Home Role of Imaging in the Era of Precision Medicine
Post
Cancel

Role of Imaging in the Era of Precision Medicine

Precision medicine is an emerging approach for treating medical disorders, which takes into account individual variability in genetic and environmental factors. Preventive or therapeutic interventions can then be directed to those who will benefit most from targeted interventions, thereby maximizing benefits and minimizing costs and complications. Precision medicine is gaining increasing recognition by clinicians, healthcare systems, pharmaceutical companies, patients, and the government. Imaging plays a critical role in precision medicine including screening, early diagnosis, guiding treatment, evaluating response to therapy, and assessing likelihood of disease recurrence. The Association of University Radiologists Radiology Research Alliance Precision Imaging Task Force convened to explore the current and future role of imaging in the era of precision medicine and summarized its finding in this article. We review the increasingly important role of imaging in various oncological and non-oncological disorders. We also highlight the challenges for radiology in the era of precision medicine.

Introduction

The current era of precision medicine is transforming the practice of medicine with its aim of early diagnosis and personalized treatments, and positively impacting the role of radiology. According to the National Academy of Sciences, “Precision medicine refers to the tailoring of medical treatment to the individual characteristics of each patient, encompassing the ability to classify individuals into subpopulations that differ in their susceptibility to a particular disease, in the biology and/or prognosis of those diseases that may develop, or in their response to a specific treatment” . Preventive or therapeutic interventions can then be directed to those who will benefit, reducing cost and minimizing side effects of therapy.

Precision medicine takes into account individual variability in genetic and environmental factors . Treatments are targeted on the basis of genetic, biomarker, phenotypic, or psychosocial characteristics that distinguish individual patients from others with similar clinical presentations . Precision medicine is receiving growing recognition by clinicians, healthcare systems, pharmaceutical companies, patients, and the government. Advances in genomics, molecular biology, information technology, and imaging are accelerating the acceptance of precision medicine. It takes less than a day to sequence a genome today, whereas it may have taken about 2 years a decade ago. Accordingly, the cost of a complete genome sequence has decreased from $10 million in 2007 to $21,000 in 2011. Based on the data collected from National Human Genome Research Institute-funded genome-sequencing groups, the cost to generate a high-quality “draft” whole human genome sequence in late 2015 was less than $1500 .

Recently, the precision medicine movement has received vital support from President Barack Obama. In the 2015 State of the Union address, the president allocated $215 million to the National Institutes of Health and other regulatory bodies to support this initiative . The initiative will help identify genomic drivers of malignancy and promote innovation in diagnosis and treatment. The goal is to “pioneer a new model of patient-powered research that promises to accelerate biomedical discoveries and provide clinicians with new tools, knowledge, and therapies to select which treatments will work best for which patients” . Ultimately, the aim of precision medicine is to administer the precise treatment to the right patient at the right time .

Imaging will play a pivotal role in precision medicine, including screening, early diagnosis, guiding treatment, evaluating response to therapy, and assessing likelihood of disease recurrence . For precision medicine to succeed, it is critically important that imaging be able to help identify and classify patients in different subgroups who have identical disease characteristics and share similar treatment response and prognosis.

Although the term “radiogenomics” is perceived by radiation oncologists to refer to the study of correlation of genetic variation with response to radiation therapy, it has a different meaning in the radiology community. In radiology, the term “radiogenomics” (also called imaging genomics) refers to the correlation of imaging phenotypes with genotypic expressions, and this is the context in which this term is being used in this review . Radiogenomic studies that help determine statistically significant linkage between imaging features and gene expressions may help create models that predict patient outcomes based on imaging features. Radiogenomics has already attracted major interest in the radiology community, with research undertaken in various cancers such as glioblastoma, breast carcinoma, and renal cell carcinoma (RCC).

Get Radiology Tree app to read full this article<

Get Radiology Tree app to read full this article<

Get Radiology Tree app to read full this article<

Get Radiology Tree app to read full this article<

Role of Imaging in Oncology in the Era of Precision Medicine

Get Radiology Tree app to read full this article<

Breast Cancer

Get Radiology Tree app to read full this article<

Get Radiology Tree app to read full this article<

Get Radiology Tree app to read full this article<

Get Radiology Tree app to read full this article<

Get Radiology Tree app to read full this article<

Brain Tumor

Get Radiology Tree app to read full this article<

Get Radiology Tree app to read full this article<

Get Radiology Tree app to read full this article<

Get Radiology Tree app to read full this article<

Get Radiology Tree app to read full this article<

Lung Cancer

Get Radiology Tree app to read full this article<

Get Radiology Tree app to read full this article<

Get Radiology Tree app to read full this article<

Get Radiology Tree app to read full this article<

Genitourinary Malignancies

Get Radiology Tree app to read full this article<

Get Radiology Tree app to read full this article<

Get Radiology Tree app to read full this article<

Get Radiology Tree app to read full this article<

Role of Imaging in Non-oncological Conditions in the Era of Precision Medicine

Get Radiology Tree app to read full this article<

Get Radiology Tree app to read full this article<

Get Radiology Tree app to read full this article<

Get Radiology Tree app to read full this article<

Challenges for Imaging in the Era of Precision Medicine

Get Radiology Tree app to read full this article<

Get Radiology Tree app to read full this article<

Get Radiology Tree app to read full this article<

Get Radiology Tree app to read full this article<

Get Radiology Tree app to read full this article<

References

  • 1. National Research Council , Division on Earth and Life Studies , Board on Life Sciences , et. al.: Toward precision medicine: building a knowledge network for biomedical research and a new taxonomy of disease.2011.The National Academies PressWashington, DC

  • 2. Obama B.: The precision medicine initiative. Available at https://www.whitehouse.gov/precision-medicine Accessed October 7, 2015

  • 3. Jameson J.L., Longo D.L.: Precision medicine—personalized, problematic, and promising. N Engl J Med 2015; 372: pp. 2229-2234.

  • 4. National Human Genome Research Institute : The cost of sequencing a human genome. Available at https://www.genome.gov/27565109/the-cost-of-sequencing-a-human-genome/ Accessed November 6, 2016

  • 5. Hunt S., Jha S.: Can precision medicine reduce overdiagnosis?. Acad Radiol 2015; 22: pp. 1040-1041.

  • 6. European Society of Radiology : Medical imaging in personalised medicine: a white paper of the research committee of the European Society of Radiology (ESR). Insights Imaging 2015; 6: pp. 141-155.

  • 7. Herold C.J., Lewin J.S., Wibmer A.G., et. al.: Imaging in the age of precision medicine: summary of the proceedings of the 10th biannual symposium of the International Society for Strategic Studies in Radiology. Radiology 2016; 279: pp. 226-238.

  • 8. Mazurowski M.A.: Radiogenomics: what it is and why it is important. J Am Coll Radiol 2015; 12: pp. 862-866.

  • 9. Kumar V., Gu Y., Basu S., et. al.: Radiomics: the process and the challenges. Magn Reson Imaging 2012; 30: pp. 1234-1248.

  • 10. Lambin P., Rios-Velazquez E., Leijenaar R., et. al.: Radiomics: extracting more information from medical images using advanced feature analysis. Eur J Cancer 2012; 48: pp. 441-446.

  • 11. De Cecco C.N., Ciolina M., Caruso D., et. al.: Performance of diffusion-weighted imaging, perfusion imaging, and texture analysis in predicting tumoral response to neoadjuvant chemoradiotherapy in rectal cancer patients studied with 3T MR: initial experience. Abdom Radiol (NY) 2016; 41: pp. 1728-1735.

  • 12. Skogen K., Schulz A., Dormagen J.B., et. al.: Diagnostic performance of texture analysis on MRI in grading cerebral gliomas. Eur J Radiol 2016; 85: pp. 824-829.

  • 13. Smith A.D., Gray M.R., del Campo S.M., et. al.: Predicting overall survival in patients with metastatic melanoma on antiangiogenic therapy and RECIST stable disease on initial posttherapy images using CT texture analysis. AJR Am J Roentgenol 2015; 205: pp. W283-W293.

  • 14. Hayano K., Tian F., Kambadakone A.R., et. al.: Texture analysis of non-contrast-enhanced computed tomography for assessing angiogenesis and survival of soft tissue sarcoma. J Comput Assist Tomogr 2015; 39: pp. 607-612.

  • 15. De Cecco C.N., Ganeshan B., Ciolina M., et. al.: Texture analysis as imaging biomarker of tumoral response to neoadjuvant chemoradiotherapy in rectal cancer patients studied with 3-T magnetic resonance. Invest Radiol 2015; 50: pp. 239-245.

  • 16. Zhang H., Graham C.M., Elci O., et. al.: Locally advanced squamous cell carcinoma of the head and neck: CT texture and histogram analysis allow independent prediction of overall survival in patients treated with induction chemotherapy. Radiology 2013; 269: pp. 801-809.

  • 17. Skogen K., Ganeshan B., Good C., et. al.: Measurements of heterogeneity in gliomas on computed tomography relationship to tumour grade. J Neurooncol 2013; 111: pp. 213-219.

  • 18. Mankoff D.A.: A definition of molecular imaging. J Nucl Med 2007; 48: pp. 18N. 21N

  • 19. American Cancer Society : Cancer Facts & Figures. Available at http://www.cancer.org/research/cancerfactsstatistics/cancerfactsfigures2016/index Accessed March 15, 2016

  • 20. Huber K.E., Carey L.A., Wazer D.E.: Breast cancer molecular subtypes in patients with locally advanced disease: impact on prognosis, patterns of recurrence, and response to therapy. Semin Radiat Oncol 2009; 19: pp. 204-210.

  • 21. Cancer Genome Atlas Network : Comprehensive molecular portraits of human breast tumours. Nature 2012; 490: pp. 61-70.

  • 22. Li H., Zhu Y., Burnside E.S., et. al.: Quantitative MRI radiomics in the prediction of molecular classifications of breast cancer subtypes in the TCGA/TCIA data set. NPJ Breast Cancer 2016; 2: pp. 16012.

  • 23. Li H., Zhu Y., Burnside E.S., et. al.: MR imaging radiomics signatures for predicting the risk of breast cancer recurrence as given by research versions of MammaPrint, Oncotype DX, and PAM50 gene assays. Radiology 2016; 281: pp. 382-391.

  • 24. Guo W., Li H., Zhu Y., et. al.: Prediction of clinical phenotypes in invasive breast carcinomas from the integration of radiomics and genomics data. J Med Imaging (Bellingham) 2015; 2: pp. 041007.

  • 25. Zhu Y., Li H., Guo W., et. al.: Deciphering genomic underpinnings of quantitative MRI-based radiomic phenotypes of invasive breast carcinoma. Sci Rep 2015; 5: pp. 17787.

  • 26. Burnside E.S., Drukker K., Li H., et. al.: Using computer-extracted image phenotypes from tumors on breast magnetic resonance imaging to predict breast cancer pathologic stage. Cancer 2016; 122: pp. 748-757.

  • 27. Mazurowski M.A., Zhang J., Grimm L.J., et. al.: Radiogenomic analysis of breast cancer: luminal B molecular subtype is associated with enhancement dynamics at MR imaging. Radiology 2014; 273: pp. 365-372.

  • 28. Chang R.F., Chen H.H., Chang Y.C., et. al.: Quantification of breast tumor heterogeneity for ER status, HER2 status, and TN molecular subtype evaluation on DCE-MRI. Magn Reson Imaging 2016; 34: pp. 809-819.

  • 29. Kato F., Kudo K., Yamashita H., et. al.: Differences in morphological features and minimum apparent diffusion coefficient values among breast cancer subtypes using 3-tesla MRI. Eur J Radiol 2016; 85: pp. 96-102.

  • 30. Wang J., Kato F., Oyama-Manabe N., et. al.: Identifying triple-negative breast cancer using background parenchymal enhancement heterogeneity on dynamic contrast-enhanced MRI: a pilot radiomics study. PLoS ONE 2015; 10: pp. e0143308.

  • 31. Agner S.C., Rosen M.A., Englander S., et. al.: Computerized image analysis for identifying triple-negative breast cancers and differentiating them from other molecular subtypes of breast cancer on dynamic contrast-enhanced MR images: a feasibility study. Radiology 2014; 272: pp. 91-99.

  • 32. Chen J.H., Agrawal G., Feig B., et. al.: Triple-negative breast cancer: MRI features in 29 patients. Ann Oncol 2007; 18: pp. 2042-2043.

  • 33. Grimm L.J., Zhang J., Mazurowski M.A.: Computational approach to radiogenomics of breast cancer: luminal A and luminal B molecular subtypes are associated with imaging features on routine breast MRI extracted using computer vision algorithms. J Magn Reson Imaging 2015; 42: pp. 902-907.

  • 34. Bahri S., Chen J.H., Yu H.J., et. al.: Can dynamic contrast-enhanced MRI (DCE-MRI) predict tumor recurrence and lymph node status in patients with breast cancer?. Ann Oncol 2008; 19: pp. 822-824.

  • 35. Sutton E.J., Oh J.H., Dashevsky B.Z., et. al.: Breast cancer subtype intertumor heterogeneity: MRI-based features predict results of a genomic assay. J Magn Reson Imaging 2015; 42: pp. 1398-1406.

  • 36. Partridge S.C., Gibbs J.E., Lu Y., et. al.: MRI measurements of breast tumor volume predict response to neoadjuvant chemotherapy and recurrence-free survival. AJR Am J Roentgenol 2005; 184: pp. 1774-1781.

  • 37. Keyaerts M., Xavier C., Heemskerk J., et. al.: Phase I study of 68Ga-HER2-nanobody for PET/CT assessment of HER2 expression in breast carcinoma. J Nucl Med 2016; 57: pp. 27-33.

  • 38. Capala J., Bouchelouche K.: Molecular imaging of HER2-positive breast cancer: a step toward an individualized “image and treat” strategy. Curr Opin Oncol 2010; 22: pp. 559-566.

  • 39. Esserman L.J., Berry D.A., DeMichele A., et. al.: Pathologic complete response predicts recurrence-free survival more effectively by cancer subset: results from the I-SPY 1 TRIAL—CALGB 150007/150012, ACRIN 6657. J Clin Oncol 2012; 30: pp. 3242-3249.

  • 40. Esserman L.J., Berry D.A., Cheang M.C., et. al.: Chemotherapy response and recurrence-free survival in neoadjuvant breast cancer depends on biomarker profiles: results from the I-SPY 1 TRIAL (CALGB 150007/150012; ACRIN 6657). Breast Cancer Res Treat 2012; 132: pp. 1049-1062.

  • 41. Veltman J., Mann R., Kok T., et. al.: Breast tumor characteristics of BRCA1 and BRCA2 gene mutation carriers on MRI. Eur Radiol 2008; 18: pp. 931-938.

  • 42. Saslow D., Boetes C., Burke W., et. al.: American Cancer Society guidelines for breast screening with MRI as an adjunct to mammography. CA Cancer J Clin 2007; 57: pp. 75-89.

  • 43. Kim M.M., Parolia A., Dunphy M.P., et. al.: Non-invasive metabolic imaging of brain tumours in the era of precision medicine. Nat Rev Clin Oncol 2016; 13: pp. 725-739.

  • 44. Hartmann C., Hentschel B., Wick W., et. al.: Patients with IDH1 wild type anaplastic astrocytomas exhibit worse prognosis than IDH1-mutated glioblastomas, and IDH1 mutation status accounts for the unfavorable prognostic effect of higher age: implications for classification of gliomas. Acta Neuropathol 2010; 120: pp. 707-718.

  • 45. Emir U.E., Larkin S.J., de Pennington N., et. al.: Noninvasive quantification of 2-hydroxyglutarate in human gliomas with IDH1 and IDH2 mutations. Cancer Res 2016; 76: pp. 43-49.

  • 46. Choi C., Ganji S.K., DeBerardinis R.J., et. al.: 2-hydroxyglutarate detection by magnetic resonance spectroscopy in IDH-mutated patients with gliomas. Nat Med 2012; 18: pp. 624-629.

  • 47. de la Fuente M.I., Young R.J., Rubel J., et. al.: Integration of 2-hydroxyglutarate-proton magnetic resonance spectroscopy into clinical practice for disease monitoring in isocitrate dehydrogenase-mutant glioma. Neuro Oncol 2016; 18: pp. 283-290.

  • 48. Natsumeda M., Igarashi H., Nomura T., et. al.: Accumulation of 2-hydroxyglutarate in gliomas correlates with survival: a study by 3.0-tesla magnetic resonance spectroscopy. Acta Neuropathol Commun 2014; 2: pp. 158.

  • 49. Rohle D., Popovici-Muller J., Palaskas N., et. al.: An inhibitor of mutant IDH1 delays growth and promotes differentiation of glioma cells. Science 2013; 340: pp. 626-630.

  • 50. Turcan S., Fabius A.W., Borodovsky A., et. al.: Efficient induction of differentiation and growth inhibition in IDH1 mutant glioma cells by the DNMT Inhibitor Decitabine. Oncotarget 2013; 4: pp. 1729-1736.

  • 51. Kool M., Korshunov A., Remke M., et. al.: Molecular subgroups of medulloblastoma: an international meta-analysis of transcriptome, genetic aberrations, and clinical data of WNT, SHH, Group 3, and Group 4 medulloblastomas. Acta Neuropathol 2012; 123: pp. 473-484.

  • 52. Bluml S., Margol A.S., Sposto R., et. al.: Molecular subgroups of medulloblastoma identification using noninvasive magnetic resonance spectroscopy. Neuro Oncol 2016; 18: pp. 126-131.

  • 53. Robinson G.W., Orr B.A., Wu G., et. al.: Vismodegib exerts targeted efficacy against recurrent sonic hedgehog-subgroup medulloblastoma: results from phase II pediatric brain tumor consortium studies PBTC-025B and PBTC-032. J Clin Oncol 2015; 33: pp. 2646-2654.

  • 54. Kieran M.W.: Targeted treatment for sonic hedgehog-dependent medulloblastoma. Neuro Oncol 2014; 16: pp. 1037-1047.

  • 55. Thon N., Kunz M., Lemke L., et. al.: Dynamic 18F-FET PET in suspected WHO grade II gliomas defines distinct biological subgroups with different clinical courses. Int J Cancer 2015; 136: pp. 2132-2145.

  • 56. Witney T.H., James M.L., Shen B., et. al.: PET imaging of tumor glycolysis downstream of hexokinase through noninvasive measurement of pyruvate kinase M2. Sci Transl Med 2015; 7: pp. 310ra169.

  • 57. Mittra E.S., Koglin N., Mosci C., et. al.: Pilot preclinical and clinical evaluation of (4S)-4-(3-[18F]fluoropropyl)-L-glutamate (18F-FSPG) for PET/CT imaging of intracranial malignancies. PLoS ONE 2016; 11: pp. e0148628.

  • 58. Koglin N., Mueller A., Berndt M., et. al.: Specific PET imaging of xC—transporter activity using a (1)(8)F-labeled glutamate derivative reveals a dominant pathway in tumor metabolism. Clin Cancer Res 2011; 17: pp. 6000-6011.

  • 59. Takeuchi S., Wada K., Toyooka T., et. al.: Increased xCT expression correlates with tumor invasion and outcome in patients with glioblastomas. Neurosurgery 2013; 72: pp. 33-41. discussion

  • 60. Challapalli A., Aboagye E.O.: Positron emission tomography imaging of tumor cell metabolism and application to therapy response monitoring. Front Oncol 2016; 6: pp. 44.

  • 61. Perreault S., Ramaswamy V., Achrol A.S., et. al.: MRI surrogates for molecular subgroups of medulloblastoma. AJNR Am J Neuroradiol 2014; 35: pp. 1263-1269.

  • 62. Carrillo J.A., Lai A., Nghiemphu P.L., et. al.: Relationship between tumor enhancement, edema, IDH1 mutational status, MGMT promoter methylation, and survival in glioblastoma. AJNR Am J Neuroradiol 2012; 33: pp. 1349-1355.

  • 63. Baldock A.L., Yagle K., Born D.E., et. al.: Invasion and proliferation kinetics in enhancing gliomas predict IDH1 mutation status. Neuro Oncol 2014; 16: pp. 779-786.

  • 64. Wang K., Wang Y., Fan X., et. al.: Radiological features combined with IDH1 status for predicting the survival outcome of glioblastoma patients. Neuro Oncol 2016; 18: pp. 589-597.

  • 65. Jamshidi N., Diehn M., Bredel M., et. al.: Illuminating radiogenomic characteristics of glioblastoma multiforme through integration of MR imaging, messenger RNA expression, and DNA copy number variation. Radiology 2014; 270: pp. 1-2.

  • 66. Zinn P.O., Mahajan B., Sathyan P., et. al.: Radiogenomic mapping of edema/cellular invasion MRI-phenotypes in glioblastoma multiforme. PLoS ONE 2011; 6: pp. e25451.

  • 67. National Lung Screening Trial Research Team, Church T.R., Black W.C., et. al.: Results of initial low-dose computed tomographic screening for lung cancer. N Engl J Med 2013; 368: pp. 1980-1991.

  • 68. National Lung Screening Trial Research Team, Aberle D.R., Adams A.M., et. al.: Reduced lung-cancer mortality with low-dose computed tomographic screening. N Engl J Med 2011; 365: pp. 395-409.

  • 69. International Early Lung Cancer Action Program Investigators, Henschke C.I., Yankelevitz D.F., et. al.: Survival of patients with stage I lung cancer detected on CT screening. N Engl J Med 2006; 355: pp. 1763-1771.

  • 70. Henschke C.I., McCauley D.I., Yankelevitz D.F., et. al.: Early Lung Cancer Action Project: overall design and findings from baseline screening. Lancet 1999; 354: pp. 99-105.

  • 71. Paez J.G., Janne P.A., Lee J.C., et. al.: EGFR mutations in lung cancer: correlation with clinical response to gefitinib therapy. Science 2004; 304: pp. 1497-1500.

  • 72. Soda M., Choi Y.L., Enomoto M., et. al.: Identification of the transforming EML4-ALK fusion gene in non-small-cell lung cancer. Nature 2007; 448: pp. 561-566.

  • 73. Politi K., Herbst R.S.: Lung cancer in the era of precision medicine. Clin Cancer Res 2015; 21: pp. 2213-2220.

  • 74. Gevaert O., Xu J., Hoang C.D., et. al.: Non-small cell lung cancer: identifying prognostic imaging biomarkers by leveraging public gene expression microarray data—methods and preliminary results. Radiology 2012; 264: pp. 387-396.

  • 75. Pao W., Miller V., Zakowski M., et. al.: EGF receptor gene mutations are common in lung cancers from “never smokers” and are associated with sensitivity of tumors to gefitinib and erlotinib. Proc Natl Acad Sci USA 2004; 101: pp. 13306-13311.

  • 76. Jackman D.M., Yeap B.Y., Sequist L.V., et. al.: Exon 19 deletion mutations of epidermal growth factor receptor are associated with prolonged survival in non-small cell lung cancer patients treated with gefitinib or erlotinib. Clin Cancer Res 2006; 12: pp. 3908-3914.

  • 77. Bean J., Brennan C., Shih J.Y., et. al.: MET amplification occurs with or without T790M mutations in EGFR mutant lung tumors with acquired resistance to gefitinib or erlotinib. Proc Natl Acad Sci USA 2007; 104: pp. 20932-20937.

  • 78. Pao W., Miller V.A., Politi K.A., et. al.: Acquired resistance of lung adenocarcinomas to gefitinib or erlotinib is associated with a second mutation in the EGFR kinase domain. PLoS Med 2005; 2: pp. e73.

  • 79. Pan Y., Xu Y., Feng S., et. al.: SKLB1206, a novel orally available multikinase inhibitor targeting EGFR activating and T790M mutants, ErbB2, ErbB4, and VEGFR2, displays potent antitumor activity both in vitro and in vivo. Mol Cancer Ther 2012; 11: pp. 952-962.

  • 80. Janjigian Y.Y., Azzoli C.G., Krug L.M., et. al.: Phase I/II trial of cetuximab and erlotinib in patients with lung adenocarcinoma and acquired resistance to erlotinib. Clin Cancer Res 2011; 17: pp. 2521-2527.

  • 81. Engelman J.A., Zejnullahu K., Gale C.M., et. al.: PF00299804, an irreversible pan-ERBB inhibitor, is effective in lung cancer models with EGFR and ERBB2 mutations that are resistant to gefitinib. Cancer Res 2007; 67: pp. 11924-11932.

  • 82. Arcila M.E., Oxnard G.R., Nafa K., et. al.: Rebiopsy of lung cancer patients with acquired resistance to EGFR inhibitors and enhanced detection of the T790M mutation using a locked nucleic acid-based assay. Clin Cancer Res 2011; 17: pp. 1169-1180.

  • 83. Marcus D.M., Rossi P.J., Nour S.G., et. al.: The impact of multiparametric pelvic magnetic resonance imaging on risk stratification in patients with localized prostate cancer. Urology 2014; 84: pp. 132-137.

  • 84. Schoots I.G., Roobol M.J., Nieboer D., et. al.: Magnetic resonance imaging-targeted biopsy may enhance the diagnostic accuracy of significant prostate cancer detection compared to standard transrectal ultrasound-guided biopsy: a systematic review and meta-analysis. Eur Urol 2015; 68: pp. 438-450.

  • 85. Valerio M., Donaldson I., Emberton M., et. al.: Detection of clinically significant prostate cancer using magnetic resonance imaging-ultrasound fusion targeted biopsy: a systematic review. Eur Urol 2015; 68: pp. 8-19.

  • 86. Abd-Alazeez M., Ahmed H.U., Arya M., et. al.: The accuracy of multiparametric MRI in men with negative biopsy and elevated PSA level—can it rule out clinically significant prostate cancer?. Urol Oncol 2014; 32: pp. 45. e17-e22

  • 87. Hoeks C.M., Schouten M.G., Bomers J.G., et. al.: Three-Tesla magnetic resonance-guided prostate biopsy in men with increased prostate-specific antigen and repeated, negative, random, systematic, transrectal ultrasound biopsies: detection of clinically significant prostate cancers. Eur Urol 2012; 62: pp. 902-909.

  • 88. Sciarra A., Panebianco V., Cattarino S., et. al.: Multiparametric magnetic resonance imaging of the prostate can improve the predictive value of the urinary prostate cancer antigen 3 test in patients with elevated prostate-specific antigen levels and a previous negative biopsy. BJU Int 2012; 110: pp. 1661-1665.

  • 89. Sonn G.A., Chang E., Natarajan S., et. al.: Value of targeted prostate biopsy using magnetic resonance-ultrasound fusion in men with prior negative biopsy and elevated prostate-specific antigen. Eur Urol 2014; 65: pp. 809-815.

  • 90. Meng X., Rosenkrantz A.B., Mendhiratta N., et. al.: Relationship between prebiopsy multiparametric magnetic resonance imaging (MRI), biopsy indication, and MRI-ultrasound fusion-targeted prostate biopsy outcomes. Eur Urol 2016; 69: pp. 512-517.

  • 91. Numao N., Yoshida S., Komai Y., et. al.: Usefulness of pre-biopsy multiparametric magnetic resonance imaging and clinical variables to reduce initial prostate biopsy in men with suspected clinically localized prostate cancer. J Urol 2013; 190: pp. 502-508.

  • 92. Da Rosa M.R., Milot L., Sugar L., et. al.: A prospective comparison of MRI-US fused targeted biopsy versus systematic ultrasound-guided biopsy for detecting clinically significant prostate cancer in patients on active surveillance. J Magn Reson Imaging 2015; 41: pp. 220-225.

  • 93. Hu J.C., Chang E., Natarajan S., et. al.: Targeted prostate biopsy in select men for active surveillance: do the Epstein criteria still apply?. J Urol 2014; 192: pp. 385-390.

  • 94. Siddiqui M.M., Truong H., Rais-Bahrami S., et. al.: Clinical implications of a multiparametric magnetic resonance imaging based nomogram applied to prostate cancer active surveillance. J Urol 2015; 193: pp. 1943-1949.

  • 95. Stoyanova R., Pollack A., Lynne C.: Using radiogenomics to characterize MRI-guided prostate cancer biopsy heterogenity. J Clin Oncol 2015; 33: abstr 25

  • 96. VanderWeele D.J., McCann S., Fan X.: Radiogenomics of prostate cancer: association between quantitative multiparametric MRI features and PTEN. J Clin Oncol 2015; 33: abstr 126

  • 97. Zumsteg Z.S., Spratt D.E., Romesser P.B., et. al.: The natural history and predictors of outcome following biochemical relapse in the dose escalation era for prostate cancer patients undergoing definitive external beam radiotherapy. Eur Urol 2015; 67: pp. 1009-1016.

  • 98. Thompson I.M., Valicenti R.K., Albertsen P., et. al.: Adjuvant and salvage radiotherapy after prostatectomy: AUA/ASTRO guideline. J Urol 2013; 190: pp. 441-449.

  • 99. Mertan F.V., Lindenberg L., Choyke P.L., et. al.: PET imaging of recurrent and metastatic prostate cancer with novel tracers. Future Oncol 2016; 12: pp. 2463-2477.

  • 100. Rowe S.P., Gorin M.A., Allaf M.E., et. al.: PET imaging of prostate-specific membrane antigen in prostate cancer: current state of the art and future challenges. Prostate Cancer Prostatic Dis 2016; 19: pp. 223-230.

  • 101. Morigi J.J., Stricker P.D., van Leeuwen P.J., et. al.: Prospective comparison of 18F-fluoromethylcholine versus 68Ga-PSMA PET/CT in prostate cancer patients who have rising PSA after curative treatment and are being considered for targeted therapy. J Nucl Med 2015; 56: pp. 1185-1190.

  • 102. Eiber M., Maurer T., Souvatzoglou M., et. al.: Evaluation of hybrid (6)(8)Ga-PSMA ligand PET/CT in 248 patients with biochemical recurrence after radical prostatectomy. J Nucl Med 2015; 56: pp. 668-674.

  • 103. Maurer T., Eiber M., Schwaiger M., et. al.: Current use of PSMA-PET in prostate cancer management. Nat Rev Urol 2016; 13: pp. 226-235.

  • 104. Shinagare A.B., Vikram R., Jaffe C., et. al.: Radiogenomics of clear cell renal cell carcinoma: preliminary findings of The Cancer Genome Atlas-Renal Cell Carcinoma (TCGA-RCC) Imaging Research Group. Abdom Imaging 2015; 40: pp. 1684-1692.

  • 105. Karlo C.A., Di Paolo P.L., Chaim J., et. al.: Radiogenomics of clear cell renal cell carcinoma: associations between CT imaging features and mutations. Radiology 2014; 270: pp. 464-471.

  • 106. Hakimi A.A., Chen Y.B., Wren J., et. al.: Clinical and pathologic impact of select chromatin-modulating tumor suppressors in clear cell renal cell carcinoma. Eur Urol 2013; 63: pp. 848-854.

  • 107. Liu Y., Bai R., Sun H., et. al.: Diffusion-weighted magnetic resonance imaging of uterine cervical cancer. J Comput Assist Tomogr 2009; 33: pp. 858-862.

  • 108. Naganawa S., Sato C., Kumada H., et. al.: Apparent diffusion coefficient in cervical cancer of the uterus: comparison with the normal uterine cervix. Eur Radiol 2005; 15: pp. 71-78.

  • 109. Sala E., Micco M., Burger I.A., et. al.: Complementary prognostic value of pelvic magnetic resonance imaging and whole-body fluorodeoxyglucose positron emission tomography/computed tomography in the pretreatment assessment of patients with cervical cancer. Int J Gynecol Cancer 2015; 25: pp. 1461-1467.

  • 110. Micco M., Vargas H.A., Burger I.A., et. al.: Combined pre-treatment MRI and 18F-FDG PET/CT parameters as prognostic biomarkers in patients with cervical cancer. Eur J Radiol 2014; 83: pp. 1169-1176.

  • 111. Burger I.A., Goldman D.A., Vargas H.A., et. al.: Incorporation of postoperative CT data into clinical models to predict 5-year overall and recurrence free survival after primary cytoreductive surgery for advanced ovarian cancer. Gynecol Oncol 2015; 138: pp. 554-559.

  • 112. Caobelli F., Alongi P., Evangelista L., et. al.: Predictive value of (18)F-FDG PET/CT in restaging patients affected by ovarian carcinoma: a multicentre study. Eur J Nucl Med Mol Imaging 2016; 43: pp. 404-413.

  • 113. Vargas H.A., Micco M., Hong S.I., et. al.: Association between morphologic CT imaging traits and prognostically relevant gene signatures in women with high-grade serous ovarian cancer: a hypothesis-generating study. Radiology 2015; 274: pp. 742-751.

  • 114. Thrall J.H.: Moreton lecture: imaging in the age of precision medicine. J Am Coll Radiol 2015; 12: pp. 1106-1111.

  • 115. Hurd M.D., Martorell P., Delavande A., et. al.: Monetary costs of dementia in the United States. N Engl J Med 2013; 368: pp. 1326-1334.

  • 116. Bateman R.J., Xiong C., Benzinger T.L., et. al.: Clinical and biomarker changes in dominantly inherited Alzheimer’s disease. N Engl J Med 2012; 367: pp. 795-804.

  • 117. Bohnen N.I., Djang D.S., Herholz K., et. al.: Effectiveness and safety of 18F-FDG PET in the evaluation of dementia: a review of the recent literature. J Nucl Med 2012; 53: pp. 59-71.

  • 118. Dubois B., Feldman H.H., Jacova C., et. al.: Revising the definition of Alzheimer’s disease: a new lexicon. Lancet Neurol 2010; 9: pp. 1118-1127.

  • 119. Klunk W.E., Engler H., Nordberg A., et. al.: Imaging brain amyloid in Alzheimer’s disease with Pittsburgh Compound-B. Ann Neurol 2004; 55: pp. 306-319.

  • 120. Li J., Shi Y., Toga A.W.: Mapping brain anatomical connectivity using diffusion magnetic resonance imaging: structural connectivity of the human brain. IEEE Signal Process Mag 2016; 33: pp. 36-51.

  • 121. Brown J.A., Terashima K.H., Burggren A.C., et. al.: Brain network local interconnectivity loss in aging APOE-4 allele carriers. Proc Natl Acad Sci USA 2011; 108: pp. 20760-20765.

  • 122. Roussotte F.F., Daianu M., Jahanshad N., et. al.: Neuroimaging and genetic risk for Alzheimer’s disease and addiction-related degenerative brain disorders. Brain Imaging Behav 2014; 8: pp. 217-233.

  • 123. Jahanshad N., Rajagopalan P., Hua X., et. al.: Genome-wide scan of healthy human connectome discovers SPON1 gene variant influencing dementia severity. Proc Natl Acad Sci USA 2013; 110: pp. 4768-4773.

  • 124. Bralten J., Arias-Vasquez A., Makkinje R., et. al.: Association of the Alzheimer’s gene SORL1 with hippocampal volume in young, healthy adults. Am J Psychiatry 2011; 168: pp. 1083-1089.

  • 125. Potkin S.G., Guffanti G., Lakatos A., et. al.: Hippocampal atrophy as a quantitative trait in a genome-wide association study identifying novel susceptibility genes for Alzheimer’s disease. PLoS ONE 2009; 4: pp. e6501.

  • 126. Ebinger M., Winter B., Wendt M., et. al.: Effect of the use of ambulance-based thrombolysis on time to thrombolysis in acute ischemic stroke: a randomized clinical trial. JAMA 2014; 311: pp. 1622-1631.

  • 127. Kidwell C.S., Jahan R., Gornbein J., et. al.: A trial of imaging selection and endovascular treatment for ischemic stroke. N Engl J Med 2013; 368: pp. 914-923.

  • 128. Demchuk A.M., Dowlatshahi D., Rodriguez-Luna D., et. al.: Prediction of haematoma growth and outcome in patients with intracerebral haemorrhage using the CT-angiography spot sign (PREDICT): a prospective observational study. Lancet Neurol 2012; 11: pp. 307-314.

  • 129. Du F.Z., Jiang R., Gu M., et. al.: The accuracy of spot sign in predicting hematoma expansion after intracerebral hemorrhage: a systematic review and meta-analysis. PLoS ONE 2014; 9: pp. e115777.

  • 130. Ivanidze J., Kesavabhotla K., Kallas O.N., et. al.: Evaluating blood-brain barrier permeability in delayed cerebral infarction after aneurysmal subarachnoid hemorrhage. AJNR Am J Neuroradiol 2015; 36: pp. 850-854.

  • 131. Jones S., Anagnostou V., Lytle K., et. al.: Personalized genomic analyses for cancer mutation discovery and interpretation. Sci Transl Med 2015; 7: pp. 283ra53.

  • 132. Lipinski K.A., Barber L.J., Davies M.N., et. al.: Cancer evolution and the limits of predictability in precision cancer medicine. Trends Cancer 2016; 2: pp. 49-63.

  • 133. Hoefnagel L.D., van der Groep P., van de Vijver M.J., et. al.: Discordance in ERalpha, PR and HER2 receptor status across different distant breast cancer metastases within the same patient. Ann Oncol 2013; 24: pp. 3017-3023.

  • 134. Gerlinger M., Rowan A.J., Horswell S., et. al.: Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N Engl J Med 2012; 366: pp. 883-892.

  • 135. Shah S.P., Morin R.D., Khattra J., et. al.: Mutational evolution in a lobular breast tumour profiled at single nucleotide resolution. Nature 2009; 461: pp. 809-813.

  • 136. Woff E., Hendlisz A., Garcia C., et. al.: Monitoring metabolic response using FDG PET-CT during targeted therapy for metastatic colorectal cancer. Eur J Nucl Med Mol Imaging 2016; 43: pp. 1792-1801.

  • 137. Hricak H.: Oncologic imaging: a guiding hand of personalized cancer care. Radiology 2011; 259: pp. 633-640.

  • 138. Huyge V., Garcia C., Alexiou J., et. al.: Heterogeneity of metabolic response to systemic therapy in metastatic breast cancer patients. Clin Oncol 2010; 22: pp. 818-827.

  • 139. Doumou G., Siddique M., Tsoumpas C., et. al.: The precision of textural analysis in (18)F-FDG-PET scans of oesophageal cancer. Eur Radiol 2015; 25: pp. 2805-2812.

  • 140. Cook G.J., O’Brien M.E., Siddique M., et. al.: Non-small cell lung cancer treated with Erlotinib: heterogeneity of (18)F-FDG uptake at PET-association with treatment response and prognosis. Radiology 2015; 276: pp. 883-893.

  • 141. Cook G.J., Yip C., Siddique M., et. al.: Are pretreatment 18F-FDG PET tumor textural features in non-small cell lung cancer associated with response and survival after chemoradiotherapy?. J Nucl Med 2013; 54: pp. 19-26.

  • 142. Chicklore S., Goh V., Siddique M., et. al.: Quantifying tumour heterogeneity in 18F-FDG PET/CT imaging by texture analysis. Eur J Nucl Med Mol Imaging 2013; 40: pp. 133-140.

  • 143. Alyass A., Turcotte M., Meyre D.: From big data analysis to personalized medicine for all: challenges and opportunities. BMC Med Genomics 2015; 8: pp. 33.

  • 144. Van Neste L., Van Criekinge W.: We are all individuals… bioinformatics in the personalized medicine era. Cell Oncol 2015; 38: pp. 29-37.

  • 145. Abramson R.G., Burton K.R., Yu J.P., et. al.: Methods and challenges in quantitative imaging biomarker development. Acad Radiol 2015; 22: pp. 25-32.

  • 146. Rosenkrantz A.B., Mendiratta-Lala M., Bartholmai B.J., et. al.: Clinical utility of quantitative imaging. Acad Radiol 2015; 22: pp. 33-49.

This post is licensed under CC BY 4.0 by the author.