正在加载图片...
PERSPECTIVES with cancer are not all the same and personalized approach to their care. 005 23. e.P.AMechani and future 60637.Us tcimitedtocnroling doe10.1038h318l 66, 20 26 trial desion N ment andng 28 As a con ch Genome Atlas 6 31. rugtargets than ever before.Indeed,the era (BOX nidable and 9 se to and clinically discrete tumour subsets Anincreasing number of drug targets . ncer [r a 12. rD10759 randomi nd pre N.et al P s that nd 14 tumour regr r ly cal d 220 es of 16 in many trials. ew para 18. 4290. 21582005 19. atory 22 LLINKS ARE ACTIVE IN THE ONLNE PDE together to meet these challenges.Patients 947-9572009 366 MAY 2010 VOLUME 9 www.nature.com/reviews/drugdisc 2010 Matmillan Publishers Limited.All rizhts reserve also resistant to EGFR-directed antibodies. These data raise the possibility of further limiting the use of these agents to patients with wild-type genetic markers in each of these segments of the EGFR signalling pathway36. Ongoing and future clinical trials with cetuximab will be limited to enrolling patients with KRAS wild-type tumours, thereby increasing the possibility of demon￾strating benefit from the drug by excluding a non-responsive patient population. Conclusion Personalized cancer care is rapidly becoming a reality in the clinical assessment and management of patients. As a consequence, the expectation is that this approach will improve treatment efficacy, reduce toxicity and minimize cost. Ongoing genome￾profiling activities such as the National Cancer Institute’s Cancer Genome Atlas and the Sanger Institute’s Cancer Genome Project hold promise to reveal more drug targets than ever before. Indeed, the number will grow even more as epigenetic targets are identified. With more than 800 anticancer drugs already in clinical devel￾opment, the challenges of the targeted therapy era (BOX 2) are formidable and include the following: • The identification of more biologically and clinically discrete tumour subsets. • An increasing number of drug targets and agents in development. • Greater use of placebo controls and randomized trials that require more patients and present greater recruitment challenges. • A longer time to reach time-to-event end points for agents that do not produce tumour regression. • More expensive clinical documentation to record progression events. • The availability of multiple lines of effective therapy making it challenging to demonstrate a survival advantage in many trials. • Greater regulatory complexity for studies that seek approval of both drugs and diagnostic tests. New paradigms of cancer biology, such as the concept of tumour stem cells, will further challenge the clinical investigator community to develop clinical end points that can be used to assess the activity of agents that may have no immediate effect on tumour mass or progression, even if they effectively eradicate the stem cell population. The cancer research, clinical and regulatory communities have an obligation to work together to meet these challenges. Patients with cancer are not all the same and each person deserves nothing less than a personalized approach to their care. Richard L. Schilsky is at the Comprehensive Cancer Center, University of Chicago, 5841 South Maryland Avenue, MC 2115, Chicago, Illinois 60637, USA. e‑mail: rschilsk@medicine.bsd.uchicago.edu doi:10.1038/nrd3181 1. Peppercorn, J., Perou, C. M. & Carey, L. A. Molecular subtypes in breast cancer evaluation and management: divide and conquer. Cancer Invest. 26, 1–10 (2008). 2. van de Vijver, M. J. et al. A gene expression signature as a predictor of survival in breast cancer. N. Engl. J. Med. 347, 1999–2009 (2002). 3. Rosenwald, A. et al. The use of molecular profiling to predict survival after chemotherapy for diffuse large B cell lymphoma. N. Engl. J. Med. 346, 1937–1947 (2002). 4. Potti, A. et al. A genomic strategy to refine prognosis in early stage non small cell lung cancer. N. Engl. J. Med. 355, 570–580 (2006). 5. Slamon, D. J. et al. Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N. Engl. J. Med. 344, 783–792 (2001). 6. Lynch, T. J. et al. Activating mutations in the epidermal growth factor receptor underlying responsiveness of non‑small‑cell lung cancer to gefitinib. N. Engl. J. Med. 350, 2129–2139 (2004). 7. Amado, R. G. et al. Wild‑type KRAS is required for panitumumab efficacy in patients with metastatic colorectal cancer. J. Clin. Oncol. 26, 1626–1634 (2008). 8. Hegi, M. E. et al. MGMT gene silencing and benefit from temozolomide in gliobalstoma. N. Engl. J. Med. 352, 997–1003 (2005). 9. O’Brien, S. G. et al. Imatinib compared with interferon and low‑dose cytarabine for newly diagnosed chronic‑ phase chronic myeloid leukemia. N. Engl. J. Med. 348, 994–1004 (2003). 10. Demetri, G. D. et al. Efficacy and safety of imatinib mesylate in advanced gastrointestinal stromal tumors. N. Engl. J. Med. 347, 472–480 (2002). 11. Paik, S. et al. A multi‑gene assay to predict recurrence of tamoxifen‑treated, node negative breast cancer. N. Engl. J. Med. 351, 2817–2826 (2004). 12. Hayes, D. F. et al. Her2 and response to paclitaxel in node positive breast cancer. N. Engl. J. Med. 357, 1496–1506 (2007). 13. Olaussen, K. A. et al. DNA repair by ERCC1 in non small cell lung cancer and cisplatin‑based adjuvant chemotherapy. N. Engl. J. Med. 355, 983–991 (2006). 14. Schuster, S. J. et al. Idiotype vaccine therapy (Biovax ID) in follicular lymphoma in first complete remission: phase III clinical trial results. J. Clin. Oncol. 27 (Jun 20 Suppl.), 2 (2009). 15. Innocenti, F. et al. Comprehensive pharmacogenetic analysis of irinotecan neutropenia and pharmacokinetics. J. Clin. Oncol. 27, 2604–2614 (2009). 16. Dezentjé, V. O., Guchelaar, H. ‑J., Nortier, J. W., van de Velde, C. J. & Gelderblom, H. Clinical implications of CYP2D6 genotyping in tamoxifen treatment for breast cancer. Clin. Cancer Res. 15, 15–21 (2009). 17. Schroth, W. et al. Breast cancer treatment outcome with adjuvant tamoxifen relative to patient CYP2D6 and CYP2C19 genotypes. J. Clin. Oncol. 25, 5187–5193 (2007). 18. Kris, M. et al. Efficacy of gefitinib, an inhibitor of the epidermal growth factor tyrosine kinase, in symptomatic patients with non small cell lung cancer. JAMA 290, 2149–2158 (2003). 19. Giaccone, G. et al. Gefitinib in combination with gemcitabine and cisplatin in advanced non‑small‑cell lung cancer: a Phase III trial — INTACT 1. J. Clin. Oncol. 22, 777–784 (2004). 20. Herbst, R. S. et al. Gefitinib in combination with paclitaxel and carboplatin in advanced non‑small‑cell lung cancer: a Phase III trial — INTACT 2. J. Clin. Oncol. 22, 785–794 (2004). 21. Mok, T. S. et al. Gefitinib or carboplatin‑paclitaxel in pulmonary adenocarcinoma. N. Engl. J. Med. 361, 947–957 (2009). 22. Kobayashi, S. et al. EGFR mutation and resistance of non small cell lung cancer to gefitinib. N. Engl. J. Med. 352, 786–792 (2005). 23. Engelman, J. A. & Janne, P. A. Mechanisms of acquired resistance to epidermal growth factor receptor tyrosine kinase inhibitors in non small cell lung cancer. Clin. Cancer Res. 14, 2895–2899 (2008). 24. Engelman, J. A. et al. MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling. Science 316, 1039–1043 (2007). 25. O’Reilly, K. E. et al. mTor inhibition induces upstream receptor tyrosine kinase signaling and activates AKT. Cancer Res. 66, 1500–1508 (2006). 26. Mandrekar, S. J. & Sargent, D. J. Clinical trial designs for predictive biomarker validation: theoretical considerations and practical challenges. J. Clin. Oncol. 27, 4027–4034 (2009). 27. Shepherd, F. A. et al. Erlotinib in previously treated non small cell lung cancer. N. Engl. J. Med. 353, 123–132 (2005). 28. Bradbury, P. A. et al. Economic analysis: randomized placebo‑controlled clinical trial of erlotinib in advanced non‑small cell lung cancer. J. Natl Cancer Inst. 102, 1–9 (2010). 29. Karapetis, C. S. et al. K‑ras mutations and benefit from cetuximab in advanced colorectal cancer. N. Engl. J. Med. 359, 1757–1765 (2008). 30. Di Fiore, F. et al. Clinical relevance of KRAS mutation detection in metastatic colorectal cancer treated by cetuximab plus chemotherapy. Br. J. Cancer 96, 1166–1169 (2007). 31. Lievre, A. et al. KRAS mutations as an independent prognostic factor in patients with advanced colorectal cancer treated with cetuximab. J. Clin. Oncol. 26, 374–379 (2008). 32. Van Cutsem, E. et al. Cetuximab and chemotherapy as initial treatment for metastatic colorectal cancer. N. Engl. J. Med. 360, 1408–1417 (2009). 33. Allegra, C. J. et al. American Society of Clinical Oncology provisional clinical opinion: testing for KRAS gene mutations in patients with metastatic colorectal carcinoma to predict response to anti‑epidermal growth factor receptor monoclonal antibody therapy. J. Clin. Oncol. 27, 2091–2096 (2009). 34. Shankaran, V., Bentrem, D. J., Mulcahy, M. F., Bennett, C. L. & Benson, A. B. III. Economic implications of kras testing in metastatic colorectal cancer (mCRC). American Association of Clincal Oncology website [online], http://www.asco.org/ ASCOv2/Meetings/Abstracts?&vmview=abst_ detail_view&confID=63&abstractID=10759 (2009). 35. Mittmann, N. et al. Prospective cost‑effectiveness analysis of cetuximab in metastatic colorectal cancer: evaluation of National Cancer Institute of Canada Clinical Trials Group CO.17 trial. J. Natl Cancer Inst. 101, 1182–1192 (2009). 36. Sartore‑Bianchi, A. et al. Multi‑determinants analysis of molecular alterations for predicting clinical benefit to EGFR‑targeted monoclonal antibodies in colorectal cancer. PLOS One 4, e7287 (2009). Competing interests statement The author declares no competing financial interests. DATABASES entrez Gene: http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?db=gene BrAF | CYP2D6 | eGFr | erCC1 | Her2 | KrAs | MGMT | PTeN | UGT1A1 OMiM: http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?db=OMiM Breast cancer | chronic myeloid leukaemia | colorectal cancer | gastointestinal stromal tumours | non-small cell lung cancer FURTHER InFORMATIOn Accelerating Development and Approval of targeted cancer therapies: http://www.brookings.edu/~/media/ Files/events/2009/0914_clinical_cancer_research/ Panel3%20ApresFiNAL.pdf the Brookings institute: http://www.brookings.edu the cancer Genome Atlas: http://cancergenome.nih.gov the cancer Genome Project: http://www.sanger.ac.uk/genetics/CGP All links Are AcTive in The online PDf Pers P ectives 366 | MAY 2010 | vOLUME 9 www.nature.com/reviews/drugdisc © 2010 Macmillan Publishers Limited. All rights reserved
<<向上翻页
©2008-现在 cucdc.com 高等教育资讯网 版权所有