and 13.9 (95% CI, 8.8-20.0) and 19.2 (95% CI, 13.1-not reached) months for EGFR-mutated and ALK-translocated patients, respectively. Seven (13.7%) patients were treated with ICI for >9 months. Toxicities were reported in 22% (11/51), including 8% (4/51) grade ≥3.In this real-world setting, analysis of ICI PFS against EGFR-mutated or ALK-translocated NSCLC patients appeared close to that observed in pretreated unselected NSCLC patients. The more promising OS probably linked to post-ICI treatments. Large prospective studies on these patient subsets are needed.
Collapse
Affiliation(s)
- Olivier Bylicki
- Service de Pneumologie, Hôpital d’Instruction des Armées Percy, Clamart
| | - Florian Guisier
- Service de Pneumologie, Centre Hospitalier Universitaire de Rouen, Rouen
| | - Isabelle Monnet
- Service de Pneumologie, Centre Hospitalier Intercommunal de Créteil, Créteil
| | | | - Radj Gervais
- Département d’oncologie, Centre François Baclesse, Caen
| | - Henri Janicot
- Service de Pneumologie, Centre Hospitalier Universitaire de Clermont-Ferrand, Clermont-Ferrand
| | - Maurice Perol
- Service d’Oncologie Thoracique, Centre Léon Bérard, Lyon
| | - Pierre Fournel
- Département d’oncologie, Institut de Cancérologie de la Loire, Saint-Priest-en-Jarez
| | - Régine Lamy
- Service de Pneumologie, Centre Hospitalier Bretagne Sud-Lorient, Lorient
| | - Jean-Bernard Auliac
- Service de Pneumologie, Centre Hospitalier F. Quesnay, Mantes-la-Jolie, France
| | - Christos Chouaid
- Service de Pneumologie, Centre Hospitalier Intercommunal de Créteil, Créteil
| |
Collapse
|
262
|
Lim ZF, Ma PC. Emerging insights of tumor heterogeneity and drug resistance mechanisms in lung cancer targeted therapy. J Hematol Oncol 2019; 12:134. [PMID: 31815659 PMCID: PMC6902404 DOI: 10.1186/s13045-019-0818-2] [Citation(s) in RCA: 352] [Impact Index Per Article: 58.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 10/31/2019] [Indexed: 02/07/2023] Open
Abstract
The biggest hurdle to targeted cancer therapy is the inevitable emergence of drug resistance. Tumor cells employ different mechanisms to resist the targeting agent. Most commonly in EGFR-mutant non-small cell lung cancer, secondary resistance mutations on the target kinase domain emerge to diminish the binding affinity of first- and second-generation inhibitors. Other alternative resistance mechanisms include activating complementary bypass pathways and phenotypic transformation. Sequential monotherapies promise to temporarily address the problem of acquired drug resistance, but evidently are limited by the tumor cells' ability to adapt and evolve new resistance mechanisms to persist in the drug environment. Recent studies have nominated a model of drug resistance and tumor progression under targeted therapy as a result of a small subpopulation of cells being able to endure the drug (minimal residual disease cells) and eventually develop further mutations that allow them to regrow and become the dominant population in the therapy-resistant tumor. This subpopulation of cells appears to have developed through a subclonal event, resulting in driver mutations different from the driver mutation that is tumor-initiating in the most common ancestor. As such, an understanding of intratumoral heterogeneity-the driving force behind minimal residual disease-is vital for the identification of resistance drivers that results from branching evolution. Currently available methods allow for a more comprehensive and holistic analysis of tumor heterogeneity in that issues associated with spatial and temporal heterogeneity can now be properly addressed. This review provides some background regarding intratumoral heterogeneity and how it leads to incomplete molecular response to targeted therapies, and proposes the use of single-cell methods, sequential liquid biopsy, and multiregion sequencing to discover the link between intratumoral heterogeneity and early adaptive drug resistance. In summary, minimal residual disease as a result of intratumoral heterogeneity is the earliest form of acquired drug resistance. Emerging technologies such as liquid biopsy and single-cell methods allow for studying targetable drivers of minimal residual disease and contribute to preemptive combinatorial targeting of both drivers of the tumor and its minimal residual disease cells.
Collapse
Affiliation(s)
- Zuan-Fu Lim
- WVU Cancer Institute, West Virginia University, Morgantown, WV, 26506, USA.,Cancer Cell Biology Program, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV, 26506, USA.,Penn State Cancer Institute, Penn State Health Milton S. Hershey Medical Center, Penn State University, P.O. Box 850, Mail Code CH46, 500 University Drive, Hershey, PA, 17033-0850, USA
| | - Patrick C Ma
- Penn State Cancer Institute, Penn State Health Milton S. Hershey Medical Center, Penn State University, P.O. Box 850, Mail Code CH46, 500 University Drive, Hershey, PA, 17033-0850, USA.
| |
Collapse
|
263
|
Blakely CM, Riess JW. Interpretation of ceritinib clinical trial results and future combination therapy strategies for ALK-rearranged NSCLC. Expert Rev Anticancer Ther 2019; 19:1061-1075. [PMID: 31809604 DOI: 10.1080/14737140.2019.1699792] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Introduction: Lung cancer is the leading cause of cancer-related deaths, with non-small cell lung cancer (NSCLC) accounting for approximately 85% of all lung cancer cases. The continued advancement of DNA sequencing technology and the discovery of multiple specific driver mutations underlying many cases of NSCLC are moving clinical intervention toward a more targeted approach. Here we focus on anaplastic lymphoma kinase (ALK), a member of the receptor tyrosine kinase family, as an oncogenic driver in NSCLC. The ALK gene is rearranged in 3-7% of NSCLCs, and targeted inhibition of ALK is a viable therapy option.Areas covered: We discuss the available treatment options for ALK-positive NSCLC with an emphasis on the second-generation ALK inhibitor ceritinib. We also discuss practical treatment strategies and possible strategies to overcome or delay resistance to ALK inhibitors.Expert opinion: With a robust treatment armamentarium for patients with ALK-positive NSCLC, emphasis has shifted to optimizing individualized treatment strategies to further enhance outcomes for these patients.
Collapse
Affiliation(s)
- Collin M Blakely
- Department of Medicine, UCSF Helen Diller Comprehensive Cancer Center, San Francisco, CA, USA
| | - Jonathan W Riess
- Department of Medicine, UC Davis Comprehensive Cancer Center, Sacramento, CA, USA
| |
Collapse
|
264
|
Spagnuolo A, Muto M, Monaco F, Colantuoni G, Gridelli C. The optional approach of oncogene-addicted non-small cell lung cancer with brain metastases in the new generation targeted therapies era. Transl Lung Cancer Res 2019; 8:1134-1151. [PMID: 32010591 PMCID: PMC6976372 DOI: 10.21037/tlcr.2019.12.24] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 11/13/2019] [Indexed: 12/11/2022]
Abstract
In recent years, the study of the molecular characteristics of non-small cell lung cancer (NSCLC) has highlighted a specific role of some genes that represent important therapeutic targets, including epidermal growth factor receptor (EGFR), anaplastic lymphoma kinase (ALK), c-ros oncogene 1 (ROS-1) and v-Raf murine sarcoma viral oncogene homolog B1 (BRAF). Patients with oncogene-addicted cancer benefit more from therapy with tyrosine kinase inhibitors (TKIs) than from chemotherapy. The brain is a preferred site for tumor spread in these patients. In addition, given greater control of extracranial disease and prolonged survival, the brain is often the first site of progression. Therefore, there is great interest in therapeutic approaches that optimize the control of intracranial disease associated with systemic drugs that, by penetrating the blood-brain barrier (BBB), may improve local control. On the latter, radiotherapy provides excellent efficacy but following the results of clinical trials with new brain penetrant drugs, the question of how and especially when to perform brain radiotherapy in patients with oncogene-addicted NSCLC remains open. Prospective studies may indicate which patients are most likely to benefit from combined use or in what sequence they will undergo systemic and radiotherapy treatment. Due to the heterogeneity of patients and the introduction of new generation TKIs, a multidisciplinary assessment for the best management of therapies in NSCLC patients with molecular driver alterations and brain metastases (BM) is required.
Collapse
Affiliation(s)
- Alessia Spagnuolo
- Division of Medical Oncology, ‘S. G. Moscati’ Hospital, Avellino, Italy
| | - Matteo Muto
- Division of Radiotherapy, ‘S. G. Moscati’ Hospital, Avellino, Italy
| | - Fabio Monaco
- Division of Radiation Protection, ‘S. G. Moscati’ Hospital, Avellino, Italy
| | | | - Cesare Gridelli
- Division of Medical Oncology, ‘S. G. Moscati’ Hospital, Avellino, Italy
| |
Collapse
|
265
|
Mogenet A, Tomasini P, Greillier L, Barlesi F. Lorlatinib: an additional option for ALK-positive non-small cell lung cancer? Transl Lung Cancer Res 2019; 8:S383-S386. [PMID: 32038920 DOI: 10.21037/tlcr.2019.05.10] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Affiliation(s)
- Alice Mogenet
- Department of Multidisciplinary Oncology & Therapeutic Innovations, Aix Marseille University, CNRS, INSERM, CRCM, APHM, Marseille, France
| | - Pascale Tomasini
- Department of Multidisciplinary Oncology & Therapeutic Innovations, Aix Marseille University, CNRS, INSERM, CRCM, APHM, Marseille, France
| | - Laurent Greillier
- Department of Multidisciplinary Oncology & Therapeutic Innovations, Aix Marseille University, CNRS, INSERM, CRCM, APHM, Marseille, France
| | - Fabrice Barlesi
- Department of Multidisciplinary Oncology & Therapeutic Innovations, Aix Marseille University, CNRS, INSERM, CRCM, APHM, Marseille, France
| |
Collapse
|
266
|
Oh CY, Klatt MG, Bourne C, Dao T, Dacek MM, Brea EJ, Mun SS, Chang AY, Korontsvit T, Scheinberg DA. ALK and RET Inhibitors Promote HLA Class I Antigen Presentation and Unmask New Antigens within the Tumor Immunopeptidome. Cancer Immunol Res 2019; 7:1984-1997. [PMID: 31540894 PMCID: PMC6891198 DOI: 10.1158/2326-6066.cir-19-0056] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 04/18/2019] [Accepted: 09/16/2019] [Indexed: 12/13/2022]
Abstract
T-cell immunotherapies are often thwarted by the limited presentation of tumor-specific antigens abetted by the downregulation of human leukocyte antigen (HLA). We showed that drugs inhibiting ALK and RET produced dose-related increases in cell-surface HLA in tumor cells bearing these mutated kinases in vitro and in vivo, as well as elevated transcript and protein expression of HLA and other antigen-processing machinery. Subsequent analysis of HLA-presented peptides after ALK and RET inhibitor treatment identified large changes in the immunopeptidome with the appearance of hundreds of new antigens, including T-cell epitopes associated with impaired peptide processing (TEIPP) peptides. ALK inhibition additionally decreased PD-L1 levels by 75%. Therefore, these oncogenes may enhance cancer formation by allowing tumors to evade the immune system by downregulating HLA expression. Altogether, RET and ALK inhibitors could enhance T-cell-based immunotherapies by upregulating HLA, decreasing checkpoint blockade ligands, and revealing new, immunogenic, cancer-associated antigens.
Collapse
Affiliation(s)
- Claire Y Oh
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Weill Cornell Medicine, New York, New York
| | - Martin G Klatt
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Christopher Bourne
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Weill Cornell Medicine, New York, New York
| | - Tao Dao
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Megan M Dacek
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Weill Cornell Medicine, New York, New York
| | - Elliott J Brea
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Weill Cornell Medicine, New York, New York
| | - Sung Soo Mun
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Aaron Y Chang
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Weill Cornell Medicine, New York, New York
| | - Tatyana Korontsvit
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - David A Scheinberg
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York.
- Weill Cornell Medicine, New York, New York
| |
Collapse
|
267
|
Petrek H, Yu A. MicroRNAs in non-small cell lung cancer: Gene regulation, impact on cancer cellular processes, and therapeutic potential. Pharmacol Res Perspect 2019; 7:e00528. [PMID: 31859460 PMCID: PMC6923806 DOI: 10.1002/prp2.528] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 09/16/2019] [Accepted: 09/18/2019] [Indexed: 12/29/2022] Open
Abstract
Lung cancer remains the most lethal cancer among men and women in the United States and worldwide. The majority of lung cancer cases are classified as non-small cell lung cancer (NSCLC). Developing new therapeutics on the basis of better understanding of NSCLC biology is critical to improve the treatment of NSCLC. MicroRNAs (miRNAs or miRs) are a superfamily of genome-derived, small noncoding RNAs that govern posttranscriptional gene expression in cells. Functional miRNAs are commonly dysregulated in NSCLC, caused by genomic deletion, methylation, or altered processing, which may lead to the changes of many cancer-related pathways and processes, such as growth and death signaling, metabolism, angiogenesis, cell cycle, and epithelial to mesenchymal transition, as well as sensitivity to current therapies. With the understanding of miRNA biology in NSCLC, there are growing interests in developing new therapeutic strategies, namely restoration of tumor suppressive miRNAs and inhibition of tumor promotive miRNAs, to combat against NSCLC. In this article, we provide an overview on the molecular features of NSCLC and current treatment options with a focus on pharmacotherapy and personalized medicine. By illustrating the roles of miRNAs in the control of NSCLC tumorigenesis and progression, we highlight the latest efforts in assessing miRNA-based therapies in animal models and discuss some critical challenges in developing RNA therapeutics.
Collapse
Affiliation(s)
- Hannah Petrek
- Department of Biochemistry & Molecular MedicineUC Davis School of MedicineSacramentoCAUSA
| | - Ai‐Ming Yu
- Department of Biochemistry & Molecular MedicineUC Davis School of MedicineSacramentoCAUSA
| |
Collapse
|
268
|
Metabolic Remodelling: An Accomplice for New Therapeutic Strategies to Fight Lung Cancer. Antioxidants (Basel) 2019; 8:antiox8120603. [PMID: 31795465 PMCID: PMC6943435 DOI: 10.3390/antiox8120603] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 11/22/2019] [Accepted: 11/27/2019] [Indexed: 12/12/2022] Open
Abstract
Metabolic remodelling is a hallmark of cancer, however little has been unravelled in its role in chemoresistance, which is a major hurdle to cancer control. Lung cancer is a leading cause of death by cancer, mainly due to the diagnosis at an advanced stage and to the development of resistance to therapy. Targeted therapeutic agents combined with comprehensive drugs are commonly used to treat lung cancer. However, resistance mechanisms are difficult to avoid. In this review, we will address some of those therapeutic regimens, resistance mechanisms that are eventually developed by lung cancer cells, metabolic alterations that have already been described in lung cancer and putative new therapeutic strategies, and the integration of conventional drugs and genetic and metabolic-targeted therapies. The oxidative stress is pivotal in this whole network. A better understanding of cancer cell metabolism and molecular adaptations underlying resistance mechanisms will provide clues to design new therapeutic strategies, including the combination of chemotherapeutic and targeted agents, considering metabolic intervenients. As cancer cells undergo a constant metabolic adaptive drift, therapeutic regimens must constantly adapt.
Collapse
|
269
|
Huber RM, Hansen KH, Paz-Ares Rodríguez L, West HL, Reckamp KL, Leighl NB, Tiseo M, Smit EF, Kim DW, Gettinger SN, Hochmair MJ, Kim SW, Langer CJ, Ahn MJ, Kim ES, Kerstein D, Groen HJM, Camidge DR. Brigatinib in Crizotinib-Refractory ALK+ NSCLC: 2-Year Follow-up on Systemic and Intracranial Outcomes in the Phase 2 ALTA Trial. J Thorac Oncol 2019; 15:404-415. [PMID: 31756496 DOI: 10.1016/j.jtho.2019.11.004] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 11/09/2019] [Indexed: 10/25/2022]
Abstract
INTRODUCTION We report updated data from a phase 2 randomized study evaluating brigatinib in crizotinib-refractory anaplastic lymphoma kinase-positive NSCLC. METHODS Patients were randomized 1:1 to take either oral brigatinib 90 mg once daily (arm A) or 180 mg once daily with a 7-day lead-in at 90 mg (arm B), stratified by central nervous system (CNS) metastases and best response to crizotinib. The primary end point was investigator-assessed confirmed objective response rate per Response Evaluation Criteria in Solid Tumors version 1.1. Secondary end points included independent review committee (IRC)-assessed progression-free survival (PFS), intracranial PFS (iPFS), and overall survival (OS). Exploratory analyses included CNS versus ex-CNS target lesion response and correlation of depth of response with PFS and OS. RESULTS Among 222 randomized patients (112 and 110 in arms A and B, respectively), 59 (27%) remained on brigatinib at analysis (median follow-up: 19.6 versus 24.3 months). At baseline, 71% and 67% had brain lesions among A and B arms, respectively. Investigator-assessed confirmed objective response rate was 46% versus 56%. Median IRC-assessed PFS was 9.2 months (95% confidence interval: 7.4-12.8) versus 16.7 months (11.6-21.4). Median OS was 29.5 months (18.2-not reached) versus 34.1 months (27.7-not reached). IRC-confirmed intracranial objective response rate in patients with measurable baseline brain lesions was 50% (13 of 26) versus 67% (12 of 18); median duration of intracranial response was 9.4 versus 16.6 months. IRC-assessed iPFS was 12.8 versus 18.4 months. Across arms, median IRC-assessed PFS was 1.9, 5.5, 11.1, 16.7, and 15.6 months for patients with no, 1%-25%, 26%-50%, 51%-75%, and 76%-100% target lesion shrinkage, respectively. No new safety findings were observed with longer follow-up. CONCLUSIONS Brigatinib (180 mg once daily with lead-in) continues to demonstrate robust PFS, long iPFS and duration of intracranial response, and high intracranial objective response rate in crizotinib-refractory patients. Depth of response may be an important end point to capture in future targeted therapy trials.
Collapse
Affiliation(s)
- Rudolf M Huber
- Division of Respiratory Medicine and Thoracic Oncology, Department of Medicine V, University Hospital of Munich, Thoracic Oncology Centre Munich, German Centre for Lung Research, Munich, Bavaria, Germany.
| | - Karin H Hansen
- Department of Clinical Oncology, Odense University Hospital, Odense, Denmark
| | | | - Howard L West
- Thoracic Oncology Program, Swedish Cancer Institute, Seattle, Washington
| | - Karen L Reckamp
- Department of Medical Oncology & Therapeutics Research, City of Hope National Medical Center, Duarte, California
| | - Natasha B Leighl
- Department of Medical Oncology, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Marcello Tiseo
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - Egbert F Smit
- Department of Pulmonary Diseases, VU University Medical Center, Amsterdam, the Netherlands
| | - Dong-Wan Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea
| | | | - Maximilian J Hochmair
- Department of Respiratory and Critical Care Medicine, Karl Landsteiner Institute of Lung Research and Pulmonary Oncology, Vienna, Austria
| | - Sang-We Kim
- Department of Oncology, Asan Medical Center, Seoul, South Korea
| | - Corey J Langer
- University of Pennsylvania Abramson Cancer Center, Philadelphia, Pennsylvania
| | - Myung-Ju Ahn
- Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Edward S Kim
- Levine Cancer Institute, Atrium Health, Charlotte, North Carolina
| | - David Kerstein
- Millennium Pharmaceuticals, Inc. (wholly owned subsidiary of Takeda Pharmaceutical Company Limited), Cambridge, Massachusetts
| | - Harry J M Groen
- University of Groningen and University Medical Center Groningen, Groningen, the Netherlands
| | | |
Collapse
|
270
|
Synthetic lethality as an engine for cancer drug target discovery. Nat Rev Drug Discov 2019; 19:23-38. [DOI: 10.1038/s41573-019-0046-z] [Citation(s) in RCA: 311] [Impact Index Per Article: 51.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/12/2019] [Indexed: 12/25/2022]
|
271
|
Tiotiu A, Billon Y, Vaillant P, Menard O, Hofman P, Mascaux C. [Therapeutic strategies in advanced ALK positive non-small cell lung cancer]. Rev Mal Respir 2019; 36:1107-1116. [PMID: 31727555 DOI: 10.1016/j.rmr.2019.02.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 02/19/2019] [Indexed: 11/30/2022]
Abstract
Anaplastic lymphoma kinase (ALK) rearrangement is a therapeutically targetable oncogenic driver found in 5% of patients with non-small-cell lung cancer (NSCLC). The objective of this paper is to synthesise current knowledge on ALK rearrangement and its impact on the management of advanced NSCLC. Several inhibitors of the tyrosine kinase of ALK (crizotinib, ceritinib, alectinib) have been approved as first line therapies in patients with advanced ALK positive NSCLC, which are associated with a better median progression-free survival than conventional chemotherapy. Unfortunately, the emergence of drug resistance leads to tumor progression. In patients with oligoprogressive disease if local ablative therapy can be effected, continuing with the same ALK tyrosine kinase inhibitor is one option. In patients with progression, clinicians may consider switching to another therapy. Rebiopsy of the tumor or liquid biopsy could be attempted to identify the mechanisms of resistance and to customize ALK-target therapy. The emergence of crizotinib drug resistance has prompted the development of next generation drugs including ceritinb, alectinib, brigatinib and lorlatinib. The ability to quickly develop targeted therapies against specific oncogenic drivers will require close co-operation between pathologists, pulmonologists and oncologists in the future to keep pace with drug discoveries and to define optimal therapeutic strategies.
Collapse
Affiliation(s)
- A Tiotiu
- Département de pneumologie, CHRU Nancy site Brabois, bâtiment de spécialités médicales, 9, rue du Morvan, 54511 Vandœuvre-lès-Nancy, France.
| | - Y Billon
- Département de pneumologie, CHRU Nancy site Brabois, bâtiment de spécialités médicales, 9, rue du Morvan, 54511 Vandœuvre-lès-Nancy, France
| | - P Vaillant
- Département de pneumologie, CHRU Nancy site Brabois, bâtiment de spécialités médicales, 9, rue du Morvan, 54511 Vandœuvre-lès-Nancy, France
| | - O Menard
- Département de pneumologie, CHRU Nancy site Brabois, bâtiment de spécialités médicales, 9, rue du Morvan, 54511 Vandœuvre-lès-Nancy, France
| | - P Hofman
- Laboratoire de pathologie clinique et expérimentale, CHU de Nice, 06000 Nice, France; Centre IRCAN, Inserm U1081 et CNRS/UMR 7284, Centre de lutte contre le cancer Antoine-Lacassagne, 06000 Nice, France; Université Côte d'Azur, 06000 Nice, France
| | - C Mascaux
- Département d'oncologie multidisciplinaire et Innovations thérapeutiques, Assistance Publique-Hôpitaux de Marseille, 13354 Marseille, France; Aix Marseille université, CNRS, Inserm, CRCM, 13354 Marseille, France
| |
Collapse
|
272
|
Liu WJ, Du Y, Wen R, Yang M, Xu J. Drug resistance to targeted therapeutic strategies in non-small cell lung cancer. Pharmacol Ther 2019; 206:107438. [PMID: 31715289 DOI: 10.1016/j.pharmthera.2019.107438] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 11/06/2019] [Indexed: 02/07/2023]
Abstract
Rapidly developing molecular biology techniques have been employed to identify cancer driver genes in specimens from patients with non-small cell lung cancer (NSCLC). Inhibitors and antibodies that specifically target driver gene-mediated signaling pathways to suppress tumor growth and progression are expected to extend the survival time and further improve the quality of life of patients. However, the health of patients with advanced and metastatic NSCLC presents significant challenges due to treatment resistance, mediated by cancer driver gene alteration, epigenetic alteration, and tumor heterogeneity. In this review, we discuss two different resistance mechanisms in NSCLC targeted therapies, namely changes in the targeted oncogenes (on-target resistance) and changes in other related signaling pathways (off-target resistance) in tumor cells. We highlight the conventional mechanisms of drug resistance elicited by the complex heterogeneous microenvironment of NSCLC during targeted therapy, including mutations in epidermal growth factor receptor (EGFR), anaplastic lymphoma kinase (ALK), the receptor tyrosine kinase ROS proto-oncogene 1 (ROS1), and the serine/threonine-protein kinase BRAF (v-Raf murine sarcoma viral oncogene homolog B). We also discuss the mechanism of action of less common oncoproteins, as in-depth understanding of these molecular mechanisms is important for optimizing treatment strategies.
Collapse
Affiliation(s)
- Wen-Juan Liu
- Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, China
| | - Yue Du
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Ru Wen
- Department of Medicine, Stanford University School of Medicine, California, USA
| | - Ming Yang
- Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, China.
| | - Jian Xu
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA.
| |
Collapse
|
273
|
Schabath MB, Dalvi TB, Dai HA, Crim AL, Midha A, Shire N, Gimbrone NT, Walker J, Greenawalt DM, Lawrence D, Rigas JR, Brody R, Potter D, Kumar NS, Huntsman SA, Gray JE. A Molecular Epidemiological Analysis Of Programmed Cell Death Ligand-1 (PD-L1) Protein Expression, Mutations And Survival In Non-Small Cell Lung Cancer. Cancer Manag Res 2019; 11:9469-9481. [PMID: 31819612 PMCID: PMC6844199 DOI: 10.2147/cmar.s218635] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 10/18/2019] [Indexed: 12/17/2022] Open
Abstract
Purpose To characterize programmed cell death ligand-1 (PD-L1) expression in relation to survival and gene mutation status in patients with advanced NSCLC. The study also explored the influence of tumor mutational burden (TMB) on PD-L1 expression and patient characteristics. Patients and methods Adult patients with histologically or cytologically documented Stage IIIB/Stage IV/recurrent/progressive NSCLC, Eastern Cooperative Oncology Group performance status 0 to 3, and >2 lines of prior systemic treatment regimens were included in this retrospective analysis. Patients were treated from 1997 to 2015 at H. Lee Moffitt Cancer Center and Research Institute, Tampa, or at 7 community centers across the United States. PD-L1 expression level was determined using the VENTANA PD-L1 (SP263) Assay. EGFR and KRAS mutation status and ALK rearrangements were determined by targeted DNA sequencing; these were obtained from clinical records where targeted DNA sequencing was not performed. TMB was calculated as the total number of somatic mutations per sample. Results From a total of 136 patients included in the study, 23.5% had tumors with high PD-L1 expression (≥25%). There were no significant differences in patient characteristics, overall survival (OS), and progression-free survival (PFS) between patients with high PD-L1 expression (median OS: 39.5 months; median PFS: 15.8 months) vs low PD-L1 expression (<25%; median OS: 38.1 months; median PFS: 18.6 months). PD-L1 expression level correlated (P=0.05) with TMB and was consistent with The Cancer Genome Atlas data. Conclusion In this retrospective analysis, survival outcomes of patients with advanced NSCLC were comparable by PD-L1 expression level. EGFR and KRAS mutation status were not found to be significantly associated with PD-L1 expression level, while TMB was weakly associated with PD-L1 expression level. Overall, PD-L1 expression level was not observed to be an independent prognostic biomarker in this cohort of patients with advanced NSCLC treated with chemotherapy.
Collapse
Affiliation(s)
- Matthew B Schabath
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA.,Department of Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | | | | | | | - Anita Midha
- Department of Personalised Healthcare and Biomarkers, AstraZeneca, Cambridge, UK
| | - Norah Shire
- Oncology R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Nicholas T Gimbrone
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Jill Walker
- Department of Precision Medicine Oncology, AstraZeneca, Cambridge, UK
| | | | - David Lawrence
- Department of Global Medicines Development, AstraZeneca, Cambridge, UK
| | - James R Rigas
- Department of Global Medical Affairs Oncology, AstraZeneca, Gaithersburg, MD, USA
| | - Robert Brody
- Department of Global Medical Affairs Oncology, AstraZeneca, Gaithersburg, MD, USA
| | - Danielle Potter
- Department of Global Medical Affairs Oncology, AstraZeneca, Gaithersburg, MD, USA
| | | | | | - Jhanelle E Gray
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| |
Collapse
|
274
|
A novel ALK inhibitor ZYY inhibits Karpas299 cell growth in vitro and in a mouse xenograft model and induces protective autophagy. Toxicol Appl Pharmacol 2019; 383:114781. [DOI: 10.1016/j.taap.2019.114781] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 10/09/2019] [Accepted: 10/10/2019] [Indexed: 12/18/2022]
|
275
|
Häckl S, Koch A, Lasch F. Empirical evaluation of the implementation of the EMA guideline on missing data in confirmatory clinical trials: Specification of mixed models for longitudinal data in study protocols. Pharm Stat 2019; 18:636-644. [PMID: 31267673 PMCID: PMC6899721 DOI: 10.1002/pst.1964] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 05/24/2019] [Accepted: 06/03/2019] [Indexed: 12/26/2022]
Abstract
In confirmatory clinical trials, the prespecification of the primary analysis model is a universally accepted scientific principle to allow strict control of the type I error. Consequently, both the ICH E9 guideline and the European Medicines Agency (EMA) guideline on missing data in confirmatory clinical trials require that the primary analysis model is defined unambiguously. This requirement applies to mixed models for longitudinal data handling missing data implicitly. To evaluate the compliance with the EMA guideline, we evaluated the model specifications in those clinical study protocols from development phases II and III submitted between 2015 and 2018 to the Ethics Committee at Hannover Medical School under the German Medicinal Products Act, which planned to use a mixed model for longitudinal data in the confirmatory testing strategy. Overall, 39 trials from different types of sponsors and a wide range of therapeutic areas were evaluated. While nearly all protocols specify the fixed and random effects of the analysis model (95%), only 77% give the structure of the covariance matrix used for modeling the repeated measurements. Moreover, the testing method (36%), the estimation method (28%), the computation method (3%), and the fallback strategy (18%) are given by less than half the study protocols. Subgroup analyses indicate that these findings are universal and not specific to clinical trial phases or size of company. Altogether, our results show that guideline compliance is to various degrees poor and consequently, strict type I error rate control at the intended level is not guaranteed.
Collapse
Affiliation(s)
- Sebastian Häckl
- Institute of BiostatisticsHannover Medical SchoolHannoverGermany
| | - Armin Koch
- Institute of BiostatisticsHannover Medical SchoolHannoverGermany
| | - Florian Lasch
- Institute of BiostatisticsHannover Medical SchoolHannoverGermany
| |
Collapse
|
276
|
Medikamentöse Therapie des metastasierten nicht-kleinzelligen Lungenkarzinoms. DER PNEUMOLOGE 2019. [DOI: 10.1007/s10405-019-00284-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
277
|
Yang J, Dong L, Du H, Li XB, Liang YX, Liu GR. ALK-TPM3 rearrangement in adult renal cell carcinoma: a case report and literature review. Diagn Pathol 2019; 14:112. [PMID: 31627758 PMCID: PMC6798478 DOI: 10.1186/s13000-019-0879-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Accepted: 08/29/2019] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Translocation-associated renal cell carcinoma involving ALK (ALK-tRCC) is a rare subtype of adult renal cell carcinoma (RCC) reported in recent years. It was recognized as a group of emerging /provisional RCC in the latest World Health Organization's classification (2016). CASE PRESENTATION A new Chinese case of ALK-tRCC was reported. The patient was a 58-year-old man with a tumor in kidney. The tumor was composed of sheets of large cells with abundant eosinophilic cytoplasm and indistinct cell borders but conspicuous intracytoplasmic vacuoles. The nuclei were enlarged with a nucleolar of grade 4. Immunohistochemically, tumor cells were diffusely positive for PAX8, keratin (AE1/AE3), epithelial membrane antigen (EMA) and CK7. Fluorescent in situ hybridization (FISH) showed a rearrangement of ALK in tumor cells. CONCLUSION ALK-tRCC is a rare subtype of adult RCC. Its diagnosis is very difficult because the histological spectrum is very wide. We suggested that RCCs should be screened for ALK expression by immunohistochemistry (IHC) for the patient might benefit from ALK inhibitors therapy.
Collapse
Affiliation(s)
- Jing Yang
- Department of pathology, Guangzhou first people's hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, Guangdong, China.
| | - Lei Dong
- Department of pathology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Hong Du
- Department of pathology, Guangzhou first people's hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, Guangdong, China
| | - Xiu-Bo Li
- Department of pathology, Guangzhou first people's hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, Guangdong, China
| | - Yan-Xiao Liang
- Department of pathology, Guangzhou first people's hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, Guangdong, China
| | - Guo-Rong Liu
- Department of pathology, Guangzhou first people's hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, Guangdong, China
| |
Collapse
|
278
|
Yang Y, Zhou J, Zhou J, Feng J, Zhuang W, Chen J, Zhao J, Zhong W, Zhao Y, Zhang Y, Song Y, Hu Y, Yu Z, Gong Y, Chen Y, Ye F, Zhang S, Cao L, Fan Y, Wu G, Guo Y, Zhou C, Ma K, Fang J, Feng W, Liu Y, Zheng Z, Li G, Wu N, Song W, Liu X, Zhao S, Ding L, Mao L, Selvaggi G, Yuan X, Fu Y, Wang T, Xiao S, Zhang L. Efficacy, safety, and biomarker analysis of ensartinib in crizotinib-resistant, ALK-positive non-small-cell lung cancer: a multicentre, phase 2 trial. THE LANCET RESPIRATORY MEDICINE 2019; 8:45-53. [PMID: 31628085 DOI: 10.1016/s2213-2600(19)30252-8] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 06/18/2019] [Accepted: 06/18/2019] [Indexed: 02/05/2023]
Abstract
BACKGROUND Ensartinib is a potent new-generation ALK inhibitor with high activity against a broad range of known crizotinib-resistant ALK mutations and CNS metastases. We aimed to assess the efficacy and safety of ensartinib in ALK-positive patients with non-small-cell lung cancer (NSCLC), in whom crizotinib therapy was unsuccessful. The associations between ensartinib efficacy and crizotinib-resistant mutations were also explored. METHODS We did a single-arm, open-label, phase 2 study at 27 centres in China. Patients were aged 18 years or older, had stage IIIb or stage IV ALK-positive NSCLC that had progressed while they were on crizotinib therapy, an Eastern Cooperative Oncology Group performance status of 2 or less, had measurable disease, and had received fewer than three previous treatments. Patients with CNS metastases were included if these metastases were asymptomatic and did not require steroid therapy. All patients received 225 mg ensartinib orally once daily on a continuous dosing schedule. The primary outcome was the proportion of patients with an objective response according to the Response Evaluation Criteria in Solid Tumors (version 1.1), as assessed by an independent review committee in all patients who received at least one dose of ensartinib with no major violations of the inclusion criteria (ie, the full analysis set). Safety was assessed in all enrolled patients who received at least one dose of ensartinib. This trial was registered with ClinicalTrials.gov, NCT03215693. FINDINGS Between Sept 28, 2017, and April 11, 2018, 160 patients were enrolled and had at least one dose of ensartinib (safety analysis set). Four patients had inclusion violations and were excluded from the efficacy analysis, which thus included 156 patients (full analysis set). 97 (62%) patients in the full analysis set had brain metastases. 76 (52% [95% CI 43-60]) of 147 patients in the full analysis set, with responses that could be assessed by the independent review committee, had an objective response. 28 (70% [53-83]) of 40 patients with measurable brain metastases as assessed by the independent review committee had an intracranial objective response. 145 (91%) of 160 patients had at least one treatment-related adverse event, which were mostly grade 1 or 2. The most common treatment-related adverse events were rash (89 [56%]), increased alanine aminotransferase concentrations (74 [46%]), and increased aspartate aminotransferase concentrations (65 [41%]). INTERPRETATION Ensartinib has activity and is well tolerated in patients with crizotinib-refractory, ALK-positive NSCLC, including those with brain metastases. The role of ensartinib in patients in whom other second-generation ALK inhibitors have been unsuccessful warrants further studies. FUNDING Betta Pharmaceuticals.
Collapse
Affiliation(s)
- Yunpeng Yang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Jianya Zhou
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Jianying Zhou
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Jifeng Feng
- Department of Medical Oncology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Wu Zhuang
- Department of Thoracic Oncology, Fujian Provincial Cancer Hospital, Fujian Medical University Cancer Hospital, Fuzhou, China
| | - Jianhua Chen
- Department of Medical Oncology-Chest, Hunan Cancer Hospital, Changsha, China
| | - Jun Zhao
- Department of Thoracic Oncology, Beijing Cancer Hospital, Beijing, China
| | - Wei Zhong
- Department of Pulmonary Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Yanqiu Zhao
- Respiratory Department of Internal Medicine, Henan Provincial Cancer Hospital, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Yiping Zhang
- Thoracic Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Yong Song
- Division of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Yi Hu
- Department of Oncology, Chinese PLA General Hospital, Beijing, China
| | - Zhuang Yu
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Youling Gong
- Department of Thoracic Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yuan Chen
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Feng Ye
- Department of Medical Oncology, Cancer Hospital, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Teaching Hospital of Fujian Medical University, Xiamen, China
| | - Shucai Zhang
- Department of Medical Oncology, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Lejie Cao
- Respiratory Medicine, The First Affiliated Hospital of the University of Science and Technology of China, Anhui Provincial Hospital, Hefei, China
| | - Yun Fan
- Thoracic Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Gang Wu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yubiao Guo
- Pulmonary & Critical Care Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Chengzhi Zhou
- Respiratory Medicine Department, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Kewei Ma
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Jian Fang
- Department of Thoracic Oncology, Beijing Cancer Hospital, Beijing, China
| | - Weineng Feng
- Department of Head and Neck and Thoracic Medical Oncology, The First People's Hospital Of Foshan, Foshan, China
| | - Yunpeng Liu
- Oncology Medicine, The First Hospital of China Medical University, Shenyang, China
| | - Zhendong Zheng
- Oncology Department, General Hospital of Northern Theater Command, Shenyang, China
| | - Gaofeng Li
- 2nd Department of Thoracic Surgery, Yunnan Cancer Hospital, Kunming, China
| | - Ning Wu
- Department of Diagnostic Radiology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Wei Song
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Xiaoqing Liu
- Department of Pulmonary Oncology, The Fifth Medical Centre Chinese PLA General Hospital, Beijing, China
| | - Shijun Zhao
- Department of Diagnostic Radiology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | | | - Li Mao
- Betta Pharmaceuticals, Hangzhou, China
| | | | | | | | - Tao Wang
- Hangzhou Repugene Technology, Hangzhou, China
| | | | - Li Zhang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.
| |
Collapse
|
279
|
Hu J, Zhang W, Liu Y, Yang Y, Tan C, Wei X, Wang Y, Tan S, Liu M, Liu K, Liu Y, Zhang H, Xiao X. LDK
378 inhibits the recruitment of myeloid‐derived suppressor cells to spleen via the p38–
GRK
2–
CCR
2 pathway in mice with sepsis. Immunol Cell Biol 2019; 97:902-915. [DOI: 10.1111/imcb.12289] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 08/28/2019] [Accepted: 08/29/2019] [Indexed: 12/29/2022]
Affiliation(s)
- Jie Hu
- Department of Anesthesiology Xiangya Hospital Central South University Changsha Hunan China
- Sepsis Translational Medicine Key Lab of Hunan Province Central South University Changsha Hunan China
- Department of Pathophysiology Xiangya School of Medicine Central South University Changsha Hunan China
| | - Wenqin Zhang
- Sepsis Translational Medicine Key Lab of Hunan Province Central South University Changsha Hunan China
- Department of Pathophysiology Xiangya School of Medicine Central South University Changsha Hunan China
| | - Yanjuan Liu
- Sepsis Translational Medicine Key Lab of Hunan Province Central South University Changsha Hunan China
- Department of Pathophysiology Xiangya School of Medicine Central South University Changsha Hunan China
| | - Yang Yang
- Sepsis Translational Medicine Key Lab of Hunan Province Central South University Changsha Hunan China
- Department of Pathophysiology Xiangya School of Medicine Central South University Changsha Hunan China
| | - Chuyi Tan
- Sepsis Translational Medicine Key Lab of Hunan Province Central South University Changsha Hunan China
- Department of Pathophysiology Xiangya School of Medicine Central South University Changsha Hunan China
| | - Xue Wei
- Sepsis Translational Medicine Key Lab of Hunan Province Central South University Changsha Hunan China
- Department of Pathophysiology Xiangya School of Medicine Central South University Changsha Hunan China
| | - Yufang Wang
- Sepsis Translational Medicine Key Lab of Hunan Province Central South University Changsha Hunan China
- Department of Pathophysiology Xiangya School of Medicine Central South University Changsha Hunan China
| | - Sipin Tan
- Sepsis Translational Medicine Key Lab of Hunan Province Central South University Changsha Hunan China
- Department of Pathophysiology Xiangya School of Medicine Central South University Changsha Hunan China
| | - Meidong Liu
- Sepsis Translational Medicine Key Lab of Hunan Province Central South University Changsha Hunan China
- Department of Pathophysiology Xiangya School of Medicine Central South University Changsha Hunan China
| | - Ke Liu
- Sepsis Translational Medicine Key Lab of Hunan Province Central South University Changsha Hunan China
- Department of Pathophysiology Xiangya School of Medicine Central South University Changsha Hunan China
| | - Ying Liu
- Sepsis Translational Medicine Key Lab of Hunan Province Central South University Changsha Hunan China
- Department of Pathophysiology Xiangya School of Medicine Central South University Changsha Hunan China
| | - Huali Zhang
- Department of Anesthesiology Xiangya Hospital Central South University Changsha Hunan China
- Sepsis Translational Medicine Key Lab of Hunan Province Central South University Changsha Hunan China
- Department of Pathophysiology Xiangya School of Medicine Central South University Changsha Hunan China
| | - Xianzhong Xiao
- Sepsis Translational Medicine Key Lab of Hunan Province Central South University Changsha Hunan China
- Department of Pathophysiology Xiangya School of Medicine Central South University Changsha Hunan China
| |
Collapse
|
280
|
Kurzrock R, Gurski LA, Carlson RW, Ettinger DS, Horwitz SM, Kumar SK, Million L, von Mehren M, Benson AB. Level of evidence used in recommendations by the National Comprehensive Cancer Network (NCCN) guidelines beyond Food and Drug Administration approvals. Ann Oncol 2019; 30:1647-1652. [PMID: 31373348 PMCID: PMC6857604 DOI: 10.1093/annonc/mdz232] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND A previous analysis of 113 National Comprehensive Cancer Network® (NCCN®) recommendations reported that NCCN frequently recommends beyond Food and Drug Administration (FDA)-approved indications (44 off-label recommendations) and claimed that the evidence for these recommendations was weak. METHODS In order to determine the strength of the evidence, we carried out an in-depth re-analysis of the 44 off-label recommendations listed in the NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines®). RESULTS Of the 44 off-label recommendations, 14 were later approved by the FDA and/or are supported by randomized controlled trial (RCT) data. In addition, 13 recommendations were either very minor extrapolations from the FDA label (n = 8) or were actually on-label (n = 5). Of the 17 remaining extrapolations, 8 were for mechanism-based agents applied in rare cancers or subsets with few available treatment options (median response rate = 43%), 7 were based on non-RCT data showing significant efficacy (>50% response rates), and 2 were later removed from the NCCN Guidelines because newer therapies with better activity and/or safety became available. CONCLUSION Off-label drug use is a frequent component of care for patients with cancer in the United States. Our findings indicate that when the NCCN recommends beyond the FDA-approved indications, the strength of the evidence supporting such recommendations is robust, with a significant subset of these drugs later becoming FDA approved or supported by RCT. Recommendations without RCT data are often for mechanism-based drugs with high response rates in rare cancers or subsets without effective therapies.
Collapse
Affiliation(s)
- R Kurzrock
- Center for Personalized Cancer Therapy, University of California San Diego, Moores Cancer Center, San Diego, USA.
| | - L A Gurski
- National Comprehensive Cancer Network, Plymouth Meeting, Pennsylvania, USA
| | - R W Carlson
- National Comprehensive Cancer Network, Plymouth Meeting, Pennsylvania, USA; Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA; Stanford Cancer Institute, Stanford, California, USA
| | - D S Ettinger
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland, USA
| | - S M Horwitz
- Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - S K Kumar
- Mayo Clinic Cancer Center, Rochester, Minnesota, USA
| | - L Million
- Stanford Cancer Institute, Stanford, California, USA
| | - M von Mehren
- Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA
| | - A B Benson
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
281
|
Liu Y, Ye X, Yu Y, Lu S. Prognostic significance of anaplastic lymphoma kinase rearrangement in patients with completely resected lung adenocarcinoma. J Thorac Dis 2019; 11:4258-4270. [PMID: 31737311 DOI: 10.21037/jtd.2019.09.65] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Background Reports of the prognostic significance of anaplastic lymphoma kinase (ALK) rearrangement in early stage lung adenocarcinoma have been contradictory. This study aimed to identify the associations of ALK rearrangement with clinicopathologic features and prognosis in patients with surgically resected stage I-IIIA lung adenocarcinoma. Methods Analysis of ALK status was performed by a fully-automated immunochemistry assay (with rabbit monoclonal Ventana D5F3 antibody) in tissue sections of 2,103 patients with surgically-resected stage I-IIIA lung adenocarcinoma. ALK positive patients were matched with negative patients in a 1:1 ratio using propensity score matching (PSM). Clinical outcomes were assessed by disease-free survival (DFS) and overall survival (OS) after surgery. Initial recurrence pattern was also investigated according to ALK status. Results Among 2,103 stage I-IIIA lung adenocarcinoma cases, 81 (3.9%) were ALK positive. ALK positivity was significantly associated with younger age (P<0.001), solid predominant adenocarcinoma (P<0.001), variants of invasive adenocarcinoma (P<0.001), higher frequency of pleura invasion (P=0.040), smaller tumor size (P=0.014), mediastinal lymph node involvement (N2; P<0.001) and later pathologic stage (IIIA; P=0.001). In the match cohort, ALK positivity was not associated with DFS [hazard ratio (HR), 0.58; 95% confidence interval (CI): 0.33-1.03, P=0.063] or OS (HR, 0.61; 95% CI: 0.22-1.67, P=0.334). Lymph node involvement (HR: 5.36, 95% CI, 3.01-9.65, P<0.001) and solid predominant adenocarcinoma subtype (HR, 2.02; 95% CI: 1.07-3.79; P=0.029) were the independent prognostic factors of inferior DFS, and lymph node involvement was the independent prognostic factors of worse OS (HR, 6.61; 95% CI: 2.43-17.94; P<0.001). ALK positive patients had a higher risk of developing tumor recurrence in liver (P=0.043). Conclusions ALK rearrangement was not an independent prognostic factor in stage I-IIIA lung adenocarcinoma patients but leaded to a higher risk of developing recurrence in liver.
Collapse
Affiliation(s)
- Yinglei Liu
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Xiangyun Ye
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Yongfeng Yu
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Shun Lu
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| |
Collapse
|
282
|
O'Bryant SE, Zhang F, Johnson LA, Hall J, Edwards M, Grammas P, Oh E, Lyketsos CG, Rissman RA. A Precision Medicine Model for Targeted NSAID Therapy in Alzheimer's Disease. J Alzheimers Dis 2019; 66:97-104. [PMID: 30198872 DOI: 10.3233/jad-180619] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND To date, the therapeutic paradigm for Alzheimer's disease (AD) has focused on a single intervention for all patients. However, a large literature in oncology supports the therapeutic benefits of a precision medicine approach to therapy. Here we test a precision-medicine approach to AD therapy. OBJECTIVE To determine if a baseline, blood-based proteomic companion diagnostic predicts response to NSAID therapy. METHODS Proteomic assays of plasma from a multicenter, randomized, double-blind, placebo-controlled, parallel group trial, with 1-year exposure to rofecoxib (25 mg once daily), naproxen (220 mg twice-daily) or placebo. RESULTS 474 participants with mild-to-moderate AD were screened with 351 enrolled into the trial. Using support vector machine (SVM) analyses, 89% of the subjects randomized to either NSAID treatment arms were correctly classified using a general NSAID companion diagnostic. Drug-specific companion diagnostics yielded 98% theragnostic accuracy in the rofecoxib arm and 97% accuracy in the naproxen arm. CONCLUSION Inflammatory-based companion diagnostics have significant potential to identify select patients with AD who have a high likelihood of responding to NSAID therapy. This work provides empirical support for a precision medicine model approach to treating AD.
Collapse
Affiliation(s)
- Sid E O'Bryant
- Department of Pharmacology & Neuroscience; Institute for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Fan Zhang
- Vermont Genetics Network, University of Vermont, VT, USA
| | - Leigh A Johnson
- Department of Pharmacology & Neuroscience; Institute for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - James Hall
- Department of Pharmacology & Neuroscience; Institute for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX, USA
| | | | - Paula Grammas
- George & Anne Ryan Institute for Neuroscience, University of Rhode Island, RI, USA
| | - Esther Oh
- Department of Medicine, Johns Hopkins University, Baltimore, MD, USA.,Department of Psychiatry, Johns Hopkins University, Baltimore, MD, USA
| | | | - Robert A Rissman
- Department of Neurosciences, UCSD School of Medicine, La Jolla, CA, USA.,VA San Diego Healthcare System, San Diego, CA, USA
| |
Collapse
|
283
|
Kuo CN, Liao YM, Kuo LN, Tsai HJ, Chang WC, Yen Y. Cancers in Taiwan: Practical insight from epidemiology, treatments, biomarkers, and cost. J Formos Med Assoc 2019; 119:1731-1741. [PMID: 31522970 DOI: 10.1016/j.jfma.2019.08.023] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 07/25/2019] [Accepted: 08/23/2019] [Indexed: 12/20/2022] Open
Abstract
Cancer is the leading cause of death in Taiwan, and the overall incidence rate has gradually increased. The four most common cancers in Taiwan are colorectal, lung, breast and liver cancers. With the rise in incidence, the clinical use and costs of all anticancer drugs have steadily increased. The costs of novel therapeutics, such as targeted therapies and immunotherapy were accounted almost two-third of all antineoplastic agents in Taiwan. Moving forward, it will be necessary to discuss the economic impacts to clinical use of new therapeutics, while continuing to monitor and improve the quality of cancer therapy. In this review, we describe the epidemiology, disease screening policies and medication treatment policies for colorectal, lung, breast and liver cancer. We focus on the recent developments in cancer therapeutics, discuss the use of biomarkers, and finally consider the costs and the recent advances of anticancer medications in Taiwan.
Collapse
Affiliation(s)
- Chun-Nan Kuo
- Department of Clinical Pharmacy, School of Pharmacy, Taipei Medical University, Taipei, Taiwan; Department of Pharmacy, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Yu-Ming Liao
- Division of Hematology and Oncology, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Li-Na Kuo
- Department of Clinical Pharmacy, School of Pharmacy, Taipei Medical University, Taipei, Taiwan; Department of Pharmacy, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Hui-Ju Tsai
- Institute of Population Health Sciences, National Health Research Institutes, Miaoli, Taiwan; Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Wei-Chiao Chang
- Department of Clinical Pharmacy, School of Pharmacy, Taipei Medical University, Taipei, Taiwan; Department of Pharmacy, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; The PhD Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan; Master Program for Clinical Pharmacogenomics and Pharmacoproteomics, School of Pharmacy, Taipei Medical University, Taipei, Taiwan; Integrative Research Center for Critical Care, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.
| | - Yun Yen
- The PhD Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
284
|
Wang H, Wang H, Jia Y, Sun R, Hong W, Zhang M, Li Z. Visual Detection of Fusion Genes by Ligation-Triggered Isothermal Exponential Amplification: A Point-of-Care Testing Method for Highly Specific and Sensitive Quantitation of Fusion Genes with a Smartphone. Anal Chem 2019; 91:12428-12434. [PMID: 31464423 DOI: 10.1021/acs.analchem.9b03061] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Fusion genes, playing a causal role in human tumorigenesis and developments, are deemed as gold standard molecular biomarkers in cancer diagnosis, therapy, and prognosis. A rapid, robust, and sensitive method of detection of fusion genes for point-of-care (POC) diagnosis is urgently needed. Here, taking the advantages of the superior specificity of the ligation reaction and the highly amplified efficiency of isothermal exponential amplification with a pH indicator, we developed a colorimetric method for visual detection of fusion genes with high sensitivity and specificity by the naked eye. More importantly, we first found that fusion genes can be accurately quantified in a wide dynamic range (2 zmol to 2 fmol) by an open-source app with a smartphone-assisted RGB (red, green, and blue value) reading mode. The proposed method for Visual detection of Fusion genes by Ligation-triggered Isothermal Exponential Amplification is termed Vis-Fusion LIEXA. We have demonstrated that the Vis-Fusion LIEXA is a practical and reliable method for accurate quantitative detection of the fusion gene in a complex biological sample at zmol level in 40 min only with a smartphone, thereby providing a user-friendly and point-of-care testing (POCT) tool for molecular diagnostics.
Collapse
Affiliation(s)
- Hui Wang
- School of Chemistry and Biological Engineering , University of Science and Technology Beijing , 30 Xueyuan Road , Haidian District, Beijing 100083 , P. R. China
| | - Honghong Wang
- School of Chemistry and Biological Engineering , University of Science and Technology Beijing , 30 Xueyuan Road , Haidian District, Beijing 100083 , P. R. China
| | - Yuting Jia
- School of Chemistry and Biological Engineering , University of Science and Technology Beijing , 30 Xueyuan Road , Haidian District, Beijing 100083 , P. R. China
| | - Ruyan Sun
- School of Chemistry and Biological Engineering , University of Science and Technology Beijing , 30 Xueyuan Road , Haidian District, Beijing 100083 , P. R. China
| | - Weixiang Hong
- School of Chemistry and Biological Engineering , University of Science and Technology Beijing , 30 Xueyuan Road , Haidian District, Beijing 100083 , P. R. China
| | - Mai Zhang
- School of Chemistry and Biological Engineering , University of Science and Technology Beijing , 30 Xueyuan Road , Haidian District, Beijing 100083 , P. R. China
| | - Zhengping Li
- School of Chemistry and Biological Engineering , University of Science and Technology Beijing , 30 Xueyuan Road , Haidian District, Beijing 100083 , P. R. China
| |
Collapse
|
285
|
Adashek JJ, LoRusso PM, Hong DS, Kurzrock R. Phase I trials as valid therapeutic options for patients with cancer. Nat Rev Clin Oncol 2019; 16:773-778. [PMID: 31477881 DOI: 10.1038/s41571-019-0262-9] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/01/2019] [Indexed: 12/17/2022]
Abstract
For many years, oncology phase I trials have been referred to as 'toxicity trials' and have been believed to have low clinical utility other than that of establishing the adverse event profile of novel therapeutic agents. The traditional distinction of clinical trials into three phases has been challenged in the past few years by the introduction of targeted therapies and immunotherapies into the routine management of patients with cancer. This transformation has especially affected early phase trials, leading to the current situation in which response rates are increasingly reported from phase I trials. In this Perspectives, we highlight key elements of phase I trials and discuss how each one of them contributes to a new paradigm whereby preliminary measurements of the clinical benefit from a novel treatment can be obtained in current phase I trials, which can therefore be considered to have a therapeutic intent.
Collapse
Affiliation(s)
- Jacob J Adashek
- Department of Internal Medicine, University of South Florida, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | | | - David S Hong
- Department of Investigational Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Razelle Kurzrock
- Center for Personalized Cancer Therapy, Department of Medicine, University of California San Diego Moores Cancer Center, La Jolla, CA, USA.
| |
Collapse
|
286
|
Ceritinib-Induced Regression of an Insulin-Like Growth Factor-Driven Neuroepithelial Brain Tumor. Int J Mol Sci 2019; 20:ijms20174267. [PMID: 31480400 PMCID: PMC6747232 DOI: 10.3390/ijms20174267] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 08/15/2019] [Accepted: 08/28/2019] [Indexed: 12/25/2022] Open
Abstract
The insulin-like growth factor (IGF) pathway plays an important role in several brain tumor entities. However, the lack of inhibitors crossing the blood–brain barrier remains a significant obstacle for clinical translation. Here, we targeted the IGF pathway using ceritinib, an off-target inhibitor of the IGF1 receptor (IGF1R) and insulin receptor (INSR), in a pediatric patient with an unclassified brain tumor and a notch receptor 1 (NOTCH1) germline mutation. Pathway analysis of the tumor revealed activation of the sonic hedgehog (SHH), the wingless and integrated-1 (WNT), the IGF, and the Notch pathway. The proliferation of the patient tumor cells (225ZL) was inhibited by arsenic trioxide (ATO), which is an inhibitor of the SHH pathway, by linsitinib, which is an inhibitor of IGF1R and INSR, and by ceritinib. 225ZL expressed INSR but not IGF1R at the protein level, and ceritinib blocked the phosphorylation of INSR. Our first personalized treatment included ATO, but because of side effects, we switched to ceritinib. After 46 days, we achieved a concentration of 1.70 µM of ceritinib in the plasma, and after 58 days, MRI confirmed that there was a response to the treatment. Ceritinib accumulated in the tumor at a concentration of 2.72 µM. Our data suggest ceritinib as a promising drug for the treatment of IGF-driven brain tumors.
Collapse
|
287
|
Pailler E, Faugeroux V, Oulhen M, Mezquita L, Laporte M, Honoré A, Lecluse Y, Queffelec P, NgoCamus M, Nicotra C, Remon J, Lacroix L, Planchard D, Friboulet L, Besse B, Farace F. Acquired Resistance Mutations to ALK Inhibitors Identified by Single Circulating Tumor Cell Sequencing in ALK-Rearranged Non-Small-Cell Lung Cancer. Clin Cancer Res 2019; 25:6671-6682. [PMID: 31439588 DOI: 10.1158/1078-0432.ccr-19-1176] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 07/04/2019] [Accepted: 08/13/2019] [Indexed: 11/16/2022]
Abstract
PURPOSE Patients with anaplastic lymphoma kinase (ALK)-rearranged non-small-cell lung cancer (NSCLC) inevitably develop resistance to ALK inhibitors. New diagnostic strategies are needed to assess resistance mechanisms and provide patients with the most effective therapy. We asked whether single circulating tumor cell (CTC) sequencing can inform on resistance mutations to ALK inhibitors and underlying tumor heterogeneity in ALK-rearranged NSCLC. EXPERIMENTAL DESIGN Resistance mutations were investigated in CTCs isolated at the single-cell level from patients at disease progression on crizotinib (n = 14) or lorlatinib (n = 3). Three strategies including filter laser-capture microdissection, fluorescence activated cell sorting, and the DEPArray were used. One hundred twenty-six CTC pools and 56 single CTCs were isolated and sequenced. Hotspot regions over 48 cancer-related genes and 14 ALK mutations were examined to identify ALK-independent and ALK-dependent resistance mechanisms. RESULTS Multiple mutations in various genes in ALK-independent pathways were predominantly identified in CTCs of crizotinib-resistant patients. The RTK-KRAS (EGFR, KRAS, BRAF genes) and TP53 pathways were recurrently mutated. In one lorlatinib-resistant patient, two single CTCs out of 12 harbored ALK compound mutations. CTC-1 harbored the ALK G1202R/F1174C compound mutation virtually similar to ALK G1202R/F1174L present in the corresponding tumor biopsy. CTC-10 harbored a second ALK G1202R/T1151M compound mutation not detected in the tumor biopsy. By copy-number analysis, CTC-1 and the tumor biopsy had similar profiles, whereas CTC-10 harbored multiple copy-number alterations and whole-genome duplication. CONCLUSIONS Our results highlight the genetic heterogeneity and clinical utility of CTCs to identify therapeutic resistance mutations in ALK-rearranged patients. Single CTC sequencing may be a unique tool to assess heterogeneous resistance mechanisms and help clinicians for treatment personalization and resistance options to ALK-targeted therapies.
Collapse
Affiliation(s)
- Emma Pailler
- Gustave Roussy, Université Paris-Saclay, "Rare Circulating Cells" Translational Platform, CNRS UMS3655 - INSERM US23 AMMICA, Villejuif, France.,INSERM, U981 "Identification of Molecular Predictors and New Targets for Cancer Treatment," Villejuif, France.,Univ Paris Sud, Université Paris-Saclay, Faculty of Medicine, Le Kremlin-Bicêtre, France
| | - Vincent Faugeroux
- Gustave Roussy, Université Paris-Saclay, "Rare Circulating Cells" Translational Platform, CNRS UMS3655 - INSERM US23 AMMICA, Villejuif, France.,INSERM, U981 "Identification of Molecular Predictors and New Targets for Cancer Treatment," Villejuif, France.,Univ Paris Sud, Université Paris-Saclay, Faculty of Medicine, Le Kremlin-Bicêtre, France
| | - Marianne Oulhen
- Gustave Roussy, Université Paris-Saclay, "Rare Circulating Cells" Translational Platform, CNRS UMS3655 - INSERM US23 AMMICA, Villejuif, France.,INSERM, U981 "Identification of Molecular Predictors and New Targets for Cancer Treatment," Villejuif, France
| | - Laura Mezquita
- Gustave Roussy, Université Paris-Saclay, Department of Medicine, Villejuif, France
| | - Mélanie Laporte
- Gustave Roussy, Université Paris-Saclay, Genomic Platform and Biobank, Department of Medical Biology and Pathology, CNRS UMS3655 - INSERM US23 AMMICA, Villejuif, France
| | - Aurélie Honoré
- Gustave Roussy, Université Paris-Saclay, Genomic Platform and Biobank, Department of Medical Biology and Pathology, CNRS UMS3655 - INSERM US23 AMMICA, Villejuif, France
| | - Yann Lecluse
- Gustave Roussy, Université Paris-Saclay, "Flow Cytometry and Imaging" Platform, CNRS UMS3655 - INSERM US23 AMMICA, Villejuif, France
| | - Pauline Queffelec
- Gustave Roussy, Université Paris-Saclay, "Rare Circulating Cells" Translational Platform, CNRS UMS3655 - INSERM US23 AMMICA, Villejuif, France.,INSERM, U981 "Identification of Molecular Predictors and New Targets for Cancer Treatment," Villejuif, France
| | - Maud NgoCamus
- Gustave Roussy, Université Paris-Saclay, Department of Medicine, Villejuif, France
| | - Claudio Nicotra
- Gustave Roussy, Université Paris-Saclay, Department of Medicine, Villejuif, France
| | - Jordi Remon
- Gustave Roussy, Université Paris-Saclay, Department of Medicine, Villejuif, France
| | - Ludovic Lacroix
- Gustave Roussy, Université Paris-Saclay, Genomic Platform and Biobank, Department of Medical Biology and Pathology, CNRS UMS3655 - INSERM US23 AMMICA, Villejuif, France
| | - David Planchard
- Gustave Roussy, Université Paris-Saclay, Department of Medicine, Villejuif, France
| | - Luc Friboulet
- INSERM, U981 "Identification of Molecular Predictors and New Targets for Cancer Treatment," Villejuif, France.,Univ Paris Sud, Université Paris-Saclay, Faculty of Medicine, Le Kremlin-Bicêtre, France
| | - Benjamin Besse
- Univ Paris Sud, Université Paris-Saclay, Faculty of Medicine, Le Kremlin-Bicêtre, France.,Gustave Roussy, Université Paris-Saclay, Department of Medicine, Villejuif, France
| | - Françoise Farace
- Gustave Roussy, Université Paris-Saclay, "Rare Circulating Cells" Translational Platform, CNRS UMS3655 - INSERM US23 AMMICA, Villejuif, France. .,INSERM, U981 "Identification of Molecular Predictors and New Targets for Cancer Treatment," Villejuif, France.,Univ Paris Sud, Université Paris-Saclay, Faculty of Medicine, Le Kremlin-Bicêtre, France
| |
Collapse
|
288
|
Jin L, Lai J, Zhang Y, Fu Y, Wang S, Dai H, Huang B. BreakID: genomics breakpoints identification to detect gene fusion events using discordant pairs and split reads. Bioinformatics 2019; 35:2859-2861. [PMID: 30601940 DOI: 10.1093/bioinformatics/bty1070] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2018] [Revised: 12/04/2018] [Accepted: 12/31/2018] [Indexed: 11/14/2022] Open
Abstract
SUMMARY Here we developed a tool called Breakpoint Identification (BreakID) to identity fusion events from targeted sequencing data. Taking discordant read pairs and split reads as supporting evidences, BreakID can identify gene fusion breakpoints at single nucleotide resolution. After validation with confirmed fusion events in cancer cell lines, we have proved that BreakID can achieve high sensitivity of 90.63% along with PPV of 100% at sequencing depth of 500× and perform better than other available fusion detection tools. We anticipate that BreakID will have an extensive popularity in the detection and analysis of fusions involved in clinical and research sequencing scenarios. AVAILABILITY AND IMPLEMENTATION Source code is freely available at https://github.com/SinOncology/BreakID. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Linfang Jin
- Department of Research and Development, Sinotech Genomics Inc., Shanghai, China
| | - Jinhuo Lai
- Department of Medical Oncology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Yang Zhang
- Department of Research and Development, Sinotech Genomics Inc., Shanghai, China
| | - Ying Fu
- Department of Research and Development, Sinotech Genomics Inc., Shanghai, China
| | - Shuhang Wang
- Thoracic Medical Oncology Department, Peking University Cancer Hospital, Beijing, China
| | - Heng Dai
- Department of Research and Development, Sinotech Genomics Inc., Shanghai, China
| | - Bingding Huang
- Department of Research and Development, Sinotech Genomics Inc., Shanghai, China
| |
Collapse
|
289
|
CT Characteristics of Non-Small Cell Lung Cancer With Anaplastic Lymphoma Kinase Rearrangement: A Systematic Review and Meta-Analysis. AJR Am J Roentgenol 2019; 213:1059-1072. [PMID: 31414902 DOI: 10.2214/ajr.19.21485] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
OBJECTIVE. The purpose of this study was to perform a systematic review and meta-analysis regarding CT features of non-small cell lung cancer (NSCLC) with anaplastic lymphoma kinase (ALK) rearrangement. MATERIALS AND METHODS. The PubMed and Embase databases were searched up to February 20, 2019. Studies that evaluated CT features of NSCLC with and without ALK rearrangement was included. Methodologic quality was assessed using Quality Assessment of Diagnostic Accuracy Studies-2. The association between CT features and ALK rearrangement was pooled in the form of the odds ratio (OR) or the mean difference (MD) using the random-effects model. Heterogeneity was examined using the inconsistency index (I2). Publication bias was examined using funnel plots and Egger tests. RESULTS. Sixteen studies were included, consisting of 3113 patients with NSCLC. The overall prevalence of patients with ALK rearrangement was 17% (528/3113). Compared with NSCLC without ALK rearrangement, on CT images those with ALK rearrangement were more frequently solid (OR = 2.86), central in location (OR = 2.72), and 3 cm or smaller (OR = 0.57); had lower contrast-enhanced CT attenuation (MD = -4.79 HU); more frequently had N2 or N3 disease (OR = 5.63), lymphangitic carcinomatosis (OR = 3.46), pleural effusion (OR = 1.91), or pleural metastasis (OR = 1.81); and less frequently had lung metastasis (OR = 0.66). Heterogeneity varied among CT features (I2 = 0-80%). No significant publication bias was seen (p = 0.15). CONCLUSION. NSCLC with ALK rearrangement had several distinctive CT features compared with that without ALK rearrangement. These CT biomarkers may help identify patients likely to have ALK rearrangement.
Collapse
|
290
|
Descourt R, Perol M, Rousseau-Bussac G, Planchard D, Mennecier B, Wislez M, Cortot A, Guisier F, Galland L, Dô P, Schott R, Dansin E, Arrondeau J, Auliac JB, Chouaid C. Brigatinib in patients with ALK-positive advanced non-small-cell lung cancer pretreated with sequential ALK inhibitors: A multicentric real-world study (BRIGALK study). Lung Cancer 2019; 136:109-114. [PMID: 31491676 DOI: 10.1016/j.lungcan.2019.08.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 07/17/2019] [Accepted: 08/13/2019] [Indexed: 10/26/2022]
Abstract
OBJECTIVES Brigatinib is a next-generation ALK inhibitor initially developed in ALK-positive NSCLC pretreated with crizotinib. MATERIALS AND METHODS This retrospective multicentric study analyzed ALK-positive advanced NSCLC patients pretreated with at least one tyrosine-kinase inhibitor, including crizotinib, and enrolled in the brigatinib French early access program. The primary endpoint was investigator-assessed progression-free survival (PFS). RESULTS 104 patients were included (mean age, 56.6 years; never smokers, 61.5%; adenocarcinoma, 98.1%). Patients had received a median of 3 previous treatment lines, including at least 2 ALK inhibitors (mainly crizotinib then ceritinib). At brigatinib initiation, 59.1% had performance status 0-1, 51.9% had ≥ 3 metastatic sites, 74.5% had central nervous system metastases (CNS) and 8.8% had carcinomatous meningitis. Median duration of brigatinib treatment was 6.7 (95% CI, 0.06-20.7) months. Median PFS was 6.6 (4.8-9.9) months for the entire population. For patients who received 2, 3-4 and >4 lines of treatment before brigatinib, PFS was 4.3 (2.5-8.9), 10.4 (5.9-13.9) and 3.8 (0.8-7.4) months, respectively. In the 91 evaluable patients, disease control rate was 78.2%. From brigatinib start, median overall survival was 17.2 (11.0-not reached) months. Among the 68 patients with progressive disease after brigatinib, CNS was involved in 29.4% of cases. Median OS from the diagnosis of NSCLC was 75.3 (38.2-174.6) months. CONCLUSION These real-world results confirm the efficacy of brigatinib in a cohort of patients heavily pretreated for ALK-positive advanced NSCLC.
Collapse
Affiliation(s)
- Renaud Descourt
- Centre Hospitalier Universitaire, Oncology Department, Brest, France.
| | | | | | - David Planchard
- Gustave Roussy, Department of Medical Oncology, Thoracic Group, Villejuif, France
| | - Bertrand Mennecier
- Centre Hospitalier Universitaire de Strasbourg, Chest Department, Strasbourg, France
| | - Marie Wislez
- AP-HP, Hôpitaux Universitaires de l'Est Parisien, Tenon Hospital, Chest Department, Paris, France; AP-HP, Hôpitaux Universitaires Paris Centre, Cochin Hospital, Thoracic Oncology Unit, Department of Pneumology, Paris, France
| | - Alexis Cortot
- Centre Hospitalier Universitaire de Lille, Thoracic Oncology Unit, Lille, France
| | - Florian Guisier
- Centre Hospitalier Universitaire de Rouen, Chest Department, Rouen, France
| | - Loïck Galland
- Georges-François-Leclerc Cancer Center, Medical Oncology Department, Dijon, France
| | - Pascal Dô
- François-Baclesse Cancer Center, Medical Oncology Department, Caen, France
| | - Roland Schott
- Paul-Strauss Cancer Center, Medical Oncology Department, Strasbourg, France
| | - Eric Dansin
- Oscar-Lambret Cancer Center, Medical Oncology Department, Lille, France
| | - Jennifer Arrondeau
- AP-HP, Hôpitaux Universitaires Paris Centre, Cochin Hospital, Thoracic Oncology Unit, Department of Pneumology, Paris, France
| | | | | |
Collapse
|
291
|
Tomasini P, Egea J, Souquet-Bressand M, Greillier L, Barlesi F. Alectinib in the treatment of ALK-positive metastatic non-small cell lung cancer: clinical trial evidence and experience with a focus on brain metastases. Ther Adv Respir Dis 2019; 13:1753466619831906. [PMID: 30786826 PMCID: PMC6385324 DOI: 10.1177/1753466619831906] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Molecular profiling of metastatic nonsquamous non-small cell lung cancer (NSCLC) is required to guide the treatment strategy. Anaplastic lymphoma kinase (ALK) gene rearrangements are found in approximately 5% of lung adenocarcinomas and are associated with specific clinical features including a high risk of brain metastases. Crizotinib was the first ALK inhibitor developed and it demonstrated improved outcomes in patients with ALK-positive advanced NSCLC in comparison with chemotherapy. However, despite an initial response, all ALK-positive NSCLC patients develop acquired resistance to crizotinib. Because the most frequent mechanism of resistance is the development of a secondary ALK mutation, second (ceritinib, alectinib, brigatinib) and third-generation (lorlatinib) ALK inhibitors were developed. Alectinib is a second-generation ALK inhibitor and was shown to be effective for a broad spectrum of ALK rearrangements and ALK mutations. It was also shown to have high intracranial efficacy. In this article, we review clinical trial evidence of alectinib efficacy as well as publications reporting the experience of alectinib in daily practice, with a focus on brain metastases.
Collapse
Affiliation(s)
- Pascale Tomasini
- Aix Marseille University, APHM, CNRS, INSERM, CRCM, Hôpital Nord, Oncologie Multidisciplinaire et Innovations Thérapeutiques, Marseille, France
| | - Julie Egea
- Aix Marseille University, APHM, CNRS, INSERM, CRCM, Hôpital Nord, Oncologie Multidisciplinaire et Innovations Thérapeutiques, Marseille, France
| | - Maxime Souquet-Bressand
- Aix Marseille University, APHM, CNRS, INSERM, CRCM, Hôpital Nord, Oncologie Multidisciplinaire et Innovations Thérapeutiques, Marseille, France
| | - Laurent Greillier
- Aix Marseille University, APHM, CNRS, INSERM, CRCM, Hôpital Nord, Oncologie Multidisciplinaire et Innovations Thérapeutiques, Marseille, France
| | - Fabrice Barlesi
- Service d'Oncologie Multidisciplinaire et Innovations Thérapeutiques, Hôpital Nord, Chemin des Bourrely, 13915 Marseille Cedex, FranceAix Marseille University, APHM, CNRS, INSERM, CRCM, Hôpital Nord, Oncologie Multidisciplinaire et Innovations Thérapeutiques, Marseille, France
| |
Collapse
|
292
|
Park H, Sholl LM, Hatabu H, Awad MM, Nishino M. Imaging of Precision Therapy for Lung Cancer: Current State of the Art. Radiology 2019; 293:15-29. [PMID: 31385753 DOI: 10.1148/radiol.2019190173] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Advances in characterization of molecular and genomic abnormalities specific to lung cancer have made precision therapy the current standard of care for lung cancer treatment. This article will provide a cutting-edge review of imaging of lung cancer in the current era of precision medicine. The focus of the article includes (a) an update on the recent advances in precision therapy for non-small cell lung cancer and their implications on imaging; (b) molecular and genomic biomarkers and pitfalls of image interpretations for lung cancer precision therapy; and (c) review of the current approaches and future directions of precision imaging for lung cancer, emphasizing emerging observations in longitudinal tumor kinetics, radiomics, and molecular and functional imaging. The article is designed to help radiologists to remain up to date in the rapidly evolving world of lung cancer therapy and serve as key members of multidisciplinary teams caring for these patients.
Collapse
Affiliation(s)
- Hyesun Park
- From the Departments of Imaging (H.P., M.N.) and Medical Oncology (M.M.A.), Dana-Farber Cancer Institute, and Departments of Radiology (H.P., H.H., M.N.), Pathology (L.M.S.), and Medicine (M.M.A.), Brigham and Women's Hospital, 450 Brookline Ave, Boston, MA 02215
| | - Lynette M Sholl
- From the Departments of Imaging (H.P., M.N.) and Medical Oncology (M.M.A.), Dana-Farber Cancer Institute, and Departments of Radiology (H.P., H.H., M.N.), Pathology (L.M.S.), and Medicine (M.M.A.), Brigham and Women's Hospital, 450 Brookline Ave, Boston, MA 02215
| | - Hiroto Hatabu
- From the Departments of Imaging (H.P., M.N.) and Medical Oncology (M.M.A.), Dana-Farber Cancer Institute, and Departments of Radiology (H.P., H.H., M.N.), Pathology (L.M.S.), and Medicine (M.M.A.), Brigham and Women's Hospital, 450 Brookline Ave, Boston, MA 02215
| | - Mark M Awad
- From the Departments of Imaging (H.P., M.N.) and Medical Oncology (M.M.A.), Dana-Farber Cancer Institute, and Departments of Radiology (H.P., H.H., M.N.), Pathology (L.M.S.), and Medicine (M.M.A.), Brigham and Women's Hospital, 450 Brookline Ave, Boston, MA 02215
| | - Mizuki Nishino
- From the Departments of Imaging (H.P., M.N.) and Medical Oncology (M.M.A.), Dana-Farber Cancer Institute, and Departments of Radiology (H.P., H.H., M.N.), Pathology (L.M.S.), and Medicine (M.M.A.), Brigham and Women's Hospital, 450 Brookline Ave, Boston, MA 02215
| |
Collapse
|
293
|
Lee VHF, Mok TSK, Goto Y, Hsue VCC, Yang L, Jiang Y, Leung DKC, Lau KS, Tse PY. Differences Between the East and the West in Managing Advanced-Stage Non-small Cell Lung Cancer. Clin Oncol (R Coll Radiol) 2019; 32:e1-e9. [PMID: 31375307 DOI: 10.1016/j.clon.2019.07.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 06/02/2019] [Accepted: 07/16/2019] [Indexed: 11/27/2022]
Abstract
Lung cancer is a common cancer associated with high mortality rates worldwide. Unfortunately, it usually presents at a late stage, precluding the chance of curative therapy. The discovery of oncogenic driver mutations in patients with non-small cell lung cancer over the past 20 years has led to new molecular targeted therapies that have dramatically improved treatment efficacy and quality of life. New generations of therapy that target the drug-resistant mutations have also quickly evolved, benefiting patients who are refractory or intolerant to first-line targeted therapy. Eastern patients, from Southeast Asia, Japan and China, are known to have a higher incidence of epidermal growth factor receptor mutation. Therefore, compared with the West, more patients would benefit from these recent advances. In contrast, survival of patients without driver mutations has benefited from advances in novel therapeutics, including the immune checkpoint inhibitors. The current review aims to highlight the recent developments in the management of advanced-stage non-small cell lung cancer and to compare the differences in clinical practice between Eastern and Western countries.
Collapse
Affiliation(s)
- V H F Lee
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong SAR, China; Clinical Oncology Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China.
| | - T S K Mok
- State Key Laboratory of Translational Oncology, Department of Clinical Oncology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Y Goto
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - V C C Hsue
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong SAR, China; Clinical Oncology Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - L Yang
- Clinical Oncology Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Y Jiang
- Clinical Oncology Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - D K C Leung
- Department of Clinical Oncology, Queen Mary Hospital, Hong Kong SAR, China
| | - K S Lau
- Department of Clinical Oncology, Queen Mary Hospital, Hong Kong SAR, China
| | - P Y Tse
- Department of Clinical Oncology, Queen Mary Hospital, Hong Kong SAR, China
| |
Collapse
|
294
|
Kim HC, Kang YR, Ji W, Kim YJ, Yoon S, Lee JC, Choi CM. Frequency and clinical features of BRAF mutations among patients with stage III/IV lung adenocarcinoma without EGFR/ALK aberrations. Onco Targets Ther 2019; 12:6045-6052. [PMID: 31440061 PMCID: PMC6666367 DOI: 10.2147/ott.s213928] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 07/03/2019] [Indexed: 11/23/2022] Open
Abstract
Purpose BRAF mutations are found in 1-5% of non-small cell lung cancers, particularly adenocarcinomas. However, information regarding this mutation is limited in patients without EGFR/ALK aberrations, who have limited treatment options. Patients and methods The medical records of 224 stage III/IV adenocarcinoma patients without EGFR/ALK aberrations and with available pathologic tissue, were retrospectively reviewed. BRAF mutations were evaluated using a PNAClampTM BRAF mutation detection kit (Panagene, Daejeon, Korea). The outcomes in the study population were compared with stage III/IV adenocarcinoma patients harboring an EGFR mutation. A case report of targeted therapy against BRAF mutations was also presented. Results A cohort of 222 adenocarcinoma patients with adequate pathologic tissue samples was analyzed. The median patient age was 63 years, 68.8% of the patients were male and 68.7% were ever-smokers. The V600E BRAF mutation was detected in 4 patients (1.8%). The 222 study patients had a poorer survival outcome compared to stage III/IV adenocarcinoma patients with an EGFR mutation (median, 12 vs 67 months, P<0.001) from a recent previous study. Moreover, a 47-year-old female with a recurrent adenocarcinoma and a BRAF V600E mutation exhibited tumor regression after a fourth line therapy with dabrafenib and trametinib, targeting agents against BRAF mutations. Conclusion Although BRAF mutations are found in 1.8% of advanced adenocarcinoma patients without EGFR/ALK aberration, they may be able to serve as a treatment target in those patients.
Collapse
Affiliation(s)
- Ho Cheol Kim
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Yeh Rim Kang
- Medical Department, Oncology, Novartis Korea Pharmaceuticals, Seoul, South Korea
| | - Wonjun Ji
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Yeon Joo Kim
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Shinkyo Yoon
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Jae Cheol Lee
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Chang-Min Choi
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea.,Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| |
Collapse
|
295
|
Gafer H, de Waard Q, Compter A, van den Heuvel M. Rapid regression of neurological symptoms in patients with metastasised ALK+ lung cancer who are treated with lorlatinib: a report of two cases. BMJ Case Rep 2019; 12:12/7/e227299. [PMID: 31345828 PMCID: PMC6663151 DOI: 10.1136/bcr-2018-227299] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Oral anaplastic lymphoma kinase (ALK) tyrosine kinase inhibitors (TKI) have shown significant benefit in the management of ALK-rearranged non-small cell lung cancer (NSCLC). However, almost all patients will experience disease progression after front-line ALK-TKIs such as crizotinib. Treatment with third generation ALK-TKI lorlatinib can have a significant clinical impact following disease progression, even in patients with a very poor performance status. Here, we review two clinical cases with metastatic ALK-rearranged NSCLC who had pulmonary disease control with first-generation ALK inhibitor. However, disease progressed rapidly in the central nervous system with severe neurological symptoms. Treatment with lorlatinib, a third-generation ALK-TKI, led to a rapid radiological and clinical cerebral response in both patients. Lorlatinib can overcome ALK resistance to crizotinib, and the presented cases suggest a potential role for lorlatinib in patients with rapidly progressive cerebral and leptomeningeal metastases.
Collapse
Affiliation(s)
- Huda Gafer
- Pulmonology, Antoni van Leeuwenhoek, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Quincy de Waard
- Pulmonology, Antoni van Leeuwenhoek, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Annette Compter
- Neurology, Antoni van Leeuwenhoek, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Michel van den Heuvel
- Pulmonology, Antoni van Leeuwenhoek, Netherlands Cancer Institute, Amsterdam, The Netherlands
| |
Collapse
|
296
|
Hamilton G, Hochmair MJ. An evaluation of brigatinib as a promising treatment option for non-small cell lung cancer. Expert Opin Pharmacother 2019; 20:1551-1561. [PMID: 31328968 DOI: 10.1080/14656566.2019.1643839] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Introduction: Brigatinib is a second-line inhibitor for the treatment of rearranged anaplastic lymphoma kinase (ALK) in lung cancer patients which has significant activity against brain metastases. This tyrosine kinase inhibitor (TKI) overcomes a wide range of ALK mutations which confer therapeutic resistance and is increasingly applied in first-line therapy due to improved benefit for patients compared to crizotinib, the current standard of care. Areas covered: The authors review the development and characteristics of brigatinib and discuss the optimal clinical use and sequence of the application of ALK inhibitors in patients progressing under therapy. Expert opinion: ALK-rearranged NSCLC can be treated with a broad range of approved and novel inhibitors at various stages of therapy, including the second-line therapeutic brigatinib. Besides this TKI, the second-line ALK inhibitors alectinib and ceritinib, as well as the third-line lorlatinib are approved for the treatment of ALK-positive NSCLC patients. The main challenge is to find sequences and combinations of ALK inhibitors which provide the best benefit for the patients.
Collapse
Affiliation(s)
- G Hamilton
- Department of Surgery, Medical University of Vienna , Vienna , Austria
| | - M J Hochmair
- Respiratory Oncology Unit, Otto Wagner Hospital , Vienna , Austria
| |
Collapse
|
297
|
Chen DT, Schell MJ, Fulp WJ, Pettersson F, Kim S, Gray JE, Haura EB. Application of Bayesian predictive probability for interim futility analysis in single-arm phase II trial. Transl Cancer Res 2019; 8:S404-S420. [PMID: 31456910 PMCID: PMC6711387 DOI: 10.21037/tcr.2019.05.17] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Background Bayesian predictive probability design, with a binary endpoint, is gaining attention for the phase II trial due to its innovative strategy. To make the Bayesian design more accessible, we elucidate this Bayesian approach with a R package to streamline a statistical plan, so biostatisticians and clinicians can easily integrate the design into clinical trial. Methods We utilize a Bayesian framework using Bayesian posterior probability and predictive probability to build a R package and develop a statistical plan for the trial design. With pre-defined sample sizes, the approach employs the posterior probability with a threshold to calculate the minimum number of responders needed at end of the study to claim efficacy. Then the predictive probability is applied to evaluate future success at interim stages and form stopping rule at each stage. Results An R package, ‘BayesianPredictiveFutility’, with associated graphical interface is developed for easy utilization of the trial design. The statistical tool generates a professional statistical plan with comprehensive results including a summary, details of study design, a series of tables and figures from stopping boundary for futility, Bayesian predictive probability, performance [probability of early termination (PET), type I error, and power], PET at each interim analysis, sensitivity analysis for predictive probability, posterior probability, sample size, and beta prior distribution. The statistical plan presents the methodology in a readable language fashion while preserving rigorous statistical arguments. The output formats (Word or PDF) are available to communicate with physicians or to be incorporated in the trial protocol. Two clinical trials in lung cancer are used to demonstrate its usefulness. Conclusions Bayesian predictive probability method presents a flexible design in clinical trial. The statistical tool brings an added value to broaden the application.
Collapse
Affiliation(s)
- Dung-Tsa Chen
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Michael J Schell
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - William J Fulp
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Fredrik Pettersson
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Sungjune Kim
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Jhanelle E Gray
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Eric B Haura
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| |
Collapse
|
298
|
Akamatsu H, Ninomiya K, Kenmotsu H, Morise M, Daga H, Goto Y, Kozuki T, Miura S, Sasaki T, Tamiya A, Teraoka S, Tsubata Y, Yoshioka H, Hattori Y, Imamura CK, Katsuya Y, Matsui R, Minegishi Y, Mizugaki H, Nosaki K, Okuma Y, Sakamoto S, Sone T, Tanaka K, Umemura S, Yamanaka T, Amano S, Hasegawa K, Morita S, Nakajima K, Maemondo M, Seto T, Yamamoto N. The Japanese Lung Cancer Society Guideline for non-small cell lung cancer, stage IV. Int J Clin Oncol 2019; 24:731-770. [PMID: 31049758 PMCID: PMC6545178 DOI: 10.1007/s10147-019-01431-z] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 03/06/2019] [Indexed: 12/23/2022]
Abstract
According to rapid development of chemotherapy in advanced non-small cell lung cancer (NSCLC), the Japan Lung Cancer Society has been updated its own guideline annually since 2010. In this latest version, all of the procedure was carried out in accordance with grading of recommendations assessment, development and evaluation (GRADE) system. It includes comprehensive literature search, systematic review, and determination of the recommendation by multidisciplinary expert panel which consisted of medical doctors, pharmacists, nurses, statisticians, and patients from patient advocacy group. Recently, we have had various types of chemotherapeutic drugs like kinase inhibitors or immune-checkpoint inhibitors. Thus, the guideline proposes to categorize patients into three entities: (1) driver oncogene-positive, (2) PD-L1 ≥ 50%, and (3) others. Based on this subgroup, 31 clinical questions were described. We believe that this attempt enables clinicians to choose appropriate treatment easier. Here, we report an English version of the Japan Lung Cancer Society Guidelines 2018 for NSCLC, stages IV.
Collapse
Affiliation(s)
- Hiroaki Akamatsu
- Internal Medicine III, Wakayama Medical University, Wakayama, Japan
| | - Kiichiro Ninomiya
- Department of Hematology, Oncology and Respiratory Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | | | - Masahiro Morise
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Aichi, Japan
| | | | - Yasushi Goto
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Toshiyuki Kozuki
- Clinical Research Center, Department of Thoracic Oncology and Medicine, National Hospital Organization Shikoku Cancer Center, Ehime, Japan
| | - Satoru Miura
- Department of Internal Medicine, Niigata Cancer Center Hospital, Niigata, Japan
| | - Takaaki Sasaki
- Respiratory Center, Asahikawa Medical University Hospital, Hokkaido, Japan
| | - Akihiro Tamiya
- National Hospital Organization Kinki-chuo Chest Medical Center, Osaka, Japan
| | - Shunsuke Teraoka
- Internal Medicine III, Wakayama Medical University, Wakayama, Japan
| | - Yukari Tsubata
- Department of Internal Medicine, Division of Medical Oncology and Respiratory Medicine, Shimane University Faculty of Medicine, Shimane, Japan
| | - Hiroshige Yoshioka
- Department of Thoracic Oncology, Kansai Medical University Hospital, Osaka, Japan
| | | | - Chiyo K Imamura
- Advanced Cancer Translational Research Institute, Showa University, Tokyo, Japan
| | - Yuki Katsuya
- Department of Surgery, University of California San Diego, California, USA
| | - Reiko Matsui
- National Cancer Center Hospital East, Chiba, Japan
| | - Yuji Minegishi
- Department of Pulmonary Medicine and Oncology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Hidenori Mizugaki
- First Department of Medicine, Hokkaido University Hospital, Hokkaido, Japan
| | - Kaname Nosaki
- Thoracic Oncology, National Hospital Organization Kyushu Cancer Center, Fukuoka, Japan
| | - Yusuke Okuma
- Department of Thoracic Oncology and Respiratory Medicine, Tokyo Metropolitan Cancer and Infectious diseases Center Komagome Hospital, Tokyo, Japan
| | | | - Takashi Sone
- Regional Respiratory Symptomatology, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| | - Kentaro Tanaka
- Research Institute for Diseases of the Chest, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shigeki Umemura
- Department of Thoracic Oncology, National Cancer Center Hospital East, Chiba, Japan
| | - Takeharu Yamanaka
- Department of Biostatistics, Yokohama City University School of Medicine, Kanagawa, Japan
| | | | | | - Satoshi Morita
- Department of Biomedical Statistics and Bioinformatics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | | | - Makoto Maemondo
- Division of Pulmonary Medicine, Allergry and Rheumatology, Department of Internal Medicine, Iwate Medical University School of Medicine, Iwate, Japan
| | - Takashi Seto
- Thoracic Oncology, National Hospital Organization Kyushu Cancer Center, Fukuoka, Japan.
| | | |
Collapse
|
299
|
Formisano L, Jansen VM, Marciano R, Bianco R. From Biology to Therapy: Improvements of Therapeutic Options in Lung Cancer. Anticancer Agents Med Chem 2019; 18:1235-1240. [PMID: 28901258 DOI: 10.2174/1871520617666170912123416] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 07/12/2017] [Accepted: 08/25/2017] [Indexed: 12/14/2022]
Abstract
Lung cancer is the leading cause of cancer-related mortality around the world, despite effective chemotherapeutic agents, the prognosis has remained poor for a long time. The discovery of molecular changes that drive lung cancer has led to a dramatic shift in the therapeutic landscape of this disease. In "in vitro" and "in vivo" models of NSCLC (Non-Small Cell Lung Cancer), angiogenesis blockade has demonstrated an excellent anti-tumor activity, thus, a number of anti-angiogenic drugs have been approved by regulatory authorities for use in clinical practice. Much more interesting is the discovery of EGFR (Epithelial Growth Factor Receptor) mutations that predict sensitivity to the anti-EGFR Tyrosine Kinase Inhibitors (TKIs), a class of drugs that has shown to significantly improve survival when compared with standard chemotherapy in the first-line treatment of metastatic NSCLC. Nevertheless, after an initial response, resistance often occurs and prognosis becomes dismal. Biomolecular studies on cell line models have led to the discovery of mutations (e.g., T790M) that confer resistance to anti-EGFR inhibitors. Fortunately, drugs that are able to circumvent this mechanism of resistance have been developed and have been recently approved for clinical use. The discovery of robust intratumor lymphocyte infiltration in NSCLC has paved the way to several strategies able to restore the immune response. Thus, agents interfering with PD-1/PD-L1 (Programmed Death) pathways make up a significant portion of the armamentarium of cancer therapies for NSCLC. In all the above-mentioned situations, the basis of the success in treating NSCLC has started from understanding of the mutational landscape of the tumor.
Collapse
Affiliation(s)
- Luigi Formisano
- Department of Clinical Medicine and Surgery, University Federico II of Naples, Italy
| | - Valerie M Jansen
- Department of Medicine, Division of Hematology-Oncology, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Roberta Marciano
- Department of Clinical Medicine and Surgery, University Federico II of Naples, Italy
| | - Roberto Bianco
- Department of Clinical Medicine and Surgery, University Federico II of Naples, Italy
| |
Collapse
|
300
|
IGF1R Is a Potential New Therapeutic Target for HGNET-BCOR Brain Tumor Patients. Int J Mol Sci 2019; 20:ijms20123027. [PMID: 31234291 PMCID: PMC6627083 DOI: 10.3390/ijms20123027] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 05/28/2019] [Accepted: 06/14/2019] [Indexed: 12/24/2022] Open
Abstract
(1) Background: The high-grade neuroepithelial tumor of the central nervous system with BCOR alteration (HGNET-BCOR) is a highly malignant tumor. Preclinical models and molecular targets are urgently required for this cancer. Previous data suggest a potential role of insulin-like growth factor (IGF) signaling in HGNET-BCOR. (2) Methods: The primary HGNET-BCOR cells PhKh1 were characterized by western blot, copy number variation, and methylation analysis and by electron microscopy. The expression of IGF2 and IGF1R was assessed by qRT-PCR. The effect of chemotherapeutics and IGF1R inhibitors on PhKh1 proliferation was tested. The phosphorylation of IGF1R and downstream molecules was assessed by western blot. (3) Results: Phkh1 cells showed a DNA methylation profile compatible with the DNA methylation class "HGNET-BCOR" and morphologic features of cellular cannibalism. IGF2 and IGF1R were highly expressed by three HGNET-BCOR tumor samples and PhKh1 cells. PhKh1 cells were particularly sensitive to vincristine, vinblastine, actinomycin D (IC50 < 10 nM for all drugs), and ceritinib (IC50 = 310 nM). Ceritinib was able to abrogate the proliferation of PhKh1 cells and blocked the phosphorylation of IGF1R and AKT. (4) Conclusion: IGF1R is as an attractive target for the development of new therapy protocols for HGNET-BCOR patients, which may include ceritinib and vinblastine.
Collapse
|