551
|
Abstract
Advances in lung cancer genomics have revolutionized the diagnosis and treatment of this heterogeneous and clinically significant group of tumors. This article provides a broad overview of the most clinically relevant oncogenic alterations in common and rare lung tumors, with an emphasis on the pathologic correlates of the major oncogenic drivers, including EGFR, KRAS, ALK, and MET. Illustrations emphasize the morphologic diversity of lung adenocarcinoma, including genotype-phenotype correlations of genomic evolution in tumorigenesis. Molecular diagnostic approaches, including PCR-based testing, massively parallel sequencing, fluorescence in situ hybridization, and immunohistochemistry are reviewed.
Collapse
Affiliation(s)
- Lynette M Sholl
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA.
| |
Collapse
|
552
|
Yin J, Paoletti X, Sargent DJ, Mandrekar SJ. Repeated measures dose-finding design with time-trend detection in the presence of correlated toxicity data. Clin Trials 2017; 14:611-620. [PMID: 28764555 DOI: 10.1177/1740774517723829] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
BACKGROUND Phase I trials are designed to determine the safety, tolerability, and recommended phase 2 dose of therapeutic agents for subsequent testing. The dose-finding paradigm has thus traditionally focused on identifying the maximum tolerable dose of an agent or combination therapy under the assumption that there is a non-decreasing relationship between dose-toxicity and dose-efficacy. The dose is typically determined based on the probability of severe toxicity observed during the first treatment cycle. A novel endpoint, the total toxicity profile, was previously developed to account for the multiple toxicity types and grades experienced in the first cycle. More recently, this was extended to a repeated measures design based on the total toxicity profile to account for longitudinal toxicities over multiple treatment cycles in the absence of within-patient correlation. METHODS In this work, we propose to extend the design in the presence of within-patient correlation. Furthermore, we provide a framework to detect a toxicity time trend (toxicity increasing, decreasing, or stable) over multiple treatment cycles. We utilize a linear mixed model in the Bayesian framework, with the addition of Bayesian risk functions for decision-making in dose assignment. RESULTS The performance of this design was evaluated using simulation studies and real data from a phase I trial. We demonstrated that using available toxicity data from all cycles of treatment improves the accuracy of maximum tolerated dose identification and allows for the detection of a time trend. The performance is consistent regardless of the strength of the within-patient correlation. In addition, the use of a quasi-continuous total toxicity profile score significantly increased the power to detect time trends compared to when binary data only were used. CONCLUSION The increased interest in molecularly targeted agents and immunotherapies in oncology necessitates innovative phase I study designs. Our proposed framework provides a tool to tackle some of the challenges presented by these novel agents, specifically through the ability to understand patterns of toxicity over time, which is important in the cases of cumulative or late toxicities.
Collapse
Affiliation(s)
- Jun Yin
- 1 Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Xavier Paoletti
- 2 Biostatistics and Epidemiology Department, INSERM CESP, OncoStat, Institut Gustave Roussy, Villejuif, France
| | - Daniel J Sargent
- 1 Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | | |
Collapse
|
553
|
Hout DR, Schweitzer BL, Lawrence K, Morris SW, Tucker T, Mazzola R, Skelton R, McMahon F, Handshoe J, Lesperance M, Karsan A, Saltman DL. Performance of a RT-PCR Assay in Comparison to FISH and Immunohistochemistry for the Detection of ALK in Non-Small Cell Lung Cancer. Cancers (Basel) 2017; 9:cancers9080099. [PMID: 28763012 PMCID: PMC5575602 DOI: 10.3390/cancers9080099] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 07/19/2017] [Accepted: 07/29/2017] [Indexed: 12/17/2022] Open
Abstract
Patients with lung cancers harboring an activating anaplastic lymphoma kinase (ALK) rearrangement respond favorably to ALK inhibitor therapy. Fluorescence in situ hybridization (FISH) and immunohistochemistry (IHC) are validated and widely used screening tests for ALK rearrangements but both methods have limitations. The ALK RGQ RT-PCR Kit (RT-PCR) is a single tube quantitative real-time PCR assay for high throughput and automated interpretation of ALK expression. In this study, we performed a direct comparison of formalin-fixed paraffin-embedded (FFPE) lung cancer specimens using all three ALK detection methods. The RT-PCR test (diagnostic cut-off ΔCt of ≤8) was shown to be highly sensitive (100%) when compared to FISH and IHC. Sequencing of RNA detected full-length ALK transcripts or EML4-ALK and KIF5B-ALK fusion variants in discordant cases in which ALK expression was detected by the ALK RT-PCR test but negative by FISH and IHC. The overall specificity of the RT-PCR test for the detection of ALK in cases without full-length ALK expression was 94% in comparison to FISH and sequencing. These data support the ALK RT-PCR test as a highly efficient and reliable diagnostic screening approach to identify patients with non-small cell lung cancer whose tumors are driven by oncogenic ALK.
Collapse
Affiliation(s)
- David R Hout
- Insight Genetics, Inc., Suite 510, 2 International Plaza, Nashville, TN 37217, USA.
| | - Brock L Schweitzer
- Insight Genetics, Inc., Suite 510, 2 International Plaza, Nashville, TN 37217, USA.
| | - Kasey Lawrence
- Insight Genetics, Inc., Suite 510, 2 International Plaza, Nashville, TN 37217, USA.
| | - Stephan W Morris
- Insight Genetics, Inc., Suite 510, 2 International Plaza, Nashville, TN 37217, USA.
| | - Tracy Tucker
- Department of Pathology and Laboratory Medicine, BC Cancer Agency, 675 West 10th Avenue, Vancouver, BC V5Z 1L3, Canada.
| | - Rosetta Mazzola
- Department of Medical Oncology, British Columbia Cancer Agency, VIC 2410 Lee Avenue, Victoria, BC V8R 6V5, Canada.
| | - Rachel Skelton
- Insight Genetics, Inc., Suite 510, 2 International Plaza, Nashville, TN 37217, USA.
| | - Frank McMahon
- Insight Genetics, Inc., Suite 510, 2 International Plaza, Nashville, TN 37217, USA.
| | - John Handshoe
- Insight Genetics, Inc., Suite 510, 2 International Plaza, Nashville, TN 37217, USA.
| | - Mary Lesperance
- Department of Mathematics and Statistics, University of Victoria, Box 1700, STN CSC, Victoria, BC V8W 2Y2, Canada.
| | - Aly Karsan
- Department of Pathology and Laboratory Medicine, BC Cancer Agency, 675 West 10th Avenue, Vancouver, BC V5Z 1L3, Canada.
| | - David L Saltman
- Department of Medical Oncology, British Columbia Cancer Agency, VIC 2410 Lee Avenue, Victoria, BC V8R 6V5, Canada.
| |
Collapse
|
554
|
Hirano T, Yamada M, Ichinose M. Chronic Cholestasis with Dilation of Intrahepatic Bile Duct Related to Administration of Ceritinib. J Thorac Oncol 2017; 12:e123-e125. [PMID: 28748821 DOI: 10.1016/j.jtho.2017.04.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 03/31/2017] [Accepted: 04/01/2017] [Indexed: 10/19/2022]
Affiliation(s)
- Taizou Hirano
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Mitsuhiro Yamada
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan.
| | - Masakazu Ichinose
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
555
|
Shackelford RE, Ansari JM, Wei EX, Alexander JS, Cotelingam J. Anaplastic lymphoma kinase rearrangements in non-small-cell lung cancer: novel applications in diagnostics and treatment. Pharmacogenomics 2017; 18:1179-1192. [PMID: 28745554 DOI: 10.2217/pgs-2017-0098] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The ALK gene, first identified as an anaplastic large cell lymphoma driver mutation, is dysregulated in nearly 20 different human malignancies, including 3-7% of non-small-cell lung cancers (NSCLC). In NSCLC, ALK commonly fuses with the EML4, forming a constitutively active tyrosine kinase that drives oncogenic progression. Recently, several ALK-inhibiting drugs have been developed that are more effective than standard chemotherapeutic regimens in treating advanced ALK-positive NSCLC. For this reason, molecular diagnostic testing for dysregulated ALK expression is a necessary part of identifying optimal NSCLC treatment options. Here, we review the molecular pathology of ALK-positive NSCLC, ALK molecular diagnostic techniques, ALK-targeted NSCLC treatments, and drug resistance mechanisms to ALK-targeted therapies.
Collapse
Affiliation(s)
| | - Junaid M Ansari
- Department of Molecular & Cellular Physiology, LSU Health Sciences Center, Shreveport, LA 71130, USA
| | - Eric X Wei
- Department of Pathology, LSU Health Shreveport, Shreveport, LA, USA
| | - Jonathan S Alexander
- Department of Molecular & Cellular Physiology, LSU Health Sciences Center, Shreveport, LA 71130, USA
| | - James Cotelingam
- Department of Pathology, LSU Health Shreveport, Shreveport, LA, USA
| |
Collapse
|
556
|
Yuan C, Ma MJ, Parker JV, Mekhail TM. Metastatic Anaplastic Lymphoma Kinase-1 (ALK-1)-Rearranged Inflammatory Myofibroblastic Sarcoma to the Brain with Leptomeningeal Involvement: Favorable Response to Serial ALK Inhibitors: A Case Report. AMERICAN JOURNAL OF CASE REPORTS 2017; 18:799-804. [PMID: 28713152 PMCID: PMC5528006 DOI: 10.12659/ajcr.903698] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Patient: Female, 18 Final Diagnosis: Inflammatory myofibroblastic sarcoma Symptoms: Headache Medication: — Clinical Procedure: Craniotomy • lobectomy Specialty: Oncology
Collapse
Affiliation(s)
- Cai Yuan
- Department of Internal Medicine, Florida Hospital Internal Medicine Residency, Orlando, FL, USA
| | - Muchou J Ma
- Department of Pathology, Florida Hospital, Orlando, FL, USA
| | - John V Parker
- Department of Obstetrics and Gynecology, Florida Hospital, Orlando, FL, USA
| | | |
Collapse
|
557
|
Gill D, Schrader J, Kelly M, Martinez F, Syed W, Wright J. Gemcitabine associated pseudocellulitis: A missed diagnosis. J Oncol Pharm Pract 2017; 24:550-552. [DOI: 10.1177/1078155217719584] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
We report a case of a 51-year-old male with past medical history significant for cholangiocarcinoma presented with two weeks of worsening bilateral lower extremity swelling and erythema. Patient has been on active chemotherapy for his cholangiocarcinoma with Gemcitabine weekly infusions. Physical exam was significant for bilaterally petechial rash coalescing into ecchymoses over the dorsum of the feet, sparing soles and toes, which dissipated into thinning petechiae more proximally. On labs he did not have any leukocytosis, his platelet count was 50 × 103/µL and basic metabolic panel was benign. Patient was started on Vancomycin for presumed cellulitis. Lower extremity ultrasound Doppler ruled out deep venous thrombosis. Patient did have biopsies bilaterally on his legs, which showed hypersensitivity reaction consistent with the diagnosis of pseudocellulitis. His Vancomycin was discontinued and his symptoms improved. Our case further supports that pseudocellulitis is underrecognized and underreported, potentially leading to unnecessary antibiotic exposure and unnecessary diagnostic work-up as seen unfortunately in our patient. Unnecessary antibiotic exposure is increasing the risk for clostridium difficile and or antibiotic resistance, therefore awareness of this reaction is critical, as to avoid unnecessary antibiotics, and costly diagnostic workups.
Collapse
Affiliation(s)
- Dalvir Gill
- Department of Internal Medicine, SUNY Upstate Medical University, USA
| | - Josh Schrader
- Department of Pharmacy, SUNY Upstate Medical University, USA
| | - Matthew Kelly
- Department of Internal Medicine, SUNY Upstate Medical University, USA
| | - Fidel Martinez
- Department of Internal Medicine, SUNY Upstate Medical University, USA
| | - Wajihuddin Syed
- Department of Internal Medicine, SUNY Upstate Medical University, USA
| | - Jonathan Wright
- Department of Internal Medicine, SUNY Upstate Medical University, USA
| |
Collapse
|
558
|
Efficacy of alectinib in central nervous system metastases in crizotinib-resistant ALK-positive non-small-cell lung cancer: Comparison of RECIST 1.1 and RANO-HGG criteria. Eur J Cancer 2017. [PMID: 28646771 DOI: 10.1016/j.ejca.2017.05.019] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
BACKGROUND Central nervous system (CNS) progression is common in patients with anaplastic lymphoma kinase-positive (ALK+) non-small-cell lung cancer (NSCLC) receiving crizotinib. Next-generation ALK inhibitors have shown activity against CNS metastases, but accurate assessment of response and progression is vital. Data from two phase II studies in crizotinib-refractory ALK+ NSCLC were pooled to examine the CNS efficacy of alectinib, a CNS-active ALK inhibitor, using Response Evaluation Criteria in Solid Tumours (RECIST version 1.1) and Response Assessment in Neuro-Oncology high-grade glioma (RANO-HGG) criteria. METHODS Both studies enrolled patients aged ≥18 years who had previously received crizotinib. NP28761 was conducted in North America and NP28673 was a global study. All patients received 600 mg oral alectinib twice daily and had baseline CNS imaging. CNS response for those with baseline CNS metastases was determined by an independent review committee. RESULTS Baseline measurable CNS disease was identified in 50 patients by RECIST and 43 by RANO-HGG. CNS objective response rate was 64.0% by RECIST (95% confidence interval [CI]: 49.2-77.1; 11 CNS complete responses [CCRs]) and 53.5% by RANO-HGG (95% CI: 37.7-68.8; eight CCRs). CNS responses were durable, with consistent estimates of median duration of 10.8 months with RECIST and 11.1 months with RANO-HGG. Of the 39 patients with measurable CNS disease by both RECIST and RANO-HGG, only three (8%) had CNS progression according to one criteria but not the other (92% concordance rate). CONCLUSION Alectinib demonstrated promising efficacy in the CNS for ALK+ NSCLC patients pretreated with crizotinib, regardless of the assessment criteria used.
Collapse
|
559
|
Santarpia M, Daffinà MG, D’Aveni A, Marabello G, Liguori A, Giovannetti E, Karachaliou N, Gonzalez Cao M, Rosell R, Altavilla G. Spotlight on ceritinib in the treatment of ALK+ NSCLC: design, development and place in therapy. Drug Des Devel Ther 2017; 11:2047-2063. [PMID: 28740365 PMCID: PMC5503498 DOI: 10.2147/dddt.s113500] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The identification of echinoderm microtubule-associated protein-like 4 (EML4) and anaplastic lymphoma kinase (ALK) fusion gene in non-small cell lung cancer (NSCLC) has radically changed the treatment of a subset of patients harboring this oncogenic driver. Crizotinib was the first ALK tyrosine kinase inhibitor to receive fast approval and is currently indicated as the first-line therapy for advanced, ALK-positive NSCLC patients. However, despite crizotinib's efficacy, patients almost invariably progress, with the central nervous system being one of the most common sites of relapse. Different mechanisms of acquired resistance have been identified, including secondary ALK mutations, ALK copy number alterations and activation of bypass tracks. Different highly potent and brain-penetrant next-generation ALK inhibitors have been developed and tested in NSCLC patients with ALK rearrangements. Ceritinib, a structurally distinct and selective ALK inhibitor, showed 20 times higher potency than crizotinib in inhibiting ALK and had activity against the most common crizotinib-resistant mutations, including L1196M and G1269A, in preclinical models. In Phase I and II studies, ceritinib demonstrated pronounced activity in both crizotinib-naïve and crizotinib-refractory patients, with responses observed regardless of the presence of ALK resistance mutations. Ceritinib was the first ALK inhibitor to be approved for the treatment of crizotinib-refractory, ALK-rearranged NSCLC, and recent results from a Phase III study have demonstrated superior efficacy compared to standard chemotherapy in the first- and second-line setting. We provide an extensive overview of ceritinib from the design of the compound through preclinical data until efficacy and toxicity results from Phase I-III clinical studies. We review the molecular alterations associated with resistance to ceritinib and highlight the importance of obtaining tumor biopsy at progression to tailor therapy based upon the underlying resistance mechanism. We finally provide an outlook on novel rational therapeutic combinations.
Collapse
Affiliation(s)
- Mariacarmela Santarpia
- Medical Oncology Unit, Department of Human Pathology “G. Barresi”, University of Messina, Messina, Italy
| | - Maria Grazia Daffinà
- Medical Oncology Unit, Department of Human Pathology “G. Barresi”, University of Messina, Messina, Italy
| | - Alessandro D’Aveni
- Medical Oncology Unit, Department of Human Pathology “G. Barresi”, University of Messina, Messina, Italy
| | - Grazia Marabello
- Medical Oncology Unit, Department of Human Pathology “G. Barresi”, University of Messina, Messina, Italy
| | - Alessia Liguori
- Medical Oncology Unit, Department of Human Pathology “G. Barresi”, University of Messina, Messina, Italy
| | - Elisa Giovannetti
- Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands
- Department of Nanoscience and Nanotechnologies, CNR-Nano, Institute of Nanoscience and Nanotechnology
- Cancer Pharmacology Lab, AIRC Start-Up Unit, University of Pisa, Pisa, Italy
| | - Niki Karachaliou
- Institute of Oncology Rosell (IOR), University Hospital Sagrat Cor
| | - Maria Gonzalez Cao
- Oncology Department, Institute of Oncology Rosell (IOR), Quirón-Dexeus University Institute, Barcelona
| | - Rafael Rosell
- Cancer Biology and Precision Medicine Program, Germans Trias i Pujol Research Institute
- Catalan Institute of Oncology, Germans Trias i Pujol University Hospital, Badalona, Spain
| | - Giuseppe Altavilla
- Medical Oncology Unit, Department of Human Pathology “G. Barresi”, University of Messina, Messina, Italy
| |
Collapse
|
560
|
Losanno T, Gridelli C. Recent advances in targeted advanced lung cancer therapy in the elderly. Expert Rev Anticancer Ther 2017; 17:787-797. [DOI: 10.1080/14737140.2017.1348232] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Tania Losanno
- Medical Oncology, Azienda Ospedaliera San Camillo Forlanini, Roma, Italy
| | | |
Collapse
|
561
|
First macrocyclic 3 rd -generation ALK inhibitor for treatment of ALK/ROS1 cancer: Clinical and designing strategy update of lorlatinib. Eur J Med Chem 2017; 134:348-356. [DOI: 10.1016/j.ejmech.2017.04.032] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 03/26/2017] [Accepted: 04/12/2017] [Indexed: 11/21/2022]
|
562
|
Gainor JF, Shaw AT. J-ALEX: alectinib versus crizotinib in ALK-positive lung cancer. Lancet 2017; 390:3-4. [PMID: 28501139 DOI: 10.1016/s0140-6736(17)31074-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 02/08/2017] [Indexed: 11/18/2022]
Affiliation(s)
- Justin F Gainor
- Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA.
| | - Alice T Shaw
- Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| |
Collapse
|
563
|
Boonstra PS, Braun TM, Taylor JMG, Kidwell KM, Bellile EL, Daignault S, Zhao L, Griffith KA, Lawrence TS, Kalemkerian GP, Schipper MJ. Statistical controversies in clinical research: building the bridge to phase II-efficacy estimation in dose-expansion cohorts. Ann Oncol 2017; 28:1427-1435. [PMID: 28200082 PMCID: PMC5834117 DOI: 10.1093/annonc/mdx045] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Regulatory agencies and others have expressed concern about the uncritical use of dose expansion cohorts (DECs) in phase I oncology trials. Nonetheless, by several metrics-prevalence, size, and number-their popularity is increasing. Although early efficacy estimation in defined populations is a common primary endpoint of DECs, the types of designs best equipped to identify efficacy signals have not been established. METHODS We conducted a simulation study of six phase I design templates with multiple DECs: three dose-assignment/adjustment mechanisms multiplied by two analytic approaches for estimating efficacy after the trial is complete. We also investigated the effect of sample size and interim futility analysis on trial performance. Identifying populations in which the treatment is efficacious (true positives) and weeding out inefficacious treatment/populations (true negatives) are competing goals in these trials. Thus, we estimated true and false positive rates for each design. RESULTS Adaptively updating the MTD during the DEC improved true positive rates by 8-43% compared with fixing the dose during the DEC phase while maintaining false positive rates. Inclusion of an interim futility analysis decreased the number of patients treated under inefficacious DECs without hurting performance. CONCLUSION A substantial gain in efficiency is obtainable using a design template that statistically models toxicity and efficacy against dose level during expansion. Design choices for dose expansion should be motivated by and based upon expected performance. Similar to the common practice in single-arm phase II trials, cohort sample sizes should be justified with respect to their primary aim and include interim analyses to allow for early stopping.
Collapse
Affiliation(s)
| | | | | | | | | | | | - L. Zhao
- Departments of Biostatistics
| | | | | | | | | |
Collapse
|
564
|
Mooradian MJ, Gainor JF. ALK Fusion Detection in Circulating Free DNA: Finding an Important Needle in the Haystack. Oncologist 2017; 22:759-761. [PMID: 28642334 PMCID: PMC5507656 DOI: 10.1634/theoncologist.2017-0178] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 05/05/2017] [Indexed: 12/29/2022] Open
Abstract
ALK rearrangements have emerged as important therapeutic targets in oncology. This commentary, in light of recently reported cases, discusses the emerging role of liquid biopsies in molecular testing and new therapeutic options for oncology patients.
Collapse
Affiliation(s)
- Meghan J Mooradian
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Justin F Gainor
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
565
|
Das S, Horn L. Plasma genotyping in patients with non-small-cell lung cancer: simplifying or confusing the diagnosis? Lung Cancer Manag 2017; 6:29-37. [PMID: 30643568 DOI: 10.2217/lmt-2016-0019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2016] [Accepted: 02/10/2017] [Indexed: 11/21/2022] Open
Abstract
The identification of driver mutations in patients with advanced non-small-cell lung cancer has changed the treatment outcomes for patients with actionable driver mutations. Lack of tissue at diagnosis, however, remains a central obstacle in making optimal treatment decisions in patients with advanced disease. Although the US FDA has approved one plasma-based test for detecting epidermal growth factor receptor mutations in patients with advanced stage disease, sensitivity of these assays remains mediocre, necessitating additional tissue testing and possible delays in patients with negative results. Serial monitoring for response and early detection of acquired resistance to targeted therapies is also possible with cell-free DNA, however the benefit of switching therapy prior to detection of changes on imaging is unknown currently.
Collapse
Affiliation(s)
- Satya Das
- 2220 Pierce Avenue, 777 Preston Research Building, Nashville, TN 37232, USA
| | - Leora Horn
- 2220 Pierce Avenue, 777 Preston Research Building, Nashville, TN 37232, USA
| |
Collapse
|
566
|
Hao L, Du B, Xi X. TRIM59 is a novel potential prognostic biomarker in patients with non-small cell lung cancer: A research based on bioinformatics analysis. Oncol Lett 2017; 14:2153-2164. [PMID: 28789440 PMCID: PMC5530082 DOI: 10.3892/ol.2017.6467] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 06/14/2017] [Indexed: 12/28/2022] Open
Abstract
Lung cancer is the leading cause of cancer-associated mortality worldwide and its prognosis is poor. Few effective biomarkers for non-small cell lung cancer (NSCLC) have been translated into the clinical practice aiming to assist in the treatment plan design and prognosis evaluation. The aim of the present study was to identify novel potential prognostic biomarkers for NSCLC. Tripartite motif 59 (TRIM59) was identified from a microarray dataset of matched-samples and was verified as an aberrantly upregulated gene in NSCLC tissue. The expression level of TRIM59 in NSCLC subtypes was observed to be significantly increased in large cell lung carcinoma and squamous cell carcinoma as compared with that in adenocarcinoma. Its expression correlated with several clinicopathological features, including gender, smoking habits, and unfavorable tumor node and pathological stages. Notably, TRIM59 demonstrated a negative correlation with survival time and its overexpression indicated a poor prognosis in NSCLC. Furthermore, univariate and multivariate Cox's regression analyses indicated that TRIM59 was an independent prognostic factor in tumor tissue as compared with age, gender, tumor stage, node stage, and metastasis. Gene set enrichment analysis and protein-protein interaction network construction revealed that TRIM59 was associated with oncogenic mammalian target of rapamycin (MTOR) and eukaryotic initiation factor 4E (EIF4E) signaling through ubiquitin C binding. In conclusion, it was revealed that TRIM59 is a novel prognostic biomarker modulating oncogenic MTOR and EIF4E signaling pathways in NSCLC. These findings provided a novel insight into the clinical application of TRIM59. Therefore, TRIM59 may serve as an independent predictor for prognosis and a potential therapeutic target for NSCLC.
Collapse
Affiliation(s)
- Ling Hao
- School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Boyu Du
- School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Xueyan Xi
- School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| |
Collapse
|
567
|
Dolly SO, Collins DC, Sundar R, Popat S, Yap TA. Advances in the Development of Molecularly Targeted Agents in Non-Small-Cell Lung Cancer. Drugs 2017; 77:813-827. [PMID: 28378229 DOI: 10.1007/s40265-017-0732-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Non-small-cell lung cancer (NSCLC) remains a significant global health challenge and the leading cause of cancer-related mortality. The traditional 'one-size-fits-all' treatment approach has now evolved into one that involves personalized strategies based on histological and molecular subtypes. The molecular era has revolutionized the treatment of patients harboring epidermal growth factor receptor (EGFR), anaplastic lymphoma kinase (ALK) and ROS1 gene aberrations. In the appropriately selected population, anti-tumor agents against these molecular targets can significantly improve progression-free survival. However, the emergence of acquired resistance is inevitable. Novel potent compounds with much improved and rational selectivity profiles, such as third-generation EGFR T790M resistance mutation-specific inhibitors, have been developed and added to the NSCLC armamentarium. To date, attempts to overcome resistance bypass pathways through downstream signaling blockade has had limited success. Furthermore, the majority of patients still do not harbor known driver genetic or epigenetic alterations and/or have no new available treatment options, with chemotherapy remaining their standard of care. Several potentially actionable driver aberrations have recently been identified, with the early clinical development of multiple inhibitors against these promising targets currently in progress. The advent of immune checkpoint inhibitors has led to significant benefit for advanced NSCLC patients with durable responses observed. Further interrogation of the underlying biology of NSCLC, coupled with modern clinical trial designs, is now required to develop novel targeted therapeutics rationally matched with predictive biomarkers of response, so as to further advance NSCLC therapeutics through the next decade.
Collapse
Affiliation(s)
| | | | - Raghav Sundar
- Royal Marsden NHS Foundation Trust, London, UK.,National University Health System, Singapore, Singapore
| | - Sanjay Popat
- Royal Marsden NHS Foundation Trust, London, UK.,National Heart and Lung Institute, Imperial College London, London, UK
| | - Timothy A Yap
- Royal Marsden NHS Foundation Trust, London, UK. .,Drug Development Unit and Lung Cancer Unit, The Institute of Cancer Research and Royal Marsden Hospital, Downs Road, London, SM2 5PT, UK.
| |
Collapse
|
568
|
Bylicki O, Paleiron N, Margery J, Guisier F, Vergnenegre A, Robinet G, Auliac JB, Gervais R, Chouaid C. Targeting the PD-1/PD-L1 Immune Checkpoint in EGFR-Mutated or ALK-Translocated Non-Small-Cell Lung Cancer. Target Oncol 2017. [DOI: 10.1007/s11523-017-0510-9] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
569
|
Abstract
Lung cancer is the most common cause of cancer-related death worldwide, less than 7% of patients survive 10 years following diagnosis across all stages of lung cancer. Late stage of diagnosis and lack of effective and personalized medicine reflect the need for a better understanding of the mechanisms that underlie lung cancer progression. Quantitative proteomics provides the relative different protein abundance in normal and cancer patients which offers the information for molecular interactions, signaling pathways, and biomarker identification. Here we introduce both theoretical and practical applications in the use of quantitative proteomics approaches, with principles of current technologies and methodologies including gel-based, label free, stable isotope labeling as well as targeted proteomics. Predictive markers of drug resistance, candidate biomarkers for diagnosis, and prognostic markers in lung cancer have also been discovered and analyzed by quantitative proteomic analysis. Moreover, construction of protein networks enables to provide an opportunity to interpret disease pathway and improve our understanding in cancer therapeutic strategies, allowing the discovery of molecular markers and new therapeutic targets for lung cancer.
Collapse
Affiliation(s)
| | - Hsueh-Fen Juan
- Institute of Molecular and Cellular Biology, National Taiwan University, Taipei, Taiwan. .,Department of Life Science, National Taiwan University, Taipei, Taiwan. .,Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, No. 1, Sec. 4, Roosevelt Road, Taipei, 10617, Taiwan.
| |
Collapse
|
570
|
Costa DB. Ascending role of next-generation ALK inhibitors. Lancet Oncol 2017; 18:837-839. [PMID: 28602777 DOI: 10.1016/s1470-2045(17)30338-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 04/07/2017] [Indexed: 12/19/2022]
Affiliation(s)
- Daniel B Costa
- Department of Medicine, Division of Hematology and Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
571
|
Cases of ALK-Rearranged Lung Cancer with 5-Year Progression-Free Survival with Crizotinib as Initial Precision Therapy. J Thorac Oncol 2017; 12:e175-e177. [PMID: 28611010 DOI: 10.1016/j.jtho.2017.06.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 06/02/2017] [Indexed: 11/21/2022]
|
572
|
Diagnosis and Treatment of Anaplastic Lymphoma Kinase-Positive Non-Small Cell Lung Cancer. Hematol Oncol Clin North Am 2017; 31:101-111. [PMID: 27912826 DOI: 10.1016/j.hoc.2016.08.012] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Anaplastic lymphoma kinase (ALK) gene rearrangements occur in a small portion of patients with non-small cell lung cancer (NSCLC). These gene rearrangements lead to constitutive activation of the ALK kinase and subsequent ALK-driven tumor formation. Patients with tumors harboring such rearrangements are highly sensitive to ALK inhibitors, such as crizotinib, ceritinib, and alectinib. Resistance to these kinase inhibitors occurs through several mechanisms, resulting in ongoing clinical challenges. This review summarizes the biology of ALK-positive lung cancer, methods for diagnosing ALK-positive NSCLC, current FDA-approved ALK inhibitors, mechanisms of resistance to ALK inhibition, and potential strategies to combat resistance.
Collapse
|
573
|
Management of ceritinib therapy and adverse events in patients with ALK-rearranged non-small cell lung cancer. Lung Cancer 2017; 111:51-58. [PMID: 28838397 DOI: 10.1016/j.lungcan.2017.06.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 06/05/2017] [Accepted: 06/08/2017] [Indexed: 12/18/2022]
Abstract
Anaplastic lymphoma kinase rearrangement (ALK+) occurs in approximately 2-7% of patients with non-small cell lung cancer (NSCLC), contributing to a considerable number of patients with ALK+ NSCLC worldwide. Ceritinib is a next generation ALK inhibitor (ALKi), approved by the European Medicines Agency in 2015. In the first-in-human, phase I study, ceritinib demonstrated rapid and durable responses in ALK patients previously treated with a different ALKi and in those who were ALKi-naive. As ceritinib is starting to be used routinely for the treatment of patients with ALK+ NSCLC, experience is growing with regard to ideal therapy management. In this review we provide a brief background to the development of ceritinib. The optimal treatment management and adverse events associated with ceritinib in clinical trials and in clinical practice are then discussed in detail, and where applicable, an expert consensus on specific recommendations are made. In clinical trials, the most common adverse events related to ceritinib are nausea, vomiting, and diarrhea. However, the majority of these are mild and, in the opinion of the authors, can be effectively managed with dose modifications. Based on clinical data, ceritinib has demonstrated efficacy as a first-line therapy and in patients who have relapsed on crizotinib, including those with brain metastases at baseline. Unfortunately, at some point, all patients experience progressive disease, with the central nervous system being a common site of metastases. Recommendations are made for continuing treatment beyond disease progression as long as a clinical benefit to patients is observed. Here, we review management of ceritinib treatment and adverse events and make recommendations on optimal management of patients.
Collapse
|
574
|
Abstract
OBJECTIVE We aimed to identify key principles of targeted therapy of protein kinases and their application to the management of solid tumors. BACKGROUND Concurrent advances in tumor genomic analysis and molecular inhibitor development have dramatically impacted the diagnosis and treatment of solid tumors, and common themes regarding the use of kinase inhibitors are developing. METHODS The list of kinase inhibitors that have been approved by the US Food and Drug Administration was reviewed and articles related to the agents were searched in the PubMed database up until December 2015. We included pivotal, randomized controlled phase 2 and 3 trials, and also pertinent preclinical studies. RESULTS Small molecule inhibitors targeted against driver kinases, overactive in selected subsets of solid tumors, elicit improved response rates and survival compared with standard chemotherapy. Disease control has been proven in the metastatic and, to a limited extent, the adjuvant setting. However, tumor eradication is rare, and duration of treatment response is limited by the development of drug resistance. CONCLUSIONS Kinase inhibitors induce response in diverse types of solid tumors. Although the agents are often effective in defined molecular subsets, cure is rare and resistance is common. This broad review provides rationale for further investigation of multimodality therapy combining kinase inhibitors with additional systemic and local therapies, including surgery.
Collapse
|
575
|
Drilon A, Nagasubramanian R, Blake JF, Ku N, Tuch BB, Ebata K, Smith S, Lauriault V, Kolakowski GR, Brandhuber BJ, Larsen PD, Bouhana KS, Winski SL, Hamor R, Wu WI, Parker A, Morales TH, Sullivan FX, DeWolf WE, Wollenberg LA, Gordon PR, Douglas-Lindsay DN, Scaltriti M, Benayed R, Raj S, Hanusch B, Schram AM, Jonsson P, Berger MF, Hechtman JF, Taylor BS, Andrews S, Rothenberg SM, Hyman DM. A Next-Generation TRK Kinase Inhibitor Overcomes Acquired Resistance to Prior TRK Kinase Inhibition in Patients with TRK Fusion-Positive Solid Tumors. Cancer Discov 2017; 7:963-972. [PMID: 28578312 DOI: 10.1158/2159-8290.cd-17-0507] [Citation(s) in RCA: 329] [Impact Index Per Article: 41.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 05/24/2017] [Accepted: 05/26/2017] [Indexed: 12/13/2022]
Abstract
Larotrectinib, a selective TRK tyrosine kinase inhibitor (TKI), has demonstrated histology-agnostic efficacy in patients with TRK fusion-positive cancers. Although responses to TRK inhibition can be dramatic and durable, duration of response may eventually be limited by acquired resistance. LOXO-195 is a selective TRK TKI designed to overcome acquired resistance mediated by recurrent kinase domain (solvent front and xDFG) mutations identified in multiple patients who have developed resistance to TRK TKIs. Activity against these acquired mutations was confirmed in enzyme and cell-based assays and in vivo tumor models. As clinical proof of concept, the first 2 patients with TRK fusion-positive cancers who developed acquired resistance mutations on larotrectinib were treated with LOXO-195 on a first-in-human basis, utilizing rapid dose titration guided by pharmacokinetic assessments. This approach led to rapid tumor responses and extended the overall duration of disease control achieved with TRK inhibition in both patients.Significance: LOXO-195 abrogated resistance in TRK fusion-positive cancers that acquired kinase domain mutations, a shared liability with all existing TRK TKIs. This establishes a role for sequential treatment by demonstrating continued TRK dependence and validates a paradigm for the accelerated development of next-generation inhibitors against validated oncogenic targets. Cancer Discov; 7(9); 963-72. ©2017 AACR.See related commentary by Parikh and Corcoran, p. 934This article is highlighted in the In This Issue feature, p. 920.
Collapse
Affiliation(s)
- Alexander Drilon
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.,Weill Cornell Medical College, New York, New York
| | | | | | - Nora Ku
- Loxo Oncology, Inc., Stamford, Connecticut
| | | | | | | | | | | | | | | | | | | | | | - Wen-I Wu
- Array BioPharma, Boulder, Colorado
| | | | | | | | | | | | | | | | - Maurizio Scaltriti
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ryma Benayed
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sandeep Raj
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Bethany Hanusch
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Alison M Schram
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Philip Jonsson
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Michael F Berger
- Weill Cornell Medical College, New York, New York.,Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York.,Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jaclyn F Hechtman
- Weill Cornell Medical College, New York, New York.,Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Barry S Taylor
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York.,Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | | | | | - David M Hyman
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York. .,Weill Cornell Medical College, New York, New York
| |
Collapse
|
576
|
Zhang X, Tian T, Sun W, Liu C, Fang X. Bmi-1 overexpression as an efficient prognostic marker in patients with nonsmall cell lung cancer. Medicine (Baltimore) 2017; 96:e7346. [PMID: 28658153 PMCID: PMC5500075 DOI: 10.1097/md.0000000000007346] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND The prognostic effect of B-cell-specific Moloney leukemia virus insertion site 1 (Bmi-1) in patients with nonsmall cell lung cancer (NSCLC) remains controversial. We thus performed a meta-analysis to reveal the correlation between Bmi-1 with clinical features and overall survival (OS) in NSCLC. METHODS Relevant studies were searched through PubMed, Embase, and Web of Science. Pooled hazard ratios (HRs) and 95% confidence intervals (CIs) as well as odds ratios (ORs) and 95% CIs were calculated by using STATA version 12.0. RESULTS Fourteen studies consisting of 1323 patients were included for quantitative analysis. The results showed that Bmi-1 was significantly associated with tumor size (n = 7, OR = 1.79, 95% CI = 1.19-2.71, P = .005, fixed effect), poor differentiation (OR = 1.61, 95% CI = 1.11-2.33, P = .011, fixed effect), and distant metastasis (n = 4, OR = 4.69, 95% CI = 1.52-14.41, P = .007, fixed effect). In addition, high Bmi-1 expression also predicted poor OS (HR = 1.62, 95% CI = 1.14-2.3, P < .001). There was no significant publication bias for any of the analyses. CONCLUSION In conclusion, Bmi-1 overexpression was correlated with tumor size, poor differentiation, distant metastasis, and worse OS in NSCLC. Therefore, Bmi-1 could be recommended as an efficient prognostic marker for NSCLC.
Collapse
|
577
|
Lin L, Zhao J, Kong N, He Y, Hu J, Huang F, Han J, Cao X. Meta-analysis of the incidence and risks of interstitial lung disease and QTc prolongation in non-small-cell lung cancer patients treated with ALK inhibitors. Oncotarget 2017; 8:57379-57385. [PMID: 28915678 PMCID: PMC5593649 DOI: 10.18632/oncotarget.18283] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2017] [Accepted: 05/03/2017] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND To conduct a systematic review and meta-analysis to assess the overall incidence and risk of interstitial lung disease (ILD) and QTc prolongation associated with anaplastic lymphoma kinase (ALK)-tyrosine kinase inhibitors (-TKIs) in non-small-cell lung cancer (NSCLC) patients. RESULTS A total of 1,770 patients from 8 clinical trials were included. The incidences of high-grade ILD and QTc prolongation was 2.5% (95% CI 1.7-3.6%), and 2.8% (95% CI 1.8-4.3%), respectively. Meta-analysis demonstrated that the use of ALK-TKIs in NSCLC patients significantly increased the risk of developing high-grade ILD (Peto OR, 3.27, 95%CI: 1.18-9.08, p = 0.023) and QTc prolongation (Peto OR 7.51, 95% CI, 2.16-26.15; p = 0.002) in comparison with chemotherapy alone. MATERIALS AND METHODS A systematic literature search was performed to identify related citations up to January 31, 2017. Data were extracted, and summary incidence rates, Peto odds ratios (Peto ORs), and 95% confidence intervals (CIs) were calculated. CONCLUSIONS The use of ALK-TKIs significantly increases the risk of developing high-grade ILD and QTc prolongation in lung cancer patients. Clinicians should pay attention to the risks of severe ILD and QTc prolongation with the administration of these drugs.
Collapse
Affiliation(s)
- Liping Lin
- Department of Oncology, Panyu Central Hospital, Guangzhou, 511400, China.,Cancer Institute of Panyu, Guangzhou, 511400, China
| | - Juanjuan Zhao
- School of Nursing, Sun Yat-sen University, Guangzhou, 510000, China
| | - Ning Kong
- Department of Ophthalmology, Panyu Central Hospital, Guangzhou, 511400, China.,Cancer Institute of Panyu, Guangzhou, 511400, China
| | - Yan He
- Department of Oncology, Panyu Central Hospital, Guangzhou, 511400, China.,Cancer Institute of Panyu, Guangzhou, 511400, China
| | - Jiazhu Hu
- Department of Oncology, Panyu Central Hospital, Guangzhou, 511400, China.,Cancer Institute of Panyu, Guangzhou, 511400, China
| | - Fuxi Huang
- Department of Oncology, Panyu Central Hospital, Guangzhou, 511400, China.,Cancer Institute of Panyu, Guangzhou, 511400, China
| | - Jianjun Han
- Department of Oncology, Panyu Central Hospital, Guangzhou, 511400, China.,Cancer Institute of Panyu, Guangzhou, 511400, China
| | - Xiaolong Cao
- Department of Oncology, Panyu Central Hospital, Guangzhou, 511400, China.,Cancer Institute of Panyu, Guangzhou, 511400, China
| |
Collapse
|
578
|
Hyman DM, Taylor BS, Baselga J. Implementing Genome-Driven Oncology. Cell 2017; 168:584-599. [PMID: 28187282 DOI: 10.1016/j.cell.2016.12.015] [Citation(s) in RCA: 345] [Impact Index Per Article: 43.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 12/09/2016] [Accepted: 12/13/2016] [Indexed: 12/18/2022]
Abstract
Early successes in identifying and targeting individual oncogenic drivers, together with the increasing feasibility of sequencing tumor genomes, have brought forth the promise of genome-driven oncology care. As we expand the breadth and depth of genomic analyses, the biological and clinical complexity of its implementation will be unparalleled. Challenges include target credentialing and validation, implementing drug combinations, clinical trial designs, targeting tumor heterogeneity, and deploying technologies beyond DNA sequencing, among others. We review how contemporary approaches are tackling these challenges and will ultimately serve as an engine for biological discovery and increase our insight into cancer and its treatment.
Collapse
Affiliation(s)
- David M Hyman
- Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA; Weill Cornell Medical College, 1300 York Avenue, New York, NY 10065, USA
| | - Barry S Taylor
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA; Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA; Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - José Baselga
- Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA; Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA; Weill Cornell Medical College, 1300 York Avenue, New York, NY 10065, USA.
| |
Collapse
|
579
|
Bordi P, Tiseo M, Rofi E, Petrini I, Restante G, Danesi R, Del Re M. Detection of ALK and KRAS Mutations in Circulating Tumor DNA of Patients With Advanced ALK-Positive NSCLC With Disease Progression During Crizotinib Treatment. Clin Lung Cancer 2017; 18:692-697. [PMID: 28601386 DOI: 10.1016/j.cllc.2017.04.013] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 04/22/2017] [Accepted: 04/25/2017] [Indexed: 11/25/2022]
Abstract
BACKGROUND In patients with anaplastic lymphoma kinase (ALK)-positive non-small-cell lung cancer (NSCLC), disease progression occurs after a median of 9 to 10 months of crizotinib treatment. Several mechanisms of resistance have been identified and include ALK mutations and amplification or the activation of bypassing signaling pathways. Rebiopsy in NSCLC patients represents a critical issue and the analysis of circulating cell-free DNA (cfDNA) has a promising role for the identification of resistance mechanisms. PATIENTS AND METHODS Twenty patients with advanced ALK-positive NSCLC were enrolled after disease progression during crizotinib treatment; cfDNA was analyzed using digital droplet polymerase chain reaction (BioRad, Hercules, CA) for ALK (p.L1196M, p.G1269A, and p.F1174L) and Kirsten rat sarcoma (KRAS) (codons 12 and 13) mutations. RESULTS ALK secondary mutations (p.L1196M, p.G1269A, and p.F1174L) were identified in 5 patients; 1 patient had 2 ALK mutations (p.L1196M and p.G1269A). Overall, 10 patients presented KRAS mutations (7 p.G12D, 2 p.G12V, and 1 p.G12C mutations, respectively). In 3 patients KRAS mutations were associated with ALK mutations. cfDNA was monitored during the treatment with second-generation ALK inhibitors and the amount of ALK as well as KRAS mutations decreased along with tumor regression. CONCLUSION ALK and KRAS mutations are associated with acquired resistance to crizotinib in ALK-positive NSCLC. In particular, ALK acquired mutations can be detected in plasma and could represent a promising tumor marker for response monitoring.
Collapse
Affiliation(s)
- Paola Bordi
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - Marcello Tiseo
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy.
| | - Eleonora Rofi
- Clinical Pharmacology and Pharmacogenetic Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Iacopo Petrini
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Giuliana Restante
- Clinical Pharmacology and Pharmacogenetic Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Romano Danesi
- Clinical Pharmacology and Pharmacogenetic Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Marzia Del Re
- Clinical Pharmacology and Pharmacogenetic Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| |
Collapse
|
580
|
Karachaliou N, Santarpia M, Gonzalez Cao M, Teixido C, Sosa AE, Berenguer J, Rodriguez Capote A, Altavilla G, Rosell R. Anaplastic lymphoma kinase inhibitors in phase I and phase II clinical trials for non-small cell lung cancer. Expert Opin Investig Drugs 2017; 26:713-722. [PMID: 28463570 DOI: 10.1080/13543784.2017.1324572] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
INTRODUCTION Crizotinib is a first-in-class ALK tyrosine kinase inhibitor (TKI), which has proven its superiority over standard platinum-based chemotherapy for the first-line therapy of ALK-rearranged non-small cell lung cancer (NSCLC) patients. The development of acquired resistance to crizotinib represents an ongoing challenge with the central nervous system being one of the most common sites of relapse. Ceritinib and alectinib are approved second-generation ALK TKIs. Several novel ALK inhibitors, more potent and with different selectivity compared to crizotinib, are currently in development. Areas covered: This review will focus on new ALK inhibitors, currently in phase 1 or 2 clinical studies. We will also comment on the mechanisms of resistance to ALK inhibition and the strategies to delay or overcome resistance. Expert opinion: The therapeutic management of ALK-rearranged NSCLC has been greatly improved. Next-generation ALK inhibitors have shown differential potency against ALK rearrangements and ALK resistance mutations. The molecular profile of the tumor at the time of disease progression to crizotinib is crucial for the sequencing of novel ALK TKIs. Ongoing clinical studies will address key issues, including the optimal therapeutic algorithm and whether combinational approaches are more effective than single ALK inhibition for the outcome of ALK-rearranged NSCLC patients.
Collapse
Affiliation(s)
- Niki Karachaliou
- a Institute of Oncology Rosell (IOR), University Hospital Sagrat Cor , Barcelona , Spain
| | - Mariacarmela Santarpia
- b Medical Oncology Unit, Department of Human Pathology 'G. Barresi,' University of Messina , Messina , Italy
| | - Maria Gonzalez Cao
- c Institute of Oncology Rosell (IOR) , Quirón-Dexeus University Institute , Barcelona , Spain
| | - Cristina Teixido
- d Pangaea Oncology , Quirón-Dexeus University Institute , Barcelona , Spain
| | - Aaron E Sosa
- a Institute of Oncology Rosell (IOR), University Hospital Sagrat Cor , Barcelona , Spain
| | - Jordi Berenguer
- d Pangaea Oncology , Quirón-Dexeus University Institute , Barcelona , Spain
| | | | - Giuseppe Altavilla
- b Medical Oncology Unit, Department of Human Pathology 'G. Barresi,' University of Messina , Messina , Italy
| | - Rafael Rosell
- f Germans Trias i Pujol Research Institute , Badalona , Spain.,g Catalan Institute of Oncology , Germans Trias i Pujol University Hospital , Badalona , Spain
| |
Collapse
|
581
|
Affiliation(s)
- Ibiayi Dagogo-Jack
- Ibiayi Dagogo-Jack and Alice T. Shaw, Massachusetts General Hospital, Boston, MA
| | - Alice T Shaw
- Ibiayi Dagogo-Jack and Alice T. Shaw, Massachusetts General Hospital, Boston, MA
| |
Collapse
|
582
|
Lu H, Villafane N, Dogruluk T, Grzeskowiak CL, Kong K, Tsang YH, Zagorodna O, Pantazi A, Yang L, Neill NJ, Kim YW, Creighton CJ, Verhaak RG, Mills GB, Park PJ, Kucherlapati R, Scott KL. Engineering and Functional Characterization of Fusion Genes Identifies Novel Oncogenic Drivers of Cancer. Cancer Res 2017; 77:3502-3512. [PMID: 28512244 DOI: 10.1158/0008-5472.can-16-2745] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 03/07/2017] [Accepted: 04/27/2017] [Indexed: 01/22/2023]
Abstract
Oncogenic gene fusions drive many human cancers, but tools to more quickly unravel their functional contributions are needed. Here we describe methodology permitting fusion gene construction for functional evaluation. Using this strategy, we engineered the known fusion oncogenes, BCR-ABL1, EML4-ALK, and ETV6-NTRK3, as well as 20 previously uncharacterized fusion genes identified in The Cancer Genome Atlas datasets. In addition to confirming oncogenic activity of the known fusion oncogenes engineered by our construction strategy, we validated five novel fusion genes involving MET, NTRK2, and BRAF kinases that exhibited potent transforming activity and conferred sensitivity to FDA-approved kinase inhibitors. Our fusion construction strategy also enabled domain-function studies of BRAF fusion genes. Our results confirmed other reports that the transforming activity of BRAF fusions results from truncation-mediated loss of inhibitory domains within the N-terminus of the BRAF protein. BRAF mutations residing within this inhibitory region may provide a means for BRAF activation in cancer, therefore we leveraged the modular design of our fusion gene construction methodology to screen N-terminal domain mutations discovered in tumors that are wild-type at the BRAF mutation hotspot, V600. We identified an oncogenic mutation, F247L, whose expression robustly activated the MAPK pathway and sensitized cells to BRAF and MEK inhibitors. When applied broadly, these tools will facilitate rapid fusion gene construction for subsequent functional characterization and translation into personalized treatment strategies. Cancer Res; 77(13); 3502-12. ©2017 AACR.
Collapse
Affiliation(s)
- Hengyu Lu
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| | - Nicole Villafane
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas.,Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas
| | - Turgut Dogruluk
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| | - Caitlin L Grzeskowiak
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| | - Kathleen Kong
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| | - Yiu Huen Tsang
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| | - Oksana Zagorodna
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| | - Angeliki Pantazi
- Division of Genetics, Brigham and Women's Hospital, Boston, Massachusetts
| | - Lixing Yang
- Department of Biomedical Informatics, Harvard Medical School, Boston, Massachusetts
| | - Nicholas J Neill
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| | - Young Won Kim
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| | - Chad J Creighton
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Roel G Verhaak
- The Jackson Laboratory, Genomic Medicine, Farmington, Connecticut
| | - Gordon B Mills
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Peter J Park
- Division of Genetics, Brigham and Women's Hospital, Boston, Massachusetts.,Department of Biomedical Informatics, Harvard Medical School, Boston, Massachusetts
| | - Raju Kucherlapati
- Division of Genetics, Brigham and Women's Hospital, Boston, Massachusetts.,Department of Genetics, Harvard Medical School, Boston, Massachusetts
| | - Kenneth L Scott
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas. .,Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
583
|
Wang VE, Young L, Ali S, Miller VA, Urisman A, Wolfe J, Bivona TG, Damato B, Fogh S, Bergsland EK. A Case of Metastatic Atypical Neuroendocrine Tumor with ALK Translocation and Diffuse Brain Metastases. Oncologist 2017; 22:768-773. [PMID: 28507205 PMCID: PMC5507651 DOI: 10.1634/theoncologist.2017-0054] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 03/08/2017] [Indexed: 01/10/2023] Open
Abstract
A challenge in precision medicine is the identification of actionable driver mutations. Alterations can be identified within the tumor tissue, by small biopsy or fine‐needle aspirates, or by noninvasive methods, such as circulating tumor cells or circulating tumor DNA. This article presents a case of atypical neuroendocrine tumor metastatic to the bone and brain for which circulating tumor DNA analysis found an ALK translocation. A challenge in precision medicine requires identification of actionable driver mutations. Critical to such effort is the deployment of sensitive and well‐validated assays for mutation detection. Although identification of such alterations within the tumor tissue remains the gold standard, many advanced non‐small cell lung cancer cases have only limited tissue samples, derived from small biopsies or fine‐needle aspirates, available for testing. More recently, noninvasive methods using either circulating tumor cells or tumor DNA (ctDNA) have become an alternative method for identifying molecular biomarkers and screening patients eligible for targeted therapies. In this article, we present a case of a 52‐year‐old never‐smoking male who presented with widely metastatic atypical neuroendocrine tumor to the bones and the brain. Molecular genotyping using DNA harvested from a bone metastasis was unsuccessful due to limited material. Subsequent ctDNA analysis revealed an ALK translocation. The clinical significance of the mutation in this particular cancer type and therapeutic strategies are discussed. Key Points. To our knowledge, this index case represents the first reported ALK translocation identified in an atypical carcinoid tumor. Liquid biopsy such as circulating tumor DNA is a feasible alternative platform for identifying sensitizing genomic alterations. Second‐generation ALK inhibitors represent a new paradigm for treating ALK‐positive patients with brain metastases.
Collapse
Affiliation(s)
- Victoria E. Wang
- Department of Medicine, University of California, San Francisco, San Francisco, California, USA
- Correspondence: Victoria E. Wang, M.D., Ph.D., Department of Medicine, University of California, San Francisco, 1600 Divisadero Street, San Francisco, California 94115, USA
| | - Lauren Young
- Foundation Medicine, Cambridge, Massachusetts, USA
| | - Siraj Ali
- Foundation Medicine, Cambridge, Massachusetts, USA
| | | | - Anatoly Urisman
- Department of Medicine Pathology, University of California, San Francisco, San Francisco, California, USA
| | - John Wolfe
- Department of Pathology, Santa Rosa Memorial Hospital, Santa Rosa, California, USA
| | - Trever G. Bivona
- Department of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Bertil Damato
- Department of Medicine Ophthalmology, University of California, San Francisco, San Francisco, California, USA
| | - Shannon Fogh
- Department of Medicine Radiation Oncology, University of California, San Francisco, San Francisco, California, USA
| | - Emily K. Bergsland
- Department of Medicine, University of California, San Francisco, San Francisco, California, USA
| |
Collapse
|
584
|
Choi SH, Kim DH, Choi YJ, Kim SY, Lee JE, Sung KJ, Kim WS, Choi CM, Rho JK, Lee JC. Multiple receptor tyrosine kinase activation related to ALK inhibitor resistance in lung cancer cells with ALK rearrangement. Oncotarget 2017; 8:58771-58780. [PMID: 28938595 PMCID: PMC5601691 DOI: 10.18632/oncotarget.17680] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 03/22/2017] [Indexed: 01/30/2023] Open
Abstract
The activation of alternative receptor tyrosine kinases (RTKs) is known to mediate resistance to ALK inhibitors. However, the role of multiple RTK activation in resistance has yet to be determined. Two crizotinib-resistant (H3122/CR-1 and H3122/CR-2) and one TAE684-resistant (H2228/TR) cell lines were established. Multi-RTK arrays and Western blots were performed to detect the activation of bypass signals. There were no secondary mutations in the sequencing. EGFR and MET were activated in H3122/CR-1 cells whereas EGFR and IGF1R were activated in H3122/CR-2 cells. Concomitant activation of MET did not contribute to resistance as crizotinib completely suppressed both p-MET and p-ALK in H3122/CR-1 cells, whose survival was not affected by crizotinib. However, combined inhibition of EGFR and ALK was effective in controlling this resistant cell line. In H3122/CR-2 cells, the inhibition of both ALK and IGF1R could effectively suppress cell growth, whereas simultaneous inhibition of ALK and EGFR brought about a less-effective suppression, indicating that IGF1R activation is the main resistance mechanism. H2228/TR cells showed activation of the HER family (EGFR, ErbB2, and ErbB3). Afatinib, a pan-HER inhibitor, was more potent in suppressing resistant cells than gefitinib when combined with crizotinib, which suggests that coactivation of ErbB2 and ErbB3 also contributes to resistance. Interestingly, all three resistant cell lines responded well to AUY922, which can inhibit ALK, EGFR, and IGF1R activity. Activation of multiple RTKs can occur during acquired resistance to ALK inhibitors, in which case the dominant or significant bypass signal should be identified to provide a more appropriate combination therapy.
Collapse
Affiliation(s)
- Se Hoon Choi
- Department of Thoracic and Cardiovascular Surgery, Asan Medical Center, University of Ulsan, College of Medicine, Seoul, Korea
| | - Dong Ha Kim
- Department of Pulmonology and Critical Care Medicine, Asan Medical Center, University of Ulsan, College of Medicine, Seoul, Korea.,Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan, College of Medicine, Seoul, Korea
| | - Yun Jung Choi
- Department of Pulmonology and Critical Care Medicine, Asan Medical Center, University of Ulsan, College of Medicine, Seoul, Korea.,Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan, College of Medicine, Seoul, Korea
| | - Seon Ye Kim
- Department of Pulmonology and Critical Care Medicine, Asan Medical Center, University of Ulsan, College of Medicine, Seoul, Korea.,Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan, College of Medicine, Seoul, Korea
| | - Jung-Eun Lee
- Department of Pulmonology and Critical Care Medicine, Asan Medical Center, University of Ulsan, College of Medicine, Seoul, Korea.,Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan, College of Medicine, Seoul, Korea
| | - Ki Jung Sung
- Department of Pulmonology and Critical Care Medicine, Asan Medical Center, University of Ulsan, College of Medicine, Seoul, Korea.,Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan, College of Medicine, Seoul, Korea
| | - Woo Sung Kim
- Department of Pulmonology and Critical Care Medicine, Asan Medical Center, University of Ulsan, College of Medicine, Seoul, Korea
| | - Chang-Min Choi
- Department of Pulmonology and Critical Care Medicine, Asan Medical Center, University of Ulsan, College of Medicine, Seoul, Korea.,Department of Oncology, Asan Medical Center, University of Ulsan, College of Medicine, Seoul, Korea
| | - Jin Kyung Rho
- Department of Pulmonology and Critical Care Medicine, Asan Medical Center, University of Ulsan, College of Medicine, Seoul, Korea.,Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan, College of Medicine, Seoul, Korea.,Department of Convergence Medicine, Asan Medical Center, University of Ulsan, College of Medicine, Seoul, Korea
| | - Jae Cheol Lee
- Department of Oncology, Asan Medical Center, University of Ulsan, College of Medicine, Seoul, Korea
| |
Collapse
|
585
|
Sokolowska-Wedzina A, Chodaczek G, Chudzian J, Borek A, Zakrzewska M, Otlewski J. High-Affinity Internalizing Human scFv-Fc Antibody for Targeting FGFR1-Overexpressing Lung Cancer. Mol Cancer Res 2017; 15:1040-1050. [DOI: 10.1158/1541-7786.mcr-16-0136] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 12/27/2016] [Accepted: 05/02/2017] [Indexed: 11/16/2022]
|
586
|
Ocular toxicities associated with targeted anticancer agents: an analysis of clinical data with management suggestions. Oncotarget 2017; 8:58709-58727. [PMID: 28938590 PMCID: PMC5601686 DOI: 10.18632/oncotarget.17634] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 03/16/2017] [Indexed: 11/25/2022] Open
Abstract
Ocular toxicities are among the most common adverse events resulting from targeted anticancer agents and are becoming increasingly relevant in the management of patients on these agents. The purpose of this study is to provide a framework for management of these challenging toxicities based on objective data from FDA labels and from analysis of the literature. All oncologic drugs approved by the FDA up to March 14, 2015, were screened for inclusion. A total of 16 drugs (12 small-molecule drugs and 4 monoclonal antibodies) were analyzed for ocular toxicity profiles based on evidence of ocular toxicity. Trials cited by FDA labels were retrieved, and a combination search in Medline, Google Scholar, the Cochrane database, and the NIH Clinical Trials Database was conducted. The majority of ocular toxicities reported were low severity, and the most common were conjunctivitis and "visual disturbances." However, severe events including incidents of blindness, retinal vascular occlusion, and corneal ulceration occurred. The frequency and severity at which ocular toxicities occur merits a more multidisciplinary approach to managing patients with agents that are known to cause ocular issues. We suggest a standardized methodology for referral and surveillance of patients who are potentially at risk of severe ocular toxicity.
Collapse
|
587
|
Kim DW, Tiseo M, Ahn MJ, Reckamp KL, Hansen KH, Kim SW, Huber RM, West HL, Groen HJM, Hochmair MJ, Leighl NB, Gettinger SN, Langer CJ, Paz-Ares Rodríguez LG, Smit EF, Kim ES, Reichmann W, Haluska FG, Kerstein D, Camidge DR. Brigatinib in Patients With Crizotinib-Refractory Anaplastic Lymphoma Kinase-Positive Non-Small-Cell Lung Cancer: A Randomized, Multicenter Phase II Trial. J Clin Oncol 2017; 35:2490-2498. [PMID: 28475456 DOI: 10.1200/jco.2016.71.5904] [Citation(s) in RCA: 448] [Impact Index Per Article: 56.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Purpose Most crizotinib-treated patients with anaplastic lymphoma kinase gene ( ALK)-rearranged non-small-cell lung cancer (ALK-positive NSCLC) eventually experience disease progression. We evaluated two regimens of brigatinib, an investigational next-generation ALK inhibitor, in crizotinib-refractory ALK-positive NSCLC. Patients and Methods Patients were stratified by brain metastases and best response to crizotinib. They were randomly assigned (1:1) to oral brigatinib 90 mg once daily (arm A) or 180 mg once daily with a 7-day lead-in at 90 mg (180 mg once daily [with lead-in]; arm B). Investigator-assessed confirmed objective response rate (ORR) was the primary end point. Results Of 222 patients enrolled (arm A: n = 112, 109 treated; arm B: n = 110, 110 treated), 154 (69%) had baseline brain metastases and 164 of 222 (74%) had received prior chemotherapy. With 8.0-month median follow-up, investigator-assessed confirmed ORR was 45% (97.5% CI, 34% to 56%) in arm A and 54% (97.5% CI, 43% to 65%) in arm B. Investigator-assessed median progression-free survival was 9.2 months (95% CI, 7.4 to 15.6) and 12.9 months (95% CI, 11.1 to not reached) in arms A and B, respectively. Independent review committee-assessed intracranial ORR in patients with measurable brain metastases at baseline was 42% (11 of 26 patients) in arm A and 67% (12 of 18 patients) in arm B. Common treatment-emergent adverse events were nausea (arm A/B, 33%/40%), diarrhea (arm A/B, 19%/38%), headache (arm A/B, 28%/27%), and cough (arm A/B, 18%/34%), and were mainly grades 1 to 2. A subset of pulmonary adverse events with early onset (median onset: day 2) occurred in 14 of 219 treated patients (all grades, 6%; grade ≥ 3, 3%); none occurred after escalation to 180 mg in arm B. Seven of 14 patients were successfully retreated with brigatinib. Conclusion Brigatinib yielded substantial whole-body and intracranial responses as well as robust progression-free survival; 180 mg (with lead-in) showed consistently better efficacy than 90 mg, with acceptable safety.
Collapse
Affiliation(s)
- Dong-Wan Kim
- Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center; Sang-We Kim, Asan Medical Center, Seoul, South Korea; Marcello Tiseo, University Hospital of Parma, Parma, Italy; Karen L. Reckamp, City of Hope, Duarte, CA; Karin Holmskov Hansen, Odense University Hospital, Odense, Denmark; Rudolf M. Huber, University Hospital of Munich, German Centre for Lung Research, Munich, Germany; Howard L. West, Swedish Cancer Institute, Seattle, WA; Harry J.M. Groen, University of Groningen, University Medical Center Groningen, Groningen; Egbert F. Smit, VU University Medical Center, Amsterdam, the Netherlands; Maximilian J. Hochmair, Otto Wagner Hospital, Vienna, Austria; Natasha B. Leighl, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Scott N. Gettinger, Yale Cancer Center, New Haven, CT; Corey J. Langer, University of Pennsylvania Abramson Cancer Center, Philadelphia, PA; Luis G. Paz-Ares Rodríguez, Hospital Universitario 12 de Octubre, Madrid, Spain; Edward S. Kim, Levine Cancer Institute, Carolinas HealthCare System, Charlotte, NC; William Reichmann, Frank G. Haluska, and David Kerstein, ARIAD Pharmaceuticals, Cambridge, MA; and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO
| | - Marcello Tiseo
- Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center; Sang-We Kim, Asan Medical Center, Seoul, South Korea; Marcello Tiseo, University Hospital of Parma, Parma, Italy; Karen L. Reckamp, City of Hope, Duarte, CA; Karin Holmskov Hansen, Odense University Hospital, Odense, Denmark; Rudolf M. Huber, University Hospital of Munich, German Centre for Lung Research, Munich, Germany; Howard L. West, Swedish Cancer Institute, Seattle, WA; Harry J.M. Groen, University of Groningen, University Medical Center Groningen, Groningen; Egbert F. Smit, VU University Medical Center, Amsterdam, the Netherlands; Maximilian J. Hochmair, Otto Wagner Hospital, Vienna, Austria; Natasha B. Leighl, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Scott N. Gettinger, Yale Cancer Center, New Haven, CT; Corey J. Langer, University of Pennsylvania Abramson Cancer Center, Philadelphia, PA; Luis G. Paz-Ares Rodríguez, Hospital Universitario 12 de Octubre, Madrid, Spain; Edward S. Kim, Levine Cancer Institute, Carolinas HealthCare System, Charlotte, NC; William Reichmann, Frank G. Haluska, and David Kerstein, ARIAD Pharmaceuticals, Cambridge, MA; and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO
| | - Myung-Ju Ahn
- Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center; Sang-We Kim, Asan Medical Center, Seoul, South Korea; Marcello Tiseo, University Hospital of Parma, Parma, Italy; Karen L. Reckamp, City of Hope, Duarte, CA; Karin Holmskov Hansen, Odense University Hospital, Odense, Denmark; Rudolf M. Huber, University Hospital of Munich, German Centre for Lung Research, Munich, Germany; Howard L. West, Swedish Cancer Institute, Seattle, WA; Harry J.M. Groen, University of Groningen, University Medical Center Groningen, Groningen; Egbert F. Smit, VU University Medical Center, Amsterdam, the Netherlands; Maximilian J. Hochmair, Otto Wagner Hospital, Vienna, Austria; Natasha B. Leighl, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Scott N. Gettinger, Yale Cancer Center, New Haven, CT; Corey J. Langer, University of Pennsylvania Abramson Cancer Center, Philadelphia, PA; Luis G. Paz-Ares Rodríguez, Hospital Universitario 12 de Octubre, Madrid, Spain; Edward S. Kim, Levine Cancer Institute, Carolinas HealthCare System, Charlotte, NC; William Reichmann, Frank G. Haluska, and David Kerstein, ARIAD Pharmaceuticals, Cambridge, MA; and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO
| | - Karen L Reckamp
- Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center; Sang-We Kim, Asan Medical Center, Seoul, South Korea; Marcello Tiseo, University Hospital of Parma, Parma, Italy; Karen L. Reckamp, City of Hope, Duarte, CA; Karin Holmskov Hansen, Odense University Hospital, Odense, Denmark; Rudolf M. Huber, University Hospital of Munich, German Centre for Lung Research, Munich, Germany; Howard L. West, Swedish Cancer Institute, Seattle, WA; Harry J.M. Groen, University of Groningen, University Medical Center Groningen, Groningen; Egbert F. Smit, VU University Medical Center, Amsterdam, the Netherlands; Maximilian J. Hochmair, Otto Wagner Hospital, Vienna, Austria; Natasha B. Leighl, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Scott N. Gettinger, Yale Cancer Center, New Haven, CT; Corey J. Langer, University of Pennsylvania Abramson Cancer Center, Philadelphia, PA; Luis G. Paz-Ares Rodríguez, Hospital Universitario 12 de Octubre, Madrid, Spain; Edward S. Kim, Levine Cancer Institute, Carolinas HealthCare System, Charlotte, NC; William Reichmann, Frank G. Haluska, and David Kerstein, ARIAD Pharmaceuticals, Cambridge, MA; and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO
| | - Karin Holmskov Hansen
- Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center; Sang-We Kim, Asan Medical Center, Seoul, South Korea; Marcello Tiseo, University Hospital of Parma, Parma, Italy; Karen L. Reckamp, City of Hope, Duarte, CA; Karin Holmskov Hansen, Odense University Hospital, Odense, Denmark; Rudolf M. Huber, University Hospital of Munich, German Centre for Lung Research, Munich, Germany; Howard L. West, Swedish Cancer Institute, Seattle, WA; Harry J.M. Groen, University of Groningen, University Medical Center Groningen, Groningen; Egbert F. Smit, VU University Medical Center, Amsterdam, the Netherlands; Maximilian J. Hochmair, Otto Wagner Hospital, Vienna, Austria; Natasha B. Leighl, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Scott N. Gettinger, Yale Cancer Center, New Haven, CT; Corey J. Langer, University of Pennsylvania Abramson Cancer Center, Philadelphia, PA; Luis G. Paz-Ares Rodríguez, Hospital Universitario 12 de Octubre, Madrid, Spain; Edward S. Kim, Levine Cancer Institute, Carolinas HealthCare System, Charlotte, NC; William Reichmann, Frank G. Haluska, and David Kerstein, ARIAD Pharmaceuticals, Cambridge, MA; and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO
| | - Sang-We Kim
- Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center; Sang-We Kim, Asan Medical Center, Seoul, South Korea; Marcello Tiseo, University Hospital of Parma, Parma, Italy; Karen L. Reckamp, City of Hope, Duarte, CA; Karin Holmskov Hansen, Odense University Hospital, Odense, Denmark; Rudolf M. Huber, University Hospital of Munich, German Centre for Lung Research, Munich, Germany; Howard L. West, Swedish Cancer Institute, Seattle, WA; Harry J.M. Groen, University of Groningen, University Medical Center Groningen, Groningen; Egbert F. Smit, VU University Medical Center, Amsterdam, the Netherlands; Maximilian J. Hochmair, Otto Wagner Hospital, Vienna, Austria; Natasha B. Leighl, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Scott N. Gettinger, Yale Cancer Center, New Haven, CT; Corey J. Langer, University of Pennsylvania Abramson Cancer Center, Philadelphia, PA; Luis G. Paz-Ares Rodríguez, Hospital Universitario 12 de Octubre, Madrid, Spain; Edward S. Kim, Levine Cancer Institute, Carolinas HealthCare System, Charlotte, NC; William Reichmann, Frank G. Haluska, and David Kerstein, ARIAD Pharmaceuticals, Cambridge, MA; and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO
| | - Rudolf M Huber
- Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center; Sang-We Kim, Asan Medical Center, Seoul, South Korea; Marcello Tiseo, University Hospital of Parma, Parma, Italy; Karen L. Reckamp, City of Hope, Duarte, CA; Karin Holmskov Hansen, Odense University Hospital, Odense, Denmark; Rudolf M. Huber, University Hospital of Munich, German Centre for Lung Research, Munich, Germany; Howard L. West, Swedish Cancer Institute, Seattle, WA; Harry J.M. Groen, University of Groningen, University Medical Center Groningen, Groningen; Egbert F. Smit, VU University Medical Center, Amsterdam, the Netherlands; Maximilian J. Hochmair, Otto Wagner Hospital, Vienna, Austria; Natasha B. Leighl, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Scott N. Gettinger, Yale Cancer Center, New Haven, CT; Corey J. Langer, University of Pennsylvania Abramson Cancer Center, Philadelphia, PA; Luis G. Paz-Ares Rodríguez, Hospital Universitario 12 de Octubre, Madrid, Spain; Edward S. Kim, Levine Cancer Institute, Carolinas HealthCare System, Charlotte, NC; William Reichmann, Frank G. Haluska, and David Kerstein, ARIAD Pharmaceuticals, Cambridge, MA; and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO
| | - Howard L West
- Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center; Sang-We Kim, Asan Medical Center, Seoul, South Korea; Marcello Tiseo, University Hospital of Parma, Parma, Italy; Karen L. Reckamp, City of Hope, Duarte, CA; Karin Holmskov Hansen, Odense University Hospital, Odense, Denmark; Rudolf M. Huber, University Hospital of Munich, German Centre for Lung Research, Munich, Germany; Howard L. West, Swedish Cancer Institute, Seattle, WA; Harry J.M. Groen, University of Groningen, University Medical Center Groningen, Groningen; Egbert F. Smit, VU University Medical Center, Amsterdam, the Netherlands; Maximilian J. Hochmair, Otto Wagner Hospital, Vienna, Austria; Natasha B. Leighl, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Scott N. Gettinger, Yale Cancer Center, New Haven, CT; Corey J. Langer, University of Pennsylvania Abramson Cancer Center, Philadelphia, PA; Luis G. Paz-Ares Rodríguez, Hospital Universitario 12 de Octubre, Madrid, Spain; Edward S. Kim, Levine Cancer Institute, Carolinas HealthCare System, Charlotte, NC; William Reichmann, Frank G. Haluska, and David Kerstein, ARIAD Pharmaceuticals, Cambridge, MA; and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO
| | - Harry J M Groen
- Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center; Sang-We Kim, Asan Medical Center, Seoul, South Korea; Marcello Tiseo, University Hospital of Parma, Parma, Italy; Karen L. Reckamp, City of Hope, Duarte, CA; Karin Holmskov Hansen, Odense University Hospital, Odense, Denmark; Rudolf M. Huber, University Hospital of Munich, German Centre for Lung Research, Munich, Germany; Howard L. West, Swedish Cancer Institute, Seattle, WA; Harry J.M. Groen, University of Groningen, University Medical Center Groningen, Groningen; Egbert F. Smit, VU University Medical Center, Amsterdam, the Netherlands; Maximilian J. Hochmair, Otto Wagner Hospital, Vienna, Austria; Natasha B. Leighl, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Scott N. Gettinger, Yale Cancer Center, New Haven, CT; Corey J. Langer, University of Pennsylvania Abramson Cancer Center, Philadelphia, PA; Luis G. Paz-Ares Rodríguez, Hospital Universitario 12 de Octubre, Madrid, Spain; Edward S. Kim, Levine Cancer Institute, Carolinas HealthCare System, Charlotte, NC; William Reichmann, Frank G. Haluska, and David Kerstein, ARIAD Pharmaceuticals, Cambridge, MA; and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO
| | - Maximilian J Hochmair
- Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center; Sang-We Kim, Asan Medical Center, Seoul, South Korea; Marcello Tiseo, University Hospital of Parma, Parma, Italy; Karen L. Reckamp, City of Hope, Duarte, CA; Karin Holmskov Hansen, Odense University Hospital, Odense, Denmark; Rudolf M. Huber, University Hospital of Munich, German Centre for Lung Research, Munich, Germany; Howard L. West, Swedish Cancer Institute, Seattle, WA; Harry J.M. Groen, University of Groningen, University Medical Center Groningen, Groningen; Egbert F. Smit, VU University Medical Center, Amsterdam, the Netherlands; Maximilian J. Hochmair, Otto Wagner Hospital, Vienna, Austria; Natasha B. Leighl, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Scott N. Gettinger, Yale Cancer Center, New Haven, CT; Corey J. Langer, University of Pennsylvania Abramson Cancer Center, Philadelphia, PA; Luis G. Paz-Ares Rodríguez, Hospital Universitario 12 de Octubre, Madrid, Spain; Edward S. Kim, Levine Cancer Institute, Carolinas HealthCare System, Charlotte, NC; William Reichmann, Frank G. Haluska, and David Kerstein, ARIAD Pharmaceuticals, Cambridge, MA; and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO
| | - Natasha B Leighl
- Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center; Sang-We Kim, Asan Medical Center, Seoul, South Korea; Marcello Tiseo, University Hospital of Parma, Parma, Italy; Karen L. Reckamp, City of Hope, Duarte, CA; Karin Holmskov Hansen, Odense University Hospital, Odense, Denmark; Rudolf M. Huber, University Hospital of Munich, German Centre for Lung Research, Munich, Germany; Howard L. West, Swedish Cancer Institute, Seattle, WA; Harry J.M. Groen, University of Groningen, University Medical Center Groningen, Groningen; Egbert F. Smit, VU University Medical Center, Amsterdam, the Netherlands; Maximilian J. Hochmair, Otto Wagner Hospital, Vienna, Austria; Natasha B. Leighl, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Scott N. Gettinger, Yale Cancer Center, New Haven, CT; Corey J. Langer, University of Pennsylvania Abramson Cancer Center, Philadelphia, PA; Luis G. Paz-Ares Rodríguez, Hospital Universitario 12 de Octubre, Madrid, Spain; Edward S. Kim, Levine Cancer Institute, Carolinas HealthCare System, Charlotte, NC; William Reichmann, Frank G. Haluska, and David Kerstein, ARIAD Pharmaceuticals, Cambridge, MA; and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO
| | - Scott N Gettinger
- Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center; Sang-We Kim, Asan Medical Center, Seoul, South Korea; Marcello Tiseo, University Hospital of Parma, Parma, Italy; Karen L. Reckamp, City of Hope, Duarte, CA; Karin Holmskov Hansen, Odense University Hospital, Odense, Denmark; Rudolf M. Huber, University Hospital of Munich, German Centre for Lung Research, Munich, Germany; Howard L. West, Swedish Cancer Institute, Seattle, WA; Harry J.M. Groen, University of Groningen, University Medical Center Groningen, Groningen; Egbert F. Smit, VU University Medical Center, Amsterdam, the Netherlands; Maximilian J. Hochmair, Otto Wagner Hospital, Vienna, Austria; Natasha B. Leighl, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Scott N. Gettinger, Yale Cancer Center, New Haven, CT; Corey J. Langer, University of Pennsylvania Abramson Cancer Center, Philadelphia, PA; Luis G. Paz-Ares Rodríguez, Hospital Universitario 12 de Octubre, Madrid, Spain; Edward S. Kim, Levine Cancer Institute, Carolinas HealthCare System, Charlotte, NC; William Reichmann, Frank G. Haluska, and David Kerstein, ARIAD Pharmaceuticals, Cambridge, MA; and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO
| | - Corey J Langer
- Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center; Sang-We Kim, Asan Medical Center, Seoul, South Korea; Marcello Tiseo, University Hospital of Parma, Parma, Italy; Karen L. Reckamp, City of Hope, Duarte, CA; Karin Holmskov Hansen, Odense University Hospital, Odense, Denmark; Rudolf M. Huber, University Hospital of Munich, German Centre for Lung Research, Munich, Germany; Howard L. West, Swedish Cancer Institute, Seattle, WA; Harry J.M. Groen, University of Groningen, University Medical Center Groningen, Groningen; Egbert F. Smit, VU University Medical Center, Amsterdam, the Netherlands; Maximilian J. Hochmair, Otto Wagner Hospital, Vienna, Austria; Natasha B. Leighl, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Scott N. Gettinger, Yale Cancer Center, New Haven, CT; Corey J. Langer, University of Pennsylvania Abramson Cancer Center, Philadelphia, PA; Luis G. Paz-Ares Rodríguez, Hospital Universitario 12 de Octubre, Madrid, Spain; Edward S. Kim, Levine Cancer Institute, Carolinas HealthCare System, Charlotte, NC; William Reichmann, Frank G. Haluska, and David Kerstein, ARIAD Pharmaceuticals, Cambridge, MA; and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO
| | - Luis G Paz-Ares Rodríguez
- Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center; Sang-We Kim, Asan Medical Center, Seoul, South Korea; Marcello Tiseo, University Hospital of Parma, Parma, Italy; Karen L. Reckamp, City of Hope, Duarte, CA; Karin Holmskov Hansen, Odense University Hospital, Odense, Denmark; Rudolf M. Huber, University Hospital of Munich, German Centre for Lung Research, Munich, Germany; Howard L. West, Swedish Cancer Institute, Seattle, WA; Harry J.M. Groen, University of Groningen, University Medical Center Groningen, Groningen; Egbert F. Smit, VU University Medical Center, Amsterdam, the Netherlands; Maximilian J. Hochmair, Otto Wagner Hospital, Vienna, Austria; Natasha B. Leighl, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Scott N. Gettinger, Yale Cancer Center, New Haven, CT; Corey J. Langer, University of Pennsylvania Abramson Cancer Center, Philadelphia, PA; Luis G. Paz-Ares Rodríguez, Hospital Universitario 12 de Octubre, Madrid, Spain; Edward S. Kim, Levine Cancer Institute, Carolinas HealthCare System, Charlotte, NC; William Reichmann, Frank G. Haluska, and David Kerstein, ARIAD Pharmaceuticals, Cambridge, MA; and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO
| | - Egbert F Smit
- Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center; Sang-We Kim, Asan Medical Center, Seoul, South Korea; Marcello Tiseo, University Hospital of Parma, Parma, Italy; Karen L. Reckamp, City of Hope, Duarte, CA; Karin Holmskov Hansen, Odense University Hospital, Odense, Denmark; Rudolf M. Huber, University Hospital of Munich, German Centre for Lung Research, Munich, Germany; Howard L. West, Swedish Cancer Institute, Seattle, WA; Harry J.M. Groen, University of Groningen, University Medical Center Groningen, Groningen; Egbert F. Smit, VU University Medical Center, Amsterdam, the Netherlands; Maximilian J. Hochmair, Otto Wagner Hospital, Vienna, Austria; Natasha B. Leighl, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Scott N. Gettinger, Yale Cancer Center, New Haven, CT; Corey J. Langer, University of Pennsylvania Abramson Cancer Center, Philadelphia, PA; Luis G. Paz-Ares Rodríguez, Hospital Universitario 12 de Octubre, Madrid, Spain; Edward S. Kim, Levine Cancer Institute, Carolinas HealthCare System, Charlotte, NC; William Reichmann, Frank G. Haluska, and David Kerstein, ARIAD Pharmaceuticals, Cambridge, MA; and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO
| | - Edward S Kim
- Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center; Sang-We Kim, Asan Medical Center, Seoul, South Korea; Marcello Tiseo, University Hospital of Parma, Parma, Italy; Karen L. Reckamp, City of Hope, Duarte, CA; Karin Holmskov Hansen, Odense University Hospital, Odense, Denmark; Rudolf M. Huber, University Hospital of Munich, German Centre for Lung Research, Munich, Germany; Howard L. West, Swedish Cancer Institute, Seattle, WA; Harry J.M. Groen, University of Groningen, University Medical Center Groningen, Groningen; Egbert F. Smit, VU University Medical Center, Amsterdam, the Netherlands; Maximilian J. Hochmair, Otto Wagner Hospital, Vienna, Austria; Natasha B. Leighl, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Scott N. Gettinger, Yale Cancer Center, New Haven, CT; Corey J. Langer, University of Pennsylvania Abramson Cancer Center, Philadelphia, PA; Luis G. Paz-Ares Rodríguez, Hospital Universitario 12 de Octubre, Madrid, Spain; Edward S. Kim, Levine Cancer Institute, Carolinas HealthCare System, Charlotte, NC; William Reichmann, Frank G. Haluska, and David Kerstein, ARIAD Pharmaceuticals, Cambridge, MA; and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO
| | - William Reichmann
- Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center; Sang-We Kim, Asan Medical Center, Seoul, South Korea; Marcello Tiseo, University Hospital of Parma, Parma, Italy; Karen L. Reckamp, City of Hope, Duarte, CA; Karin Holmskov Hansen, Odense University Hospital, Odense, Denmark; Rudolf M. Huber, University Hospital of Munich, German Centre for Lung Research, Munich, Germany; Howard L. West, Swedish Cancer Institute, Seattle, WA; Harry J.M. Groen, University of Groningen, University Medical Center Groningen, Groningen; Egbert F. Smit, VU University Medical Center, Amsterdam, the Netherlands; Maximilian J. Hochmair, Otto Wagner Hospital, Vienna, Austria; Natasha B. Leighl, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Scott N. Gettinger, Yale Cancer Center, New Haven, CT; Corey J. Langer, University of Pennsylvania Abramson Cancer Center, Philadelphia, PA; Luis G. Paz-Ares Rodríguez, Hospital Universitario 12 de Octubre, Madrid, Spain; Edward S. Kim, Levine Cancer Institute, Carolinas HealthCare System, Charlotte, NC; William Reichmann, Frank G. Haluska, and David Kerstein, ARIAD Pharmaceuticals, Cambridge, MA; and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO
| | - Frank G Haluska
- Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center; Sang-We Kim, Asan Medical Center, Seoul, South Korea; Marcello Tiseo, University Hospital of Parma, Parma, Italy; Karen L. Reckamp, City of Hope, Duarte, CA; Karin Holmskov Hansen, Odense University Hospital, Odense, Denmark; Rudolf M. Huber, University Hospital of Munich, German Centre for Lung Research, Munich, Germany; Howard L. West, Swedish Cancer Institute, Seattle, WA; Harry J.M. Groen, University of Groningen, University Medical Center Groningen, Groningen; Egbert F. Smit, VU University Medical Center, Amsterdam, the Netherlands; Maximilian J. Hochmair, Otto Wagner Hospital, Vienna, Austria; Natasha B. Leighl, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Scott N. Gettinger, Yale Cancer Center, New Haven, CT; Corey J. Langer, University of Pennsylvania Abramson Cancer Center, Philadelphia, PA; Luis G. Paz-Ares Rodríguez, Hospital Universitario 12 de Octubre, Madrid, Spain; Edward S. Kim, Levine Cancer Institute, Carolinas HealthCare System, Charlotte, NC; William Reichmann, Frank G. Haluska, and David Kerstein, ARIAD Pharmaceuticals, Cambridge, MA; and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO
| | - David Kerstein
- Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center; Sang-We Kim, Asan Medical Center, Seoul, South Korea; Marcello Tiseo, University Hospital of Parma, Parma, Italy; Karen L. Reckamp, City of Hope, Duarte, CA; Karin Holmskov Hansen, Odense University Hospital, Odense, Denmark; Rudolf M. Huber, University Hospital of Munich, German Centre for Lung Research, Munich, Germany; Howard L. West, Swedish Cancer Institute, Seattle, WA; Harry J.M. Groen, University of Groningen, University Medical Center Groningen, Groningen; Egbert F. Smit, VU University Medical Center, Amsterdam, the Netherlands; Maximilian J. Hochmair, Otto Wagner Hospital, Vienna, Austria; Natasha B. Leighl, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Scott N. Gettinger, Yale Cancer Center, New Haven, CT; Corey J. Langer, University of Pennsylvania Abramson Cancer Center, Philadelphia, PA; Luis G. Paz-Ares Rodríguez, Hospital Universitario 12 de Octubre, Madrid, Spain; Edward S. Kim, Levine Cancer Institute, Carolinas HealthCare System, Charlotte, NC; William Reichmann, Frank G. Haluska, and David Kerstein, ARIAD Pharmaceuticals, Cambridge, MA; and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO
| | - D Ross Camidge
- Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center; Sang-We Kim, Asan Medical Center, Seoul, South Korea; Marcello Tiseo, University Hospital of Parma, Parma, Italy; Karen L. Reckamp, City of Hope, Duarte, CA; Karin Holmskov Hansen, Odense University Hospital, Odense, Denmark; Rudolf M. Huber, University Hospital of Munich, German Centre for Lung Research, Munich, Germany; Howard L. West, Swedish Cancer Institute, Seattle, WA; Harry J.M. Groen, University of Groningen, University Medical Center Groningen, Groningen; Egbert F. Smit, VU University Medical Center, Amsterdam, the Netherlands; Maximilian J. Hochmair, Otto Wagner Hospital, Vienna, Austria; Natasha B. Leighl, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Scott N. Gettinger, Yale Cancer Center, New Haven, CT; Corey J. Langer, University of Pennsylvania Abramson Cancer Center, Philadelphia, PA; Luis G. Paz-Ares Rodríguez, Hospital Universitario 12 de Octubre, Madrid, Spain; Edward S. Kim, Levine Cancer Institute, Carolinas HealthCare System, Charlotte, NC; William Reichmann, Frank G. Haluska, and David Kerstein, ARIAD Pharmaceuticals, Cambridge, MA; and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO
| |
Collapse
|
588
|
Jang J, Son JB, To C, Bahcall M, Kim SY, Kang SY, Mushajiang M, Lee Y, Jänne PA, Choi HG, Gray NS. Discovery of a potent dual ALK and EGFR T790M inhibitor. Eur J Med Chem 2017; 136:497-510. [PMID: 28528303 DOI: 10.1016/j.ejmech.2017.04.079] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 04/27/2017] [Accepted: 04/30/2017] [Indexed: 12/13/2022]
Abstract
The mutational activations of anaplastic lymphoma kinase (ALK) and epidermal growth factor receptor (EGFR) are validated oncogenic events and the targets of approved drugs to treat non-small cell lung cancer (NSCLC). Here we report highly potent dual small molecule inhibitors of both ALK and EGFR, particularly the T790M mutant which confers resistance to first generation EGFR inhibitors. Dual ALK/EGFR inhibitors may provide an efficient approach to prevent resistance that arises as a consequence of clinically reported reciprocal activation mechanisms. Our lead compound 7c displayed remarkable inhibitory activities against both ALK and EGFR in enzymatic and cellular assays. We demonstrate that 7c is capable of recapitulating the signaling effects and antiproliferative activity of combined treatment with the approved ALK inhibitor ceritinib and T790M EGFR inhibitor osimertinib against patient-derived non-small cell lung cancer cell line, DFCI032 which harbors both EML4-ALK and activated EGFR.
Collapse
Affiliation(s)
- Jaebong Jang
- Department of Biological Chemistry & Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, United States; Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, United States
| | - Jung Beom Son
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea
| | - Ciric To
- Harvard Medical School, Boston, MA 02115, United States; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, United States
| | - Magda Bahcall
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, United States
| | - So Young Kim
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea
| | - Seock Yong Kang
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea
| | - Mierzhati Mushajiang
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, United States
| | - Younho Lee
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea
| | - Pasi A Jänne
- Harvard Medical School, Boston, MA 02115, United States; Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, United States; Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, MA 02215, United States; Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, United States
| | - Hwan Geun Choi
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea.
| | - Nathanael S Gray
- Department of Biological Chemistry & Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, United States; Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, United States.
| |
Collapse
|
589
|
Woo CG, Seo S, Kim SW, Jang SJ, Park KS, Song JY, Lee B, Richards MW, Bayliss R, Lee DH, Choi J. Differential protein stability and clinical responses of EML4-ALK fusion variants to various ALK inhibitors in advanced ALK-rearranged non-small cell lung cancer. Ann Oncol 2017; 28:791-797. [PMID: 28039177 DOI: 10.1093/annonc/mdw693] [Citation(s) in RCA: 164] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Indexed: 11/14/2022] Open
Abstract
Background Anaplastic lymphoma kinase (ALK) inhibition using crizotinib has become the standard of care in advanced ALK-rearranged non-small cell lung cancer (NSCLC), but the treatment outcomes and duration of response vary widely. Echinoderm microtubule-associated protein-like 4 (EML4)-ALK is the most common translocation, and the fusion variants show different sensitivity to crizotinib in vitro. However, there are only limited data on the specific EML4-ALK variants and clinical responses of patients to various ALK inhibitors. Patients and methods By multiplex reverse-transcriptase PCR, which detects 12 variants of known EML4-ALK rearrangements, we retrospectively determined ALK fusion variants in 54 advanced ALK rearrangement-positive NSCLCs. We subdivided the patients into two groups (variants 1/2/others and variants 3a/b) by protein stability and evaluated correlations of the variant status with clinical responses to crizotinib, alectinib, or ceritinib. Moreover, we established the EML4-ALK variant-expressing system and analyzed patterns of sensitivity of the variants to ALK inhibitors. Results Of the 54 tumors analyzed, EML4-ALK variants 3a/b (44.4%) was the most common type, followed by variants 1 (33.3%) and 2 (11.1%). The 2-year progression-free survival (PFS) rate was 76.0% [95% confidence interval (CI) 56.8-100] in group EML4-ALK variants 1/2/others versus 26.4% (95% CI 10.5-66.6) in group variants 3a/b (P = 0.034) among crizotinib-treated patients. Meanwhile, the 2-year PFS rate was 69.0% (95% CI 49.9-95.4) in group variants 1/2/others versus 32.7% (95% CI 15.6-68.4) in group variants 3a/b (P = 0.108) among all crizotinib-, alectinib-, and ceritinib-treated patients. Variant 3a- or 5a-harboring cells were resistant to ALK inhibitors with >10-fold higher half maximal inhibitory concentration in vitro. Conclusion Our findings show that group EML4-ALK variants 3a/b may be a major source of ALK inhibitor resistance in the clinic. The variant-specific genotype of the EML4-ALK fusion allows for more precise stratification of patients with advanced NSCLC.
Collapse
Affiliation(s)
- C G Woo
- Department of Pathology, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon, Gyeonggi-do, South Korea
| | - S Seo
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - S W Kim
- Department of Surgery, Seoul National University College of Medicine, Daehak-ro 101, Jongno-gu, Seoul, South Korea
| | - S J Jang
- Molecular Imaging Research Center, Korea Institute of Radiological and Medical Sciences, Seoul 01812, South Korea
| | - K S Park
- Department of Biomedical Science, College of Life Science, CHA University, Seoul, South Korea
| | - J Y Song
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - B Lee
- Division of Surgical Oncology, Department of Surgery, City of Hope, Duarte, California, USA
| | - M W Richards
- Astbury Centre for Structural Molecular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - R Bayliss
- Astbury Centre for Structural Molecular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - D H Lee
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul 152-703, South Korea
| | - J Choi
- epartment of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| |
Collapse
|
590
|
Pailler E, Oulhen M, Borget I, Remon J, Ross K, Auger N, Billiot F, Ngo Camus M, Commo F, Lindsay CR, Planchard D, Soria JC, Besse B, Farace F. Circulating Tumor Cells with Aberrant ALK Copy Number Predict Progression-Free Survival during Crizotinib Treatment in ALK-Rearranged Non-Small Cell Lung Cancer Patients. Cancer Res 2017; 77:2222-2230. [PMID: 28461563 DOI: 10.1158/0008-5472.can-16-3072] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 01/04/2017] [Accepted: 02/03/2017] [Indexed: 11/16/2022]
Abstract
The duration and magnitude of clinical response are unpredictable in ALK-rearranged non-small cell lung cancer (NSCLC) patients treated with crizotinib, although all patients invariably develop resistance. Here, we evaluated whether circulating tumor cells (CTC) with aberrant ALK-FISH patterns [ALK-rearrangement, ALK-copy number gain (ALK-CNG)] monitored on crizotinib could predict progression-free survival (PFS) in a cohort of ALK-rearranged patients. Thirty-nine ALK-rearranged NSCLC patients treated with crizotinib as first ALK inhibitor were recruited prospectively. Blood samples were collected at baseline and at an early time-point (2 months) on crizotinib. Aberrant ALK-FISH patterns were examined in CTCs using immunofluorescence staining combined with filter-adapted FISH after filtration enrichment. CTCs were classified into distinct subsets according to the presence of ALK-rearrangement and/or ALK-CNG signals. No significant association between baseline numbers of ALK-rearranged or ALK-CNG CTCs and PFS was observed. However, we observed a significant association between the decrease in CTC number with ALK-CNG on crizotinib and a longer PFS (likelihood ratio test, P = 0.025). In multivariate analysis, the dynamic change of CTC with ALK-CNG was the strongest factor associated with PFS (HR, 4.485; 95% confidence interval, 1.543-13.030, P = 0.006). Although not dominant, ALK-CNG has been reported to be one of the mechanisms of acquired resistance to crizotinib in tumor biopsies. Our results suggest that the dynamic change in the numbers of CTCs with ALK-CNG may be a predictive biomarker for crizotinib efficacy in ALK-rearranged NSCLC patients. Serial molecular analysis of CTC shows promise for real-time patient monitoring and clinical outcome prediction in this population. Cancer Res; 77(9); 2222-30. ©2017 AACR.
Collapse
Affiliation(s)
- Emma Pailler
- Gustave Roussy, Université Paris-Saclay, "Circulating Tumor 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-Bicetre, France
| | - Marianne Oulhen
- Gustave Roussy, Université Paris-Saclay, "Circulating Tumor Cells" Translational Platform, CNRS UMS3655 - INSERM US23 AMMICA, Villejuif, France.,INSERM, U981 "Identification of Molecular Predictors and new Targets for Cancer Treatment," Villejuif, France
| | - Isabelle Borget
- Gustave Roussy, Université Paris-Saclay, Department of Biostatistics and Epidemiology, Villejuif, France.,Univ Paris-Sud, Université Paris-Saclay, Faculty of Pharmacy, Châtenay-Malabry, France
| | - Jordi Remon
- Gustave Roussy, Université Paris-Saclay, Department of Medicine, Villejuif, France
| | - Kirsty Ross
- Gustave Roussy, Université Paris-Saclay, "Circulating Tumor Cells" Translational Platform, CNRS UMS3655 - INSERM US23 AMMICA, Villejuif, France.,INSERM, U981 "Identification of Molecular Predictors and new Targets for Cancer Treatment," Villejuif, France
| | - Nathalie Auger
- Gustave Roussy, Université Paris-Saclay, Department of Biopathology, Villejuif, France
| | - Fanny Billiot
- Gustave Roussy, Université Paris-Saclay, "Circulating Tumor 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 Ngo Camus
- Gustave Roussy, Université Paris-Saclay, Department of Medicine, Villejuif, France
| | - Frédéric Commo
- INSERM, U981 "Identification of Molecular Predictors and new Targets for Cancer Treatment," Villejuif, France
| | - Colin R Lindsay
- Gustave Roussy, Université Paris-Saclay, "Circulating Tumor Cells" Translational Platform, CNRS UMS3655 - INSERM US23 AMMICA, Villejuif, France.,Gustave Roussy, Université Paris-Saclay, Department of Medicine, Villejuif, France
| | - David Planchard
- Gustave Roussy, Université Paris-Saclay, Department of Medicine, Villejuif, France
| | - Jean-Charles Soria
- 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-Bicetre, France.,Gustave Roussy, Université Paris-Saclay, Department of Medicine, Villejuif, France
| | - Benjamin Besse
- Gustave Roussy, Université Paris-Saclay, Department of Medicine, Villejuif, France
| | - Françoise Farace
- Gustave Roussy, Université Paris-Saclay, "Circulating Tumor 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-Bicetre, France
| |
Collapse
|
591
|
Vachani A, Sequist LV, Spira A. AJRCCM: 100-Year Anniversary. The Shifting Landscape for Lung Cancer: Past, Present, and Future. Am J Respir Crit Care Med 2017; 195:1150-1160. [PMID: 28459327 PMCID: PMC5439022 DOI: 10.1164/rccm.201702-0433ci] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 03/16/2017] [Indexed: 12/13/2022] Open
Abstract
The past century has witnessed a transformative shift in lung cancer from a rare reportable disease to the leading cause of cancer death among men and women worldwide. This historic shift reflects the increase in tobacco consumption worldwide, spurring public health efforts over the past several decades directed at tobacco cessation and control. Although most lung cancers are still diagnosed at a late stage, there have been significant advances in screening high-risk smokers, diagnostic modalities, and chemopreventive approaches. Improvements in surgery and radiation are advancing our ability to manage early-stage disease, particularly among patients considered unfit for traditional open resection. Arguably, the most dramatic progress has occurred on the therapeutic side, with the development of targeted and immune-based therapy over the past decade. This article reviews the major shifts in the lung cancer landscape over the past 100 years. Although many ongoing clinical challenges remain, this review will also highlight emerging molecular and imaging-based approaches that represent opportunities to transform the prevention, early detection, and treatment of lung cancer in the years ahead.
Collapse
Affiliation(s)
- Anil Vachani
- Division of Pulmonary, Allergy, and Critical Care Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Lecia V. Sequist
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts; and
| | - Avrum Spira
- Section of Computational Biomedicine, Department of Medicine, Boston University Medical Center, Boston, Massachusetts
| |
Collapse
|
592
|
Gotwals P, Cameron S, Cipolletta D, Cremasco V, Crystal A, Hewes B, Mueller B, Quaratino S, Sabatos-Peyton C, Petruzzelli L, Engelman JA, Dranoff G. Prospects for combining targeted and conventional cancer therapy with immunotherapy. Nat Rev Cancer 2017; 17:286-301. [PMID: 28338065 DOI: 10.1038/nrc.2017.17] [Citation(s) in RCA: 718] [Impact Index Per Article: 89.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Over the past 25 years, research in cancer therapeutics has largely focused on two distinct lines of enquiry. In one approach, efforts to understand the underlying cell-autonomous, genetic drivers of tumorigenesis have led to the development of clinically important targeted agents that result in profound, but often not durable, tumour responses in genetically defined patient populations. In the second parallel approach, exploration of the mechanisms of protective tumour immunity has provided several therapeutic strategies - most notably the 'immune checkpoint' antibodies that reverse the negative regulators of T cell function - that accomplish durable clinical responses in subsets of patients with various tumour types. The integration of these potentially complementary research fields provides new opportunities to improve cancer treatments. Targeted and immune-based therapies have already transformed the standard-of-care for several malignancies. However, additional insights into the effects of targeted therapies, along with conventional chemotherapy and radiation therapy, on the induction of antitumour immunity will help to advance the design of combination strategies that increase the rate of complete and durable clinical response in patients.
Collapse
Affiliation(s)
- Philip Gotwals
- Exploratory Immuno-Oncology, Novartis Institutes for BioMedical Research
| | - Scott Cameron
- Translational Clinical Oncology, Novartis Institutes for BioMedical Research
| | - Daniela Cipolletta
- Exploratory Immuno-Oncology, Novartis Institutes for BioMedical Research
| | - Viviana Cremasco
- Exploratory Immuno-Oncology, Novartis Institutes for BioMedical Research
| | - Adam Crystal
- Translational Clinical Oncology, Novartis Institutes for BioMedical Research
| | - Becker Hewes
- Translational Clinical Oncology, Novartis Institutes for BioMedical Research
| | - Britta Mueller
- Exploratory Immuno-Oncology, Novartis Institutes for BioMedical Research
| | - Sonia Quaratino
- Translational Clinical Oncology, Novartis Institutes for BioMedical Research
| | | | - Lilli Petruzzelli
- Translational Clinical Oncology, Novartis Institutes for BioMedical Research
| | - Jeffrey A Engelman
- Oncology, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, Massachusetts 02139, USA
| | - Glenn Dranoff
- Exploratory Immuno-Oncology, Novartis Institutes for BioMedical Research
| |
Collapse
|
593
|
Chen G, Chen X, Zhang Y, Yan F, Fang W, Yang Y, Hong S, Miao S, Wu M, Huang X, Luo Y, Zhou C, Gong R, Huang Y, Zhou N, Zhao H, Zhang L. A large, single-center, real-world study of clinicopathological characteristics and treatment in advanced ALK-positive non-small-cell lung cancer. Cancer Med 2017; 6:953-961. [PMID: 28374971 PMCID: PMC5430086 DOI: 10.1002/cam4.1059] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 02/19/2017] [Accepted: 02/22/2017] [Indexed: 11/10/2022] Open
Abstract
Crizotinib has achieved astonishing success in advanced non-small-cell lung cancer (NSCLC) patients harboring anaplastic lymphoma kinase (ALK) rearrangement. However, no real-world studies described the clinicopathological characteristics and treatment of such patients in China. Patients were consecutively collected from Sun Yat-sen University Cancer Center. Chi-square test was applied to explore the relationship between ALK fusion status and metastasis sites. Kaplan-Meier methods and multivariable analyses were used to estimate progression-free survival (PFS). A total of 291 advanced NSCLC patients (ALK (+), N = 97; both ALK & epidermal growth factor receptor (EGFR) (-), N = 194) were enrolled. The occurrence of brain metastasis in ALK-positive patients was significantly higher than double-negative ones both at baseline (26.5% vs. 16.5%, P = 0.038) and during treatment (25.8% vs. 11.9%, P = 0.003), but opposite for pleural effusion (6.2% vs. 26.9%, P < 0.001 at baseline; 3.1% vs. 10.3%, P = 0.031 during treatment). ALK-positive patients of 53.6% used crizotinib, whereas others only received chemotherapy (37.1%) or supportive care (9.3%). Usage of crizotinib prolonged PFS compared with chemotherapy in ALK-positive patients (median PFS 17.6 m vs. 4.8 m, P < 0.001). ALK-positive NSCLC had more brain metastasis and less pleural effusion than double-negative ones. Crizotinib showed better PFS than chemotherapy in advanced ALK-positive NSCLC at any line. However, half advanced ALK-positive patients never received crizotinib, which was grim and need improving.
Collapse
|
594
|
Lim C, Sekhon HS, Cutz JC, Hwang DM, Kamel-Reid S, Carter RF, Santos GDC, Waddell T, Binnie M, Patel M, Paul N, Chung T, Brade A, El-Maraghi R, Sit C, Tsao MS, Leighl NB. Improving molecular testing and personalized medicine in non-small-cell lung cancer in Ontario. ACTA ACUST UNITED AC 2017; 24:103-110. [PMID: 28490924 DOI: 10.3747/co.24.3495] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Although molecular testing has become standard in managing advanced nonsquamous non-small-cell lung cancer (nsclc), most patients undergo minimally invasive procedures, and the diagnostic tumour specimens available for testing are usually limited. A knowledge translation initiative to educate diagnostic specialists about sampling techniques and laboratory processes was undertaken to improve the uptake and application of molecular testing in advanced lung cancer. METHODS A multidisciplinary panel of physician experts including pathologists, respirologists, interventional thoracic radiologists, thoracic surgeons, medical oncologists, and radiation oncologists developed a specialty-specific education program, adapting international clinical guidelines to the local Ontario context. Expert recommendations from the program are reported here. RESULTS Panel experts agreed that specialists procuring samples for lung cancer diagnosis should choose biopsy techniques that maximize tumour cellularity, and that conservation strategies to maximize tissue for molecular testing should be used in tissue processing. The timeliness of molecular reporting can be improved by pathologist-initiated reflex testing upon confirmation of nonsquamous nsclc and by prompt transportation of specimens to designated molecular diagnostic centres. To coordinate timely molecular testing and optimal treatment, collaboration and communication between all clinicians involved in diagnosing patients with advanced lung cancer are mandatory. CONCLUSIONS Knowledge transfer to diagnostic lung cancer specialists could potentially improve molecular testing and treatment for advanced lung cancer patients.
Collapse
Affiliation(s)
- C Lim
- Division of Medical Oncology, Princess Margaret Cancer Centre, University of Toronto, Toronto
| | - H S Sekhon
- Department of Pathology and Laboratory Medicine, University of Ottawa, Ottawa
| | - J C Cutz
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton
| | - D M Hwang
- Laboratory Medicine Program, University Health Network, University of Toronto, Toronto
| | - S Kamel-Reid
- Laboratory Medicine Program, University Health Network, University of Toronto, Toronto.,Molecular Diagnostics Laboratory, University Health Network, Toronto
| | - R F Carter
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton.,LifeLabs Genetics, Toronto
| | - G da Cunha Santos
- Laboratory Medicine Program, University Health Network, University of Toronto, Toronto
| | - T Waddell
- Division of Thoracic Surgery, University of Toronto, Toronto
| | - M Binnie
- Division of Respirology, University of Toronto, Toronto
| | - M Patel
- Division of Respirology, Trillium Health Partners, Mississauga
| | - N Paul
- Joint Department of Medical Imaging, University Health Network, Mount Sinai Hospital and Women's College Hospital, Toronto
| | - T Chung
- Joint Department of Medical Imaging, University Health Network, Mount Sinai Hospital and Women's College Hospital, Toronto
| | - A Brade
- Department of Radiation Oncology, University of Toronto, Toronto
| | - R El-Maraghi
- Simcoe Muskoka Regional Cancer Centre, Barrie; and
| | - C Sit
- Lung Cancer Canada, Toronto, ON
| | - M S Tsao
- Laboratory Medicine Program, University Health Network, University of Toronto, Toronto
| | - N B Leighl
- Division of Medical Oncology, Princess Margaret Cancer Centre, University of Toronto, Toronto
| |
Collapse
|
595
|
Wang HQ, Halilovic E, Li X, Liang J, Cao Y, Rakiec DP, Ruddy DA, Jeay S, Wuerthner JU, Timple N, Kasibhatla S, Li N, Williams JA, Sellers WR, Huang A, Li F. Combined ALK and MDM2 inhibition increases antitumor activity and overcomes resistance in human ALK mutant neuroblastoma cell lines and xenograft models. eLife 2017; 6. [PMID: 28425916 PMCID: PMC5435462 DOI: 10.7554/elife.17137] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Accepted: 04/18/2017] [Indexed: 12/02/2022] Open
Abstract
The efficacy of ALK inhibitors in patients with ALK-mutant neuroblastoma is limited, highlighting the need to improve their effectiveness in these patients. To this end, we sought to develop a combination strategy to enhance the antitumor activity of ALK inhibitor monotherapy in human neuroblastoma cell lines and xenograft models expressing activated ALK. Herein, we report that combined inhibition of ALK and MDM2 induced a complementary set of anti-proliferative and pro-apoptotic proteins. Consequently, this combination treatment synergistically inhibited proliferation of TP53 wild-type neuroblastoma cells harboring ALK amplification or mutations in vitro, and resulted in complete and durable responses in neuroblastoma xenografts derived from these cells. We further demonstrate that concurrent inhibition of MDM2 and ALK was able to overcome ceritinib resistance conferred by MYCN upregulation in vitro and in vivo. Together, combined inhibition of ALK and MDM2 may provide an effective treatment for TP53 wild-type neuroblastoma with ALK aberrations. DOI:http://dx.doi.org/10.7554/eLife.17137.001
Collapse
Affiliation(s)
- Hui Qin Wang
- Disease Area Oncology, Novartis Institutes for BioMedical Research, Cambridge, United States
| | - Ensar Halilovic
- Disease Area Oncology, Novartis Institutes for BioMedical Research, Cambridge, United States
| | - Xiaoyan Li
- Disease Area Oncology, Novartis Institutes for BioMedical Research, Cambridge, United States
| | - Jinsheng Liang
- Disease Area Oncology, Novartis Institutes for BioMedical Research, Cambridge, United States
| | - Yichen Cao
- Disease Area Oncology, Novartis Institutes for BioMedical Research, Cambridge, United States
| | - Daniel P Rakiec
- Disease Area Oncology, Novartis Institutes for BioMedical Research, Cambridge, United States
| | - David A Ruddy
- Disease Area Oncology, Novartis Institutes for BioMedical Research, Cambridge, United States
| | - Sebastien Jeay
- Disease Area Oncology, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Jens U Wuerthner
- Disease Area Oncology, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Noelito Timple
- Genomics Institute of the Novartis Research Foundation, San Diego, United States
| | - Shailaja Kasibhatla
- Genomics Institute of the Novartis Research Foundation, San Diego, United States
| | - Nanxin Li
- Genomics Institute of the Novartis Research Foundation, San Diego, United States
| | - Juliet A Williams
- Disease Area Oncology, Novartis Institutes for BioMedical Research, Cambridge, United States
| | - William R Sellers
- Disease Area Oncology, Novartis Institutes for BioMedical Research, Cambridge, United States
| | - Alan Huang
- Disease Area Oncology, Novartis Institutes for BioMedical Research, Cambridge, United States
| | - Fang Li
- Disease Area Oncology, Novartis Institutes for BioMedical Research, Cambridge, United States
| |
Collapse
|
596
|
Lau YY, Gu W, Lin T, Viraswami-Appanna K, Cai C, Scott JW, Shi M. Assessment of drug-drug interaction potential between ceritinib and proton pump inhibitors in healthy subjects and in patients with ALK-positive non-small cell lung cancer. Cancer Chemother Pharmacol 2017; 79:1119-1128. [PMID: 28424965 DOI: 10.1007/s00280-017-3308-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 04/11/2017] [Indexed: 12/31/2022]
Abstract
PURPOSE The impact of proton pump inhibitors (PPIs) on the pharmacokinetics (PK) and efficacy of ceritinib was evaluated. METHODS A healthy subject drug-drug interaction (DDI) study was conducted to assess the effect of esomeprazole on the PK of a single 750 mg dose of ceritinib. To further investigate the impact of PPIs on the PK and efficacy of ceritinib in ALK-positive cancer patients, two subgroup analyses were performed. Analysis 1 evaluated ceritinib steady-state trough concentration (Ctrough,ss) and overall response rate (ORR) by concomitant use of PPIs in patients from the ASCEND-1, -2, and -3 studies; analysis 2 evaluated ceritinib single-dose and steady-state AUC0-24h and C max by concomitant PPI use in patients from ASCEND-1 using a definition of PPI usage similar to that used in the healthy subject study. RESULTS In the healthy subject study, co-administration of a single 750 mg dose of ceritinib with esomeprazole 40 mg for 6 days decreased ceritinib AUC0-∞ by 76% and C max by 79%. However, based on subgroup analysis 1, patients had similar C trough,ss and ORR regardless of concomitant PPI usage. Based on analysis 2, co-administration of a single 750 mg ceritinib dose with PPIs for 6 days in patients suggested less effect on ceritinib exposure than that observed in healthy subjects as AUC0-24h decreased by 30% and C max decreased by 25%. No clinically meaningful effect on steady-state exposure was observed after daily dosing. CONCLUSIONS Long-term administration of ceritinib with PPIs does not adversely affect the PK and efficacy of ceritinib in ALK-positive cancer patients.
Collapse
Affiliation(s)
- Yvonne Y Lau
- Novartis Pharmaceuticals Corporation, One Health Plaza, East Hanover, NJ, 07936-1080, USA.
| | - Wen Gu
- Novartis Pharmaceuticals Corporation, One Health Plaza, East Hanover, NJ, 07936-1080, USA
| | - Tiffany Lin
- Novartis Pharmaceuticals Corporation, One Health Plaza, East Hanover, NJ, 07936-1080, USA
| | | | - Can Cai
- Novartis Pharmaceuticals Corporation, One Health Plaza, East Hanover, NJ, 07936-1080, USA
| | - Jeffrey W Scott
- Novartis Pharmaceuticals Corporation, One Health Plaza, East Hanover, NJ, 07936-1080, USA
| | - Michael Shi
- Novartis Pharmaceuticals Corporation, One Health Plaza, East Hanover, NJ, 07936-1080, USA
| |
Collapse
|
597
|
Lim SH, Yoh KA, Lee JS, Ahn MJ, Kim YJ, Kim SH, Zhang J, Patel D, Swallow E, Kageleiry A, Galebach P, Lee D, Stein K, Degun R, Park K. Characteristics and outcomes of ALK
+ non-small cell lung cancer patients in Korea. Asia Pac J Clin Oncol 2017; 13:e239-e245. [DOI: 10.1111/ajco.12645] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 09/06/2016] [Accepted: 10/03/2016] [Indexed: 11/30/2022]
Affiliation(s)
- Sung Hee Lim
- Division of Hematology-Oncology; Department of Medicine; Samsung Medical Center; Sungkyunkwan University School of Medicine; Seoul Republic of Korea
| | - Kyung Ah Yoh
- Seoul National University Bundang Hospital; Seongnam Republic of Korea
| | - Jong Seok Lee
- Seoul National University Bundang Hospital; Seongnam Republic of Korea
| | - Myung-ju Ahn
- Division of Hematology-Oncology; Department of Medicine; Samsung Medical Center; Sungkyunkwan University School of Medicine; Seoul Republic of Korea
| | - Yu Jung Kim
- Seoul National University Bundang Hospital; Seongnam Republic of Korea
| | - Se Hyun Kim
- Seoul National University Bundang Hospital; Seongnam Republic of Korea
| | - Jie Zhang
- Novartis Pharmaceuticals Corporation; East Hanover; New Jersey USA
| | | | | | | | | | | | - Karen Stein
- Novartis Pharmaceuticals Corporation; East Hanover; New Jersey USA
| | | | - Keunchil Park
- Division of Hematology-Oncology; Department of Medicine; Samsung Medical Center; Sungkyunkwan University School of Medicine; Seoul Republic of Korea
- Innovative Cancer Medicine Institute; Seoul Republic of Korea
| |
Collapse
|
598
|
Zheng J, Zhou J, Zhu Y, Shen Q, Zhou J. Significant Radiologic Response of Pancreatic Metastasis After Targeted Therapy of Ceritinib (LDK378) for ALK-Rearranged Lung Adenocarcinoma Presenting With Hyperglycemia. Oncol Res 2017; 25:545-550. [PMID: 28438234 PMCID: PMC7841023 DOI: 10.3727/096504016x14801968368898] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Pancreatic metastasis from non-small cell lung cancer (NSCLC) is usually asymptomatic or presents with abdominal pain, acute pancreatitis, or jaundice. A lung primary is associated with worse survival compared to pancreatic metastases from other organs. Surgical treatment of solitary metastasis to the pancreas from NSCLC has been reviewed in several studies, one of which had a notable disease-free interval. To our knowledge, there are no prior reports of targeted therapy of pancreatic metastasis of NSCLC followed by a significant response. Herein we report the case of a 31-year-old female with a solitary pancreatic metastasis from ALK-rearranged lung adenocarcinoma despite treatment with chemotherapy and crizotinib; she presented with symptoms of hyperglycemia. Targeted therapy with ceritinib (LDK378) led to symptomatic improvement and a significant radiologic response in the lung and pancreas, but not in the brain.
Collapse
Affiliation(s)
- Jing Zheng
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, P.R. China
| | - Jianya Zhou
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, P.R. China
| | - Yanping Zhu
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, P.R. China
| | - Qian Shen
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, P.R. China
| | - Jianying Zhou
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, P.R. China
| |
Collapse
|
599
|
Aggarwal C, Borghaei H. Treatment Paradigms for Advanced Non-Small Cell Lung Cancer at Academic Medical Centers: Involvement in Clinical Trial Endpoint Design. Oncologist 2017; 22:700-708. [PMID: 28408617 PMCID: PMC5469580 DOI: 10.1634/theoncologist.2016-0345] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 01/03/2017] [Indexed: 12/25/2022] Open
Abstract
Selection of appropriate clinical endpoints for examining the efficacy of investigational agents for non‐small cell lung cancer is of vital importance in clinical trial design. This review provides an overview of the study designs of clinical trials for approved agents in non‐small cell lung cancer and focuses on the validity of alternative endpoints for such trials. Based on the positive results of various clinical trials, treatment options for non‐small cell lung cancer (NSCLC) have expanded greatly over the last 25 years. While regulatory approvals of chemotherapeutic agents for NSCLC have largely been based on improvements in overall survival, recent approvals of many targeted agents for NSCLC (afatinib, crizotinib, ceritinib, osimertinib) have been based on surrogate endpoints such as progression‐free survival and objective response. As such, selection of appropriate clinical endpoints for examining the efficacy of investigational agents for NSCLC is of vital importance in clinical trial design. This review provides an overview of clinical trial endpoints previously utilized for approved agents for NSCLC and highlights the key efficacy results for these trials. Trends for more recent approvals in NSCLC, including those for the immunotherapeutic agents nivolumab and pembrolizumab, are also discussed. The results of a correlative analysis of endpoints from 18 clinical trials that supported approvals of investigational agents in clinical trials for NSCLC are also presented. Implications for Practice. While improving survival remains the ultimate goal of oncology clinical trials, overall survival may not always be the most feasible or appropriate endpoint to assess patient response. Recently, several investigational agents, both targeted agents and immunotherapies, have gained U.S. Food and Drug Administration approval in non‐small cell lung cancer based on alternate endpoints such as progression‐free survival or response rate. An understanding of the assessment of response and trial endpoint choice is important for future oncology clinical trial design.
Collapse
Affiliation(s)
- Charu Aggarwal
- Division of Hematology/Oncology, Abramson Cancer Center, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Hossein Borghaei
- Department of Medical Oncology, Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA
| |
Collapse
|
600
|
Sundar R, Chénard-Poirier M, Collins DC, Yap TA. Imprecision in the Era of Precision Medicine in Non-Small Cell Lung Cancer. Front Med (Lausanne) 2017; 4:39. [PMID: 28443282 PMCID: PMC5385461 DOI: 10.3389/fmed.2017.00039] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 03/22/2017] [Indexed: 12/29/2022] Open
Abstract
Over the past decade, major advances have been made in the management of advanced non-small cell lung cancer (NSCLC). There has been a particular focus on the identification and targeting of putative driver aberrations, which has propelled NSCLC to the forefront of precision medicine. Several novel molecularly targeted agents have now achieved regulatory approval, while many others are currently in late-phase clinical trial testing. These antitumor therapies have significantly impacted the clinical outcomes of advanced NSCLC and provided patients with much hope for the future. Despite this, multiple deficiencies still exist in our knowledge of this complex disease, and further research is urgently required to overcome these critical issues. This review traces the path undertaken by the different therapeutics assessed in NSCLC and the impact of precision medicine in this disease. We also discuss the areas of "imprecision" that still exist in NSCLC and the modern hypothesis-testing studies being conducted to address these key challenges.
Collapse
Affiliation(s)
- Raghav Sundar
- Royal Marsden Hospital, London, UK
- Department of Haematology-Oncology, National University Cancer Institute of Singapore, Singapore, Singapore
| | | | | | - Timothy A. Yap
- Royal Marsden Hospital, London, UK
- The Institute of Cancer Research, London, UK
| |
Collapse
|