11.07.2015 Views

2DkcTXceO

2DkcTXceO

2DkcTXceO

SHOW MORE
SHOW LESS

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

372 Statistics in a new eraular compounds and “bedside” refers to new treatments developed after preclinicalanimal studies and Phase I, II, and III trials involving human subjects—isamaximofevidence-basedtranslationalmedicalresearch.The development of imatinib, the first drug to target the genetic defectsof a particular cancer while leaving healthy cells unharmed, exemplifies thismaxim and has revolutionized the treatment of cancer. A Phase I clinicaltrial treating CML (chronic myeloid leukemia) patients with the drug beganin June 1998, and within six months remissions had occurred in all patientsas determined by their white blood cell counts returning to normal. In asubsequent five-year study on survival, which followed 553 CML patients whohad received imatinib as their primary therapy, only 5% of the patients diedfrom CML and 11% died from all causes during the five-year period. Moreover,there were few significant side effects; see Druker et al. (2006). Such remarkablesuccess of targeted therapies has led to hundreds of kinase inhibitors andother targeted drugs that are in various stages of development in the presentanticancer drug pipeline.Most new targeted treatments, however, have resulted in only modestclinical benefit, with less than 50% remission rates and less than one yearof progression-free survival, unlike a few cases such as trastuzumab in HER2-positive breast cancer, imatinib in CML and GIST, and gefitinib and erlotinibin non-small cell lung cancer. While the targeted treatments are devised to attackspecific targets, the “one size fits all” treatment regimens commonly usedmay have diminished their effectiveness, and genomic-guided and risk-adaptedpersonalized therapies that are tailored for individual patients are expectedto substantially improve the effectiveness of these treatments. To achieve thispotential for personalized therapies, the first step is to identify and measurethe relevant biomarkers. The markers can be individual genes or proteins orgene expression signatures. The next step is to select drugs (standard cytotoxins,monoclonal antibodies, kinase inhibitors and other targeted drugs) basedon the genetics of the disease in individual patients and biomarkers of drugsensitivity and resistance. The third step is to design clinical trials to providedata for the development and verification of personalized therapies. This isan active area of research and several important developments are reviewedin Lai and Lavori (2011) and Lai et al. (2012). It is an ongoing project of oursat Stanford’s Center for Innovative Study Design.Despite the sequential nature of Phase I–III trials, in which Phase I studiesare used to determine a safe dose or dosage regimen, Phase II trials are usedto evaluate the efficacy of the drug for particular indications (endpoints) inpatients with the disease, and Phase III trials aim to demonstrate the effectivenessof the drug for its approval by the regulatory agency, the trials areoften planned separately, treating each trial as an independent study whosedesign depends on studies in previous phases. Since the sample sizes of thetrials are often inadequate because of separate planning, an alternative strategyis to expand a trial seamlessly from one phase into the next phase, asin the Phase II–III cancer trial designs of Inoue et al. (2002) and Lai et al.

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!