201
|
Frega S, Bonanno L, Guarneri V, Conte P, Pasello G. Therapeutic perspectives for brain metastases in non-oncogene addicted non-small cell lung cancer (NSCLC): Towards a less dismal future? Crit Rev Oncol Hematol 2018; 128:19-29. [DOI: 10.1016/j.critrevonc.2018.05.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 05/14/2018] [Indexed: 12/16/2022] Open
|
202
|
Efficacy of ALK inhibitors on NSCLC brain metastases: A systematic review and pooled analysis of 21 studies. PLoS One 2018; 13:e0201425. [PMID: 30052658 PMCID: PMC6063430 DOI: 10.1371/journal.pone.0201425] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Accepted: 07/16/2018] [Indexed: 12/21/2022] Open
Abstract
Background Patients with anaplastic lymphoma kinase rearranged (ALK+) non-small cell lung cancer (NSCLC) have a higher risk of developing brain metastases (BMs) than patients with other NSCLC sub-types. ALK inhibitors have activity in BMs due to ALK+ NSCLC. We performed a systematic review of the literature with the aim of assessing the efficacy of ALK inhibitors on BMs. Material and methods A systematic search of the literature was performed using the databases Pubmed, EMBASE, Web of Science, The Cochrane Library, and SCOPUS. Relevant publications reporting activity of ALK inhibitors in NSCLC BMs were retrieved. Data were pooled using the number of events/number of evaluable patients according to fixed or random effect models. Intracranial tumour response was assessed through overall response rate (ORR), disease control rate (DCR: ORR + stable disease rate), median progression-free survival (PFS), and overall survival (OS). The primary endpoint was intracranial overall response rate (IC ORR). Results A total of 1,016 patients with BMs from 21 studies were analysed. In patients receiving ALK inhibitors in the first line setting, the pooled IC ORR was 39.17% (95%CI 13.1–65.2%), while the pooled IC ORR observed in further lines was 44.2% (95%CI 33.3–55.1%). Intracranial disease control rate (IC DCR) was 70.3% and 78.2% in naïve and pre-treated patients, respectively. Patients who had not received brain radiation attained an IC ORR of 49.0%. Conclusions Based on these data, ALK inhibitors are effective in both naive and pre-treated patients with similar IC ORR and IC DCR, irrespective of the line of therapy.
Collapse
|
203
|
Kim M, Baek M, Kim DJ. Protein Tyrosine Signaling and its Potential Therapeutic Implications in Carcinogenesis. Curr Pharm Des 2018. [PMID: 28625132 DOI: 10.2174/1381612823666170616082125] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Protein tyrosine phosphorylation is a crucial signaling mechanism that plays a role in epithelial carcinogenesis. Protein tyrosine kinases (PTKs) control various cellular processes including growth, differentiation, metabolism, and motility by activating major signaling pathways including STAT3, AKT, and MAPK. Genetic mutation of PTKs and/or prolonged activation of PTKs and their downstream pathways can lead to the development of epithelial cancer. Therefore, PTKs became an attractive target for cancer prevention. PTK inhibitors are continuously being developed, and they are currently used for the treatment of cancers that show a high expression of PTKs. Protein tyrosine phosphatases (PTPs), the homeostatic counterpart of PTKs, negatively regulate the rate and duration of phosphotyrosine signaling. PTPs initially were considered to be only housekeeping enzymes with low specificity. However, recent studies have demonstrated that PTPs can function as either tumor suppressors or tumor promoters, depending on their target substrates. Together, both PTK and PTP signal transduction pathways are potential therapeutic targets for cancer prevention and treatment.
Collapse
Affiliation(s)
- Mihwa Kim
- Department of Biomedical Sciences, School of Medicine, University of Texas Rio Grande Valley, Edinburg, TX, USA
| | - Minwoo Baek
- Department of Biomedical Sciences, School of Medicine, University of Texas Rio Grande Valley, Edinburg, TX, USA
| | - Dae Joon Kim
- Department of Biomedical Sciences, School of Medicine, University of Texas Rio Grande Valley, Edinburg, TX, USA
| |
Collapse
|
204
|
Structure and energy based quantitative missense variant effect analysis provides insights into drug resistance mechanisms of anaplastic lymphoma kinase mutations. Sci Rep 2018; 8:10664. [PMID: 30006516 PMCID: PMC6045602 DOI: 10.1038/s41598-018-28752-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 06/29/2018] [Indexed: 02/05/2023] Open
Abstract
Anaplastic lymphoma kinase (ALK) is considered as a validated molecular target in multiple malignancies, such as non-small cell lung cancer (NSCLC). However, the effectiveness of molecularly targeted therapies using ALK inhibitors is almost universally limited by drug resistance. Drug resistance to molecularly targeted therapies has now become a major obstacle to effective cancer treatment and personalized medicine. It is of particular importance to provide an improved understanding on the mechanisms of resistance of ALK inhibitors, thus rational new therapeutic strategies can be developed to combat resistance. We used state-of-the-art computational approaches to systematically explore the mutational effects of ALK mutations on drug resistance properties. We found the activation of ALK was increased by substitution with destabilizing mutations, creating the capacity to confer drug resistance to inhibitors. In addition, results implied that evolutionary constraints might affect the drug resistance properties. Moreover, an extensive profile of drugs against ALK mutations was constructed to give better understanding of the mechanism of drug resistance based on structural transitions and energetic variation. Our work hopes to provide an up-to-date mechanistic framework for understanding the mechanisms of drug resistance induced by ALK mutations, thus tailor treatment decisions after the emergence of resistance in ALK-dependent diseases.
Collapse
|
205
|
Barriers to Effective Drug Treatment for Brain Metastases: A Multifactorial Problem in the Delivery of Precision Medicine. Pharm Res 2018; 35:177. [PMID: 30003344 DOI: 10.1007/s11095-018-2455-9] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 06/30/2018] [Indexed: 12/12/2022]
Abstract
The treatment of metastatic lesions in the brain represents a serious unmet medical need in the field of neuro-oncology. Even though many effective compounds have demonstrated success in treating peripheral (non-CNS) tumors with targeted agents, one aspect of this lack of success in the brain may be related to poor delivery of otherwise effective compounds. Many factors can influence the brain delivery of these agents, but one key barrier is a heterogeneously "leaky" BBB that expresses efflux transporters that limit the BBB permeability for many targeted agents. Future success in therapeutics for brain metastases must take into account the adequate delivery of "active, free drug" to the target, and may include combinations of targeted drugs that are appropriate to address each individual patient's tumor type. This review discusses some issues that are pertinent to precision medicine for brain metastases, using specific examples of tumor types that have a high incidence of brain metastases.
Collapse
|
206
|
Indian consensus statement for treatment of advanced non small cell lung cancer: First line, maintenance, and second line. Indian J Cancer 2018; 54:89-103. [PMID: 29199671 DOI: 10.4103/ijc.ijc_136_17] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
The management of advanced nonsmall cell lung cancer (NSCLC) patients is becoming complex with the identification of driver mutations and targeted therapies. The expert group of academic medical oncologists used data from published literature, practical experience to arrive at practical consensus recommendations to treat advanced NSCLC for use by the community oncologists.
Collapse
|
207
|
Lin JJ, Zhu VW, Schoenfeld AJ, Yeap BY, Saxena A, Ferris LA, Dagogo-Jack I, Farago AF, Taber A, Traynor A, Menon S, Gainor JF, Lennerz JK, Plodkowski AJ, Digumarthy SR, Ou SHI, Shaw AT, Riely GJ. Brigatinib in Patients With Alectinib-Refractory ALK-Positive NSCLC. J Thorac Oncol 2018; 13:1530-1538. [PMID: 29935304 DOI: 10.1016/j.jtho.2018.06.005] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 06/06/2018] [Accepted: 06/11/2018] [Indexed: 12/14/2022]
Abstract
INTRODUCTION The second-generation anaplastic lymphoma kinase (ALK) inhibitor alectinib recently showed superior efficacy compared to the first-generation ALK inhibitor crizotinib in advanced ALK-rearranged NSCLC, establishing alectinib as the new standard first-line therapy. Brigatinib, another second-generation ALK inhibitor, has shown substantial activity in patients with crizotinib-refractory ALK-positive NSCLC; however, its activity in the alectinib-refractory setting is unknown. METHODS A multicenter, retrospective study was performed at three institutions. Patients were eligible if they had advanced, alectinib-refractory ALK-positive NSCLC and were treated with brigatinib. Medical records were reviewed to determine clinical outcomes. RESULTS Twenty-two patients were eligible for this study. Confirmed objective responses to brigatinib were observed in 3 of 18 patients (17%) with measurable disease. Nine patients (50%) had stable disease on brigatinib. The median progression-free survival was 4.4 months (95% confidence interval [CI]: 1.8-5.6 months) with a median duration of treatment of 5.7 months (95% CI: 1.8-6.2 months). Among 9 patients in this study who underwent post-alectinib/pre-brigatinib biopsies, 5 had an ALK I1171X or V1180L resistance mutation; of these, 1 had a confirmed partial response and 3 had stable disease on brigatinib. One patient had an ALK G1202R mutation in a post-alectinib/pre-brigatinib biopsy, and had progressive disease as the best overall response to brigatinib. CONCLUSIONS Brigatinib has limited clinical activity in alectinib-refractory ALK-positive NSCLC. Additional studies are needed to establish biomarkers of response to brigatinib and to identify effective therapeutic options for alectinib-resistant ALK-positive NSCLC patients.
Collapse
Affiliation(s)
- Jessica J Lin
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Viola W Zhu
- Chao Family Comprehensive Cancer Center, University of California Irvine School of Medicine, Orange, California
| | - Adam J Schoenfeld
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Beow Y Yeap
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Ashish Saxena
- Department of Medicine, Weill Cornell Medicine, New York, New York
| | - Lorin A Ferris
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Ibiayi Dagogo-Jack
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Anna F Farago
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Angela Taber
- Lifespan Cancer Institute, Providence, Rhode Island
| | - Anne Traynor
- Department of Medicine, University of Wisconsin Carbone Cancer Center, Madison, Wisconsin
| | - Smitha Menon
- Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Justin F Gainor
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Jochen K Lennerz
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts
| | - Andrew J Plodkowski
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Subba R Digumarthy
- Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts
| | - Sai-Hong Ignatius Ou
- Chao Family Comprehensive Cancer Center, University of California Irvine School of Medicine, Orange, California
| | - Alice T Shaw
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts.
| | - Gregory J Riely
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
208
|
Horn L, Infante JR, Reckamp KL, Blumenschein GR, Leal TA, Waqar SN, Gitlitz BJ, Sanborn RE, Whisenant JG, Du L, Neal JW, Gockerman JP, Dukart G, Harrow K, Liang C, Gibbons JJ, Holzhausen A, Lovly CM, Wakelee HA. Ensartinib (X-396) in ALK-Positive Non-Small Cell Lung Cancer: Results from a First-in-Human Phase I/II, Multicenter Study. Clin Cancer Res 2018; 24:2771-2779. [PMID: 29563138 PMCID: PMC6004248 DOI: 10.1158/1078-0432.ccr-17-2398] [Citation(s) in RCA: 139] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 12/14/2017] [Accepted: 03/13/2018] [Indexed: 11/16/2022]
Abstract
Purpose: Evaluate safety and determine the recommended phase II dose (RP2D) of ensartinib (X-396), a potent anaplastic lymphoma kinase (ALK) tyrosine kinase inhibitor (TKI), and evaluate preliminary pharmacokinetics and antitumor activity in a first-in-human, phase I/II clinical trial primarily in patients with non-small cell lung cancer (NSCLC).Patients and Methods: In dose escalation, ensartinib was administered at doses of 25 to 250 mg once daily in patients with advanced solid tumors; in dose expansion, patients with advanced ALK-positive NSCLC were administered 225 mg once daily. Patients who had received prior ALK TKI(s) and patients with brain metastases were eligible.Results: Thirty-seven patients enrolled in dose escalation, and 60 enrolled in dose expansion. The most common treatment-related toxicities were rash (56%), nausea (36%), pruritus (28%), vomiting (26%), and fatigue (22%); 23% of patients experienced a treatment-related grade 3 to 4 toxicity (primarily rash and pruritus). The maximum tolerated dose was not reached, but the RP2D was chosen as 225 mg based on the frequency of rash observed at 250 mg without improvement in activity. Among the ALK-positive efficacy evaluable patients treated at ≥200 mg, the response rate (RR) was 60%, and median progression-free survival (PFS) was 9.2 months. RR in ALK TKI-naïve patients was 80%, and median PFS was 26.2 months. In patients with prior crizotinib only, the RR was 69% and median PFS was 9.0 months. Responses were also observed in the central nervous system, with an intracranial RR of 64%.Conclusions: Ensartinib was active and generally well tolerated in patients with ALK-positive NSCLC. Clin Cancer Res; 24(12); 2771-9. ©2018 AACR.
Collapse
Affiliation(s)
- Leora Horn
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee. .,Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | | | | | - George R. Blumenschein
- Department of Thoracic/Head and Neck Medical Oncology, University of Texas M.D. Anderson Cancer Center, Houston, TX
| | - Ticiana A. Leal
- Department of Medicine, University of Wisconsin, Madison, WI
| | - Saiama N. Waqar
- Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO
| | - Barbara J. Gitlitz
- Department of Medicine, University of Southern California Keck School of Medicine, Los Angeles, CA
| | - Rachel E. Sanborn
- Providence Thoracic Oncology Program, Earle A. Chiles Research Institute, Providence Cancer Center, Providence, OR
| | - Jennifer G. Whisenant
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN,Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Liping Du
- Department of Biostatistics, Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, TN
| | - Joel W. Neal
- Department of Medicine, Stanford Cancer Institute, Stanford University, Stanford, CA
| | | | - Gary Dukart
- Xcovery Holding Company, Palm Beach Gardens, FL
| | | | - Chris Liang
- Xcovery Holding Company, Palm Beach Gardens, FL
| | | | | | - Christine M. Lovly
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN,Department of Medicine, Vanderbilt University Medical Center, Nashville, TN,Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, TN
| | - Heather A. Wakelee
- Department of Medicine, Stanford Cancer Institute, Stanford University, Stanford, CA
| |
Collapse
|
209
|
O'Kane GM, Leighl NB. Systemic Therapy of Lung Cancer CNS Metastases Using Molecularly Targeted Agents and Immune Checkpoint Inhibitors. CNS Drugs 2018; 32:527-542. [PMID: 29799091 DOI: 10.1007/s40263-018-0526-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Central nervous system (CNS) metastases most commonly arise from lung cancer, with the majority of patients affected during their disease course. The prognosis for patients with untreated brain metastases is poor, with surgical resection and/or radiotherapy as classic therapeutic options. However, the value of systemic therapy in the management of CNS metastases from lung cancer is growing. Novel targeted agents for the treatment of non-small cell lung cancer (NSCLC) have demonstrated activity in treating patients with CNS involvement, and are potential alternatives to radiation and surgery. These agents include anaplastic lymphoma kinase (ALK) inhibitors such as alectinib, crizotinib, ceritinib, lorlatinib, and others; epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors, including the recently developed third-generation inhibitor osimertinib, and even immune checkpoint inhibitors such as nivolumab, pembrolizumab, and atezolizumab. This review summarizes current activity of systemic agents in the management of CNS metastases from NSCLC, as well as potential mechanisms of action of these small and large molecules.
Collapse
Affiliation(s)
- Grainne M O'Kane
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, 7W-389, 700 University Avenue, Toronto, ON, M5G 1Z5, Canada. Grainne.O'
| | - Natasha B Leighl
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, 7W-389, 700 University Avenue, Toronto, ON, M5G 1Z5, Canada
| |
Collapse
|
210
|
Melosky B. Rapidly changing treatment algorithms for metastatic nonsquamous non-small-cell lung cancer. Curr Oncol 2018; 25:S68-S76. [PMID: 29910649 PMCID: PMC6001773 DOI: 10.3747/co.25.3839] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Background The treatment paradigm for metastatic nonsquamous non-small-cell lung cancer (nsclc) continues to change. Algorithms published only 6 months ago are outdated today and are dramatically different from those published a few years ago. New driver mutations continue to be identified, and the development of therapies to inhibit oncogenic addiction is ongoing. Patient survival is improving as treatments become more personalized and effective. Methods This review looks at the outcomes of recent trials and discusses treatment options for patients with metastatic nsclc of nonsquamous histology. Algorithms continue to change quickly, and an attempt is made to keep the paradigm current and applicable into the near future. Results Treatment algorithms for nsclc tumours with EGFR mutations, ALK rearrangements, and ROS1 rearrangements, and for wild-type tumours are presented. A future algorithm based on new immunotherapy data is proposed. Conclusions The treatment algorithm for EGFR mutation is changing with the proven efficacy of osimertinib for the acquired T790M mutation. All patients taking first- or second-generation epidermal growth factor receptor tyrosine kinase inhibitors must be tested. The treatment algorithm for ALK rearrangement has changed with the proven superiority of alectinib compared with crizotinib in the first-line setting. The approval of crizotinib for ROS1 rearrangements now means that patients also must be tested for that mutation. The biomarker for checkpoint inhibitors continues to be PD-L1 by immunohistochemistry stain, but whether testing will be necessary for patient selection if chemotherapy combinations are implemented will be determined soon.
Collapse
Affiliation(s)
- B. Melosky
- Medical Oncology, BC Cancer–Vancouver Centre, Vancouver, BC
| |
Collapse
|
211
|
Abdallah SM, Wong A. Brain metastases in non-small-cell lung cancer: are tyrosine kinase inhibitors and checkpoint inhibitors now viable options? Curr Oncol 2018; 25:S103-S114. [PMID: 29910653 PMCID: PMC6001769 DOI: 10.3747/co.25.3733] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Significant progress has been made in the treatment of stage iv non-small-cell lung cancer (nsclc); however, the prognosis of patients with brain metastases remains poor. Resection and radiation therapy remain standard options. This issue is an important one because 10% of patients with nsclc have brain metastases at diagnosis, and 25%-40% develop brain metastases during their disease. Standard chemotherapy does not cross the blood-brain barrier. However, there is new hope that tyrosine kinase inhibitors (tkis) used in patients with identified targetable mutations such as mutations of EGFR and rearrangements of ALK could have activity in the central nervous system (cns). Furthermore, immunotherapy is increasingly becoming a standard option for patients with nsclc, and interest about the intracranial activity of those agents is growing. This review presents current data about the cns activity of the available major tkis and immunotherapy agents.
Collapse
Affiliation(s)
| | - A. Wong
- Medical Oncology, McGill University Health Centre, Montreal, and
- Medical Oncology, Hôpital du Suroît, Valleyfield, QC
| |
Collapse
|
212
|
Rybarczyk-Kasiuchnicz A, Ramlau R. Current views on molecularly targeted therapy for lung cancer - a review of literature from the last five years. KARDIOCHIRURGIA I TORAKOCHIRURGIA POLSKA = POLISH JOURNAL OF CARDIO-THORACIC SURGERY 2018; 15:119-124. [PMID: 30069193 PMCID: PMC6066673 DOI: 10.5114/kitp.2018.76478] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 02/25/2018] [Indexed: 12/26/2022]
Abstract
Lung cancer is the main cause of cancer-related deaths in Poland. Epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs) are a new group of agents for non-small-cell lung cancer (NSCLC) patients. Determining the predictive value of activating mutations in the EGFR and ROS-1 genes and ALK rearrangement in non-small-cell lung cancer has enabled the identification of patients likely to achieve true clinical benefits. EGFR-TKIs may produce objective response in more than 60% of patients and prolong progression-free survival to 10 months in mutation-positive patients. No improvement of overall survival was shown in randomized trials. The era of immunotherapy implementing PD-1 and PD-L1 inhibitors has changed the face of lung cancer therapy. We aimed to review the literature on the use of EGFR-TKIs and immunotherapeutic agents for NSCLC patients.
Collapse
Affiliation(s)
| | - Rodryg Ramlau
- Department of Oncology and Pulmonology, Poznan University of Medical Sciences, Poznan, Poland
| |
Collapse
|
213
|
Abstract
The treatment of advanced non-small-cell lung cancer (nsclc) has undergone a paradigm shift since the early 2000s. The identification of molecular subtypes of the disease, based on oncogenic drivers, has led to the development of personalized medicine and the ability to deliver molecularly targeted therapies to patients. In the 10 years that have elapsed since the discovery of the ALK gene in a patient with nsclc, several active drugs have moved rapidly from bench to bedside, and multiple others are currently in clinical trials. Those developments have led to important improvements in patient outcomes, while simultaneously raising key questions about the optimal treatment for ALK-positive nsclc. The inevitable emergence of resistance to alk-directed therapy is central to ongoing research and daily clinical practice for affected patients. In the present review, we highlight the current treatment landscape, the available and emerging clinical trials, and the evolving clinical decision-making in ALK-positive nsclc, with a focus on Canadian practice.
Collapse
Affiliation(s)
- J.M. Rothenstein
- RS McLaughlin Durham Regional Cancer Centre, Oshawa, and Department of Oncology, Queen’s University, Kingston, ON
| | - N. Chooback
- Department of Medical Oncology, BC Cancer, Vancouver, BC
| |
Collapse
|
214
|
Kiura K, Imamura F, Kagamu H, Matsumoto S, Hida T, Nakagawa K, Satouchi M, Okamoto I, Takenoyama M, Fujisaka Y, Kurata T, Ito M, Tokushige K, Hatano B, Nishio M. Phase 3 study of ceritinib vs chemotherapy in ALK-rearranged NSCLC patients previously treated with chemotherapy and crizotinib (ASCEND-5): Japanese subset. Jpn J Clin Oncol 2018; 48:367-375. [PMID: 29474558 PMCID: PMC5892855 DOI: 10.1093/jjco/hyy016] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 01/30/2018] [Indexed: 01/19/2023] Open
Abstract
Background In the global, Phase 3, ASCEND-5 study, ceritinib improved progression-free survival (PFS) vs chemotherapy in patients with anaplastic lymphoma kinase (ALK)-rearranged non-small cell lung cancer (NSCLC) who had previously progressed on crizotinib and platinum-based chemotherapy. Here, we report efficacy and safety in a subset of Japanese patients from the ASCEND-5 study. Methods Patients with advanced ALK-rearranged NSCLC received oral ceritinib 750 mg/day or chemotherapy (intravenous pemetrexed 500 mg/m2 or docetaxel 75 mg/m2 [investigator’s choice], every 21 days). Results Among the 231 patients, 29 were Japanese, of which, 11 were treated with ceritinib and 18 were treated with chemotherapy (5 with pemetrexed and 13 with docetaxel). All the patients received prior crizotinib and one or two lines of prior chemotherapy for advanced disease. Median follow-up time was 16.6 months for ceritinib arm and 16.4 months for chemotherapy arm in the overall population. The median PFS by blinded independent review committee was 9.8 months (95% CI, 4.3–14.0) in ceritinib arm vs 1.6 months (95% CI, 1.4–3.0) in chemotherapy arm. Grade 3 or 4 adverse events, suspected to be study drug related, were reported in 36.4% of ceritinib arm and 72.2% of chemotherapy arm, respectively. No Grade 3 or 4 events of diarrhea, nausea and vomiting were reported in both the treatment arms. Adverse events leading to study drug discontinuation were reported in one patient in each arm: Grade 3 central-nervous system metastases in ceritinib-treated patient and Grade 3 febrile neutropenia in chemotherapy-treated patient. Conclusions Consistent with overall population, ceritinib demonstrated better efficacy compared with the standard second-line chemotherapy in Japanese patients with crizotinib-resistant ALK+ NSCLC. ClinicalTrials.gov identifier NCT01828112
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Isamu Okamoto
- Research Institute for Diseases of the Chest, Graduate School of Medical Sciences, Kyushu University, Fukuoka
| | | | | | | | | | | | | | | |
Collapse
|
215
|
Peters S, Zimmermann S. Management of Resistance to First-Line Anaplastic Lymphoma Kinase Tyrosine Kinase Inhibitor Therapy. Curr Treat Options Oncol 2018; 19:37. [DOI: 10.1007/s11864-018-0553-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
216
|
Yu HA, Planchard D, Lovly CM. Sequencing Therapy for Genetically Defined Subgroups of Non-Small Cell Lung Cancer. Am Soc Clin Oncol Educ Book 2018; 38:726-739. [PMID: 30231382 PMCID: PMC10172876 DOI: 10.1200/edbk_201331] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2023]
Abstract
The practice of precision medicine for patients with metastatic non-small cell lung cancer (NSCLC), particularly those patients with adenocarcinoma histology (the predominant subtype of NSCLC), has become the accepted standard of care worldwide. Implementation of prospective tumor molecular profiling and rational therapeutic decision-making based on the presence of recurrently detected oncogenic "driver" alterations in the tumor genome has revolutionized the way that lung cancer is diagnosed and treated in the clinic. Over the past two decades, there has been a deluge of therapeutically actionable driver alterations and accompanying small molecule inhibitors to target these drivers. Herein, we synthesize a large and rapidly growing body of literature regarding therapeutic inhibition of driver mutations. We focus on established targets, including EGFR, anaplastic lymphoma kinase (ALK), ROS1, BRAF, RET, MET, HER2, and neurotrophic tyrosine kinase receptor (NTRK), with a particular emphasis on the sequencing of small molecule inhibitors in these genetically defined cohorts of patients with lung cancer.
Collapse
Affiliation(s)
- Helena A Yu
- From the Department of Medicine, Memorial Sloan Kettering Cancer Center, Weil Cornell Medical College, New York, NY; Department of Medical Oncology, Institut Gustave Roussy, Villejuif, France; Department of Medicine, Division of Hematology and Oncology, Vanderbilt University Medical Center, Vanderbilt Ingram Cancer Center, Nashville, TN
| | - David Planchard
- From the Department of Medicine, Memorial Sloan Kettering Cancer Center, Weil Cornell Medical College, New York, NY; Department of Medical Oncology, Institut Gustave Roussy, Villejuif, France; Department of Medicine, Division of Hematology and Oncology, Vanderbilt University Medical Center, Vanderbilt Ingram Cancer Center, Nashville, TN
| | - Christine M Lovly
- From the Department of Medicine, Memorial Sloan Kettering Cancer Center, Weil Cornell Medical College, New York, NY; Department of Medical Oncology, Institut Gustave Roussy, Villejuif, France; Department of Medicine, Division of Hematology and Oncology, Vanderbilt University Medical Center, Vanderbilt Ingram Cancer Center, Nashville, TN
| |
Collapse
|
217
|
Liao BC, Lin CC, Yang JCH. Treating brain metastases in non-small cell lung cancer patients: what have we learnt from pharmaceutical recent clinical trials? Expert Opin Pharmacother 2018; 19:851-864. [PMID: 29726292 DOI: 10.1080/14656566.2018.1472765] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
INTRODUCTION Brain metastases (BMs) develop in up to 40% of patients with non-small cell lung cancer (NSCLC). In many recent practice-changing clinical trials, patients with BM were included; however, only few trials reported intracranial efficacies in either post hoc or pre-planned analysis. Clinically meaningful intracranial efficacy data of novel agents have not been completely disclosed. AREAS COVERED The authors performed a systemic review of recent pharmaceutical clinical trials, mainly pivotal or practice-changing trials. Some of the prospective clinical trials focused on patients with NSCLC and BM. The authors collected and compared intracranial efficacy reports of chemotherapy, epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs), ALK inhibitors, and immune checkpoint inhibitors. EXPERT OPINION Many clinical trials, especially those on 'brain-active' EGFR-TKIs and ALK inhibitors, have robust reports of intracranial efficacies either as post hoc or pre-planned analysis. Physicians should interpret this data with caution and apply the results to patients accordingly. For the design of future clinical trials, enrolling patients with only BM, incorporating novel risk classifications, pre-planning intracranial efficacy endpoints, reporting prior local brain therapies, and applying novel response evaluation criteria are emerging trends in this area.
Collapse
Affiliation(s)
- Bin-Chi Liao
- a Department of Oncology , National Taiwan University Hospital , Taipei , Taiwan.,b National Taiwan University Cancer Center, College of Medicine, National Taiwan University , Taipei , Taiwan.,c Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University , Taipei , Taiwan
| | - Chia-Chi Lin
- a Department of Oncology , National Taiwan University Hospital , Taipei , Taiwan.,c Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University , Taipei , Taiwan
| | - James Chih-Hsin Yang
- a Department of Oncology , National Taiwan University Hospital , Taipei , Taiwan.,b National Taiwan University Cancer Center, College of Medicine, National Taiwan University , Taipei , Taiwan.,c Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University , Taipei , Taiwan.,d Graduate Institute of Oncology, College of Medicine, National Taiwan University , Taipei , Taiwan
| |
Collapse
|
218
|
Camidge DR, Kim DW, Tiseo M, Langer CJ, Ahn MJ, Shaw AT, Huber RM, Hochmair MJ, Lee DH, Bazhenova LA, Gold KA, Ou SHI, West HL, Reichmann W, Haney J, Clackson T, Kerstein D, Gettinger SN. Exploratory Analysis of Brigatinib Activity in Patients With Anaplastic Lymphoma Kinase-Positive Non-Small-Cell Lung Cancer and Brain Metastases in Two Clinical Trials. J Clin Oncol 2018; 36:2693-2701. [PMID: 29768119 DOI: 10.1200/jco.2017.77.5841] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Purpose In patients with crizotinib-treated, anaplastic lymphoma kinase gene ( ALK)-rearranged non-small-cell lung cancer (ALK-positive NSCLC), initial disease progression often occurs in the CNS. We evaluated brigatinib, a next-generation ALK inhibitor, in patients with ALK-positive NSCLC with brain metastases. Patients and Methods Patients with ALK-positive NSCLC received brigatinib (90 to 240 mg total daily) in a phase I/II trial (phI/II; ClinicalTrials.gov identifier: NCT01449461) and in the subsequent randomized phase II trial ALTA (ALK in Lung Cancer Trial of AP26113; ClinicalTrials.gov identifier: NCT02094573; patients in arm A received 90 mg once daily; patients in arm B received 180 mg once daily with 7-day lead-in at 90 mg). Primary end points (systemic objective response rates [ORRs]) were previously reported. Independent review committees assessed intracranial efficacy in patients with baseline brain metastases. Results Most patients with ALK-positive NSCLC had baseline brain metastases (50 of 79 [63%], phI/II; 80 of 112 [71%] and 73 of 110 [66%] in ALTA arms A and B, respectively), many of whom had no prior brain radiotherapy (23 of 50 [46%], phI/II; 32 of 80 [40%], ALTA arm A; 30 of 73 [41%], arm B). All patients, except four in phI/II, had received crizotinib. Among patients with measurable (≥ 10 mm) brain metastases, confirmed intracranial ORR was 53% (eight of 15; 95% CI, 27% to 79%) in phI/II, 46% (12 of 26; 95% CI, 27% to 67%) in ALTA arm A, and 67% (12 of 18; 95% CI, 41% to 87%) in arm B. Intracranial ORRs were similar in subsets without prior radiation or progression postradiation. Among patients with any baseline brain metastases, median intracranial progression-free survival (iPFS) was 14.6 months (95% CI, 12.7 to 36.8 months), phI/II; 15.6 months (95% CI, 9.0 to 18.3 months), ALTA arm A; 18.4 months (95% CI, 12.8 months to not reached), ALTA arm B. Conclusion Brigatinib yielded substantial intracranial responses and durable iPFS in ALK-positive, crizotinib-treated NSCLC, with highest iPFS in patients receiving 180 mg once daily (with lead-in).
Collapse
Affiliation(s)
- D Ross Camidge
- D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO; Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center; Dae Ho Lee, Asan Medical Center, Seoul, South Korea; Marcello Tiseo, University Hospital of Parma, Parma, Italy; Corey J. Langer, University of Pennsylvania, Philadelphia, PA; Alice T. Shaw, Massachusetts General Hospital, Boston; William Reichmann, Jeff Haney, Tim Clackson, and David Kerstein, ARIAD Pharmaceuticals, Cambridge, MA; Rudolf M. Huber, University Hospital of Munich, Munich, Germany; Maximilian J. Hochmair, Otto Wagner Hospital, Vienna, Austria; Lyudmila A. Bazhenova and Kathryn A. Gold, University of California San Diego Moores Cancer Center, La Jolla; Sai-Hong Ignatius Ou, University of California Irvine School of Medicine, Irvine, CA; Howard L. West, Swedish Cancer Institute, Seattle, WA; and Scott N. Gettinger, Yale Cancer Center, New Haven, CT
| | - Dong-Wan Kim
- D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO; Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center; Dae Ho Lee, Asan Medical Center, Seoul, South Korea; Marcello Tiseo, University Hospital of Parma, Parma, Italy; Corey J. Langer, University of Pennsylvania, Philadelphia, PA; Alice T. Shaw, Massachusetts General Hospital, Boston; William Reichmann, Jeff Haney, Tim Clackson, and David Kerstein, ARIAD Pharmaceuticals, Cambridge, MA; Rudolf M. Huber, University Hospital of Munich, Munich, Germany; Maximilian J. Hochmair, Otto Wagner Hospital, Vienna, Austria; Lyudmila A. Bazhenova and Kathryn A. Gold, University of California San Diego Moores Cancer Center, La Jolla; Sai-Hong Ignatius Ou, University of California Irvine School of Medicine, Irvine, CA; Howard L. West, Swedish Cancer Institute, Seattle, WA; and Scott N. Gettinger, Yale Cancer Center, New Haven, CT
| | - Marcello Tiseo
- D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO; Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center; Dae Ho Lee, Asan Medical Center, Seoul, South Korea; Marcello Tiseo, University Hospital of Parma, Parma, Italy; Corey J. Langer, University of Pennsylvania, Philadelphia, PA; Alice T. Shaw, Massachusetts General Hospital, Boston; William Reichmann, Jeff Haney, Tim Clackson, and David Kerstein, ARIAD Pharmaceuticals, Cambridge, MA; Rudolf M. Huber, University Hospital of Munich, Munich, Germany; Maximilian J. Hochmair, Otto Wagner Hospital, Vienna, Austria; Lyudmila A. Bazhenova and Kathryn A. Gold, University of California San Diego Moores Cancer Center, La Jolla; Sai-Hong Ignatius Ou, University of California Irvine School of Medicine, Irvine, CA; Howard L. West, Swedish Cancer Institute, Seattle, WA; and Scott N. Gettinger, Yale Cancer Center, New Haven, CT
| | - Corey J Langer
- D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO; Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center; Dae Ho Lee, Asan Medical Center, Seoul, South Korea; Marcello Tiseo, University Hospital of Parma, Parma, Italy; Corey J. Langer, University of Pennsylvania, Philadelphia, PA; Alice T. Shaw, Massachusetts General Hospital, Boston; William Reichmann, Jeff Haney, Tim Clackson, and David Kerstein, ARIAD Pharmaceuticals, Cambridge, MA; Rudolf M. Huber, University Hospital of Munich, Munich, Germany; Maximilian J. Hochmair, Otto Wagner Hospital, Vienna, Austria; Lyudmila A. Bazhenova and Kathryn A. Gold, University of California San Diego Moores Cancer Center, La Jolla; Sai-Hong Ignatius Ou, University of California Irvine School of Medicine, Irvine, CA; Howard L. West, Swedish Cancer Institute, Seattle, WA; and Scott N. Gettinger, Yale Cancer Center, New Haven, CT
| | - Myung-Ju Ahn
- D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO; Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center; Dae Ho Lee, Asan Medical Center, Seoul, South Korea; Marcello Tiseo, University Hospital of Parma, Parma, Italy; Corey J. Langer, University of Pennsylvania, Philadelphia, PA; Alice T. Shaw, Massachusetts General Hospital, Boston; William Reichmann, Jeff Haney, Tim Clackson, and David Kerstein, ARIAD Pharmaceuticals, Cambridge, MA; Rudolf M. Huber, University Hospital of Munich, Munich, Germany; Maximilian J. Hochmair, Otto Wagner Hospital, Vienna, Austria; Lyudmila A. Bazhenova and Kathryn A. Gold, University of California San Diego Moores Cancer Center, La Jolla; Sai-Hong Ignatius Ou, University of California Irvine School of Medicine, Irvine, CA; Howard L. West, Swedish Cancer Institute, Seattle, WA; and Scott N. Gettinger, Yale Cancer Center, New Haven, CT
| | - Alice T Shaw
- D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO; Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center; Dae Ho Lee, Asan Medical Center, Seoul, South Korea; Marcello Tiseo, University Hospital of Parma, Parma, Italy; Corey J. Langer, University of Pennsylvania, Philadelphia, PA; Alice T. Shaw, Massachusetts General Hospital, Boston; William Reichmann, Jeff Haney, Tim Clackson, and David Kerstein, ARIAD Pharmaceuticals, Cambridge, MA; Rudolf M. Huber, University Hospital of Munich, Munich, Germany; Maximilian J. Hochmair, Otto Wagner Hospital, Vienna, Austria; Lyudmila A. Bazhenova and Kathryn A. Gold, University of California San Diego Moores Cancer Center, La Jolla; Sai-Hong Ignatius Ou, University of California Irvine School of Medicine, Irvine, CA; Howard L. West, Swedish Cancer Institute, Seattle, WA; and Scott N. Gettinger, Yale Cancer Center, New Haven, CT
| | - Rudolf M Huber
- D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO; Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center; Dae Ho Lee, Asan Medical Center, Seoul, South Korea; Marcello Tiseo, University Hospital of Parma, Parma, Italy; Corey J. Langer, University of Pennsylvania, Philadelphia, PA; Alice T. Shaw, Massachusetts General Hospital, Boston; William Reichmann, Jeff Haney, Tim Clackson, and David Kerstein, ARIAD Pharmaceuticals, Cambridge, MA; Rudolf M. Huber, University Hospital of Munich, Munich, Germany; Maximilian J. Hochmair, Otto Wagner Hospital, Vienna, Austria; Lyudmila A. Bazhenova and Kathryn A. Gold, University of California San Diego Moores Cancer Center, La Jolla; Sai-Hong Ignatius Ou, University of California Irvine School of Medicine, Irvine, CA; Howard L. West, Swedish Cancer Institute, Seattle, WA; and Scott N. Gettinger, Yale Cancer Center, New Haven, CT
| | - Maximilian J Hochmair
- D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO; Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center; Dae Ho Lee, Asan Medical Center, Seoul, South Korea; Marcello Tiseo, University Hospital of Parma, Parma, Italy; Corey J. Langer, University of Pennsylvania, Philadelphia, PA; Alice T. Shaw, Massachusetts General Hospital, Boston; William Reichmann, Jeff Haney, Tim Clackson, and David Kerstein, ARIAD Pharmaceuticals, Cambridge, MA; Rudolf M. Huber, University Hospital of Munich, Munich, Germany; Maximilian J. Hochmair, Otto Wagner Hospital, Vienna, Austria; Lyudmila A. Bazhenova and Kathryn A. Gold, University of California San Diego Moores Cancer Center, La Jolla; Sai-Hong Ignatius Ou, University of California Irvine School of Medicine, Irvine, CA; Howard L. West, Swedish Cancer Institute, Seattle, WA; and Scott N. Gettinger, Yale Cancer Center, New Haven, CT
| | - Dae Ho Lee
- D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO; Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center; Dae Ho Lee, Asan Medical Center, Seoul, South Korea; Marcello Tiseo, University Hospital of Parma, Parma, Italy; Corey J. Langer, University of Pennsylvania, Philadelphia, PA; Alice T. Shaw, Massachusetts General Hospital, Boston; William Reichmann, Jeff Haney, Tim Clackson, and David Kerstein, ARIAD Pharmaceuticals, Cambridge, MA; Rudolf M. Huber, University Hospital of Munich, Munich, Germany; Maximilian J. Hochmair, Otto Wagner Hospital, Vienna, Austria; Lyudmila A. Bazhenova and Kathryn A. Gold, University of California San Diego Moores Cancer Center, La Jolla; Sai-Hong Ignatius Ou, University of California Irvine School of Medicine, Irvine, CA; Howard L. West, Swedish Cancer Institute, Seattle, WA; and Scott N. Gettinger, Yale Cancer Center, New Haven, CT
| | - Lyudmila A Bazhenova
- D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO; Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center; Dae Ho Lee, Asan Medical Center, Seoul, South Korea; Marcello Tiseo, University Hospital of Parma, Parma, Italy; Corey J. Langer, University of Pennsylvania, Philadelphia, PA; Alice T. Shaw, Massachusetts General Hospital, Boston; William Reichmann, Jeff Haney, Tim Clackson, and David Kerstein, ARIAD Pharmaceuticals, Cambridge, MA; Rudolf M. Huber, University Hospital of Munich, Munich, Germany; Maximilian J. Hochmair, Otto Wagner Hospital, Vienna, Austria; Lyudmila A. Bazhenova and Kathryn A. Gold, University of California San Diego Moores Cancer Center, La Jolla; Sai-Hong Ignatius Ou, University of California Irvine School of Medicine, Irvine, CA; Howard L. West, Swedish Cancer Institute, Seattle, WA; and Scott N. Gettinger, Yale Cancer Center, New Haven, CT
| | - Kathryn A Gold
- D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO; Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center; Dae Ho Lee, Asan Medical Center, Seoul, South Korea; Marcello Tiseo, University Hospital of Parma, Parma, Italy; Corey J. Langer, University of Pennsylvania, Philadelphia, PA; Alice T. Shaw, Massachusetts General Hospital, Boston; William Reichmann, Jeff Haney, Tim Clackson, and David Kerstein, ARIAD Pharmaceuticals, Cambridge, MA; Rudolf M. Huber, University Hospital of Munich, Munich, Germany; Maximilian J. Hochmair, Otto Wagner Hospital, Vienna, Austria; Lyudmila A. Bazhenova and Kathryn A. Gold, University of California San Diego Moores Cancer Center, La Jolla; Sai-Hong Ignatius Ou, University of California Irvine School of Medicine, Irvine, CA; Howard L. West, Swedish Cancer Institute, Seattle, WA; and Scott N. Gettinger, Yale Cancer Center, New Haven, CT
| | - Sai-Hong Ignatius Ou
- D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO; Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center; Dae Ho Lee, Asan Medical Center, Seoul, South Korea; Marcello Tiseo, University Hospital of Parma, Parma, Italy; Corey J. Langer, University of Pennsylvania, Philadelphia, PA; Alice T. Shaw, Massachusetts General Hospital, Boston; William Reichmann, Jeff Haney, Tim Clackson, and David Kerstein, ARIAD Pharmaceuticals, Cambridge, MA; Rudolf M. Huber, University Hospital of Munich, Munich, Germany; Maximilian J. Hochmair, Otto Wagner Hospital, Vienna, Austria; Lyudmila A. Bazhenova and Kathryn A. Gold, University of California San Diego Moores Cancer Center, La Jolla; Sai-Hong Ignatius Ou, University of California Irvine School of Medicine, Irvine, CA; Howard L. West, Swedish Cancer Institute, Seattle, WA; and Scott N. Gettinger, Yale Cancer Center, New Haven, CT
| | - Howard L West
- D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO; Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center; Dae Ho Lee, Asan Medical Center, Seoul, South Korea; Marcello Tiseo, University Hospital of Parma, Parma, Italy; Corey J. Langer, University of Pennsylvania, Philadelphia, PA; Alice T. Shaw, Massachusetts General Hospital, Boston; William Reichmann, Jeff Haney, Tim Clackson, and David Kerstein, ARIAD Pharmaceuticals, Cambridge, MA; Rudolf M. Huber, University Hospital of Munich, Munich, Germany; Maximilian J. Hochmair, Otto Wagner Hospital, Vienna, Austria; Lyudmila A. Bazhenova and Kathryn A. Gold, University of California San Diego Moores Cancer Center, La Jolla; Sai-Hong Ignatius Ou, University of California Irvine School of Medicine, Irvine, CA; Howard L. West, Swedish Cancer Institute, Seattle, WA; and Scott N. Gettinger, Yale Cancer Center, New Haven, CT
| | - William Reichmann
- D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO; Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center; Dae Ho Lee, Asan Medical Center, Seoul, South Korea; Marcello Tiseo, University Hospital of Parma, Parma, Italy; Corey J. Langer, University of Pennsylvania, Philadelphia, PA; Alice T. Shaw, Massachusetts General Hospital, Boston; William Reichmann, Jeff Haney, Tim Clackson, and David Kerstein, ARIAD Pharmaceuticals, Cambridge, MA; Rudolf M. Huber, University Hospital of Munich, Munich, Germany; Maximilian J. Hochmair, Otto Wagner Hospital, Vienna, Austria; Lyudmila A. Bazhenova and Kathryn A. Gold, University of California San Diego Moores Cancer Center, La Jolla; Sai-Hong Ignatius Ou, University of California Irvine School of Medicine, Irvine, CA; Howard L. West, Swedish Cancer Institute, Seattle, WA; and Scott N. Gettinger, Yale Cancer Center, New Haven, CT
| | - Jeff Haney
- D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO; Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center; Dae Ho Lee, Asan Medical Center, Seoul, South Korea; Marcello Tiseo, University Hospital of Parma, Parma, Italy; Corey J. Langer, University of Pennsylvania, Philadelphia, PA; Alice T. Shaw, Massachusetts General Hospital, Boston; William Reichmann, Jeff Haney, Tim Clackson, and David Kerstein, ARIAD Pharmaceuticals, Cambridge, MA; Rudolf M. Huber, University Hospital of Munich, Munich, Germany; Maximilian J. Hochmair, Otto Wagner Hospital, Vienna, Austria; Lyudmila A. Bazhenova and Kathryn A. Gold, University of California San Diego Moores Cancer Center, La Jolla; Sai-Hong Ignatius Ou, University of California Irvine School of Medicine, Irvine, CA; Howard L. West, Swedish Cancer Institute, Seattle, WA; and Scott N. Gettinger, Yale Cancer Center, New Haven, CT
| | - Tim Clackson
- D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO; Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center; Dae Ho Lee, Asan Medical Center, Seoul, South Korea; Marcello Tiseo, University Hospital of Parma, Parma, Italy; Corey J. Langer, University of Pennsylvania, Philadelphia, PA; Alice T. Shaw, Massachusetts General Hospital, Boston; William Reichmann, Jeff Haney, Tim Clackson, and David Kerstein, ARIAD Pharmaceuticals, Cambridge, MA; Rudolf M. Huber, University Hospital of Munich, Munich, Germany; Maximilian J. Hochmair, Otto Wagner Hospital, Vienna, Austria; Lyudmila A. Bazhenova and Kathryn A. Gold, University of California San Diego Moores Cancer Center, La Jolla; Sai-Hong Ignatius Ou, University of California Irvine School of Medicine, Irvine, CA; Howard L. West, Swedish Cancer Institute, Seattle, WA; and Scott N. Gettinger, Yale Cancer Center, New Haven, CT
| | - David Kerstein
- D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO; Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center; Dae Ho Lee, Asan Medical Center, Seoul, South Korea; Marcello Tiseo, University Hospital of Parma, Parma, Italy; Corey J. Langer, University of Pennsylvania, Philadelphia, PA; Alice T. Shaw, Massachusetts General Hospital, Boston; William Reichmann, Jeff Haney, Tim Clackson, and David Kerstein, ARIAD Pharmaceuticals, Cambridge, MA; Rudolf M. Huber, University Hospital of Munich, Munich, Germany; Maximilian J. Hochmair, Otto Wagner Hospital, Vienna, Austria; Lyudmila A. Bazhenova and Kathryn A. Gold, University of California San Diego Moores Cancer Center, La Jolla; Sai-Hong Ignatius Ou, University of California Irvine School of Medicine, Irvine, CA; Howard L. West, Swedish Cancer Institute, Seattle, WA; and Scott N. Gettinger, Yale Cancer Center, New Haven, CT
| | - Scott N Gettinger
- D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO; Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center; Dae Ho Lee, Asan Medical Center, Seoul, South Korea; Marcello Tiseo, University Hospital of Parma, Parma, Italy; Corey J. Langer, University of Pennsylvania, Philadelphia, PA; Alice T. Shaw, Massachusetts General Hospital, Boston; William Reichmann, Jeff Haney, Tim Clackson, and David Kerstein, ARIAD Pharmaceuticals, Cambridge, MA; Rudolf M. Huber, University Hospital of Munich, Munich, Germany; Maximilian J. Hochmair, Otto Wagner Hospital, Vienna, Austria; Lyudmila A. Bazhenova and Kathryn A. Gold, University of California San Diego Moores Cancer Center, La Jolla; Sai-Hong Ignatius Ou, University of California Irvine School of Medicine, Irvine, CA; Howard L. West, Swedish Cancer Institute, Seattle, WA; and Scott N. Gettinger, Yale Cancer Center, New Haven, CT
| |
Collapse
|
219
|
Burudpakdee C, Wong W, Seetasith A, Corvino F, Yeh W, Gubens M. Economic impact of preventing brain metastases with alectinib in ALK-positive non-small cell lung cancer. Lung Cancer 2018; 119:103-111. [DOI: 10.1016/j.lungcan.2018.03.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 02/05/2018] [Accepted: 03/08/2018] [Indexed: 10/17/2022]
|
220
|
Ippen FM, D'Andrea M, Brastianos PK. Personalized medicine in brain metastases: a plea for more translational studies. Per Med 2018; 15:141-143. [PMID: 29798715 DOI: 10.2217/pme-2017-0087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Affiliation(s)
- Franziska M Ippen
- Divisions of Hematology/Oncology & Neuro-Oncology, Departments of Medicine & Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Megan D'Andrea
- Divisions of Hematology/Oncology & Neuro-Oncology, Departments of Medicine & Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Priscilla K Brastianos
- Divisions of Hematology/Oncology & Neuro-Oncology, Departments of Medicine & Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA
| |
Collapse
|
221
|
Costa RB, Costa RLB, Talamantes SM, Kaplan JB, Bhave MA, Rademaker A, Miller C, Carneiro BA, Mahalingam D, Chae YK. Systematic review and meta-analysis of selected toxicities of approved ALK inhibitors in metastatic non-small cell lung cancer. Oncotarget 2018; 9:22137-22146. [PMID: 29774128 PMCID: PMC5955140 DOI: 10.18632/oncotarget.25154] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Accepted: 04/04/2018] [Indexed: 12/19/2022] Open
Abstract
Introduction Anaplastic lymphoma kinase (ALK) inhibitors are the mainstay treatment for patients with non-small cell lung carcinoma (NSCLC) harboring a rearrangement of the ALK gene or the ROS1 oncogenes. With the recent publication of pivotal trials leading to the approval of these compounds in different indications, their toxicity profile warrants an update. Materials and Methods A systematic literature search was performed in July 2017. Studies evaluating US FDA approved doses of one of the following ALK inhibitors: Crizotinib, Ceritinib, Alectinib or Brigatinib as monotherapy were included. Data were analyzed using random effects meta-analysis for absolute risks (AR), study heterogeneity, publication bias and differences among treatments. Results Fifteen trials with a total of 2,005 patients with evaluable toxicity data were included in this report. There was significant heterogeneity amongst different studies. The pooled AR of death and severe adverse events were 0.5% and 34.5%, respectively. Grade 3/4 nausea, vomiting, diarrhea, and constipation were uncommon: 2.6%, 2.5%, 2.7%, 1.2%, respectively. Conclusions ALK inhibitors have an acceptable safety profile with a low risk of treatment-related deaths. Important differences in toxicity profile were detected amongst the different drugs.
Collapse
Affiliation(s)
- Rubens Barros Costa
- Developmental Therapeutics Program, Northwestern University, Chicago, IL, USA
| | - Ricardo L B Costa
- Department of Breast Oncology, Lee Moffitt Cancer Center, Tampa, USA
| | | | - Jason B Kaplan
- Developmental Therapeutics Program, Northwestern University, Chicago, IL, USA
| | - Manali A Bhave
- Developmental Therapeutics Program, Northwestern University, Chicago, IL, USA
| | - Alfred Rademaker
- Northwestern University, Department of Preventive Medicine, Chicago, IL, USA
| | - Corinne Miller
- Galter Health Sciences Library, Northwestern University, Chicago, IL, USA
| | | | | | - Young Kwang Chae
- Developmental Therapeutics Program, Northwestern University, Chicago, IL, USA
| |
Collapse
|
222
|
Lin JJ, Zhu VW, Yoda S, Yeap BY, Schrock AB, Dagogo-Jack I, Jessop NA, Jiang GY, Le LP, Gowen K, Stephens PJ, Ross JS, Ali SM, Miller VA, Johnson ML, Lovly CM, Hata AN, Gainor JF, Iafrate AJ, Shaw AT, Ou SHI. Impact of EML4-ALK Variant on Resistance Mechanisms and Clinical Outcomes in ALK-Positive Lung Cancer. J Clin Oncol 2018; 36:1199-1206. [PMID: 29373100 PMCID: PMC5903999 DOI: 10.1200/jco.2017.76.2294] [Citation(s) in RCA: 253] [Impact Index Per Article: 36.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Purpose Advanced anaplastic lymphoma kinase ( ALK) fusion-positive non-small-cell lung cancers (NSCLCs) are effectively treated with ALK tyrosine kinase inhibitors (TKIs). However, clinical outcomes in these patients vary, and the benefit of TKIs is limited as a result of acquired resistance. Emerging data suggest that the ALK fusion variant may affect clinical outcome, but the molecular basis for this association is unknown. Patients and Methods We identified 129 patients with ALK-positive NSCLC with known ALK variants. ALK resistance mutations and clinical outcomes on ALK TKIs were retrospectively evaluated according to ALK variant. A Foundation Medicine data set of 577 patients with ALK-positive NSCLC was also examined. Results The most frequent ALK variants were EML4-ALK variant 1 in 55 patients (43%) and variant 3 in 51 patients (40%). We analyzed 77 tumor biopsy specimens from patients with variants 1 and 3 who had progressed on an ALK TKI. ALK resistance mutations were significantly more common in variant 3 than in variant 1 (57% v 30%; P = .023). In particular, ALK G1202R was more common in variant 3 than in variant 1 (32% v 0%; P < .001). Analysis of the Foundation Medicine database revealed similar associations of variant 3 with ALK resistance mutation and with G1202R ( P = .010 and .015, respectively). Among patients treated with the third-generation ALK TKI lorlatinib, variant 3 was associated with a significantly longer progression-free survival than variant 1 (hazard ratio, 0.31; 95% CI, 0.12 to 0.79; P = .011). Conclusion Specific ALK variants may be associated with the development of ALK resistance mutations, particularly G1202R, and provide a molecular link between variant and clinical outcome. ALK variant thus represents a potentially important factor in the selection of next-generation ALK inhibitors.
Collapse
Affiliation(s)
- Jessica J. Lin
- Jessica J. Lin, Satoshi Yoda, Beow Y. Yeap, Ibiayi Dagogo-Jack, Nicholas A. Jessop, Ginger Y. Jiang, Long P. Le, Aaron N. Hata, Justin F. Gainor, Anthony J. Iafrate, and Alice T. Shaw, Massachusetts General Hospital, Boston; Alexa B. Schrock, Kyle Gowen, Philip J. Stephens, Jeffrey S. Ross, Siraj M. Ali, and Vincent A. Miller, Foundation Medicine, Cambridge, MA; Viola W. Zhu and Sai-Hong Ignatius Ou, Chao Family Comprehensive Cancer Center, University of California Irvine School of Medicine, Orange, CA; Melissa L. Johnson, Sarah Cannon Research Institute; and Christine M. Lovly, Vanderbilt-Ingram Cancer Center, Nashville, TN
| | - Viola W. Zhu
- Jessica J. Lin, Satoshi Yoda, Beow Y. Yeap, Ibiayi Dagogo-Jack, Nicholas A. Jessop, Ginger Y. Jiang, Long P. Le, Aaron N. Hata, Justin F. Gainor, Anthony J. Iafrate, and Alice T. Shaw, Massachusetts General Hospital, Boston; Alexa B. Schrock, Kyle Gowen, Philip J. Stephens, Jeffrey S. Ross, Siraj M. Ali, and Vincent A. Miller, Foundation Medicine, Cambridge, MA; Viola W. Zhu and Sai-Hong Ignatius Ou, Chao Family Comprehensive Cancer Center, University of California Irvine School of Medicine, Orange, CA; Melissa L. Johnson, Sarah Cannon Research Institute; and Christine M. Lovly, Vanderbilt-Ingram Cancer Center, Nashville, TN
| | - Satoshi Yoda
- Jessica J. Lin, Satoshi Yoda, Beow Y. Yeap, Ibiayi Dagogo-Jack, Nicholas A. Jessop, Ginger Y. Jiang, Long P. Le, Aaron N. Hata, Justin F. Gainor, Anthony J. Iafrate, and Alice T. Shaw, Massachusetts General Hospital, Boston; Alexa B. Schrock, Kyle Gowen, Philip J. Stephens, Jeffrey S. Ross, Siraj M. Ali, and Vincent A. Miller, Foundation Medicine, Cambridge, MA; Viola W. Zhu and Sai-Hong Ignatius Ou, Chao Family Comprehensive Cancer Center, University of California Irvine School of Medicine, Orange, CA; Melissa L. Johnson, Sarah Cannon Research Institute; and Christine M. Lovly, Vanderbilt-Ingram Cancer Center, Nashville, TN
| | - Beow Y. Yeap
- Jessica J. Lin, Satoshi Yoda, Beow Y. Yeap, Ibiayi Dagogo-Jack, Nicholas A. Jessop, Ginger Y. Jiang, Long P. Le, Aaron N. Hata, Justin F. Gainor, Anthony J. Iafrate, and Alice T. Shaw, Massachusetts General Hospital, Boston; Alexa B. Schrock, Kyle Gowen, Philip J. Stephens, Jeffrey S. Ross, Siraj M. Ali, and Vincent A. Miller, Foundation Medicine, Cambridge, MA; Viola W. Zhu and Sai-Hong Ignatius Ou, Chao Family Comprehensive Cancer Center, University of California Irvine School of Medicine, Orange, CA; Melissa L. Johnson, Sarah Cannon Research Institute; and Christine M. Lovly, Vanderbilt-Ingram Cancer Center, Nashville, TN
| | - Alexa B. Schrock
- Jessica J. Lin, Satoshi Yoda, Beow Y. Yeap, Ibiayi Dagogo-Jack, Nicholas A. Jessop, Ginger Y. Jiang, Long P. Le, Aaron N. Hata, Justin F. Gainor, Anthony J. Iafrate, and Alice T. Shaw, Massachusetts General Hospital, Boston; Alexa B. Schrock, Kyle Gowen, Philip J. Stephens, Jeffrey S. Ross, Siraj M. Ali, and Vincent A. Miller, Foundation Medicine, Cambridge, MA; Viola W. Zhu and Sai-Hong Ignatius Ou, Chao Family Comprehensive Cancer Center, University of California Irvine School of Medicine, Orange, CA; Melissa L. Johnson, Sarah Cannon Research Institute; and Christine M. Lovly, Vanderbilt-Ingram Cancer Center, Nashville, TN
| | - Ibiayi Dagogo-Jack
- Jessica J. Lin, Satoshi Yoda, Beow Y. Yeap, Ibiayi Dagogo-Jack, Nicholas A. Jessop, Ginger Y. Jiang, Long P. Le, Aaron N. Hata, Justin F. Gainor, Anthony J. Iafrate, and Alice T. Shaw, Massachusetts General Hospital, Boston; Alexa B. Schrock, Kyle Gowen, Philip J. Stephens, Jeffrey S. Ross, Siraj M. Ali, and Vincent A. Miller, Foundation Medicine, Cambridge, MA; Viola W. Zhu and Sai-Hong Ignatius Ou, Chao Family Comprehensive Cancer Center, University of California Irvine School of Medicine, Orange, CA; Melissa L. Johnson, Sarah Cannon Research Institute; and Christine M. Lovly, Vanderbilt-Ingram Cancer Center, Nashville, TN
| | - Nicholas A. Jessop
- Jessica J. Lin, Satoshi Yoda, Beow Y. Yeap, Ibiayi Dagogo-Jack, Nicholas A. Jessop, Ginger Y. Jiang, Long P. Le, Aaron N. Hata, Justin F. Gainor, Anthony J. Iafrate, and Alice T. Shaw, Massachusetts General Hospital, Boston; Alexa B. Schrock, Kyle Gowen, Philip J. Stephens, Jeffrey S. Ross, Siraj M. Ali, and Vincent A. Miller, Foundation Medicine, Cambridge, MA; Viola W. Zhu and Sai-Hong Ignatius Ou, Chao Family Comprehensive Cancer Center, University of California Irvine School of Medicine, Orange, CA; Melissa L. Johnson, Sarah Cannon Research Institute; and Christine M. Lovly, Vanderbilt-Ingram Cancer Center, Nashville, TN
| | - Ginger Y. Jiang
- Jessica J. Lin, Satoshi Yoda, Beow Y. Yeap, Ibiayi Dagogo-Jack, Nicholas A. Jessop, Ginger Y. Jiang, Long P. Le, Aaron N. Hata, Justin F. Gainor, Anthony J. Iafrate, and Alice T. Shaw, Massachusetts General Hospital, Boston; Alexa B. Schrock, Kyle Gowen, Philip J. Stephens, Jeffrey S. Ross, Siraj M. Ali, and Vincent A. Miller, Foundation Medicine, Cambridge, MA; Viola W. Zhu and Sai-Hong Ignatius Ou, Chao Family Comprehensive Cancer Center, University of California Irvine School of Medicine, Orange, CA; Melissa L. Johnson, Sarah Cannon Research Institute; and Christine M. Lovly, Vanderbilt-Ingram Cancer Center, Nashville, TN
| | - Long P. Le
- Jessica J. Lin, Satoshi Yoda, Beow Y. Yeap, Ibiayi Dagogo-Jack, Nicholas A. Jessop, Ginger Y. Jiang, Long P. Le, Aaron N. Hata, Justin F. Gainor, Anthony J. Iafrate, and Alice T. Shaw, Massachusetts General Hospital, Boston; Alexa B. Schrock, Kyle Gowen, Philip J. Stephens, Jeffrey S. Ross, Siraj M. Ali, and Vincent A. Miller, Foundation Medicine, Cambridge, MA; Viola W. Zhu and Sai-Hong Ignatius Ou, Chao Family Comprehensive Cancer Center, University of California Irvine School of Medicine, Orange, CA; Melissa L. Johnson, Sarah Cannon Research Institute; and Christine M. Lovly, Vanderbilt-Ingram Cancer Center, Nashville, TN
| | - Kyle Gowen
- Jessica J. Lin, Satoshi Yoda, Beow Y. Yeap, Ibiayi Dagogo-Jack, Nicholas A. Jessop, Ginger Y. Jiang, Long P. Le, Aaron N. Hata, Justin F. Gainor, Anthony J. Iafrate, and Alice T. Shaw, Massachusetts General Hospital, Boston; Alexa B. Schrock, Kyle Gowen, Philip J. Stephens, Jeffrey S. Ross, Siraj M. Ali, and Vincent A. Miller, Foundation Medicine, Cambridge, MA; Viola W. Zhu and Sai-Hong Ignatius Ou, Chao Family Comprehensive Cancer Center, University of California Irvine School of Medicine, Orange, CA; Melissa L. Johnson, Sarah Cannon Research Institute; and Christine M. Lovly, Vanderbilt-Ingram Cancer Center, Nashville, TN
| | - Philip J. Stephens
- Jessica J. Lin, Satoshi Yoda, Beow Y. Yeap, Ibiayi Dagogo-Jack, Nicholas A. Jessop, Ginger Y. Jiang, Long P. Le, Aaron N. Hata, Justin F. Gainor, Anthony J. Iafrate, and Alice T. Shaw, Massachusetts General Hospital, Boston; Alexa B. Schrock, Kyle Gowen, Philip J. Stephens, Jeffrey S. Ross, Siraj M. Ali, and Vincent A. Miller, Foundation Medicine, Cambridge, MA; Viola W. Zhu and Sai-Hong Ignatius Ou, Chao Family Comprehensive Cancer Center, University of California Irvine School of Medicine, Orange, CA; Melissa L. Johnson, Sarah Cannon Research Institute; and Christine M. Lovly, Vanderbilt-Ingram Cancer Center, Nashville, TN
| | - Jeffrey S. Ross
- Jessica J. Lin, Satoshi Yoda, Beow Y. Yeap, Ibiayi Dagogo-Jack, Nicholas A. Jessop, Ginger Y. Jiang, Long P. Le, Aaron N. Hata, Justin F. Gainor, Anthony J. Iafrate, and Alice T. Shaw, Massachusetts General Hospital, Boston; Alexa B. Schrock, Kyle Gowen, Philip J. Stephens, Jeffrey S. Ross, Siraj M. Ali, and Vincent A. Miller, Foundation Medicine, Cambridge, MA; Viola W. Zhu and Sai-Hong Ignatius Ou, Chao Family Comprehensive Cancer Center, University of California Irvine School of Medicine, Orange, CA; Melissa L. Johnson, Sarah Cannon Research Institute; and Christine M. Lovly, Vanderbilt-Ingram Cancer Center, Nashville, TN
| | - Siraj M. Ali
- Jessica J. Lin, Satoshi Yoda, Beow Y. Yeap, Ibiayi Dagogo-Jack, Nicholas A. Jessop, Ginger Y. Jiang, Long P. Le, Aaron N. Hata, Justin F. Gainor, Anthony J. Iafrate, and Alice T. Shaw, Massachusetts General Hospital, Boston; Alexa B. Schrock, Kyle Gowen, Philip J. Stephens, Jeffrey S. Ross, Siraj M. Ali, and Vincent A. Miller, Foundation Medicine, Cambridge, MA; Viola W. Zhu and Sai-Hong Ignatius Ou, Chao Family Comprehensive Cancer Center, University of California Irvine School of Medicine, Orange, CA; Melissa L. Johnson, Sarah Cannon Research Institute; and Christine M. Lovly, Vanderbilt-Ingram Cancer Center, Nashville, TN
| | - Vincent A. Miller
- Jessica J. Lin, Satoshi Yoda, Beow Y. Yeap, Ibiayi Dagogo-Jack, Nicholas A. Jessop, Ginger Y. Jiang, Long P. Le, Aaron N. Hata, Justin F. Gainor, Anthony J. Iafrate, and Alice T. Shaw, Massachusetts General Hospital, Boston; Alexa B. Schrock, Kyle Gowen, Philip J. Stephens, Jeffrey S. Ross, Siraj M. Ali, and Vincent A. Miller, Foundation Medicine, Cambridge, MA; Viola W. Zhu and Sai-Hong Ignatius Ou, Chao Family Comprehensive Cancer Center, University of California Irvine School of Medicine, Orange, CA; Melissa L. Johnson, Sarah Cannon Research Institute; and Christine M. Lovly, Vanderbilt-Ingram Cancer Center, Nashville, TN
| | - Melissa L. Johnson
- Jessica J. Lin, Satoshi Yoda, Beow Y. Yeap, Ibiayi Dagogo-Jack, Nicholas A. Jessop, Ginger Y. Jiang, Long P. Le, Aaron N. Hata, Justin F. Gainor, Anthony J. Iafrate, and Alice T. Shaw, Massachusetts General Hospital, Boston; Alexa B. Schrock, Kyle Gowen, Philip J. Stephens, Jeffrey S. Ross, Siraj M. Ali, and Vincent A. Miller, Foundation Medicine, Cambridge, MA; Viola W. Zhu and Sai-Hong Ignatius Ou, Chao Family Comprehensive Cancer Center, University of California Irvine School of Medicine, Orange, CA; Melissa L. Johnson, Sarah Cannon Research Institute; and Christine M. Lovly, Vanderbilt-Ingram Cancer Center, Nashville, TN
| | - Christine M. Lovly
- Jessica J. Lin, Satoshi Yoda, Beow Y. Yeap, Ibiayi Dagogo-Jack, Nicholas A. Jessop, Ginger Y. Jiang, Long P. Le, Aaron N. Hata, Justin F. Gainor, Anthony J. Iafrate, and Alice T. Shaw, Massachusetts General Hospital, Boston; Alexa B. Schrock, Kyle Gowen, Philip J. Stephens, Jeffrey S. Ross, Siraj M. Ali, and Vincent A. Miller, Foundation Medicine, Cambridge, MA; Viola W. Zhu and Sai-Hong Ignatius Ou, Chao Family Comprehensive Cancer Center, University of California Irvine School of Medicine, Orange, CA; Melissa L. Johnson, Sarah Cannon Research Institute; and Christine M. Lovly, Vanderbilt-Ingram Cancer Center, Nashville, TN
| | - Aaron N. Hata
- Jessica J. Lin, Satoshi Yoda, Beow Y. Yeap, Ibiayi Dagogo-Jack, Nicholas A. Jessop, Ginger Y. Jiang, Long P. Le, Aaron N. Hata, Justin F. Gainor, Anthony J. Iafrate, and Alice T. Shaw, Massachusetts General Hospital, Boston; Alexa B. Schrock, Kyle Gowen, Philip J. Stephens, Jeffrey S. Ross, Siraj M. Ali, and Vincent A. Miller, Foundation Medicine, Cambridge, MA; Viola W. Zhu and Sai-Hong Ignatius Ou, Chao Family Comprehensive Cancer Center, University of California Irvine School of Medicine, Orange, CA; Melissa L. Johnson, Sarah Cannon Research Institute; and Christine M. Lovly, Vanderbilt-Ingram Cancer Center, Nashville, TN
| | - Justin F. Gainor
- Jessica J. Lin, Satoshi Yoda, Beow Y. Yeap, Ibiayi Dagogo-Jack, Nicholas A. Jessop, Ginger Y. Jiang, Long P. Le, Aaron N. Hata, Justin F. Gainor, Anthony J. Iafrate, and Alice T. Shaw, Massachusetts General Hospital, Boston; Alexa B. Schrock, Kyle Gowen, Philip J. Stephens, Jeffrey S. Ross, Siraj M. Ali, and Vincent A. Miller, Foundation Medicine, Cambridge, MA; Viola W. Zhu and Sai-Hong Ignatius Ou, Chao Family Comprehensive Cancer Center, University of California Irvine School of Medicine, Orange, CA; Melissa L. Johnson, Sarah Cannon Research Institute; and Christine M. Lovly, Vanderbilt-Ingram Cancer Center, Nashville, TN
| | - Anthony J. Iafrate
- Jessica J. Lin, Satoshi Yoda, Beow Y. Yeap, Ibiayi Dagogo-Jack, Nicholas A. Jessop, Ginger Y. Jiang, Long P. Le, Aaron N. Hata, Justin F. Gainor, Anthony J. Iafrate, and Alice T. Shaw, Massachusetts General Hospital, Boston; Alexa B. Schrock, Kyle Gowen, Philip J. Stephens, Jeffrey S. Ross, Siraj M. Ali, and Vincent A. Miller, Foundation Medicine, Cambridge, MA; Viola W. Zhu and Sai-Hong Ignatius Ou, Chao Family Comprehensive Cancer Center, University of California Irvine School of Medicine, Orange, CA; Melissa L. Johnson, Sarah Cannon Research Institute; and Christine M. Lovly, Vanderbilt-Ingram Cancer Center, Nashville, TN
| | - Alice T. Shaw
- Jessica J. Lin, Satoshi Yoda, Beow Y. Yeap, Ibiayi Dagogo-Jack, Nicholas A. Jessop, Ginger Y. Jiang, Long P. Le, Aaron N. Hata, Justin F. Gainor, Anthony J. Iafrate, and Alice T. Shaw, Massachusetts General Hospital, Boston; Alexa B. Schrock, Kyle Gowen, Philip J. Stephens, Jeffrey S. Ross, Siraj M. Ali, and Vincent A. Miller, Foundation Medicine, Cambridge, MA; Viola W. Zhu and Sai-Hong Ignatius Ou, Chao Family Comprehensive Cancer Center, University of California Irvine School of Medicine, Orange, CA; Melissa L. Johnson, Sarah Cannon Research Institute; and Christine M. Lovly, Vanderbilt-Ingram Cancer Center, Nashville, TN.,Corresponding author: Alice T. Shaw, MD, PhD, Massachusetts General Hospital Cancer Center, Department of Thoracic Oncology, 32 Fruit St, Boston, MA 02114; e-mail:
| | - Sai-Hong Ignatius Ou
- Jessica J. Lin, Satoshi Yoda, Beow Y. Yeap, Ibiayi Dagogo-Jack, Nicholas A. Jessop, Ginger Y. Jiang, Long P. Le, Aaron N. Hata, Justin F. Gainor, Anthony J. Iafrate, and Alice T. Shaw, Massachusetts General Hospital, Boston; Alexa B. Schrock, Kyle Gowen, Philip J. Stephens, Jeffrey S. Ross, Siraj M. Ali, and Vincent A. Miller, Foundation Medicine, Cambridge, MA; Viola W. Zhu and Sai-Hong Ignatius Ou, Chao Family Comprehensive Cancer Center, University of California Irvine School of Medicine, Orange, CA; Melissa L. Johnson, Sarah Cannon Research Institute; and Christine M. Lovly, Vanderbilt-Ingram Cancer Center, Nashville, TN
| |
Collapse
|
223
|
Ricciuti B, De Giglio A, Mecca C, Arcuri C, Marini S, Metro G, Baglivo S, Sidoni A, Bellezza G, Crinò L, Chiari R. Precision medicine against ALK-positive non-small cell lung cancer: beyond crizotinib. Med Oncol 2018; 35:72. [PMID: 29666949 DOI: 10.1007/s12032-018-1133-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 04/11/2018] [Indexed: 12/31/2022]
Abstract
Anaplastic lymphoma kinase (ALK) rearrangements represent the molecular driver of a subset of non-small cell lung cancers (NSCLCs). Despite the initial response, virtually all ALK-positive patients develop an acquired resistance to the ALK inhibitor crizotinib, usually within 12 months. Several next-generation ALK inhibitors have been developed in order to overcome crizotinib limitation, providing an unprecedented survival for this subset of patients. The aim of this review to summarize the current knowledge on ALK tyrosine kinase inhibitors (TKIs) in the treatment of advanced ALK-positive NSCLC, focusing on the role of novel ALK inhibitors in this setting. In addition, we will discuss their role in the pharmacological management of ALK-positive brain metastasis. Next-generation ALK inhibitors showed an impressive clinical activity in ALK-positive NSCLC, also against the sanctuary site of CNS. Sequential therapy with ALK TKIs appears to be effective in patients who fail a first ALK TKI and translates in clinically meaningful benefit. However, these agents display different activity profiles against crizotinib resistance mutation; therefore re-genotyping the disease at progression in order to administer the right TKI to the right patient is going to be necessary to correctly tailor the treatment. To avoid repeated invasive procedure, noninvasive methods to detect and monitor ALK rearrangement are under clinical investigation.
Collapse
Affiliation(s)
- Biagio Ricciuti
- Department of Medical Oncology, Santa Maria della Misericordia Hospital, University of Perugia, Piazzale L. Severi n. 1, 06132, Perugia, Italy.
| | - Andrea De Giglio
- Department of Medical Oncology, Santa Maria della Misericordia Hospital, University of Perugia, Piazzale L. Severi n. 1, 06132, Perugia, Italy
| | - Carmen Mecca
- Department of Experimental Medicine, Perugia Medical School, University of Perugia, Piazzale L. Severi n. 1, 06132, Perugia, Italy
| | - Cataldo Arcuri
- Department of Experimental Medicine, Perugia Medical School, University of Perugia, Piazzale L. Severi n. 1, 06132, Perugia, Italy
| | - Sabrina Marini
- Department of Medical Oncology, Santa Maria della Misericordia Hospital, University of Perugia, Piazzale L. Severi n. 1, 06132, Perugia, Italy
| | - Giulio Metro
- Department of Medical Oncology, Santa Maria della Misericordia Hospital, University of Perugia, Piazzale L. Severi n. 1, 06132, Perugia, Italy
| | - Sara Baglivo
- Department of Medical Oncology, Santa Maria della Misericordia Hospital, University of Perugia, Piazzale L. Severi n. 1, 06132, Perugia, Italy
| | - Angelo Sidoni
- Division of Pathology and Histology, Department of Experimental Medicine, Perugia Medical School, University of Perugia, Piazzale L. Severi n. 1, 06132, Perugia, Italy
| | - Guido Bellezza
- Division of Pathology and Histology, Department of Experimental Medicine, Perugia Medical School, University of Perugia, Piazzale L. Severi n. 1, 06132, Perugia, Italy
| | - Lucio Crinò
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori, Meldola FC, Italy
| | - Rita Chiari
- Department of Medical Oncology, Santa Maria della Misericordia Hospital, University of Perugia, Piazzale L. Severi n. 1, 06132, Perugia, Italy
| |
Collapse
|
224
|
Remon J, Besse B. Brain Metastases in Oncogene-Addicted Non-Small Cell Lung Cancer Patients: Incidence and Treatment. Front Oncol 2018; 8:88. [PMID: 29696132 PMCID: PMC5904204 DOI: 10.3389/fonc.2018.00088] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 03/14/2018] [Indexed: 12/11/2022] Open
Abstract
Brain metastases (BM) are common in non-small cell lung cancer patients including in molecularly selected populations, such as EGFR-mutant and ALK-rearranged tumors. They are associated with a reduced quality of life, and are commonly the first site of progression for patients receiving tyrosine kinase inhibitors (TKIs). In this review, we summarize incidence of BM and intracranial efficacy with TKI agents according to oncogene driver mutations, focusing on important clinical issues, notably optimal first-line treatment in oncogene-addicted lung tumors with upfront BM (local therapies followed by TKI vs. TKI monotherapy). We also discuss the potential role of newly emerging late-generation TKIs as new standard treatment in oncogene-addicted lung cancer tumors compared with sequential strategies.
Collapse
Affiliation(s)
- J. Remon
- Medical Oncology Department, Hospital Universitari Vall d’Hebron, Barcelona, Spain
| | - Benjamin Besse
- Cancer Medicine Department, Institut Gustave Roussy, Villejuif, France
- University Paris-Sud, Orsay, France
| |
Collapse
|
225
|
Kim HK, Ahn MJ. Brigatinib for treatment of anaplastic lymphoma kinase-rearranged metastatic non-small cell lung cancer. Expert Opin Orphan Drugs 2018. [DOI: 10.1080/21678707.2018.1451325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Affiliation(s)
- Hee Kyung Kim
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
- Division of Oncology, Department of Internal Medicine, Chungbuk National University Hospital, Chungbuk National University College of Medicine, Cheongju, Korea
| | - Myung-Ju Ahn
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
226
|
Ziogas DC, Tsiara A, Tsironis G, Lykka M, Liontos M, Bamias A, Dimopoulos MA. Treating ALK-positive non-small cell lung cancer. ANNALS OF TRANSLATIONAL MEDICINE 2018; 6:141. [PMID: 29862230 DOI: 10.21037/atm.2017.11.34] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Targeting genomic alterations, such as epidermal growth factor receptor (EGFR) mutations and anaplastic lymphoma kinase (ALK) gene rearrangements, have radically changed the treatment of patients with non-small cell lung cancer (NSCLC). In the case of ALK-rearranged gene, subsequent rapid development of effective genotype-directed therapies with ALK tyrosine kinase inhibitors (TKIs) triggered major advances in the personalized molecularly based approach of NSCLC. Crizotinib was the first-in-class ALK TKI with proven superiority over standard platinum-based chemotherapy for the 1st-line therapy of ALK-rearranged NSCLC patients. However, the acquired resistance to crizotinib and its diminished efficacy to the central nervous system (CNS) relapse led to the development of several novel ALK inhibitors, more potent and with different selectivity compared to crizotinib. To date, four ALK TKIs, crizotinib, ceritinib, alectinib and brigatinib have received approval from the Food and Drug Administration (FDA) and/or the European Medicines Agency (EMA) and even more agents are currently under investigation for the treatment of ALK-rearranged NSCLC. However, the optimal frontline approach and the exact sequence of ALK inhibitors are still under consideration. Recently announced results of phase III trials recognized higher efficacy of alectinib compared to crizotinib in first-line setting, even in patients with CNS involvement. In this review, we will discuss the current knowledge regarding the biology of the ALK-positive NSCLC, the available therapeutic inhibitors and we will focus on the raised issues from their use in clinical practise.
Collapse
Affiliation(s)
- Dimitrios C Ziogas
- Department of Clinical Therapeutics, Alexandra General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Anna Tsiara
- Department of Clinical Therapeutics, Alexandra General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Georgios Tsironis
- Department of Clinical Therapeutics, Alexandra General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Maria Lykka
- Department of Clinical Therapeutics, Alexandra General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Michalis Liontos
- Department of Clinical Therapeutics, Alexandra General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Aristotelis Bamias
- Department of Clinical Therapeutics, Alexandra General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Meletios-Athanasios Dimopoulos
- Department of Clinical Therapeutics, Alexandra General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| |
Collapse
|
227
|
Du X, Shao Y, Qin H, Tai Y, Gao H. ALK-rearrangement in non-small-cell lung cancer (NSCLC). Thorac Cancer 2018; 9:423-430. [PMID: 29488330 PMCID: PMC5879058 DOI: 10.1111/1759-7714.12613] [Citation(s) in RCA: 175] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Revised: 01/28/2018] [Accepted: 01/29/2018] [Indexed: 12/31/2022] Open
Abstract
The ALK gene encodes a transmembrane tyrosine kinase receptor. ALK is physiologically expressed in the nervous system during embryogenesis, but its expression decreases postnatally. ALK first emerged in the field of oncology in 1994 when it was identified to fuse to NPM1 in anaplastic large-cell lymphoma. Since then, ALK has been associated with other types of cancers, including non-small-cell lung cancer (NSCLC). More than 19 different ALK fusion partners have been discovered in NSCLC, including EML4, KIF5B, KLC1, and TPR. Most of these ALK fusions in NSCLC patients respond well to the ALK inhibitor, crizotinib. In this paper, we reviewed fusion partner genes with ALK, detection methods for ALK-rearrangement (ALK-R), and the ALK-tyrosine kinase inhibitor, crizotinib, used in NSCLC patients.
Collapse
Affiliation(s)
- Xue Du
- Department of Pathology, Cancer Center of People's Liberation Army of ChinaAffiliated Hospital of Academy of Military Medical SciencesBeijingChina
| | - Yun Shao
- Department of Pathology, Cancer Center of People's Liberation Army of ChinaAffiliated Hospital of Academy of Military Medical SciencesBeijingChina
| | - Hai‐Feng Qin
- Department of Lung Cancer, Cancer Center of People's Liberation Army of ChinaAffiliated Hospital of Academy of Military Medical SciencesBeijingChina
| | - Yan‐Hong Tai
- Department of Pathology, Cancer Center of People's Liberation Army of ChinaAffiliated Hospital of Academy of Military Medical SciencesBeijingChina
| | - Hong‐Jun Gao
- Department of Lung Cancer, Cancer Center of People's Liberation Army of ChinaAffiliated Hospital of Academy of Military Medical SciencesBeijingChina
| |
Collapse
|
228
|
Wallerstedt SM, Henriksson M. Balancing early access with uncertainties in evidence for drugs authorized by prospective case series - systematic review of reimbursement decisions. Br J Clin Pharmacol 2018; 84:1146-1155. [PMID: 29381234 PMCID: PMC5980547 DOI: 10.1111/bcp.13531] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 01/18/2018] [Accepted: 01/24/2018] [Indexed: 12/13/2022] Open
Abstract
Aims To review clinical and cost‐effectiveness evidence underlying reimbursement decisions relating to drugs whose authorization mainly is based on evidence from prospective case series. Methods A systematic review of all new drugs evaluated in 2011–2016 within a health care profession‐driven resource prioritization process, with a market approval based on prospective case series, and a reimbursement decision by the Swedish Dental and Pharmaceutical Benefits Agency (TLV). Public assessment reports from the European Medicines Agency, published pivotal studies, and TLV, Scottish Medicines Consortium and National Institute of Health and Care Excellence decisions and guidance documents were reviewed. Results Six drug cases were assessed (brentuximab vedotin, bosutinib, ponatinib, idelalisib, vismodegib, ceritinib). The validity of the pivotal studies was hampered by the use of surrogate primary outcomes and the absence of recruitment information. To quantify drug treatment effect sizes, the reimbursement agencies primarily used data from another source in indirect comparisons. TLV granted reimbursement in five cases, compared with five in five cases for Scottish Medicines Consortium and four in five cases for National Institute of Health and Care Excellence. Decision modifiers, contributing to granted reimbursement despite hugely uncertain cost‐effectiveness ratios, were, for example, small population size, occasionally linked to budget impact, severity of disease, end of life and improved life expectancy. Conclusion For drugs whose authorization is based on prospective case series, most applications for reimbursement within public health care are granted. The underlying evidence has limitations over and above the design per se, and decision modifiers are frequently referred to in the value‐based pricing decision making.
Collapse
Affiliation(s)
- Susanna M Wallerstedt
- Department of Pharmacology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Department of Clinical Pharmacology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Martin Henriksson
- Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
229
|
Poetz O, Dieze T, Hammer H, Weiß F, Sommersdorf C, Templin MF, Esdar C, Zimmermann A, Stevanovic S, Bedke J, Stenzl A, Joos TO. Peptide-Based Sandwich Immunoassay for the Quantification of the Membrane Transporter Multidrug Resistance Protein 1. Anal Chem 2018; 90:5788-5794. [DOI: 10.1021/acs.analchem.8b00152] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Affiliation(s)
- Oliver Poetz
- NMI Natural and Medical Sciences Institute at the University of Tuebingen, Markwiesenstrasse 55, 72770 Reutlingen, Germany
- SIGNATOPE GmbH Markwiesenstrasse 55, 72770 Reutlingen, Germany
| | - Theresa Dieze
- NMI Natural and Medical Sciences Institute at the University of Tuebingen, Markwiesenstrasse 55, 72770 Reutlingen, Germany
| | - Helen Hammer
- NMI Natural and Medical Sciences Institute at the University of Tuebingen, Markwiesenstrasse 55, 72770 Reutlingen, Germany
- SIGNATOPE GmbH Markwiesenstrasse 55, 72770 Reutlingen, Germany
| | - Frederik Weiß
- NMI Natural and Medical Sciences Institute at the University of Tuebingen, Markwiesenstrasse 55, 72770 Reutlingen, Germany
- SIGNATOPE GmbH Markwiesenstrasse 55, 72770 Reutlingen, Germany
| | - Cornelia Sommersdorf
- NMI Natural and Medical Sciences Institute at the University of Tuebingen, Markwiesenstrasse 55, 72770 Reutlingen, Germany
- SIGNATOPE GmbH Markwiesenstrasse 55, 72770 Reutlingen, Germany
| | - Markus F. Templin
- NMI Natural and Medical Sciences Institute at the University of Tuebingen, Markwiesenstrasse 55, 72770 Reutlingen, Germany
| | - Christina Esdar
- Merck KGaA, Frankfurter Strasse 250, 64293 Darmstadt, Germany
| | | | - Stefan Stevanovic
- Eberhard Karls University, Department of Immunology, 72076 Tübingen, Germany
| | - Jens Bedke
- Eberhard Karls University, Department of Urology, 72076 Tübingen, Germany
| | - Arnulf Stenzl
- Eberhard Karls University, Department of Urology, 72076 Tübingen, Germany
| | - Thomas O. Joos
- NMI Natural and Medical Sciences Institute at the University of Tuebingen, Markwiesenstrasse 55, 72770 Reutlingen, Germany
- SIGNATOPE GmbH Markwiesenstrasse 55, 72770 Reutlingen, Germany
| |
Collapse
|
230
|
Lazaro T, Brastianos PK. Immunotherapy and targeted therapy in brain metastases: emerging options in precision medicine. CNS Oncol 2018; 6:139-151. [PMID: 28425754 DOI: 10.2217/cns-2016-0038] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Brain metastases (BM) continue to represent an unmet clinical need in oncology. Immunotherapy and targeted therapy hold great promise in the treatment of BM. Emerging data are confirming the activity of these agents in patients with BM. Genomic studies have confirmed that clinically actionable mutations are present in BM and they can be used in clinical studies to link targeted therapies with their genetic targets. Furthermore, as molecular signatures associated with sensitivity and resistance to immunotherapies are developed, we will better be able to select BM patients who will most benefit from these therapies. Understanding the genetic and immune evolution within BM should drive the next generation of immunotherapy and target therapy, as well as increase the accuracy of the selection process for these therapies.
Collapse
Affiliation(s)
- Tyler Lazaro
- Department of Medicine, Division of Hematology & Oncology, Massachusetts General Hospital, Boston, MA 02144, USA.,Department of Neurology, Division of Neuro-Oncology, Massachusetts General Hospital, Boston, MA 02144, USA
| | - Priscilla K Brastianos
- Department of Medicine, Division of Hematology & Oncology, Massachusetts General Hospital, Boston, MA 02144, USA.,Department of Neurology, Division of Neuro-Oncology, Massachusetts General Hospital, Boston, MA 02144, USA
| |
Collapse
|
231
|
Marta GN, da Cunha Colombo Bonadio RR, Martins RE, Zuppani HB, de Castro G. Cystic brain metastases in ALK-rearranged non-small cell lung cancer. Ecancermedicalscience 2018; 12:818. [PMID: 29662531 PMCID: PMC5880229 DOI: 10.3332/ecancer.2018.818] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2018] [Indexed: 11/26/2022] Open
Abstract
The central nervous system (CNS) is a common site of disease progression in patients with non-small-cell lung cancer (NSCLC) with anaplastic lymphoma kinase (ALK)-rearrangement treated with crizotinib. Cystic brain metastases (CBM) have been recently identified as one possible variant of this disease. An illustrative case report is presented along with a literature review performed in order to track relevant papers about CBM in ALK-rearranged NSCLC, including possible pathophysiology, differential diagnosis and treatment options for this condition. Three case reports have been published describing six ALK-rearranged NSCLC patients presenting with CBM, all of which were under treatment with crizotinib by the time of CBM diagnosis. Treatment with CNS-penetrating tyrosine kinase inhibitors (TKIs) resulted in CNS disease control in three of the six cases reported either as single therapy or in combination with radiation therapy (RT). Investigation of differential diagnoses of CBM might be necessary, which include inflammatory and demyelinating disorders, primary brain tumours and infectious diseases, especially neurocysticercosis that might mimic CBM images. Treatment options include RT, CNS-penetrating TKIs and invasive procedures, such as stereotactic drainage. Thus, CBM are associated with ALK-rearranged NSCLC, particularly in patients who use crizotinib and should prompt investigation of differential diagnosis. CNS-penetrating TKIs are effective in the control of solid brain metastases and also seem to be active in CBM as single therapy or in combination with RT.
Collapse
Affiliation(s)
| | | | | | | | - Gilberto de Castro
- Instituto do Câncer do Estado de São Paulo, São Paulo, SP 01246-000, Brazil
| |
Collapse
|
232
|
Jo J, Kim SH, Kim YJ, Lee J, Kim M, Keam B, Kim TM, Kim DW, Heo DS, Chung JH, Jeon YK, Lee JS. Efficacy of Pemetrexed-based Chemotherapy in Comparison to Non-Pemetrexed-based Chemotherapy in Advanced, ALK+ Non-Small Cell Lung Cancer. Yonsei Med J 2018; 59:202-210. [PMID: 29436187 PMCID: PMC5823821 DOI: 10.3349/ymj.2018.59.2.202] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 12/26/2017] [Accepted: 12/31/2017] [Indexed: 01/07/2023] Open
Abstract
PURPOSE Previous retrospective studies suggest that anaplastic lymphoma kinase (ALK) mutation-positive (ALK+) non-small cell lung cancer (NSCLC) patients are sensitive to pemetrexed. To determine its efficacy, we retrospectively evaluated clinical outcomes of pemetrexed-based chemotherapy in patients with ALK+ NSCLC. MATERIALS AND METHODS We identified 126 patients with advanced, ALK+ NSCLC who received first-line cytotoxic chemotherapy. We compared response, progression-free survival (PFS), and overall survival (OS) rates according to chemotherapy regimens. Furthermore, we evaluated intracranial time to tumor progression (TTP) and proportion of ALK+ cells as prognostic factors. RESULTS Forty-eight patients received pemetrexed-based chemotherapy, while 78 received other regimens as first-line treatment. The pemetrexed-based chemotherapy group showed superior overall response (44.7% vs. 14.3%, p<0.001) and disease control (85.1% vs. 62.3%, p=0.008) rates. The pemetrexed-based chemotherapy group also exhibited longer PFS (6.6 months vs. 3.8 months, p<0.001); OS rates were not significantly different. The lack of exposure to second-generation ALK inhibitors and intracranial metastasis on initial diagnosis were independent negative prognostic factors of OS. Intracranial TTP was similar between the treatment groups (32.7 months vs. 35.7 months, p=0.733). Patients who harbored a greater number of ALK+ tumor cells (≥70%) showed prolonged OS on univariate analysis (not reached vs. 44.8 months, p=0.041), but not on multivariate analysis (hazard ratio: 0.19, 95% confidence interval: 0.03-1.42; p=0.106). CONCLUSION Pemetrexed-based regimens may prolong PFS in patients with ALK+ NSCLC as a first-line treatment, but are not associated with prolonged OS. Exposure to second-generation ALK inhibitors may improve OS rates in patients with ALK+ NSCLC.
Collapse
Affiliation(s)
- Jaemin Jo
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Se Hyun Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Yu Jung Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Juhyun Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Miso Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Bhumsuk Keam
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Tae Min Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Dong Wan Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Dae Seog Heo
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Jin Haeng Chung
- Department of Pathology, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Yoon Kyung Jeon
- Department of Pathology, Seoul National University Hospital, Seoul, Korea
| | - Jong Seok Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea.
| |
Collapse
|
233
|
Affiliation(s)
- Martin Früh
- Martin Früh, Cantonal Hospital of St Gallen, St Gallen, Switzerland; and Solange Peters, University Hospital of Lausanne, Lausanne, Switzerland
| | - Solange Peters
- Martin Früh, Cantonal Hospital of St Gallen, St Gallen, Switzerland; and Solange Peters, University Hospital of Lausanne, Lausanne, Switzerland
| |
Collapse
|
234
|
Pellegrino B, Facchinetti F, Bordi P, Silva M, Gnetti L, Tiseo M. Lung Toxicity in Non-Small-Cell Lung Cancer Patients Exposed to ALK Inhibitors: Report of a Peculiar Case and Systematic Review of the Literature. Clin Lung Cancer 2018; 19:e151-e161. [PMID: 29174221 DOI: 10.1016/j.cllc.2017.10.008] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2017] [Revised: 10/10/2017] [Accepted: 10/19/2017] [Indexed: 01/17/2023]
Abstract
Lung toxicity is a potential fatal effect involving non-small-cell lung cancer (NSCLC) patients exposed to tyrosine kinase inhibitors (TKIs). Moving from our experience regarding a patient who developed lung toxicity while receiving 2 different anaplastic lymphoma kinase (ALK)-TKIs, we performed a systematic review to assess the epidemiologic magnitude and the clinical significance of such toxicity in NSCLC patients treated with ALK-TKIs. Studies were identified using MEDLINE and additional sources (European Society for Medical Oncology, American Society of Clinical Oncology, and World Conference on Lung Cancer abstracts) in agreement with Preferred Reporting Items for Systematic Reviews and Meta-Analyses and Cochrane guidelines. Lung toxicity was reported in 105 of 4943 NSCLC patients (2.1%). Crizotinib was responsible for pulmonary adverse events (AEs) in 1.8% of exposed patients (49 of 2706). With the limit of a lower number of treated patients (n = 359), brigatinib resulted as the most frequently involved in lung toxicity (7%; n = 25). Pulmonary AEs during therapy with ceritinib, alectinib, and lorlatinib occurred in 1.1%, 2.6%, and 1.8% of the patients, respectively. Sixty-five percent of cases accounted for Grade 3 or 4 events, with a mortality rate of 9%. Radiological patterns of pneumonia were reported in 25 patients, whereas imaging evocative of interstitial lung disease in 37. Overall, 26 of 105 patients (25%) permanently discontinued treatment because of lung toxicity. Lung toxicity is a rare albeit potentially severe side effect in NSCLC patients receiving ALK-TKIs, apparently more frequent with brigatinib. Its early recognition and treatment are crucial for the best outcome of this subgroup of patients, whose overall prognosis is being improved by the availability of several targeted agents.
Collapse
Affiliation(s)
| | | | - Paola Bordi
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - Mario Silva
- Radiology Unit, University Hospital of Parma, Parma, Italy
| | - Letizia Gnetti
- Pathology Section, University Hospital of Parma, Parma, Italy
| | - Marcello Tiseo
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| |
Collapse
|
235
|
Sharma GG, Mota I, Mologni L, Patrucco E, Gambacorti-Passerini C, Chiarle R. Tumor Resistance against ALK Targeted Therapy-Where It Comes From and Where It Goes. Cancers (Basel) 2018; 10:E62. [PMID: 29495603 PMCID: PMC5876637 DOI: 10.3390/cancers10030062] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 02/25/2018] [Accepted: 02/26/2018] [Indexed: 12/12/2022] Open
Abstract
Anaplastic lymphoma kinase (ALK) is a validated molecular target in several ALK-rearranged malignancies, particularly in non-small-cell lung cancer (NSCLC), which has generated considerable interest and effort in developing ALK tyrosine kinase inhibitors (TKI). Crizotinib was the first ALK inhibitor to receive FDA approval for ALK-positive NSCLC patients treatment. However, the clinical benefit observed in targeting ALK in NSCLC is almost universally limited by the emergence of drug resistance with a median of occurrence of approximately 10 months after the initiation of therapy. Thus, to overcome crizotinib resistance, second/third-generation ALK inhibitors have been developed and received, or are close to receiving, FDA approval. However, even when treated with these new inhibitors tumors became resistant, both in vitro and in clinical settings. The elucidation of the diverse mechanisms through which resistance to ALK TKI emerges, has informed the design of novel therapeutic strategies to improve patients disease outcome. This review summarizes the currently available knowledge regarding ALK physiologic function/structure and neoplastic transforming role, as well as an update on ALK inhibitors and resistance mechanisms along with possible therapeutic strategies that may overcome the development of resistance.
Collapse
Affiliation(s)
- Geeta Geeta Sharma
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza 20900, Italy.
| | - Ines Mota
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin 10124, Italy.
| | - Luca Mologni
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza 20900, Italy.
- Galkem Srl, Monza 20900, Italy.
| | - Enrico Patrucco
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin 10124, Italy.
| | - Carlo Gambacorti-Passerini
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza 20900, Italy.
- Galkem Srl, Monza 20900, Italy.
- Hematology and Clinical Research Unit, San Gerardo Hospital, Monza 20900, Italy.
| | - Roberto Chiarle
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin 10124, Italy.
- Department of Pathology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
236
|
De Carlo E, Del Savio MC, Polesel J, Da Ros V, Berto E, Santarossa S, Chimienti E, Fratino L, Bearz A. Outcomes of ALK positive lung cancer patients treated with crizotinib or second-generation ALK inhibitor: a monoinstitutional experience. Oncotarget 2018; 9:15340-15349. [PMID: 29632648 PMCID: PMC5880608 DOI: 10.18632/oncotarget.24573] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 02/21/2018] [Indexed: 11/30/2022] Open
Abstract
Rearrangement in the anaplastic lymphoma kinase (ALK) gene is one of the oncogenic drivers in non-small cell lung cancer (NSCLC) patients. Several ALK inhibitors (ALKis) have been developed and have demonstrated their efficacy, however the best treatment strategy for ALK positive NSCLC patients has yet to be determined. Our retrospective study has investigated the outcome of 40 ALK-rearranged NSCLC patients treated with two different sequential strategies in our Institute; a “classical group”, treated with crizotinib followed by second or third generation ALKis, and the “experimental group”, treated upfront with a second generation ALK inhibitor. The primary endpoints investigated were Progression-free survival (PFS) and intracranial activity. The analysis has revealed a significant improvement in PFS (p = 0.050) in the experimental group, furthermore none of these patients developed brain metastasis. There was no statistically significant difference in OS, but all patients in the experimental group were still alive after a median follow up of 15 months. Our retrospective analysis suggests that systemic and intracranial efficacy tends to be better in the experimental group; randomized prospective studies could confirm our observations.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Lucia Fratino
- Medical Oncology Department, CRO-IRCCS, Aviano, Italy
| | | |
Collapse
|
237
|
Metro G, Passaro A, Lo Russo G, Bonanno L, Giusti R, Gregorc V, Capelletto E, Martelli O, Cecere FL, Giannarelli D, Luciani A, Bearz A, Tuzi A, Scotti V, Tonini G, Galetta D, Carta A, Soto Parra H, Rebonato A, Morabito A, Chiari R. Ceritinib compassionate use for patients with crizotinib-refractory, anaplastic lymphoma kinase-positive advanced non-small-cell lung cancer. Future Oncol 2018; 14:353-361. [DOI: 10.2217/fon-2017-0441] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: Ceritinib was evaluated within a compassionate use program of Italian patients. Patients & methods: 70 patients with anaplastic lymphoma kinase-positive crizotinib-refractory advanced non-small-cell lung cancer received ceritinib. Results: Overall response was 40.6%, median progression-free survival was 8.2 months and median survival was 15.5 months. Dose reduction due to treatment-related adverse events occurred in 50.8% of patients starting at 750 mg/day. No significantly different progression-free survival was observed between patients who underwent any time dose reduction (n = 38) versus those who remained on the recommended dose of 750 mg/day (n = 32; p = 0.07). Conclusion: The efficacy of ceritinib compassionate use program resembled that of clinical trials. Dose reductions and adjustments did not appear to negatively affect clinical outcome.
Collapse
Affiliation(s)
- Giulio Metro
- Medical Oncology, Santa Maria della Misericordia Hospital, AOU di Perugia, Perugia, Italy
| | - Antonio Passaro
- Division of Thoracic Oncology, European Institute of Oncology, Milano, Italy
| | - Giuseppe Lo Russo
- Division of Thoracic Oncology, Fondazione IRCCS, Istituto Nazionale Tumori, Milano, Italy
| | - Laura Bonanno
- Medical Oncology 2, Istituto Oncologico Veneto IRCCS, Padova (PD), Italy
| | | | - Vanesa Gregorc
- Department of Medical Oncology, IRCCS, San Raffaele Hospital, Milano, Italy
| | - Enrica Capelletto
- Department of Oncology, University of Torino, AOU San Luigi Orbassano, Torino, Italy
| | - Olga Martelli
- Medical Oncology, AO San Giovanni Addolorata, Roma, Italy
| | - Fabiana L Cecere
- Medical Oncology 1, Regina Elena National Cancer Institute, Roma, Italy
| | - Diana Giannarelli
- Medical Oncology 1, Regina Elena National Cancer Institute, Roma, Italy
| | | | | | | | - Vieri Scotti
- Department of Oncology, Division of Radiotherapy, AOU Careggi, Firenze, Italy
| | - Giuseppe Tonini
- Oncologia Medica, Policlinico Universitario Campus Bio-Medico, Roma, Italy
| | - Domenico Galetta
- Division of Thoracic Oncology, IRCCS Oncologico Giovanni Paolo II, Bari, Italy
| | | | | | - Alberto Rebonato
- Department of Diagnostic Imaging, Santa Maria della Misericordia Hospital, University of Perugia, Perugia, Italy
| | - Alessandro Morabito
- Division of Thoracic Oncology, Istituto Nazionale Tumori, Fondazione G. Pascale, IRCCS, Napoli, Italy
| | - Rita Chiari
- Medical Oncology, Santa Maria della Misericordia Hospital, AOU di Perugia, Perugia, Italy
| |
Collapse
|
238
|
Vestergaard HH, Christensen MR, Lassen UN. A systematic review of targeted agents for non-small cell lung cancer. Acta Oncol 2018; 57:176-186. [PMID: 29172833 DOI: 10.1080/0284186x.2017.1404634] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
BACKGROUND advanced-stage non-small cell lung cancer (NSCLC) is characterized by having limited treatment options and thus a poor prognosis. However, new treatment options, in the form of targeted agents (TA), have emerged during recent years. This systematic review aims to provide an overview of the accessible literature in PubMed evaluating TA used on NSCLC patients, and the resulting survival outcomes. METHOD this systematic literature review was conducted by reviewing all relevant literature in PubMed. Six separate searches were performed: Three searches where controlled entry terms were used and three free text searches. Furthermore, other relevant publications were included manually. A total of seventy-two studies met the search criteria and were thus further analyzed and evaluated. RESULTS In the included studies, various TAs and their effect on different molecular targets have been evaluated. Clinical responses vary considerably among the different genetic aberrations. The majority of studies evaluated TA for epidermal growth factor receptor (EGFR) mutations and TA for echinoderm microtubule-associated protein-like 4-anaplastic lymphoma kinase (EML4-ALK) rearrangements. Studies regarding the use of TA for Rat sarcoma (RAS), rapidly accelerated fibrosarcoma (RAF), ROS proto-oncogene 1 (ROS1) rearrangement, Receptor tyrosine-protein kinase erbB-2 (ERBB2), Phosphatidylinositol 3-kinase (PIK3CA)/v-akt murine thymoma viral oncogene homolog; protein kinase B(AKT)/Phosphatase and tensin homolog deleted on chromosome 10(PTEN), The mammalian target of rapamycin (mTOR), and Mesenchymal-epithelial transition factor (MET) were included as well. In general, studies comparing treatment outcomes in EGFR-mutated patients and EML4-ALK (ALK) rearranged patients after use of either TA or standard chemotherapy, present significant better results after TA. CONCLUSIONS This systematic review provides an overview of available literature in PubMed regarding NSCLC and TA. Included studies point toward that TA appears to be a promising therapeutic tool in treating NSCLC patients and use of TA is expected to result in improved treatment outcomes.
Collapse
|
239
|
Jindal V, Gupta S. Expected Paradigm Shift in Brain Metastases Therapy-Immune Checkpoint Inhibitors. Mol Neurobiol 2018; 55:7072-7078. [PMID: 29383686 DOI: 10.1007/s12035-018-0905-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 01/11/2018] [Indexed: 01/02/2023]
Abstract
Brain metastasis (BM) is one of the dreadful complications of malignancies. The prognosis after BM is extremely poor and life expectancy is meager. Currently, our treatment modalities are limited to radiotherapy and surgical resection, which also has poor outcomes and leads to various neurological deficits and affects the quality of life of patients. New treatment modality, i.e., immune checkpoint inhibitors, has brought revolution in management of melanoma, renal cancer, and non-small cell lung cancer (NSCLC). Immune checkpoint inhibitors basically enhance the immune response of the body to fight against cancers. Immune response in the brain is highly regulated; therefore, it is challenging to use immune-modulator drugs in BM. The microenvironment of BM is rich in cytotoxic T lymphocytes and which is the target of immune checkpoint inhibitors. Few studies have shown some hope regarding use of immune checkpoint inhibitors in management of BM. It works through inhibiting immune check point gates, i.e., CTLA-4 (cytotoxic T-lymphocyte-associated protein) and PD-1/PD-L1 (programmed cell death protein-1/program death ligand-1). This article explains the basic mechanism of immune check point inhibitors, rationale behind their usage in BM, and some of the clinical studies which have shown the efficacy of immune check point inhibitors in BM.
Collapse
Affiliation(s)
- Vishal Jindal
- St. Vincent Hospital, 123 Summer Street, Worcester, 01608, USA.
| | - Sorab Gupta
- Einstein Medical Center, 5501 Old York Rd, Philadelphia, 19141, USA
| |
Collapse
|
240
|
Makita Y, Teratani M, Murata S, Hoashi Y, Matsumoto S, Kawamata Y. Antitumor activity of kinetochore-associated protein 2 siRNA against lung cancer patient-derived tumor xenografts. Oncol Lett 2018. [PMID: 29541240 DOI: 10.3892/ol.2018.7890] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
It has been widely reported that patient-derived tumor xenografts (PDXs) are more similar to tumor tissues than conventional cancer cell lines. Kinetochore-associated protein 2 (KNTC2) is known to be upregulated specifically in tumor tissues of cancer patients and is recognized as a potential target for cancer therapy. Previously, in vivo antitumor activities of KNTC2 short interfering RNA encapsulated into a lipid nanoparticle (KNTC2-LNP) were reported in orthotopic hepatocellular carcinoma mouse models. However, it remains unclear whether KNTC2-LNP exhibits antitumor activities against lung cancer PDXs. In the present study, the antitumor activities of KNTC2-LNP were clarified in a three-dimensional culture system and a subcutaneous tumor model of lung cancer PDX, LC-60, which was resistant to erlotinib. Growth inhibitory activities of KNTC2-LNP were associated with knockdown activities. Furthermore, KNTC2-LNP also exhibited in vivo antitumor activity against another lung cancer PDX, LC-45, which was sensitive to erlotinib. These results suggest that KNTC2 is a promising target for patients with lung cancer.
Collapse
Affiliation(s)
- Yukimasa Makita
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa 251-8555, Japan
| | - Mika Teratani
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa 251-8555, Japan
| | - Shumpei Murata
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa 251-8555, Japan
| | - Yasutaka Hoashi
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa 251-8555, Japan
| | - Satoru Matsumoto
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa 251-8555, Japan
| | - Yuji Kawamata
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa 251-8555, Japan
| |
Collapse
|
241
|
Abstract
Drug resistance inevitably limits the efficacy of all targeted therapies including tyrosine kinase inhibitors (TKIs). Understanding the biological underpinnings of TKI resistance is key to the successful development of future therapeutic strategies. Traditionally, mechanisms of TKI resistance have been viewed under a dichotomous lens. Tumor cells are TKI-sensitive or TKI-refractory, exhibit intrinsic or acquired resistance, and accumulate alterations within or outside the target to promote their survival. Such classifications facilitate our comprehension of an otherwise complex biology, but are likely an oversimplification. Recent studies underscore the multifaceted, genetically heterogeneous nature of TKI resistance, which evolves dynamically with changes in therapy. In this Review, we provide a broad framework for understanding the diverse mechanisms of resistance at play in oncogene-driven lung cancers.
Collapse
Affiliation(s)
- Jessica J Lin
- Department of Thoracic Oncology, Massachusetts General Hospital Cancer Center, 32 Fruit Street, Boston, MA 02114, USA
| | - Alice T Shaw
- Department of Thoracic Oncology, Massachusetts General Hospital Cancer Center, 32 Fruit Street, Boston, MA 02114, USA
| |
Collapse
|
242
|
Dagogo-Jack I, Brannon AR, Ferris LA, Campbell CD, Lin JJ, Schultz KR, Ackil J, Stevens S, Dardaei L, Yoda S, Hubbeling H, Digumarthy SR, Riester M, Hata AN, Sequist LV, Lennes IT, Iafrate AJ, Heist RS, Azzoli CG, Farago AF, Engelman JA, Lennerz JK, Benes CH, Leary RJ, Shaw AT, Gainor JF. Tracking the Evolution of Resistance to ALK Tyrosine Kinase Inhibitors through Longitudinal Analysis of Circulating Tumor DNA. JCO Precis Oncol 2018; 2018. [PMID: 29376144 DOI: 10.1200/po.17.00160] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Purpose ALK rearrangements predict for sensitivity to ALK tyrosine kinase inhibitors (TKIs). However, responses to ALK TKIs are generally short-lived. Serial molecular analysis is an informative strategy for identifying genetic mediators of resistance. Although multiple studies support the clinical benefits of repeat tissue sampling, the clinical utility of longitudinal circulating tumor DNA analysis has not been established in ALK-positive lung cancer. Methods Using a 566-gene hybrid-capture next-generation sequencing (NGS) assay, we performed longitudinal analysis of plasma specimens from 22 ALK-positive patients with acquired resistance to ALK TKIs to track the evolution of resistance during treatment. To determine tissue-plasma concordance, we compared plasma findings to results of repeat biopsies. Results At progression, we detected an ALK fusion in plasma from 19 (86%) of 22 patients, and identified ALK resistance mutations in plasma specimens from 11 (50%) patients. There was 100% agreement between tissue- and plasma-detected ALK fusions. Among 16 cases where contemporaneous plasma and tissue specimens were available, we observed 100% concordance between ALK mutation calls. ALK mutations emerged and disappeared during treatment with sequential ALK TKIs, suggesting that plasma mutation profiles were dependent on the specific TKI administered. ALK G1202R, the most frequent plasma mutation detected after progression on a second-generation TKI, was consistently suppressed during treatment with lorlatinib. Conclusions Plasma genotyping by NGS is an effective method for detecting ALK fusions and ALK mutations in patients progressing on ALK TKIs. The correlation between plasma ALK mutations and response to distinct ALK TKIs highlights the potential for plasma analysis to guide selection of ALK-directed therapies.
Collapse
Affiliation(s)
| | - A Rose Brannon
- Novartis Institutes of BioMedical Research, Cambridge, MA
| | - Lorin A Ferris
- Department of Medicine, Massachusetts General Hospital, Boston, MA
| | | | - Jessica J Lin
- Department of Medicine, Massachusetts General Hospital, Boston, MA
| | | | - Jennifer Ackil
- Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Sara Stevens
- Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Leila Dardaei
- Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Satoshi Yoda
- Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Harper Hubbeling
- Department of Medicine, Massachusetts General Hospital, Boston, MA
| | | | - Markus Riester
- Novartis Institutes of BioMedical Research, Cambridge, MA
| | - Aaron N Hata
- Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Lecia V Sequist
- Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Inga T Lennes
- Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - A John Iafrate
- Department of Pathology, Massachusetts General Hospital, Boston, MA
| | - Rebecca S Heist
- Department of Medicine, Massachusetts General Hospital, Boston, MA
| | | | - Anna F Farago
- Department of Medicine, Massachusetts General Hospital, Boston, MA
| | | | - Jochen K Lennerz
- Department of Pathology, Massachusetts General Hospital, Boston, MA
| | - Cyril H Benes
- Department of Medicine, Massachusetts General Hospital, Boston, MA
| | | | - Alice T Shaw
- Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Justin F Gainor
- Department of Medicine, Massachusetts General Hospital, Boston, MA
| |
Collapse
|
243
|
Cadranel J, Cortot AB, Lena H, Mennecier B, Do P, Dansin E, Mazieres J, Chouaid C, Perol M, Barlesi F, Robinet G, Friard S, Thiberville L, Audigier-Valette C, Vergnenegre A, Westeel V, Slimane K, Buturuga A, Moro-Sibilot D, Besse B. Real-life experience of ceritinib in crizotinib-pretreated ALK+ advanced non-small cell lung cancer patients. ERJ Open Res 2018; 4:00058-2017. [PMID: 29450203 PMCID: PMC5810621 DOI: 10.1183/23120541.00058-2017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 11/21/2017] [Indexed: 12/13/2022] Open
Abstract
Here we report our experience of ceritinib in crizotinib-pretreated patients with anaplastic lymphoma kinase (ALK) positive (ALK+) non-small cell lung cancer (NSCLC) in a French temporary authorisation for use (TAU) study. The French TAU study included crizotinib-pretreated patients with advanced ALK+ or ROS proto-oncogene 1 positive (ROS1+) tumours. Patients received oral ceritinib (750 mg·day-1 as a starting dose) and best tumour response (as evaluated by the investigator) and safety were reported every 3 months. A total of 242 TAUs were granted from March 12, 2013 to August 05, 2015. Of the 242 patients, 228 had ALK+ NSCLC and 13 had ROS1+ NSCLC. The median age of ALK+ patients (n=214) was 58.5 years, 51.9% were female, 70.8% had an Eastern Cooperative Oncology Group (ECOG) performance status (PS) of 0-1 and 50.0% had brain metastases. Of the 149 efficacy evaluable ALK+ NSCLC patients, 5.4% had a complete response (CR), 47.0% had a partial response (PR) and 22.8% had stable disease (SD). At September 05, 2015, the median duration of ceritinib treatment (n=182) was 3.9 months but 5.5 months for patients (n=71) with a follow-up of ≥12 months. Higher objective response rate (ORR) was observed for patients with ECOG PS 0 to 1 (55.0% versus 42.4%) and those receiving prior crizotinib for >5 months (51.6% versus 36.1%). Treatment-related adverse events (AEs) were reported in 118 of 208 patients (56.7%), the most common being diarrhoea (22.1%) and hepatic toxicity (19.7%). Ceritinib (750 mg·day-1) demonstrated efficacy similar efficacy to ASCEND-1, ASCEND-2 and phase 3 ASCEND-5 trials with manageable safety in crizotinib-pretreated patients with ALK+ NSCLC.
Collapse
Affiliation(s)
- Jacques Cadranel
- Service de Pneumologie, Hôpital Tenon, Assistance Publique-Hôpitaux de Paris (AP-HP) and Université Pierre-et-Marie-Curie (Paris 6), Paris, France
| | - Alexis B. Cortot
- Thoracic Oncology, Centre Hospitalier Universitaire (CHU) and Université de Lille, Lille, France
| | - Hervé Lena
- Pneumologie, CHU and INSERM ERL440-OSS, Rennes, France
| | | | - Pascal Do
- Pathologies Thoraciques, Centre François Baclesse, Caen, France
| | - Eric Dansin
- Département de cancérologie cervico-faciale et thoracique, CLCC Oscar Lambret, Lille, France
| | | | - Christos Chouaid
- Service de Pneumologie, Centre Hospitalier Intercommunal Cretéil (CHIC), Créteil, France
| | - Maurice Perol
- Cancérologie poumon, tumeurs thoraciques, Centre Léon Bérard, Lyon, France
| | - Fabrice Barlesi
- Multidisciplinary Oncology & Therapeutic Innovations Dept, Assistance Publique-Hôpitaux de Marseille (AP-HM) and Université Aix-Marseille, Marseille, France
| | | | | | | | | | | | | | | | | | | | - Benjamin Besse
- Institut d'Oncologie Thoracique, Institut Gustave Roussy, Villejuif, France
- Université Paris-Sud, Orsay, France
| |
Collapse
|
244
|
Peters S, Zimmermann S. Lung Cancer. SIDE EFFECTS OF MEDICAL CANCER THERAPY 2018:85-103. [DOI: 10.1007/978-3-319-70253-7_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
245
|
Second-Line Treatment Options in Non-Small-Cell Lung Cancer: Report From an International Experts Panel Meeting of the Italian Association of Thoracic Oncology. Clin Lung Cancer 2017; 19:301-314. [PMID: 29396237 DOI: 10.1016/j.cllc.2017.12.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 12/05/2017] [Accepted: 12/17/2017] [Indexed: 11/24/2022]
Abstract
Non-small-cell lung cancer (NSCLC) patients inevitably progress to first-line therapy and further active treatments are warranted. In the past few years, new second-line therapies, beyond chemotherapy agents, have become available in clinical practice. To date, several options for the second-line treatment of non-oncogene-addicted NSCLC patients ranging from chemotherapy in combination with antivascular endothelial growth factor receptor to immunotherapeutics are available. In oncogene-driven tumors, the better knowledge of mechanisms of acquired resistance to earlier tyrosine kinase inhibitors is leading to novel active inhibitors now available/in development. The second-line algorithm treatment of NSCLC becomes very intricate and the selection of proper patients with one of the new available therapeutic options is of paramount importance to personalize and optimize the treatment. In this review we discuss the second-line treatment opportunities of addicted as well as not-addicted NSCLC.
Collapse
|
246
|
Sun YW, Xu J, Zhou J, Liu WJ. Targeted drugs for systemic therapy of lung cancer with brain metastases. Oncotarget 2017; 9:5459-5472. [PMID: 29435193 PMCID: PMC5797064 DOI: 10.18632/oncotarget.23616] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 10/28/2017] [Indexed: 02/07/2023] Open
Abstract
Brain metastases are very common in lung cancer patients. The condition of these patients is complicated and difficult to treat, and adverse reactions following treatment can affect the nervous system, which severely reduces quality of life. Lung cancers are categorized as small cell lung cancers and non-small cell lung cancers. Patients with brain metastasis of small cell lung cancers are generally treated with brain radiotherapy and systemic chemotherapy, but stage III/IV patients with brain metastasis of non-small cell lung cancers are generally not responsive to radiotherapy or chemotherapy. With the recent development of targeted drugs, tumor molecular profile detection allows the selection of appropriate targeted drugs for adjuvant pharmacological treatment of brain metastasis in lung cancer patients. In recent years, immune checkpoint inhibitors have emerged and have been approved by the Food and Drug Administration (FDA) for the treatment of certain cancers, but their efficacy in lung cancer patients with brain metastases still needs to be confirmed. This paper focuses on highlighting drugs for targeted therapy of brain metastasis in lung cancer patients and their molecular targets and mechanisms of drug resistance.
Collapse
Affiliation(s)
- Ya-Wen Sun
- Shandong Cancer Hospital and Institute, Shandong Cancer Hospital Affiliated to Shandong University, Jinan, China
| | - Jian Xu
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA.,Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jun Zhou
- University of South Carolina, Computer Science and Engineering Department, Columbia, SC, USA
| | - Wen-Juan Liu
- Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
247
|
Liu B, Yuan M, Sun Y, Cheng Z, Zhang Z, Hou S, Wang X, Liu J. Incidence and risk of hepatic toxicities associated with anaplastic lymphoma kinase inhibitors in the treatment of non-small-cell lung cancer: a systematic review and meta-analysis. Oncotarget 2017; 9:9480-9488. [PMID: 29507704 PMCID: PMC5823621 DOI: 10.18632/oncotarget.23840] [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: 05/27/2017] [Accepted: 07/26/2017] [Indexed: 11/25/2022] Open
Abstract
Background Two anaplastic lymphoma kinase (ALK)-tyrosine kinase inhibitors (-TKIs) have been approved for the treatment of patients with ALK-rearranged (ALK-positive) advanced non-small cell lung cancer (NSCLC). Severe hepatotoxicity has been observed in several clinical studies. We aim to assess the incidence and risk of liver toxicity with these drugs by a systematic review and meta-analysis of clinical trials. Materials and Methods The databases of PubMed, Web of Science and abstracts presented at oncology conferences’ proceedings were searched for relevant studies from January 2000 to January 2017. Summary incidence rates, relative risks (RRs), and 95% confidence intervals (CIs) were calculated by using either random effects or fixed effect models. Results A total of 1,908 patients from 10 clinical trials were included. The incidences of all-grade aspartate aminotransferase (AST) and alanine transaminase (ALT) elevation were 25.2% (95% CI 17.7–34.7%), and 26.0% (95% CI 17.8–36.3%), respectively. The incidences of high-grade (grade 3 and 4) AST and ALT elevation were 7.0% (95% CI: 5.4–9.0%), and 9.9% (95%CI: 5.6–16.7%), respectively. Sub-group analysis according to ALK-TKIs showed that the incidence of liver toxicities associated with ceritinib was higher than that of crizotinib and alectinib. In comparison with chemotherapy, ALK-TKIs significantly increased the risk of developing all-grade and high-grade AST elevation (RR, 2.30, 95%CI: 1.87–2.83, p < 0.001; RR 10.14, 95% CI: 3.9–26.39, p < 0.001) and ALT elevation (RR 2.37, 95%CI: 1.97–2.86, p < 0.001; RR 7.34, 95% CI: 3.95–13.63, p < 0.001), respectively. Conclusions The use of ALK-TKIs significantly increases the risk of developing all-grade and high-grade liver toxicities in lung cancer patients.
Collapse
Affiliation(s)
- Bing Liu
- Department of Thoracic Surgery, Linyi Central Hospital, Yishui 276400, Shandong Province, China
| | - Maoxi Yuan
- Department of Thoracic Surgery, Linyi Central Hospital, Yishui 276400, Shandong Province, China
| | - Yi Sun
- Department of Thoracic Surgery, Linyi Central Hospital, Yishui 276400, Shandong Province, China
| | - Ziming Cheng
- Department of Thoracic Surgery, Linyi Central Hospital, Yishui 276400, Shandong Province, China
| | - Zaiyong Zhang
- Department of Thoracic Surgery, Linyi Central Hospital, Yishui 276400, Shandong Province, China
| | - Shizheng Hou
- Department of Thoracic Surgery, Linyi Central Hospital, Yishui 276400, Shandong Province, China
| | - Xiangdong Wang
- Department of Thoracic Surgery, Linyi Central Hospital, Yishui 276400, Shandong Province, China
| | - Jingfeng Liu
- Department of Thoracic Surgery, Linyi Central Hospital, Yishui 276400, Shandong Province, China
| |
Collapse
|
248
|
Le X, Rangachari D, Costa DB. Moving more potent and less toxic options to the frontline in the management of advanced lung cancer. J Thorac Dis 2017; 9:2812-2818. [PMID: 29221246 DOI: 10.21037/jtd.2017.08.79] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Xiuning Le
- Division of Hematology/Oncology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, US
| | - Deepa Rangachari
- Division of Hematology/Oncology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, US
| | - Daniel B Costa
- Division of Hematology/Oncology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, US
| |
Collapse
|
249
|
Abstract
Lung cancer is the number one cause of cancer-related death in both men and women. However, over the last few years, we have witnessed improved outcomes that are largely attributable to early detection, increased efforts in tobacco control, improved surgical approaches, and the development of novel targeted therapies. Currently, there are several novel therapies in clinical practice, including those targeting actionable mutations and more recently immunotherapeutic agents. Immunotherapy represents the most significant step forward in eradicating this deadly disease. Given the ever-changing landscape of lung cancer management, here we present an overview of the most recent advances in the management of non-small cell lung cancer.
Collapse
Affiliation(s)
- Samira Shojaee
- Division of Pulmonary and Critical Care Medicine, Virginia Commonwealth University, Richmond, USA
| | - Patrick Nana-Sinkam
- Division of Pulmonary and Critical Care Medicine, Virginia Commonwealth University, Richmond, USA
| |
Collapse
|
250
|
Sgambato A, Casaluce F, Maione P, Gridelli C. Targeted therapies in non-small cell lung cancer: a focus on ALK/ROS1 tyrosine kinase inhibitors. Expert Rev Anticancer Ther 2017; 18:71-80. [PMID: 29187012 DOI: 10.1080/14737140.2018.1412260] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
INTRODUCTION Anaplastic lymphoma kinase (ALK) and ROS1 rearrangements define important molecular subgroups of advanced non-small cell lung cancer (NSCLC). The identification of these genetic driver alterations created new potential for highly active therapeutic interventions. After discovery of ALK rearrangements in NSCLC, it was recognized that these confer sensitivity to ALK inhibition. Areas covered: Crizotinib, the first-in-class ALK/ROS1/MET inhibitor, was initially approved as second-line treatment of ALK-positive advanced NSCLC but after this, it was firmly established as the standard first-line therapy for advanced ALK-positive NSCLC. After initial response to crizotinib, tumors inevitably relapse. Next-generation ALK inhibitors, more potent and brain-penetrable than crizotinib, may be effective in re-inducing remissions when cancers are still addicted to ALK. Ceritinib and alectinib are approved for metastatic ALK positive NSCLC patients, while brigatinib received granted accelerated approval by the United States Food and Drug Administration. Regarding ROS1 rearrangement, to date crizotinib is the only ALK-tyrosine kinase inhibitor receiving indication as treatment of ROS1 positive advanced NSCLC. Expert commentary: Although novel ALK-inhibitors are under clinical investigation compared to crizotinib as front-line treatment for ALK-positive NSCLC, nowadays the current standard first-line therapy for these patients is crizotinib. Further research will clarify the best management of ALK-positive NSCLC, above all who progress on first-line crizotinib.
Collapse
Affiliation(s)
- Assunta Sgambato
- a Division of Medical Oncology , 'S. G. Moscati' Hospital , Avellino , Italy
| | - Francesca Casaluce
- a Division of Medical Oncology , 'S. G. Moscati' Hospital , Avellino , Italy
| | - Paolo Maione
- a Division of Medical Oncology , 'S. G. Moscati' Hospital , Avellino , Italy
| | - Cesare Gridelli
- a Division of Medical Oncology , 'S. G. Moscati' Hospital , Avellino , Italy
| |
Collapse
|