851
|
Shimazawa R, Ikeda M. Drug–diagnostic co-development: challenges and issues. Expert Rev Mol Diagn 2016; 16:187-204. [DOI: 10.1586/14737159.2016.1132163] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
852
|
Wang W, Jiang X, Song Z, Zhang Y. Patients harboring EGFR mutation after primary resistance to crizotinib and response to EGFR-tyrosine kinase inhibitor. Onco Targets Ther 2016; 9:211-5. [PMID: 26811689 PMCID: PMC4712969 DOI: 10.2147/ott.s97100] [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: 12/12/2022] Open
Abstract
Anaplastic lymphoma kinase (ALK) rearrangement lung cancer responds to ALK tyrosine kinase inhibitors. It is known that many cases ultimately acquired resistance to crizotinib. However, a case of primary resistance is rare. We present a case of harboring exon 19 deletion in epidermal growth factor receptor in ALK rearranged lung adenocarcinoma, who experienced a partial tumor response to icotinib after failure with crizotinib therapy and chemotherapy. Considering the partial response, we conclude that it is important to find the cause of resistance to crizotinib. We detected gene mutations with plasma by the next-generation sequencing; the next-generation sequencing demonstrates an attractive system to identify mutations improving the outcome of patients with a deadly disease.
Collapse
Affiliation(s)
- Wenxian Wang
- Department of Chemotherapy, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, People's Republic of China
| | - Xiaowen Jiang
- Department of Chemotherapy, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, People's Republic of China
| | - Zhengbo Song
- Department of Chemotherapy, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, People's Republic of China; Key Laboratory Diagnosis and Treatment Technology on Thoracic Oncology, Hangzhou, Zhejiang, People's Republic of China
| | - Yiping Zhang
- Department of Chemotherapy, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, People's Republic of China; Key Laboratory Diagnosis and Treatment Technology on Thoracic Oncology, Hangzhou, Zhejiang, People's Republic of China
| |
Collapse
|
853
|
Shaw AT, Friboulet L, Leshchiner I, Gainor JF, Bergqvist S, Brooun A, Burke BJ, Deng YL, Liu W, Dardaei L, Frias RL, Schultz KR, Logan J, James LP, Smeal T, Timofeevski S, Katayama R, Iafrate AJ, Le L, McTigue M, Getz G, Johnson TW, Engelman JA. Resensitization to Crizotinib by the Lorlatinib ALK Resistance Mutation L1198F. N Engl J Med 2016; 374:54-61. [PMID: 26698910 PMCID: PMC4773904 DOI: 10.1056/nejmoa1508887] [Citation(s) in RCA: 389] [Impact Index Per Article: 43.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
In a patient who had metastatic anaplastic lymphoma kinase (ALK)-rearranged lung cancer, resistance to crizotinib developed because of a mutation in the ALK kinase domain. This mutation is predicted to result in a substitution of cysteine by tyrosine at amino acid residue 1156 (C1156Y). Her tumor did not respond to a second-generation ALK inhibitor, but it did respond to lorlatinib (PF-06463922), a third-generation inhibitor. When her tumor relapsed, sequencing of the resistant tumor revealed an ALK L1198F mutation in addition to the C1156Y mutation. The L1198F substitution confers resistance to lorlatinib through steric interference with drug binding. However, L1198F paradoxically enhances binding to crizotinib, negating the effect of C1156Y and resensitizing resistant cancers to crizotinib. The patient received crizotinib again, and her cancer-related symptoms and liver failure resolved. (Funded by Pfizer and others; ClinicalTrials.gov number, NCT01970865.).
Collapse
Affiliation(s)
- Alice T Shaw
- Massachusetts General Hospital Cancer Center (A.T.S., L.F., J.F.G., L.D., R.L.F., K.R.S., J.L., G.G., J.A.E.) and the Department of Pathology (A.J.I., L.L., G.G.), Massachusetts General Hospital, Boston, and the Broad Institute of the Massachusetts Institute of Technology and Harvard, Cambridge (I.L., G.G.) - all in Massachusetts; Pfizer Worldwide Research and Development, La Jolla, CA (S.B., A.B., B.J.B., Y.-L.D., W.L., L.P.J., T.S., S.T., M.M., T.W.J.); and the Japanese Foundation for Cancer Research, Tokyo (R.K.)
| | - Luc Friboulet
- Massachusetts General Hospital Cancer Center (A.T.S., L.F., J.F.G., L.D., R.L.F., K.R.S., J.L., G.G., J.A.E.) and the Department of Pathology (A.J.I., L.L., G.G.), Massachusetts General Hospital, Boston, and the Broad Institute of the Massachusetts Institute of Technology and Harvard, Cambridge (I.L., G.G.) - all in Massachusetts; Pfizer Worldwide Research and Development, La Jolla, CA (S.B., A.B., B.J.B., Y.-L.D., W.L., L.P.J., T.S., S.T., M.M., T.W.J.); and the Japanese Foundation for Cancer Research, Tokyo (R.K.)
| | - Ignaty Leshchiner
- Massachusetts General Hospital Cancer Center (A.T.S., L.F., J.F.G., L.D., R.L.F., K.R.S., J.L., G.G., J.A.E.) and the Department of Pathology (A.J.I., L.L., G.G.), Massachusetts General Hospital, Boston, and the Broad Institute of the Massachusetts Institute of Technology and Harvard, Cambridge (I.L., G.G.) - all in Massachusetts; Pfizer Worldwide Research and Development, La Jolla, CA (S.B., A.B., B.J.B., Y.-L.D., W.L., L.P.J., T.S., S.T., M.M., T.W.J.); and the Japanese Foundation for Cancer Research, Tokyo (R.K.)
| | - Justin F Gainor
- Massachusetts General Hospital Cancer Center (A.T.S., L.F., J.F.G., L.D., R.L.F., K.R.S., J.L., G.G., J.A.E.) and the Department of Pathology (A.J.I., L.L., G.G.), Massachusetts General Hospital, Boston, and the Broad Institute of the Massachusetts Institute of Technology and Harvard, Cambridge (I.L., G.G.) - all in Massachusetts; Pfizer Worldwide Research and Development, La Jolla, CA (S.B., A.B., B.J.B., Y.-L.D., W.L., L.P.J., T.S., S.T., M.M., T.W.J.); and the Japanese Foundation for Cancer Research, Tokyo (R.K.)
| | - Simon Bergqvist
- Massachusetts General Hospital Cancer Center (A.T.S., L.F., J.F.G., L.D., R.L.F., K.R.S., J.L., G.G., J.A.E.) and the Department of Pathology (A.J.I., L.L., G.G.), Massachusetts General Hospital, Boston, and the Broad Institute of the Massachusetts Institute of Technology and Harvard, Cambridge (I.L., G.G.) - all in Massachusetts; Pfizer Worldwide Research and Development, La Jolla, CA (S.B., A.B., B.J.B., Y.-L.D., W.L., L.P.J., T.S., S.T., M.M., T.W.J.); and the Japanese Foundation for Cancer Research, Tokyo (R.K.)
| | - Alexei Brooun
- Massachusetts General Hospital Cancer Center (A.T.S., L.F., J.F.G., L.D., R.L.F., K.R.S., J.L., G.G., J.A.E.) and the Department of Pathology (A.J.I., L.L., G.G.), Massachusetts General Hospital, Boston, and the Broad Institute of the Massachusetts Institute of Technology and Harvard, Cambridge (I.L., G.G.) - all in Massachusetts; Pfizer Worldwide Research and Development, La Jolla, CA (S.B., A.B., B.J.B., Y.-L.D., W.L., L.P.J., T.S., S.T., M.M., T.W.J.); and the Japanese Foundation for Cancer Research, Tokyo (R.K.)
| | - Benjamin J Burke
- Massachusetts General Hospital Cancer Center (A.T.S., L.F., J.F.G., L.D., R.L.F., K.R.S., J.L., G.G., J.A.E.) and the Department of Pathology (A.J.I., L.L., G.G.), Massachusetts General Hospital, Boston, and the Broad Institute of the Massachusetts Institute of Technology and Harvard, Cambridge (I.L., G.G.) - all in Massachusetts; Pfizer Worldwide Research and Development, La Jolla, CA (S.B., A.B., B.J.B., Y.-L.D., W.L., L.P.J., T.S., S.T., M.M., T.W.J.); and the Japanese Foundation for Cancer Research, Tokyo (R.K.)
| | - Ya-Li Deng
- Massachusetts General Hospital Cancer Center (A.T.S., L.F., J.F.G., L.D., R.L.F., K.R.S., J.L., G.G., J.A.E.) and the Department of Pathology (A.J.I., L.L., G.G.), Massachusetts General Hospital, Boston, and the Broad Institute of the Massachusetts Institute of Technology and Harvard, Cambridge (I.L., G.G.) - all in Massachusetts; Pfizer Worldwide Research and Development, La Jolla, CA (S.B., A.B., B.J.B., Y.-L.D., W.L., L.P.J., T.S., S.T., M.M., T.W.J.); and the Japanese Foundation for Cancer Research, Tokyo (R.K.)
| | - Wei Liu
- Massachusetts General Hospital Cancer Center (A.T.S., L.F., J.F.G., L.D., R.L.F., K.R.S., J.L., G.G., J.A.E.) and the Department of Pathology (A.J.I., L.L., G.G.), Massachusetts General Hospital, Boston, and the Broad Institute of the Massachusetts Institute of Technology and Harvard, Cambridge (I.L., G.G.) - all in Massachusetts; Pfizer Worldwide Research and Development, La Jolla, CA (S.B., A.B., B.J.B., Y.-L.D., W.L., L.P.J., T.S., S.T., M.M., T.W.J.); and the Japanese Foundation for Cancer Research, Tokyo (R.K.)
| | - Leila Dardaei
- Massachusetts General Hospital Cancer Center (A.T.S., L.F., J.F.G., L.D., R.L.F., K.R.S., J.L., G.G., J.A.E.) and the Department of Pathology (A.J.I., L.L., G.G.), Massachusetts General Hospital, Boston, and the Broad Institute of the Massachusetts Institute of Technology and Harvard, Cambridge (I.L., G.G.) - all in Massachusetts; Pfizer Worldwide Research and Development, La Jolla, CA (S.B., A.B., B.J.B., Y.-L.D., W.L., L.P.J., T.S., S.T., M.M., T.W.J.); and the Japanese Foundation for Cancer Research, Tokyo (R.K.)
| | - Rosa L Frias
- Massachusetts General Hospital Cancer Center (A.T.S., L.F., J.F.G., L.D., R.L.F., K.R.S., J.L., G.G., J.A.E.) and the Department of Pathology (A.J.I., L.L., G.G.), Massachusetts General Hospital, Boston, and the Broad Institute of the Massachusetts Institute of Technology and Harvard, Cambridge (I.L., G.G.) - all in Massachusetts; Pfizer Worldwide Research and Development, La Jolla, CA (S.B., A.B., B.J.B., Y.-L.D., W.L., L.P.J., T.S., S.T., M.M., T.W.J.); and the Japanese Foundation for Cancer Research, Tokyo (R.K.)
| | - Kate R Schultz
- Massachusetts General Hospital Cancer Center (A.T.S., L.F., J.F.G., L.D., R.L.F., K.R.S., J.L., G.G., J.A.E.) and the Department of Pathology (A.J.I., L.L., G.G.), Massachusetts General Hospital, Boston, and the Broad Institute of the Massachusetts Institute of Technology and Harvard, Cambridge (I.L., G.G.) - all in Massachusetts; Pfizer Worldwide Research and Development, La Jolla, CA (S.B., A.B., B.J.B., Y.-L.D., W.L., L.P.J., T.S., S.T., M.M., T.W.J.); and the Japanese Foundation for Cancer Research, Tokyo (R.K.)
| | - Jennifer Logan
- Massachusetts General Hospital Cancer Center (A.T.S., L.F., J.F.G., L.D., R.L.F., K.R.S., J.L., G.G., J.A.E.) and the Department of Pathology (A.J.I., L.L., G.G.), Massachusetts General Hospital, Boston, and the Broad Institute of the Massachusetts Institute of Technology and Harvard, Cambridge (I.L., G.G.) - all in Massachusetts; Pfizer Worldwide Research and Development, La Jolla, CA (S.B., A.B., B.J.B., Y.-L.D., W.L., L.P.J., T.S., S.T., M.M., T.W.J.); and the Japanese Foundation for Cancer Research, Tokyo (R.K.)
| | - Leonard P James
- Massachusetts General Hospital Cancer Center (A.T.S., L.F., J.F.G., L.D., R.L.F., K.R.S., J.L., G.G., J.A.E.) and the Department of Pathology (A.J.I., L.L., G.G.), Massachusetts General Hospital, Boston, and the Broad Institute of the Massachusetts Institute of Technology and Harvard, Cambridge (I.L., G.G.) - all in Massachusetts; Pfizer Worldwide Research and Development, La Jolla, CA (S.B., A.B., B.J.B., Y.-L.D., W.L., L.P.J., T.S., S.T., M.M., T.W.J.); and the Japanese Foundation for Cancer Research, Tokyo (R.K.)
| | - Tod Smeal
- Massachusetts General Hospital Cancer Center (A.T.S., L.F., J.F.G., L.D., R.L.F., K.R.S., J.L., G.G., J.A.E.) and the Department of Pathology (A.J.I., L.L., G.G.), Massachusetts General Hospital, Boston, and the Broad Institute of the Massachusetts Institute of Technology and Harvard, Cambridge (I.L., G.G.) - all in Massachusetts; Pfizer Worldwide Research and Development, La Jolla, CA (S.B., A.B., B.J.B., Y.-L.D., W.L., L.P.J., T.S., S.T., M.M., T.W.J.); and the Japanese Foundation for Cancer Research, Tokyo (R.K.)
| | - Sergei Timofeevski
- Massachusetts General Hospital Cancer Center (A.T.S., L.F., J.F.G., L.D., R.L.F., K.R.S., J.L., G.G., J.A.E.) and the Department of Pathology (A.J.I., L.L., G.G.), Massachusetts General Hospital, Boston, and the Broad Institute of the Massachusetts Institute of Technology and Harvard, Cambridge (I.L., G.G.) - all in Massachusetts; Pfizer Worldwide Research and Development, La Jolla, CA (S.B., A.B., B.J.B., Y.-L.D., W.L., L.P.J., T.S., S.T., M.M., T.W.J.); and the Japanese Foundation for Cancer Research, Tokyo (R.K.)
| | - Ryohei Katayama
- Massachusetts General Hospital Cancer Center (A.T.S., L.F., J.F.G., L.D., R.L.F., K.R.S., J.L., G.G., J.A.E.) and the Department of Pathology (A.J.I., L.L., G.G.), Massachusetts General Hospital, Boston, and the Broad Institute of the Massachusetts Institute of Technology and Harvard, Cambridge (I.L., G.G.) - all in Massachusetts; Pfizer Worldwide Research and Development, La Jolla, CA (S.B., A.B., B.J.B., Y.-L.D., W.L., L.P.J., T.S., S.T., M.M., T.W.J.); and the Japanese Foundation for Cancer Research, Tokyo (R.K.)
| | - A John Iafrate
- Massachusetts General Hospital Cancer Center (A.T.S., L.F., J.F.G., L.D., R.L.F., K.R.S., J.L., G.G., J.A.E.) and the Department of Pathology (A.J.I., L.L., G.G.), Massachusetts General Hospital, Boston, and the Broad Institute of the Massachusetts Institute of Technology and Harvard, Cambridge (I.L., G.G.) - all in Massachusetts; Pfizer Worldwide Research and Development, La Jolla, CA (S.B., A.B., B.J.B., Y.-L.D., W.L., L.P.J., T.S., S.T., M.M., T.W.J.); and the Japanese Foundation for Cancer Research, Tokyo (R.K.)
| | - Long Le
- Massachusetts General Hospital Cancer Center (A.T.S., L.F., J.F.G., L.D., R.L.F., K.R.S., J.L., G.G., J.A.E.) and the Department of Pathology (A.J.I., L.L., G.G.), Massachusetts General Hospital, Boston, and the Broad Institute of the Massachusetts Institute of Technology and Harvard, Cambridge (I.L., G.G.) - all in Massachusetts; Pfizer Worldwide Research and Development, La Jolla, CA (S.B., A.B., B.J.B., Y.-L.D., W.L., L.P.J., T.S., S.T., M.M., T.W.J.); and the Japanese Foundation for Cancer Research, Tokyo (R.K.)
| | - Michele McTigue
- Massachusetts General Hospital Cancer Center (A.T.S., L.F., J.F.G., L.D., R.L.F., K.R.S., J.L., G.G., J.A.E.) and the Department of Pathology (A.J.I., L.L., G.G.), Massachusetts General Hospital, Boston, and the Broad Institute of the Massachusetts Institute of Technology and Harvard, Cambridge (I.L., G.G.) - all in Massachusetts; Pfizer Worldwide Research and Development, La Jolla, CA (S.B., A.B., B.J.B., Y.-L.D., W.L., L.P.J., T.S., S.T., M.M., T.W.J.); and the Japanese Foundation for Cancer Research, Tokyo (R.K.)
| | - Gad Getz
- Massachusetts General Hospital Cancer Center (A.T.S., L.F., J.F.G., L.D., R.L.F., K.R.S., J.L., G.G., J.A.E.) and the Department of Pathology (A.J.I., L.L., G.G.), Massachusetts General Hospital, Boston, and the Broad Institute of the Massachusetts Institute of Technology and Harvard, Cambridge (I.L., G.G.) - all in Massachusetts; Pfizer Worldwide Research and Development, La Jolla, CA (S.B., A.B., B.J.B., Y.-L.D., W.L., L.P.J., T.S., S.T., M.M., T.W.J.); and the Japanese Foundation for Cancer Research, Tokyo (R.K.)
| | - Ted W Johnson
- Massachusetts General Hospital Cancer Center (A.T.S., L.F., J.F.G., L.D., R.L.F., K.R.S., J.L., G.G., J.A.E.) and the Department of Pathology (A.J.I., L.L., G.G.), Massachusetts General Hospital, Boston, and the Broad Institute of the Massachusetts Institute of Technology and Harvard, Cambridge (I.L., G.G.) - all in Massachusetts; Pfizer Worldwide Research and Development, La Jolla, CA (S.B., A.B., B.J.B., Y.-L.D., W.L., L.P.J., T.S., S.T., M.M., T.W.J.); and the Japanese Foundation for Cancer Research, Tokyo (R.K.)
| | - Jeffrey A Engelman
- Massachusetts General Hospital Cancer Center (A.T.S., L.F., J.F.G., L.D., R.L.F., K.R.S., J.L., G.G., J.A.E.) and the Department of Pathology (A.J.I., L.L., G.G.), Massachusetts General Hospital, Boston, and the Broad Institute of the Massachusetts Institute of Technology and Harvard, Cambridge (I.L., G.G.) - all in Massachusetts; Pfizer Worldwide Research and Development, La Jolla, CA (S.B., A.B., B.J.B., Y.-L.D., W.L., L.P.J., T.S., S.T., M.M., T.W.J.); and the Japanese Foundation for Cancer Research, Tokyo (R.K.)
| |
Collapse
|
854
|
Rooney C, Sethi T. Advances in molecular biology of lung disease: aiming for precision therapy in non-small cell lung cancer. Chest 2016; 148:1063-1072. [PMID: 26182407 DOI: 10.1378/chest.14-2663] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Lung cancer is the principal cause of cancer-related mortality in the developed world, accounting for almost one-quarter of all cancer deaths. Traditional treatment algorithms have largely relied on histologic subtype and have comprised pragmatic chemotherapy regimens with limited efficacy. However, because our understanding of the molecular basis of disease in non-small cell lung cancer (NSCLC) has improved exponentially, it has become apparent that NSCLC can be radically subdivided, or molecularly characterized, based on recurrent driver mutations occurring in specific oncogenes. We know that the presence of such mutations leads to constitutive activation of aberrant signaling proteins that initiate, progress, and sustain tumorigenesis. This persistence of the malignant phenotype is referred to as "oncogene addiction." On this basis, a paradigm shift in treatment approach has occurred. Rational, targeted therapies have been developed, the first being tyrosine kinase inhibitors (TKIs), which entered the clinical arena > 10 years ago. These were tremendously successful, significantly affecting the natural history of NSCLC and improving patient outcomes. However, the benefits of these drugs are somewhat limited by the emergence of adaptive resistance mechanisms, and efforts to tackle this phenomenon are ongoing. A better understanding of all types of oncogene-driven NSCLC and the occurrence of TKI resistance will help us to further develop second- and third-generation small molecule inhibitors and will expand our range of precision therapies for this disease.
Collapse
Affiliation(s)
- Claire Rooney
- Division of Asthma, Allergy and Lung Biology, King's College London, London, England
| | - Tariq Sethi
- Division of Asthma, Allergy and Lung Biology, King's College London, London, England; Department of Respiratory Medicine, King's Health Partners, London, England.
| |
Collapse
|
855
|
[News about targeted therapies in non-small-cell lung cancer in 2015 (except immuno-therapy)]. Ann Pathol 2016; 36:63-72. [PMID: 26775573 DOI: 10.1016/j.annpat.2015.11.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 11/08/2015] [Indexed: 01/06/2023]
Abstract
Recently, developments of therapies that target abnormally activated signaling pathways are increasing for patients with non-small cell lung cancer. EGFR mutations are found in about 10% of lung cancers, especially in adenocarcinoma, women and non-smokers. Three EGFR inhibitors (erlotinib, gefitinib and afatinib) received a European marketing authorization for up to first line treatment of EGFR mutated NSCLC. Effectiveness of EGFR inhibitors is higher than conventional chemotherapy. Third generation EGFR inhibitors (rociletinib, AZD9291) are effective for patients who develop a resistance mutation such as T790M resistance mutation; they obtained temporary authorization for use in France in 2015. The EML4-ALK translocation is found in about 5% of NSCLC and more particularly in adenocarcinoma of young non-smoking patients. Crizotinib is a new therapeutic standard in ALK translocated NSCLC in second line. Ceritinib is a 2nd generation ALK inhibitor which received a European marketing authorization for up to treatment of ALK translocated NSCLC after progression with crizotinib. INCA supports ACSé program evaluating the efficacy of crizotinib in NSCLC amplified for MET or translocated for ROS1 and ACSé program evaluating the efficacy of vemurafenib in tumors non melanoma mutated V600E BRAF. The role of other biomarkers such as KRAS, BRAF, HER2 and PI3KCA mutations remains to be defined in NSCLC.
Collapse
|
856
|
Gardiner RE, Jahangeer S, Forde P, Ariffin AB, Bird B, Soden D, Hinchion J. Low immunogenicity in non-small cell lung cancer; do new developments and novel treatments have a role? Cancer Metastasis Rev 2016; 34:129-44. [PMID: 25726003 DOI: 10.1007/s10555-015-9550-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Approximately 1.6 million new cases of lung cancer are diagnosed annually (Jemal et al. CA: A Cancer Journal for Clinicians, 61, 69-90, 2011) and it remains the leading cause of cancer-related mortality worldwide. Despite decades of bench and clinical research to attempt to improve outcome for locally advanced, good performance status patients, the 5-year survival remains less than 15 % (Molina et al. 2008). Immune checkpoint inhibitor (ICH) therapies have shown a significant promise in preclinical and clinical trails to date in the treatment of non-small cell lung cancer (NSCLC). The idea of combining these systemic immune therapies with local ablative techniques is one that is gaining momentum. Electrochemotherapy (ECT) is a unique atraumatic local therapy that has had very promising objective response rates and a number of advantages including but not limited to its immunostimulatory effects. ECT in combination with ICHs offers a novel approach for dealing with this difficult disease process.
Collapse
Affiliation(s)
- R E Gardiner
- Cork Cancer Research Centre, University College Cork, Cork, Ireland,
| | | | | | | | | | | | | |
Collapse
|
857
|
Muller IB, De Langen AJ, Honeywell RJ, Giovannetti E, Peters GJ. Overcoming crizotinib resistance in ALK-rearranged NSCLC with the second-generation ALK-inhibitor ceritinib. Expert Rev Anticancer Ther 2016; 16:147-157. [PMID: 26654422 DOI: 10.1586/14737140.2016.1131612] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
In up to 5% of non-small cell lung cancer (NSCLC) patients, the EML4-ALK translocation drives tumor progression. Treatment with the ALK inhibitor crizotinib is more effective than standard chemotherapy. However, resistance to crizotinib occurs after approximately 8 months. Ceritinib is the first second-generation ALK inhibitor approved for treatment of crizotinib-resistant NSCLC. Ceritinib inhibits two of the most common ALK-mutants that confer resistance to crizotinib: L1196 M and G1269A. Cells with ALK expression are more sensitive to ceritinib than crizotinib (IC50 25 nM vs. 150 nM, respectively). Alternative second-generation ALK inhibitors such as Alectinib, Brigatinib and PF-06463922 are currently in development, each affecting different crizotinib-resistant ALK target mutations. Genetic identification of crizotinib-resistant mutants is essential for selecting the optimal treatment strategy in NSCLC patients to overcome resistance and to increase progression-free survival.
Collapse
Affiliation(s)
- Ittai B Muller
- a Department of Medical Oncology , VU University Medical Center , Amsterdam , The Netherlands
| | - Adrianus J De Langen
- b Department of Medical Pulmonology , VU University Medical Center , Amsterdam , The Netherlands
| | - Richard J Honeywell
- a Department of Medical Oncology , VU University Medical Center , Amsterdam , The Netherlands
| | - Elisa Giovannetti
- a Department of Medical Oncology , VU University Medical Center , Amsterdam , The Netherlands
- c Cancer Pharmacology Lab , AIRC Start-Up Unit, DIPINT , Pisa , Italy
| | - Godefridus J Peters
- a Department of Medical Oncology , VU University Medical Center , Amsterdam , The Netherlands
| |
Collapse
|
858
|
Awad MM, Oxnard GR, Jackman DM, Savukoski DO, Hall D, Shivdasani P, Heng JC, Dahlberg SE, Jänne PA, Verma S, Christensen J, Hammerman PS, Sholl LM. MET Exon 14 Mutations in Non-Small-Cell Lung Cancer Are Associated With Advanced Age and Stage-Dependent MET Genomic Amplification and c-Met Overexpression. J Clin Oncol 2016; 34:721-30. [PMID: 26729443 DOI: 10.1200/jco.2015.63.4600] [Citation(s) in RCA: 512] [Impact Index Per Article: 56.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
PURPOSE Non-small-cell lung cancers (NSCLCs) harboring mutations in MET exon 14 and its flanking introns may respond to c-Met inhibitors. We sought to describe the clinical, pathologic, and genomic characteristics of patients with cancer with MET exon 14 mutations. PATIENTS AND METHODS We interrogated next-generation sequencing results from 6,376 cancers to identify those harboring MET exon 14 mutations. Clinical characteristics of MET exon 14 mutated NSCLCs were compared with those of NSCLCs with activating mutations in KRAS and EGFR. Co-occurring genomic mutations and copy number alterations were identified. c-Met immunohistochemistry and real-time polymerase chain reaction to detect exon 14 skipping were performed where sufficient tissue was available. RESULTS MET exon 14 mutations were identified in 28 of 933 nonsquamous NSCLCs (3.0%) and were not seen in other cancer types in this study. Patients with MET exon 14-mutated NSCLC were significantly older (median age, 72.5 years) than patients with EGFR-mutant (median age, 61 years; P < .001) or KRAS-mutant NSCLC (median age, 65 years; P < .001). Among patients with MET exon 14 mutations, 68% were women, and 36% were never-smokers. Stage IV MET exon 14-mutated NSCLCs were significantly more likely to have concurrent MET genomic amplification (mean ratio of MET to chromosome 7, 4.3) and strong c-Met immunohistochemical expression (mean H score, 253) than stage IA to IIIB MET exon 14-mutated NSCLCs (mean ratio of MET to chromosome 7, 1.4; P = .007; mean H score, 155; P = .002) and stage IV MET exon 14-wild-type NSCLCs (mean ratio of MET to chromosome 7, 1.2; P < .001; mean H score, 142; P < .001). A patient whose lung cancer harbored a MET exon 14 mutation with concurrent genomic amplification of the mutated MET allele experienced a major partial response to the c-Met inhibitor crizotinib. CONCLUSION MET exon 14 mutations represent a clinically unique molecular subtype of NSCLC. Prospective clinical trials with c-Met inhibitors will be necessary to validate MET exon 14 mutations as an important therapeutic target in NSCLC.
Collapse
Affiliation(s)
- Mark M Awad
- Mark M. Awad, Geoffrey R. Oxnard, David M. Jackman, Jennifer C. Heng, Suzanne E. Dahlberg, Pasi A. Jänne, and Peter S. Hammerman, Dana-Farber Cancer Institute; Mark M. Awad, Geoffrey R. Oxnard, David M. Jackman, Daniel O. Savukoski, Dimity Hall, Priyanka Shivdasani, Pasi A. Jänne, Peter S. Hammerman, and Lynette M. Sholl, Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Suman Verma, ResearchDX, Irvine; and James Christensen, Mirati Therapeutics, San Diego, CA.
| | - Geoffrey R Oxnard
- Mark M. Awad, Geoffrey R. Oxnard, David M. Jackman, Jennifer C. Heng, Suzanne E. Dahlberg, Pasi A. Jänne, and Peter S. Hammerman, Dana-Farber Cancer Institute; Mark M. Awad, Geoffrey R. Oxnard, David M. Jackman, Daniel O. Savukoski, Dimity Hall, Priyanka Shivdasani, Pasi A. Jänne, Peter S. Hammerman, and Lynette M. Sholl, Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Suman Verma, ResearchDX, Irvine; and James Christensen, Mirati Therapeutics, San Diego, CA
| | - David M Jackman
- Mark M. Awad, Geoffrey R. Oxnard, David M. Jackman, Jennifer C. Heng, Suzanne E. Dahlberg, Pasi A. Jänne, and Peter S. Hammerman, Dana-Farber Cancer Institute; Mark M. Awad, Geoffrey R. Oxnard, David M. Jackman, Daniel O. Savukoski, Dimity Hall, Priyanka Shivdasani, Pasi A. Jänne, Peter S. Hammerman, and Lynette M. Sholl, Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Suman Verma, ResearchDX, Irvine; and James Christensen, Mirati Therapeutics, San Diego, CA
| | - Daniel O Savukoski
- Mark M. Awad, Geoffrey R. Oxnard, David M. Jackman, Jennifer C. Heng, Suzanne E. Dahlberg, Pasi A. Jänne, and Peter S. Hammerman, Dana-Farber Cancer Institute; Mark M. Awad, Geoffrey R. Oxnard, David M. Jackman, Daniel O. Savukoski, Dimity Hall, Priyanka Shivdasani, Pasi A. Jänne, Peter S. Hammerman, and Lynette M. Sholl, Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Suman Verma, ResearchDX, Irvine; and James Christensen, Mirati Therapeutics, San Diego, CA
| | - Dimity Hall
- Mark M. Awad, Geoffrey R. Oxnard, David M. Jackman, Jennifer C. Heng, Suzanne E. Dahlberg, Pasi A. Jänne, and Peter S. Hammerman, Dana-Farber Cancer Institute; Mark M. Awad, Geoffrey R. Oxnard, David M. Jackman, Daniel O. Savukoski, Dimity Hall, Priyanka Shivdasani, Pasi A. Jänne, Peter S. Hammerman, and Lynette M. Sholl, Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Suman Verma, ResearchDX, Irvine; and James Christensen, Mirati Therapeutics, San Diego, CA
| | - Priyanka Shivdasani
- Mark M. Awad, Geoffrey R. Oxnard, David M. Jackman, Jennifer C. Heng, Suzanne E. Dahlberg, Pasi A. Jänne, and Peter S. Hammerman, Dana-Farber Cancer Institute; Mark M. Awad, Geoffrey R. Oxnard, David M. Jackman, Daniel O. Savukoski, Dimity Hall, Priyanka Shivdasani, Pasi A. Jänne, Peter S. Hammerman, and Lynette M. Sholl, Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Suman Verma, ResearchDX, Irvine; and James Christensen, Mirati Therapeutics, San Diego, CA
| | - Jennifer C Heng
- Mark M. Awad, Geoffrey R. Oxnard, David M. Jackman, Jennifer C. Heng, Suzanne E. Dahlberg, Pasi A. Jänne, and Peter S. Hammerman, Dana-Farber Cancer Institute; Mark M. Awad, Geoffrey R. Oxnard, David M. Jackman, Daniel O. Savukoski, Dimity Hall, Priyanka Shivdasani, Pasi A. Jänne, Peter S. Hammerman, and Lynette M. Sholl, Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Suman Verma, ResearchDX, Irvine; and James Christensen, Mirati Therapeutics, San Diego, CA
| | - Suzanne E Dahlberg
- Mark M. Awad, Geoffrey R. Oxnard, David M. Jackman, Jennifer C. Heng, Suzanne E. Dahlberg, Pasi A. Jänne, and Peter S. Hammerman, Dana-Farber Cancer Institute; Mark M. Awad, Geoffrey R. Oxnard, David M. Jackman, Daniel O. Savukoski, Dimity Hall, Priyanka Shivdasani, Pasi A. Jänne, Peter S. Hammerman, and Lynette M. Sholl, Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Suman Verma, ResearchDX, Irvine; and James Christensen, Mirati Therapeutics, San Diego, CA
| | - Pasi A Jänne
- Mark M. Awad, Geoffrey R. Oxnard, David M. Jackman, Jennifer C. Heng, Suzanne E. Dahlberg, Pasi A. Jänne, and Peter S. Hammerman, Dana-Farber Cancer Institute; Mark M. Awad, Geoffrey R. Oxnard, David M. Jackman, Daniel O. Savukoski, Dimity Hall, Priyanka Shivdasani, Pasi A. Jänne, Peter S. Hammerman, and Lynette M. Sholl, Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Suman Verma, ResearchDX, Irvine; and James Christensen, Mirati Therapeutics, San Diego, CA
| | - Suman Verma
- Mark M. Awad, Geoffrey R. Oxnard, David M. Jackman, Jennifer C. Heng, Suzanne E. Dahlberg, Pasi A. Jänne, and Peter S. Hammerman, Dana-Farber Cancer Institute; Mark M. Awad, Geoffrey R. Oxnard, David M. Jackman, Daniel O. Savukoski, Dimity Hall, Priyanka Shivdasani, Pasi A. Jänne, Peter S. Hammerman, and Lynette M. Sholl, Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Suman Verma, ResearchDX, Irvine; and James Christensen, Mirati Therapeutics, San Diego, CA
| | - James Christensen
- Mark M. Awad, Geoffrey R. Oxnard, David M. Jackman, Jennifer C. Heng, Suzanne E. Dahlberg, Pasi A. Jänne, and Peter S. Hammerman, Dana-Farber Cancer Institute; Mark M. Awad, Geoffrey R. Oxnard, David M. Jackman, Daniel O. Savukoski, Dimity Hall, Priyanka Shivdasani, Pasi A. Jänne, Peter S. Hammerman, and Lynette M. Sholl, Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Suman Verma, ResearchDX, Irvine; and James Christensen, Mirati Therapeutics, San Diego, CA
| | - Peter S Hammerman
- Mark M. Awad, Geoffrey R. Oxnard, David M. Jackman, Jennifer C. Heng, Suzanne E. Dahlberg, Pasi A. Jänne, and Peter S. Hammerman, Dana-Farber Cancer Institute; Mark M. Awad, Geoffrey R. Oxnard, David M. Jackman, Daniel O. Savukoski, Dimity Hall, Priyanka Shivdasani, Pasi A. Jänne, Peter S. Hammerman, and Lynette M. Sholl, Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Suman Verma, ResearchDX, Irvine; and James Christensen, Mirati Therapeutics, San Diego, CA
| | - Lynette M Sholl
- Mark M. Awad, Geoffrey R. Oxnard, David M. Jackman, Jennifer C. Heng, Suzanne E. Dahlberg, Pasi A. Jänne, and Peter S. Hammerman, Dana-Farber Cancer Institute; Mark M. Awad, Geoffrey R. Oxnard, David M. Jackman, Daniel O. Savukoski, Dimity Hall, Priyanka Shivdasani, Pasi A. Jänne, Peter S. Hammerman, and Lynette M. Sholl, Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Suman Verma, ResearchDX, Irvine; and James Christensen, Mirati Therapeutics, San Diego, CA
| |
Collapse
|
859
|
Lung Cancer Genomics in the Era of Accelerated Targeted Drug Development. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 890:1-23. [PMID: 26703796 DOI: 10.1007/978-3-319-24932-2_1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Lung cancer is the leading cause of cancer-related deaths in the United States and the 5-year overall survival outlook for a patient has not improved in several decades. Recently, however, molecular and genomic profiling of the lung tumors has revealed recurring somatic mutations. As a result the therapeutic landscape of lung cancer is undergoing a paradigm shift from a purely histology-based understanding of the disease to subtype distinctions based on tumor genetics, which has launched cancer-specific, mechanism-based targeted therapies with clear benefit to patients. While targeted therapy advancements are being made at an ever increasing rate, a new challenge in the form of drug resistance has also emerged. This review summarizes the current literature for these issues.
Collapse
|
860
|
Venur VA, Ahluwalia MS. Targeted Therapy in Brain Metastases: Ready for Primetime? Am Soc Clin Oncol Educ Book 2016; 35:e123-e130. [PMID: 27249714 DOI: 10.1200/edbk_100006] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Brain metastasis is a serious complication of cancer that causes significant morbidity for patients. Over the last decade, numerous new driver somatic mutations have been recognized and targeted therapies are changing the landscape of treatment in lung cancer, breast cancer, and melanoma, which are also the three most common cancers that result in brain metastases. The common actionable mutations include the EGFR mutation and anaplastic lymphoma kinase (ALK) translocations in non-small cell lung cancer, the HER2 mutation in breast cancer, and the BRAF mutation in melanoma. However, most of the early trials with targeted agents excluded patients with brain metastases. With a better understanding of the biology, several recent trials of targeted therapy that focus on brain metastases have been reported and others are ongoing. Novel agents with better penetration across the blood-brain barrier are currently being investigated for patients with brain metastases. In this review, we discuss the current state of use and future directions of targeted therapies in brain metastases.
Collapse
Affiliation(s)
- Vyshak A Venur
- From the Division of Hematology and Oncology, Department of Internal Medicine, University of Iowa Hospitals and Clinics, Iowa City, IA; Burkhardt Brain Tumor and Neuro-Oncology Center, Department of Medicine, Neurologic Institute, Cleveland Clinic, Cleveland, OH; Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
| | - Manmeet S Ahluwalia
- From the Division of Hematology and Oncology, Department of Internal Medicine, University of Iowa Hospitals and Clinics, Iowa City, IA; Burkhardt Brain Tumor and Neuro-Oncology Center, Department of Medicine, Neurologic Institute, Cleveland Clinic, Cleveland, OH; Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
| |
Collapse
|
861
|
Abstract
Lung cancer is the leading cause of cancer-associated mortality in the USA. The median age at diagnosis of lung cancer is 70 years, and thus, about one-half of patients with lung cancer fall into the elderly subgroup. There is dearth of high level of evidence regarding the management of lung cancer in the elderly in the three broad stages of the disease including early-stage, locally advanced, and metastatic disease. A major reason for the lack of evidence is the underrepresentation of elderly in prospective randomized clinical trials. Due to the typical decline in physical and physiologic function associated with aging, most elderly do not meet the stringent eligibility criteria set forth in age-unselected clinical trials. In addition to performance status, ideally, comorbidity, cognitive, and psychological function, polypharmacy, social support, and patient preferences should be taken into account before applying prevailing treatment paradigms often derived in younger, healthier patients to the care of the elderly patient with lung cancer. The purpose of this chapter was to review the existing evidence of management of early-stage, locally advanced disease, and metastatic lung cancer in the elderly.
Collapse
Affiliation(s)
- Archana Rao
- Department of Medicine, Upstate Medical University, 750 E Adams Street, Syracuse, NY, 13210, USA
| | - Namita Sharma
- Department of Medicine, Upstate Medical University, 750 E Adams Street, Syracuse, NY, 13210, USA
| | - Ajeet Gajra
- Department of Medicine, Upstate Medical University, 750 E Adams Street, Syracuse, NY, 13210, USA.
| |
Collapse
|
862
|
The role of a meal on gastrointestinal toxicity and maximum tolerated dose. Lancet Oncol 2016; 17:e2-3. [DOI: 10.1016/s1470-2045(15)00552-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 11/19/2015] [Indexed: 12/24/2022]
|
863
|
Guérin A, Sasane M, Dea K, Zhang J, Culver K, Nitulescu R, Wu EQ, Macalalad AR. The economic burden of brain metastasis among lung cancer patients in the United States. J Med Econ 2016; 19:526-36. [PMID: 26735844 DOI: 10.3111/13696998.2016.1138962] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
OBJECTIVE Brain metastases among lung cancer patients can impair cognitive and functional ability, complicate care, and reduce survival. This study focuses on the economic burden of brain metastasis in lung cancer-direct healthcare costs to payers and indirect costs to patients, payers, and employers-in the US. METHODS Retrospective study using claims data from over 60 self-insured Fortune 500 companies across all US census regions (January 1999-March 2013). Adult, non-elderly lung cancer patients with brain metastasis were evaluated over two study periods: (1) pre-diagnosis (≤30 days prior to first observed lung cancer diagnosis to ≤30 days prior to first-observed brain metastasis diagnosis) and (2) post-diagnosis (≤30 days prior to first observed brain metastasis diagnosis to end of continuous eligibility or observation). OUTCOME MEASURES Healthcare costs to payers and resource utilization, salary loss to patients, disability payouts for payers, and productivity loss to employers. RESULTS A total of 132 patients were followed for a median of 8.4 and 6.6 months in the pre- and post-diagnosis periods, respectively. At diagnosis of brain metastasis, 21.2% of patients were on leave of absence and 6.1% on long-term disability leave. Substantial differences were observed in the pre- vs post-diagnosis periods. Specifically, patients incurred much greater healthcare utilization in the post-diagnosis period, resulting in $25,579 higher medical costs per-patient-per-6-months (PPP6M). During this period, patients missed significantly more work days, generating an incremental burden of $2853 PPP6M in salary loss for patients, $2557 PPP6M in disability payments for payers, and $4570 PPP6M in productivity loss for employers. LIMITATIONS Type of primary lung cancer and extent of brain metastasis could not be assessed in the data. The analysis was also limited to patients with comprehensive disability coverage. CONCLUSIONS Development of brain metastasis among lung cancer patients is associated with a substantial economic burden to payers, patients, and employers.
Collapse
Affiliation(s)
- A Guérin
- a a Analysis Group, Inc. , Montreal , QC , Canada
| | - M Sasane
- b b Novartis Pharmaceuticals Corporation , East Hanover , NJ , USA
| | - K Dea
- a a Analysis Group, Inc. , Montreal , QC , Canada
| | - J Zhang
- b b Novartis Pharmaceuticals Corporation , East Hanover , NJ , USA
| | - K Culver
- b b Novartis Pharmaceuticals Corporation , East Hanover , NJ , USA
| | - R Nitulescu
- a a Analysis Group, Inc. , Montreal , QC , Canada
| | - E Q Wu
- c c Analysis Group, Inc. , Boston , MA , USA
| | | |
Collapse
|
864
|
Anaplastic Lymphoma Kinase (ALK) Signaling in Lung Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 893:179-187. [PMID: 26667344 DOI: 10.1007/978-3-319-24223-1_9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Chromosomal rearrangement in the anaplastic lymphoma kinase (ALK) gene was identified as an oncogenic driver in non-small cell lung cancer (NSCLC) in 2007. A multi-targeted ALK/ROS1/MET inhibitor, crizotinib, targeting this activated tyrosine kinase has led to significant clinical benefit including tumor shrinkage and prolonged survival without disease progression and has been approved by US FDA since 2011 for the treatment of advanced ALK-rearranged NSCLC (Ou et al. Oncologist 17:1351-1375, 2012). Knowledge gained from treating ALK-rearranged NSCLC patients including the presenting clinicopathologic characteristics, methods of detecting ALK-rearranged NSCLC, pattern of relapse and acquired resistance mechanisms while on crizotinib, and the clinical activities of more potent ALK inhibitors has led us to a detailed and ever expanding knowledge of the ALK signaling pathway in lung cancer but also raising many more questions that remained to be answered in the future. This book chapter will provide a concise summary of the importance of ALK signaling pathway in lung cancer. Understanding the ALK signaling pathway in lung cancer will likely provide the roadmap to the management of major epithelial malignancies driven by receptor tyrosine kinase rearrangement.
Collapse
|
865
|
Abstract
Targeted therapies have become standard therapies for patients with non-small cell lung cancer (NSCLC). A phase III trial of carboplatin and paclitaxel with and without bevacizumab in patients with advanced NSCLC with non-squamous histology demonstrated a statistically significant improvement in efficacy. In patients with NSCLC with an activating epidermal growth factor receptor (EGFR) mutation (defined as exon 19 deletion and exon 21 L858R point mutation), phase III trials of EGFR tyrosine kinase inhibitors (TKI) compared to platinum-based chemotherapy have demonstrated superior efficacy in the first-line setting. In patients with NSCLC with anaplastic lymphoma kinase (ALK) rearrangements, phase III trials of crizotinib have demonstrated superior efficacy compared to platinum-pemetrexed in the first-line setting and standard chemotherapy in the second-line setting. A second-generation ALK inhibitor, ceritinib, is available for patients who have progressed after or were intolerant of crizotinib. Crizotinib has also demonstrated activity on patients with ROS1 rearrangements, and BRAF inhibitors (dabrafenib, vemurafenib) have demonstrated activity in patients with NSCLC with BRAF V600E mutation. The oncogenic mutations that are susceptible to targeted therapy are mainly found in non-squamous NSCLC. The development of targeted therapy in patients with squamous NSCLC has been more challenging due to the genomic complexity observed in the squamous histology and the low prevalence of EGFR, ALK, and ROS1 molecular alterations. A phase III trial of cisplatin and gemcitabine with and without necitumumab in patients with advanced NSCLC with squamous histology demonstrated a statistically significant improvement in progression-free and overall survival.
Collapse
Affiliation(s)
- Thomas E Stinchcombe
- University of North Carolina at Chapel Hill, 170 Manning Drive, Physician's Office Building, 3rd Floor, Chapel Hill, NC, 27599-7305, USA.
| |
Collapse
|
866
|
Abstract
Identification of driver mutations in adenocarcinoma of the lung has revolutionized the treatment of this disease. It is now standard of care to look for activating mutations in epidermal growth factor receptor (EGFR), and translocations in anaplastic lymphoma kinase (ALK) or ROS1 in all newly diagnosed adenocarcinoma of the lung, and in many patients with squamous cell carcinoma as well. Recognition of multiple other lung cancer driver mutations has also expanded treatment options. Targeted treatments of these mutations lead to rapid and prolonged responses, but resistance inevitably develops. Until recently, traditional chemotherapy was the only alternative at that time, but better understanding of resistance mechanisms has lead to additional therapeutic options. These mechanisms of resistance and treatments are the focus of this chapter. Understanding of mechanisms of chemotherapy resistance is touched upon, along with a brief discussion of immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Gabriel Rivera
- Stanford University, Kaiser Permanente Fresno, Stanford, USA
| | - Heather A Wakelee
- Stanford University, Stanford Cancer Institute, 875 Blake Wilbur Drive, Stanford, CA, 94305-5826, USA.
| |
Collapse
|
867
|
Chan ELY, Chin CHY, Lui VWY. An update of ALK inhibitors in human clinical trials. Future Oncol 2016; 12:71-81. [DOI: 10.2217/fon.15.293] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The proto-oncogenic ALK is a druggable receptor tyrosine kinase for cancer treatment. Two small molecule inhibitors of ALK, crizotinib and ceritinib, have been recently approved for the treatment of metastatic non-small-cell lung cancer, with marked improvement of progression-free survival of patients. Independent case reports also indicate their potential therapeutic activity in other ALK-rearranged cancers. Numerous single-agent and combination therapy trials are ongoing in lung and many other cancers. Results of these trials are greatly anticipated. Here, we summarize our current understanding of ALK signaling, genomic aberrations in cancer and emerging mechanisms of drug resistance. We will also provide a timely review on all ALK inhibitors and their current status of development in clinical settings.
Collapse
Affiliation(s)
- Eason Leong Yin Chan
- Pharmacogenomics & Precision Therapeutics Laboratory, Department of Pharmacology & Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR
| | - Claudia Ho Yi Chin
- Pharmacogenomics & Precision Therapeutics Laboratory, Department of Pharmacology & Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR
| | - Vivian Wai Yan Lui
- Pharmacogenomics & Precision Therapeutics Laboratory, Department of Pharmacology & Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR
| |
Collapse
|
868
|
Abstract
Understanding the molecular landscape of cancer has facilitated the development of diagnostic, prognostic, and predictive biomarkers for clinical oncology. Developments in next-generation DNA sequencing technologies have increased the speed and reduced the cost of sequencing the nucleic acids of cancer cells. This has unlocked opportunities to characterize the genomic and transcriptomic landscapes of cancer for basic science research through projects like The Cancer Genome Atlas. The cancer genome includes DNA-based alterations, such as point mutations or gene duplications. The cancer transcriptome involves RNA-based alterations, including changes in messenger RNAs. Together, the genome and transcriptome can provide a comprehensive view of an individual patient's cancer that is beginning to impact real-time clinical decision-making. The authors discuss several opportunities for translating this basic science knowledge into clinical practice, including a molecular classification of cancer, heritable risk of cancer, eligibility for targeted therapies, and the development of innovative, genomic-based clinical trials. In this review, key applications and new directions are outlined for translating the cancer genome and transcriptome into patient care in the clinic.
Collapse
Affiliation(s)
- Sameek Roychowdhury
- Department of Internal Medicine, Division of Medical Oncology, The Ohio State University, Columbus, Ohio 43210, USA
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210 USA
- Department of Pharmacology, The Ohio State University, Columbus, Ohio 43210 USA
| | - Arul M. Chinnaiyan
- Michigan Center for Translational Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Howard Hughes Medical Institute, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Urology, University of Michigan Medical School, Ann Arbor, MI 48109 USA
- Center for Computational Medicine and Biology, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
869
|
Sullivan I, Planchard D. ALK inhibitors in non-small cell lung cancer: the latest evidence and developments. Ther Adv Med Oncol 2016; 8:32-47. [PMID: 26753004 PMCID: PMC4699265 DOI: 10.1177/1758834015617355] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
The treatment of patients with advanced non-small cell lung cancer (NSCLC) harbouring chromosomal rearrangements of ALK (anaplastic lymphoma kinase) was revolutionized by crizotinib, a small molecule inhibitor of ALK, ROS1 and MET. Unfortunately, the disease progressed within the first 12 months in most of the patients because of the development of crizotinib resistance in the majority of patients and the emergence of acquired resistance mutations in most of them. Many of them had been reported even before its approval leading to the rapid development of second-generation ALK inhibitors for crizotinib-resistant NSCLC. In the last few years, novel potent ALK inhibitors with promising results and a good toxicity profile have become available: ceritinib (LDK378), alectinib (RG7853/AF-802/RO5424802/CH5424802), brigatinib (AP26113), entrectinib (RXDX-101, NMS-E628), PF-06463922, ASP3026, TSR-011, X-376/X-396 and CEP-28122/CEP-37440. Moreover, HSP90 (90 kDa heat shock protein) inhibitors have demonstrated clinical activity in patients with ALK+ NSCLC. This review focuses on the molecular and clinical properties of this new generation of ALK inhibitors under development in the clinic.
Collapse
Affiliation(s)
| | - David Planchard
- Gustave Roussy – Medical Oncology, 114 rue Édouard Vaillant, Villejuif 94805, France
| |
Collapse
|
870
|
Personalized Medicine in Respiratory Disease: Role of Proteomics. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2015; 102:115-46. [PMID: 26827604 DOI: 10.1016/bs.apcsb.2015.11.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Respiratory diseases affect humanity globally, with chronic lung diseases (e.g., asthma, chronic obstructive pulmonary disease, idiopathic pulmonary fibrosis, among others) and lung cancer causing extensive morbidity and mortality. These conditions are highly heterogeneous and require an early diagnosis. However, initial symptoms are nonspecific, and the clinical diagnosis is made late frequently. Over the last few years, personalized medicine has emerged as a medical care approach that uses novel technology aiming to personalize treatments according to the particular patient's medical needs. This review highlights the contributions of proteomics toward the understanding of personalized medicine in respiratory disease and its potential applications in the clinic.
Collapse
|
871
|
Lau YY, Gu W, Lin T, Song D, Yu R, Scott JW. Effects of meal type on the oral bioavailability of the ALK inhibitor ceritinib in healthy adult subjects. J Clin Pharmacol 2015; 56:559-66. [DOI: 10.1002/jcph.619] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 08/11/2015] [Indexed: 01/30/2023]
Affiliation(s)
- Yvonne Y. Lau
- Novartis Pharmaceuticals Corporation; East Hanover NJ USA
| | - Wen Gu
- Novartis Pharmaceuticals Corporation; East Hanover NJ USA
| | - Tiffany Lin
- Novartis Pharmaceuticals Corporation; East Hanover NJ USA
| | - Dongweon Song
- Novartis Pharmaceuticals Corporation; East Hanover NJ USA
| | - Richard Yu
- Novartis Pharmaceuticals Corporation; East Hanover NJ USA
| | | |
Collapse
|
872
|
SEOM clinical guidelines for the treatment of non-small cell lung cancer (NSCLC) 2015. Clin Transl Oncol 2015; 17:1020-9. [PMID: 26691657 PMCID: PMC4689744 DOI: 10.1007/s12094-015-1455-z] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 11/12/2015] [Indexed: 12/19/2022]
Abstract
Lung cancer is the most common cancer worldwide as well as the leading cause of cancer related deaths as reported by Torre et al (CA Cancer J Clin 65:87–108, 2015]. Non-small cell lung cancer (NSCLC) accounts for up to 85 % of all lung cancers. Multiple advances in the staging, diagnostic procedures, therapeutic options, as well as molecular knowledge have been achieved during the past years, although the overall outlook has not greatly changed for the majority of patients with the overall 5-year survival having marginally increased over the last decade from 15.7 to 17.4 % as reported by Howlader et al. (SEER Cancer Statistics Review 2015).
Collapse
|
873
|
Alectinib in ALK-positive, crizotinib-resistant, non-small-cell lung cancer: a single-group, multicentre, phase 2 trial. Lancet Oncol 2015; 17:234-242. [PMID: 26708155 PMCID: PMC4752892 DOI: 10.1016/s1470-2045(15)00488-x] [Citation(s) in RCA: 510] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Revised: 11/05/2015] [Accepted: 11/06/2015] [Indexed: 01/10/2023]
Abstract
BACKGROUND Alectinib--a highly selective, CNS-active, ALK inhibitor-showed promising clinical activity in crizotinib-naive and crizotinib-resistant patients with ALK-rearranged (ALK-positive) non-small-cell lung cancer (NSCLC). We aimed to assess the safety and efficacy of alectinib in patients with ALK-positive NSCLC who progressed on previous crizotinib. METHODS We did a phase 2 study at 27 centres in the USA and Canada. We enrolled patients aged 18 years or older with stage IIIB-IV, ALK-positive NSCLC who had progressed after crizotinib. Patients were treated with oral alectinib 600 mg twice daily until progression, death, or withdrawal. The primary endpoint was the proportion of patients achieving an objective response by an independent review committee using Response Evaluation Criteria in Solid Tumors, version 1.1. Response endpoints were assessed in the response-evaluable population (ie, patients with measurable disease at baseline who received at least one dose of study drug), and efficacy and safety analyses were done in the intention-to-treat population (all enrolled patients). This study is registered with ClinicalTrials.gov, number NCT01871805. The study is ongoing and patients are still receiving treatment. FINDINGS Between Sept 4, 2013, and Aug 4, 2014, 87 patients were enrolled into the study (intention-to-treat population). At the time of the primary analysis (median follow-up 4·8 months [IQR 3·3-7·1]), 33 of 69 patients with measurable disease at baseline had a confirmed partial response; thus, the proportion of patients achieving an objective response by the independent review committee was 48% (95% CI 36-60). Adverse events were predominantly grade 1 or 2, most commonly constipation (31 [36%]), fatigue (29 [33%]), myalgia 21 [24%]), and peripheral oedema 20 [23%]). The most common grade 3 and 4 adverse events were changes in laboratory values, including increased blood creatine phosphokinase (seven [8%]), increased alanine aminotransferase (five [6%]), and increased aspartate aminotransferase (four [5%]). Two patients died: one had a haemorrhage (judged related to study treatment), and one had disease progression and a history of stroke (judged unrelated to treatment). INTERPRETATION Alectinib showed clinical activity and was well tolerated in patients with ALK-positive NSCLC who had progressed on crizotinib. Therefore, alectinib could be a suitable treatment for patients with ALK-positive disease who have progressed on crizotinib. FUNDING F Hoffmann-La Roche.
Collapse
|
874
|
Landi L, Cappuzzo F. Ceritinib for the treatment of patients with anaplastic lymphoma kinase (ALK)-positive metastatic non-small cell lung cancer. Expert Rev Clin Pharmacol 2015; 9:203-14. [DOI: 10.1586/17512433.2016.1122518] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
|
875
|
Amin AD, Rajan SS, Groysman MJ, Pongtornpipat P, Schatz JH. Oncogene Overdose: Too Much of a Bad Thing for Oncogene-Addicted Cancer Cells. BIOMARKERS IN CANCER 2015; 7:25-32. [PMID: 26688666 PMCID: PMC4681422 DOI: 10.4137/bic.s29326] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Revised: 08/17/2015] [Accepted: 08/19/2015] [Indexed: 01/22/2023]
Abstract
Acquired resistance to targeted inhibitors remains a major, and inevitable, obstacle in the treatment of oncogene-addicted cancers. Newer-generation inhibitors may help overcome resistance mutations, and inhibitor combinations can target parallel pathways, but durable benefit to patients remains elusive in most clinical scenarios. Now, recent studies suggest a third approach may be available in some cases—exploitation of oncogene overexpression that may arise to promote resistance. Here, we discuss the importance of maintaining oncogenic signaling at “just-right” levels in cells, with too much signaling, or oncogene overdose, being potentially as detrimental as too little. This is highlighted in particular by recent studies of mutant-BRAF in melanoma and the fusion kinase nucleophosmin–anaplastic lymphoma kinase (NPM–ALK) in anaplastic large cell lymphoma. Oncogene overdose may be exploitable to prolong tumor control through intermittent dosing in some cases, and studies of acute lymphoid leukemias suggest that it may be specifically pharmacologically inducible.
Collapse
Affiliation(s)
- Amit Dipak Amin
- Department of Medicine, Division of Hematology-Oncology, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Soumya S Rajan
- Sheila and David Fuente Graduate Program in Cancer Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Matthew J Groysman
- Undergraduate Biology Research Program, University of Arizona Cancer Center, Tucson, AZ, USA
| | | | - Jonathan H Schatz
- Department of Medicine, Division of Hematology-Oncology, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
876
|
Minguet J, Smith KH, Bramlage P. Targeted therapies for treatment of non-small cell lung cancer-Recent advances and future perspectives. Int J Cancer 2015; 138:2549-61. [DOI: 10.1002/ijc.29915] [Citation(s) in RCA: 158] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 10/01/2015] [Accepted: 10/29/2015] [Indexed: 12/17/2022]
Affiliation(s)
- Joan Minguet
- European Institute of Cancer Research (EICR); Carrer Del Passeig, 2 Terrassa 08221 Spain
- Institute of Pharmacology and Preventive Medicine (IPPMED); Bahnhofstr. 20 Cloppenburg 49661 Germany
| | - Katherine H. Smith
- European Institute of Cancer Research (EICR); Carrer Del Passeig, 2 Terrassa 08221 Spain
| | - Peter Bramlage
- European Institute of Cancer Research (EICR); Carrer Del Passeig, 2 Terrassa 08221 Spain
- Institute of Pharmacology and Preventive Medicine (IPPMED); Bahnhofstr. 20 Cloppenburg 49661 Germany
| |
Collapse
|
877
|
Stover DG, Wagle N. Precision medicine in breast cancer: genes, genomes, and the future of genomically driven treatments. Curr Oncol Rep 2015; 17:15. [PMID: 25708799 DOI: 10.1007/s11912-015-0438-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Remarkable progress in sequencing technology over the past 20 years has made it possible to comprehensively profile tumors and identify clinically relevant genomic alterations. In breast cancer, the most common malignancy affecting women, we are now increasingly able to use this technology to help specify the use of therapies that target key molecular and genetic dependencies. Large sequencing studies have confirmed the role of well-known cancer-related genes and have also revealed numerous other genes that are recurrently mutated in breast cancer. This growing understanding of patient-to-patient variability at the genomic level in breast cancer is advancing our ability to direct the appropriate treatment to the appropriate patient at the appropriate time--a hallmark of "precision cancer medicine." This review focuses on the technological advances that have catalyzed these developments, the landscape of mutations in breast cancer, the clinical impact of genomic profiling, and the incorporation of genomic information into clinical care and clinical trials.
Collapse
Affiliation(s)
- Daniel G Stover
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, 02215, USA
| | | |
Collapse
|
878
|
Katayama R, Sakashita T, Yanagitani N, Ninomiya H, Horiike A, Friboulet L, Gainor JF, Motoi N, Dobashi A, Sakata S, Tambo Y, Kitazono S, Sato S, Koike S, John Iafrate A, Mino-Kenudson M, Ishikawa Y, Shaw AT, Engelman JA, Takeuchi K, Nishio M, Fujita N. P-glycoprotein Mediates Ceritinib Resistance in Anaplastic Lymphoma Kinase-rearranged Non-small Cell Lung Cancer. EBioMedicine 2015; 3:54-66. [PMID: 26870817 PMCID: PMC4739423 DOI: 10.1016/j.ebiom.2015.12.009] [Citation(s) in RCA: 116] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 11/27/2015] [Accepted: 12/11/2015] [Indexed: 12/17/2022] Open
Abstract
The anaplastic lymphoma kinase (ALK) fusion oncogene is observed in 3%–5% of non-small cell lung cancer (NSCLC). Crizotinib and ceritinib, a next-generation ALK tyrosine kinase inhibitor (TKI) active against crizotinib-refractory patients, are clinically available for the treatment of ALK-rearranged NSCLC patients, and multiple next-generation ALK-TKIs are currently under clinical evaluation. These ALK-TKIs exhibit robust clinical activity in ALK-rearranged NSCLC patients; however, the emergence of ALK-TKI resistance restricts the therapeutic effect. To date, various secondary mutations or bypass pathway activation-mediated resistance have been identified, but large parts of the resistance mechanism are yet to be identified. Here, we report the discovery of p-glycoprotein (P-gp/ABCB1) overexpression as a ceritinib resistance mechanism in ALK-rearranged NSCLC patients. P-gp exported ceritinib and its overexpression conferred ceritinib and crizotinib resistance, but not to PF-06463922 or alectinib, which are next-generation ALK inhibitors. Knockdown of ABCB1 or P-gp inhibitors sensitizes the patient-derived cancer cells to ceritinib, in vitro and in vivo. P-gp overexpression was identified in three out of 11 cases with in ALK-rearranged crizotinib or ceritinib resistant NSCLC patients. Our study suggests that alectinib, PF-06463922, or P-gp inhibitor with ceritinib could overcome the ceritinib or crizotinib resistance mediated by P-gp overexpression. Ceritinib resistant patient-derived cancer cells overexpress P-gp without having mutation in ALK and other major oncogenes. P-gp overexpression conferred the resistance to ceritinib and crizotinib but not to alectinib and PF-06463922. Ceritinib is a substrate of P-gp, and P-gp-inhibitors or knockdown of P-gp reversed ceritinib resistance. P-gp overexpression was observed in 3 out of 11 crizotinib- or ceritinib-resistant ALK-rearranged NSCLC patients. For treatment of ALK-rearranged NSCLC, two ALK-TKIs, crizotinib and ceritinib are currently in use, but the emergence of acquired resistance limits the efficacy of ALK-TKIs. Except for the resistance-associated mutations in ALK, ALK-TKIs resistance mechanisms are still largely unknown. Here we identified P-gp overexpression mediating resistance in three ceritinib-resistant ALK-rearranged NSCLC patients. P-gp overexpression conferred ceritinib and crizotinib resistance but did not confer alectinib and PF-06463922 resistance, and treatment using P-gp inhibitor with ceritinib, or alectinib- or PF-06463922- monotherapy overcame the resistance, suggesting that P-gp expression could be an important determinant in the future treatment strategies.
Collapse
Key Words
- (sh)RNA, small hairpin
- ABC, adenosine triphosphate (ATP)-binding cassette
- ALK
- ALK, anaplastic lymphoma kinase
- ATP, adenosine triphosphate
- BAC, bronchioloalveolar carcinoma
- BBB, blood–brain barrier
- BCRP, breast cancer resistance protein
- CAF, cyclophosphamide, doxorubicin, and fluorouracil
- CSCs, cancer stem/initiating cells
- CT, computed tomography
- Ceritinib
- Crizotinib
- EGFR, epidermal growth factor receptor
- FBS, fetal bovine serum
- FISH, fluorescence in situ hybridization
- IC50, half-maximal inhibitory concentration
- IHC, immunohistochemical
- IRB, institutional review board
- K562/VCR, K562-derived vincristine-resistant
- LCNEC, large cell neuroendocrine carcinoma
- MRP1, multidrug Resistance-associated Protein 1
- ORR, overall response rate
- OS, overall survival
- P-glycoprotein
- P-gp, P-glycoprotein
- PFS, progression-free survival
- ROS1, v-ros avian ur2 sarcoma virus oncogene homolog 1
- RPMI, Roswell Park Memorial Institute
- Resistance
- SP, side population
- TKI, tyrosine kinase inhibitor
- TNM, tumor-node-metastasis
- Tyrosine kinase
Collapse
Affiliation(s)
- Ryohei Katayama
- Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan.
| | - Takuya Sakashita
- Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan; Department of Medical Genome Science, Graduate School of Frontier Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Noriko Yanagitani
- Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan
| | - Hironori Ninomiya
- The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan
| | - Atsushi Horiike
- Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan
| | - Luc Friboulet
- Cancer Center, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Justin F Gainor
- Cancer Center, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Noriko Motoi
- The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan
| | - Akito Dobashi
- The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan
| | - Seiji Sakata
- The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan
| | - Yuichi Tambo
- Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan
| | - Satoru Kitazono
- Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan
| | - Shigeo Sato
- Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan
| | - Sumie Koike
- Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan
| | - A John Iafrate
- Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Mari Mino-Kenudson
- Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Yuichi Ishikawa
- The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan
| | - Alice T Shaw
- Cancer Center, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Jeffrey A Engelman
- Cancer Center, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Kengo Takeuchi
- The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan
| | - Makoto Nishio
- Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan.
| | - Naoya Fujita
- Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan.
| |
Collapse
|
879
|
Adding bevacizumab to chemotherapy effectively control radioresistant brain metastases in ALK-positive lung adenocarcinoma. J Thorac Oncol 2015; 10:e21-2. [PMID: 25789840 DOI: 10.1097/jto.0000000000000449] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
880
|
Noonan SA, Camidge DR. PROFILE 1014: lessons for the new era of lung cancer clinical research. Transl Lung Cancer Res 2015; 4:642-8. [PMID: 26629438 DOI: 10.3978/j.issn.2218-6751.2015.05.02] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
PROFILE 1014 compared crizotinib to up to six cycles of standard platinum-pemetrexed chemotherapy as the first line treatment of advanced anaplastic lymphoma kinase positive (ALK+) non-small cell lung cancer (NSCLC). Overall, PROFILE 1014 has taught us many valuable lessons about the natural history of ALK+ NSCLC, the effectiveness of key therapies and the positive ways in which clinical research in oncogene addicted subtypes of cancer continue to evolve. These lessons include (I) confirming the benefit of using personalized medicine approaches compared to chemotherapy that had already been established in EGFR mutant disease and in ALK+ disease in later lines of therapy; (II) demonstrating that molecular preselection can also affect outcomes from standard chemotherapy in addition to from targeted therapy. Specifically, the benefit of the control arm (platinum-pemetrexed), although inferior to that of crizotinib, was remarkable and expands the dataset on the increased sensitivity of ALK+ NSCLC to pemetrexed; (III) identifying the central nervous system (CNS) as a key battleground for metastatic NSCLC, especially for ALK+ disease. In PROFILE 1014 CNS time to progression (TTP) was included as a prominent secondary endpoint, which showed no difference between crizotinib and chemotherapy but all CNS lesions at baseline had to be both stable and treated, so any apparent stabilizing effect of the drug may be confounded. Ongoing studies with other ALK inhibitors vs. crizotinib that include untreated CNS diseases will provide greater clarity on the true effect of these drugs in the brain.
Collapse
Affiliation(s)
- Sinead A Noonan
- Division of Medical Oncology, Department of Medicine, University of Colorado, Aurora, USA
| | - D Ross Camidge
- Division of Medical Oncology, Department of Medicine, University of Colorado, Aurora, USA
| |
Collapse
|
881
|
Klempner SJ, Raufi A, Ou SHI. Moving molecularly directed therapies to the first-line in ALK-positive lung cancer: crizotinib is just the beginning. Transl Lung Cancer Res 2015; 4:649-52. [PMID: 26629439 DOI: 10.3978/j.issn.2218-6751.2015.07.03] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The increasing appreciation of oncogenic driver alterations in non-small cell lung cancer (NSCLC) has resulted in a rapid expansion of therapeutic compounds. Epidermal growth factor receptor (EGFR) and anaplastic lymphoma kinase (ALK) alterations are the prototypical examples and have driven the paradigm shift in NSCLC management. Early phase studies in previously treated ALK+ patients demonstrated activity and recently Solomon et al. confirmed the superiority of crizotinib over chemotherapy in first line treatment. The phase III PROFILE 1014 represents the culmination of the rapid development of crizotinib and provides lessons for future generation ALK inhibitors and other molecularly directed therapies in NSCLC. Important considerations for second and third generation inhibitors include the ability to overcome known resistance mechanisms, CNS activity, improvement in side effect profile, and safety in possible combination strategies.
Collapse
Affiliation(s)
- Samuel J Klempner
- 1 Department of Medicine, Division of Hematology-Oncology, 2 Department of Medicine, University of California Irvine School of Medicine, Orange, CA 92868, USA
| | - Alexander Raufi
- 1 Department of Medicine, Division of Hematology-Oncology, 2 Department of Medicine, University of California Irvine School of Medicine, Orange, CA 92868, USA
| | - Sai-Hong Ignatius Ou
- 1 Department of Medicine, Division of Hematology-Oncology, 2 Department of Medicine, University of California Irvine School of Medicine, Orange, CA 92868, USA
| |
Collapse
|
882
|
Patel JN, Ersek JL, Kim ES. Lung cancer biomarkers, targeted therapies and clinical assays. Transl Lung Cancer Res 2015; 4:503-14. [PMID: 26629419 DOI: 10.3978/j.issn.2218-6751.2015.06.02] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Until recently, the majority of genomic cancer research has been in discovery and validation; however, as our knowledge of tumor molecular profiling improves, the idea of genomic application in the clinic becomes increasingly tangible, paralleled with the drug development of newer targeted therapies. A number of profiling methodologies exist to identify biomarkers found within the patient (germ-line DNA) and tumor (somatic DNA). Subsequently, commercially available clinical assays to test for both germ-line and somatic alterations that are prognostic and/or predictive of disease outcome, toxicity or treatment response have significantly increased. This review aims to summarize clinically relevant cancer biomarkers that serve as targets for therapy and their potential relationship to lung cancer. In order to realize the full potential of genomic cancer medicine, it is imperative that clinicians understand these intricate molecular pathways, the therapeutic implication of mutations within these pathways, and the availability of clinical assays to identify such biomarkers.
Collapse
Affiliation(s)
- Jai N Patel
- Levine Cancer Institute, Carolinas HealthCare System, Charlotte, NC, USA
| | - Jennifer L Ersek
- Levine Cancer Institute, Carolinas HealthCare System, Charlotte, NC, USA
| | - Edward S Kim
- Levine Cancer Institute, Carolinas HealthCare System, Charlotte, NC, USA
| |
Collapse
|
883
|
Hall RD, Le TM, Haggstrom DE, Gentzler RD. Angiogenesis inhibition as a therapeutic strategy in non-small cell lung cancer (NSCLC). Transl Lung Cancer Res 2015; 4:515-23. [PMID: 26629420 DOI: 10.3978/j.issn.2218-6751.2015.06.09] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
In many cancers, including non-small cell lung cancer (NSCLC), tumor angiogenesis pathways have been identified as important therapeutic targets. Angiogenesis is essential in the process of primary tumor growth, proliferation and metastasis. One of the best characterized group of protein factors for angiogenesis include the members of the vascular endothelial growth factor (VEGF) family, consisting of VEGF-(A-D), and placenta growth factor (PIGF). Targeting tumor angiogenesis has been approached through two primary methods, monoclonal antibodies that block VEGF-vascular endothelial growth factor receptor (VEGFR) binding or small molecule tyrosine kinase inhibitors (TKIs) that inhibit the downstream VEGFR mediated signaling. Many TKIs inhibit multiple pro-angiogenic and pro-proliferative pathways such as the mitogen activated protein (MAP) kinase pathway. Bevacizumab and ramucirumab, monoclonal antibodies targeting VEGF and the VEGFR, respectively, have each led to improvements in overall survival (OS) for NSCLC when added to standard first and second line chemotherapy, respectively. Small incremental gains seen with both bevacizumab and ramucirumab may be further improved upon by incorporating novel agents and treatment strategies, and many additional trials are ongoing.
Collapse
Affiliation(s)
- Richard D Hall
- 1 Division of Hematology/Oncology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA ; 2 Levine Cancer Institute, Carolinas Healthcare System, Charlotte, NC, USA
| | - Tri M Le
- 1 Division of Hematology/Oncology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA ; 2 Levine Cancer Institute, Carolinas Healthcare System, Charlotte, NC, USA
| | - Daniel E Haggstrom
- 1 Division of Hematology/Oncology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA ; 2 Levine Cancer Institute, Carolinas Healthcare System, Charlotte, NC, USA
| | - Ryan D Gentzler
- 1 Division of Hematology/Oncology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA ; 2 Levine Cancer Institute, Carolinas Healthcare System, Charlotte, NC, USA
| |
Collapse
|
884
|
Chuang JC, Neal JW. Crizotinib as first line therapy for advanced ALK-positive non-small cell lung cancers. Transl Lung Cancer Res 2015; 4:639-41. [PMID: 26629437 DOI: 10.3978/j.issn.2218-6751.2015.03.06] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Affiliation(s)
- Jody C Chuang
- 1 Division of Hematology and Oncology, Stanford Hospital & Clinics, Stanford, CA, USA ; 2 Department of Medicine, Division of Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Joel W Neal
- 1 Division of Hematology and Oncology, Stanford Hospital & Clinics, Stanford, CA, USA ; 2 Department of Medicine, Division of Oncology, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
885
|
Tobin NP, Foukakis T, De Petris L, Bergh J. The importance of molecular markers for diagnosis and selection of targeted treatments in patients with cancer. J Intern Med 2015; 278:545-70. [PMID: 26373821 DOI: 10.1111/joim.12429] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The past 30 years have seen the introduction of a number of cancer therapies with the aim of restricting the growth and spread of primary and metastatic tumours. A shared commonality among these therapies is their targeting of various aspects of the cancer hallmarks, that is traits that are essential to successful tumour propagation and dissemination. The evolution of molecular-scale technology has been central to the identification of new cancer targets, and it is not a coincidence that improved therapies have emerged at the same time as gene expression arrays and DNA sequencing have enhanced our understanding of cancer genetics. Modern tumour pathology is now viewed at the molecular level ranging from IHC biomarkers, to gene signature classifiers and gene mutations, all of which provide crucial information about which patients will respond to targeted therapy regimens. In this review, we briefly discuss the general types of targeted therapies used in a clinical setting and provide a short background on immunohistochemical, gene expression and DNA sequencing technologies, before focusing on three tumour types: breast, lung and colorectal cancers. For each of these cancer types, we provide a background to the disease along with an overview of the current standard therapies and then focus on the relevant targeted therapies and the pathways they inhibit. Finally, we highlight several strategies that are pivotal to the successful development of targeted anti-cancer drugs.
Collapse
Affiliation(s)
- N P Tobin
- Department of Oncology and Pathology, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - T Foukakis
- Department of Oncology and Pathology, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - L De Petris
- Department of Oncology and Pathology, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - J Bergh
- Department of Oncology and Pathology, Karolinska Institutet and University Hospital, Stockholm, Sweden
| |
Collapse
|
886
|
Huang JQ, Liang HL, Zhang XC, Xie Z, Jin TE. Synergistic antitumor activity of pro-apoptotic agent PAC-1 with cisplatinum by the activation of CASP3 in pulmonary adenocarcinoma cell line H1299. Asia Pac J Clin Oncol 2015; 12:41-51. [PMID: 26620316 DOI: 10.1111/ajco.12419] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Jian-qing Huang
- Affiliated Cancer Hospital; Guangzhou Medical University; Guangzhou China
| | - Hong-ling Liang
- Affiliated Cancer Hospital; Guangzhou Medical University; Guangzhou China
| | - Xu-chao Zhang
- Guangdong Lung Cancer Institute; Guangdong General Hospital; Guangzhou China
| | - Zhi Xie
- Guangdong Lung Cancer Institute; Guangdong General Hospital; Guangzhou China
| | - Tian-en Jin
- Affiliated Cancer Hospital; Guangzhou Medical University; Guangzhou China
| |
Collapse
|
887
|
Ulivi P, Chiadini E, Dazzi C, Dubini A, Costantini M, Medri L, Puccetti M, Capelli L, Calistri D, Verlicchi A, Gamboni A, Papi M, Mariotti M, De Luigi N, Scarpi E, Bravaccini S, Turolla GM, Amadori D, Crinò L, Delmonte A. Nonsquamous, Non-Small-Cell Lung Cancer Patients Who Carry a Double Mutation of EGFR, EML4-ALK or KRAS: Frequency, Clinical-Pathological Characteristics, and Response to Therapy. Clin Lung Cancer 2015; 17:384-390. [PMID: 26712101 DOI: 10.1016/j.cllc.2015.11.004] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 11/09/2015] [Accepted: 11/12/2015] [Indexed: 10/22/2022]
Abstract
BACKGROUND Epidermal growth factor receptor (EGFR) and v-Ki-ras2 Kirsten rat sarcoma viral oncogene homolog (KRAS) mutations, and echinoderm microtubule-associated protein-like 4 (EML4) anaplastic lymphoma kinase (ALK) translocation are generally considered to be mutually exclusive. However, concomitant mutations are found in a small number of patients and the effect of these on response to targeted therapy is still unknown. PATIENTS AND METHODS We considered 380 non-small-cell lung cancer (NSCLC) patients who underwent nonsequential testing for EGFR and EML4-ALK translocation. KRAS mutation analysis was also performed on 282 patients. RESULTS We found 1.6%, 1.1%, and 2.5% of patients who showed a double mutation comprising EGFR and EML4-ALK, EGFR and KRAS, and EML4-ALK and KRAS, respectively. Twenty-eight patients with EGFR mutation underwent first-line therapy with a tyrosine kinase receptor; a clinical benefit was observed in 81.8% of patients with EGFR mutations only and in 67% of those who also showed an EML4-ALK translocation. Twelve patients with an EML4-ALK translocation received crizotinib and 7 of these had disease progression within 3 months (2 had a concomitant KRAS mutation and 1 had a concomitant EGFR mutation). Two patients showed stable disease, 1 of whom also had a KRAS mutation. Two patients obtained a partial response and 1 had a complete response; all harbored an EML4-ALK translocation only. The median overall survival of patients who carried an EML4-ALK translocation alone or concomitant with a KRAS mutation was 57.1 (range, 10.7-not reached) and 10.7 (range, 4.6-not reached) months, respectively. CONCLUSION Concomitant EGFR, EML4-ALK, or KRAS mutations can occur in NSCLC. Concomitant KRAS mutation and EML4-ALK translocation represents the most common double alteration and confers a poor prognosis.
Collapse
Affiliation(s)
- Paola Ulivi
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy.
| | - Elisa Chiadini
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Claudio Dazzi
- Medical Oncology Unit, S.Maria delle Croci Hospital, Ravenna, Italy
| | | | | | - Laura Medri
- Pathology Unit, Morgagni-Pierantoni Hospital, Forlì, Italy
| | | | - Laura Capelli
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Daniele Calistri
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | | | | | - Maximilian Papi
- Department of Oncology, Per gli Infermi Hospital, Rimini, Italy
| | - Marita Mariotti
- Medical Oncology Unit, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Nicoletta De Luigi
- Medical Oncology Unit, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Emanuela Scarpi
- Unit of Biostatistics and Clinical Trials, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Sara Bravaccini
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | | | - Dino Amadori
- Medical Oncology Unit, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Lucio Crinò
- Division of Medical Oncology, Santa Maria della Misericordia Hospital, Perugia, Italy
| | - Angelo Delmonte
- Medical Oncology Unit, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| |
Collapse
|
888
|
Mohindra NA, Patel JD. Towards manageable toxicities from targeted lung cancer treatment. Lung Cancer Manag 2015. [DOI: 10.2217/lmt.15.30] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Targeted agents are now considered standard of care for patients whose tumors possess a sensitizing mutation in EGFR or ALK rearrangement. As the toxicity profiles of these agents differ significantly from that of cytotoxic chemotherapy, physicians need to be cognizant of the clinically relevant adverse events and manage them aggressively. Early recognition of these toxicities is vital to ensure medication compliance and maintain quality of life for patients. As more novel agents enter the treatment armamentarium, such as third-generation EGFR and ALK inhibitors, it will be important for physicians to understand class-specific toxicities and rare but serious side effects associated with these drugs.
Collapse
Affiliation(s)
- Nisha A Mohindra
- Division of Hematology/Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Robert H Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Jyoti D Patel
- Division of Hematology/Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Robert H Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
889
|
Shi W, Dicker AP. CNS Metastases in Patients With Non-Small-Cell Lung Cancer and ALK Gene Rearrangement. J Clin Oncol 2015; 34:107-9. [PMID: 26628475 DOI: 10.1200/jco.2015.63.9682] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Wenyin Shi
- Thomas Jefferson University, Philadelphia, PA
| | | |
Collapse
|
890
|
Shea M, Costa DB, Rangachari D. Management of advanced non-small cell lung cancers with known mutations or rearrangements: latest evidence and treatment approaches. Ther Adv Respir Dis 2015; 10:113-29. [PMID: 26620497 DOI: 10.1177/1753465815617871] [Citation(s) in RCA: 125] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Precision oncology is now the evidence-based standard of care for the management of many advanced non-small cell lung cancers (NSCLCs). Expert consensus has defined minimum requirements for routine testing and identification of epidermal growth factor (EGFR) mutations (15% of tumors harbor EGFR exon 19 deletions or exon 21 L858R substitutions) and anaplastic lymphoma kinase (ALK) rearrangements (5% of tumors) in advanced lung adenocarcinomas (ACs). Application of palliative targeted therapies with oral tyrosine kinase inhibitors (TKIs) in advanced/metastatic lung ACs harboring abnormalities in EGFR (gefitinib, erlotinib, afatinib) and ALK/ROS1/MET (crizotinib) has consistently led to more favorable outcomes compared with traditional cytotoxic agents. In addition, mutations leading to resistance to first-line EGFR and ALK TKIs can now be successfully inhibited by soon to be approved third-generation EGFR TKIs (osimertinib, rociletinib) and second-generation ALK TKIs (ceritinib, alectinib). Notably, increasing feasibility, accessibility, and application of molecular profiling technologies has permitted dynamic growth in the identification of actionable driver oncogenes. Emerging genomic aberrations for which TKIs have shown impressive results in clinical trials and expansion of drug labels for approved agents are awaited include ROS1 rearrangements (1-2% of tumors, drug: crizotinib) and BRAF-V600E mutations (1-3% of tumors, drugs: vemurafenib, dafrafenib + trametinib). Evolving genomic events in which TKI responses have been reported in smaller series include MET exon 14 skipping mutations (2-4% of tumors, drug: crizotinib); high-level MET amplification (1-2% of tumors, drug: crizotinib); RET rearrangements (1% of tumors, drug: cabozantinib); and ERBB2 mutations (2-3% of tumors, drug: afatinib), among others. Unfortunately, the most common genomic event in NSCLC, KRAS mutations (25-30% of tumors), is not targetable with approved or in development small molecule inhibitors. Here, we review currently approved, emerging, and evolving systemic precision therapies matched with their driver oncogenes for the management of advanced NSCLC.
Collapse
Affiliation(s)
- Meghan Shea
- Division of Hematology/Oncology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School; Boston, MA, USA
| | - Daniel B Costa
- Division of Hematology/Oncology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School; Boston, MA, USA
| | - Deepa Rangachari
- Division of Hematology/Oncology, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Shapiro 9, Boston, MA 02215, USA
| |
Collapse
|
891
|
Liu SY, Mok T, Wu YL. Novel targeted agents for the treatment of lung cancer in China. Cancer 2015; 121 Suppl 17:3089-96. [PMID: 26331815 DOI: 10.1002/cncr.29522] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 05/19/2015] [Accepted: 05/21/2015] [Indexed: 11/09/2022]
Abstract
Since the discovery of epidermal growth factor receptor mutations and epidermal growth factor receptor tyrosine kinase inhibitors, lung cancer treatment in China has entered the precision era. However, novel targeted agents based on novel biomarkers still need to be developed. In the current study, the authors review the biomarker features of lung cancer in the Chinese population and the novel agents that are aimed at novel targets. Furthermore, they discuss issues that require improvement for the production of novel drugs and provided future directions for development in China.
Collapse
Affiliation(s)
- Si-Yang Liu
- Guangdong Lung Cancer Institute, Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Guangdong General Hospital and Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Tony Mok
- State Key Laboratory of Southern China, The Chinese University of Hong Kong, Sir Y.K. Pau Cancer Center, Prince of Wales Hospital, Hong Kong, China
| | - Yi-Long Wu
- Guangdong Lung Cancer Institute, Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Guangdong General Hospital and Guangdong Academy of Medical Sciences, Guangzhou, China
| |
Collapse
|
892
|
Carper MB, Claudio PP. Clinical potential of gene mutations in lung cancer. Clin Transl Med 2015; 4:33. [PMID: 26603430 PMCID: PMC4658345 DOI: 10.1186/s40169-015-0074-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 10/15/2015] [Indexed: 12/11/2022] Open
Abstract
Lung cancer is the most common cancer type worldwide and the leading cause of cancer related deaths in the United States. The majority of newly diagnosed patients present with late stage metastatic lung cancer that is inoperable and resistant to therapies. High-throughput genomic technologies have made the identification of genetic mutations that promote lung cancer progression possible. Identification of the mutations that drive lung cancer provided new targets for non-small cell lung cancer (NSCLC) treatment and led to the development of targeted therapies such as tyrosine kinase inhibitors that can be used to combat the molecular changes that promote cancer progression. Development of targeted therapies is not the only clinical benefit of gene analysis studies. Biomarkers identified from gene analysis can be used for early lung cancer detection, determine patient’s prognosis and response to therapy, and monitor disease progression. Biomarkers can be used to identify the NSCLC patient population that would most benefit from treatment (targeted therapies or chemotherapies), providing clinicians tools that can be used to develop a personalized treatment plan. This review explores the clinical potential of NSCLC genetic studies on diagnosing and treating NSCLC.
Collapse
Affiliation(s)
- Miranda B Carper
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| | - Pier Paolo Claudio
- Department of Radiation Oncology, The University of Mississippi Medical Center Cancer Institute, 350 W Woodrow Wilson Ave, Jackson, MS, 39213, USA. .,Department of Biomedical Sciences, University of Mississippi, National Center for Natural Products Research, Oxford, MS, USA.
| |
Collapse
|
893
|
EMT is associated with, but does not drive resistance to ALK inhibitors among EML4-ALK non-small cell lung cancer. Mol Oncol 2015; 10:601-9. [PMID: 26639656 DOI: 10.1016/j.molonc.2015.11.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Revised: 10/30/2015] [Accepted: 11/10/2015] [Indexed: 11/20/2022] Open
Abstract
ALK gene fusion occurs in approximately 3-7% of non-small cell lung cancer (NSCLC). For patients with ALK positive NCSLC, crizotinib and ceritinib are FDA approved ALK inhibitors, however, patients inevitably acquire resistance to such therapies typically within one to two years. Interrogation of in vitro ALK-positive NSCLC cell line models of acquired resistance to first and second-generation ALK inhibitors revealed acquired epithelial-to-mesenchymal transition (EMT) mechanisms. Here we demonstrated that knockdown of upregulated mesenchymal markers in acquired resistant lines decreased the invasive and migratory capabilities of the cells, however, it did not restore sensitivity to ALK inhibitors. Removing drug for 5 weeks from H3122 cell line that acquired resistance to ceritinib restored its sensitivity to ceritinib. In addition, HSP90 inhibitors ganetespib and 17-AAG were potent in inducing cell death in cell lines resistant to crizotinib and ceritinib. Taken together, EMT does not drive resistance to ALK inhibitors and HSP90 inhibition demonstrates more efficacy when further ALK inhibition may not. This study warrants more exploration of HSP90 inhibitors for ALK-positive patients who progress on 1st and 2nd line ALK inhibitor therapy.
Collapse
|
894
|
Ou SHI, Ahn JS, De Petris L, Govindan R, Yang JCH, Hughes B, Lena H, Moro-Sibilot D, Bearz A, Ramirez SV, Mekhail T, Spira A, Bordogna W, Balas B, Morcos PN, Monnet A, Zeaiter A, Kim DW. Alectinib in Crizotinib-Refractory ALK-Rearranged Non-Small-Cell Lung Cancer: A Phase II Global Study. J Clin Oncol 2015; 34:661-8. [PMID: 26598747 DOI: 10.1200/jco.2015.63.9443] [Citation(s) in RCA: 485] [Impact Index Per Article: 48.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
PURPOSE Crizotinib confers improved progression-free survival compared with chemotherapy in anaplastic lymphoma kinase (ALK)-rearranged non-small-cell lung cancer (NSCLC), but progression invariably occurs. We investigated the efficacy and safety of alectinib, a potent and selective ALK inhibitor with excellent CNS penetration, in patients with crizotinib-refractory ALK-positive NSCLC. PATIENTS AND METHODS Alectinib 600 mg was administered orally twice daily. The primary end point was objective response rate (ORR) by central independent review committee (IRC). RESULTS Of the 138 patients treated, 84 patients (61%) had CNS metastases at baseline, and 122 were response evaluable (RE) by IRC. ORR by IRC was 50% (95% CI, 41% to 59%), and the median duration of response (DOR) was 11.2 months (95% CI, 9.6 months to not reached). In 96 patients (79%) previously treated with chemotherapy, the ORR was 45% (95% CI, 35% to 55%). Median IRC-assessed progression-free survival for all 138 patients was 8.9 months (95% CI, 5.6 to 11.3 months). CNS disease control rate was 83% (95% CI, 74% to 91%), and the median CNS DOR was 10.3 months (95% CI, 7.6 to 11.2 months). CNS ORR in 35 patients with baseline measurable CNS lesions was 57% (95% CI, 39% to 74%). Of the 23 patients with baseline CNS metastases (measurable or nonmeasurable) and no prior radiation, 10 (43%) had a complete CNS response. At 12 months, the cumulative CNS progression rate (24.8%) was lower than the cumulative non-CNS progression rate (33.2%) for all patients. Common adverse events were constipation (33%), fatigue (26%), and peripheral edema (25%); most were grade 1 to 2. CONCLUSION Alectinib is highly active and well tolerated in patients with advanced, crizotinib-refractory ALK-positive NSCLC, including those with CNS metastases.
Collapse
Affiliation(s)
- Sai-Hong Ignatius Ou
- Sai-Hong Ignatius Ou, University of California Irvine School of Medicine, Orange, CA; Jin Seok Ahn, Sungkyunkwan University School of Medicine; Luigi De Petris, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden; Ramaswamy Govindan, Washington University School of Medicine, St Louis, MO; James Chih-Hsin Yang, National Taiwan University, Taipei, Taiwan; Brett Hughes, The Prince Charles Hospital, Chermside, and University of Queensland, Queensland, Australia; Hervé Lena, Centre Hospitalier Universitaire de Rennes, Rennes; Denis Moro-Sibilot, Centre Hospitalier Universitaire de Grenoble, Institut National de la Santé et de la Recherche Médicale U823, Grenoble, France; Alessandra Bearz, National Cancer Institute, Aviano, Italy; Santiago Viteri Ramirez, Quiron-Dexeus University Hospital, Barcelona, Spain; Tarek Mekhail, Florida Hospital Cancer Institute, Orlando, FL; Alexander Spira, Virginia Cancer Specialists, Fairfax, VA; and Walter Bordogna, Bogdana Balas, Peter N. Morcos, Annabelle Monnet, and Ali Zeaiter, F. Hoffmann-La Roche, Basel, Switzerland; Dong-Wan Kim, Seoul National University Hospital, Seoul, South Korea.
| | - Jin Seok Ahn
- Sai-Hong Ignatius Ou, University of California Irvine School of Medicine, Orange, CA; Jin Seok Ahn, Sungkyunkwan University School of Medicine; Luigi De Petris, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden; Ramaswamy Govindan, Washington University School of Medicine, St Louis, MO; James Chih-Hsin Yang, National Taiwan University, Taipei, Taiwan; Brett Hughes, The Prince Charles Hospital, Chermside, and University of Queensland, Queensland, Australia; Hervé Lena, Centre Hospitalier Universitaire de Rennes, Rennes; Denis Moro-Sibilot, Centre Hospitalier Universitaire de Grenoble, Institut National de la Santé et de la Recherche Médicale U823, Grenoble, France; Alessandra Bearz, National Cancer Institute, Aviano, Italy; Santiago Viteri Ramirez, Quiron-Dexeus University Hospital, Barcelona, Spain; Tarek Mekhail, Florida Hospital Cancer Institute, Orlando, FL; Alexander Spira, Virginia Cancer Specialists, Fairfax, VA; and Walter Bordogna, Bogdana Balas, Peter N. Morcos, Annabelle Monnet, and Ali Zeaiter, F. Hoffmann-La Roche, Basel, Switzerland; Dong-Wan Kim, Seoul National University Hospital, Seoul, South Korea
| | - Luigi De Petris
- Sai-Hong Ignatius Ou, University of California Irvine School of Medicine, Orange, CA; Jin Seok Ahn, Sungkyunkwan University School of Medicine; Luigi De Petris, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden; Ramaswamy Govindan, Washington University School of Medicine, St Louis, MO; James Chih-Hsin Yang, National Taiwan University, Taipei, Taiwan; Brett Hughes, The Prince Charles Hospital, Chermside, and University of Queensland, Queensland, Australia; Hervé Lena, Centre Hospitalier Universitaire de Rennes, Rennes; Denis Moro-Sibilot, Centre Hospitalier Universitaire de Grenoble, Institut National de la Santé et de la Recherche Médicale U823, Grenoble, France; Alessandra Bearz, National Cancer Institute, Aviano, Italy; Santiago Viteri Ramirez, Quiron-Dexeus University Hospital, Barcelona, Spain; Tarek Mekhail, Florida Hospital Cancer Institute, Orlando, FL; Alexander Spira, Virginia Cancer Specialists, Fairfax, VA; and Walter Bordogna, Bogdana Balas, Peter N. Morcos, Annabelle Monnet, and Ali Zeaiter, F. Hoffmann-La Roche, Basel, Switzerland; Dong-Wan Kim, Seoul National University Hospital, Seoul, South Korea
| | - Ramaswamy Govindan
- Sai-Hong Ignatius Ou, University of California Irvine School of Medicine, Orange, CA; Jin Seok Ahn, Sungkyunkwan University School of Medicine; Luigi De Petris, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden; Ramaswamy Govindan, Washington University School of Medicine, St Louis, MO; James Chih-Hsin Yang, National Taiwan University, Taipei, Taiwan; Brett Hughes, The Prince Charles Hospital, Chermside, and University of Queensland, Queensland, Australia; Hervé Lena, Centre Hospitalier Universitaire de Rennes, Rennes; Denis Moro-Sibilot, Centre Hospitalier Universitaire de Grenoble, Institut National de la Santé et de la Recherche Médicale U823, Grenoble, France; Alessandra Bearz, National Cancer Institute, Aviano, Italy; Santiago Viteri Ramirez, Quiron-Dexeus University Hospital, Barcelona, Spain; Tarek Mekhail, Florida Hospital Cancer Institute, Orlando, FL; Alexander Spira, Virginia Cancer Specialists, Fairfax, VA; and Walter Bordogna, Bogdana Balas, Peter N. Morcos, Annabelle Monnet, and Ali Zeaiter, F. Hoffmann-La Roche, Basel, Switzerland; Dong-Wan Kim, Seoul National University Hospital, Seoul, South Korea
| | - James Chih-Hsin Yang
- Sai-Hong Ignatius Ou, University of California Irvine School of Medicine, Orange, CA; Jin Seok Ahn, Sungkyunkwan University School of Medicine; Luigi De Petris, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden; Ramaswamy Govindan, Washington University School of Medicine, St Louis, MO; James Chih-Hsin Yang, National Taiwan University, Taipei, Taiwan; Brett Hughes, The Prince Charles Hospital, Chermside, and University of Queensland, Queensland, Australia; Hervé Lena, Centre Hospitalier Universitaire de Rennes, Rennes; Denis Moro-Sibilot, Centre Hospitalier Universitaire de Grenoble, Institut National de la Santé et de la Recherche Médicale U823, Grenoble, France; Alessandra Bearz, National Cancer Institute, Aviano, Italy; Santiago Viteri Ramirez, Quiron-Dexeus University Hospital, Barcelona, Spain; Tarek Mekhail, Florida Hospital Cancer Institute, Orlando, FL; Alexander Spira, Virginia Cancer Specialists, Fairfax, VA; and Walter Bordogna, Bogdana Balas, Peter N. Morcos, Annabelle Monnet, and Ali Zeaiter, F. Hoffmann-La Roche, Basel, Switzerland; Dong-Wan Kim, Seoul National University Hospital, Seoul, South Korea
| | - Brett Hughes
- Sai-Hong Ignatius Ou, University of California Irvine School of Medicine, Orange, CA; Jin Seok Ahn, Sungkyunkwan University School of Medicine; Luigi De Petris, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden; Ramaswamy Govindan, Washington University School of Medicine, St Louis, MO; James Chih-Hsin Yang, National Taiwan University, Taipei, Taiwan; Brett Hughes, The Prince Charles Hospital, Chermside, and University of Queensland, Queensland, Australia; Hervé Lena, Centre Hospitalier Universitaire de Rennes, Rennes; Denis Moro-Sibilot, Centre Hospitalier Universitaire de Grenoble, Institut National de la Santé et de la Recherche Médicale U823, Grenoble, France; Alessandra Bearz, National Cancer Institute, Aviano, Italy; Santiago Viteri Ramirez, Quiron-Dexeus University Hospital, Barcelona, Spain; Tarek Mekhail, Florida Hospital Cancer Institute, Orlando, FL; Alexander Spira, Virginia Cancer Specialists, Fairfax, VA; and Walter Bordogna, Bogdana Balas, Peter N. Morcos, Annabelle Monnet, and Ali Zeaiter, F. Hoffmann-La Roche, Basel, Switzerland; Dong-Wan Kim, Seoul National University Hospital, Seoul, South Korea
| | - Hervé Lena
- Sai-Hong Ignatius Ou, University of California Irvine School of Medicine, Orange, CA; Jin Seok Ahn, Sungkyunkwan University School of Medicine; Luigi De Petris, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden; Ramaswamy Govindan, Washington University School of Medicine, St Louis, MO; James Chih-Hsin Yang, National Taiwan University, Taipei, Taiwan; Brett Hughes, The Prince Charles Hospital, Chermside, and University of Queensland, Queensland, Australia; Hervé Lena, Centre Hospitalier Universitaire de Rennes, Rennes; Denis Moro-Sibilot, Centre Hospitalier Universitaire de Grenoble, Institut National de la Santé et de la Recherche Médicale U823, Grenoble, France; Alessandra Bearz, National Cancer Institute, Aviano, Italy; Santiago Viteri Ramirez, Quiron-Dexeus University Hospital, Barcelona, Spain; Tarek Mekhail, Florida Hospital Cancer Institute, Orlando, FL; Alexander Spira, Virginia Cancer Specialists, Fairfax, VA; and Walter Bordogna, Bogdana Balas, Peter N. Morcos, Annabelle Monnet, and Ali Zeaiter, F. Hoffmann-La Roche, Basel, Switzerland; Dong-Wan Kim, Seoul National University Hospital, Seoul, South Korea
| | - Denis Moro-Sibilot
- Sai-Hong Ignatius Ou, University of California Irvine School of Medicine, Orange, CA; Jin Seok Ahn, Sungkyunkwan University School of Medicine; Luigi De Petris, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden; Ramaswamy Govindan, Washington University School of Medicine, St Louis, MO; James Chih-Hsin Yang, National Taiwan University, Taipei, Taiwan; Brett Hughes, The Prince Charles Hospital, Chermside, and University of Queensland, Queensland, Australia; Hervé Lena, Centre Hospitalier Universitaire de Rennes, Rennes; Denis Moro-Sibilot, Centre Hospitalier Universitaire de Grenoble, Institut National de la Santé et de la Recherche Médicale U823, Grenoble, France; Alessandra Bearz, National Cancer Institute, Aviano, Italy; Santiago Viteri Ramirez, Quiron-Dexeus University Hospital, Barcelona, Spain; Tarek Mekhail, Florida Hospital Cancer Institute, Orlando, FL; Alexander Spira, Virginia Cancer Specialists, Fairfax, VA; and Walter Bordogna, Bogdana Balas, Peter N. Morcos, Annabelle Monnet, and Ali Zeaiter, F. Hoffmann-La Roche, Basel, Switzerland; Dong-Wan Kim, Seoul National University Hospital, Seoul, South Korea
| | - Alessandra Bearz
- Sai-Hong Ignatius Ou, University of California Irvine School of Medicine, Orange, CA; Jin Seok Ahn, Sungkyunkwan University School of Medicine; Luigi De Petris, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden; Ramaswamy Govindan, Washington University School of Medicine, St Louis, MO; James Chih-Hsin Yang, National Taiwan University, Taipei, Taiwan; Brett Hughes, The Prince Charles Hospital, Chermside, and University of Queensland, Queensland, Australia; Hervé Lena, Centre Hospitalier Universitaire de Rennes, Rennes; Denis Moro-Sibilot, Centre Hospitalier Universitaire de Grenoble, Institut National de la Santé et de la Recherche Médicale U823, Grenoble, France; Alessandra Bearz, National Cancer Institute, Aviano, Italy; Santiago Viteri Ramirez, Quiron-Dexeus University Hospital, Barcelona, Spain; Tarek Mekhail, Florida Hospital Cancer Institute, Orlando, FL; Alexander Spira, Virginia Cancer Specialists, Fairfax, VA; and Walter Bordogna, Bogdana Balas, Peter N. Morcos, Annabelle Monnet, and Ali Zeaiter, F. Hoffmann-La Roche, Basel, Switzerland; Dong-Wan Kim, Seoul National University Hospital, Seoul, South Korea
| | - Santiago Viteri Ramirez
- Sai-Hong Ignatius Ou, University of California Irvine School of Medicine, Orange, CA; Jin Seok Ahn, Sungkyunkwan University School of Medicine; Luigi De Petris, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden; Ramaswamy Govindan, Washington University School of Medicine, St Louis, MO; James Chih-Hsin Yang, National Taiwan University, Taipei, Taiwan; Brett Hughes, The Prince Charles Hospital, Chermside, and University of Queensland, Queensland, Australia; Hervé Lena, Centre Hospitalier Universitaire de Rennes, Rennes; Denis Moro-Sibilot, Centre Hospitalier Universitaire de Grenoble, Institut National de la Santé et de la Recherche Médicale U823, Grenoble, France; Alessandra Bearz, National Cancer Institute, Aviano, Italy; Santiago Viteri Ramirez, Quiron-Dexeus University Hospital, Barcelona, Spain; Tarek Mekhail, Florida Hospital Cancer Institute, Orlando, FL; Alexander Spira, Virginia Cancer Specialists, Fairfax, VA; and Walter Bordogna, Bogdana Balas, Peter N. Morcos, Annabelle Monnet, and Ali Zeaiter, F. Hoffmann-La Roche, Basel, Switzerland; Dong-Wan Kim, Seoul National University Hospital, Seoul, South Korea
| | - Tarek Mekhail
- Sai-Hong Ignatius Ou, University of California Irvine School of Medicine, Orange, CA; Jin Seok Ahn, Sungkyunkwan University School of Medicine; Luigi De Petris, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden; Ramaswamy Govindan, Washington University School of Medicine, St Louis, MO; James Chih-Hsin Yang, National Taiwan University, Taipei, Taiwan; Brett Hughes, The Prince Charles Hospital, Chermside, and University of Queensland, Queensland, Australia; Hervé Lena, Centre Hospitalier Universitaire de Rennes, Rennes; Denis Moro-Sibilot, Centre Hospitalier Universitaire de Grenoble, Institut National de la Santé et de la Recherche Médicale U823, Grenoble, France; Alessandra Bearz, National Cancer Institute, Aviano, Italy; Santiago Viteri Ramirez, Quiron-Dexeus University Hospital, Barcelona, Spain; Tarek Mekhail, Florida Hospital Cancer Institute, Orlando, FL; Alexander Spira, Virginia Cancer Specialists, Fairfax, VA; and Walter Bordogna, Bogdana Balas, Peter N. Morcos, Annabelle Monnet, and Ali Zeaiter, F. Hoffmann-La Roche, Basel, Switzerland; Dong-Wan Kim, Seoul National University Hospital, Seoul, South Korea
| | - Alexander Spira
- Sai-Hong Ignatius Ou, University of California Irvine School of Medicine, Orange, CA; Jin Seok Ahn, Sungkyunkwan University School of Medicine; Luigi De Petris, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden; Ramaswamy Govindan, Washington University School of Medicine, St Louis, MO; James Chih-Hsin Yang, National Taiwan University, Taipei, Taiwan; Brett Hughes, The Prince Charles Hospital, Chermside, and University of Queensland, Queensland, Australia; Hervé Lena, Centre Hospitalier Universitaire de Rennes, Rennes; Denis Moro-Sibilot, Centre Hospitalier Universitaire de Grenoble, Institut National de la Santé et de la Recherche Médicale U823, Grenoble, France; Alessandra Bearz, National Cancer Institute, Aviano, Italy; Santiago Viteri Ramirez, Quiron-Dexeus University Hospital, Barcelona, Spain; Tarek Mekhail, Florida Hospital Cancer Institute, Orlando, FL; Alexander Spira, Virginia Cancer Specialists, Fairfax, VA; and Walter Bordogna, Bogdana Balas, Peter N. Morcos, Annabelle Monnet, and Ali Zeaiter, F. Hoffmann-La Roche, Basel, Switzerland; Dong-Wan Kim, Seoul National University Hospital, Seoul, South Korea
| | - Walter Bordogna
- Sai-Hong Ignatius Ou, University of California Irvine School of Medicine, Orange, CA; Jin Seok Ahn, Sungkyunkwan University School of Medicine; Luigi De Petris, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden; Ramaswamy Govindan, Washington University School of Medicine, St Louis, MO; James Chih-Hsin Yang, National Taiwan University, Taipei, Taiwan; Brett Hughes, The Prince Charles Hospital, Chermside, and University of Queensland, Queensland, Australia; Hervé Lena, Centre Hospitalier Universitaire de Rennes, Rennes; Denis Moro-Sibilot, Centre Hospitalier Universitaire de Grenoble, Institut National de la Santé et de la Recherche Médicale U823, Grenoble, France; Alessandra Bearz, National Cancer Institute, Aviano, Italy; Santiago Viteri Ramirez, Quiron-Dexeus University Hospital, Barcelona, Spain; Tarek Mekhail, Florida Hospital Cancer Institute, Orlando, FL; Alexander Spira, Virginia Cancer Specialists, Fairfax, VA; and Walter Bordogna, Bogdana Balas, Peter N. Morcos, Annabelle Monnet, and Ali Zeaiter, F. Hoffmann-La Roche, Basel, Switzerland; Dong-Wan Kim, Seoul National University Hospital, Seoul, South Korea
| | - Bogdana Balas
- Sai-Hong Ignatius Ou, University of California Irvine School of Medicine, Orange, CA; Jin Seok Ahn, Sungkyunkwan University School of Medicine; Luigi De Petris, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden; Ramaswamy Govindan, Washington University School of Medicine, St Louis, MO; James Chih-Hsin Yang, National Taiwan University, Taipei, Taiwan; Brett Hughes, The Prince Charles Hospital, Chermside, and University of Queensland, Queensland, Australia; Hervé Lena, Centre Hospitalier Universitaire de Rennes, Rennes; Denis Moro-Sibilot, Centre Hospitalier Universitaire de Grenoble, Institut National de la Santé et de la Recherche Médicale U823, Grenoble, France; Alessandra Bearz, National Cancer Institute, Aviano, Italy; Santiago Viteri Ramirez, Quiron-Dexeus University Hospital, Barcelona, Spain; Tarek Mekhail, Florida Hospital Cancer Institute, Orlando, FL; Alexander Spira, Virginia Cancer Specialists, Fairfax, VA; and Walter Bordogna, Bogdana Balas, Peter N. Morcos, Annabelle Monnet, and Ali Zeaiter, F. Hoffmann-La Roche, Basel, Switzerland; Dong-Wan Kim, Seoul National University Hospital, Seoul, South Korea
| | - Peter N Morcos
- Sai-Hong Ignatius Ou, University of California Irvine School of Medicine, Orange, CA; Jin Seok Ahn, Sungkyunkwan University School of Medicine; Luigi De Petris, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden; Ramaswamy Govindan, Washington University School of Medicine, St Louis, MO; James Chih-Hsin Yang, National Taiwan University, Taipei, Taiwan; Brett Hughes, The Prince Charles Hospital, Chermside, and University of Queensland, Queensland, Australia; Hervé Lena, Centre Hospitalier Universitaire de Rennes, Rennes; Denis Moro-Sibilot, Centre Hospitalier Universitaire de Grenoble, Institut National de la Santé et de la Recherche Médicale U823, Grenoble, France; Alessandra Bearz, National Cancer Institute, Aviano, Italy; Santiago Viteri Ramirez, Quiron-Dexeus University Hospital, Barcelona, Spain; Tarek Mekhail, Florida Hospital Cancer Institute, Orlando, FL; Alexander Spira, Virginia Cancer Specialists, Fairfax, VA; and Walter Bordogna, Bogdana Balas, Peter N. Morcos, Annabelle Monnet, and Ali Zeaiter, F. Hoffmann-La Roche, Basel, Switzerland; Dong-Wan Kim, Seoul National University Hospital, Seoul, South Korea
| | - Annabelle Monnet
- Sai-Hong Ignatius Ou, University of California Irvine School of Medicine, Orange, CA; Jin Seok Ahn, Sungkyunkwan University School of Medicine; Luigi De Petris, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden; Ramaswamy Govindan, Washington University School of Medicine, St Louis, MO; James Chih-Hsin Yang, National Taiwan University, Taipei, Taiwan; Brett Hughes, The Prince Charles Hospital, Chermside, and University of Queensland, Queensland, Australia; Hervé Lena, Centre Hospitalier Universitaire de Rennes, Rennes; Denis Moro-Sibilot, Centre Hospitalier Universitaire de Grenoble, Institut National de la Santé et de la Recherche Médicale U823, Grenoble, France; Alessandra Bearz, National Cancer Institute, Aviano, Italy; Santiago Viteri Ramirez, Quiron-Dexeus University Hospital, Barcelona, Spain; Tarek Mekhail, Florida Hospital Cancer Institute, Orlando, FL; Alexander Spira, Virginia Cancer Specialists, Fairfax, VA; and Walter Bordogna, Bogdana Balas, Peter N. Morcos, Annabelle Monnet, and Ali Zeaiter, F. Hoffmann-La Roche, Basel, Switzerland; Dong-Wan Kim, Seoul National University Hospital, Seoul, South Korea
| | - Ali Zeaiter
- Sai-Hong Ignatius Ou, University of California Irvine School of Medicine, Orange, CA; Jin Seok Ahn, Sungkyunkwan University School of Medicine; Luigi De Petris, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden; Ramaswamy Govindan, Washington University School of Medicine, St Louis, MO; James Chih-Hsin Yang, National Taiwan University, Taipei, Taiwan; Brett Hughes, The Prince Charles Hospital, Chermside, and University of Queensland, Queensland, Australia; Hervé Lena, Centre Hospitalier Universitaire de Rennes, Rennes; Denis Moro-Sibilot, Centre Hospitalier Universitaire de Grenoble, Institut National de la Santé et de la Recherche Médicale U823, Grenoble, France; Alessandra Bearz, National Cancer Institute, Aviano, Italy; Santiago Viteri Ramirez, Quiron-Dexeus University Hospital, Barcelona, Spain; Tarek Mekhail, Florida Hospital Cancer Institute, Orlando, FL; Alexander Spira, Virginia Cancer Specialists, Fairfax, VA; and Walter Bordogna, Bogdana Balas, Peter N. Morcos, Annabelle Monnet, and Ali Zeaiter, F. Hoffmann-La Roche, Basel, Switzerland; Dong-Wan Kim, Seoul National University Hospital, Seoul, South Korea
| | - Dong-Wan Kim
- Sai-Hong Ignatius Ou, University of California Irvine School of Medicine, Orange, CA; Jin Seok Ahn, Sungkyunkwan University School of Medicine; Luigi De Petris, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden; Ramaswamy Govindan, Washington University School of Medicine, St Louis, MO; James Chih-Hsin Yang, National Taiwan University, Taipei, Taiwan; Brett Hughes, The Prince Charles Hospital, Chermside, and University of Queensland, Queensland, Australia; Hervé Lena, Centre Hospitalier Universitaire de Rennes, Rennes; Denis Moro-Sibilot, Centre Hospitalier Universitaire de Grenoble, Institut National de la Santé et de la Recherche Médicale U823, Grenoble, France; Alessandra Bearz, National Cancer Institute, Aviano, Italy; Santiago Viteri Ramirez, Quiron-Dexeus University Hospital, Barcelona, Spain; Tarek Mekhail, Florida Hospital Cancer Institute, Orlando, FL; Alexander Spira, Virginia Cancer Specialists, Fairfax, VA; and Walter Bordogna, Bogdana Balas, Peter N. Morcos, Annabelle Monnet, and Ali Zeaiter, F. Hoffmann-La Roche, Basel, Switzerland; Dong-Wan Kim, Seoul National University Hospital, Seoul, South Korea
| |
Collapse
|
895
|
Loong HH, Mok K, Leung LKS, Mok TSK. Crizotinib in the management of advanced-stage non-small-cell lung cancer. Future Oncol 2015; 11:735-45. [PMID: 25757678 DOI: 10.2217/fon.14.314] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
ABSTRACT Rearrangement of ALK gene has been identified as exerting a potent transforming effect as driver oncogene in patients with non-small-cell lung cancer (NSCLC). Crizotinib is a small-molecule oral inhibitor of ALK, c-Met/HGF receptor and ROS1 receptor kinases. Its efficacy in ALK-rearranged NSCLC has been established. Crizotinib's effect on ROS1 receptor kinases and c-Met with relevance to NSCLC is also actively being explored. Resistance mechanisms such as secondary gatekeeper mutations in ALK gene and activation of other oncogenes have been identified to confer acquired resistance to crizotinib. This article reviews the pharmacological properties of crizotinib, preclinical and clinical results that led to its approval in ALK-positive NSCLC and current directions of clinical research in overcoming crizotinib resistance.
Collapse
Affiliation(s)
- Herbert H Loong
- Department of Clinical Oncology, State Key Laboratory in Oncology in South China, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
| | | | | | | |
Collapse
|
896
|
Hembrough T, Liao WL, Hartley CP, Ma PC, Velcheti V, Lanigan C, Thyparambil S, An E, Monga M, Krizman D, Burrows J, Tafe LJ. Quantification of Anaplastic Lymphoma Kinase Protein Expression in Non-Small Cell Lung Cancer Tissues from Patients Treated with Crizotinib. Clin Chem 2015; 62:252-61. [PMID: 26585927 DOI: 10.1373/clinchem.2015.245860] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 10/15/2015] [Indexed: 12/17/2022]
Abstract
BACKGROUND Crizotinib has antitumor activity in ALK (anaplastic lymphoma receptor tyrosine kinase)-rearranged non-small cell lung cancer (NSCLC). The current diagnostic test for ALK rearrangement is breakapart fluorescence in situ hybridization (FISH), but FISH has low throughput and is not always reflective of protein concentrations. The emergence of multiple clinically relevant biomarkers in NSCLC necessitates efficient testing of scarce tissue samples. We developed an anaplastic lymphoma kinase (ALK) protein assay that uses multiplexed selected reaction monitoring (SRM) to quantify absolute amounts of ALK in formalin-fixed paraffin-embedded (FFPE) tumor tissue. METHODS After validation in formalin-fixed cell lines, the SRM assay was used to quantify concentrations of ALK in 18 FFPE NSCLC samples that had been tested for ALK by FISH and immunohistochemistry. Results were correlated with patient response to crizotinib. RESULTS We detected ALK in 11 of 14 NSCLC samples with known ALK rearrangements by FISH. Absolute ALK concentrations correlated with clinical response in 5 of 8 patients treated with crizotinib. The SRM assay did not detect ALK in 3 FISH-positive patients who had not responded to crizotinib. In 1 of these cases, DNA sequencing revealed a point mutation that predicts a nonfunctional ALK fusion protein. The SRM assay did not detect ALK in any tumor tissue with a negative ALK status by FISH or immunohistochemistry. CONCLUSIONS ALK concentrations measured by SRM correlate with crizotinib response in NSCLC patients. The ALK SRM proteomic assay, which may be multiplexed with other clinically relevant proteins, allows for rapid identification of patients potentially eligible for targeted therapies.
Collapse
Affiliation(s)
- Todd Hembrough
- OncoPlex Diagnostics, Rockville, MD; NantOmics, LLC, Rockville, MD
| | - Wei-Li Liao
- OncoPlex Diagnostics, Rockville, MD; NantOmics, LLC, Rockville, MD
| | - Christopher P Hartley
- Department of Pathology, Dartmouth-Hitchcock Medical Center, Lebanon, NH; current affiliation: Department of Pathology and Laboratory Medicine, Medical College of Wisconsin, Milwaukee, WI
| | - Patrick C Ma
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Department of Hematology and Medical Oncology, Taussig Cancer Institute, and current affiliation: Mary Babb Randolph Cancer Center, West Virginia University, Morgantown, WV
| | - Vamsidhar Velcheti
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, and
| | - Christopher Lanigan
- Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH
| | | | - Eunkyung An
- OncoPlex Diagnostics, Rockville, MD; NantOmics, LLC, Rockville, MD
| | - Manish Monga
- Mary Babb Randolph Cancer Center, West Virginia University, Morgantown, WV
| | - David Krizman
- OncoPlex Diagnostics, Rockville, MD; NantOmics, LLC, Rockville, MD
| | | | - Laura J Tafe
- Department of Pathology, Dartmouth-Hitchcock Medical Center, Lebanon, NH; Geisel School of Medicine at Dartmouth, Hanover, NH;
| |
Collapse
|
897
|
Giroux Leprieur E, Fallet V, Wislez M. [Modalities of use of ceritinib (Zykadia™), a 2nd generation ALK inhibitor, in advanced stage non-small cell lung cancer]. Bull Cancer 2015; 102:1053-7. [PMID: 26597476 DOI: 10.1016/j.bulcan.2015.09.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 09/16/2015] [Indexed: 11/30/2022]
Abstract
Around 4% of advanced non-small cell lung cancers (NSCLC) harbor a ALK rearrangement, with high sensitivity to ALK inhibitor as crizotinib. However, the vast majority of these tumors end with a tumor progression after several months of treatment with crizotinib. Ceritinib is a 2nd generation ALK inhibitor, which showed high efficiency in NSCLC with ALK rearrangement. Results from phase I trial showed a response rate at 58% in these tumors, with a similar rate for previously crizotinib-treated patients or crizotinib-naïve patients. Moreover, cerebral responses were observed with ceritinib. Preliminary date from a phase 2 trial confirmed these results. These promising results allowed a European marketing authorization (autorisation de mise sur le marché [AMM]) since May 2015 for the treatment of advanced NSCLC with ALK rearrangement and resistance or intolerance to crizotinib.
Collapse
Affiliation(s)
- Etienne Giroux Leprieur
- AP-HP, hôpital Ambroise-Paré, service de pneumologie et oncologie thoracique, 9, avenue Charles-de-Gaulle, 92100 Boulogne-Billancourt, France; Université Versailles - Saint-Quentin-en-Yvelines, laboratoire EA4340, biomarqueurs en cancérologie et onco-hématologie, 9, avenue Charles-de-Gaulle, 92100 Boulogne-Billancourt, France
| | - Vincent Fallet
- Groupe hospitalier Paris - Saint-Joseph, service de pneumologie, 185, rue Raymond-Losserand, 75014 Paris, France
| | - Marie Wislez
- AP-HP, hôpital Tenon, service de pneumologie, 4, rue de la Chine, 75020 Paris, France; UPMC université Paris 06, GRC n(o) 04, Theranoscan, Sorbonne universités, 75252 Paris, France.
| |
Collapse
|
898
|
Postel-Vinay S, Aspeslagh S, Lanoy E, Robert C, Soria JC, Marabelle A. Challenges of phase 1 clinical trials evaluating immune checkpoint-targeted antibodies. Ann Oncol 2015; 27:214-24. [PMID: 26578728 DOI: 10.1093/annonc/mdv550] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 10/27/2015] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Immunostimulatory monoclonal antibodies (imAbs) targeting immune checkpoint molecules are revolutionizing oncology not only regarding cancer therapeutics and clinical care, but also from a drug development point of view. A handful of first-generation molecules have been approved so far based on their tremendous efficacy, after an expedited development phase that has challenged most paradigms established in the era of conventional cytotoxic therapy and to some extent molecularly targeted agents. A huge wave of second-generation imAbs is just entering into phase 1 trials now, in monotherapy or in combination. In order to maximize their chances of success in early phase trials, and eventually for patients' benefit, their clinical development has to benefit from lessons learnt from previous imAbs phase 1 trials. MATERIALS AND METHODS We reviewed the early clinical development of anti-cytotoxic T-lymphocyte antigen 4 and anti-programmed death-1 receptor/ligand. Particularities of each agent, including safety, dose--toxicity and dose--efficacy relationships, scheduling, pharmacokinetics (PK), pharmacodynamics (PD), trial design, biomarkers, response assessment and overall drug development strategies, are described and challenged. RESULTS As opposed to conventional cytotoxic agents, dose of imAbs is not linearly associated with efficacy and toxicity. Therefore, the definition of a minimal immunologically active dose could be proposed. Traditional patient eligibility criteria might also be revisited as the toxicity profile and mechanism of toxicity--immune-related adverse events--are mostly known and their physiopathology somehow less unexpected than with molecularly targeted small molecules. Most challenging are the comprehensive investigation of complex PK and PD characteristics as well as the definition of patient selection biomarkers. Finally, the early focus on efficacy (and not only dose confirmation) in expansion cohorts challenges the traditional phase 1/2/3 drug development process. CONCLUSION Several drug development paradigms have been challenged by imAbs. Here, we discuss novel approaches for an efficient and successful drug development of these agents.
Collapse
Affiliation(s)
- S Postel-Vinay
- DITEP (Département d'Innovations Thérapeutiques et Essais Précoces), Gustave Roussy, Villejuif Faculty of Medicine, Université Paris Saclay, Université Paris-Sud, Paris Inserm Unit U981, Gustave Roussy, Villejuif
| | - S Aspeslagh
- DITEP (Département d'Innovations Thérapeutiques et Essais Précoces), Gustave Roussy, Villejuif
| | - E Lanoy
- Biostatistics and Epidemiology Unit, Gustave-Roussy, Villejuif Inserm Unit U1018, CESP, Université Paris-Sud, Université Paris-Saclay, Villejuif
| | - C Robert
- Faculty of Medicine, Université Paris Saclay, Université Paris-Sud, Paris Inserm Unit U981, Gustave Roussy, Villejuif Department of Medical Oncology, Gustave Roussy, Villejuif
| | - J-C Soria
- DITEP (Département d'Innovations Thérapeutiques et Essais Précoces), Gustave Roussy, Villejuif Faculty of Medicine, Université Paris Saclay, Université Paris-Sud, Paris Inserm Unit U981, Gustave Roussy, Villejuif
| | - A Marabelle
- DITEP (Département d'Innovations Thérapeutiques et Essais Précoces), Gustave Roussy, Villejuif Inserm Unit U1015, Gustave Roussy, Villejuif, France
| |
Collapse
|
899
|
Zhang Q, Wu YL. Safety and efficacy of targeted agents monotherapy in advanced NSCLC. Expert Rev Clin Pharmacol 2015; 9:143-55. [PMID: 26558941 DOI: 10.1586/17512433.2016.1101340] [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: 01/07/2023]
Abstract
The emergence of targetted therapy has revolutionised the treatment of advanced NSCLC. Increasing numbers of driver genes and related targetted agents have supplied more powerful weapons for conquering NSCLC. Depending on whether there are clear targets and therapeutic effects, we can now rank targetted agents into three categories: agents with explicit targets and pure effects, agents with theoretical targets but with no effective biomarkers, agents with vague targets and lower effects. The latest clinical data on the safety and efficacy of monotherapy with three kinds of agents will be reviewed respectively in this article.
Collapse
Affiliation(s)
- Qi Zhang
- a Graduate School , Southern Medical University , Guangzhou , Guangdong , PR China.,b Guangdong Lung Cancer Institute , Guangdong General Hospital and Guangdong Academy of Medical Sciences , Guangzhou , Guangdong , PR China
| | - Yi-Long Wu
- b Guangdong Lung Cancer Institute , Guangdong General Hospital and Guangdong Academy of Medical Sciences , Guangzhou , Guangdong , PR China
| |
Collapse
|
900
|
|