201
|
Rossi G, Jocollé G, Conti A, Tiseo M, Zito Marino F, Donati G, Franco R, Bono F, Barbisan F, Facchinetti F. Detection of ROS1 rearrangement in non-small cell lung cancer: current and future perspectives. LUNG CANCER (AUCKLAND, N.Z.) 2017; 8:45-55. [PMID: 28740441 PMCID: PMC5508815 DOI: 10.2147/lctt.s120172] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
ROS1 rearrangement characterizes a small subset (1%-2%) of non-small cell lung cancer and is associated with slight/never smoking patients and adenocarcinoma histology. Identification of ROS1 rearrangement is mandatory to permit targeted therapy with specific inhibitors, demonstrating a significantly better survival when compared with conventional chemotherapy. Detection of ROS1 rearrangement is based on in situ (immunohistochemistry, fluorescence in situ hybridization) and extractive non-in situ assays. While fluorescence in situ hybridization still represents the gold standard in clinical trials, this technique may fail to recognize rearrangements of ROS1 with some gene fusion partner. On the other hand, immunohistochemistry is the most cost-effective screening technique, but it seems to be characterized by low specificity. Extractive molecular assays are expensive and laborious methods, but they specifically recognize almost all ROS1 fusions using a limited amount of mRNA even from formalin-fixed, paraffin-embedded tumor tissues. This review is a discussion on the present and futuristic diagnostic scenario of ROS1 identification in lung cancer.
Collapse
Affiliation(s)
| | - Genny Jocollé
- Oncology Unit, Azienda USL Valle d’Aosta, Regional Hospital “Parini”, Aosta
| | | | - Marcello Tiseo
- Medical Oncology Unit, University Hospital of Parma, Parma
| | - Federica Zito Marino
- Pathology Unit, Istituto Nazionale Tumori Fondazione G. Pascale
- Pathology Unit, Luigi Vanvitelli University of Campania, Naples
| | - Giovanni Donati
- Unit of Thoracic and Senology Surgery, Azienda USL Valle d’Aosta, Regional Hospital “Parini”, Aosta
| | - Renato Franco
- Pathology Unit, Istituto Nazionale Tumori Fondazione G. Pascale
- Pathology Unit, Luigi Vanvitelli University of Campania, Naples
| | - Francesca Bono
- Unit of Pathologic Anatomy, San Gerardo Hospital, IRCCS, Monza
| | - Francesca Barbisan
- Pathology Unit, University Hospital, Azienda Ospedali Riuniti, Ancona, Italy
| | - Francesco Facchinetti
- Medical Oncology Unit, University Hospital of Parma, Parma
- INSERM, U981, Gustave Roussy Cancer Campus, Villejuif, France
| |
Collapse
|
202
|
First macrocyclic 3 rd -generation ALK inhibitor for treatment of ALK/ROS1 cancer: Clinical and designing strategy update of lorlatinib. Eur J Med Chem 2017; 134:348-356. [DOI: 10.1016/j.ejmech.2017.04.032] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 03/26/2017] [Accepted: 04/12/2017] [Indexed: 11/21/2022]
|
203
|
Woo JS, Reddy OL, Koo M, Xiong Y, Li F, Xu H. Application of Immunohistochemistry in the Diagnosis of Pulmonary and Pleural Neoplasms. Arch Pathol Lab Med 2017. [PMID: 28644685 DOI: 10.5858/arpa.2016-0550-ra] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
CONTEXT - A vast majority of neoplasms arising from lung or pleura are initially diagnosed based on the histologic evaluation of small transbronchial, endobronchial, or needle core biopsies. Although most diagnoses can be determined by morphology alone, immunohistochemistry can be a valuable diagnostic tool in the workup of problematic cases. OBJECTIVE - To provide a practical approach in the interpretation and immunohistochemical selection of lung/pleura-based neoplasms obtained from small biopsy samples. DATA SOURCES - A literature review of previously published articles and the personal experience of the authors were used in this review article. CONCLUSION - Immunohistochemistry is a useful diagnostic tool in the workup of small biopsies from the lung and pleura sampled by small biopsy techniques.
Collapse
|
204
|
Dolly SO, Collins DC, Sundar R, Popat S, Yap TA. Advances in the Development of Molecularly Targeted Agents in Non-Small-Cell Lung Cancer. Drugs 2017; 77:813-827. [PMID: 28378229 DOI: 10.1007/s40265-017-0732-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Non-small-cell lung cancer (NSCLC) remains a significant global health challenge and the leading cause of cancer-related mortality. The traditional 'one-size-fits-all' treatment approach has now evolved into one that involves personalized strategies based on histological and molecular subtypes. The molecular era has revolutionized the treatment of patients harboring epidermal growth factor receptor (EGFR), anaplastic lymphoma kinase (ALK) and ROS1 gene aberrations. In the appropriately selected population, anti-tumor agents against these molecular targets can significantly improve progression-free survival. However, the emergence of acquired resistance is inevitable. Novel potent compounds with much improved and rational selectivity profiles, such as third-generation EGFR T790M resistance mutation-specific inhibitors, have been developed and added to the NSCLC armamentarium. To date, attempts to overcome resistance bypass pathways through downstream signaling blockade has had limited success. Furthermore, the majority of patients still do not harbor known driver genetic or epigenetic alterations and/or have no new available treatment options, with chemotherapy remaining their standard of care. Several potentially actionable driver aberrations have recently been identified, with the early clinical development of multiple inhibitors against these promising targets currently in progress. The advent of immune checkpoint inhibitors has led to significant benefit for advanced NSCLC patients with durable responses observed. Further interrogation of the underlying biology of NSCLC, coupled with modern clinical trial designs, is now required to develop novel targeted therapeutics rationally matched with predictive biomarkers of response, so as to further advance NSCLC therapeutics through the next decade.
Collapse
Affiliation(s)
| | | | - Raghav Sundar
- Royal Marsden NHS Foundation Trust, London, UK.,National University Health System, Singapore, Singapore
| | - Sanjay Popat
- Royal Marsden NHS Foundation Trust, London, UK.,National Heart and Lung Institute, Imperial College London, London, UK
| | - Timothy A Yap
- Royal Marsden NHS Foundation Trust, London, UK. .,Drug Development Unit and Lung Cancer Unit, The Institute of Cancer Research and Royal Marsden Hospital, Downs Road, London, SM2 5PT, UK.
| |
Collapse
|
205
|
Lim SM, Kim HR, Lee JS, Lee KH, Lee YG, Min YJ, Cho EK, Lee SS, Kim BS, Choi MY, Shim HS, Chung JH, La Choi Y, Lee MJ, Kim M, Kim JH, Ali SM, Ahn MJ, Cho BC. Open-Label, Multicenter, Phase II Study of Ceritinib in Patients With Non-Small-Cell Lung Cancer Harboring ROS1 Rearrangement. J Clin Oncol 2017; 35:2613-2618. [PMID: 28520527 DOI: 10.1200/jco.2016.71.3701] [Citation(s) in RCA: 236] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Purpose ROS1 rearrangement is a distinct molecular subset of non-small-cell lung cancer (NSCLC). We investigated the efficacy and safety of ceritinib in patients with ROS1-rearranged NSCLC. Patients and Methods We enrolled 32 patients with advanced NSCLC who tested positive for ROS1 rearrangement by fluorescent in situ hybridization. Ceritinib 750 mg was administered once daily. The primary end point was objective response rate. The secondary end points were disease control rate; duration of response; progression-free survival; overall survival; toxicity; and concordance among fluorescent in situ hybridization, immunohistochemistry, and next-generation sequencing. Results Between June 7, 2013, and February 1, 2016, 404 patients underwent ROS1 prescreening, and 32 patients with ROS1 rearrangement were enrolled. All patients except two were crizotinib-naïve. At the time of data cutoff, the median follow-up was 14.0 months, and 18 patients (56%) had discontinued treatment. Of the 32 patients enrolled, 28 were evaluable for response by independent radiologic review. Objective response rate was 62% (95% CI, 45% to 77%), with one complete response and 19 partial responses; duration of response was 21.0 months (95% CI, 17 to 25 months); and disease control rate was 81% (95% CI, 65% to 91%). The median progression-free survival was 9.3 months (95% CI, 0 to 22 months) for all patients and 19.3 months (95% CI, 1 to 37 months) for crizotinib-naïve patients. The median overall survival was 24 months (95% CI, 5 to 43 months). Of the eight patients with brain metastases, intracranial disease control was reported in five (63%; 95% CI, 31% to 86%). The most common adverse events (majority, grade 1 or 2) for all treated patients were diarrhea (78%), nausea (59%), and anorexia (56%). Conclusion Ceritinib demonstrated potent clinical activity in patients with ROS1-rearranged NSCLC who were heavily treated previously with multiple lines of chemotherapy.
Collapse
Affiliation(s)
- Sun Min Lim
- Sun Min Lim, Hye Ryun Kim, Hyo Sup Shim, Joo-Hang Kim, and Byoung Chul Cho, Yonsei University College of Medicine; Yun-Gyoo Lee, Yoon La Choi, and Myung-Ju Ahn, Sungkyunkwan University School of Medicine; Bong-Seog Kim, VHS Medical Center; Min Jeong Lee and Maria Kim, Yonsei University Health System, Seoul; Jong-Seok Lee and Jin-Haeng Chung, Seoul National University Bundang Hospital, Bundang; Ki Hyeong Lee, Chungbuk National University, Cheongju; Young Joo Min, University of Ulsan College of Medicine, Ulsan; Eun Kyung Cho, Gachon Medical School, Inchon; Sung Sook Lee, Inje University College of Medicine; Moon Young Choi, Inje University, Busan; Jin-Haeng Chung, Seoul National University College of Medicine, Seongnam, Republic of Korea; and Siraj M. Ali, Foundation Medicine, Cambridge, MA
| | - Hye Ryun Kim
- Sun Min Lim, Hye Ryun Kim, Hyo Sup Shim, Joo-Hang Kim, and Byoung Chul Cho, Yonsei University College of Medicine; Yun-Gyoo Lee, Yoon La Choi, and Myung-Ju Ahn, Sungkyunkwan University School of Medicine; Bong-Seog Kim, VHS Medical Center; Min Jeong Lee and Maria Kim, Yonsei University Health System, Seoul; Jong-Seok Lee and Jin-Haeng Chung, Seoul National University Bundang Hospital, Bundang; Ki Hyeong Lee, Chungbuk National University, Cheongju; Young Joo Min, University of Ulsan College of Medicine, Ulsan; Eun Kyung Cho, Gachon Medical School, Inchon; Sung Sook Lee, Inje University College of Medicine; Moon Young Choi, Inje University, Busan; Jin-Haeng Chung, Seoul National University College of Medicine, Seongnam, Republic of Korea; and Siraj M. Ali, Foundation Medicine, Cambridge, MA
| | - Jong-Seok Lee
- Sun Min Lim, Hye Ryun Kim, Hyo Sup Shim, Joo-Hang Kim, and Byoung Chul Cho, Yonsei University College of Medicine; Yun-Gyoo Lee, Yoon La Choi, and Myung-Ju Ahn, Sungkyunkwan University School of Medicine; Bong-Seog Kim, VHS Medical Center; Min Jeong Lee and Maria Kim, Yonsei University Health System, Seoul; Jong-Seok Lee and Jin-Haeng Chung, Seoul National University Bundang Hospital, Bundang; Ki Hyeong Lee, Chungbuk National University, Cheongju; Young Joo Min, University of Ulsan College of Medicine, Ulsan; Eun Kyung Cho, Gachon Medical School, Inchon; Sung Sook Lee, Inje University College of Medicine; Moon Young Choi, Inje University, Busan; Jin-Haeng Chung, Seoul National University College of Medicine, Seongnam, Republic of Korea; and Siraj M. Ali, Foundation Medicine, Cambridge, MA
| | - Ki Hyeong Lee
- Sun Min Lim, Hye Ryun Kim, Hyo Sup Shim, Joo-Hang Kim, and Byoung Chul Cho, Yonsei University College of Medicine; Yun-Gyoo Lee, Yoon La Choi, and Myung-Ju Ahn, Sungkyunkwan University School of Medicine; Bong-Seog Kim, VHS Medical Center; Min Jeong Lee and Maria Kim, Yonsei University Health System, Seoul; Jong-Seok Lee and Jin-Haeng Chung, Seoul National University Bundang Hospital, Bundang; Ki Hyeong Lee, Chungbuk National University, Cheongju; Young Joo Min, University of Ulsan College of Medicine, Ulsan; Eun Kyung Cho, Gachon Medical School, Inchon; Sung Sook Lee, Inje University College of Medicine; Moon Young Choi, Inje University, Busan; Jin-Haeng Chung, Seoul National University College of Medicine, Seongnam, Republic of Korea; and Siraj M. Ali, Foundation Medicine, Cambridge, MA
| | - Yun-Gyoo Lee
- Sun Min Lim, Hye Ryun Kim, Hyo Sup Shim, Joo-Hang Kim, and Byoung Chul Cho, Yonsei University College of Medicine; Yun-Gyoo Lee, Yoon La Choi, and Myung-Ju Ahn, Sungkyunkwan University School of Medicine; Bong-Seog Kim, VHS Medical Center; Min Jeong Lee and Maria Kim, Yonsei University Health System, Seoul; Jong-Seok Lee and Jin-Haeng Chung, Seoul National University Bundang Hospital, Bundang; Ki Hyeong Lee, Chungbuk National University, Cheongju; Young Joo Min, University of Ulsan College of Medicine, Ulsan; Eun Kyung Cho, Gachon Medical School, Inchon; Sung Sook Lee, Inje University College of Medicine; Moon Young Choi, Inje University, Busan; Jin-Haeng Chung, Seoul National University College of Medicine, Seongnam, Republic of Korea; and Siraj M. Ali, Foundation Medicine, Cambridge, MA
| | - Young Joo Min
- Sun Min Lim, Hye Ryun Kim, Hyo Sup Shim, Joo-Hang Kim, and Byoung Chul Cho, Yonsei University College of Medicine; Yun-Gyoo Lee, Yoon La Choi, and Myung-Ju Ahn, Sungkyunkwan University School of Medicine; Bong-Seog Kim, VHS Medical Center; Min Jeong Lee and Maria Kim, Yonsei University Health System, Seoul; Jong-Seok Lee and Jin-Haeng Chung, Seoul National University Bundang Hospital, Bundang; Ki Hyeong Lee, Chungbuk National University, Cheongju; Young Joo Min, University of Ulsan College of Medicine, Ulsan; Eun Kyung Cho, Gachon Medical School, Inchon; Sung Sook Lee, Inje University College of Medicine; Moon Young Choi, Inje University, Busan; Jin-Haeng Chung, Seoul National University College of Medicine, Seongnam, Republic of Korea; and Siraj M. Ali, Foundation Medicine, Cambridge, MA
| | - Eun Kyung Cho
- Sun Min Lim, Hye Ryun Kim, Hyo Sup Shim, Joo-Hang Kim, and Byoung Chul Cho, Yonsei University College of Medicine; Yun-Gyoo Lee, Yoon La Choi, and Myung-Ju Ahn, Sungkyunkwan University School of Medicine; Bong-Seog Kim, VHS Medical Center; Min Jeong Lee and Maria Kim, Yonsei University Health System, Seoul; Jong-Seok Lee and Jin-Haeng Chung, Seoul National University Bundang Hospital, Bundang; Ki Hyeong Lee, Chungbuk National University, Cheongju; Young Joo Min, University of Ulsan College of Medicine, Ulsan; Eun Kyung Cho, Gachon Medical School, Inchon; Sung Sook Lee, Inje University College of Medicine; Moon Young Choi, Inje University, Busan; Jin-Haeng Chung, Seoul National University College of Medicine, Seongnam, Republic of Korea; and Siraj M. Ali, Foundation Medicine, Cambridge, MA
| | - Sung Sook Lee
- Sun Min Lim, Hye Ryun Kim, Hyo Sup Shim, Joo-Hang Kim, and Byoung Chul Cho, Yonsei University College of Medicine; Yun-Gyoo Lee, Yoon La Choi, and Myung-Ju Ahn, Sungkyunkwan University School of Medicine; Bong-Seog Kim, VHS Medical Center; Min Jeong Lee and Maria Kim, Yonsei University Health System, Seoul; Jong-Seok Lee and Jin-Haeng Chung, Seoul National University Bundang Hospital, Bundang; Ki Hyeong Lee, Chungbuk National University, Cheongju; Young Joo Min, University of Ulsan College of Medicine, Ulsan; Eun Kyung Cho, Gachon Medical School, Inchon; Sung Sook Lee, Inje University College of Medicine; Moon Young Choi, Inje University, Busan; Jin-Haeng Chung, Seoul National University College of Medicine, Seongnam, Republic of Korea; and Siraj M. Ali, Foundation Medicine, Cambridge, MA
| | - Bong-Seog Kim
- Sun Min Lim, Hye Ryun Kim, Hyo Sup Shim, Joo-Hang Kim, and Byoung Chul Cho, Yonsei University College of Medicine; Yun-Gyoo Lee, Yoon La Choi, and Myung-Ju Ahn, Sungkyunkwan University School of Medicine; Bong-Seog Kim, VHS Medical Center; Min Jeong Lee and Maria Kim, Yonsei University Health System, Seoul; Jong-Seok Lee and Jin-Haeng Chung, Seoul National University Bundang Hospital, Bundang; Ki Hyeong Lee, Chungbuk National University, Cheongju; Young Joo Min, University of Ulsan College of Medicine, Ulsan; Eun Kyung Cho, Gachon Medical School, Inchon; Sung Sook Lee, Inje University College of Medicine; Moon Young Choi, Inje University, Busan; Jin-Haeng Chung, Seoul National University College of Medicine, Seongnam, Republic of Korea; and Siraj M. Ali, Foundation Medicine, Cambridge, MA
| | - Moon Young Choi
- Sun Min Lim, Hye Ryun Kim, Hyo Sup Shim, Joo-Hang Kim, and Byoung Chul Cho, Yonsei University College of Medicine; Yun-Gyoo Lee, Yoon La Choi, and Myung-Ju Ahn, Sungkyunkwan University School of Medicine; Bong-Seog Kim, VHS Medical Center; Min Jeong Lee and Maria Kim, Yonsei University Health System, Seoul; Jong-Seok Lee and Jin-Haeng Chung, Seoul National University Bundang Hospital, Bundang; Ki Hyeong Lee, Chungbuk National University, Cheongju; Young Joo Min, University of Ulsan College of Medicine, Ulsan; Eun Kyung Cho, Gachon Medical School, Inchon; Sung Sook Lee, Inje University College of Medicine; Moon Young Choi, Inje University, Busan; Jin-Haeng Chung, Seoul National University College of Medicine, Seongnam, Republic of Korea; and Siraj M. Ali, Foundation Medicine, Cambridge, MA
| | - Hyo Sup Shim
- Sun Min Lim, Hye Ryun Kim, Hyo Sup Shim, Joo-Hang Kim, and Byoung Chul Cho, Yonsei University College of Medicine; Yun-Gyoo Lee, Yoon La Choi, and Myung-Ju Ahn, Sungkyunkwan University School of Medicine; Bong-Seog Kim, VHS Medical Center; Min Jeong Lee and Maria Kim, Yonsei University Health System, Seoul; Jong-Seok Lee and Jin-Haeng Chung, Seoul National University Bundang Hospital, Bundang; Ki Hyeong Lee, Chungbuk National University, Cheongju; Young Joo Min, University of Ulsan College of Medicine, Ulsan; Eun Kyung Cho, Gachon Medical School, Inchon; Sung Sook Lee, Inje University College of Medicine; Moon Young Choi, Inje University, Busan; Jin-Haeng Chung, Seoul National University College of Medicine, Seongnam, Republic of Korea; and Siraj M. Ali, Foundation Medicine, Cambridge, MA
| | - Jin-Haeng Chung
- Sun Min Lim, Hye Ryun Kim, Hyo Sup Shim, Joo-Hang Kim, and Byoung Chul Cho, Yonsei University College of Medicine; Yun-Gyoo Lee, Yoon La Choi, and Myung-Ju Ahn, Sungkyunkwan University School of Medicine; Bong-Seog Kim, VHS Medical Center; Min Jeong Lee and Maria Kim, Yonsei University Health System, Seoul; Jong-Seok Lee and Jin-Haeng Chung, Seoul National University Bundang Hospital, Bundang; Ki Hyeong Lee, Chungbuk National University, Cheongju; Young Joo Min, University of Ulsan College of Medicine, Ulsan; Eun Kyung Cho, Gachon Medical School, Inchon; Sung Sook Lee, Inje University College of Medicine; Moon Young Choi, Inje University, Busan; Jin-Haeng Chung, Seoul National University College of Medicine, Seongnam, Republic of Korea; and Siraj M. Ali, Foundation Medicine, Cambridge, MA
| | - Yoon La Choi
- Sun Min Lim, Hye Ryun Kim, Hyo Sup Shim, Joo-Hang Kim, and Byoung Chul Cho, Yonsei University College of Medicine; Yun-Gyoo Lee, Yoon La Choi, and Myung-Ju Ahn, Sungkyunkwan University School of Medicine; Bong-Seog Kim, VHS Medical Center; Min Jeong Lee and Maria Kim, Yonsei University Health System, Seoul; Jong-Seok Lee and Jin-Haeng Chung, Seoul National University Bundang Hospital, Bundang; Ki Hyeong Lee, Chungbuk National University, Cheongju; Young Joo Min, University of Ulsan College of Medicine, Ulsan; Eun Kyung Cho, Gachon Medical School, Inchon; Sung Sook Lee, Inje University College of Medicine; Moon Young Choi, Inje University, Busan; Jin-Haeng Chung, Seoul National University College of Medicine, Seongnam, Republic of Korea; and Siraj M. Ali, Foundation Medicine, Cambridge, MA
| | - Min Jeong Lee
- Sun Min Lim, Hye Ryun Kim, Hyo Sup Shim, Joo-Hang Kim, and Byoung Chul Cho, Yonsei University College of Medicine; Yun-Gyoo Lee, Yoon La Choi, and Myung-Ju Ahn, Sungkyunkwan University School of Medicine; Bong-Seog Kim, VHS Medical Center; Min Jeong Lee and Maria Kim, Yonsei University Health System, Seoul; Jong-Seok Lee and Jin-Haeng Chung, Seoul National University Bundang Hospital, Bundang; Ki Hyeong Lee, Chungbuk National University, Cheongju; Young Joo Min, University of Ulsan College of Medicine, Ulsan; Eun Kyung Cho, Gachon Medical School, Inchon; Sung Sook Lee, Inje University College of Medicine; Moon Young Choi, Inje University, Busan; Jin-Haeng Chung, Seoul National University College of Medicine, Seongnam, Republic of Korea; and Siraj M. Ali, Foundation Medicine, Cambridge, MA
| | - Maria Kim
- Sun Min Lim, Hye Ryun Kim, Hyo Sup Shim, Joo-Hang Kim, and Byoung Chul Cho, Yonsei University College of Medicine; Yun-Gyoo Lee, Yoon La Choi, and Myung-Ju Ahn, Sungkyunkwan University School of Medicine; Bong-Seog Kim, VHS Medical Center; Min Jeong Lee and Maria Kim, Yonsei University Health System, Seoul; Jong-Seok Lee and Jin-Haeng Chung, Seoul National University Bundang Hospital, Bundang; Ki Hyeong Lee, Chungbuk National University, Cheongju; Young Joo Min, University of Ulsan College of Medicine, Ulsan; Eun Kyung Cho, Gachon Medical School, Inchon; Sung Sook Lee, Inje University College of Medicine; Moon Young Choi, Inje University, Busan; Jin-Haeng Chung, Seoul National University College of Medicine, Seongnam, Republic of Korea; and Siraj M. Ali, Foundation Medicine, Cambridge, MA
| | - Joo-Hang Kim
- Sun Min Lim, Hye Ryun Kim, Hyo Sup Shim, Joo-Hang Kim, and Byoung Chul Cho, Yonsei University College of Medicine; Yun-Gyoo Lee, Yoon La Choi, and Myung-Ju Ahn, Sungkyunkwan University School of Medicine; Bong-Seog Kim, VHS Medical Center; Min Jeong Lee and Maria Kim, Yonsei University Health System, Seoul; Jong-Seok Lee and Jin-Haeng Chung, Seoul National University Bundang Hospital, Bundang; Ki Hyeong Lee, Chungbuk National University, Cheongju; Young Joo Min, University of Ulsan College of Medicine, Ulsan; Eun Kyung Cho, Gachon Medical School, Inchon; Sung Sook Lee, Inje University College of Medicine; Moon Young Choi, Inje University, Busan; Jin-Haeng Chung, Seoul National University College of Medicine, Seongnam, Republic of Korea; and Siraj M. Ali, Foundation Medicine, Cambridge, MA
| | - Siraj M Ali
- Sun Min Lim, Hye Ryun Kim, Hyo Sup Shim, Joo-Hang Kim, and Byoung Chul Cho, Yonsei University College of Medicine; Yun-Gyoo Lee, Yoon La Choi, and Myung-Ju Ahn, Sungkyunkwan University School of Medicine; Bong-Seog Kim, VHS Medical Center; Min Jeong Lee and Maria Kim, Yonsei University Health System, Seoul; Jong-Seok Lee and Jin-Haeng Chung, Seoul National University Bundang Hospital, Bundang; Ki Hyeong Lee, Chungbuk National University, Cheongju; Young Joo Min, University of Ulsan College of Medicine, Ulsan; Eun Kyung Cho, Gachon Medical School, Inchon; Sung Sook Lee, Inje University College of Medicine; Moon Young Choi, Inje University, Busan; Jin-Haeng Chung, Seoul National University College of Medicine, Seongnam, Republic of Korea; and Siraj M. Ali, Foundation Medicine, Cambridge, MA
| | - Myung-Ju Ahn
- Sun Min Lim, Hye Ryun Kim, Hyo Sup Shim, Joo-Hang Kim, and Byoung Chul Cho, Yonsei University College of Medicine; Yun-Gyoo Lee, Yoon La Choi, and Myung-Ju Ahn, Sungkyunkwan University School of Medicine; Bong-Seog Kim, VHS Medical Center; Min Jeong Lee and Maria Kim, Yonsei University Health System, Seoul; Jong-Seok Lee and Jin-Haeng Chung, Seoul National University Bundang Hospital, Bundang; Ki Hyeong Lee, Chungbuk National University, Cheongju; Young Joo Min, University of Ulsan College of Medicine, Ulsan; Eun Kyung Cho, Gachon Medical School, Inchon; Sung Sook Lee, Inje University College of Medicine; Moon Young Choi, Inje University, Busan; Jin-Haeng Chung, Seoul National University College of Medicine, Seongnam, Republic of Korea; and Siraj M. Ali, Foundation Medicine, Cambridge, MA
| | - Byoung Chul Cho
- Sun Min Lim, Hye Ryun Kim, Hyo Sup Shim, Joo-Hang Kim, and Byoung Chul Cho, Yonsei University College of Medicine; Yun-Gyoo Lee, Yoon La Choi, and Myung-Ju Ahn, Sungkyunkwan University School of Medicine; Bong-Seog Kim, VHS Medical Center; Min Jeong Lee and Maria Kim, Yonsei University Health System, Seoul; Jong-Seok Lee and Jin-Haeng Chung, Seoul National University Bundang Hospital, Bundang; Ki Hyeong Lee, Chungbuk National University, Cheongju; Young Joo Min, University of Ulsan College of Medicine, Ulsan; Eun Kyung Cho, Gachon Medical School, Inchon; Sung Sook Lee, Inje University College of Medicine; Moon Young Choi, Inje University, Busan; Jin-Haeng Chung, Seoul National University College of Medicine, Seongnam, Republic of Korea; and Siraj M. Ali, Foundation Medicine, Cambridge, MA
| |
Collapse
|
206
|
Vaishnavi A, Schubert L, Rix U, Marek LA, Le AT, Keysar SB, Glogowska MJ, Smith MA, Kako S, Sumi NJ, Davies KD, Ware KE, Varella-Garcia M, Haura EB, Jimeno A, Heasley LE, Aisner DL, Doebele RC. EGFR Mediates Responses to Small-Molecule Drugs Targeting Oncogenic Fusion Kinases. Cancer Res 2017; 77:3551-3563. [PMID: 28428274 DOI: 10.1158/0008-5472.can-17-0109] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 03/23/2017] [Accepted: 04/14/2017] [Indexed: 02/07/2023]
Abstract
Oncogenic kinase fusions of ALK, ROS1, RET, and NTRK1 act as drivers in human lung and other cancers. Residual tumor burden following treatment of ALK or ROS1+ lung cancer patients with oncogene-targeted therapy ultimately enables the emergence of drug-resistant clones, limiting the long-term effectiveness of these therapies. To determine the signaling mechanisms underlying incomplete tumor cell killing in oncogene-addicted cancer cells, we investigated the role of EGFR signaling in drug-naïve cancer cells harboring these oncogene fusions. We defined three distinct roles for EGFR in the response to oncogene-specific therapies. First, EGF-mediated activation of EGFR blunted fusion kinase inhibitor binding and restored fusion kinase signaling complexes. Second, fusion kinase inhibition shifted adaptor protein binding from the fusion oncoprotein to EGFR. Third, EGFR enabled bypass signaling to critical downstream pathways such as MAPK. While evidence of EGFR-mediated bypass signaling has been reported after ALK and ROS1 blockade, our results extended this effect to RET and NTRK1 blockade and uncovered the other additional mechanisms in gene fusion-positive lung cancer cells, mouse models, and human clinical specimens before the onset of acquired drug resistance. Collectively, our findings show how EGFR signaling can provide a critical adaptive survival mechanism that allows cancer cells to evade oncogene-specific inhibitors, providing a rationale to cotarget EGFR to reduce the risks of developing drug resistance. Cancer Res; 77(13); 3551-63. ©2017 AACR.
Collapse
Affiliation(s)
- Aria Vaishnavi
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| | - Laura Schubert
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| | - Uwe Rix
- Department of Drug Discovery, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Lindsay A Marek
- Department of Craniofacial Biology, University of Colorado School of Dental Medicine, Aurora, Colorado
| | - Anh T Le
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| | - Stephen B Keysar
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| | - Magdalena J Glogowska
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| | - Matthew A Smith
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Severine Kako
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| | - Natalia J Sumi
- Department of Drug Discovery, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Kurtis D Davies
- Department of Pathology, University of Colorado School of Medicine, Aurora, Colorado
| | - Kathryn E Ware
- Department of Craniofacial Biology, University of Colorado School of Dental Medicine, Aurora, Colorado
| | - Marileila Varella-Garcia
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| | - Eric B Haura
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Antonio Jimeno
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| | - Lynn E Heasley
- Department of Craniofacial Biology, University of Colorado School of Dental Medicine, Aurora, Colorado
| | - Dara L Aisner
- Department of Pathology, University of Colorado School of Medicine, Aurora, Colorado
| | - Robert C Doebele
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado.
| |
Collapse
|
207
|
Jiang L, Su X, Zhang T, Yin X, Zhang M, Fu H, Han H, Sun Y, Dong L, Qian J, Xu Y, Fu X, Gavine PR, Zhou Y, Tian K, Huang J, Shen D, Jiang H, Yao Y, Han B, Gu Y. PD-L1 expression and its relationship with oncogenic drivers in non-small cell lung cancer (NSCLC). Oncotarget 2017; 8:26845-26857. [PMID: 28460468 PMCID: PMC5432301 DOI: 10.18632/oncotarget.15839] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 02/20/2017] [Indexed: 01/05/2023] Open
Abstract
In order to explore the potential patient population who could benefit from anti PD-1/PD-L1 mono or combination therapies, this study aimed to profile a panel of immunotherapy related biomarkers (PD-1, PD-L1, CTLA-4 and CD8) and targeted therapy biomarkers (EGFR, KRAS, ALK, ROS1 and MET) in NSCLC.Tumor samples from 297 NSCLC patients, including 156 adenocarcinomas (AD) and 129 squamous cell carcinomas (SCC), were analyzed using immunohistochemistry, immunofluorescence, sequencing and fluorescence in situ hybridization.43.1% of NSCLC patients had PD-L1 positive staining on ≥ 5% tumor cells (TC). Furthermore, dual color immunofluorescence revealed that the majority of PD-L1/CD8 dual positive tumor infiltrating lymphocytes (TIL) had infiltrated into the tumor core. Finally, combined analysis of all eight biomarkers showed that tumor PD-L1 positivity overlapped with known alterations in NSCLC oncogenic tumor drivers in 26% of SCC and 76% of AD samples.Our illustration of the eight biomarkers' overlap provides an intuitive overview of NSCLC for personalized therapeutic strategies using anti-PD-1/PD-L1 immune therapies, either as single agents, or in combination with targeted therapies. For the first time, we also report that PD-L1 and CD8 dual positive TILs are predominantly located within the tumor core.
Collapse
MESH Headings
- Aged
- Aged, 80 and over
- B7-H1 Antigen/genetics
- B7-H1 Antigen/metabolism
- Biomarkers, Tumor
- CTLA-4 Antigen/genetics
- CTLA-4 Antigen/metabolism
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/metabolism
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Non-Small-Cell Lung/surgery
- Cell Transformation, Neoplastic/genetics
- Female
- Gene Amplification
- Gene Expression
- Humans
- Immunohistochemistry
- In Situ Hybridization, Fluorescence
- Lung Neoplasms/genetics
- Lung Neoplasms/metabolism
- Lung Neoplasms/pathology
- Lung Neoplasms/surgery
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Male
- Middle Aged
- Mutation
- Neoplasm Grading
- Neoplasm Staging
- Programmed Cell Death 1 Receptor/genetics
- Programmed Cell Death 1 Receptor/metabolism
Collapse
Affiliation(s)
- Liyan Jiang
- Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xinying Su
- Asia & Emerging Markets, iMed, AstraZeneca, Shanghai, China
| | - Tianwei Zhang
- Asia & Emerging Markets, iMed, AstraZeneca, Shanghai, China
| | - Xiaolu Yin
- Asia & Emerging Markets, iMed, AstraZeneca, Shanghai, China
| | | | - Haihua Fu
- Asia & Emerging Markets, iMed, AstraZeneca, Shanghai, China
| | - Hulin Han
- Asia & Emerging Markets, iMed, AstraZeneca, Shanghai, China
| | - Yun Sun
- Asia & Emerging Markets, iMed, AstraZeneca, Shanghai, China
| | - Lili Dong
- Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jialin Qian
- Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yanhua Xu
- Global Medicines Development, AstraZeneca, Shanghai, China
| | - Xuan Fu
- Global Medicines Development, AstraZeneca, Shanghai, China
| | - Paul R. Gavine
- Asia & Emerging Markets, iMed, AstraZeneca, Shanghai, China
| | - Yanbin Zhou
- The First Affiliated Hospital, Sun Yat-Sen University, Guangdong, China
| | - Kun Tian
- General Hospital of Chengdu Military Region of PLA, Sichuan, China
| | - Jiaqi Huang
- R&D, MedImmune, AstraZeneca, Gaithersburg, MD, USA
| | - Dong Shen
- R&D, MedImmune, AstraZeneca, Gaithersburg, MD, USA
| | - Haiyi Jiang
- Global Medicines Development, AstraZeneca, Shanghai, China
| | - Yihong Yao
- R&D, MedImmune, AstraZeneca, Gaithersburg, MD, USA
| | - Baohui Han
- Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yi Gu
- Asia & Emerging Markets, iMed, AstraZeneca, Shanghai, China
| |
Collapse
|
208
|
Calvayrac O, Pradines A, Pons E, Mazières J, Guibert N. Molecular biomarkers for lung adenocarcinoma. Eur Respir J 2017; 49:49/4/1601734. [PMID: 28381431 DOI: 10.1183/13993003.01734-2016] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 12/28/2016] [Indexed: 12/28/2022]
Abstract
The identification of oncogenic driver alterations that underlie sensitivity to small inhibitors has led to growing interest in identifying additional targetable oncogenes in nonsmall cell lung cancer. Although the therapeutic impact of the discovery of these alterations has now been widely demonstrated, the epidemiological data associated with each of these biomarkers remain insufficiently studied. In this review, we discuss the techniques used to discover each of these candidate oncogenes, their prevalence in nonsmall cell lung cancer, and briefly outline the epidemiological features of the major oncogenes and ways in which their identification can determine therapeutic strategies.
Collapse
Affiliation(s)
- Olivier Calvayrac
- Inserm, Centre de Recherche en Cancérologie de Toulouse, CRCT UMR-1037, Toulouse, France
| | - Anne Pradines
- Inserm, Centre de Recherche en Cancérologie de Toulouse, CRCT UMR-1037, Toulouse, France.,Institut Universitaire du Cancer, Toulouse, France.,Institut Claudius Regaud, IUCT-Oncopole, Laboratoire de Biologie Médicale Oncologique, Toulouse, France
| | - Elvire Pons
- Inserm, Centre de Recherche en Cancérologie de Toulouse, CRCT UMR-1037, Toulouse, France.,Institut Claudius Regaud, IUCT-Oncopole, Laboratoire de Biologie Médicale Oncologique, Toulouse, France
| | - Julien Mazières
- Inserm, Centre de Recherche en Cancérologie de Toulouse, CRCT UMR-1037, Toulouse, France .,Institut Universitaire du Cancer, Toulouse, France.,Hôpital Larrey, Centre Hospitalier Universitaire, Université Paul Sabatier, Toulouse, France
| | - Nicolas Guibert
- Inserm, Centre de Recherche en Cancérologie de Toulouse, CRCT UMR-1037, Toulouse, France.,Institut Universitaire du Cancer, Toulouse, France.,Hôpital Larrey, Centre Hospitalier Universitaire, Université Paul Sabatier, Toulouse, France
| |
Collapse
|
209
|
Oncogene addiction in non-small cell lung cancer: Focus on ROS1 inhibition. Cancer Treat Rev 2017; 55:83-95. [DOI: 10.1016/j.ctrv.2017.02.010] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 02/09/2017] [Accepted: 02/26/2017] [Indexed: 12/21/2022]
|
210
|
Paik PK. Something Old, Something New, Something Borrowed, Something Fused: Novel EGFR Rearrangements in Lung Adenocarcinomas. Cancer Discov 2017; 6:574-5. [PMID: 27261479 DOI: 10.1158/2159-8290.cd-16-0478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Mutations in EGFR stand as the archetype for somatic alterations that lead to oncogene addiction and that predict for response to targeted therapies. In this issue of Cancer Discovery, Konduri and colleagues report on a pair of novel oncogenic and actionable EGFR fusion events in a series of patients with lung adenocarcinomas, casting new light on this model gene. Cancer Discov; 6(6); 574-5. ©2016 AACRSee related article by Konduri et al., p. 601.
Collapse
Affiliation(s)
- Paul K Paik
- Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York. Weill Cornell Medical College, New York, New York.
| |
Collapse
|
211
|
Dagogo-Jack I, Engelman JA, Shaw AT. Overcoming On-Target Resistance to Tyrosine Kinase Inhibitors in Lung Cancer. ANNUAL REVIEW OF CANCER BIOLOGY-SERIES 2017. [DOI: 10.1146/annurev-cancerbio-050216-122044] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Advances in genomics, an improved understanding of malignant transformation, and the development of potent small molecule inhibitors capable of targeting key kinases have led to the adoption of genotype-guided approaches for the treatment of advanced cancers. As regulators of complex signaling networks, tyrosine kinases are among the most attractive targets. Moreover, insight into the conserved three-dimensional structures of these kinases and their mechanism of activation has facilitated the development of selective tyrosine kinase inhibitors (TKIs). TKIs have shown robust clinical activity in many different oncogene-addicted cancers; however, resistance invariably develops. In a significant proportion of patients, resistance results from acquired genetic alterations within the kinase target that allow cancer cells to escape TKI-mediated growth suppression. In this review, we discuss clinically observed and preclinical on-target resistance events in oncogene-driven solid tumors and describe current and future therapeutic strategies to overcome this type of resistance.
Collapse
Affiliation(s)
- Ibiayi Dagogo-Jack
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts 02114;,
| | | | - Alice T. Shaw
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts 02114;,
| |
Collapse
|
212
|
Hardin C, Wang F, Cheng H. Precision drug development in ROS1-positive lung cancer. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2017. [DOI: 10.1080/23808993.2017.1322899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
213
|
Domblides C, Antoine M, Lavole A, Cadranel J, Wislez M. [Crizotinib for ROS1-rearranged non-small cell lung cancer patients]. Bull Cancer 2017; 104:303-310. [PMID: 28237354 DOI: 10.1016/j.bulcan.2017.01.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2016] [Revised: 01/16/2017] [Accepted: 01/18/2017] [Indexed: 11/16/2022]
Abstract
ROS1 fusions are rare mutations that preferentially concern young and non-smoker women. The ROS1-rearranged protein conserves an intact tyrosine kinase domain, leading to the constitutive activation of the ROS1 tyrosine kinase function and of its downstream pathways, that are known to be involved in tumorigenesis. These molecular abnormalities have shown their oncogenic potential in animals' models and in human, with an early effect on carcinogenesis. Several partners have been identified. Patients with non-small cell lung cancers (NSCLC) harbouring ROS1 alterations can receive specific targeted therapies. Indeed, crizotinib has recently been approved in France in advanced ROS1-rearranged NSCLC. We propose a review of the oncogenic role of ROS1 rearrangements, the different methods for its diagnosis, and the available treatments.
Collapse
Affiliation(s)
- Charlotte Domblides
- GH HUEP, AP-HP, hôpital Tenon, service de pneumologie, 4, rue de la Chine, 75970 Paris, France
| | - Martine Antoine
- Hôpital Tenon, Sorbonne universités, UPMC université Paris 06, Theranoscan, GRC n(o) 04, 4, rue de la Chine, 75252 Paris, France; Hôpital Tenon, AP-HP, GH HUEP, service d'anatomie pathologique, 75970 Paris, France
| | - Armelle Lavole
- GH HUEP, AP-HP, hôpital Tenon, service de pneumologie, 4, rue de la Chine, 75970 Paris, France
| | - Jacques Cadranel
- GH HUEP, AP-HP, hôpital Tenon, service de pneumologie, 4, rue de la Chine, 75970 Paris, France; Hôpital Tenon, Sorbonne universités, UPMC université Paris 06, Theranoscan, GRC n(o) 04, 4, rue de la Chine, 75252 Paris, France
| | - Marie Wislez
- GH HUEP, AP-HP, hôpital Tenon, service de pneumologie, 4, rue de la Chine, 75970 Paris, France; Hôpital Tenon, Sorbonne universités, UPMC université Paris 06, Theranoscan, GRC n(o) 04, 4, rue de la Chine, 75252 Paris, France.
| |
Collapse
|
214
|
Luo LX, Fan XX, Li Y, Peng X, Ji YC, Hsiao WWL, Liu L, Leung ELH, Yao XJ. Identification of mitoxantrone as a new inhibitor of ROS1 fusion protein in non-small cell lung cancer cells. MEDCHEMCOMM 2017; 8:621-624. [PMID: 30108778 DOI: 10.1039/c6md00643d] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2016] [Accepted: 01/09/2017] [Indexed: 01/09/2023]
Abstract
Mitoxantrone, an FDA-approved drug for multiple sclerosis and hormone refractory prostate cancer, is identified as a potent inhibitor of ROS1 fusion protein by in silico screening in non-small cell lung cancer cells. Mitoxantrone can suppress the phosphorylation of ROS1 and subsequently inhibit its downstream signaling pathway and thus induce cell apoptosis.
Collapse
Affiliation(s)
- Lian-Xiang Luo
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute For Applied Research in Medicine and Health , Macau University of Science and Technology , Macau SAR , China . ; ; ; ; Tel: +853 8897 2438
| | - Xing-Xing Fan
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute For Applied Research in Medicine and Health , Macau University of Science and Technology , Macau SAR , China . ; ; ; ; Tel: +853 8897 2438
| | - Ying Li
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute For Applied Research in Medicine and Health , Macau University of Science and Technology , Macau SAR , China . ; ; ; ; Tel: +853 8897 2438
| | - Xia Peng
- State Key Laboratory of Drug Research , Shanghai Institute of Materia Medica , Chinese Academic of Sciences , Shanghai , China
| | - Yin-Chun Ji
- State Key Laboratory of Drug Research , Shanghai Institute of Materia Medica , Chinese Academic of Sciences , Shanghai , China
| | - Wendy Wen-Luan Hsiao
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute For Applied Research in Medicine and Health , Macau University of Science and Technology , Macau SAR , China . ; ; ; ; Tel: +853 8897 2438
| | - Liang Liu
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute For Applied Research in Medicine and Health , Macau University of Science and Technology , Macau SAR , China . ; ; ; ; Tel: +853 8897 2438
| | - Elaine Lai-Han Leung
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute For Applied Research in Medicine and Health , Macau University of Science and Technology , Macau SAR , China . ; ; ; ; Tel: +853 8897 2438
| | - Xiao-Jun Yao
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute For Applied Research in Medicine and Health , Macau University of Science and Technology , Macau SAR , China . ; ; ; ; Tel: +853 8897 2438
| |
Collapse
|
215
|
Clavé S, Gimeno J, Muñoz-Mármol AM, Vidal J, Reguart N, Carcereny E, Pijuan L, Menéndez S, Taus Á, Mate JL, Serrano S, Albanell J, Espinet B, Arriola E, Salido M. ROS1 copy number alterations are frequent in non-small cell lung cancer. Oncotarget 2016; 7:8019-28. [PMID: 26783962 PMCID: PMC4884972 DOI: 10.18632/oncotarget.6921] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 01/06/2016] [Indexed: 01/15/2023] Open
Abstract
Objectives We aimed to determine the prevalence and partners of ROS1 rearrangements, to explore the correlation between FISH and IHC assays, and to investigate clinical implications of ROS1 copy number alterations (CNAs). Methods A total of 314 NSCLC patients were screened using ROS1 FISH break-apart probes. Of these, 47 surgical tumors were included in TMAs to analyze ROS1 heterogeneity assessed either by FISH and IHC, and chromosome 6 aneusomy. To characterize ROS1 partners, probes for CD74, EZR, SLC34A2 and SDC3 genes were developed. ROS1 positive FISH cases were screened also by IHC. Results Five patients were ROS1 positive (1.8%). We identified two known fusion partners in three patients: CD74 and SLC34A2. Four out of five ROS1 rearranged patients were female, never smokers and with adenocarcinoma histology. Rearranged cases were also positive by IHC as well. According to ROS1 CNAs, we found a prevalence of 37.8% gains/amplifications and 25.1% deletions. Conclusions This study point out the high prevalence of ROS1 CNAs in a large series of NSCLC. ROS1 gains, amplifications and deletions, most of them due to chromosome 6 polysomy or monosomy, were heterogeneous within a tumor and had no impact on overall survival.
Collapse
Affiliation(s)
- Sergi Clavé
- Laboratori de Citogenètica Molecular, Servei de Patologia, Hospital del Mar, Barcelona, Spain.,Programa de Recerca en Càncer, IMIM (Institut Hospital del Mar de Investigacions Mèdiques), Barcelona, Spain
| | - Javier Gimeno
- Servei de Patologia, Hospital del Mar, Barcelona, Spain
| | - Ana M Muñoz-Mármol
- Servei de Anatomia Patològica, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Joana Vidal
- Servei de Oncologia Mèdica, Hospital del Mar, Barcelona, Spain
| | - Noemí Reguart
- Servei de Oncologia Mèdica, ICMHO, Hospital Clinic Barcelona, Barcelona, Spain
| | - Enric Carcereny
- Departament de Oncologia Mèdica, Institut Català de Oncologia (ICO), Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Lara Pijuan
- Servei de Patologia, Hospital del Mar, Barcelona, Spain
| | - Sílvia Menéndez
- Programa de Recerca en Càncer, IMIM (Institut Hospital del Mar de Investigacions Mèdiques), Barcelona, Spain.,Servei de Patologia, Hospital del Mar, Barcelona, Spain
| | - Álvaro Taus
- Servei de Oncologia Mèdica, Hospital del Mar, Barcelona, Spain
| | - José Luís Mate
- Servei de Anatomia Patològica, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | | | - Joan Albanell
- Programa de Recerca en Càncer, IMIM (Institut Hospital del Mar de Investigacions Mèdiques), Barcelona, Spain.,Servei de Oncologia Mèdica, Hospital del Mar, Barcelona, Spain
| | - Blanca Espinet
- Laboratori de Citogenètica Molecular, Servei de Patologia, Hospital del Mar, Barcelona, Spain.,Programa de Recerca en Càncer, IMIM (Institut Hospital del Mar de Investigacions Mèdiques), Barcelona, Spain
| | - Edurne Arriola
- Programa de Recerca en Càncer, IMIM (Institut Hospital del Mar de Investigacions Mèdiques), Barcelona, Spain.,Cancer Sciences Unit, University of Southampton. Southampton, United Kingdom
| | - Marta Salido
- Laboratori de Citogenètica Molecular, Servei de Patologia, Hospital del Mar, Barcelona, Spain.,Programa de Recerca en Càncer, IMIM (Institut Hospital del Mar de Investigacions Mèdiques), Barcelona, Spain
| |
Collapse
|
216
|
Gow CH, Hsieh MS, Wu SG, Shih JY. A comprehensive analysis of clinical outcomes in lung cancer patients harboring a MET exon 14 skipping mutation compared to other driver mutations in an East Asian population. Lung Cancer 2016; 103:82-89. [PMID: 28024701 DOI: 10.1016/j.lungcan.2016.12.001] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 11/10/2016] [Accepted: 12/02/2016] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Recurrent somatic splice-site alterations at MET exon 14 (METΔ14), which result in exon skipping and MET proto-oncogene, receptor tyrosine kinase (MET) activation, have been characterised. However, their demographic features and clinical outcomes in East Asian lung cancer patients have yet to be determined. METHODS A one-step reverse transcription-polymerase chain reaction (RT-PCR), using RNA samples from 850 East Asian lung cancer patients, was performed in order to detect METΔ14 and five other major driver mutations, including those in the EGFR, KRAS, ALK, HER2, and ROS1 genes. Immunohistochemistry (IHC) was used to confirm the overexpression of MET in patients harbouring the METΔ14 mutation. We analysed the demographic data and clinical outcomes of METΔ14 mutation positive lung cancer patients and compared them to those of METΔ14 mutation negative lung cancer patients. RESULTS In total, 27 lung adenocarcinoma (ADC) patients and 1 squamous cell carcinoma patient with the METΔ14 mutation were identified. The overall incidence was 3.3% for lung cancer and 4.0% for lung ADC. IHC demonstrated that the majority of lung cancer patients harboring a METΔ14 mutation exhibited a strong cytoplasmic expression of MET. METΔ14 mutation positive patients were generally quite elderly individuals. Stage IV METΔ14 mutation positive lung cancer patients receiving no specific anti-MET therapy were observed to have a similar overall survival (OS) compared to patients in the all negative group (P>0.05). In the multivariate analysis, mutation status was found not to be a major risk factor for OS in lung cancer patients without appropriate tyrosine kinase inhibitors treatment. CONCLUSIONS The OS of METΔ14 mutation positive lung cancer patients is comparable to that of the major driver gene mutation negative lung cancer patients.
Collapse
Affiliation(s)
- Chien-Hung Gow
- Department of Internal Medicine, Far Eastern Memorial Hospital, New Taipei City, Taiwan; Department of Healthcare Information and Management, Ming-Chuan University, Taiwan; Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Min-Shu Hsieh
- Department of Pathology, National Taiwan University Hospital, Taipei, Taiwan
| | - Shang-Gin Wu
- Department of Internal Medicine, National Taiwan University Hospital Yun-Lin, Branch, Yun-Lin, Taiwan; Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan; Graduate Institute of Clinical Medicine, National Taiwan University, Taipei, Taiwan
| | - Jin-Yuan Shih
- Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan; Graduate Institute of Clinical Medicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
217
|
Michels S, Wolf J. Stratified Treatment in Lung Cancer. Oncol Res Treat 2016; 39:760-766. [PMID: 27889787 DOI: 10.1159/000453406] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 11/15/2016] [Indexed: 11/19/2022]
Abstract
Even though great efforts have been made to improve chemotherapy-based treatment approaches for lung cancer, the prognosis of patients with advanced and metastasized disease remains particularly poor. In recent years, a growing number of genetic aberrations driving lung cancer have been identified. Targeted inhibition of some of these aberrations, most prominently mutated EGFR and ALK, by tyrosine kinase inhibitors has dramatically increased efficacy and tolerability of systemic lung cancer treatment in subsets of patients. However, the duration of response is limited due to the acquisition of molecular mechanisms of resistance to targeted treatment. Modern next-generation inhibitors aim to break resistance. A deep understanding of the mechanisms of treatment failure is imperative to the development of new approaches. In this review, we focus on the current status of stratified therapy in lung cancer and highlight new, potentially promising treatment approaches.
Collapse
Affiliation(s)
- Sebastian Michels
- Lung Cancer Group Cologne, Department I for Internal Medicine, Center for Integrated Oncology, University Hospital of Cologne, Cologne, Germany
| | | |
Collapse
|
218
|
Selinger CI, Li BT, Pavlakis N, Links M, Gill AJ, Lee A, Clarke S, Tran TN, Lum T, Yip PY, Horvath L, Yu B, Kohonen-Corish MRJ, O'Toole SA, Cooper WA. Screening for ROS1 gene rearrangements in non-small-cell lung cancers using immunohistochemistry with FISH confirmation is an effective method to identify this rare target. Histopathology 2016; 70:402-411. [PMID: 27599111 DOI: 10.1111/his.13076] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 09/02/2016] [Indexed: 12/26/2022]
Abstract
AIMS To assess the prevalence of ROS1 rearrangements in a retrospective and prospective diagnostic Australian cohort and evaluate the effectiveness of immunohistochemical screening. METHODS AND RESULTS A retrospective cohort of 278 early stage lung adenocarcinomas and an additional 104 prospective non-small-cell lung cancer (NSCLC) cases referred for routine molecular testing were evaluated. ROS1 immunohistochemistry (IHC) was performed (D4D6 clone, Cell Signaling Technology) on all cases as well as fluorescence in-situ hybridization (FISH) using the ZytoVision and Abbott Molecular ROS1 FISH probes, with ≥15% of cells with split signals considered positive for rearrangement. Eighty-eight cases (32%) from the retrospective cohort showed staining by ROS1 IHC, and one case (0.4%) showed ROS1 rearrangement by FISH. Nineteen of the prospective diagnostic cases showed ROS1 IHC staining, 12 (12%) cases of which were confirmed as ROS1 rearranged by FISH. There were no ROS1 rearranged cases that showed no expression of ROS1 with IHC. The ROS1 rearranged cases in the prospective cohort were all EGFR wild-type and anaplastic lymphoma kinase (ALK) rearrangement-negative. The sensitivity of ROS1 IHC in the retrospective cohort was 100% and specificity was 76%. CONCLUSIONS ROS1 rearrangements are rare events in lung adenocarcinomas. Selection of cases for ROS1 FISH testing, by excluding EGFR/ALK-positive cases and use of IHC to screen for potentially positive cases, can be used to enrich for the likelihood of identifying a ROS1 rearranged lung cancer and prevent the need to undertake expensive and time-consuming FISH testing in all cases.
Collapse
Affiliation(s)
- Christina I Selinger
- Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
| | - Bob T Li
- Department of Medical Oncology, Royal North Shore Hospital, St Leonards, NSW, Australia.,Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, USA.,Sydney Medical School, University of Sydney, Camperdown, NSW, Australia
| | - Nick Pavlakis
- Department of Medical Oncology, Royal North Shore Hospital, St Leonards, NSW, Australia.,Sydney Medical School, University of Sydney, Camperdown, NSW, Australia
| | - Matthew Links
- Department of Medical Oncology, St George Hospital, Kogarah, NSW, Australia
| | - Anthony J Gill
- Sydney Medical School, University of Sydney, Camperdown, NSW, Australia.,Cancer Diagnosis and Pathology Group, Kolling Institute of Medical Research, Royal North Shore Hospital, St Leonards, NSW, Australia
| | - Adrian Lee
- Department of Medical Oncology, Royal North Shore Hospital, St Leonards, NSW, Australia
| | - Stephen Clarke
- Department of Medical Oncology, Royal North Shore Hospital, St Leonards, NSW, Australia.,Sydney Medical School, University of Sydney, Camperdown, NSW, Australia
| | - Thang N Tran
- Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
| | - Trina Lum
- Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
| | - Po Y Yip
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.,School of Medicine, University of Western Sydney, Sydney, NSW, Australia.,Chris O'Brien Lifehouse, Camperdown, NSW, Australia.,Macarthur Cancer Therapy Centre, Campbelltown Hospital, Camperdown, NSW, Australia
| | - Lisa Horvath
- Sydney Medical School, University of Sydney, Camperdown, NSW, Australia.,The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.,Medical Oncology, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
| | - Bing Yu
- Sydney Medical School, University of Sydney, Camperdown, NSW, Australia.,Department of Medical Genomics, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
| | - Maija R J Kohonen-Corish
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.,School of Medicine, University of Western Sydney, Sydney, NSW, Australia.,St Vincent's Clinical School, University of New South Wales, Darlinghurst, NSW, Australia
| | - Sandra A O'Toole
- Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Camperdown, NSW, Australia.,Sydney Medical School, University of Sydney, Camperdown, NSW, Australia.,The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Wendy A Cooper
- Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Camperdown, NSW, Australia.,Sydney Medical School, University of Sydney, Camperdown, NSW, Australia.,School of Medicine, University of Western Sydney, Sydney, NSW, Australia
| |
Collapse
|
219
|
Crizotinib for ROS1 patients: One small step in biomarker testing, one giant leap for advanced NSCLC patients. Lung Cancer 2016; 104:131-133. [PMID: 27863911 DOI: 10.1016/j.lungcan.2016.11.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 11/10/2016] [Indexed: 12/12/2022]
|
220
|
Bunn PA. Karnofsky Award 2016: A Lung Cancer Journey, 1973 to 2016. J Clin Oncol 2016; 35:243-252. [PMID: 28056194 DOI: 10.1200/jco.2016.70.4064] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Affiliation(s)
- Paul A Bunn
- From University of Colorado Cancer Center, Aurora, CO
| |
Collapse
|
221
|
Bubendorf L, Büttner R, Al-Dayel F, Dietel M, Elmberger G, Kerr K, López-Ríos F, Marchetti A, Öz B, Pauwels P, Penault-Llorca F, Rossi G, Ryška A, Thunnissen E. Testing for ROS1 in non-small cell lung cancer: a review with recommendations. Virchows Arch 2016; 469:489-503. [PMID: 27535289 PMCID: PMC5082594 DOI: 10.1007/s00428-016-2000-3] [Citation(s) in RCA: 169] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 07/07/2016] [Accepted: 08/02/2016] [Indexed: 10/26/2022]
Abstract
Rearrangements of the ROS1 gene occur in 1-2 % of non-small cell lung cancers (NSCLCs). Crizotinib, a highly effective inhibitor of ROS1 kinase activity, is now FDA-approved for the treatment of patients with advanced ROS1-positive NSCLC. Consequently, focus on ROS1 testing is growing. Most laboratories currently rely on fluorescence in situ hybridisation (FISH) assays using a dual-colour break-apart probe to detect ROS1 rearrangements. Given the rarity of these rearrangements in NSCLC, detection of elevated ROS1 protein levels by immunohistochemistry may provide cost-effective screening prior to confirmatory FISH testing. Non-in situ testing approaches also hold potential as stand-alone methods or complementary tests, including multiplex real-time PCR assays and next-generation sequencing (NGS) platforms which include commercial test kits covering a range of fusion genes. In order to ensure high-quality biomarker testing, appropriate tissue handling, adequate control materials and participation in external quality assessment programmes are essential, irrespective of the testing technique employed. ROS1 testing is often only considered after negative tests for EGFR mutation and ALK gene rearrangement, based on the assumption that these oncogenic driver events tend to be exclusive. However, as the use of ROS1 inhibitors becomes routine, accurate and timely detection of ROS1 gene rearrangements will be critical for the optimal treatment of patients with NSCLC. As NGS techniques are introduced into routine diagnostic practice, ROS1 fusion gene testing will be provided as part of the initial testing package.
Collapse
Affiliation(s)
- Lukas Bubendorf
- Institute of Pathology, University Hospital Basel, Basel, Switzerland
| | - Reinhard Büttner
- Institute of Pathology, University Hospital Cologne and Network Genomic Medicine, Cologne, Germany
| | - Fouad Al-Dayel
- Department of Pathology and Laboratory Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Manfred Dietel
- Institute of Pathology, Charité Campus Mitte, Berlin, Germany
| | - Göran Elmberger
- Department of Pathology and Cytology, Karolinska University Hospital, Stockholm, Sweden
| | - Keith Kerr
- Department of Pathology, Aberdeen University Medical School, Aberdeen, UK
| | - Fernando López-Ríos
- Laboratorio de Dianas Terapéuticas, Hospital Universitario HM Sanchinarro, C/Oña, 10, 28050, Madrid, Spain.
| | - Antonio Marchetti
- Center of Predictive Molecular Medicine, University-Foundation, Chieti, Italy
| | - Büge Öz
- Cerrahpasa Medical Faculty, Istanbul University, Istanbul, Turkey
| | - Patrick Pauwels
- Institute of Pathology, University Hospital Antwerp, Edegem, Belgium
| | | | - Giulio Rossi
- Unit of Pathologic Anatomy, Azienda USL Valle d'Aosta, Aosta, Italy
| | - Aleš Ryška
- The Fingerland Department of Pathology, Charles University Faculty of Medicine and Faculty Hospital in Hradec Kralove, Hradec Kralove, Czech Republic
| | - Erik Thunnissen
- Department of Pathology, VU University Medical Centre, Amsterdam, The Netherlands
| |
Collapse
|
222
|
Zhong E, Huang H. Crizotinib in ROS1 rearranged non-small cell lung cancer (NSCLC), from response to resistance. BMJ Case Rep 2016; 2016:bcr-2016-217322. [PMID: 27797842 DOI: 10.1136/bcr-2016-217322] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
We examined an immediate, but short-lived, response to crizotinib, a drug with a new indication for ROS1 rearranged non-small cell lung cancer (NSCLC) in a middle-aged non-smoker. The patient presented with metastatic NSCLC and extensive disease in multiple organs. He was treated with crizotinib 250 mg twice a day. Within 2-3 days, his condition rapidly improved, which was evident in a CT scan 2 months later. However, after 3 months of treatment, his condition deteriorated dramatically. The patient did not respond to ceritinib, a second-line drug that targets anaplastic lymphoma kinase, and died shortly after. This case demonstrated an impressive but brief response to crizotinib.
Collapse
Affiliation(s)
- Eric Zhong
- Quality Medical Oncology, Flushing, New York, USA.,Brown University, Providence, Rhode Island, USA
| | - Hua Huang
- Quality Medical Oncology, Flushing, New York, USA.,New York Presbyterian Queens, Flushing, New York, USA
| |
Collapse
|
223
|
Wiesweg M, Eberhardt WEE, Reis H, Ting S, Savvidou N, Skiba C, Herold T, Christoph DC, Meiler J, Worm K, Kasper S, Theegarten D, Hense J, Hager T, Darwiche K, Oezkan F, Aigner C, Welter S, Kühl H, Stuschke M, Schmid KW, Schuler M. High Prevalence of Concomitant Oncogene Mutations in Prospectively Identified Patients with ROS1-Positive Metastatic Lung Cancer. J Thorac Oncol 2016; 12:54-64. [PMID: 27575422 DOI: 10.1016/j.jtho.2016.08.137] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 08/17/2016] [Accepted: 08/18/2016] [Indexed: 02/01/2023]
Abstract
OBJECTIVES Chromosomal rearrangements involving ROS1 define a rare entity of lung adenocarcinomas with exquisite sensitivity to molecularly targeted therapy. We report clinical outcomes and genomic findings of patients with ROS1-positive lung cancer who were prospectively identified within a multiplex biomarker profiling program at the West German Cancer Center. METHODS Standardized immunohistochemical (IHC) analysis, fluorescence in situ hybridization (FISH), and hotspot mutation analyses were performed in 1345 patients with advanced cancer, including 805 patients with metastatic lung adenocarcinoma. Clinical and epidemiological data were retrieved from the institutional database. RESULTS ROS1 positivity by IHC analysis was detected in 25 patients with lung cancer (4.8% of lung adenocarcinomas), including 13 patients (2.5%) with ROS1 FISH positivity with a cutoff of at least 15% of events. Of the ROS1 IHC analysis-positive cases, 36% presented with concomitant oncogenic driver mutations involving EGFR (six cases, five of which were clinically validated by response to EGFR-targeting agents), KRAS (two cases), phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha gene (PIK3CA), and BRAF. Three cases initially classified as ROS1 FISH-negative passed the threshold of 15% positive events when repeat biopsies were analyzed at progression. The median overall survival of the ROS1-positive patients (104 months) was significantly superior to that of the 261 patients with EGFR/anaplastic lymphoma kinase/ROS1-negative lung adenocarcinoma (24.4 months, p = 0.044). Interestingly, the overall survival of the 13 ROS1-positive patients with lung cancer from initiation of pemetrexed-based chemotherapy was significantly prolonged when compared with that of 169 pemetrexed-treated patients with EGFR/anaplastic lymphoma kinase/ROS1-negative adenocarcinoma (p = 0.01). CONCLUSIONS ROS1-positive metastatic lung adenocarcinomas frequently harbor concomitant oncogenic driver mutations. Levels of ROS1 FISH-positive events are variable over time. This heterogeneity provides additional therapeutic options if discovered by multiplex biomarker testing and repeat biopsies.
Collapse
Affiliation(s)
- Marcel Wiesweg
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Wilfried E E Eberhardt
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany; Division of Thoracic Oncology, West German Lung Center, Ruhrlandklinik, University Hospital Essen, Essen, Germany
| | - Henning Reis
- Institute of Pathology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Saskia Ting
- Institute of Pathology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Nikoleta Savvidou
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Charlotte Skiba
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Thomas Herold
- Institute of Pathology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany; German Cancer Consortium (DKTK), Partner Site University Hospital Essen, Essen, Germany
| | - Daniel C Christoph
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Johannes Meiler
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Karl Worm
- Institute of Pathology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Stefan Kasper
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Dirk Theegarten
- Institute of Pathology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Jörg Hense
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Thomas Hager
- Institute of Pathology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Kaid Darwiche
- Division of Interventional Pneumology, West German Lung Center, Ruhrlandklinik, University Hospital Essen, Essen, Germany
| | - Filiz Oezkan
- Division of Interventional Pneumology, West German Lung Center, Ruhrlandklinik, University Hospital Essen, Essen, Germany
| | - Clemens Aigner
- Division of Thoracic Surgery, West German Lung Center, Ruhrlandklinik, University Hospital Essen, Essen, Germany
| | - Stefan Welter
- Division of Thoracic Surgery, West German Lung Center, Ruhrlandklinik, University Hospital Essen, Essen, Germany
| | - Hilmar Kühl
- Institute for Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Martin Stuschke
- German Cancer Consortium (DKTK), Partner Site University Hospital Essen, Essen, Germany; Department of Radiotherapy, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Kurt W Schmid
- Institute of Pathology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany; German Cancer Consortium (DKTK), Partner Site University Hospital Essen, Essen, Germany
| | - Martin Schuler
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany; Division of Thoracic Oncology, West German Lung Center, Ruhrlandklinik, University Hospital Essen, Essen, Germany; German Cancer Consortium (DKTK), Partner Site University Hospital Essen, Essen, Germany.
| |
Collapse
|
224
|
Wu J, Lin Y, He X, Yang H, He P, Fu X, Li G, Gu X. Comparison of detection methods and follow-up study on the tyrosine kinase inhibitors therapy in non-small cell lung cancer patients with ROS1 fusion rearrangement. BMC Cancer 2016; 16:599. [PMID: 27488371 PMCID: PMC4973062 DOI: 10.1186/s12885-016-2582-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2015] [Accepted: 07/19/2016] [Indexed: 01/15/2023] Open
Abstract
Background The screening of ROS proto-oncogene 1, receptor tyrosine kinase(ROS1) fusion rearrangement might be potentially beneficial for an effective therapy against non-small cell lung cancer (NSCLC). However, the three main ROS1 rearrangement detection methods have limitations, and no routine protocol for the detection of ROS1 rearrangement in NSCLC is available. In this study, our aims were to compare immunohistochemistry (IHC), fluorescent in situ hybridization (FISH) and quantitative real-time polymerase chain reaction (qRT-PCR) in their ability to detect ROS1 rearrangement in NSCLC, and discuss the clinical characteristics and histopathology of the patients with ROS1 rearrangement. Moreover, the effects of tyrosine kinase inhibitors (TKIs) therapy on the patients with ROS1 rearrangement and advanced stage disease (III b–IV) were investigated. Methods Patients with a previously diagnosed NSCLC were recruited in this study from November 2013 to October 2015. IHC was performed using the D4D6 monoclonal antibody (mAb) in an automatic IHC instrument, while FISH and qRT-PCR were carried out to confirm the IHC results. FISH and qRT-PCR positive cases underwent direct sequencing. After detection, patients with advanced ROS1 rearranged NSCLC had received TKI therapy. Results Two hundred and thirty-eight patients were included in this study. ROS1 rearrangement was detected in 10 patients. The concordant rate of FISH and qRT-PCR results was 100 %, while in the FISH and IHC results high congruence was present when IHC showed a diffusely (≥60 % tumor cells) 2–3+ cytoplasmic reactivity pattern. Patients harboring ROS1 rearrangement were mostly young (8/10), females (7/10) and non-smokers (7/10) with adenocarcinoma (10/10) and acinar pattern. Most of their tumor were in intermediate grade (6/8). Among these 10 patients, three of them in stage IV with ROS1 rearrangement gained benefits from ROS1 TKI therapy. Conclusions IHC, FISH and qRT-PCR can reliably detect ROS1 rearrangement in NSCLC, while IHC can be used as a preliminary screening tool. These results supported the efficacy of ROS1 TKI therapy in treating advanced NSCLC patients with ROS1 rearrangement. Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-2582-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jieyu Wu
- Department of Pathology, the First Affiliated Hospital of Guangzhou Medical University, No. 151, Yanjiangxi Road, Guangzhou, 510120, China
| | - Yunen Lin
- Department of Pathology, the First Affiliated Hospital of Guangzhou Medical University, No. 151, Yanjiangxi Road, Guangzhou, 510120, China
| | - Xinming He
- Department of Pathology, the First Affiliated Hospital of Guangzhou Medical University, No. 151, Yanjiangxi Road, Guangzhou, 510120, China
| | - Haihong Yang
- Department of Cardiothoracic Surgery, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ping He
- Department of Pathology, the First Affiliated Hospital of Guangzhou Medical University, No. 151, Yanjiangxi Road, Guangzhou, 510120, China
| | - Xinge Fu
- Department of Pathology, the First Affiliated Hospital of Guangzhou Medical University, No. 151, Yanjiangxi Road, Guangzhou, 510120, China
| | - Guangqiu Li
- Department of Pathology, the First Affiliated Hospital of Guangzhou Medical University, No. 151, Yanjiangxi Road, Guangzhou, 510120, China
| | - Xia Gu
- Department of Pathology, the First Affiliated Hospital of Guangzhou Medical University, No. 151, Yanjiangxi Road, Guangzhou, 510120, China.
| |
Collapse
|
225
|
Zhu YC, Xu CW, Ye XQ, Yin MX, Zhang JX, Du KQ, Zhang ZH, Hu J. Lung cancer with concurrent EGFR mutation and ROS1 rearrangement: a case report and review of the literature. Onco Targets Ther 2016; 9:4301-5. [PMID: 27486332 PMCID: PMC4956058 DOI: 10.2147/ott.s109415] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
ROS1 rearrangement has recently emerged as a new molecular subtype in non-small cell lung cancer, and is predominantly found in lung adenocarcinomas compared with other oncogenes such as EGFR, KRAS, or ALK. Patients who have both mutations are extremely rare. Here we report a 50-year-old female diagnosed with adenocarcinoma with sarcomatoid differentiation, who was shown to have EGFR and ROS1 mutations. The patient was treated surgically and received three cycles of adjuvant postoperative chemotherapy. In addition, we reviewed the previously reported cases and related literature. This presentation will provide further understanding of the underlying molecular biology and optimal treatment for non-small cell lung cancer patients with more than one driver mutation.
Collapse
Affiliation(s)
- You-Cai Zhu
- Department of Thoracic Surgery, The First Affiliated Hospital of Medical School of Zhejiang University, Hangzhou; Department of Thoracic Surgery, Chinese People's Armed Police Force, Zhejiang Corps Hospital, Jiaxing, Zhejiang
| | - Chun-Wei Xu
- Department of Pathology, Affiliated Hospital of Academy of Military Medical Sciences, Beijing
| | - Xiao-Qian Ye
- Department of Pathology, Chinese People's Armed Police Force, Zhejiang Corps Hospital, Jiaxing, Zhejiang, People's Republic of China
| | - Man-Xiang Yin
- Department of Pathology, Chinese People's Armed Police Force, Zhejiang Corps Hospital, Jiaxing, Zhejiang, People's Republic of China
| | - Jin-Xian Zhang
- Department of Thoracic Surgery, Chinese People's Armed Police Force, Zhejiang Corps Hospital, Jiaxing, Zhejiang
| | - Kai-Qi Du
- Department of Thoracic Surgery, Chinese People's Armed Police Force, Zhejiang Corps Hospital, Jiaxing, Zhejiang
| | - Zhi-Hao Zhang
- Department of Thoracic Surgery, Chinese People's Armed Police Force, Zhejiang Corps Hospital, Jiaxing, Zhejiang
| | - Jian Hu
- Department of Thoracic Surgery, The First Affiliated Hospital of Medical School of Zhejiang University, Hangzhou
| |
Collapse
|
226
|
Chen YF, Hsieh MS, Wu SG, Chang YL, Yu CJ, Yang JCH, Yang PC, Shih JY. Efficacy of Pemetrexed-Based Chemotherapy in Patients with ROS1 Fusion–Positive Lung Adenocarcinoma Compared with in Patients Harboring Other Driver Mutations in East Asian Populations. J Thorac Oncol 2016; 11:1140-52. [DOI: 10.1016/j.jtho.2016.03.022] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Revised: 03/24/2016] [Accepted: 03/25/2016] [Indexed: 02/07/2023]
|
227
|
Waxman ES, Fossella FV. Biomarkers/Molecular Targets, Immunotherapy, and Treatments for Non-Small Cell Lung Cancer. J Adv Pract Oncol 2016; 7:514-524. [PMID: 29282427 PMCID: PMC5737399 DOI: 10.6004/jadpro.2016.7.5.4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
For decades, the prognosis for patients with advanced-stage non-small cell lung cancer (NSCLC) was bleak, with chemotherapy offering limited benefit and much toxicity. Now, with mutational testing, new generations of targeted therapies, and emerging immunotherapies, the treatment horizon for these patients has greatly expanded. In this article, the authors review molecular targets, biomarkers, as well as immune checkpoint inhibitors, which are having a major impact on the management of this patient population.
Collapse
|
228
|
Viola P, Maurya M, Croud J, Gazdova J, Suleman N, Lim E, Newsom-Davis T, Plowman N, Rice A, Montero MA, Gonzalez de Castro D, Popat S, Nicholson AG. A Validation Study for the Use of ROS1 Immunohistochemical Staining in Screening for ROS1 Translocations in Lung Cancer. J Thorac Oncol 2016; 11:1029-39. [DOI: 10.1016/j.jtho.2016.03.019] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Revised: 03/17/2016] [Accepted: 03/31/2016] [Indexed: 12/21/2022]
|
229
|
Kazandjian D, Blumenthal GM, Luo L, He K, Fran I, Lemery S, Pazdur R. Benefit-Risk Summary of Crizotinib for the Treatment of Patients With ROS1 Alteration-Positive, Metastatic Non-Small Cell Lung Cancer. Oncologist 2016; 21:974-80. [PMID: 27328934 PMCID: PMC4978556 DOI: 10.1634/theoncologist.2016-0101] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 04/12/2016] [Indexed: 12/22/2022] Open
Abstract
The FDA has expanded the crizotinib metastatic non-small cell lung cancer indication to include treatment of patients whose tumors harbor a ROS1 rearrangement. The approval was based on a clinically meaningful, durable objective response rate (66%) in a multicenter, single-arm clinical trial. Patients received crizotinib 250 mg twice daily; the median duration of exposure and of response was 34.4 and 18.3 months, respectively. On March 11, 2016, after an expedited 5-month review, the U.S. Food and Drug Administration expanded the crizotinib metastatic non-small cell lung cancer (mNSCLC) indication to include the treatment of patients whose tumors harbor a ROS1 rearrangement. The approval was based on a clinically meaningful, durable objective response rate (ORR) in a multicenter, single-arm clinical trial (ROS1 cohort of Trial PROFILE 1001) in patients with ROS1-positive mNSCLC. The trial enrolled 50 patients (age range: 25–77 years) whose tumors were prospectively determined to have a ROS1 gene rearrangement by break-apart fluorescence in situ hybridization (96%) or reverse transcriptase polymerase chain reaction (4%) clinical trial assays. Crizotinib demonstrated an ORR of 66% (95% confidence interval [CI]: 51%–79%) with a median duration of response of 18.3 months by independent radiology review and 72% (95% CI: 58%–84%) by investigator review. Patients received crizotinib 250 mg twice daily and had a median duration of exposure of 34.4 months. The toxicity profile in ROS1-positive patients was generally consistent with the randomized safety data in the U.S. Product Insert from two ALK-positive mNSCLC trials. The most common (≥25%) adverse reactions and laboratory test abnormalities included vision disorders, elevation of alanine transaminase and aspartate transaminase levels, nausea, hypophosphatemia, diarrhea, edema, vomiting, constipation, neutropenia, and fatigue. There were no treatment-related deaths. A favorable benefit-to-risk evaluation led to the traditional approval of crizotinib for this new supplemental indication. Implications for Practice: Given the results from the ROS1 cohort of the clinical trial PROFILE 1001, crizotinib represents a new treatment option and the first approved therapy for patients with metastatic non-small cell lung cancer whose tumors are ROS1 positive. Crizotinib demonstrated efficacy irrespective of prior treatment status.
Collapse
Affiliation(s)
- Dickran Kazandjian
- Office of Hematology and Oncology Products and Office of Biostatistics, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Gideon M Blumenthal
- Office of Hematology and Oncology Products and Office of Biostatistics, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Lola Luo
- Office of Hematology and Oncology Products and Office of Biostatistics, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Kun He
- Office of Hematology and Oncology Products and Office of Biostatistics, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Ingrid Fran
- Office of Hematology and Oncology Products and Office of Biostatistics, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Steven Lemery
- Office of Hematology and Oncology Products and Office of Biostatistics, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Richard Pazdur
- Office of Hematology and Oncology Products and Office of Biostatistics, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| |
Collapse
|
230
|
McCoach CE, Bivona TG, Blakely CM, Doebele RC. Neoadjuvant Oncogene-Targeted Therapy in Early Stage Non-Small-Cell Lung Cancer as a Strategy to Improve Clinical Outcome and Identify Early Mechanisms of Resistance. Clin Lung Cancer 2016; 17:466-469. [PMID: 27378174 DOI: 10.1016/j.cllc.2016.05.025] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 05/31/2016] [Indexed: 01/23/2023]
Abstract
Evaluations of resistance mechanisms to targeted treatments in non-small-cell lung cancer (NSCLC) are necessary for development of improved treatment after disease progression and to help delay progression of disease. Populations of cells that survive after initial treatment form the basis of resistance via outgrowth of resistant clones or activation of alternative signaling pathways. In this report we describe a clinical trial approach in which patients with epidermal growth factor receptor (EGFR), anaplastic lymphoma kinase (ALK), C-ros-1 proto-oncogene (ROS1), and hepatocyte growth factor receptor (MET) exon 14 alterations and early stage (IA-IIIA) NSCLC will be treated with induction EGFR tyrosine kinase inhibitor (TKI) or crizotinib, a TKI that inhibits ALK, ROS1, and MET. We will evaluate resected tumor samples for pathologic response to induction therapy, overall response rate, and disease-free survival. Additionally, we will assess patients for early evidence of resistance to targeted therapy in terms of activation of alternative signaling pathways and for identification of resistance clones in remnant cell populations.
Collapse
Affiliation(s)
- Caroline E McCoach
- Division of Medical Oncology, Department of Medicine, University of Colorado, Denver, Aurora, CO.
| | - Trever G Bivona
- Division of Hematology and Oncology, Department of Medicine, University of California, San Francisco, San Francisco, CA; Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA
| | - Collin M Blakely
- Division of Hematology and Oncology, Department of Medicine, University of California, San Francisco, San Francisco, CA
| | - Robert C Doebele
- Division of Medical Oncology, Department of Medicine, University of Colorado, Denver, Aurora, CO
| |
Collapse
|
231
|
Abstract
The ROS1 gene belongs to the sevenless subfamily of tyrosine kinase insulin receptor genes. A literature review identified a ROS1 fusion in 2.54% of the patients with lung adenocarcinoma and even higher frequencies in spitzoid neoplasms and inflammatory myofibroblastic tumors. At present, 26 genes were found to fuse with ROS1, some of them already known to fuse with RET and ALK. All the fusion proteins retain the ROS1 kinase domain, but rarely its transmembrane domain. Most of the partners have dimerization domains that are retained in the fusion, presumably leading to constitutive ROS1 tyrosine kinase activation. Some partners have transmembrane domains that are retained or not in the chimeric proteins. Therefore, different ROS1 fusions have distinct subcellular localization, suggesting that they may activate different substrates in vivo.
Collapse
Affiliation(s)
- Arnaud Uguen
- Faculté de Médecine et des Sciences de la Santé, Université de Brest, Brest, France.,Institut National de la Santé et de la Recherche Médicale (INSERM), U1078, Brest, France.,Service d'Anatomie et Cytologie Pathologiques, Hôpital Morvan, CHRU Brest, Brest, France
| | - Marc De Braekeleer
- Faculté de Médecine et des Sciences de la Santé, Université de Brest, Brest, France.,Institut National de la Santé et de la Recherche Médicale (INSERM), U1078, Brest, France.,Service de Cytogénétique et Biologie de la Reproduction, Hôpital Morvan, CHRU Brest, Brest, France
| |
Collapse
|
232
|
Lim SM, Yoo JE, Lim KH, Meng Tai DW, Cho BC, Park YN. Rare Incidence of ROS1 Rearrangement in Cholangiocarcinoma. Cancer Res Treat 2016; 49:185-192. [PMID: 27121721 PMCID: PMC5266400 DOI: 10.4143/crt.2015.497] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 04/02/2016] [Indexed: 12/23/2022] Open
Abstract
Purpose The recent discovery and characterization of an oncogenic ROS1 gene rearrangement has raised significant interest because small molecule inhibitors are effective in these tumors. The aim of this study was to determine frequency and clinicopathological features associated with ROS1 rearrangement in patients with cholangiocarcinoma (CCA). Materials and Methods A total of 261 patients who underwent surgery for CCA between October 1997 and August 2013 were identified from an international, multi-institutional database. ROS1 rearrangement was evaluated by break-apart fluorescence in situ hybridization using tissue microarrays of these patients. Results Of 261 CCA evaluated, three cases (1.1%) showed ROS1 rearrangement by fluorescence in situ hybridization (FISH), all of which were derived from intrahepatic origin. ROS1 protein expression was observed in 38 samples (19.1%). Significantly larger tumor size was observed in ROS1 immunohistochemistry (IHC)–negative patients compared with ROS1 IHC–positive patients. ROS1 FISH–positive patients had a single tumor with a median size of 4 cm and well-to-moderate differentiation. Overall, there was no difference in terms of baseline characteristics, overall survival, and recurrence-free survival between ROS1-positive and -negative patients. Conclusion ROS1 rearrangement was detected in 1.1% of CCA patients. Although rare, conduct of clinical trials using ROS1 inhibitors in these genetically unique patients is warranted.
Collapse
Affiliation(s)
- Sun Min Lim
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea.,Division of Medical Oncology, Department of Internal Medicine, CHA Bundang Medical Center, CHA University, Seongnam, Korea
| | - Jeong Eun Yoo
- Department of Pathology, Yonsei University College of Medicine, Seoul, Korea
| | - Kiat Hon Lim
- Department of Pathology, Singapore General Hospital, Singapore
| | - David Wai Meng Tai
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - Byoung Chul Cho
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Young Nyun Park
- Department of Pathology, Yonsei University College of Medicine, Seoul, Korea.,BK21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea.,Integrated Genomic Research Center for Metabolic Regulation, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
233
|
Dziadziuszko R, Le AT, Wrona A, Jassem J, Camidge DR, Varella-Garcia M, Aisner DL, Doebele RC. An Activating KIT Mutation Induces Crizotinib Resistance in ROS1-Positive Lung Cancer. J Thorac Oncol 2016; 11:1273-1281. [PMID: 27068398 DOI: 10.1016/j.jtho.2016.04.001] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 03/09/2016] [Accepted: 04/01/2016] [Indexed: 01/29/2023]
Abstract
INTRODUCTION Patients with non-small cell lung cancer (NSCLC) harboring ROS proto-oncogene 1, receptor tyrosine kinase gene (ROS1) chromosomal rearrangements benefit from treatment with the ROS1 inhibitor crizotinib. Limited data exist on the spectrum of resistance mechanisms in ROS1-positive NSCLC. To delineate mechanisms of acquired resistance, we analyzed biopsy samples of tumor lesions that progressed while patients were receiving crizotinib. METHODS An activating mutation in the KIT proto-oncogene receptor tyrosine kinase (KIT) (p.D816G) was identified by SNaPshot sequencing in a tumor sample from a patient with ROS1-positive NSCLC identified by fluorescence in situ hybridization whose disease progressed after initial response to crizotinib. In vitro studies included evaluation of KIT mRNA expression by quantitative reverse-transcriptase polymerase chain reactions, transduction of Ba/F3 cells and NSCLC cell lines with KIT-expressing lentiviral plasmids, immunoblotting, and cellular proliferation assays. RESULTS KIT(D816G) is an activating mutation that induces autophosphorylation and cell proliferation. Expression of the mutant KIT(D816G) receptor in ROS1-positive NSCLC cell lines led to constitutively activated KIT as measured by phosphorylation of the KIT receptor. Expression of the KIT(D816G) rendered the HCC78 and CUTO2 cell lines resistant to crizotinib, and only dual inhibition of ROS1 and KIT with crizotinib and ponatinib could resensitize the cells to inhibition of proliferation. The oncogenic switch observed in ROS1-positive cell lines was not immediate and required pharmacologic inactivation of ROS1. CONCLUSIONS Activation of KIT by a gain-of-function somatic mutation is a novel mechanism of resistance to crizotinib in ROS1-rearranged NSCLC. This bypass signaling pathway serves as a ROS1-independent mechanism of resistance, similarly to previously identified epidermal growth factor receptor or Kirsten rat sarcoma viral oncogene homolog/neuroblastoma RAS viral oncogene homolog signaling pathways, and can potentially be targeted by KIT inhibitors.
Collapse
Affiliation(s)
- Rafal Dziadziuszko
- Department of Oncology and Radiotherapy, Medical University of Gdańsk, Gdańsk, Poland
| | - Anh T Le
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine, University of Colorado, Aurora, Colorado
| | - Anna Wrona
- Department of Oncology and Radiotherapy, Medical University of Gdańsk, Gdańsk, Poland
| | - Jacek Jassem
- Department of Oncology and Radiotherapy, Medical University of Gdańsk, Gdańsk, Poland
| | - D Ross Camidge
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine, University of Colorado, Aurora, Colorado
| | - Marileila Varella-Garcia
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine, University of Colorado, Aurora, Colorado; Department of Pathology, University of Colorado School of Medicine, University of Colorado, Aurora, Colorado
| | - Dara L Aisner
- Department of Pathology, University of Colorado School of Medicine, University of Colorado, Aurora, Colorado
| | - Robert C Doebele
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine, University of Colorado, Aurora, Colorado.
| |
Collapse
|
234
|
Lai Z, Markovets A, Ahdesmaki M, Chapman B, Hofmann O, McEwen R, Johnson J, Dougherty B, Barrett JC, Dry JR. VarDict: a novel and versatile variant caller for next-generation sequencing in cancer research. Nucleic Acids Res 2016; 44:e108. [PMID: 27060149 PMCID: PMC4914105 DOI: 10.1093/nar/gkw227] [Citation(s) in RCA: 608] [Impact Index Per Article: 67.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2015] [Accepted: 03/22/2016] [Indexed: 12/22/2022] Open
Abstract
Accurate variant calling in next generation sequencing (NGS) is critical to understand cancer genomes better. Here we present VarDict, a novel and versatile variant caller for both DNA- and RNA-sequencing data. VarDict simultaneously calls SNV, MNV, InDels, complex and structural variants, expanding the detected genetic driver landscape of tumors. It performs local realignments on the fly for more accurate allele frequency estimation. VarDict performance scales linearly to sequencing depth, enabling ultra-deep sequencing used to explore tumor evolution or detect tumor DNA circulating in blood. In addition, VarDict performs amplicon aware variant calling for polymerase chain reaction (PCR)-based targeted sequencing often used in diagnostic settings, and is able to detect PCR artifacts. Finally, VarDict also detects differences in somatic and loss of heterozygosity variants between paired samples. VarDict reprocessing of The Cancer Genome Atlas (TCGA) Lung Adenocarcinoma dataset called known driver mutations in KRAS, EGFR, BRAF, PIK3CA and MET in 16% more patients than previously published variant calls. We believe VarDict will greatly facilitate application of NGS in clinical cancer research.
Collapse
Affiliation(s)
- Zhongwu Lai
- Oncology iMed, AstraZeneca, Waltham, MA 02451, USA
| | | | | | - Brad Chapman
- Bioinformatics Core, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Oliver Hofmann
- Bioinformatics Core, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Bearsden Glasgow, G61 1QH, UK
| | | | | | | | | | | |
Collapse
|
235
|
Zhao W, Choi YL, Song JY, Zhu Y, Xu Q, Zhang F, Jiang L, Cheng J, Zheng G, Mao M. ALK, ROS1 and RET rearrangements in lung squamous cell carcinoma are very rare. Lung Cancer 2016; 94:22-7. [DOI: 10.1016/j.lungcan.2016.01.011] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 01/14/2016] [Accepted: 01/18/2016] [Indexed: 01/11/2023]
|
236
|
Tiash S, Chua MJ, Chowdhury EH. Knockdown of ROS1 gene sensitizes breast tumor growth to doxorubicin in a syngeneic mouse model. Int J Oncol 2016; 48:2359-66. [PMID: 27035628 DOI: 10.3892/ijo.2016.3452] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 01/05/2016] [Indexed: 11/06/2022] Open
Abstract
Treatment of breast cancer, the second leading cause of female deaths worldwide, with classical drugs is often accompanied by treatment failure and relapse of disease condition. Development of chemoresistance and drug toxicity compels compromising the drug concentration below the threshold level with the consequence of therapeutic inefficacy. Moreover, amplification and over-activation of proto-oncogenes in tumor cells make the treatment more challenging. The oncogene, ROS1 which is highly expressed in diverse types of cancers including breast carcinoma, functions as a survival protein aiding cancer progression. Thus we speculated that selective silencing of ROS1 gene by carrier-mediated delivery of siRNA might sensitize the cancer cells to the classical drugs at a relatively low concentration. In this investigation we showed that intracellular delivery of c-ROS1-targeting siRNA using pH-sensitive inorganic nanoparticles of carbonate apatite sensitizes mouse breast cancer cells (4T1) to doxorubicin, but not to cisplatin or paclitaxel, with the highest enhancement in chemosensitivity obtained at 40 nM of the drug concentration. Although intravenous administrations of ROS1-loaded nanoparticles reduced growth of the tumor, a further substantial effect on growth retardation was noted when the mice were treated with the siRNA- and Dox-bound particles, thus suggesting that silencing of ROS1 gene could sensitize the mouse breast cancer cells both in vitro and in vivo to doxorubicin as a result of synergistic effect of the gene knockdown and the drug action, eventually preventing activation of the survival pathway protein, AKT1. Our findings therefore provide valuable insight into the potential cross-talk between the pathways of ROS1 and doxorubicin for future development of effective therapeutics for breast cancer.
Collapse
Affiliation(s)
- Snigdha Tiash
- Advanced Engineering Platform (AEP) and Jeffrey Cheah School of Medicine and Health Sciences, Monash University (Sunway campus), Petaling Jaya, Selangor 46150, Malaysia
| | - Ming Jang Chua
- Advanced Engineering Platform (AEP) and Jeffrey Cheah School of Medicine and Health Sciences, Monash University (Sunway campus), Petaling Jaya, Selangor 46150, Malaysia
| | - Ezharul Hoque Chowdhury
- Advanced Engineering Platform (AEP) and Jeffrey Cheah School of Medicine and Health Sciences, Monash University (Sunway campus), Petaling Jaya, Selangor 46150, Malaysia
| |
Collapse
|
237
|
Mendes R, Carreira B, Baptista PV, Fernandes AR. Non-small cell lung cancer biomarkers and targeted therapy - two faces of the same coin fostered by nanotechnology. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2016. [DOI: 10.1080/23808993.2016.1159914] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
238
|
EGFR, KRAS and ROS1 variants coexist in a lung adenocarcinoma patient. Lung Cancer 2016; 95:94-7. [PMID: 27040858 DOI: 10.1016/j.lungcan.2016.03.005] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 03/02/2016] [Accepted: 03/11/2016] [Indexed: 01/11/2023]
Abstract
The c-ros oncogene 1 (ROS1) fusion is almost mutually exclusive to epidermal growth factor receptor (EGFR), anaplastic lymphoma kinase (ALK) or Kirsten rat sarcoma viral oncogene homolog (KRAS) mutation in non-small cell lung cancer (NSCLC), and it is not seen in the literature for patients to exhibit three mutations. The present study reported a case of a 53-year-old male diagnosed with adenocarcinoma, exhibiting combined EGFR, KRAS mutations and ROS1 rearrangement. At the first line therapy, the patient was treated with crizotinib because of the KRAS mutation that is a known resistant factor of EGFR-TKI resistance, but no responsive. At the second line therapy, EGFR-TKI Icotinib revealed a good response until now. To the best of to our knowledge, this is the first case report of a patient with concurrent EGFR, KRAS mutations and ROS1 fusion. This patient had an excellent response to Icotinib but not crizotinib, suggesting that the EGFR mutation was the oncogenic driver but ROS1 fusion and KRAS mutation not.
Collapse
|
239
|
Inoue M, Toki H, Matsui J, Togashi Y, Dobashi A, Fukumura R, Gondo Y, Minowa O, Tanaka N, Mori S, Takeuchi K, Noda T. Mouse models for ROS1-fusion-positive lung cancers and their application to the analysis of multikinase inhibitor efficiency. Carcinogenesis 2016; 37:452-60. [PMID: 26964870 DOI: 10.1093/carcin/bgw028] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2015] [Accepted: 02/16/2016] [Indexed: 12/16/2022] Open
Abstract
ROS1-fusion genes, resulting from chromosomal rearrangement, have been reported in 1-2% of human non-small cell lung cancer cases. More than 10 distinct ROS1-fusion genes, including break-point variants, have been identified to date. In this study, to investigate the in vivo oncogenic activities of one of the most frequently detected fusions, CD74-ROS1, as well as another SDC4-ROS1 fusion that has also been reported in several studies, we generated transgenic (TG) mouse strains that express either of the two ROS1-fusion genes specifically in lung alveolar type II cells. Mice in all TG lines developed tumorigenic nodules in the lung, and a few strains of both TG mouse lines demonstrated early-onset nodule development (multiple tumor lesions present in the lung at 2-4 weeks after birth); therefore, these two strains were selected for further investigation. Tumors developed progressively in the untreated TG mice of both lines, whereas those receiving oral administration of an ALK/MET/ROS1 inhibitor, crizotinib, and an ALK/ROS1 inhibitor, ASP3026, showed marked reduction in the tumor burden. Collectively, these data suggest that each of these two ROS1-fusion genes acts as a driver for the pathogenesis of lung adenocarcinoma in vivo The TG mice developed in this study are expected to serve as valuable tools for exploring novel therapeutic agents against ROS1-fusion-positive lung cancer.
Collapse
Affiliation(s)
- Maki Inoue
- Team for Advanced Development and Evaluation of Human Disease Models, RIKEN BioResource Center, Tsukuba 305-0074, Japan, Division of Cell Biology
| | - Hideaki Toki
- Team for Advanced Development and Evaluation of Human Disease Models, RIKEN BioResource Center, Tsukuba 305-0074, Japan
| | - Junko Matsui
- Team for Advanced Development and Evaluation of Human Disease Models, RIKEN BioResource Center, Tsukuba 305-0074, Japan
| | - Yuki Togashi
- Pathology Project for Molecular Targets and Division of Pathology, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan
| | | | - Ryutaro Fukumura
- Mutagenesis and Genomics Team, RIKEN BioResource Center, Tsukuba 305-0074, Japan and
| | - Yoichi Gondo
- Mutagenesis and Genomics Team, RIKEN BioResource Center, Tsukuba 305-0074, Japan and
| | - Osamu Minowa
- Team for Advanced Development and Evaluation of Human Disease Models, RIKEN BioResource Center, Tsukuba 305-0074, Japan
| | - Norio Tanaka
- Project for Development of Innovative Research on Cancer Therapeutics, The Genome Center, Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan
| | - Seiichi Mori
- Project for Development of Innovative Research on Cancer Therapeutics, The Genome Center, Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan
| | - Kengo Takeuchi
- Pathology Project for Molecular Targets and Division of Pathology, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan
| | - Tetsuo Noda
- Team for Advanced Development and Evaluation of Human Disease Models, RIKEN BioResource Center, Tsukuba 305-0074, Japan, Division of Cell Biology,
| |
Collapse
|
240
|
Tomasini P, Fina F, Greillier L, Barlesi F. New oncogenes drivers in lung cancer—new therapeutic targets. CURRENT PULMONOLOGY REPORTS 2016. [DOI: 10.1007/s13665-016-0137-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
241
|
Sun H, Chen P, Li D, Li Y, Hou T. Directly Binding Rather than Induced-Fit Dominated Binding Affinity Difference in (S)- and (R)-Crizotinib Bound MTH1. J Chem Theory Comput 2016; 12:851-60. [DOI: 10.1021/acs.jctc.5b00973] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
| | | | | | - Youyong Li
- Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, Suzhou, Jiangsu 215123, China
| | | |
Collapse
|
242
|
Cargnelutti M, Corso S, Pergolizzi M, Mévellec L, Aisner DL, Dziadziuszko R, Varella-Garcia M, Comoglio PM, Doebele RC, Vialard J, Giordano S. Activation of RAS family members confers resistance to ROS1 targeting drugs. Oncotarget 2016; 6:5182-94. [PMID: 25691052 PMCID: PMC4467141 DOI: 10.18632/oncotarget.3311] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 12/29/2014] [Indexed: 01/09/2023] Open
Abstract
The ROS1 tyrosine kinase is activated in lung cancer as a consequence of chromosomal rearrangement. Although high response rates and disease control have been observed in lung cancer patients bearing rearranged ROS1 tumors (ROS1+) treated with the kinase inhibitor crizotinib, many of these patients eventually relapse.To identify mechanisms of resistance to ROS1 inhibitors we generated resistant cells from HCC78 lung cancer cells bearing the SLC34A2-ROS1 rearrangement. We found that activation of the RAS pathway in the HCC78 cell model, due to either KRAS/NRAS mutations or to KRAS amplification, rendered the cells resistant to ROS1 inhibition. These cells were cross-resistant to different ROS1 inhibitors, but sensitive to inhibitors of the RAS signaling pathway. Interestingly, we identified focal KRAS amplification in a biopsy of a tumor from a patient that had become resistant to crizotinib treatment.Altogether our data suggest that the activation of members of the RAS family can confer resistance to ROS1 inhibitors. This has important clinical implications as: (i) RAS genetic alterations in ROS1+ primary tumors are likely negative predictors of efficacy for targeted drugs and (ii) this kind of resistance is unlikely to be overcome by the use of more specific or more potent ROS1 targeting drugs.
Collapse
Affiliation(s)
| | - Simona Corso
- Candiolo Cancer Institute - FPO, IRCCS, Torino, Italy.,Department of Oncology, University of Torino, Italy
| | | | | | - Dara L Aisner
- Department of Pathology, University of Colorado School of Medicine, Aurora, CO, USA
| | | | - Marileila Varella-Garcia
- Department of Pathology, University of Colorado School of Medicine, Aurora, CO, USA.,Department of Medicine, Division of Medical Oncology University of Colorado School of Medicine, Aurora, CO, USA
| | - Paolo M Comoglio
- Candiolo Cancer Institute - FPO, IRCCS, Torino, Italy.,Department of Oncology, University of Torino, Italy
| | - Robert C Doebele
- Department of Medicine, Division of Medical Oncology University of Colorado School of Medicine, Aurora, CO, USA
| | - Jorge Vialard
- Janssen Research & Development, a division of Janssen Pharmaceutica NV, Beerse, Belgium
| | - Silvia Giordano
- Candiolo Cancer Institute - FPO, IRCCS, Torino, Italy.,Department of Oncology, University of Torino, Italy
| |
Collapse
|
243
|
Abstract
Lung cancer is the leading cause of cancer-related deaths in United States, accounting for more than one-fourth of the deaths annually. Although comparatively rare and relatively less studied, genetic abnormalities other than epidermal growth factor receptor (EGFR) mutations, anaplastic lymphoma kinase (ALK) rearrangements, and Kirsten rat sarcoma (KRAS) mutations account for significant proportion of the driver mutations identified thus far. The targeted agents against B-rapidly accelerated fibrosarcoma (BRAF) V600E mutation, MNNG-HOS transforming gene (MET) pathway, ROS1 rearrangement, rearranged during transfection (RET) rearrangement, and HER2 pathways offer promising therapeutic options. Recruiting patients with these rarer mutations to well-designed, large multicenter trials to further validate the use of targeted agents remains a challenge. The clinical data and ongoing trials with these agents are reviewed in this article.
Collapse
Affiliation(s)
- Nabin Khanal
- a Department of Internal Medicine , Creighton University Medical Center , Omaha , NE , USA
| | - Apar Kishor Ganti
- b Division of Oncology-Hematology, Department of Internal Medicine , VA-Nebraska Western Iowa Health Care System , Omaha , NE , USA.,c Division of Oncology-Hematology, Department of Internal Medicine , University of Nebraska Medical Center , Omaha , NE , USA
| |
Collapse
|
244
|
Awad MM, Oxnard GR, Jackman DM, Savukoski DO, Hall D, Shivdasani P, Heng JC, Dahlberg SE, Jänne PA, Verma S, Christensen J, Hammerman PS, Sholl LM. MET Exon 14 Mutations in Non-Small-Cell Lung Cancer Are Associated With Advanced Age and Stage-Dependent MET Genomic Amplification and c-Met Overexpression. J Clin Oncol 2016; 34:721-30. [PMID: 26729443 DOI: 10.1200/jco.2015.63.4600] [Citation(s) in RCA: 512] [Impact Index Per Article: 56.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
PURPOSE Non-small-cell lung cancers (NSCLCs) harboring mutations in MET exon 14 and its flanking introns may respond to c-Met inhibitors. We sought to describe the clinical, pathologic, and genomic characteristics of patients with cancer with MET exon 14 mutations. PATIENTS AND METHODS We interrogated next-generation sequencing results from 6,376 cancers to identify those harboring MET exon 14 mutations. Clinical characteristics of MET exon 14 mutated NSCLCs were compared with those of NSCLCs with activating mutations in KRAS and EGFR. Co-occurring genomic mutations and copy number alterations were identified. c-Met immunohistochemistry and real-time polymerase chain reaction to detect exon 14 skipping were performed where sufficient tissue was available. RESULTS MET exon 14 mutations were identified in 28 of 933 nonsquamous NSCLCs (3.0%) and were not seen in other cancer types in this study. Patients with MET exon 14-mutated NSCLC were significantly older (median age, 72.5 years) than patients with EGFR-mutant (median age, 61 years; P < .001) or KRAS-mutant NSCLC (median age, 65 years; P < .001). Among patients with MET exon 14 mutations, 68% were women, and 36% were never-smokers. Stage IV MET exon 14-mutated NSCLCs were significantly more likely to have concurrent MET genomic amplification (mean ratio of MET to chromosome 7, 4.3) and strong c-Met immunohistochemical expression (mean H score, 253) than stage IA to IIIB MET exon 14-mutated NSCLCs (mean ratio of MET to chromosome 7, 1.4; P = .007; mean H score, 155; P = .002) and stage IV MET exon 14-wild-type NSCLCs (mean ratio of MET to chromosome 7, 1.2; P < .001; mean H score, 142; P < .001). A patient whose lung cancer harbored a MET exon 14 mutation with concurrent genomic amplification of the mutated MET allele experienced a major partial response to the c-Met inhibitor crizotinib. CONCLUSION MET exon 14 mutations represent a clinically unique molecular subtype of NSCLC. Prospective clinical trials with c-Met inhibitors will be necessary to validate MET exon 14 mutations as an important therapeutic target in NSCLC.
Collapse
Affiliation(s)
- Mark M Awad
- Mark M. Awad, Geoffrey R. Oxnard, David M. Jackman, Jennifer C. Heng, Suzanne E. Dahlberg, Pasi A. Jänne, and Peter S. Hammerman, Dana-Farber Cancer Institute; Mark M. Awad, Geoffrey R. Oxnard, David M. Jackman, Daniel O. Savukoski, Dimity Hall, Priyanka Shivdasani, Pasi A. Jänne, Peter S. Hammerman, and Lynette M. Sholl, Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Suman Verma, ResearchDX, Irvine; and James Christensen, Mirati Therapeutics, San Diego, CA.
| | - Geoffrey R Oxnard
- Mark M. Awad, Geoffrey R. Oxnard, David M. Jackman, Jennifer C. Heng, Suzanne E. Dahlberg, Pasi A. Jänne, and Peter S. Hammerman, Dana-Farber Cancer Institute; Mark M. Awad, Geoffrey R. Oxnard, David M. Jackman, Daniel O. Savukoski, Dimity Hall, Priyanka Shivdasani, Pasi A. Jänne, Peter S. Hammerman, and Lynette M. Sholl, Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Suman Verma, ResearchDX, Irvine; and James Christensen, Mirati Therapeutics, San Diego, CA
| | - David M Jackman
- Mark M. Awad, Geoffrey R. Oxnard, David M. Jackman, Jennifer C. Heng, Suzanne E. Dahlberg, Pasi A. Jänne, and Peter S. Hammerman, Dana-Farber Cancer Institute; Mark M. Awad, Geoffrey R. Oxnard, David M. Jackman, Daniel O. Savukoski, Dimity Hall, Priyanka Shivdasani, Pasi A. Jänne, Peter S. Hammerman, and Lynette M. Sholl, Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Suman Verma, ResearchDX, Irvine; and James Christensen, Mirati Therapeutics, San Diego, CA
| | - Daniel O Savukoski
- Mark M. Awad, Geoffrey R. Oxnard, David M. Jackman, Jennifer C. Heng, Suzanne E. Dahlberg, Pasi A. Jänne, and Peter S. Hammerman, Dana-Farber Cancer Institute; Mark M. Awad, Geoffrey R. Oxnard, David M. Jackman, Daniel O. Savukoski, Dimity Hall, Priyanka Shivdasani, Pasi A. Jänne, Peter S. Hammerman, and Lynette M. Sholl, Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Suman Verma, ResearchDX, Irvine; and James Christensen, Mirati Therapeutics, San Diego, CA
| | - Dimity Hall
- Mark M. Awad, Geoffrey R. Oxnard, David M. Jackman, Jennifer C. Heng, Suzanne E. Dahlberg, Pasi A. Jänne, and Peter S. Hammerman, Dana-Farber Cancer Institute; Mark M. Awad, Geoffrey R. Oxnard, David M. Jackman, Daniel O. Savukoski, Dimity Hall, Priyanka Shivdasani, Pasi A. Jänne, Peter S. Hammerman, and Lynette M. Sholl, Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Suman Verma, ResearchDX, Irvine; and James Christensen, Mirati Therapeutics, San Diego, CA
| | - Priyanka Shivdasani
- Mark M. Awad, Geoffrey R. Oxnard, David M. Jackman, Jennifer C. Heng, Suzanne E. Dahlberg, Pasi A. Jänne, and Peter S. Hammerman, Dana-Farber Cancer Institute; Mark M. Awad, Geoffrey R. Oxnard, David M. Jackman, Daniel O. Savukoski, Dimity Hall, Priyanka Shivdasani, Pasi A. Jänne, Peter S. Hammerman, and Lynette M. Sholl, Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Suman Verma, ResearchDX, Irvine; and James Christensen, Mirati Therapeutics, San Diego, CA
| | - Jennifer C Heng
- Mark M. Awad, Geoffrey R. Oxnard, David M. Jackman, Jennifer C. Heng, Suzanne E. Dahlberg, Pasi A. Jänne, and Peter S. Hammerman, Dana-Farber Cancer Institute; Mark M. Awad, Geoffrey R. Oxnard, David M. Jackman, Daniel O. Savukoski, Dimity Hall, Priyanka Shivdasani, Pasi A. Jänne, Peter S. Hammerman, and Lynette M. Sholl, Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Suman Verma, ResearchDX, Irvine; and James Christensen, Mirati Therapeutics, San Diego, CA
| | - Suzanne E Dahlberg
- Mark M. Awad, Geoffrey R. Oxnard, David M. Jackman, Jennifer C. Heng, Suzanne E. Dahlberg, Pasi A. Jänne, and Peter S. Hammerman, Dana-Farber Cancer Institute; Mark M. Awad, Geoffrey R. Oxnard, David M. Jackman, Daniel O. Savukoski, Dimity Hall, Priyanka Shivdasani, Pasi A. Jänne, Peter S. Hammerman, and Lynette M. Sholl, Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Suman Verma, ResearchDX, Irvine; and James Christensen, Mirati Therapeutics, San Diego, CA
| | - Pasi A Jänne
- Mark M. Awad, Geoffrey R. Oxnard, David M. Jackman, Jennifer C. Heng, Suzanne E. Dahlberg, Pasi A. Jänne, and Peter S. Hammerman, Dana-Farber Cancer Institute; Mark M. Awad, Geoffrey R. Oxnard, David M. Jackman, Daniel O. Savukoski, Dimity Hall, Priyanka Shivdasani, Pasi A. Jänne, Peter S. Hammerman, and Lynette M. Sholl, Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Suman Verma, ResearchDX, Irvine; and James Christensen, Mirati Therapeutics, San Diego, CA
| | - Suman Verma
- Mark M. Awad, Geoffrey R. Oxnard, David M. Jackman, Jennifer C. Heng, Suzanne E. Dahlberg, Pasi A. Jänne, and Peter S. Hammerman, Dana-Farber Cancer Institute; Mark M. Awad, Geoffrey R. Oxnard, David M. Jackman, Daniel O. Savukoski, Dimity Hall, Priyanka Shivdasani, Pasi A. Jänne, Peter S. Hammerman, and Lynette M. Sholl, Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Suman Verma, ResearchDX, Irvine; and James Christensen, Mirati Therapeutics, San Diego, CA
| | - James Christensen
- Mark M. Awad, Geoffrey R. Oxnard, David M. Jackman, Jennifer C. Heng, Suzanne E. Dahlberg, Pasi A. Jänne, and Peter S. Hammerman, Dana-Farber Cancer Institute; Mark M. Awad, Geoffrey R. Oxnard, David M. Jackman, Daniel O. Savukoski, Dimity Hall, Priyanka Shivdasani, Pasi A. Jänne, Peter S. Hammerman, and Lynette M. Sholl, Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Suman Verma, ResearchDX, Irvine; and James Christensen, Mirati Therapeutics, San Diego, CA
| | - Peter S Hammerman
- Mark M. Awad, Geoffrey R. Oxnard, David M. Jackman, Jennifer C. Heng, Suzanne E. Dahlberg, Pasi A. Jänne, and Peter S. Hammerman, Dana-Farber Cancer Institute; Mark M. Awad, Geoffrey R. Oxnard, David M. Jackman, Daniel O. Savukoski, Dimity Hall, Priyanka Shivdasani, Pasi A. Jänne, Peter S. Hammerman, and Lynette M. Sholl, Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Suman Verma, ResearchDX, Irvine; and James Christensen, Mirati Therapeutics, San Diego, CA
| | - Lynette M Sholl
- Mark M. Awad, Geoffrey R. Oxnard, David M. Jackman, Jennifer C. Heng, Suzanne E. Dahlberg, Pasi A. Jänne, and Peter S. Hammerman, Dana-Farber Cancer Institute; Mark M. Awad, Geoffrey R. Oxnard, David M. Jackman, Daniel O. Savukoski, Dimity Hall, Priyanka Shivdasani, Pasi A. Jänne, Peter S. Hammerman, and Lynette M. Sholl, Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Suman Verma, ResearchDX, Irvine; and James Christensen, Mirati Therapeutics, San Diego, CA
| |
Collapse
|
245
|
Lung Cancer Genomics in the Era of Accelerated Targeted Drug Development. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 890:1-23. [PMID: 26703796 DOI: 10.1007/978-3-319-24932-2_1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Lung cancer is the leading cause of cancer-related deaths in the United States and the 5-year overall survival outlook for a patient has not improved in several decades. Recently, however, molecular and genomic profiling of the lung tumors has revealed recurring somatic mutations. As a result the therapeutic landscape of lung cancer is undergoing a paradigm shift from a purely histology-based understanding of the disease to subtype distinctions based on tumor genetics, which has launched cancer-specific, mechanism-based targeted therapies with clear benefit to patients. While targeted therapy advancements are being made at an ever increasing rate, a new challenge in the form of drug resistance has also emerged. This review summarizes the current literature for these issues.
Collapse
|
246
|
Rodriguez-Canales J, Parra-Cuentas E, Wistuba II. Diagnosis and Molecular Classification of Lung Cancer. Cancer Treat Res 2016; 170:25-46. [PMID: 27535388 DOI: 10.1007/978-3-319-40389-2_2] [Citation(s) in RCA: 166] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Lung cancer is a complex disease composed of diverse histological and molecular types with clinical relevance. The advent of large-scale molecular profiling has been helpful to identify novel molecular targets that can be applied to the treatment of particular lung cancer patients and has helped to reshape the pathological classification of lung cancer. Novel directions include the immunotherapy revolution, which has opened the door for new opportunities for cancer therapy and is also redefining the classification of multiple tumors, including lung cancer. In the present chapter, we will review the main current basis of the pathological diagnosis and classification of lung cancer incorporating the histopathological and molecular dimensions of the disease.
Collapse
Affiliation(s)
- Jaime Rodriguez-Canales
- Department of Translational Molecular Pathology, Unit 951, The University of Texas MD Anderson Cancer Center, 2130 Holcombe Blvd., Houston, TX, 77030, USA
| | - Edwin Parra-Cuentas
- Department of Translational Molecular Pathology, Unit 951, The University of Texas MD Anderson Cancer Center, 2130 Holcombe Blvd., Houston, TX, 77030, USA
| | - Ignacio I Wistuba
- Department of Translational Molecular Pathology, Unit 951, The University of Texas MD Anderson Cancer Center, 2130 Holcombe Blvd., Houston, TX, 77030, USA.
| |
Collapse
|
247
|
Emerging Biomarkers in Personalized Therapy of Lung Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 890:25-36. [DOI: 10.1007/978-3-319-24932-2_2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
248
|
Cao B, Wei P, Liu Z, Bi R, Lu Y, Zhang L, Zhang J, Yang Y, Shen C, Du X, Zhou X. Detection of lung adenocarcinoma with ROS1 rearrangement by IHC, FISH, and RT-PCR and analysis of its clinicopathologic features. Onco Targets Ther 2015; 9:131-8. [PMID: 26770062 PMCID: PMC4706119 DOI: 10.2147/ott.s94997] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Objective To detect ROS1 rearrangement using three different assays, including immunohistochemistry (IHC), fluorescence in situ hybridization (FISH), and reverse transcription polymerase chain reaction (RT-PCR), and to analyze the clinicopathologic features of ROS1 rearrangement in patients with lung adenocarcinoma. Methods One hundred eighty-three consecutive patients with lung adenocarcinoma with operation and follow-up data were analyzed for ROS1 rearrangement by IHC, FISH, and RT-PCR. PCR products of the RT-PCR-positive samples were sequenced for confirmation of the specific fusion partners. Results Three of the 183 (1.64%) cases were identified to be positive for ROS1 rearrangement through all three methods. The fusion patterns were CD74 e6-ROS1 e32, CD74 e6-ROS1 e34, and TPM3 e8-ROS1 e35, respectively. FISH-positive cases showed two types of signals, single 3′ signals (green) and split red and green signals. Using FISH as a standard method, the sensitivity and specificity of ROS1 IHC with 1+ staining or more were 100% and 96.67%, respectively. The sensitivity and specificity of RT-PCR were both 100%. Univariate analysis identified female sex (P=0.044), Stage I disease (P<0.001), and ROS1-negative status (P=0.022) to be significantly associated with longer overall survival. Conclusion IHC, FISH, and RT-PCR are all effective methods for the detection of ROS1 rearrangement. IHC would be a useful screening method in routine pathologic laboratories. RT-PCR can detect exact fusion patterns. ROS1 rearrangement may be a worse prognostic factor. The exact correlation of ROS1 rearrangement with prognosis and whether different fusion types are correlated with different responses to targeted therapy need to be further investigated.
Collapse
Affiliation(s)
- Bing Cao
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China; Institute of Pathology, Fudan University, Shanghai, People's Republic of China
| | - Ping Wei
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China; Institute of Pathology, Fudan University, Shanghai, People's Republic of China
| | - Zebing Liu
- Department of Pathology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, People's Republic of China
| | - Rui Bi
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China; Institute of Pathology, Fudan University, Shanghai, People's Republic of China
| | - Yongming Lu
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China; Institute of Pathology, Fudan University, Shanghai, People's Republic of China
| | - Ling Zhang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China; Institute of Pathology, Fudan University, Shanghai, People's Republic of China
| | - Jing Zhang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China; Institute of Pathology, Fudan University, Shanghai, People's Republic of China
| | - Yusi Yang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China; Institute of Pathology, Fudan University, Shanghai, People's Republic of China
| | - Chen Shen
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China; Institute of Pathology, Fudan University, Shanghai, People's Republic of China
| | - Xiang Du
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China; Institute of Pathology, Fudan University, Shanghai, People's Republic of China
| | - Xiaoyan Zhou
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China; Institute of Pathology, Fudan University, Shanghai, People's Republic of China
| |
Collapse
|
249
|
Loong HH, Mok K, Leung LKS, Mok TSK. Crizotinib in the management of advanced-stage non-small-cell lung cancer. Future Oncol 2015; 11:735-45. [PMID: 25757678 DOI: 10.2217/fon.14.314] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
ABSTRACT Rearrangement of ALK gene has been identified as exerting a potent transforming effect as driver oncogene in patients with non-small-cell lung cancer (NSCLC). Crizotinib is a small-molecule oral inhibitor of ALK, c-Met/HGF receptor and ROS1 receptor kinases. Its efficacy in ALK-rearranged NSCLC has been established. Crizotinib's effect on ROS1 receptor kinases and c-Met with relevance to NSCLC is also actively being explored. Resistance mechanisms such as secondary gatekeeper mutations in ALK gene and activation of other oncogenes have been identified to confer acquired resistance to crizotinib. This article reviews the pharmacological properties of crizotinib, preclinical and clinical results that led to its approval in ALK-positive NSCLC and current directions of clinical research in overcoming crizotinib resistance.
Collapse
Affiliation(s)
- Herbert H Loong
- Department of Clinical Oncology, State Key Laboratory in Oncology in South China, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
| | | | | | | |
Collapse
|
250
|
Abstract
INTRODUCTION Patients with non-small cell lung cancer (NSCLC) harboring anaplastic lymphoma kinase (ALK) rearrangement selectively respond to ALK inhibitors. Thus, identification of ALK rearrangements has become a standard diagnostic test in advanced NSCLC patients. Our institution has been a referral center in Spain for ALK determination by Fluorescent in situ hybridization (FISH). The aim of our study was to assess the feasibility and the FISH patterns of the ALK gene and to evaluate the clinical and pathological features of patients with ALK alterations. METHODS Between 2010 and 2014, 1092 samples were evaluated for ALK using FISH technique (927 histological samples, 165 cytological samples). Correlation with available clinical-pathological information was assessed. RESULTS ALK rearrangement was found in 35 patients (3.2%). Cytological samples (using either direct smears or cell blocks), were more frequently non-assessable than histological samples (69% versus 89%, respectively) (p < 0.001). Within the ALK-rearranged cases the majority were female, non-smokers, and stage IV. CONCLUSIONS Although assessable in cytological samples, biopsies are preferred when available for ALK evaluation by FISH. The ALK translocation prevalence and the associated clinico-pathological features in Spanish NSCLC patients are similar to those previously reported.
Collapse
|