2101
|
Lee J, Kim HC, Hong JY, Wang K, Kim SY, Jang J, Kim ST, Park JO, Lim HY, Kang WK, Park YS, Lee J, Lee WY, Park YA, Huh JW, Yun SH, Do IG, Kim SH, Balasubramanian S, Stephens PJ, Ross JS, Li GG, Hornby Z, Ali SM, Miller VA, Kim KM, Ou SHI. Detection of novel and potentially actionable anaplastic lymphoma kinase (ALK) rearrangement in colorectal adenocarcinoma by immunohistochemistry screening. Oncotarget 2016; 6:24320-32. [PMID: 26172300 PMCID: PMC4695188 DOI: 10.18632/oncotarget.4462] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 06/19/2015] [Indexed: 11/29/2022] Open
Abstract
Purpose Anaplastic lymphoma kinase (ALK) rearrangement has been detected in colorectal carcinoma (CRC) using advanced molecular diagnostics tests including exon scanning, fluorescence in situ hybridization (FISH), and next generation sequencing (NGS). We investigated if immunohistochemistry (IHC) can be used to detect ALK rearrangement in gastrointestinal malignancies. Experimental designs Tissue microarrays (TMAs) from consecutive gastric carcinoma (GC) and CRC patients who underwent surgical resection at Samsung Medical Center, Seoul, Korea were screened by IHC using ALK monoclonal antibody 5A4. IHC positive cases were confirmed by FISH, nCounter assays, and NGS-based comprehensive genomic profiling (CGP). ALK IHC was further applied to CRC patients enrolled in a pathway-directed therapeutic trial. Results Four hundred thirty-two GC and 172 CRC cases were screened by IHC. No GC sample was ALK IHC positive. One CRC (0.6%) was ALK IHC positive (3+) that was confirmed by ALK FISH and a novel CAD-ALK (C35; A20) fusion variant that resulted from a paracentric inversion event inv(2)(p22–21p23) was identified by CGP. One out of 50 CRC patients enrolled in a pathway-directed therapeutic trial was ALK IHC positive (3+) confirmed by ALK FISH and found to harbor the EML4-ALK (E21, A20) fusion variant by CGP. Growth of a tumor cell line derived from this EML4-ALK CRC patient was inhibited by ALK inhibitors crizotinib and entrectinib. Conclusions ALK IHC is a viable screening strategy for identifying ALK rearrangement in CRC. ALK rearrangement is a potential actionable driver mutation in CRC based on survival inhibition of patient tumor-derived cell line by potent ALK inhibitors.
Collapse
Affiliation(s)
- Jeeyun Lee
- Department of Medicine, Division of Hematology-Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Hee Cheol Kim
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jung Yong Hong
- Department of Internal Medicine, Chung-Ang University College of Medicine, Dongjak-Gu, Seoul, Republic of Korea
| | - Kai Wang
- Foundation Medicine Inc, Cambridge, Massachusetts, USA
| | - Sun Young Kim
- Department of Medicine, Division of Hematology-Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jiryeon Jang
- Department of Medicine, Division of Hematology-Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Seung Tae Kim
- Department of Medicine, Division of Hematology-Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Joon Oh Park
- Department of Medicine, Division of Hematology-Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Ho Yeong Lim
- Department of Medicine, Division of Hematology-Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Won Ki Kang
- Department of Medicine, Division of Hematology-Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Young Suk Park
- Department of Medicine, Division of Hematology-Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jiyun Lee
- Department of Medicine, Division of Hematology-Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Woo Yong Lee
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Yoon Ah Park
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jung Wook Huh
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Seong Hyeon Yun
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - In-Gu Do
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Seok Hyung Kim
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | | | | | - Jeffrey S Ross
- Foundation Medicine Inc, Cambridge, Massachusetts, USA.,Department of Pathology and Laboratory Medicine, Albany Medical College, Albany, New York, USA
| | | | | | - Siraj M Ali
- Foundation Medicine Inc, Cambridge, Massachusetts, USA
| | | | - Kyoung-Mee Kim
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.,Innovative Cancer Medicine Institute, Samsung Medical Center, Seoul, Korea
| | - Sai-Hong Ignatius Ou
- Chao Family Comprehensive Cancer Center, University of California Irvine School of Medicine, Orange, California, USA
| |
Collapse
|
2102
|
Mattsson JSM, Brunnström H, Jabs V, Edlund K, Jirström K, Mindus S, la Fleur L, Pontén F, Karlsson MG, Karlsson C, Koyi H, Brandén E, Botling J, Helenius G, Micke P, Svensson MA. Inconsistent results in the analysis of ALK rearrangements in non-small cell lung cancer. BMC Cancer 2016; 16:603. [PMID: 27495736 PMCID: PMC4974795 DOI: 10.1186/s12885-016-2646-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2016] [Accepted: 07/28/2016] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Identification of targetable EML4-ALK fusion proteins has revolutionized the treatment of a minor subgroup of non-small cell lung cancer (NSCLC) patients. Although fluorescence in situ hybridization (FISH) is regarded as the gold standard for detection of ALK rearrangements, ALK immunohistochemistry (IHC) is often used as screening tool in clinical practice. In order to unbiasedly analyze the diagnostic impact of such a screening strategy, we compared ALK IHC with ALK FISH in three large representative Swedish NSCLC cohorts incorporating clinical parameters and gene expression data. METHODS ALK rearrangements were detected using FISH on tissue microarrays (TMAs), including tissue from 851 NSCLC patients. In parallel, ALK protein expression was detected using IHC, applying the antibody clone D5F3 with two different protocols (the FDA approved Ventana CDx assay and our in house Dako IHC protocol). Gene expression microarray data (Affymetrix) was available for 194 patients. RESULTS ALK rearrangements were detected in 1.7 % in the complete cohort and 2.0 % in the non-squamous cell carcinoma subgroup. ALK protein expression was observed in 1.8 and 1.4 % when applying the Ventana assay or the in house Dako protocol, respectively. The specificity and accuracy of IHC was high (> 98 %), while the sensitivity was between 69 % (Ventana) and 62 % (in house Dako protocol). Furthermore, only 67 % of the ALK IHC positive cases were positive with both IHC assays. Gene expression analysis revealed that 6/194 (3 %) tumors showed high ALK gene expression (≥ 6 AU) and of them only three were positive by either FISH or IHC. CONCLUSION The overall frequency of ALK rearrangements based on FISH was lower than previously reported. The sensitivity of both IHC assays was low, and the concordance between the FISH and the IHC assays poor, questioning current strategies to screen with IHC prior to FISH or completely replace FISH by IHC.
Collapse
Affiliation(s)
- Johanna S M Mattsson
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85, Uppsala, Sweden.
| | - Hans Brunnström
- Department of Clinical Sciences Lund, Division of Oncology and Pathology, Lund University, Lund, Sweden.,Department of Pathology, Regional Laboratories Region Skåne, SE-221 85, Lund, Sweden
| | - Verena Jabs
- Department of Statistics, TU Dortmund University, Dortmund, Germany
| | - Karolina Edlund
- Leibniz Research Centre for Working Environment and Human Factors (IfADo) at Dortmund TU, Dortmund, Germany
| | - Karin Jirström
- Department of Clinical Sciences Lund, Division of Oncology and Pathology, Lund University, Lund, Sweden
| | - Stephanie Mindus
- Department of Medical Sciences, Respiratory, Allergy and Sleep Research, Uppsala University, Uppsala, Sweden
| | - Linnéa la Fleur
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85, Uppsala, Sweden
| | - Fredrik Pontén
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85, Uppsala, Sweden
| | - Mats G Karlsson
- Department of Research and Education, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | | | - Hirsh Koyi
- Department of Respiratory Medicine, Gävle hospital, Gävle; Centre for Research and Development, Uppsala University/County Council of Gävleborg, Gävle, Sweden
| | - Eva Brandén
- Department of Respiratory Medicine, Gävle hospital, Gävle; Centre for Research and Development, Uppsala University/County Council of Gävleborg, Gävle, Sweden
| | - Johan Botling
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85, Uppsala, Sweden
| | - Gisela Helenius
- Department of Laboratory Medicine, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Patrick Micke
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85, Uppsala, Sweden
| | - Maria A Svensson
- Clinical Research Center, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| |
Collapse
|
2103
|
Savic S, Bubendorf L. Common Fluorescence In Situ Hybridization Applications in Cytology. Arch Pathol Lab Med 2016; 140:1323-1330. [DOI: 10.5858/arpa.2016-0202-ra] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Context.—
Fluorescence in situ hybridization (FISH) is a well-established method for detection of genomic aberrations in diagnostic, prognostic, and predictive marker testing.
Objective.—
To review common applications of FISH in cytology.
Data Sources.—
The published literature was reviewed.
Conclusions.—
Cytology is particularly well suited for all kinds of FISH applications, which is highlighted in respiratory tract cytology with an increasing demand for predictive FISH testing in lung cancer. Fluorescence in situ hybridization is the gold standard for detection of predictive anaplastic lymphoma kinase gene (ALK) rearrangements, and the same evaluation criteria as in histology apply to cytology. Several other gene rearrangements, including ROS proto-oncogene 1 receptor tyrosine kinase (ROS1), are becoming clinically important and share the same underlining cytogenetic mechanisms with ALK. MET amplification is one of the most common mechanisms of acquired resistance to epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors and can be targeted by crizotinib. As genomic aberrations are a hallmark of malignant cells, FISH is a valuable objective ancillary diagnostic tool. In urinary tract cytology, atypical urothelial cells equivocal for malignancy are a common diagnostic dilemma and multitarget FISH can help clarify such cells. Diagnosis of malignant mesothelioma remains one of the most challenging fields in effusion cytology, and ancillary FISH is useful in establishing the diagnosis. Fluorescence in situ hybridization is a morphology-based technique, and the prerequisite for reliable FISH results is a targeted evaluation of the cells in question (eg, cancer or atypical cells). Cytopathologists and cytotechnicians should therefore be involved in molecular testing in order to select the best material and to provide their morphologic expertise.
Collapse
Affiliation(s)
- Spasenija Savic
- From the Institute of Pathology, University Hospital Basel, Basel, Switzerland
| | | |
Collapse
|
2104
|
Ciccarese C, Massari F, Tortora G. 2015 and human cancer: back to overall survival. Future Oncol 2016; 12:1751-4. [DOI: 10.2217/fon-2015-0085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Chiara Ciccarese
- Medical Oncology, Azienda Ospedaliera Universitaria Integrata, University of Verona, Verona, Italy
| | - Francesco Massari
- Medical Oncology, Azienda Ospedaliera Universitaria Integrata, University of Verona, Verona, Italy
| | - Giampaolo Tortora
- Medical Oncology, Azienda Ospedaliera Universitaria Integrata, University of Verona, Verona, Italy
| |
Collapse
|
2105
|
Dorantes-Heredia R, Ruiz-Morales JM, Cano-García F. Histopathological transformation to small-cell lung carcinoma in non-small cell lung carcinoma tumors. Transl Lung Cancer Res 2016; 5:401-12. [PMID: 27652204 PMCID: PMC5009079 DOI: 10.21037/tlcr.2016.07.10] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Lung cancer is the principal cause of cancer-related death worldwide. The use of targeted therapies, especially tyrosine kinase inhibitors (TKIs), in specific groups of patients has dramatically improved the prognosis of this disease, although inevitably some patients will develop resistance to these drugs during active treatment. The most common cancer-associated acquired mutation is the epidermal growth factor receptor (EGFR) Thr790Met (T790M) mutation. During active treatment with targeted therapies, histopathological transformation to small-cell lung carcinoma (SCLC) can occur in 3-15% of patients with non-small-cell lung carcinoma (NSCLC) tumors. By definition, SCLC is a high-grade tumor with specific histological and genetic characteristics. In the majority of cases, a good-quality hematoxylin and eosin (H&E) stain is enough to establish a diagnosis. Immunohistochemistry (IHC) is used to confirm the diagnosis and exclude other neoplasia such as sarcomatoid carcinomas, large-cell carcinoma, basaloid squamous-cell carcinoma, chronic inflammation, malignant melanoma, metastatic carcinoma, sarcoma, and lymphoma. A loss of the tumor-suppressor protein retinoblastoma 1 (RB1) is found in 100% of human SCLC tumors; therefore, it has an essential role in tumorigenesis and tumor development. Other genetic pathways probably involved in the histopathological transformation include neurogenic locus notch homolog (NOTCH) and achaete-scute homolog 1 (ASCL1). Histological transformation to SCLC can be suspected in NSCLC patients who clinically deteriorate during active treatment. Biopsy of any new lesion in this clinical setting is highly recommended to rule out a SCLC transformation. New studies are trying to assess this histological transformation by noninvasive measures such as measuring the concentration of serum neuron-specific enolase.
Collapse
|
2106
|
Cabanillas ME, Busaidy NL, Khan SA, Gunn GB, Dadu R, Rao SN, Waguespack SG. Molecular diagnostics and anaplastic thyroid carcinoma: the time has come to harvest the high hanging fruit. INTERNATIONAL JOURNAL OF ENDOCRINE ONCOLOGY 2016. [DOI: 10.2217/ije-2016-0007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Targeted therapies have played a major role in cancer therapeutics, starting with the discovery of a drug against BCR–ABL rearrangements in chronic myelogenous leukemia. This led to the first approval of a targeted agent in cancer and since, many others have followed. Anaplastic thyroid cancer (ATC) is an aggressive carcinoma with few curative options. Although previous cytotoxic chemotherapy and kinase inhibitor therapies have not proven efficacious in ATC, some of the newer drugs appear to be promising. A case report and a comprehensive review of the current standard of care, genetics, modern therapeutic drugs and clinical trials are presented, in order to outline where we currently stand and where the future lies in the quest for a cure for ATC.
Collapse
Affiliation(s)
- Maria E Cabanillas
- Department of Endocrine Neoplasia & Hormonal Disorders, The University of Texas MD Anderson Cancer Center; Houston, TX, USA
| | - Naifa L Busaidy
- Department of Endocrine Neoplasia & Hormonal Disorders, The University of Texas MD Anderson Cancer Center; Houston, TX, USA
| | - Saad A Khan
- The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - G Brandon Gunn
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center; Houston, TX, USA
| | - Ramona Dadu
- Department of Endocrine Neoplasia & Hormonal Disorders, The University of Texas MD Anderson Cancer Center; Houston, TX, USA
| | - Sarika N Rao
- Department of Endocrine Neoplasia & Hormonal Disorders, The University of Texas MD Anderson Cancer Center; Houston, TX, USA
| | - Steven G Waguespack
- Department of Endocrine Neoplasia & Hormonal Disorders, The University of Texas MD Anderson Cancer Center; Houston, TX, USA
| |
Collapse
|
2107
|
Rolfo C, Caparica R. B-RAF mutation in non-small cell lung cancer: the sleeping beauty is waking up. Transl Lung Cancer Res 2016; 5:367-9. [PMID: 27650165 PMCID: PMC5009081 DOI: 10.21037/tlcr.2016.07.04] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 06/23/2016] [Indexed: 12/30/2022]
Affiliation(s)
- Christian Rolfo
- Phase I-Early Clinical Trials Unit, Oncology Department, Antwerp University Hospital & Center for Oncological Research of Antwerp University (CORE), Edegem, Belgium
| | - Rafael Caparica
- Phase I-Early Clinical Trials Unit, Oncology Department, Antwerp University Hospital & Center for Oncological Research of Antwerp University (CORE), Edegem, Belgium
| |
Collapse
|
2108
|
Zago G, Muller M, van den Heuvel M, Baas P. New targeted treatments for non-small-cell lung cancer - role of nivolumab. Biologics 2016; 10:103-17. [PMID: 27536062 PMCID: PMC4975160 DOI: 10.2147/btt.s87878] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Non-small-cell lung cancer (NSCLC) is often diagnosed at an advanced stage of disease, where it is no longer amenable to curative treatment. During the last decades, the survival has only improved significantly for lung cancer patients who have tumors harboring a driver mutation. Therefore, there is a clear unmet need for effective therapies for patients with no mutation. Immunotherapy has emerged as an effective treatment for different cancer types. Nivolumab, a monoclonal inhibitory antibody against PD-1 receptor, can prolong survival of NSCLC patients, with a manageable toxicity profile. In two Phase III trials, nivolumab was compared to docetaxel in patients with, respectively, squamous (CheckMate 017) and non-squamous NSCLC (CheckMate 057). In both trials, nivolumab significantly reduced the risk of death compared to docetaxel (41% and 27% lower risk of death for squamous and non-squamous NSCLC, respectively). Therefore, nivolumab has been approved in the US and in Europe as second-line treatment for advanced NSCLC. Unfortunately, accurate predictive factors for patient selection are lacking, making it difficult to decide who will benefit and who will not. Currently, there are many ongoing trials that evaluate the efficacy of nivolumab in different settings and in combination with other agents. This paper reviews the present literature about the role of nivolumab in the treatment of NSCLC. Particular attention has been given to efficacy studies, toxicity profile, and current and emerging predictive factors.
Collapse
Affiliation(s)
- Giulia Zago
- Department of Thoracic Oncology, The Netherlands Cancer Institute, Antoni van Leeuwenhoek (NKI-AvL), Amsterdam, the Netherlands
- Medical Oncology 2, Istituto Oncologico Veneto (IOV), Padova, Italy
| | - Mirte Muller
- Department of Thoracic Oncology, The Netherlands Cancer Institute, Antoni van Leeuwenhoek (NKI-AvL), Amsterdam, the Netherlands
| | - Michel van den Heuvel
- Department of Thoracic Oncology, The Netherlands Cancer Institute, Antoni van Leeuwenhoek (NKI-AvL), Amsterdam, the Netherlands
| | - Paul Baas
- Department of Thoracic Oncology, The Netherlands Cancer Institute, Antoni van Leeuwenhoek (NKI-AvL), Amsterdam, the Netherlands
| |
Collapse
|
2109
|
Molecular characterization of pulmonary sarcomatoid carcinoma: analysis of 33 cases. Mod Pathol 2016; 29:824-31. [PMID: 27174587 DOI: 10.1038/modpathol.2016.89] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 04/08/2016] [Accepted: 04/14/2016] [Indexed: 12/12/2022]
Abstract
Several targetable genetic alterations have been found in lung cancer, predominantly in adenocarcinomas, which have led to important therapeutic advancements with the advent of targeted therapy. In contrast, the molecular features and presence of targetable genetic abnormalities in pulmonary sarcomatoid carcinomas are largely unknown. Thirty-three cases of pulmonary sarcomatoid carcinoma were tested for approximately 2800 mutations in 50 oncogenes and tumor-suppressor genes, including EGFR, KRAS, NRAS, TP53, BRAF, ERBB2, JAK3, AKT1, ATM, MET, KIT, and PIK3CA. ALK immunostaining was performed, and ALK FISH was performed on cases with any degree of staining. Twenty-four of the 33 cases (72%) had at least one genetic abnormality: 19 cases (58%) had TP53 mutations; 10 cases (30%) had KRAS mutations; AKT1, JAK3, BRAF, NRAS, and PIK3CA mutations were observed in 1 case each (3%). Six of the 19 cases (32%) with a mutation in TP53 had simultaneous mutations in KRAS (18%). The cases with alterations in JAK3, BRAF, and NRAS also had mutations in TP53. The case showing a mutation in PIK3CA had a mutation in KRAS. No EGFR mutations were observed. One case had ALK gene rearrangement. ALK rearrangement was observed in a single case of sarcomatoid carcinoma (3%), which has currently available targeted therapy. Four tumors had mutations in genes with experimental molecular-based therapy, including BRAF, NRAS, PIK3CA, and AKT1. Testing for targetable mutations should be considered for patients with pulmonary sarcomatoid carcinoma, as a subset may benefit from currently approved drugs or clinical trials of novel therapeutic options available for other types of lung cancer.
Collapse
|
2110
|
Izzedine H, El-Fekih RK, Perazella MA. The renal effects of ALK inhibitors. Invest New Drugs 2016; 34:643-9. [PMID: 27468827 DOI: 10.1007/s10637-016-0379-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 07/20/2016] [Indexed: 12/18/2022]
Abstract
Anaplastic lymphoma kinase 1 (ALK-1) is a member of the insulin receptor tyrosine kinase family. In clinical practice, three small molecule inhibitors of ALK-1 are used, namely crizotinib, ceritinib and alectinib. Several more agents are in active pre-clinical and clinical studies. Crizotinib is approved for the treatment of advanced ALK-positive non-small cell lung cancer (NSCLC). According to the package insert and published literature, treatment with crizotinib appears to be associated with kidney failure as well as an increased risk for the development and progression of renal cysts. In addition, this agent is associated with development of peripheral edema and rare electrolyte disorders. This review focuses on the adverse renal effects of Crizotinib in clinical practice.
Collapse
Affiliation(s)
- Hassan Izzedine
- Department of Nephrology, Monceau Park International Clinic Paris, Paris, France.
| | | | - Mark A Perazella
- Department of Nephrology, Yale University School of Medicine, New Haven, CT, 06520, USA
| |
Collapse
|
2111
|
Bansal P, Rusthoven C, Boumber Y, Gan GN. The role of local ablative therapy in oligometastatic non-small-cell lung cancer: hype or hope. Future Oncol 2016; 12:2713-2727. [PMID: 27467543 DOI: 10.2217/fon-2016-0219] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
In recent years, the emergence of the oligometastatic state has called into question whether patients found to have a limited or low metastatic tumor burden may benefit from locally ablative therapy (LAT). In the past two decades, stereotactic body radiation therapy has been increasingly used to safely deliver LAT and provide high local control in nonoperable non-small-cell lung cancer patients. Mostly retrospective analyses suggest that using LAT for oligometastatic disease in non-small-cell lung cancer offers excellent local control and may provide an improvement in progression-free survival. Any meaningful improvement in cancer-specific survival remains debatable. We examine the role of integrating LAT in this patient population and the rationale behind its use in combination with targeted therapy and immunotherapy.
Collapse
Affiliation(s)
- Pranshu Bansal
- Department of Internal Medicine, Division of Hematology/Oncology, University of New Mexico School of Medicine, University of New Mexico Comprehensive Cancer Center, Albuquerque, NM 87131, USA.,Hematology/Oncology Fellowship Program, University of New Mexico School of Medicine, University of New Mexico Comprehensive Cancer Center, Albuquerque, NM 87131, USA
| | - Chad Rusthoven
- Department of Radiation Oncology, University of Colorado School of Medicine, University of Colorado, Aurora, CO, USA
| | - Yanis Boumber
- Department of Internal Medicine, Division of Hematology/Oncology, University of New Mexico School of Medicine, University of New Mexico Comprehensive Cancer Center, Albuquerque, NM 87131, USA.,Cancer Genetics, Epigenetics & Genomics Research Program, University of New Mexico Comprehensive Cancer Center, Albuquerque, NM 87131, USA
| | - Gregory N Gan
- Department of Internal Medicine, Division of Hematology/Oncology, University of New Mexico School of Medicine, University of New Mexico Comprehensive Cancer Center, Albuquerque, NM 87131, USA.,Section of Radiation Oncology, University of New Mexico Comprehensive Cancer Center, Albuquerque, NM 87131, USA.,Cancer Therapeutics: Technology, Discovery & Targeted Delivery Program, University of New Mexico Comprehensive Cancer Center, Albuquerque, NM 87131, USA
| |
Collapse
|
2112
|
Tang N, Wang Z. Comparison of bevacizumab plus chemotherapy with chemotherapy alone in advanced non-small-lung cancer patients. Onco Targets Ther 2016; 9:4671-9. [PMID: 27536131 PMCID: PMC4973774 DOI: 10.2147/ott.s110339] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Bevacizumab plus chemotherapy was approved by the US Food and Drug Administration (FDA) as a first-line treatment for advanced nonsquamous, non-small-cell lung cancer (NSCLC) in 2006. This study retrospectively compared the efficacy of bevacizumab plus chemotherapy with chemotherapy alone as the first-line and second-line treatment as well as the maintenance treatment for advanced NSCLC patients. A total of 1,352 patients were included and we analyzed the efficacy evaluation according to the criteria of the Response Evaluation Criteria In Solid Tumors (RECIST), survival, and adverse reactions. The data showed that for bevacizumab plus chemotherapy as the first-line treatment, the median progression-free survival (mPFS) and median overall survival (mOS) were 11.5 and 17.0 months, respectively, compared to 7.0 and 14 months, respectively, in patients who received chemotherapy alone (P<0.01). With bevacizumab plus chemotherapy as maintenance treatment, the mPFS and mOS were 6.0 and 17.4 months, respectively, compared to 3.0 and 15.0 months, respectively, with chemotherapy alone (P<0.01). With bevacizumab plus chemotherapy as the second-line treatment, the mPFS was 3.0 months compared to only 2.0 months with chemotherapy alone (P<0.01). The overall responses to the different regimens showed that the remission rate with bevacizumab plus chemotherapy was higher than that with chemotherapy alone (31.8% vs 25.5%, P<0.05), although there was no statistical difference in the disease control rate with either first- or second-line treatment. In conclusion, chemotherapy plus bevacizumab as the first-line and maintenance treatment, led to better curative rates and tolerable adverse reactions compared with chemotherapy alone in advanced NSCLC patients. Bevacizumab combined with cytotoxic drugs was suitable as the second-line treatment for such patients.
Collapse
Affiliation(s)
- Ning Tang
- Department of Oncology, Shandong Cancer Hospital and Institute, Jinan, Shandong, People's Republic of China
| | - Zhehai Wang
- Department of Oncology, Shandong Cancer Hospital and Institute, Jinan, Shandong, People's Republic of China
| |
Collapse
|
2113
|
Schvartsman G, Ferrarotto R, Massarelli E. Checkpoint inhibitors in lung cancer: latest developments and clinical potential. Ther Adv Med Oncol 2016; 8:460-473. [PMID: 27800034 DOI: 10.1177/1758834016661164] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Lung cancer is the leading cause of cancer death in the United States. The vast majority of patients are diagnosed with metastatic disease with a 5-year survival rate of less than 5%. After first-line chemotherapy or biomarker-matched targeted therapy, only suitable for a small group of patients, further systemic therapy options rendered very limited, if any, benefit until recently. Checkpoint inhibitors have significantly improved outcomes in patients with metastatic non-small cell lung cancer (NSCLC) and are currently an established second-line therapeutic option. In this manuscript, we review the mechanism of action of checkpoint inhibitors, present the available data with approved and experimental agents, discuss the progress that has already been made in the field, as well as toxicity awareness, and future perspectives.
Collapse
Affiliation(s)
- Gustavo Schvartsman
- Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Renata Ferrarotto
- Department of Thoracic Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Erminia Massarelli
- Department of Medical Oncology, City of Hope Comprehensive Cancer Center, 1500 East Duarte Road, Duarte, CA 91010, USA
| |
Collapse
|
2114
|
Tyrosine Kinase Receptor Landscape in Lung Cancer: Therapeutical Implications. DISEASE MARKERS 2016; 2016:9214056. [PMID: 27528792 PMCID: PMC4977389 DOI: 10.1155/2016/9214056] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 06/08/2016] [Accepted: 06/09/2016] [Indexed: 12/24/2022]
Abstract
Lung cancer is a heterogeneous disease responsible for the most cases of cancer-related deaths. The majority of patients are clinically diagnosed at advanced stages, with a poor survival rate. For this reason, the identification of oncodrivers and novel biomarkers is decisive for the future clinical management of this pathology. The rise of high throughput technologies popularly referred to as “omics” has accelerated the discovery of new biomarkers and drivers for this pathology. Within them, tyrosine kinase receptors (TKRs) have proven to be of importance as diagnostic, prognostic, and predictive tools and, due to their molecular nature, as therapeutic targets. Along this review, the role of TKRs in the different lung cancer histologies, research on improvement of anti-TKR therapy, and the current approaches to manage anti-TKR resistance will be discussed.
Collapse
|
2115
|
Lin JJ, Shaw AT. Differential Sensitivity to Crizotinib: Does EML4-ALK Fusion Variant Matter? J Clin Oncol 2016; 34:3363-5. [PMID: 27458283 DOI: 10.1200/jco.2016.68.5891] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Jessica J Lin
- Massachusetts General Hospital Cancer Center, Boston, MA
| | - Alice T Shaw
- Massachusetts General Hospital Cancer Center, Boston, MA
| |
Collapse
|
2116
|
Hellmann MD, Li BT, Chaft JE, Kris MG. Chemotherapy remains an essential element of personalized care for persons with lung cancers. Ann Oncol 2016; 27:1829-35. [PMID: 27456296 DOI: 10.1093/annonc/mdw271] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 07/02/2016] [Indexed: 12/14/2022] Open
Abstract
Molecularly targeted and immunotherapies have improved the care of patients with lung cancers. These successes have rallied calls to replace or avoid chemotherapy. Yet, even in this era of precision medicine and exciting advances, cytotoxic chemotherapies remain an essential component of lung cancer treatment. In the setting of locoregional disease, chemotherapy is the only systemic therapy thus far proven to enhance curability when combined with surgery or radiation. In the metastatic setting, chemotherapy can improve the length and quality of life in many patients. Chemotherapy remains the mainstay of care for individuals whose cancers with oncogenic drivers have acquired resistance to targeted agents. Chemotherapy also has the potential to modulate the immune system to enhance the effectiveness of immune checkpoint inhibitors. In this context, chemotherapy should be framed as a critical component of the armamentarium available for optimizing cancer care rather than an unfortunate anachronism. We examine the role of chemotherapy with precision medicine in the current care of patients with lung cancers, as well as opportunities for future integration in combinations with targeted agents, angiogenesis inhibitors, immunotherapies, and antibody drug conjugates.
Collapse
Affiliation(s)
- M D Hellmann
- Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, USA
| | - B T Li
- Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, USA
| | - J E Chaft
- Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, USA
| | - M G Kris
- Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, USA
| |
Collapse
|
2117
|
Cameron L, Solomon B. New Treatment Options for ALK-Rearranged Non-Small Cell Lung Cancer. Curr Treat Options Oncol 2016; 16:49. [PMID: 26318457 DOI: 10.1007/s11864-015-0367-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
OPINION STATEMENT ALK rearrangements are present in 3-5% of patients with non-small cell lung cancer (NSCLC) and after epidermal growth factor receptor (EGFR) mutations represent the second molecular target in NSCLC to be validated through phase III clinical trials. The PROFILE 1014 international multicentre phase III trial demonstrated the superiority of crizotinib over standard chemotherapy, establishing crizotinib as standard first-line therapy for patients with advanced ALK-positive NSCLC and indicating the requirement for ALK testing to guide selection of optimal first-line therapy for non-squamous NSCLC. Despite impressive and durable responses, progression on treatment reflecting the development of acquired resistance is inevitable. There are several mechanisms of resistance including ALK kinase mutation or copy number gain, activation of bypass pathways and potentially pharmacokinetic failure of therapy (most commonly in CNS). A broad array of newer generation ALK inhibitors are in development that appear effective in the crizotinib-resistant setting including in patients with intracranial progression. These agents, including ceritinib and alectinib, have a higher potency against ALK kinase than crizotinib, activity against mutations that confer resistance to crizotinib and potentially improved CNS penetration. While in selected patients, continued therapy with crizotinib after local ablative treatments of oligo-progressive systemic or CNS disease may be an option, for many patients use of a newer generation compound will be effective. First-line treatment with newer generation ALK inhibitors may have potential advantages over sequential treatment after crizotinib; however, the optimal sequence of therapy with ALK inhibitors has not been determined and is being explored in ongoing phase III studies.
Collapse
Affiliation(s)
- Laird Cameron
- Department of Medical Oncology, Peter MacCallum Cancer Centre, St Andrew's Place, East Melbourne, VIC, 3002, Australia
| | | |
Collapse
|
2118
|
Non-Invasive Methods to Monitor Mechanisms of Resistance to Tyrosine Kinase Inhibitors in Non-Small-Cell Lung Cancer: Where Do We Stand? Int J Mol Sci 2016; 17:ijms17071186. [PMID: 27455248 PMCID: PMC4964555 DOI: 10.3390/ijms17071186] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 06/28/2016] [Accepted: 07/15/2016] [Indexed: 12/22/2022] Open
Abstract
The induction of resistance mechanisms represents an important problem for the targeted therapy of patients with non-small-cell lung cancer (NSCLC). The best-known resistance mechanism induced during treatment with epidermal growth factor receptor (EGFR)-tyrosine kinase inhibitors (TKIs) is EGFR T790M mutation for which specific drugs are have been developed. However, other molecular alterations have also been reported as induced resistance mechanisms to EGFR-TKIs. Similarly, there is growing evidence of acquired resistance mechanisms to anaplastic lymphoma kinase (ALK)-TKI treatment. A better understanding of these acquired resistance mechanisms is essential in clinical practice as patients could be treated with specific drugs that are active against the induced alterations. The use of free circulating tumor nucleic acids or circulating tumor cells (CTCs) enables resistance mechanisms to be characterized in a non-invasive manner and reduces the need for tumor re-biopsy. This review discusses the main resistance mechanisms to TKIs and provides a comprehensive overview of innovative strategies to evaluate known resistance mechanisms in free circulating nucleic acids or CTCs and potential future orientations for these non-invasive approaches.
Collapse
|
2119
|
Pugliese SB, Neal JW, Kwong BY. Management of Dermatologic Complications of Lung Cancer Therapies. Curr Treat Options Oncol 2016; 16:50. [PMID: 26338208 DOI: 10.1007/s11864-015-0368-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OPINION STATEMENT In recent years, oncogene-directed targeted agents and immunotherapies have expanded the treatment armamentarium for advanced lung cancer and, in particular, non-small cell lung cancer (NSCLC). Along with extended survival, these agents are accompanied by a host of cutaneous complications that affect the skin, hair, and nails. These skin complications range from the well-characterized papulopustular (acneiform) eruption of the epidermal growth factor receptor (EGFR) inhibitors to the emerging characterization of lichenoid skin eruptions seen during treatment with antibodies targeting the programmed cell death protein 1 (PD-1) and programmed cell death protein 1 ligand (PD-L1). When promptly recognized and accurately diagnosed, most cutaneous adverse events can be managed with supportive treatments, avoiding the need to interrupt antitumor therapy. Furthermore, preemptive management of skin problems can lead to significantly decreased severity of many cutaneous complications of these therapies. We encourage close collaboration between dermatologists and oncologists to better characterize cutaneous toxicity, select appropriate management, and avoid unnecessary dose reduction or discontinuation while simultaneously improving patient quality of life.
Collapse
|
2120
|
Broutin S, Stewart A, Thavasu P, Paci A, Bidart JM, Banerji U. Insights into significance of combined inhibition of MEK and m-TOR signalling output in KRAS mutant non-small-cell lung cancer. Br J Cancer 2016; 115:549-52. [PMID: 27441499 PMCID: PMC4997545 DOI: 10.1038/bjc.2016.220] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 06/14/2016] [Accepted: 06/23/2016] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND We aimed to understand the dependence of MEK and m-TOR inhibition in EGFR(WT)/ALK(non-rearranged) NSCLC cell lines. METHODS In a panel of KRAS(M) and KRAS(WT) NSCLC cell lines, we determined growth inhibition (GI) following maximal reduction in p-ERK and p-S6RP caused by trametinib (MEK inhibitor) and AZD2014 (m-TOR inhibitor), respectively. RESULTS GI caused by maximal m-TOR inhibition was significantly greater than GI caused by maximal MEK inhibition in the cell line panel (52% vs 18%, P<10(-4)). There was no significant difference in GI caused by maximal m-TOR compared with maximal m-TOR+MEK inhibition. However, GI caused by the combination was significantly greater in the KRAS(M) cell lines (79% vs 61%, P=0.017). CONCLUSIONS m-TOR inhibition was more critical to GI than MEK inhibition in EGFR(WT)/ALK(non-rearranged) NSCLC cells. The combination of MEK and m-TOR inhibition was most effective in KRAS(M) cells.
Collapse
Affiliation(s)
- Sophie Broutin
- Gustave Roussy, Université Paris-Saclay, Département de Biologie et Pathologie Médicales, Villejuif, F-94805, France.,The Institute of Cancer Research, Clinical Pharmacology and Trials Team, London, SM2 5PT, UK
| | - Adam Stewart
- The Institute of Cancer Research, Clinical Pharmacology and Trials Team, London, SM2 5PT, UK
| | - Parames Thavasu
- The Institute of Cancer Research, Clinical Pharmacology and Trials Team, London, SM2 5PT, UK
| | - Angelo Paci
- Gustave Roussy, Université Paris-Saclay, Département de Biologie et Pathologie Médicales, Villejuif, F-94805, France
| | - Jean-Michel Bidart
- Gustave Roussy, Université Paris-Saclay, Département de Biologie et Pathologie Médicales, Villejuif, F-94805, France
| | - Udai Banerji
- The Institute of Cancer Research, Clinical Pharmacology and Trials Team, London, SM2 5PT, UK
| |
Collapse
|
2121
|
Uguen A, Andrieu-Key S, Vergne F, Descourt R, Quéré G, Quintin-Roué I, Key S, Guéguen P, Talagas M, De Braekeleer M, Marcorelles P. ALK ambiguous-positive non-small cell lung cancers are tumors challenged by diagnostic and therapeutic issues. Oncol Rep 2016; 36:1427-34. [PMID: 27460205 DOI: 10.3892/or.2016.4962] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 04/19/2016] [Indexed: 11/06/2022] Open
Abstract
Searching for ALK rearrangements using the approved fluorescent in situ hybridization (FISH) test and complementary immunohistochemistry (IHC) has become the rule to treat patients with advanced non‑small cell lung cancer (NSCLC) with anti‑ALK targeted therapy. The concordance between the two techniques is reported to be strong but imperfect. We report our experience with cases of ALK‑rearranged lung adenocarcinomas pointing out particularly ambiguous cases. FISH and IHC data on ALK but also c‑MET IHC as well as EGFR and KRAS mutation screening are considered, together with response to crizotinib treatment. We classified the 55 FISH ALK‑rearranged tumors into two groups according to the FISH and IHC results: a concordant FISH+IHC+ group (31 tumors) and an ambiguous group (24 tumors). These tumors were considered as 'ambiguous' ALK‑positive due to negative (21 tumors) or non‑contributive (3 tumors) IHC. In addition, the percentage of FISH-positive nuclei was between 15 and 20% in 17 tumors belonging to one or the other group (now called borderline tumors). We discuss the accuracy of the different tests with intent to determine whether ambiguous and borderline tumors are real positive ALK‑rearranged tumors. To conclude, ambiguous ALK‑positive lung cancers are challenging tumors with diagnosis and therapeutic issues that can justify parallel FISH, IHC and molecular screening strategy.
Collapse
Affiliation(s)
| | - Sophie Andrieu-Key
- Department of Pathology, Brest University Hospital, F‑29609 Brest, France
| | | | | | - Gilles Quéré
- Department of Oncology, CHRU Brest, F‑29220 Brest, France
| | | | - Stéphane Key
- Department of Oncology, CHRU Brest, F‑29220 Brest, France
| | | | - Matthieu Talagas
- Department of Pathology, Brest University Hospital, F‑29609 Brest, France
| | | | | |
Collapse
|
2122
|
Fujiwara Y, Hamada A, Mizugaki H, Aikawa H, Hata T, Horinouchi H, Kanda S, Goto Y, Itahashi K, Nokihara H, Yamamoto N, Ohe Y. Pharmacokinetic profiles of significant adverse events with crizotinib in Japanese patients with ABCB1 polymorphism. Cancer Sci 2016; 107:1117-23. [PMID: 27270784 PMCID: PMC4982581 DOI: 10.1111/cas.12983] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 05/24/2016] [Accepted: 06/06/2016] [Indexed: 01/11/2023] Open
Abstract
Crizotinib is a standard treatment for advanced ALK-positive non-small-cell lung cancer (NSCLC). We undertook this study to investigate the pharmacokinetics of crizotinib and clinical and pharmacogenomic factors that may increase the risk of adverse events (AEs). We defined clinically significant AEs as grade 4 hematological toxicity, grade ≥3 non-hematological toxicity, and any grade of interstitial lung disease. Eight subjects with ALK-positive NSCLC scheduled to receive crizotinib 250 mg twice daily were studied. Six patients were female and two were male, and most of the patients had low body weight with a median body weight of 46.8 kg (range, 42.4-61.0 kg). All patients developed AEs, five developing six clinically significant AEs. Six patients required dose reduction. In pharmacokinetic analysis, blood samples were obtained on days 1 and 15. The mean area under the plasma concentration-time curve from 0-12 h (AUC0-12 ) on day 15 was significantly increased in patients with clinically significant AEs (n = 5) compared with those without (n = 3) (P = 0.04). Genetic polymorphisms of ABCB1 were analyzed. One patient with the ABCB1 1236TT-2677TT-3435TT genotype was an outlier, with an AUC0-12 and peak concentrations on day 15 of 2.84× and 2.61× the mean, respectively, compared with those with other genotypes. Our results suggest that some Japanese NSCLC patients treated with crizotinib developed clinically significant toxicities that were related to altered pharmacokinetics parameters due to genotype and body weight factors.
Collapse
Affiliation(s)
- Yutaka Fujiwara
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan.,Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan
| | - Akinobu Hamada
- Division of Clinical Pharmacology and Translational Research, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Tokyo, Japan
| | - Hidenori Mizugaki
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan.,First Department of Medicine, Hokkaido University School of Medicine, Sapporo, Japan
| | - Hiroaki Aikawa
- Division of Clinical Pharmacology and Translational Research, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Tokyo, Japan
| | - Toshiyuki Hata
- Division of Clinical Pharmacology and Translational Research, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Tokyo, Japan
| | - Hidehito Horinouchi
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Shintaro Kanda
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Yasushi Goto
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Kota Itahashi
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Hiroshi Nokihara
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Noboru Yamamoto
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan.,Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan
| | - Yuichiro Ohe
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| |
Collapse
|
2123
|
Crinò L, Ahn MJ, De Marinis F, Groen HJM, Wakelee H, Hida T, Mok T, Spigel D, Felip E, Nishio M, Scagliotti G, Branle F, Emeremni C, Quadrigli M, Zhang J, Shaw AT. Multicenter Phase II Study of Whole-Body and Intracranial Activity With Ceritinib in Patients With ALK-Rearranged Non-Small-Cell Lung Cancer Previously Treated With Chemotherapy and Crizotinib: Results From ASCEND-2. J Clin Oncol 2016; 34:2866-73. [PMID: 27432917 DOI: 10.1200/jco.2015.65.5936] [Citation(s) in RCA: 279] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
PURPOSE Phase I data (ASCEND-1) showed ceritinib efficacy in patients with ALK-rearranged non-small-cell lung cancer (NSCLC), regardless of brain metastases status and with or without prior therapy with an inhibitor of the ALK protein. Data are presented from a phase II trial (ASCEND-2) in which ceritinib efficacy and safety were evaluated in patients who had ALK-rearranged NSCLC previously treated with at least one platinum-based chemotherapy and who had experienced progression during crizotinib treatment as their last prior therapy. PATIENTS AND METHODS Patients with advanced ALK-rearranged NSCLC, including those with asymptomatic or neurologically stable baseline brain metastases, received oral ceritinib 750 mg/d. Whole-body and intracranial responses were investigator assessed (according to RECIST version 1.1). Patient-reported outcomes were evaluated with the Lung Cancer Symptom Scale and European Organisation for Research and Treatment of Cancer surveys (the core-30 and the 13-item lung cancer-specific quality-of-life questionnaires). RESULTS All 140 patients enrolled had received two or more previous treatment regimens, and all patients had received crizotinib. The median duration of exposure and the follow-up time with ceritinib were 8.8 months (range, 0.1 to 19.4 months) and 11.3 months (range, 0.1 to 18.9 months), respectively. Investigator-assessed overall response rate was 38.6% (95% CI, 30.5% to 47.2%). Secondary end points, all investigator assessed, included disease control rate (77.1%; 95% CI, 69.3% to 83.8%), time to response (median, 1.8 months; range, 1.6 to 5.6 months), duration of response (median, 9.7 months; 95% CI, 7.1 to 11.1 months), and progression-free survival (median, 5.7 months; 95% CI, 5.4 to 7.6 months). Of 100 patients with baseline brain metastases, 20 had active target lesions at baseline; investigator-assessed intracranial overall response rate was 45.0% (95% CI, 23.1% to 68.5%). The most common adverse events (majority, grade 1 or 2) for all treated patients were nausea (81.4%), diarrhea (80.0%), and vomiting (62.9%). Patient-reported outcomes showed a trend toward improved symptom burden. The global quality-of-life score was maintained during treatment. CONCLUSION Consistent with its activity in ASCEND-1, ceritinib treatment provided clinically meaningful and durable responses with manageable tolerability in chemotherapy- and crizotinib-pretreated patients, including those with brain metastases.
Collapse
Affiliation(s)
- Lucio Crinò
- Lucio Crinò, University Medical School of Perugia, Azienda Ospedale Perugia, Perugia; Filippo De Marinis, European Institute of Oncology, Milan; Giorgio Scagliotti, University of Torino, Torino, Italy; Myung-Ju Ahn, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, South Korea; Harry J.M. Groen, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Heather Wakelee, Stanford University Medical Center, Stanford, CA; Toyoaki Hida, Aichi Cancer Center, Nagoya; Makoto Nishio, Japanese Foundation for Cancer Research, Tokyo, Japan; Tony Mok, Chinese University of Hong Kong, Shatin, China; David Spigel, Sarah Cannon Research Institute, Nashville, TN; Enriqueta Felip, Vall d'Hebron University, Barcelona, Spain; Fabrice Branle and Massimiliano Quadrigli, Novartis Pharma AG, Basel, Switzerland; Chetachi Emeremni and Jie Zhang, Novartis Pharma, East Hanover, NJ; and Alice T. Shaw, Massachusetts General Hospital, Boston MA.
| | - Myung-Ju Ahn
- Lucio Crinò, University Medical School of Perugia, Azienda Ospedale Perugia, Perugia; Filippo De Marinis, European Institute of Oncology, Milan; Giorgio Scagliotti, University of Torino, Torino, Italy; Myung-Ju Ahn, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, South Korea; Harry J.M. Groen, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Heather Wakelee, Stanford University Medical Center, Stanford, CA; Toyoaki Hida, Aichi Cancer Center, Nagoya; Makoto Nishio, Japanese Foundation for Cancer Research, Tokyo, Japan; Tony Mok, Chinese University of Hong Kong, Shatin, China; David Spigel, Sarah Cannon Research Institute, Nashville, TN; Enriqueta Felip, Vall d'Hebron University, Barcelona, Spain; Fabrice Branle and Massimiliano Quadrigli, Novartis Pharma AG, Basel, Switzerland; Chetachi Emeremni and Jie Zhang, Novartis Pharma, East Hanover, NJ; and Alice T. Shaw, Massachusetts General Hospital, Boston MA
| | - Filippo De Marinis
- Lucio Crinò, University Medical School of Perugia, Azienda Ospedale Perugia, Perugia; Filippo De Marinis, European Institute of Oncology, Milan; Giorgio Scagliotti, University of Torino, Torino, Italy; Myung-Ju Ahn, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, South Korea; Harry J.M. Groen, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Heather Wakelee, Stanford University Medical Center, Stanford, CA; Toyoaki Hida, Aichi Cancer Center, Nagoya; Makoto Nishio, Japanese Foundation for Cancer Research, Tokyo, Japan; Tony Mok, Chinese University of Hong Kong, Shatin, China; David Spigel, Sarah Cannon Research Institute, Nashville, TN; Enriqueta Felip, Vall d'Hebron University, Barcelona, Spain; Fabrice Branle and Massimiliano Quadrigli, Novartis Pharma AG, Basel, Switzerland; Chetachi Emeremni and Jie Zhang, Novartis Pharma, East Hanover, NJ; and Alice T. Shaw, Massachusetts General Hospital, Boston MA
| | - Harry J M Groen
- Lucio Crinò, University Medical School of Perugia, Azienda Ospedale Perugia, Perugia; Filippo De Marinis, European Institute of Oncology, Milan; Giorgio Scagliotti, University of Torino, Torino, Italy; Myung-Ju Ahn, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, South Korea; Harry J.M. Groen, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Heather Wakelee, Stanford University Medical Center, Stanford, CA; Toyoaki Hida, Aichi Cancer Center, Nagoya; Makoto Nishio, Japanese Foundation for Cancer Research, Tokyo, Japan; Tony Mok, Chinese University of Hong Kong, Shatin, China; David Spigel, Sarah Cannon Research Institute, Nashville, TN; Enriqueta Felip, Vall d'Hebron University, Barcelona, Spain; Fabrice Branle and Massimiliano Quadrigli, Novartis Pharma AG, Basel, Switzerland; Chetachi Emeremni and Jie Zhang, Novartis Pharma, East Hanover, NJ; and Alice T. Shaw, Massachusetts General Hospital, Boston MA
| | - Heather Wakelee
- Lucio Crinò, University Medical School of Perugia, Azienda Ospedale Perugia, Perugia; Filippo De Marinis, European Institute of Oncology, Milan; Giorgio Scagliotti, University of Torino, Torino, Italy; Myung-Ju Ahn, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, South Korea; Harry J.M. Groen, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Heather Wakelee, Stanford University Medical Center, Stanford, CA; Toyoaki Hida, Aichi Cancer Center, Nagoya; Makoto Nishio, Japanese Foundation for Cancer Research, Tokyo, Japan; Tony Mok, Chinese University of Hong Kong, Shatin, China; David Spigel, Sarah Cannon Research Institute, Nashville, TN; Enriqueta Felip, Vall d'Hebron University, Barcelona, Spain; Fabrice Branle and Massimiliano Quadrigli, Novartis Pharma AG, Basel, Switzerland; Chetachi Emeremni and Jie Zhang, Novartis Pharma, East Hanover, NJ; and Alice T. Shaw, Massachusetts General Hospital, Boston MA
| | - Toyoaki Hida
- Lucio Crinò, University Medical School of Perugia, Azienda Ospedale Perugia, Perugia; Filippo De Marinis, European Institute of Oncology, Milan; Giorgio Scagliotti, University of Torino, Torino, Italy; Myung-Ju Ahn, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, South Korea; Harry J.M. Groen, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Heather Wakelee, Stanford University Medical Center, Stanford, CA; Toyoaki Hida, Aichi Cancer Center, Nagoya; Makoto Nishio, Japanese Foundation for Cancer Research, Tokyo, Japan; Tony Mok, Chinese University of Hong Kong, Shatin, China; David Spigel, Sarah Cannon Research Institute, Nashville, TN; Enriqueta Felip, Vall d'Hebron University, Barcelona, Spain; Fabrice Branle and Massimiliano Quadrigli, Novartis Pharma AG, Basel, Switzerland; Chetachi Emeremni and Jie Zhang, Novartis Pharma, East Hanover, NJ; and Alice T. Shaw, Massachusetts General Hospital, Boston MA
| | - Tony Mok
- Lucio Crinò, University Medical School of Perugia, Azienda Ospedale Perugia, Perugia; Filippo De Marinis, European Institute of Oncology, Milan; Giorgio Scagliotti, University of Torino, Torino, Italy; Myung-Ju Ahn, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, South Korea; Harry J.M. Groen, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Heather Wakelee, Stanford University Medical Center, Stanford, CA; Toyoaki Hida, Aichi Cancer Center, Nagoya; Makoto Nishio, Japanese Foundation for Cancer Research, Tokyo, Japan; Tony Mok, Chinese University of Hong Kong, Shatin, China; David Spigel, Sarah Cannon Research Institute, Nashville, TN; Enriqueta Felip, Vall d'Hebron University, Barcelona, Spain; Fabrice Branle and Massimiliano Quadrigli, Novartis Pharma AG, Basel, Switzerland; Chetachi Emeremni and Jie Zhang, Novartis Pharma, East Hanover, NJ; and Alice T. Shaw, Massachusetts General Hospital, Boston MA
| | - David Spigel
- Lucio Crinò, University Medical School of Perugia, Azienda Ospedale Perugia, Perugia; Filippo De Marinis, European Institute of Oncology, Milan; Giorgio Scagliotti, University of Torino, Torino, Italy; Myung-Ju Ahn, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, South Korea; Harry J.M. Groen, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Heather Wakelee, Stanford University Medical Center, Stanford, CA; Toyoaki Hida, Aichi Cancer Center, Nagoya; Makoto Nishio, Japanese Foundation for Cancer Research, Tokyo, Japan; Tony Mok, Chinese University of Hong Kong, Shatin, China; David Spigel, Sarah Cannon Research Institute, Nashville, TN; Enriqueta Felip, Vall d'Hebron University, Barcelona, Spain; Fabrice Branle and Massimiliano Quadrigli, Novartis Pharma AG, Basel, Switzerland; Chetachi Emeremni and Jie Zhang, Novartis Pharma, East Hanover, NJ; and Alice T. Shaw, Massachusetts General Hospital, Boston MA
| | - Enriqueta Felip
- Lucio Crinò, University Medical School of Perugia, Azienda Ospedale Perugia, Perugia; Filippo De Marinis, European Institute of Oncology, Milan; Giorgio Scagliotti, University of Torino, Torino, Italy; Myung-Ju Ahn, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, South Korea; Harry J.M. Groen, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Heather Wakelee, Stanford University Medical Center, Stanford, CA; Toyoaki Hida, Aichi Cancer Center, Nagoya; Makoto Nishio, Japanese Foundation for Cancer Research, Tokyo, Japan; Tony Mok, Chinese University of Hong Kong, Shatin, China; David Spigel, Sarah Cannon Research Institute, Nashville, TN; Enriqueta Felip, Vall d'Hebron University, Barcelona, Spain; Fabrice Branle and Massimiliano Quadrigli, Novartis Pharma AG, Basel, Switzerland; Chetachi Emeremni and Jie Zhang, Novartis Pharma, East Hanover, NJ; and Alice T. Shaw, Massachusetts General Hospital, Boston MA
| | - Makoto Nishio
- Lucio Crinò, University Medical School of Perugia, Azienda Ospedale Perugia, Perugia; Filippo De Marinis, European Institute of Oncology, Milan; Giorgio Scagliotti, University of Torino, Torino, Italy; Myung-Ju Ahn, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, South Korea; Harry J.M. Groen, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Heather Wakelee, Stanford University Medical Center, Stanford, CA; Toyoaki Hida, Aichi Cancer Center, Nagoya; Makoto Nishio, Japanese Foundation for Cancer Research, Tokyo, Japan; Tony Mok, Chinese University of Hong Kong, Shatin, China; David Spigel, Sarah Cannon Research Institute, Nashville, TN; Enriqueta Felip, Vall d'Hebron University, Barcelona, Spain; Fabrice Branle and Massimiliano Quadrigli, Novartis Pharma AG, Basel, Switzerland; Chetachi Emeremni and Jie Zhang, Novartis Pharma, East Hanover, NJ; and Alice T. Shaw, Massachusetts General Hospital, Boston MA
| | - Giorgio Scagliotti
- Lucio Crinò, University Medical School of Perugia, Azienda Ospedale Perugia, Perugia; Filippo De Marinis, European Institute of Oncology, Milan; Giorgio Scagliotti, University of Torino, Torino, Italy; Myung-Ju Ahn, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, South Korea; Harry J.M. Groen, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Heather Wakelee, Stanford University Medical Center, Stanford, CA; Toyoaki Hida, Aichi Cancer Center, Nagoya; Makoto Nishio, Japanese Foundation for Cancer Research, Tokyo, Japan; Tony Mok, Chinese University of Hong Kong, Shatin, China; David Spigel, Sarah Cannon Research Institute, Nashville, TN; Enriqueta Felip, Vall d'Hebron University, Barcelona, Spain; Fabrice Branle and Massimiliano Quadrigli, Novartis Pharma AG, Basel, Switzerland; Chetachi Emeremni and Jie Zhang, Novartis Pharma, East Hanover, NJ; and Alice T. Shaw, Massachusetts General Hospital, Boston MA
| | - Fabrice Branle
- Lucio Crinò, University Medical School of Perugia, Azienda Ospedale Perugia, Perugia; Filippo De Marinis, European Institute of Oncology, Milan; Giorgio Scagliotti, University of Torino, Torino, Italy; Myung-Ju Ahn, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, South Korea; Harry J.M. Groen, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Heather Wakelee, Stanford University Medical Center, Stanford, CA; Toyoaki Hida, Aichi Cancer Center, Nagoya; Makoto Nishio, Japanese Foundation for Cancer Research, Tokyo, Japan; Tony Mok, Chinese University of Hong Kong, Shatin, China; David Spigel, Sarah Cannon Research Institute, Nashville, TN; Enriqueta Felip, Vall d'Hebron University, Barcelona, Spain; Fabrice Branle and Massimiliano Quadrigli, Novartis Pharma AG, Basel, Switzerland; Chetachi Emeremni and Jie Zhang, Novartis Pharma, East Hanover, NJ; and Alice T. Shaw, Massachusetts General Hospital, Boston MA
| | - Chetachi Emeremni
- Lucio Crinò, University Medical School of Perugia, Azienda Ospedale Perugia, Perugia; Filippo De Marinis, European Institute of Oncology, Milan; Giorgio Scagliotti, University of Torino, Torino, Italy; Myung-Ju Ahn, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, South Korea; Harry J.M. Groen, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Heather Wakelee, Stanford University Medical Center, Stanford, CA; Toyoaki Hida, Aichi Cancer Center, Nagoya; Makoto Nishio, Japanese Foundation for Cancer Research, Tokyo, Japan; Tony Mok, Chinese University of Hong Kong, Shatin, China; David Spigel, Sarah Cannon Research Institute, Nashville, TN; Enriqueta Felip, Vall d'Hebron University, Barcelona, Spain; Fabrice Branle and Massimiliano Quadrigli, Novartis Pharma AG, Basel, Switzerland; Chetachi Emeremni and Jie Zhang, Novartis Pharma, East Hanover, NJ; and Alice T. Shaw, Massachusetts General Hospital, Boston MA
| | - Massimiliano Quadrigli
- Lucio Crinò, University Medical School of Perugia, Azienda Ospedale Perugia, Perugia; Filippo De Marinis, European Institute of Oncology, Milan; Giorgio Scagliotti, University of Torino, Torino, Italy; Myung-Ju Ahn, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, South Korea; Harry J.M. Groen, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Heather Wakelee, Stanford University Medical Center, Stanford, CA; Toyoaki Hida, Aichi Cancer Center, Nagoya; Makoto Nishio, Japanese Foundation for Cancer Research, Tokyo, Japan; Tony Mok, Chinese University of Hong Kong, Shatin, China; David Spigel, Sarah Cannon Research Institute, Nashville, TN; Enriqueta Felip, Vall d'Hebron University, Barcelona, Spain; Fabrice Branle and Massimiliano Quadrigli, Novartis Pharma AG, Basel, Switzerland; Chetachi Emeremni and Jie Zhang, Novartis Pharma, East Hanover, NJ; and Alice T. Shaw, Massachusetts General Hospital, Boston MA
| | - Jie Zhang
- Lucio Crinò, University Medical School of Perugia, Azienda Ospedale Perugia, Perugia; Filippo De Marinis, European Institute of Oncology, Milan; Giorgio Scagliotti, University of Torino, Torino, Italy; Myung-Ju Ahn, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, South Korea; Harry J.M. Groen, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Heather Wakelee, Stanford University Medical Center, Stanford, CA; Toyoaki Hida, Aichi Cancer Center, Nagoya; Makoto Nishio, Japanese Foundation for Cancer Research, Tokyo, Japan; Tony Mok, Chinese University of Hong Kong, Shatin, China; David Spigel, Sarah Cannon Research Institute, Nashville, TN; Enriqueta Felip, Vall d'Hebron University, Barcelona, Spain; Fabrice Branle and Massimiliano Quadrigli, Novartis Pharma AG, Basel, Switzerland; Chetachi Emeremni and Jie Zhang, Novartis Pharma, East Hanover, NJ; and Alice T. Shaw, Massachusetts General Hospital, Boston MA
| | - Alice T Shaw
- Lucio Crinò, University Medical School of Perugia, Azienda Ospedale Perugia, Perugia; Filippo De Marinis, European Institute of Oncology, Milan; Giorgio Scagliotti, University of Torino, Torino, Italy; Myung-Ju Ahn, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, South Korea; Harry J.M. Groen, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Heather Wakelee, Stanford University Medical Center, Stanford, CA; Toyoaki Hida, Aichi Cancer Center, Nagoya; Makoto Nishio, Japanese Foundation for Cancer Research, Tokyo, Japan; Tony Mok, Chinese University of Hong Kong, Shatin, China; David Spigel, Sarah Cannon Research Institute, Nashville, TN; Enriqueta Felip, Vall d'Hebron University, Barcelona, Spain; Fabrice Branle and Massimiliano Quadrigli, Novartis Pharma AG, Basel, Switzerland; Chetachi Emeremni and Jie Zhang, Novartis Pharma, East Hanover, NJ; and Alice T. Shaw, Massachusetts General Hospital, Boston MA
| |
Collapse
|
2124
|
Kim SM, Kim H, Yun MR, Kang HN, Pyo KH, Park HJ, Lee JM, Choi HM, Ellinghaus P, Ocker M, Paik S, Kim HR, Cho BC. Activation of the Met kinase confers acquired drug resistance in FGFR-targeted lung cancer therapy. Oncogenesis 2016; 5:e241. [PMID: 27429073 PMCID: PMC5399172 DOI: 10.1038/oncsis.2016.48] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 05/30/2016] [Accepted: 06/06/2016] [Indexed: 12/11/2022] Open
Abstract
Aberrant fibroblast growth factor receptor (FGFR) activation/expression is a common feature in lung cancer (LC). In this study, we evaluated the antitumor activity of and the mechanisms underlying acquired resistance to two potent selective FGFR inhibitors, AZD4547 and BAY116387, in LC cell lines. The antitumor activity of AZD4547 and BAY1163877 was screened in 24 LC cell lines, including 5 with FGFR1 amplification. Two cell lines containing FGFR1 amplifications, H1581 and DMS114, were sensitive to FGFR inhibitors (IC50<250 nm). Clones of FGFR1-amplified H1581 cells resistant to AZD4547 or BAY116387 (H1581AR and H1581BR cells, respectively) were established. Receptor tyrosine kinase (RTK) array and immunoblotting analyses showed strong overexpression and activation of Met in H1581AR/BR cells, compared with that in the parental cells. Gene set enrichment analysis against the Kyoto Encyclopedia of Genes and Genomes (KEGG) database showed that cytokine-cytokine receptor interaction pathways were significantly enriched in H1581AR/BR cells, with Met contributing significantly to the core enrichment. Genomic DNA quantitative PCR and fluorescent in situ hybridization analyses showed MET amplification in H1581AR, but not in H1581BR, cells. Met amplification drives acquired resistance to AZD4547 in H1581AR cells by activating ErbB3. Combination treatment with FGFR inhibitors and an anaplastic lymphoma kinase (ALK)/Met inhibitor, crizotinib, or Met-specific short interfering RNA (siRNA) synergistically inhibited cell proliferation in both H1581AR and H1581BR cells. Conversely, ectopic expression of Met in H1581 cells conferred resistance to AZD4547 and BAY1163877. Acquired resistance to FGFR inhibitors not only altered cellular morphology, but also promoted migration and invasion of resistant clones, in part by inducing epithelial-to-mesenchymal transition. Taken together, our data suggest that Met activation is sufficient to bypass dependency on FGFR signaling. Concurrent inhibition of the Met and FGFR pathways may have synergistic clinical benefits when targeting FGFR-dependent LC.
Collapse
Affiliation(s)
- S-M Kim
- JE-UK Institute for Cancer Research, JEUK Co., Ltd., Gumi, Kyungbuk, Korea
| | - H Kim
- JE-UK Institute for Cancer Research, JEUK Co., Ltd., Gumi, Kyungbuk, Korea
| | - M R Yun
- JE-UK Institute for Cancer Research, JEUK Co., Ltd., Gumi, Kyungbuk, Korea
| | - H N Kang
- JE-UK Institute for Cancer Research, JEUK Co., Ltd., Gumi, Kyungbuk, Korea
| | - K-H Pyo
- JE-UK Institute for Cancer Research, JEUK Co., Ltd., Gumi, Kyungbuk, Korea
| | - H J Park
- JE-UK Institute for Cancer Research, JEUK Co., Ltd., Gumi, Kyungbuk, Korea
| | - J M Lee
- JE-UK Institute for Cancer Research, JEUK Co., Ltd., Gumi, Kyungbuk, Korea
| | - H M Choi
- JE-UK Institute for Cancer Research, JEUK Co., Ltd., Gumi, Kyungbuk, Korea
| | - P Ellinghaus
- Bayer Pharma AG, Global Drug Discovery, Wuppertal, Germany
| | - M Ocker
- Bayer Pharma AG, Global Drug Discovery, Wuppertal, Germany
| | - S Paik
- Division of Pathology NSABP, Pittsburgh, PA, USA
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea
| | - H R Kim
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea
| | - B C Cho
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
2125
|
Gainor JF, Dardaei L, Yoda S, Friboulet L, Leshchiner I, Katayama R, Dagogo-Jack I, Gadgeel S, Schultz K, Singh M, Chin E, Parks M, Lee D, DiCecca RH, Lockerman E, Huynh T, Logan J, Ritterhouse LL, Le LP, Muniappan A, Digumarthy S, Channick C, Keyes C, Getz G, Dias-Santagata D, Heist RS, Lennerz J, Sequist LV, Benes CH, Iafrate AJ, Mino-Kenudson M, Engelman JA, Shaw AT. Molecular Mechanisms of Resistance to First- and Second-Generation ALK Inhibitors in ALK-Rearranged Lung Cancer. Cancer Discov 2016; 6:1118-1133. [PMID: 27432227 DOI: 10.1158/2159-8290.cd-16-0596] [Citation(s) in RCA: 865] [Impact Index Per Article: 96.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 07/14/2016] [Indexed: 11/16/2022]
Abstract
Advanced, anaplastic lymphoma kinase (ALK)-positive lung cancer is currently treated with the first-generation ALK inhibitor crizotinib followed by more potent, second-generation ALK inhibitors (e.g., ceritinib and alectinib) upon progression. Second-generation inhibitors are generally effective even in the absence of crizotinib-resistant ALK mutations, likely reflecting incomplete inhibition of ALK by crizotinib in many cases. Herein, we analyzed 103 repeat biopsies from ALK-positive patients progressing on various ALK inhibitors. We find that each ALK inhibitor is associated with a distinct spectrum of ALK resistance mutations and that the frequency of one mutation, ALKG1202R, increases significantly after treatment with second-generation agents. To investigate strategies to overcome resistance to second-generation ALK inhibitors, we examine the activity of the third-generation ALK inhibitor lorlatinib in a series of ceritinib-resistant, patient-derived cell lines, and observe that the presence of ALK resistance mutations is highly predictive for sensitivity to lorlatinib, whereas those cell lines without ALK mutations are resistant. SIGNIFICANCE Secondary ALK mutations are a common resistance mechanism to second-generation ALK inhibitors and predict for sensitivity to the third-generation ALK inhibitor lorlatinib. These findings highlight the importance of repeat biopsies and genotyping following disease progression on targeted therapies, particularly second-generation ALK inhibitors. Cancer Discov; 6(10); 1118-33. ©2016 AACRSee related commentary by Qiao and Lovly, p. 1084This article is highlighted in the In This Issue feature, p. 1069.
Collapse
Affiliation(s)
- Justin F Gainor
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Leila Dardaei
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Satoshi Yoda
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Luc Friboulet
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts. Gustave Roussy Cancer Campus, Université Paris Saclay, INSERM U981, Paris, France
| | - Ignaty Leshchiner
- Broad Institute of the Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts
| | - Ryohei Katayama
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts. Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Ibiayi Dagogo-Jack
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Shirish Gadgeel
- Department of Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan
| | - Katherine Schultz
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Manrose Singh
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Emily Chin
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Melissa Parks
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Dana Lee
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Richard H DiCecca
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Elizabeth Lockerman
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Tiffany Huynh
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts
| | - Jennifer Logan
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | | | - Long P Le
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts
| | - Ashok Muniappan
- Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts
| | - Subba Digumarthy
- Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts
| | - Colleen Channick
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Colleen Keyes
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Gad Getz
- Broad Institute of the Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts
| | - Dora Dias-Santagata
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts
| | - Rebecca S Heist
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Jochen Lennerz
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts
| | - Lecia V Sequist
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Cyril H Benes
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - A John Iafrate
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts
| | - Mari Mino-Kenudson
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts
| | - Jeffrey A Engelman
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts.
| | - Alice T Shaw
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts.
| |
Collapse
|
2126
|
Langsch S, Baumgartner U, Haemmig S, Schlup C, Schäfer SC, Berezowska S, Rieger G, Dorn P, Tschan MP, Vassella E. miR-29b Mediates NF-κB Signaling in KRAS-Induced Non–Small Cell Lung Cancers. Cancer Res 2016; 76:4160-9. [DOI: 10.1158/0008-5472.can-15-2580] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 05/04/2016] [Indexed: 11/16/2022]
Abstract
Abstract
A global understanding of miRNA function in EGFR signaling pathways may provide insights into improving the management of KRAS-mutant lung cancers, which remain relatively recalcitrant to treatment. To identify miRNAs implicated in EGFR signaling, we transduced bronchial epithelial BEAS-2B cells with retroviral vectors expressing KRASG12V and monitored miRNA expression patterns by microarray analysis. Through this approach, we defined miR-29b as an important target for upregulation by mutant KRAS in non–small cell lung cancers. Cell biologic analyses showed that pharmacologic inhibition of EGFR or MEK was sufficient to reduce levels of miR-29b, while PI3K inhibition had no effect. In KRASG12V-transduced BEAS-2B cells, introduction of anti-miR-29b constructs increased the sensitivity to apoptosis, arguing that miR-29b mediated apoptotic resistance conferred by mutant KRAS. Mechanistic investigations traced this effect to the ability of miR-29b to target TNFAIP3/A20, a negative regulator of NF-κB signaling. Accordingly, overexpression of an miR-29b–refractory isoform of TNFAIP3 restored NF-κB and extrinsic apoptosis, confirming that TNFAIP3 is a functionally relevant target of miR-29b. We also noted that miR-29b could confer sensitivity to intrinsic apoptosis triggered by exposure to cisplatin, a drug used widely in lung cancer treatment. Thus, miR-29b expression may tilt cells from extrinsic to intrinsic mechanisms of apoptosis. Overall, our results reveal a complexity in cancer for miR-29b, which can act as either an oncogene or tumor suppressor gene depending on signaling context. Cancer Res; 76(14); 4160–9. ©2016 AACR.
Collapse
Affiliation(s)
- Stephanie Langsch
- 1Institute of Pathology, University of Bern, Bern, Switzerland
- 2Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Ulrich Baumgartner
- 1Institute of Pathology, University of Bern, Bern, Switzerland
- 2Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Stefan Haemmig
- 1Institute of Pathology, University of Bern, Bern, Switzerland
| | - Cornelia Schlup
- 1Institute of Pathology, University of Bern, Bern, Switzerland
| | | | | | - Gregor Rieger
- 1Institute of Pathology, University of Bern, Bern, Switzerland
| | - Patrick Dorn
- 3Universitätsklinik für Thoraxchirurgie, Inselspital Bern, Bern, Switzerland
| | - Mario P. Tschan
- 1Institute of Pathology, University of Bern, Bern, Switzerland
| | - Erik Vassella
- 1Institute of Pathology, University of Bern, Bern, Switzerland
| |
Collapse
|
2127
|
Altered expression of programmed death-ligand 1 after neo-adjuvant chemotherapy in patients with lung squamous cell carcinoma. Lung Cancer 2016; 99:166-71. [PMID: 27565935 DOI: 10.1016/j.lungcan.2016.07.013] [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: 03/12/2016] [Revised: 06/16/2016] [Accepted: 07/14/2016] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Programmed death-ligand 1 (PD-L1) is known to be over-expressed in non-small cell lung cancer (NSCLC). However, the impact of chemotherapy on the altered status of PD-L1 expression has not been examined for NSCLC. The present study was intended to examine the impact of neoadjuvant chemotherapy on PD-L1 expression and its prognostic significance in lung squamous cell carcinoma (SCC). PATIENTS AND METHODS Matched tumor samples were obtained from 76 SCC patients prior to and after neoadjuvant chemotherapy. The expression of PD-L1 was evaluated by immunohistochemistry. Survival analysis was performed by the Kaplan-Meier method. RESULTS Prior to neoadjuvant chemotherapy, PD-L1 expression was identified in 52.6% (40/76) of SCC patients while 61.8% (47/76) were positive for PD-L1 expression after neoadjuvant chemotherapy. Nine patients switched from negative to positive while another two patients' samples showed the reverse of the above result. Multivariate analysis demonstrated that postoperative expression of PD-L1 was an independent prognostic factor for overall survival (HR=0.50, P=0.003), but not for PD-L1 expression prior to neoadjuvant chemotherapy. CONCLUSION Neoadjuvant chemotherapy may up-regulate the expression of PD-L1. As compared with the status of PD-L1 expression prior to chemotherapy, the postoperative expression of PD-L1 is a better prognostic factor for overall survival in SCC.
Collapse
|
2128
|
Zappa C, Mousa SA. Non-small cell lung cancer: current treatment and future advances. Transl Lung Cancer Res 2016; 5:288-300. [PMID: 27413711 DOI: 10.21037/tlcr.2016.06.07] [Citation(s) in RCA: 1223] [Impact Index Per Article: 135.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Lung cancer has a poor prognosis; over half of people diagnosed with lung cancer die within one year of diagnosis and the 5-year survival is less than 18%. Non-small cell lung cancer (NSCLC) accounts for the majority of all lung cancer cases. Risk factors for developing NSCLC have been identified, with cigarette smoking being a major factor along with other environmental and genetic risk factors. Depending on the staging of lung cancer, patients are eligible for certain treatments ranging from surgery to radiation to chemotherapy as well as targeted therapy. With the advancement of genetics and biomarkers testing, specific mutations have been identified to better target treatment for individual patients. This review discusses current treatments including surgery, chemotherapy, radiotherapy, and immunotherapy as well as how biomarker testing has helped improve survival in patients with NSCLC.
Collapse
Affiliation(s)
- Cecilia Zappa
- The Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, New York, USA
| | - Shaker A Mousa
- The Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, New York, USA
| |
Collapse
|
2129
|
Gainor JF. Alectinib-a new chapter in the management of ALK-positive lung cancer. Transl Lung Cancer Res 2016; 5:343-6. [PMID: 27411885 DOI: 10.21037/tlcr.2016.03.05] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Justin F Gainor
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
2130
|
Facchinetti F, Tiseo M, Di Maio M, Graziano P, Bria E, Rossi G, Novello S. Tackling ALK in non-small cell lung cancer: the role of novel inhibitors. Transl Lung Cancer Res 2016; 5:301-21. [PMID: 27413712 DOI: 10.21037/tlcr.2016.06.10] [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] [Indexed: 12/12/2022]
Abstract
Crizotinib is an oral inhibitor of anaplastic lymphoma kinase (ALK) with remarkable clinical activity in patients suffering from ALK-rearranged non-small cell lung cancer (NSCLC), accounting to its superiority compared to chemotherapy. Unfortunately, virtually all ALK-rearranged tumors acquire resistance to crizotinib, frequently within one year since the treatment initiation. To date, therapeutic strategies to overcome crizotinib resistance have focused on the use of more potent and structurally different compounds. Second-generation ALK inhibitors such as ceritinib (LDK378), alectinib (CH5424802/RO5424802) and brigatinib (AP26113) have shown relevant clinical activity, consequently fostering their rapid clinical development and their approval by health agencies. The third-generation inhibitor lorlatinib (PF-06463922), selectively active against ALK and ROS1, harbors impressive biological potency; its efficacy in reversing resistance to crizotinib and to other ALK inhibitors is being proven by early clinical trials. The NTRK1-3 and ROS1 inhibitor entrectinib (RXDX-101) has been reported to act against NSCLC harboring ALK fusion proteins too. Despite the quick development of these novel agents, several issues remain to be discussed in the treatment of patients suffering from ALK-rearranged NSCLC. This position paper will discuss the development, the current evidence and approvals, as long as the future perspectives of new ALK inhibitors beyond crizotinib. Clinical behaviors of ALK-rearranged NSCLC vary significantly among patients and differential molecular events responsible of crizotinib resistance account for the most important quote of this heterogeneity. The precious availability of a wide range of active anti-ALK compounds should be approached in a critical and careful perspective, in order to develop treatment strategies tailored on the disease evolution of every single patient.
Collapse
Affiliation(s)
- Francesco Facchinetti
- 1 INSERM U981, Gustave Roussy Cancer Campus, Université Paris-Sud, Villejuif, France ; 2 Medical Oncology Unit, University Hospital of Parma, Parma, Italy ; 3 Department of Oncology, AOU San Luigi (Orbassano), University of Turin, Italy ; 4 Medical Oncology, AO Ordine Mauriziano, Turin, Italy ; 5 Division of Pathology, IRCCS "Casa Sollievo della Sofferenza", San Giovanni Rotondo, Foggia, Italy ; 6 Medical Oncology, Azienda Ospedaliera Universitaria Integrata, University of Verona, Verona, Italy ; 7 Operative Unit of Pathology, Azienda USL Valle d'Aosta, Aosta, Italy
| | - Marcello Tiseo
- 1 INSERM U981, Gustave Roussy Cancer Campus, Université Paris-Sud, Villejuif, France ; 2 Medical Oncology Unit, University Hospital of Parma, Parma, Italy ; 3 Department of Oncology, AOU San Luigi (Orbassano), University of Turin, Italy ; 4 Medical Oncology, AO Ordine Mauriziano, Turin, Italy ; 5 Division of Pathology, IRCCS "Casa Sollievo della Sofferenza", San Giovanni Rotondo, Foggia, Italy ; 6 Medical Oncology, Azienda Ospedaliera Universitaria Integrata, University of Verona, Verona, Italy ; 7 Operative Unit of Pathology, Azienda USL Valle d'Aosta, Aosta, Italy
| | - Massimo Di Maio
- 1 INSERM U981, Gustave Roussy Cancer Campus, Université Paris-Sud, Villejuif, France ; 2 Medical Oncology Unit, University Hospital of Parma, Parma, Italy ; 3 Department of Oncology, AOU San Luigi (Orbassano), University of Turin, Italy ; 4 Medical Oncology, AO Ordine Mauriziano, Turin, Italy ; 5 Division of Pathology, IRCCS "Casa Sollievo della Sofferenza", San Giovanni Rotondo, Foggia, Italy ; 6 Medical Oncology, Azienda Ospedaliera Universitaria Integrata, University of Verona, Verona, Italy ; 7 Operative Unit of Pathology, Azienda USL Valle d'Aosta, Aosta, Italy
| | - Paolo Graziano
- 1 INSERM U981, Gustave Roussy Cancer Campus, Université Paris-Sud, Villejuif, France ; 2 Medical Oncology Unit, University Hospital of Parma, Parma, Italy ; 3 Department of Oncology, AOU San Luigi (Orbassano), University of Turin, Italy ; 4 Medical Oncology, AO Ordine Mauriziano, Turin, Italy ; 5 Division of Pathology, IRCCS "Casa Sollievo della Sofferenza", San Giovanni Rotondo, Foggia, Italy ; 6 Medical Oncology, Azienda Ospedaliera Universitaria Integrata, University of Verona, Verona, Italy ; 7 Operative Unit of Pathology, Azienda USL Valle d'Aosta, Aosta, Italy
| | - Emilio Bria
- 1 INSERM U981, Gustave Roussy Cancer Campus, Université Paris-Sud, Villejuif, France ; 2 Medical Oncology Unit, University Hospital of Parma, Parma, Italy ; 3 Department of Oncology, AOU San Luigi (Orbassano), University of Turin, Italy ; 4 Medical Oncology, AO Ordine Mauriziano, Turin, Italy ; 5 Division of Pathology, IRCCS "Casa Sollievo della Sofferenza", San Giovanni Rotondo, Foggia, Italy ; 6 Medical Oncology, Azienda Ospedaliera Universitaria Integrata, University of Verona, Verona, Italy ; 7 Operative Unit of Pathology, Azienda USL Valle d'Aosta, Aosta, Italy
| | - Giulio Rossi
- 1 INSERM U981, Gustave Roussy Cancer Campus, Université Paris-Sud, Villejuif, France ; 2 Medical Oncology Unit, University Hospital of Parma, Parma, Italy ; 3 Department of Oncology, AOU San Luigi (Orbassano), University of Turin, Italy ; 4 Medical Oncology, AO Ordine Mauriziano, Turin, Italy ; 5 Division of Pathology, IRCCS "Casa Sollievo della Sofferenza", San Giovanni Rotondo, Foggia, Italy ; 6 Medical Oncology, Azienda Ospedaliera Universitaria Integrata, University of Verona, Verona, Italy ; 7 Operative Unit of Pathology, Azienda USL Valle d'Aosta, Aosta, Italy
| | - Silvia Novello
- 1 INSERM U981, Gustave Roussy Cancer Campus, Université Paris-Sud, Villejuif, France ; 2 Medical Oncology Unit, University Hospital of Parma, Parma, Italy ; 3 Department of Oncology, AOU San Luigi (Orbassano), University of Turin, Italy ; 4 Medical Oncology, AO Ordine Mauriziano, Turin, Italy ; 5 Division of Pathology, IRCCS "Casa Sollievo della Sofferenza", San Giovanni Rotondo, Foggia, Italy ; 6 Medical Oncology, Azienda Ospedaliera Universitaria Integrata, University of Verona, Verona, Italy ; 7 Operative Unit of Pathology, Azienda USL Valle d'Aosta, Aosta, Italy
| |
Collapse
|
2131
|
Zhang YC, Zhou Q, Wu YL. Efficacy of crizotinib in first-line treatment of adults with ALK-positive advanced NSCLC. Expert Opin Pharmacother 2016; 17:1693-701. [PMID: 27359268 DOI: 10.1080/14656566.2016.1208171] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
INTRODUCTION The treatment of advanced non-small cell lung cancer (NSCLC) has evolved from palliative cytotoxic chemotherapy to precise medicine based on genetic alternations over the last decade. Anaplastic lymphoma kinase (ALK) rearrangement characterizes a molecular subset of NSCLC with an impressive response to crizotinib. AREAS COVERED To analyze the efficacy of crizotinib in first-line treatment of adults with advanced ALK-positive NSCLC, updated data on development and recent advances of first-line crizotinib in this subset population are reviewed. EXPERT OPINION To date, crizotinib should be established as a standard of care in previously untreated advanced NSCLC with ALK-rearrangement. However, the efficacy of first-line crizotinib is limited by acquired resistance. Second generation ALK inhibitors have demonstrated clinical activity in both crizotinib-refractory and crizotinib naïve setting. How to maximize first-line benefit for advanced ALK-positive NSCLC remains challenging. Combinational strategy, advances in companion diagnostics and optimization of ALK inhibitors might contribute to improve outcome in this subset of patients in future.
Collapse
Affiliation(s)
- Yi-Chen Zhang
- a Graduate School , Southern Medical University , Guangzhou , PR China.,b Guangdong Lung Cancer Institute , Guangdong General Hospital and Guangdong Academy of Medical Sciences , Guangzhou , PR China
| | - Qing Zhou
- b Guangdong Lung Cancer Institute , Guangdong General Hospital and Guangdong Academy of Medical Sciences , Guangzhou , PR China
| | - Yi-Long Wu
- b Guangdong Lung Cancer Institute , Guangdong General Hospital and Guangdong Academy of Medical Sciences , Guangzhou , PR China
| |
Collapse
|
2132
|
Larkins E, Blumenthal GM, Chen H, He K, Agarwal R, Gieser G, Stephens O, Zahalka E, Ringgold K, Helms W, Shord S, Yu J, Zhao H, Davis G, McKee AE, Keegan P, Pazdur R. FDA Approval: Alectinib for the Treatment of Metastatic, ALK-Positive Non-Small Cell Lung Cancer Following Crizotinib. Clin Cancer Res 2016; 22:5171-5176. [PMID: 27413075 DOI: 10.1158/1078-0432.ccr-16-1293] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 06/25/2016] [Indexed: 11/16/2022]
Abstract
On December 11, 2015, the FDA granted accelerated approval to alectinib (Alecensa; Genentech) for the treatment of patients with anaplastic lymphoma receptor tyrosine kinase (ALK)-positive, metastatic non-small cell lung cancer (NSCLC) who have progressed on or are intolerant to crizotinib. This approval was based on two single-arm trials including 225 patients treated with alectinib 600 mg orally twice daily. The objective response rates (ORR) by an independent review committee in these studies were 38% [95% confidence interval (CI), 28-49] and 44% (95% CI, 36-53); the median durations of response (DOR) were 7.5 months and 11.2 months. In a pooled analysis of 51 patients with measurable disease in the central nervous system (CNS) at baseline, the CNS ORR was 61% (95% CI, 46-74); the CNS DOR was 9.1 months. The primary safety analysis population included 253 patients. The most common adverse reactions were fatigue (41%), constipation (34%), edema (30%), and myalgia (29%). The most common laboratory abnormalities were anemia (56%), increased aspartate aminotransferase (51%), increased alkaline phosphatase (47%), increased creatine phosphokinase (43%), hyperbilirubinemia (39%), hyperglycemia (36%), increased alanine aminotransferase (34%), and hypocalcemia (32%). Dose reductions due to adverse reactions occurred in 12% of patients, whereas 27% of patients had alectinib dosing interrupted for adverse reactions. Permanent discontinuation of alectinib due to adverse reactions occurred in only 6% of patients. With the clinically meaningful ORR and DOR as well as the safety profile observed in these trials, alectinib was determined to have a favorable benefit-risk profile for the treatment of the indicated population. Clin Cancer Res; 22(21); 5171-6. ©2016 AACR.
Collapse
Affiliation(s)
- Erin Larkins
- Office of Hematology and Oncology Products, U.S. Food and Drug Administration, Silver Spring, Maryland.
| | - Gideon M Blumenthal
- Office of Hematology and Oncology Products, U.S. Food and Drug Administration, Silver Spring, Maryland
| | - Huanyu Chen
- Office of Biostatistics, U.S. Food and Drug Administration, Silver Spring, Maryland
| | - Kun He
- Office of Biostatistics, U.S. Food and Drug Administration, Silver Spring, Maryland
| | - Rajiv Agarwal
- Office of Pharmaceutical Quality, U.S. Food and Drug Administration, Silver Spring, Maryland
| | - Gerlie Gieser
- Office of Pharmaceutical Quality, U.S. Food and Drug Administration, Silver Spring, Maryland
| | - Olen Stephens
- Office of Pharmaceutical Quality, U.S. Food and Drug Administration, Silver Spring, Maryland
| | - Eias Zahalka
- Office of Hematology and Oncology Products, U.S. Food and Drug Administration, Silver Spring, Maryland
| | - Kimberly Ringgold
- Office of Hematology and Oncology Products, U.S. Food and Drug Administration, Silver Spring, Maryland
| | - Whitney Helms
- Office of Hematology and Oncology Products, U.S. Food and Drug Administration, Silver Spring, Maryland
| | - Stacy Shord
- Office of Translational Sciences, U.S. Food and Drug Administration, Silver Spring, Maryland
| | - Jingyu Yu
- Office of Translational Sciences, U.S. Food and Drug Administration, Silver Spring, Maryland
| | - Hong Zhao
- Office of Translational Sciences, U.S. Food and Drug Administration, Silver Spring, Maryland
| | - Gina Davis
- Office of Hematology and Oncology Products, U.S. Food and Drug Administration, Silver Spring, Maryland
| | - Amy E McKee
- Office of Hematology and Oncology Products, U.S. Food and Drug Administration, Silver Spring, Maryland
| | - Patricia Keegan
- Office of Hematology and Oncology Products, U.S. Food and Drug Administration, Silver Spring, Maryland
| | - Richard Pazdur
- Office of Hematology and Oncology Products, U.S. Food and Drug Administration, Silver Spring, Maryland
| |
Collapse
|
2133
|
Ni J, Xie S, Ramkissoon SH, Luu V, Sun Y, Bandopadhayay P, Beroukhim R, Roberts TM, Stiles CD, Segal RA, Ligon KL, Hahn WC, Zhao JJ. Tyrosine receptor kinase B is a drug target in astrocytomas. Neuro Oncol 2016; 19:22-30. [PMID: 27402815 DOI: 10.1093/neuonc/now139] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Astrocytomas are the most common primary human brain tumors. Receptor tyrosine kinases (RTKs), including tyrosine receptor kinase B (TrkB, also known as tropomyosin-related kinase B; encoded by neurotrophic tyrosine kinase receptor type 2 [NTRK2]), are frequently mutated by rearrangement/fusion in high-grade and low-grade astrocytomas. We found that activated TrkB can contribute to the development of astrocytoma and might serve as a therapeutic target in this tumor type. METHODS To identify RTKs capable of inducing astrocytoma formation, a library of human tyrosine kinases was screened for the ability to transform murine Ink4a-/-/Arf-/- astrocytes. Orthotopic allograft studies were conducted to evaluate the effects of RTKs on the development of astrocytoma. Since TrkB was identified as a driver of astrocytoma formation, the effect of the Trk inhibitors AZD1480 and RXDX-101 was assessed in astrocytoma cells expressing activated TrkB. RNA sequencing, real-time PCR, western blotting, and enzyme-linked immunosorbent assays were conducted to characterize NTRK2 in astrocytomas. RESULTS Activated TrkB cooperated with Ink4a/Arf loss to induce the formation of astrocytomas through a mechanism mediated by activation of signal transducer and activator of transcription 3 (STAT3). TrkB activation positively correlated with Ccl2 expression. TrkB-induced astrocytomas remained dependent on TrkB signaling for survival, highlighting a role of NTRK2 as an addictive oncogene. Furthermore, the QKI-NTRK2 fusion associated with human astrocytoma transformed Ink4a-/-/Arf-/- astrocytes, and this process was also mediated via STAT3 signaling. CONCLUSIONS Our findings provide evidence that constitutively activated NTRK2 alleles, notably the human tumor-associated QKI-NTRK2 fusion, can cooperate with Ink4a/Arf loss to drive astrocytoma formation. Therefore, we propose NTRK2 as a potential therapeutic target in the subset of astrocytoma patients defined by QKI-NTRK2 fusion.
Collapse
Affiliation(s)
- Jing Ni
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts (J.N., S.X., V.L., Y.S., P.B., R.B., T.M.R., C.D.S., R.A.S., J.J.Z.); Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts (J.N., S.X., V.L., T.M.R., J.J.Z.); Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts (S.H.R., R.B., K.L.L., W.C.H.); Broad Institute, Boston, Massachusetts (P.B., R.B., W.C.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (S.H.R., K.L.L.); Department of Pathology, Boston Children's Hospital, Boston, Massachusetts (K.L.L.)
| | - Shaozhen Xie
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts (J.N., S.X., V.L., Y.S., P.B., R.B., T.M.R., C.D.S., R.A.S., J.J.Z.); Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts (J.N., S.X., V.L., T.M.R., J.J.Z.); Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts (S.H.R., R.B., K.L.L., W.C.H.); Broad Institute, Boston, Massachusetts (P.B., R.B., W.C.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (S.H.R., K.L.L.); Department of Pathology, Boston Children's Hospital, Boston, Massachusetts (K.L.L.)
| | - Shakti H Ramkissoon
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts (J.N., S.X., V.L., Y.S., P.B., R.B., T.M.R., C.D.S., R.A.S., J.J.Z.); Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts (J.N., S.X., V.L., T.M.R., J.J.Z.); Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts (S.H.R., R.B., K.L.L., W.C.H.); Broad Institute, Boston, Massachusetts (P.B., R.B., W.C.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (S.H.R., K.L.L.); Department of Pathology, Boston Children's Hospital, Boston, Massachusetts (K.L.L.)
| | - Victor Luu
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts (J.N., S.X., V.L., Y.S., P.B., R.B., T.M.R., C.D.S., R.A.S., J.J.Z.); Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts (J.N., S.X., V.L., T.M.R., J.J.Z.); Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts (S.H.R., R.B., K.L.L., W.C.H.); Broad Institute, Boston, Massachusetts (P.B., R.B., W.C.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (S.H.R., K.L.L.); Department of Pathology, Boston Children's Hospital, Boston, Massachusetts (K.L.L.)
| | - Yu Sun
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts (J.N., S.X., V.L., Y.S., P.B., R.B., T.M.R., C.D.S., R.A.S., J.J.Z.); Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts (J.N., S.X., V.L., T.M.R., J.J.Z.); Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts (S.H.R., R.B., K.L.L., W.C.H.); Broad Institute, Boston, Massachusetts (P.B., R.B., W.C.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (S.H.R., K.L.L.); Department of Pathology, Boston Children's Hospital, Boston, Massachusetts (K.L.L.)
| | - Pratiti Bandopadhayay
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts (J.N., S.X., V.L., Y.S., P.B., R.B., T.M.R., C.D.S., R.A.S., J.J.Z.); Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts (J.N., S.X., V.L., T.M.R., J.J.Z.); Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts (S.H.R., R.B., K.L.L., W.C.H.); Broad Institute, Boston, Massachusetts (P.B., R.B., W.C.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (S.H.R., K.L.L.); Department of Pathology, Boston Children's Hospital, Boston, Massachusetts (K.L.L.)
| | - Rameen Beroukhim
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts (J.N., S.X., V.L., Y.S., P.B., R.B., T.M.R., C.D.S., R.A.S., J.J.Z.); Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts (J.N., S.X., V.L., T.M.R., J.J.Z.); Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts (S.H.R., R.B., K.L.L., W.C.H.); Broad Institute, Boston, Massachusetts (P.B., R.B., W.C.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (S.H.R., K.L.L.); Department of Pathology, Boston Children's Hospital, Boston, Massachusetts (K.L.L.)
| | - Thomas M Roberts
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts (J.N., S.X., V.L., Y.S., P.B., R.B., T.M.R., C.D.S., R.A.S., J.J.Z.); Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts (J.N., S.X., V.L., T.M.R., J.J.Z.); Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts (S.H.R., R.B., K.L.L., W.C.H.); Broad Institute, Boston, Massachusetts (P.B., R.B., W.C.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (S.H.R., K.L.L.); Department of Pathology, Boston Children's Hospital, Boston, Massachusetts (K.L.L.)
| | - Charles D Stiles
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts (J.N., S.X., V.L., Y.S., P.B., R.B., T.M.R., C.D.S., R.A.S., J.J.Z.); Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts (J.N., S.X., V.L., T.M.R., J.J.Z.); Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts (S.H.R., R.B., K.L.L., W.C.H.); Broad Institute, Boston, Massachusetts (P.B., R.B., W.C.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (S.H.R., K.L.L.); Department of Pathology, Boston Children's Hospital, Boston, Massachusetts (K.L.L.)
| | - Rosalind A Segal
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts (J.N., S.X., V.L., Y.S., P.B., R.B., T.M.R., C.D.S., R.A.S., J.J.Z.); Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts (J.N., S.X., V.L., T.M.R., J.J.Z.); Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts (S.H.R., R.B., K.L.L., W.C.H.); Broad Institute, Boston, Massachusetts (P.B., R.B., W.C.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (S.H.R., K.L.L.); Department of Pathology, Boston Children's Hospital, Boston, Massachusetts (K.L.L.)
| | - Keith L Ligon
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts (J.N., S.X., V.L., Y.S., P.B., R.B., T.M.R., C.D.S., R.A.S., J.J.Z.); Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts (J.N., S.X., V.L., T.M.R., J.J.Z.); Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts (S.H.R., R.B., K.L.L., W.C.H.); Broad Institute, Boston, Massachusetts (P.B., R.B., W.C.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (S.H.R., K.L.L.); Department of Pathology, Boston Children's Hospital, Boston, Massachusetts (K.L.L.)
| | - William C Hahn
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts (J.N., S.X., V.L., Y.S., P.B., R.B., T.M.R., C.D.S., R.A.S., J.J.Z.); Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts (J.N., S.X., V.L., T.M.R., J.J.Z.); Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts (S.H.R., R.B., K.L.L., W.C.H.); Broad Institute, Boston, Massachusetts (P.B., R.B., W.C.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (S.H.R., K.L.L.); Department of Pathology, Boston Children's Hospital, Boston, Massachusetts (K.L.L.)
| | - Jean J Zhao
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts (J.N., S.X., V.L., Y.S., P.B., R.B., T.M.R., C.D.S., R.A.S., J.J.Z.); Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts (J.N., S.X., V.L., T.M.R., J.J.Z.); Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts (S.H.R., R.B., K.L.L., W.C.H.); Broad Institute, Boston, Massachusetts (P.B., R.B., W.C.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (S.H.R., K.L.L.); Department of Pathology, Boston Children's Hospital, Boston, Massachusetts (K.L.L.)
| |
Collapse
|
2134
|
Park S, Kim HJ, Choi CM, Lee DH, Kim SW, Lee JS, Kim WS, Choi SH, Rho JK, Lee JC. Predictive factors for a long-term response duration in non-squamous cell lung cancer patients treated with pemetrexed. BMC Cancer 2016; 16:417. [PMID: 27388008 PMCID: PMC4936194 DOI: 10.1186/s12885-016-2457-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 06/24/2016] [Indexed: 12/19/2022] Open
Abstract
Background Pemetrexed is widely used for the treatment of advanced non-squamous non-small-cell lung cancer (NSCLC). However, factors that can predict the benefits of pemetrexed therapy have not yet been defined. Methods We compared the clinical and molecule pathological characteristics of good and poor responders among a cohort of 1,848 non-squamous NSCLC patients who had received at least two cycles of pemetrexed therapy between November 2006 and February 2015. Among these cases, 92 good responders who were the top 5 % in terms of progression-free survival (PFS) and 222 poor responders who had progressive disease after only 2 cycles of therapy were selected for the analysis. Results The median PFS of the good responders was 29.9 months (range; 20.9–90.0) and the median number of cycle was 37 (range; 18–129). Although 53.5 % of patients showed stable disease (SD), this response was sustained (median PFS in SD, 29.6 months). A never-smoking status was related to better survival outcome, whereas EGFR mutation, two or more metastatic sites, and intra-abdominal metastasis were each associated with a poor PFS. ALK translocation showed a tendency for a positive impact on response to pemetrexed, whereas metastatic lesion to liver, adrenal gland or bone showed a tendency for a negative impact despite not reaching our threshold for statistical significance. Conclusions Predictive factors, such as smoking status, the status of genetic alteration and tumor burden, should be considered when administering pemetrexed therapy for non-squamous NSCLC.
Collapse
Affiliation(s)
- Sojung Park
- Department of Pulmonary and Critical Care Medicine, University of Ulsan, College of Medicine, Asan Medical Center, 388-1 Pungnap-Dong, Songpa-gu, Seoul, 05505, South Korea
| | - Hyun Jung Kim
- Department of Internal Medicine, Kyungpook National University Hospital, Kyungpook National University School of Medicine, 130 Dongdeok-ro, Jung-gu, Daegu, 41944, South Korea
| | - Chang-Min Choi
- Department of Pulmonary and Critical Care Medicine, University of Ulsan, College of Medicine, Asan Medical Center, 388-1 Pungnap-Dong, Songpa-gu, Seoul, 05505, South Korea.,Department of Oncology, University of Ulsan, College of Medicine, Asan Medical Center, 388-1 Pungnap-Dong, Songpa-gu, Seoul, 05505, South Korea
| | - Dae Ho Lee
- Department of Oncology, University of Ulsan, College of Medicine, Asan Medical Center, 388-1 Pungnap-Dong, Songpa-gu, Seoul, 05505, South Korea
| | - Sang-We Kim
- Department of Oncology, University of Ulsan, College of Medicine, Asan Medical Center, 388-1 Pungnap-Dong, Songpa-gu, Seoul, 05505, South Korea
| | - Jung-Shin Lee
- Department of Oncology, University of Ulsan, College of Medicine, Asan Medical Center, 388-1 Pungnap-Dong, Songpa-gu, Seoul, 05505, South Korea
| | - Woo Sung Kim
- Department of Pulmonary and Critical Care Medicine, University of Ulsan, College of Medicine, Asan Medical Center, 388-1 Pungnap-Dong, Songpa-gu, Seoul, 05505, South Korea
| | - Se Hoon Choi
- Department of Thoracic and Cardiovascular Surgery, University of Ulsan, College of Medicine, Asan Medical Center, 388-1 Pungnap-Dong, Songpa-gu, Seoul, 05505, South Korea
| | - Jin Kyung Rho
- Asan Institute for Life Sciences, Asan Medical Center, 388-1 Pungnap-Dong, Songpa-gu, Seoul, 05505, South Korea
| | - Jae Cheol Lee
- Department of Oncology, University of Ulsan, College of Medicine, Asan Medical Center, 388-1 Pungnap-Dong, Songpa-gu, Seoul, 05505, South Korea.
| |
Collapse
|
2135
|
Lee DW, Lee KH, Kim JW, Keam B. Molecular Targeted Therapies for the Treatment of Leptomeningeal Carcinomatosis: Current Evidence and Future Directions. Int J Mol Sci 2016; 17:ijms17071074. [PMID: 27399673 PMCID: PMC4964450 DOI: 10.3390/ijms17071074] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 06/26/2016] [Accepted: 06/28/2016] [Indexed: 12/16/2022] Open
Abstract
Leptomeningeal carcinomatosis (LMC) is the multifocal seeding of cerebrospinal fluid and leptomeninges by malignant cells. The incidence of LMC is approximately 5% in patients with malignant tumors overall and the rate is increasing due to increasing survival time of cancer patients. Eradication of the disease is not yet possible, so the treatment goals of LMC are to improve neurologic symptoms and to prolong survival. A standard treatment for LMC has not been established due to low incidences of LMC, the rapidly progressing nature of the disease, heterogeneous populations with LMC, and a lack of randomized clinical trial results. Treatment options for LMC include intrathecal chemotherapy, systemic chemotherapy, and radiation therapy, but the prognoses remain poor with a median survival of <3 months. Recently, molecular targeted agents have been applied in the clinic and have shown groundbreaking results in specific patient groups epidermal growth factor receptor (EGFR)-targeted therapy or an anaplastic lymphoma kinase (ALK) inhibitor in lung cancer, human epidermal growth factor receptor 2 (HER2)-directed therapy in breast cancer, and CD20-targeted therapy in B cell lymphoma). Moreover, there are results indicating that the use of these agents under proper dose and administration routes can be effective for managing LMC. In this article, we review molecular targeted agents for managing LMC.
Collapse
Affiliation(s)
- Dae-Won Lee
- Department of Internal Medicine, Seoul National University Hospital, Seoul 03080, Korea.
| | - Kyung-Hun Lee
- Department of Internal Medicine, Seoul National University Hospital, Seoul 03080, Korea.
- Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Korea.
| | - Jin Wook Kim
- Department of Neurosurgery, Seoul National University Hospital, Seoul 03080, Korea.
| | - Bhumsuk Keam
- Department of Internal Medicine, Seoul National University Hospital, Seoul 03080, Korea.
- Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Korea.
| |
Collapse
|
2136
|
Kiesewetter B, Raderer M, Steger GG, Bartsch R, Pirker R, Zöchbauer-Müller S, Prager G, Krainer M, Preusser M, Schmidinger M, Zielinski CC. The European Society for Medical Oncology Magnitude of Clinical Benefit Scale in daily practice: a single institution, real-life experience at the Medical University of Vienna. ESMO Open 2016; 1:e000066. [PMID: 27843624 PMCID: PMC5070236 DOI: 10.1136/esmoopen-2016-000066] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 05/09/2016] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND The European Society for Medical Oncology (ESMO) Magnitude of Clinical Benefit Scale (MCBS) has been designed to stratify the therapeutic benefit of a certain drug registered for the treatment of cancer. However, though internally validated, this tool has not yet been evaluated for its feasibility in the daily practice of a major center of medical oncology. METHODS The practicability of the MCBS for advanced oncological diseases at the Clinical Division of Oncology, Medical University of Vienna, which constitutes one of the largest oncological centres in Europe, was analysed in a three-step approach. First, retrospectively collected data were analysed to gain an overview of treatments in regular use. Second, data were scored by using the MCBS. Third, the ensuing results were evaluated within corresponding programme directorships to assess feasibility in a real-life clinical context. RESULTS In the majority of tumour entities, the MCBS results reported earlier are consistent with daily clinical practice. Thus, in metastatic breast cancer or advanced lung cancer, there was a high level of clinical benefit for first-line treatment standards, and these results reflected well real-life experience. However, analyses based on the first version of the MCBS are limited if it comes to salvage treatment in tumour entities in which optimal sequencing of potential treatment options is of major importance, as in metastatic colorectal or renal cell cancer. In contrast to this, it is remarkable that certain novel therapies such as nivolumab assessed for heavily pretreated advanced renal cancer reached the highest level of clinical benefit due to prolongation in survival and a favourable toxicity profile. The MCBS clearly underlines the potential benefit of these compounds. CONCLUSIONS The MCBS is an excellent tool for daily clinical practice of a tertiary referral centre. It supports treatment decisions based on the clinical benefit to be expected from a novel approach such as immunotherapy in as yet untested indications.
Collapse
Affiliation(s)
- Barbara Kiesewetter
- Clinical Division of Oncology, Department of Medicine I , Comprehensive Cancer Center, Medical University of Vienna-General Hospital , Vienna , Austria
| | - Markus Raderer
- Clinical Division of Oncology, Department of Medicine I , Comprehensive Cancer Center, Medical University of Vienna-General Hospital , Vienna , Austria
| | - Günther G Steger
- Clinical Division of Oncology, Department of Medicine I , Comprehensive Cancer Center, Medical University of Vienna-General Hospital , Vienna , Austria
| | - Rupert Bartsch
- Clinical Division of Oncology, Department of Medicine I , Comprehensive Cancer Center, Medical University of Vienna-General Hospital , Vienna , Austria
| | - Robert Pirker
- Clinical Division of Oncology, Department of Medicine I , Comprehensive Cancer Center, Medical University of Vienna-General Hospital , Vienna , Austria
| | - Sabine Zöchbauer-Müller
- Clinical Division of Oncology, Department of Medicine I , Comprehensive Cancer Center, Medical University of Vienna-General Hospital , Vienna , Austria
| | - Gerald Prager
- Clinical Division of Oncology, Department of Medicine I , Comprehensive Cancer Center, Medical University of Vienna-General Hospital , Vienna , Austria
| | - Michael Krainer
- Clinical Division of Oncology, Department of Medicine I , Comprehensive Cancer Center, Medical University of Vienna-General Hospital , Vienna , Austria
| | - Matthias Preusser
- Clinical Division of Oncology, Department of Medicine I , Comprehensive Cancer Center, Medical University of Vienna-General Hospital , Vienna , Austria
| | - Manuela Schmidinger
- Clinical Division of Oncology, Department of Medicine I , Comprehensive Cancer Center, Medical University of Vienna-General Hospital , Vienna , Austria
| | - Christoph C Zielinski
- Clinical Division of Oncology, Department of Medicine I , Comprehensive Cancer Center, Medical University of Vienna-General Hospital , Vienna , Austria
| |
Collapse
|
2137
|
Yang Y, Wu N, Shen J, Teixido C, Sun X, Lin Z, Qian X, Zou Z, Guan W, Yu L, Rosell R, Liu B, Wei J. MET overexpression and amplification define a distinct molecular subgroup for targeted therapies in gastric cancer. Gastric Cancer 2016; 19:778-88. [PMID: 26404902 DOI: 10.1007/s10120-015-0545-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 09/07/2015] [Indexed: 02/07/2023]
Abstract
BACKGROUND Currently, only trastuzumab, ramucirumab, and apatinib effectively treat gastric cancer. Thus, additional novel targets are required for this disease. METHODS We investigated the immunohistochemical and fluorescence in situ hybridization expression of MET, ROS1, and ALK in four gastric cell lines and a cohort of 98 gastric cancer patients. Crizotinib response was studied in vitro and in vivo. RESULTS Crizotinib potently inhibited in vitro cell growth in only one cell line, which also showed MET amplification. A positive correlation between crizotinib sensitivity and MET overexpression was observed (P = 0.045) in the histoculture drug response assay. Meanwhile, patient-derived tumor xenograft mouse models transplanted with tissues with higher MET protein expression displayed a highly selective sensitivity to crizotinib. In the 98 patients, MET overexpression was found in 42 (42.9 %) and MET was amplified in 4 (4.1 %). ROS1 and ALK overexpression were found in 25 (25.5 %) and 0 patients, respectively. However, none of the patients screened harbored ALK or ROS1 rearrangements. No significant association was found between overall survival and MET or ROS1 status. We also observed a stage IV gastric cancer patient with MET amplification who experienced tumor shrinkage and clinical benefit after 3 weeks of crizotinib as fourth-line treatment. CONCLUSIONS Crizotinib may induce clinically relevant anticancer effects in MET-overexpressed or MET-amplified gastric cancer patients.
Collapse
Affiliation(s)
- Yang Yang
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University and Clinical Cancer Institute of Nanjing University, 321 Zhongshan Road, Nanjing, 210008, China
| | - Nandie Wu
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University and Clinical Cancer Institute of Nanjing University, 321 Zhongshan Road, Nanjing, 210008, China
| | - Jie Shen
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University and Clinical Cancer Institute of Nanjing University, 321 Zhongshan Road, Nanjing, 210008, China
| | - Cristina Teixido
- Pangaea Biotech, USP Dexeus University Institute, Barcelona, Spain
| | - Xia Sun
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University and Clinical Cancer Institute of Nanjing University, 321 Zhongshan Road, Nanjing, 210008, China
| | - Zihan Lin
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University and Clinical Cancer Institute of Nanjing University, 321 Zhongshan Road, Nanjing, 210008, China
| | - Xiaoping Qian
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University and Clinical Cancer Institute of Nanjing University, 321 Zhongshan Road, Nanjing, 210008, China
| | - Zhengyun Zou
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University and Clinical Cancer Institute of Nanjing University, 321 Zhongshan Road, Nanjing, 210008, China
| | - Wenxian Guan
- Department of General Surgery, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Lixia Yu
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University and Clinical Cancer Institute of Nanjing University, 321 Zhongshan Road, Nanjing, 210008, China
| | - Rafael Rosell
- Pangaea Biotech, USP Dexeus University Institute, Barcelona, Spain
- Catalan Institute of Oncology, Hospital Germans Trias i Pujol, Medical Oncology Service, Badalona, Spain
| | - Baorui Liu
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University and Clinical Cancer Institute of Nanjing University, 321 Zhongshan Road, Nanjing, 210008, China
| | - Jia Wei
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University and Clinical Cancer Institute of Nanjing University, 321 Zhongshan Road, Nanjing, 210008, China.
| |
Collapse
|
2138
|
Planchard D, Besse B, Groen HJM, Souquet PJ, Quoix E, Baik CS, Barlesi F, Kim TM, Mazieres J, Novello S, Rigas JR, Upalawanna A, D'Amelio AM, Zhang P, Mookerjee B, Johnson BE. Dabrafenib plus trametinib in patients with previously treated BRAF(V600E)-mutant metastatic non-small cell lung cancer: an open-label, multicentre phase 2 trial. Lancet Oncol 2016; 17:984-993. [PMID: 27283860 PMCID: PMC4993103 DOI: 10.1016/s1470-2045(16)30146-2] [Citation(s) in RCA: 635] [Impact Index Per Article: 70.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Revised: 05/06/2016] [Accepted: 05/06/2016] [Indexed: 12/12/2022]
Abstract
BACKGROUND BRAF mutations act as an oncogenic driver via the mitogen-activated protein kinase (MAPK) pathway in non-small cell lung cancer (NSCLC). BRAF inhibition has shown antitumour activity in patients with BRAF(V600E)-mutant NSCLC. Dual MAPK pathway inhibition with BRAF and MEK inhibitors in BRAF(V600E)-mutant NSCLC might improve efficacy over BRAF inhibitor monotherapy based on observations in BRAF(V600)-mutant melanoma. We aimed to assess the antitumour activity and safety of dabrafenib plus trametinib in patients with BRAF(V600E)-mutant NSCLC. METHODS In this phase 2, multicentre, non-randomised, open-label study, we enrolled adult patients (aged ≥18 years) with pretreated metastatic stage IV BRAF(V600E)-mutant NSCLC who had documented tumour progression after at least one previous platinum-based chemotherapy and had had no more than three previous systemic anticancer therapies. Patients with previous BRAF or MEK inhibitor treatment were ineligible. Patients with brain metastases were allowed to enrol only if the lesions were asymptomatic, untreated (or stable more than 3 weeks after local therapy if treated), and measured less than 1 cm. Enrolled patients received oral dabrafenib (150 mg twice daily) plus oral trametinib (2 mg once daily) in continuous 21-day cycles until disease progression, unacceptable adverse events, withdrawal of consent, or death. The primary endpoint was investigator-assessed overall response, which was assessed by intention to treat in the protocol-defined population (patients who received second-line or later treatment); safety was also assessed in this population and was assessed at least once every 3 weeks, with adverse events, laboratory values, and vital signs graded according to the Common Terminology Criteria for Adverse Events version 4.0. The study is ongoing but no longer recruiting patients. This trial is registered with ClinicalTrials.gov, number NCT01336634. FINDINGS Between Dec 20, 2013, and Jan 14, 2015, 59 patients from 30 centres in nine countries across North America, Europe, and Asia met eligibility criteria. Two patients who had previously been untreated due to protocol deviation were excluded; thus, 57 eligible patients were enrolled. 36 patients (63·2% [95% CI 49·3-75·6]) achieved an investigator-assessed overall response. Serious adverse events were reported in 32 (56%) of 57 patients and included pyrexia in nine (16%), anaemia in three (5%), confusional state in two (4%), decreased appetite in two (4%), haemoptysis in two (4%), hypercalcaemia in two (4%), nausea in two (4%), and cutaneous squamous cell carcinoma in two (4%). The most common grade 3-4 adverse events were neutropenia in five patients (9%), hyponatraemia in four (7%), and anaemia in three (5%). Four patients died during the study from fatal adverse events judged to be unrelated to treatment (one retroperitoneal haemorrhage, one subarachnoid haemorrhage, one respiratory distress, and one from disease progression that was more severe than typical progression, as assessed by the investigator). INTERPRETATION Dabrafenib plus trametinib could represent a new targeted therapy with robust antitumour activity and a manageable safety profile in patients with BRAF(V600E)-mutant NSCLC. FUNDING GlaxoSmithKline.
Collapse
MESH Headings
- Adenocarcinoma/drug therapy
- Adenocarcinoma/genetics
- Adenocarcinoma/secondary
- Adult
- Aged
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Biomarkers, Tumor/genetics
- Carcinoma, Large Cell/drug therapy
- Carcinoma, Large Cell/genetics
- Carcinoma, Large Cell/secondary
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/secondary
- Female
- Follow-Up Studies
- Humans
- Imidazoles/administration & dosage
- Lung Neoplasms/drug therapy
- Lung Neoplasms/genetics
- Lung Neoplasms/pathology
- Lymphatic Metastasis
- Male
- Middle Aged
- Mutation/genetics
- Neoplasm Recurrence, Local/drug therapy
- Neoplasm Recurrence, Local/genetics
- Neoplasm Recurrence, Local/pathology
- Neoplasm Staging
- Oximes/administration & dosage
- Prognosis
- Proto-Oncogene Proteins B-raf/genetics
- Pyridones/administration & dosage
- Pyrimidinones/administration & dosage
- Survival Rate
Collapse
Affiliation(s)
| | - Benjamin Besse
- Gustave Roussy, Villejuif, France; Paris-Sud University, Orsay, France
| | - Harry J M Groen
- University of Groningen, Groningen, Netherlands; University Medical Center Groningen, Groningen, the Netherlands
| | | | - Elisabeth Quoix
- University Hospital, Nouvel Hôpital Civil, Strasbourg, France
| | | | - Fabrice Barlesi
- Aix-Marseille University, Assistance Publique Hôpitaux de Marseille, Multidisciplinary Oncology & Therapeutic Innovations Department, Hôpital Nord, Marseille, France
| | - Tae Min Kim
- Seoul National University Hospital, Seoul, South Korea
| | - Julien Mazieres
- Rangueil-Larrey Hospital, Toulouse, France; Paul Sabatier University, Toulouse, France
| | - Silvia Novello
- Department of Oncology, University of Turin, Orbassano, Italy
| | - James R Rigas
- The Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | | | | | - Pingkuan Zhang
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA
| | | | | |
Collapse
|
2139
|
Wang W, Wu W, Zhang Y. Response to crizotinib in a lung adenocarcinoma patient harboring EML4-ALK translocation with adnexal metastasis: A Case Report. Medicine (Baltimore) 2016; 95:e4221. [PMID: 27472693 PMCID: PMC5265830 DOI: 10.1097/md.0000000000004221] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Lung cancer with ovarian metastasis or adnexal metastasis harboring anaplastic lymphoma kinase (ALK) gene translocation is rare. Crizotinib, a novel ALK tyrosine kinase inhibitor, has already shown an impressive single-agent activity in ALK positive lung cancer. METHODS To summarize the case of clinical data and treatment of a 33-year-old woman with pelvic adnexal metastasis NSCLC. RESULTS Histological examination of the tumors showed lung adenocarcinoma. The right lung biopsy tissue and left adnexal mass biopsy tissue both revealed the presence of an ALK rearrangement by Ventana (D5F3) ALK immunohistochemistry assay (Ventana Medical Systems, Roche, Inc., Tuscon, AZ). The patient experienced a remarkable tumor response to crizotinib treatment. CONCLUSION Although the adnexal location is an uncommon metastasis site from lung cancer, oncologists should be aware of the possibility of such metastasis for female patients with ALK rearrangement NSCLC. Considering this remarkable response, we conclude that the presence of adnexal metastasis in NSCLC patients with ALK rearrangement should be attentive.
Collapse
Affiliation(s)
| | - Wei Wu
- Department of Pathology, Zhejiang Cancer Hospital
| | - Yiping Zhang
- Department of Chemotherapy
- Key Laboratory Diagnosis and Treatment Technology on Thoracic Oncology, Hangzhou, Zhejiang, China
- Correspondence: Yiping Zhang, Department of Chemotherapy, Zhejiang Cancer Hospital, 38 guangji road, Gongshu District, Hangzhou 310022, China (e-mail: )
| |
Collapse
|
2140
|
Chen YL, Lee CT, Lu CC, Yang SC, Chen WL, Lee YC, Yang CH, Peng SL, Su WC, Chow NH, Ho CL. Epidermal Growth Factor Receptor Mutation and Anaplastic Lymphoma Kinase Gene Fusion: Detection in Malignant Pleural Effusion by RNA or PNA Analysis. PLoS One 2016; 11:e0158125. [PMID: 27352172 PMCID: PMC4924845 DOI: 10.1371/journal.pone.0158125] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 06/11/2016] [Indexed: 11/29/2022] Open
Abstract
Analyzing EGFR mutations and detecting ALK gene fusion are indispensable when planning to treat pulmonary adenocarcinoma. Malignant pleural effusion (MPE) is a devastating complication of lung cancer and sometimes the only source for mutation analysis. The percentage of tumor cells in the pleural effusion may be low; therefore, mutant enrichment is required for a successful analysis. The EGFR mutation status in MPE was determined using three methods: (1) PCR sequencing of genomic DNA (direct sequencing), (2) mutant-enriched PCR sequencing of genomic DNA using peptide nucleic acid (PNA-sequencing), and (3) PCR sequencing of cDNA after reverse transcription for cellular RNA (RNA-sequencing). RT-PCR was also used to test cases for ALK gene fusion. PNA-sequencing and RNA-sequencing had similar analytical sensitivities (< 1%), which indicates similar enrichment capabilities. The clinical sensitivity in 133 cases when detecting the common EGFR exon 19 and exon 21 mutations was 56.4% (75/133) for direct sequencing, 63.2% (84/133) for PNA-sequencing, and 65.4% (87/133) for RNA-sequencing. RT-PCR and sequencing showed 5 cases (3.8%) with ALK gene fusion. All had wild-type EGFR. For EGFR analysis of MPE, RNA-sequencing is at least as sensitive as PNA-sequencing but not limited to specific mutations. Detecting ALK fusion can be incorporated in the same RNA workflow. Therefore, RNA is a better source for comprehensive molecular diagnoses in MPE.
Collapse
Affiliation(s)
- Yi-Lin Chen
- Molecular Diagnostics Laboratory, Department of Pathology, National Cheng Kung University Hospital, Tainan, Taiwan
- Department of Medical Laboratory Science and Biotechnology, National Cheng Kung University Hospital, Tainan, Taiwan
- College of Medicine, Molecular Medicine Core Laboratory, Research Center of Clinical Medicine, National Cheng Kung University Hospital, Tainan, Taiwan
- The Association of Medical Technologists, Tainan, Taiwan
| | - Chung-Ta Lee
- Molecular Diagnostics Laboratory, Department of Pathology, National Cheng Kung University Hospital, Tainan, Taiwan
- College of Medicine, Molecular Medicine Core Laboratory, Research Center of Clinical Medicine, National Cheng Kung University Hospital, Tainan, Taiwan
- The Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Cheng-Chan Lu
- Molecular Diagnostics Laboratory, Department of Pathology, National Cheng Kung University Hospital, Tainan, Taiwan
- College of Medicine, Molecular Medicine Core Laboratory, Research Center of Clinical Medicine, National Cheng Kung University Hospital, Tainan, Taiwan
- The Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University Hospital, Tainan, Taiwan
- The Institute of Molecular Medical, College of Medicine, National Cheng Kung University Hospital, Tainan, Taiwan
- The Association of Medical Technologists, Tainan, Taiwan
| | - Shu-Ching Yang
- Molecular Diagnostics Laboratory, Department of Pathology, National Cheng Kung University Hospital, Tainan, Taiwan
- College of Medicine, Molecular Medicine Core Laboratory, Research Center of Clinical Medicine, National Cheng Kung University Hospital, Tainan, Taiwan
- The Association of Medical Technologists, Tainan, Taiwan
| | - Wan-Li Chen
- Molecular Diagnostics Laboratory, Department of Pathology, National Cheng Kung University Hospital, Tainan, Taiwan
- College of Medicine, Molecular Medicine Core Laboratory, Research Center of Clinical Medicine, National Cheng Kung University Hospital, Tainan, Taiwan
- The Association of Medical Technologists, Tainan, Taiwan
| | - Yang-Cheng Lee
- Division of Hematology/Oncology, Department of Internal Medicine, Tainan Municipal Hospital, Tainan, Taiwan
| | | | - Shu-Ling Peng
- Molecular Diagnostics Laboratory, Department of Pathology, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Wu-Chou Su
- Molecular Diagnostics Laboratory, Department of Pathology, National Cheng Kung University Hospital, Tainan, Taiwan
- College of Medicine, Molecular Medicine Core Laboratory, Research Center of Clinical Medicine, National Cheng Kung University Hospital, Tainan, Taiwan
- The Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University Hospital, Tainan, Taiwan
- The Institute of Molecular Medical, College of Medicine, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Nan-Haw Chow
- Molecular Diagnostics Laboratory, Department of Pathology, National Cheng Kung University Hospital, Tainan, Taiwan
- College of Medicine, Molecular Medicine Core Laboratory, Research Center of Clinical Medicine, National Cheng Kung University Hospital, Tainan, Taiwan
- The Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University Hospital, Tainan, Taiwan
- The Institute of Molecular Medical, College of Medicine, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Chung-Liang Ho
- Molecular Diagnostics Laboratory, Department of Pathology, National Cheng Kung University Hospital, Tainan, Taiwan
- Department of Medical Laboratory Science and Biotechnology, National Cheng Kung University Hospital, Tainan, Taiwan
- College of Medicine, Molecular Medicine Core Laboratory, Research Center of Clinical Medicine, National Cheng Kung University Hospital, Tainan, Taiwan
- The Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University Hospital, Tainan, Taiwan
- The Institute of Molecular Medical, College of Medicine, National Cheng Kung University Hospital, Tainan, Taiwan
- * E-mail:
| |
Collapse
|
2141
|
Gettinger S, Rizvi NA, Chow LQ, Borghaei H, Brahmer J, Ready N, Gerber DE, Shepherd FA, Antonia S, Goldman JW, Juergens RA, Laurie SA, Nathan FE, Shen Y, Harbison CT, Hellmann MD. Nivolumab Monotherapy for First-Line Treatment of Advanced Non-Small-Cell Lung Cancer. J Clin Oncol 2016; 34:2980-7. [PMID: 27354485 DOI: 10.1200/jco.2016.66.9929] [Citation(s) in RCA: 408] [Impact Index Per Article: 45.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
PURPOSE Nivolumab, a programmed death-1 (PD-1) immune checkpoint inhibitor antibody, has demonstrated improved survival over docetaxel in previously treated advanced non-small-cell lung cancer (NSCLC). First-line monotherapy with nivolumab for advanced NSCLC was evaluated in the phase I, multicohort, Checkmate 012 trial. METHODS Fifty-two patients received nivolumab 3 mg/kg intravenously every 2 weeks until progression or unacceptable toxicity; postprogression treatment was permitted per protocol. The primary objective was to assess safety; secondary objectives included objective response rate (ORR) and 24-week progression-free survival (PFS) rate; overall survival (OS) was an exploratory end point. RESULTS Any-grade treatment-related adverse events (AEs) occurred in 71% of patients, most commonly: fatigue (29%), rash (19%), nausea (14%), diarrhea (12%), pruritus (12%), and arthralgia (10%). Ten patients (19%) reported grade 3 to 4 treatment-related AEs; grade 3 rash was the only grade 3 to 4 event occurring in more than one patient (n = 2; 4%). Six patients (12%) discontinued because of a treatment-related AE. The confirmed ORR was 23% (12 of 52), including four ongoing complete responses. Nine of 12 responses (75%) occurred by first tumor assessment (week 11); eight (67%) were ongoing (range, 5.3+ to 25.8+ months) at the time of data lock. ORR was 28% (nine of 32) in patients with any degree of tumor PD-ligand 1 expression and 14% (two of 14) in patients with no PD-ligand 1 expression. Median PFS was 3.6 months, and the 24-week PFS rate was 41% (95% CI, 27 to 54). Median OS was 19.4 months, and the 1-year and 18-month OS rates were 73% (95% CI, 59 to 83) and 57% (95% CI, 42 to 70), respectively. CONCLUSION First-line nivolumab monotherapy demonstrated a tolerable safety profile and durable responses in first-line advanced NSCLC.
Collapse
Affiliation(s)
- Scott Gettinger
- Scott Gettinger, Yale Cancer Center, New Haven, CT; Naiyer A. Rizvi and Matthew D. Hellmann, Memorial Sloan Kettering Cancer Center, New York, NY; Laura Q. Chow, University of Washington, Seattle, WA; Hossein Borghaei, Fox Chase Cancer Center, Philadelphia, PA; Julie Brahmer, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD; Neal Ready, Duke University Medical Center, Durham, NC; David E. Gerber, University of Texas Southwestern Medical Center, Dallas, TX; Frances A. Shepherd, Princess Margaret Cancer Centre, Toronto; Rosalyn A. Juergens, Juravinski Cancer Centre at McMaster University, Hamilton; Scott A. Laurie, Ottawa Hospital Cancer Centre, Ottawa, Ontario, Canada; Scott Antonia, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Jonathan W. Goldman, University of California, Los Angeles, Los Angeles, CA; and Faith E. Nathan, Yun Shen, and Christopher T. Harbison, Bristol-Myers Squibb, Princeton, NJ.
| | - Naiyer A Rizvi
- Scott Gettinger, Yale Cancer Center, New Haven, CT; Naiyer A. Rizvi and Matthew D. Hellmann, Memorial Sloan Kettering Cancer Center, New York, NY; Laura Q. Chow, University of Washington, Seattle, WA; Hossein Borghaei, Fox Chase Cancer Center, Philadelphia, PA; Julie Brahmer, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD; Neal Ready, Duke University Medical Center, Durham, NC; David E. Gerber, University of Texas Southwestern Medical Center, Dallas, TX; Frances A. Shepherd, Princess Margaret Cancer Centre, Toronto; Rosalyn A. Juergens, Juravinski Cancer Centre at McMaster University, Hamilton; Scott A. Laurie, Ottawa Hospital Cancer Centre, Ottawa, Ontario, Canada; Scott Antonia, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Jonathan W. Goldman, University of California, Los Angeles, Los Angeles, CA; and Faith E. Nathan, Yun Shen, and Christopher T. Harbison, Bristol-Myers Squibb, Princeton, NJ
| | - Laura Q Chow
- Scott Gettinger, Yale Cancer Center, New Haven, CT; Naiyer A. Rizvi and Matthew D. Hellmann, Memorial Sloan Kettering Cancer Center, New York, NY; Laura Q. Chow, University of Washington, Seattle, WA; Hossein Borghaei, Fox Chase Cancer Center, Philadelphia, PA; Julie Brahmer, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD; Neal Ready, Duke University Medical Center, Durham, NC; David E. Gerber, University of Texas Southwestern Medical Center, Dallas, TX; Frances A. Shepherd, Princess Margaret Cancer Centre, Toronto; Rosalyn A. Juergens, Juravinski Cancer Centre at McMaster University, Hamilton; Scott A. Laurie, Ottawa Hospital Cancer Centre, Ottawa, Ontario, Canada; Scott Antonia, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Jonathan W. Goldman, University of California, Los Angeles, Los Angeles, CA; and Faith E. Nathan, Yun Shen, and Christopher T. Harbison, Bristol-Myers Squibb, Princeton, NJ
| | - Hossein Borghaei
- Scott Gettinger, Yale Cancer Center, New Haven, CT; Naiyer A. Rizvi and Matthew D. Hellmann, Memorial Sloan Kettering Cancer Center, New York, NY; Laura Q. Chow, University of Washington, Seattle, WA; Hossein Borghaei, Fox Chase Cancer Center, Philadelphia, PA; Julie Brahmer, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD; Neal Ready, Duke University Medical Center, Durham, NC; David E. Gerber, University of Texas Southwestern Medical Center, Dallas, TX; Frances A. Shepherd, Princess Margaret Cancer Centre, Toronto; Rosalyn A. Juergens, Juravinski Cancer Centre at McMaster University, Hamilton; Scott A. Laurie, Ottawa Hospital Cancer Centre, Ottawa, Ontario, Canada; Scott Antonia, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Jonathan W. Goldman, University of California, Los Angeles, Los Angeles, CA; and Faith E. Nathan, Yun Shen, and Christopher T. Harbison, Bristol-Myers Squibb, Princeton, NJ
| | - Julie Brahmer
- Scott Gettinger, Yale Cancer Center, New Haven, CT; Naiyer A. Rizvi and Matthew D. Hellmann, Memorial Sloan Kettering Cancer Center, New York, NY; Laura Q. Chow, University of Washington, Seattle, WA; Hossein Borghaei, Fox Chase Cancer Center, Philadelphia, PA; Julie Brahmer, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD; Neal Ready, Duke University Medical Center, Durham, NC; David E. Gerber, University of Texas Southwestern Medical Center, Dallas, TX; Frances A. Shepherd, Princess Margaret Cancer Centre, Toronto; Rosalyn A. Juergens, Juravinski Cancer Centre at McMaster University, Hamilton; Scott A. Laurie, Ottawa Hospital Cancer Centre, Ottawa, Ontario, Canada; Scott Antonia, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Jonathan W. Goldman, University of California, Los Angeles, Los Angeles, CA; and Faith E. Nathan, Yun Shen, and Christopher T. Harbison, Bristol-Myers Squibb, Princeton, NJ
| | - Neal Ready
- Scott Gettinger, Yale Cancer Center, New Haven, CT; Naiyer A. Rizvi and Matthew D. Hellmann, Memorial Sloan Kettering Cancer Center, New York, NY; Laura Q. Chow, University of Washington, Seattle, WA; Hossein Borghaei, Fox Chase Cancer Center, Philadelphia, PA; Julie Brahmer, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD; Neal Ready, Duke University Medical Center, Durham, NC; David E. Gerber, University of Texas Southwestern Medical Center, Dallas, TX; Frances A. Shepherd, Princess Margaret Cancer Centre, Toronto; Rosalyn A. Juergens, Juravinski Cancer Centre at McMaster University, Hamilton; Scott A. Laurie, Ottawa Hospital Cancer Centre, Ottawa, Ontario, Canada; Scott Antonia, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Jonathan W. Goldman, University of California, Los Angeles, Los Angeles, CA; and Faith E. Nathan, Yun Shen, and Christopher T. Harbison, Bristol-Myers Squibb, Princeton, NJ
| | - David E Gerber
- Scott Gettinger, Yale Cancer Center, New Haven, CT; Naiyer A. Rizvi and Matthew D. Hellmann, Memorial Sloan Kettering Cancer Center, New York, NY; Laura Q. Chow, University of Washington, Seattle, WA; Hossein Borghaei, Fox Chase Cancer Center, Philadelphia, PA; Julie Brahmer, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD; Neal Ready, Duke University Medical Center, Durham, NC; David E. Gerber, University of Texas Southwestern Medical Center, Dallas, TX; Frances A. Shepherd, Princess Margaret Cancer Centre, Toronto; Rosalyn A. Juergens, Juravinski Cancer Centre at McMaster University, Hamilton; Scott A. Laurie, Ottawa Hospital Cancer Centre, Ottawa, Ontario, Canada; Scott Antonia, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Jonathan W. Goldman, University of California, Los Angeles, Los Angeles, CA; and Faith E. Nathan, Yun Shen, and Christopher T. Harbison, Bristol-Myers Squibb, Princeton, NJ
| | - Frances A Shepherd
- Scott Gettinger, Yale Cancer Center, New Haven, CT; Naiyer A. Rizvi and Matthew D. Hellmann, Memorial Sloan Kettering Cancer Center, New York, NY; Laura Q. Chow, University of Washington, Seattle, WA; Hossein Borghaei, Fox Chase Cancer Center, Philadelphia, PA; Julie Brahmer, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD; Neal Ready, Duke University Medical Center, Durham, NC; David E. Gerber, University of Texas Southwestern Medical Center, Dallas, TX; Frances A. Shepherd, Princess Margaret Cancer Centre, Toronto; Rosalyn A. Juergens, Juravinski Cancer Centre at McMaster University, Hamilton; Scott A. Laurie, Ottawa Hospital Cancer Centre, Ottawa, Ontario, Canada; Scott Antonia, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Jonathan W. Goldman, University of California, Los Angeles, Los Angeles, CA; and Faith E. Nathan, Yun Shen, and Christopher T. Harbison, Bristol-Myers Squibb, Princeton, NJ
| | - Scott Antonia
- Scott Gettinger, Yale Cancer Center, New Haven, CT; Naiyer A. Rizvi and Matthew D. Hellmann, Memorial Sloan Kettering Cancer Center, New York, NY; Laura Q. Chow, University of Washington, Seattle, WA; Hossein Borghaei, Fox Chase Cancer Center, Philadelphia, PA; Julie Brahmer, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD; Neal Ready, Duke University Medical Center, Durham, NC; David E. Gerber, University of Texas Southwestern Medical Center, Dallas, TX; Frances A. Shepherd, Princess Margaret Cancer Centre, Toronto; Rosalyn A. Juergens, Juravinski Cancer Centre at McMaster University, Hamilton; Scott A. Laurie, Ottawa Hospital Cancer Centre, Ottawa, Ontario, Canada; Scott Antonia, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Jonathan W. Goldman, University of California, Los Angeles, Los Angeles, CA; and Faith E. Nathan, Yun Shen, and Christopher T. Harbison, Bristol-Myers Squibb, Princeton, NJ
| | - Jonathan W Goldman
- Scott Gettinger, Yale Cancer Center, New Haven, CT; Naiyer A. Rizvi and Matthew D. Hellmann, Memorial Sloan Kettering Cancer Center, New York, NY; Laura Q. Chow, University of Washington, Seattle, WA; Hossein Borghaei, Fox Chase Cancer Center, Philadelphia, PA; Julie Brahmer, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD; Neal Ready, Duke University Medical Center, Durham, NC; David E. Gerber, University of Texas Southwestern Medical Center, Dallas, TX; Frances A. Shepherd, Princess Margaret Cancer Centre, Toronto; Rosalyn A. Juergens, Juravinski Cancer Centre at McMaster University, Hamilton; Scott A. Laurie, Ottawa Hospital Cancer Centre, Ottawa, Ontario, Canada; Scott Antonia, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Jonathan W. Goldman, University of California, Los Angeles, Los Angeles, CA; and Faith E. Nathan, Yun Shen, and Christopher T. Harbison, Bristol-Myers Squibb, Princeton, NJ
| | - Rosalyn A Juergens
- Scott Gettinger, Yale Cancer Center, New Haven, CT; Naiyer A. Rizvi and Matthew D. Hellmann, Memorial Sloan Kettering Cancer Center, New York, NY; Laura Q. Chow, University of Washington, Seattle, WA; Hossein Borghaei, Fox Chase Cancer Center, Philadelphia, PA; Julie Brahmer, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD; Neal Ready, Duke University Medical Center, Durham, NC; David E. Gerber, University of Texas Southwestern Medical Center, Dallas, TX; Frances A. Shepherd, Princess Margaret Cancer Centre, Toronto; Rosalyn A. Juergens, Juravinski Cancer Centre at McMaster University, Hamilton; Scott A. Laurie, Ottawa Hospital Cancer Centre, Ottawa, Ontario, Canada; Scott Antonia, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Jonathan W. Goldman, University of California, Los Angeles, Los Angeles, CA; and Faith E. Nathan, Yun Shen, and Christopher T. Harbison, Bristol-Myers Squibb, Princeton, NJ
| | - Scott A Laurie
- Scott Gettinger, Yale Cancer Center, New Haven, CT; Naiyer A. Rizvi and Matthew D. Hellmann, Memorial Sloan Kettering Cancer Center, New York, NY; Laura Q. Chow, University of Washington, Seattle, WA; Hossein Borghaei, Fox Chase Cancer Center, Philadelphia, PA; Julie Brahmer, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD; Neal Ready, Duke University Medical Center, Durham, NC; David E. Gerber, University of Texas Southwestern Medical Center, Dallas, TX; Frances A. Shepherd, Princess Margaret Cancer Centre, Toronto; Rosalyn A. Juergens, Juravinski Cancer Centre at McMaster University, Hamilton; Scott A. Laurie, Ottawa Hospital Cancer Centre, Ottawa, Ontario, Canada; Scott Antonia, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Jonathan W. Goldman, University of California, Los Angeles, Los Angeles, CA; and Faith E. Nathan, Yun Shen, and Christopher T. Harbison, Bristol-Myers Squibb, Princeton, NJ
| | - Faith E Nathan
- Scott Gettinger, Yale Cancer Center, New Haven, CT; Naiyer A. Rizvi and Matthew D. Hellmann, Memorial Sloan Kettering Cancer Center, New York, NY; Laura Q. Chow, University of Washington, Seattle, WA; Hossein Borghaei, Fox Chase Cancer Center, Philadelphia, PA; Julie Brahmer, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD; Neal Ready, Duke University Medical Center, Durham, NC; David E. Gerber, University of Texas Southwestern Medical Center, Dallas, TX; Frances A. Shepherd, Princess Margaret Cancer Centre, Toronto; Rosalyn A. Juergens, Juravinski Cancer Centre at McMaster University, Hamilton; Scott A. Laurie, Ottawa Hospital Cancer Centre, Ottawa, Ontario, Canada; Scott Antonia, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Jonathan W. Goldman, University of California, Los Angeles, Los Angeles, CA; and Faith E. Nathan, Yun Shen, and Christopher T. Harbison, Bristol-Myers Squibb, Princeton, NJ
| | - Yun Shen
- Scott Gettinger, Yale Cancer Center, New Haven, CT; Naiyer A. Rizvi and Matthew D. Hellmann, Memorial Sloan Kettering Cancer Center, New York, NY; Laura Q. Chow, University of Washington, Seattle, WA; Hossein Borghaei, Fox Chase Cancer Center, Philadelphia, PA; Julie Brahmer, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD; Neal Ready, Duke University Medical Center, Durham, NC; David E. Gerber, University of Texas Southwestern Medical Center, Dallas, TX; Frances A. Shepherd, Princess Margaret Cancer Centre, Toronto; Rosalyn A. Juergens, Juravinski Cancer Centre at McMaster University, Hamilton; Scott A. Laurie, Ottawa Hospital Cancer Centre, Ottawa, Ontario, Canada; Scott Antonia, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Jonathan W. Goldman, University of California, Los Angeles, Los Angeles, CA; and Faith E. Nathan, Yun Shen, and Christopher T. Harbison, Bristol-Myers Squibb, Princeton, NJ
| | - Christopher T Harbison
- Scott Gettinger, Yale Cancer Center, New Haven, CT; Naiyer A. Rizvi and Matthew D. Hellmann, Memorial Sloan Kettering Cancer Center, New York, NY; Laura Q. Chow, University of Washington, Seattle, WA; Hossein Borghaei, Fox Chase Cancer Center, Philadelphia, PA; Julie Brahmer, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD; Neal Ready, Duke University Medical Center, Durham, NC; David E. Gerber, University of Texas Southwestern Medical Center, Dallas, TX; Frances A. Shepherd, Princess Margaret Cancer Centre, Toronto; Rosalyn A. Juergens, Juravinski Cancer Centre at McMaster University, Hamilton; Scott A. Laurie, Ottawa Hospital Cancer Centre, Ottawa, Ontario, Canada; Scott Antonia, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Jonathan W. Goldman, University of California, Los Angeles, Los Angeles, CA; and Faith E. Nathan, Yun Shen, and Christopher T. Harbison, Bristol-Myers Squibb, Princeton, NJ
| | - Matthew D Hellmann
- Scott Gettinger, Yale Cancer Center, New Haven, CT; Naiyer A. Rizvi and Matthew D. Hellmann, Memorial Sloan Kettering Cancer Center, New York, NY; Laura Q. Chow, University of Washington, Seattle, WA; Hossein Borghaei, Fox Chase Cancer Center, Philadelphia, PA; Julie Brahmer, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD; Neal Ready, Duke University Medical Center, Durham, NC; David E. Gerber, University of Texas Southwestern Medical Center, Dallas, TX; Frances A. Shepherd, Princess Margaret Cancer Centre, Toronto; Rosalyn A. Juergens, Juravinski Cancer Centre at McMaster University, Hamilton; Scott A. Laurie, Ottawa Hospital Cancer Centre, Ottawa, Ontario, Canada; Scott Antonia, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Jonathan W. Goldman, University of California, Los Angeles, Los Angeles, CA; and Faith E. Nathan, Yun Shen, and Christopher T. Harbison, Bristol-Myers Squibb, Princeton, NJ
| |
Collapse
|
2142
|
Campo M, Al-Halabi H, Khandekar M, Shaw AT, Sequist LV, Willers H. Integration of Stereotactic Body Radiation Therapy With Tyrosine Kinase Inhibitors in Stage IV Oncogene-Driven Lung Cancer. Oncologist 2016; 21:964-73. [PMID: 27354669 DOI: 10.1634/theoncologist.2015-0508] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 03/18/2016] [Indexed: 02/07/2023] Open
Abstract
UNLABELLED : Genotype-based selection of patients for targeted therapies has had a substantial impact on the treatment of non-small cell lung cancers (NSCLCs). Tyrosine kinase inhibitors (TKIs) directed at cancers driven by oncogenes, such as epidermal growth factor receptor mutations or anaplastic lymphoma kinase rearrangements, often achieve dramatic responses and result in prolonged survival compared with chemotherapy. However, TKI resistance invariably develops. Disease progression can be limited to only one or a few sites and might not be symptomatic, raising the important question of whether this type of oligoprogression warrants a change in systemic therapy or consideration of local treatment. Recent clinical observations suggest a growing role for stereotactic body radiation therapy (SBRT) in the treatment of oligoprogressive and perhaps even oligopersistent disease (primary and/or metastases) in oncogene-driven NSCLC. SBRT might allow patients to continue with existing TKI treatments longer and delay the need to switch to other systemic options. We review the current data with regard to the use of SBRT for metastatic NSCLC and particularly oncogene-driven disease. Although there is great promise in the marriage of targeted therapies with SBRT, prospective data are urgently needed. In the meantime, such strategies are being used in carefully selected patients, with risk-adapted SBRT dose-fractionation regimens used to optimize the therapeutic index. IMPLICATIONS FOR PRACTICE Stereotactic body radiation therapy (SBRT) or SBRT-like treatments are increasingly being used for oligoprogression in patients with oncogene-driven non-small cell lung cancer. This approach allows patients to extend the duration of tyrosine kinase inhibitor therapy and has the potential to prolong survival times. Careful patient selection and risk-adapted radiation dosing is of critical importance to minimize toxicity and preserve patient quality of life.
Collapse
Affiliation(s)
- Meghan Campo
- Hematology/Oncology Fellowship Program, Dana-Farber/Partners CancerCare, Boston, Massachusetts, USA
| | - Hani Al-Halabi
- Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Melin Khandekar
- Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Alice T Shaw
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Lecia V Sequist
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Henning Willers
- Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
2143
|
Cortinovis D, Abbate M, Bidoli P, Capici S, Canova S. Targeted therapies and immunotherapy in non-small-cell lung cancer. Ecancermedicalscience 2016; 10:648. [PMID: 27433281 PMCID: PMC4929979 DOI: 10.3332/ecancer.2016.648] [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: 04/10/2016] [Indexed: 01/23/2023] Open
Abstract
Non-small-cell lung cancer is still considered a difficult disease to manage because of its aggressiveness and resistance to common therapies. Chemotherapy remains the gold standard in nearly 80% of lung cancers, but clinical outcomes are discouraging, and the impact on median overall survival (OS) barely reaches 12 months. At the end of the last century, the discovery of oncogene-driven tumours completely changed the therapeutic landscape in lung cancers, harbouring specific gene mutations/translocations. Epidermal growth factors receptor (EGFR) common mutations first and anaplastic lymphoma kinase (ALK) translocations later led new insights in lung cancer biology knowledge. The use of specific tyrosine kinases inhibitors overturned the biological behaviour of EGFR mutation positive tumours and became a preclinical model to understand the heterogeneity of lung cancers and the mechanisms of drug resistance. In this review, we summarise the employment of targeted agents against the most representative biomolecular alterations and provide some criticisms of the therapeutic strategies.
Collapse
Affiliation(s)
- D Cortinovis
- Medical Oncology Unit, AOU San Gerardo, via Giambattista Pergolesi 33, 20900 Monza, Italy
| | - M Abbate
- Medical Oncology Unit, AOU San Gerardo, via Giambattista Pergolesi 33, 20900 Monza, Italy
| | - P Bidoli
- Medical Oncology Unit, AOU San Gerardo, via Giambattista Pergolesi 33, 20900 Monza, Italy
| | - S Capici
- Medical Oncology Unit, AOU San Gerardo, via Giambattista Pergolesi 33, 20900 Monza, Italy
| | - S Canova
- Medical Oncology Unit, AOU San Gerardo, via Giambattista Pergolesi 33, 20900 Monza, Italy
| |
Collapse
|
2144
|
Zaarour M, Nazha B, Weerasinghe C, Moussaly E, Terjanian T. Anaplastic lymphoma kinase inhibitors in elderly patients with advanced non-small cell lung cancer. Expert Rev Anticancer Ther 2016; 16:877-83. [PMID: 27328177 DOI: 10.1080/14737140.2016.1204235] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
INTRODUCTION Non-small cell lung cancer (NSCLC) is the leading cause of cancer-related deaths in the USA and worldwide. At diagnosis, half of the patients are over 70 years of age, and most present with advanced disease for which chemotherapy provides modest benefit with significant toxicity. Older patients often have more comorbidities than their younger counterparts and tend to be excluded from clinical trials. AREAS COVERED A small percentage (less than 7%) of patients with NSCLC have the anaplastic lymphoma kinase (ALK) rearrangement. Compared to the general NSCLC population, this clinically distinct group has a relatively younger median age of 51 years at diagnosis. As such, elderly patients with ALK-positive disease are both a minority within this group and are expected to be underrepresented in clinical trials. Expert commentary: Despite promising results in the general population, data about the efficacy and safety of ALK inhibitors in the elderly population remains scarce. In our review, we briefly discuss the current evidence of ALK inhibitors in the general population and we shed light on this subgroup of elderly patients with advanced ALK-positive disease.
Collapse
Affiliation(s)
- Mazen Zaarour
- a Department of Medicine , Staten Island University Hospital Northwell Health , Staten Island , NY , USA
| | - Bassel Nazha
- a Department of Medicine , Staten Island University Hospital Northwell Health , Staten Island , NY , USA
| | - Chanudi Weerasinghe
- a Department of Medicine , Staten Island University Hospital Northwell Health , Staten Island , NY , USA
| | - Elias Moussaly
- a Department of Medicine , Staten Island University Hospital Northwell Health , Staten Island , NY , USA
| | - Terenig Terjanian
- b Department of Medicine, Division of Hematology/Oncology , Staten Island University Hospital Northwell Health , Staten Island , NY , USA
| |
Collapse
|
2145
|
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
|
2146
|
Abstract
INTRODUCTION Anaplastic lymphoma kinase (ALK) rearrangements are present in about 5% of advanced non-small-cell lung cancer (NSCLC) patients. Despite the initial response, after a median of 1-2 years, ALK-positive patients developed an acquired resistance to the ALK-inhibitor crizotinib. Among the most promising second-generation ALK-inhibitors, alectinib is being investigated in crizotinib-naïve and -resistant ALK-positive NSCLC patients. AREAS COVERED The current state-of-the-art of ALK-inhibitors treatment, and in particular the role of alectinib in this setting, is reviewed and discussed. A structured search of bibliographic databases for peer-reviewed research literature and of main meetings using a focused review question was undertaken. Expert commentary: Alectinib reports promising results with a good safety profile, becoming a potentially very important option for ALK-translocated NSCLC patients. The preliminary results from the J-ALEX phase III randomized trial performed in ALK-rearranged NSCLC Japanese patients showed a better activity and tolerability of alectinib versus crizotinib.
Collapse
Affiliation(s)
- Antonio Rossi
- a Division of Medical Oncology , "S.G. Moscati" Hospital , Avellino , Italy
| |
Collapse
|
2147
|
Alectinib's activity against CNS metastases from ALK-positive non-small cell lung cancer: a single institution case series. J Neurooncol 2016; 129:355-61. [PMID: 27324494 DOI: 10.1007/s11060-016-2184-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 06/09/2016] [Indexed: 10/21/2022]
Abstract
In the present study we assessed the activity of the next-generation anaplastic lymphoma kinase (ALK)-tyrosine kinase inhibitor (-TKI) alectinib, in patients with ALK-postive, advanced non-small cell lung cancer (NSCLC) and central nervous system (CNS) metastases. NSCLCs with ALK-positive disease, as assessed by fluorescence in situ hybridization, and CNS metastases were treated with alectinib 600 mg BID. Included patients were followed prospectively in order to evaluate the efficacy of the drug, with particular emphasis on activity in the CNS. Eleven consecutive patients were enrolled. The majority of them were pretreated with crizotinib (n = 10, 90.9 %), and cranial radiotherapy (n = 8, 72.7 %). Six of the seven patients with measurable CNS disease experienced a CNS response, including three patients who were naïve for cranial radiation. Median duration of response was 8 months. For the whole population, median CNS-progression-free survival (-PFS), systemic-PFS, overall-PFS, overall survival, and 1-year survival were 8, 11, 8, 13 months, and 31.1 %, respectively. Two patients experiencing a CNS response were assessed for alectinib's concentrations in serum and cerebro-spinal fluid (CSF), and showed a CSF-to-serum ratio ranging from 0.001 to 0.003 ng/mL. Alectinib is highly active against CNS metastases from ALK-positive NSCLCs, irrespective of prior treatment(s) with ALK-TKI(s) and/or cranial radiotherapy. The low CSF-to-serum ratio of alectinib suggests that measuring the concentrations of the drug in the CSF may not be a reliable surrogate of its distribution into the CNS.
Collapse
|
2148
|
Schmidt KT, Chau CH, Price DK, Figg WD. Precision Oncology Medicine: The Clinical Relevance of Patient-Specific Biomarkers Used to Optimize Cancer Treatment. J Clin Pharmacol 2016; 56:1484-1499. [PMID: 27197880 DOI: 10.1002/jcph.765] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 05/06/2016] [Accepted: 05/09/2016] [Indexed: 12/22/2022]
Abstract
Precision medicine in oncology is the result of an increasing awareness of patient-specific clinical features coupled with the development of genomic-based diagnostics and targeted therapeutics. Companion diagnostics designed for specific drug-target pairs were the first to widely utilize clinically applicable tumor biomarkers (eg, HER2, EGFR), directing treatment for patients whose tumors exhibit a mutation susceptible to an FDA-approved targeted therapy (eg, trastuzumab, erlotinib). Clinically relevant germline mutations in drug-metabolizing enzymes and transporters (eg, TPMT, DPYD) have been shown to impact drug response, providing a rationale for individualized dosing to optimize treatment. The use of multigene expression-based assays to analyze an array of prognostic biomarkers has been shown to help direct treatment decisions, especially in breast cancer (eg, Oncotype DX). More recently, the use of next-generation sequencing to detect many potential "actionable" cancer molecular alterations is further shifting the 1 gene-1 drug paradigm toward a more comprehensive, multigene approach. Currently, many clinical trials (eg, NCI-MATCH, NCI-MPACT) are assessing novel diagnostic tools with a combination of different targeted therapeutics while also examining tumor biomarkers that were previously unexplored in a variety of cancer histologies. Results from ongoing trials such as the NCI-MATCH will help determine the clinical utility and future development of the precision-medicine approach.
Collapse
Affiliation(s)
- Keith T Schmidt
- Clinical Pharmacology Program, Office of the Clinical Director, NIH, Bethesda, MD, USA
| | - Cindy H Chau
- Molecular Pharmacology Section, Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Douglas K Price
- Molecular Pharmacology Section, Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - William D Figg
- Clinical Pharmacology Program, Office of the Clinical Director, NIH, Bethesda, MD, USA
- Molecular Pharmacology Section, Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| |
Collapse
|
2149
|
Giroux-Leprieur E, Fallet V, Cadranel J, Wislez M. Spotlight on crizotinib in the first-line treatment of ALK-positive advanced non-small-cell lung cancer: patients selection and perspectives. LUNG CANCER-TARGETS AND THERAPY 2016; 7:83-90. [PMID: 28210164 PMCID: PMC5310701 DOI: 10.2147/lctt.s99303] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Around 4% of advanced non-small-cell lung cancers (NSCLCs) have an ALK rearrangement at the time of diagnosis. This molecular feature is more frequent in young patients, with no/light smoking habit and with adenocarcinoma pathological subtype. Crizotinib is a tyrosine kinase inhibitor, targeting ALK, ROS1, RON, and MET. The preclinical efficacy results led to a fast-track clinical development. The US Food and Drug Administration (FDA) approval was achieved after the Phase I clinical trial in 2011 in ALK-rearranged advanced NSCLC progressing after a first-line treatment. In 2013, the randomized Phase III trial PROFILE-1007 confirmed the efficacy of crizotinib in ALK-rearranged NSCLC, compared to cytotoxic chemotherapy, in second-line setting or more. In 2014, the PROFILE-1014 trial showed the superiority of crizotinib in the first-line setting compared to the pemetrexed platinum doublet chemotherapy. The response rate was 74%, and the progression-free survival was 10.9 months with crizotinib. Based on these results, crizotinib received approval from the FDA and European Medicines Agency for first-line treatment of ALK-rearranged NSCLC. The various molecular mechanisms at the time of the progression (ALK mutations or amplification, ALK-independent mechanisms) encourage performing re-biopsy at the time of progression under crizotinib. The best treatment strategy at the progression (crizotinib continuation beyond progression, switch to second-generation tyrosine kinase inhibitors, or cytotoxic chemotherapy) depends on the phenotype of the progression, the molecular status, and the physical condition of the patient.
Collapse
Affiliation(s)
- Etienne Giroux-Leprieur
- Respiratory Diseases and Thoracic Oncology Department, APHP-Ambroise Paré Hospital, Boulogne-Billancourt, France; EA4340 Laboratory, UVSQ, Paris-Saclay University, France
| | - Vincent Fallet
- Respiratory Diseases Department, APHP - Tenon Hospital, Paris, France; Sorbonne University, GRC 04, UPMC Univ Paris 06, France
| | - Jacques Cadranel
- Respiratory Diseases Department, APHP - Tenon Hospital, Paris, France; Sorbonne University, GRC 04, UPMC Univ Paris 06, France
| | - Marie Wislez
- Respiratory Diseases Department, APHP - Tenon Hospital, Paris, France; Sorbonne University, GRC 04, UPMC Univ Paris 06, France
| |
Collapse
|
2150
|
Abstract
Management paradigms for metastatic non-small cell lung cancer (mNSCLC) are evolving. Locally ablative therapies are now being increasingly integrated into combined-modality treatment strategies for mNSCLC patients with limited burdens of metastatic foci, termed oligometastases. Concurrently, techniques allowing for precise high-dose radiotherapy delivered over 1 to 5 total treatments, termed stereotactic body radiation therapy (SBRT) or stereotactic ablative radiation therapy (SABR), have emerged as a powerful means of noninvasive tumor ablation with broad patient candidacy. Strong rationale exists for ablative therapy in the setting of oligometastatic NSCLC, including patterns-of-failure analyses and data supporting local ablation of oligoprogressive disease for patients with oncogene-addicted mNSCLC treated with tyrosine kinase inhibitors. In this article, we examine the theoretical basis for ablation of oligometastatic NSCLC and review the growing clinical literature of mNSCLC patients treated with ablative radiation therapy.
Collapse
|