Home Methods and Challenges in Quantitative Imaging Biomarker Development
Post
Cancel

Methods and Challenges in Quantitative Imaging Biomarker Development

Academic radiology is poised to play an important role in the development and implementation of quantitative imaging (QI) tools. This article, drafted by the Association of University Radiologists Radiology Research Alliance Quantitative Imaging Task Force, reviews current issues in QI biomarker research. We discuss motivations for advancing QI, define key terms, present a framework for QI biomarker research, and outline challenges in QI biomarker development. We conclude by describing where QI research and development is currently taking place and discussing the paramount role of academic radiology in this rapidly evolving field.

Medical imaging has evolved dramatically since the first roentgenogram nearly 125 years ago . Modern techniques including ultrasound, computed tomography (CT), magnetic resonance imaging (MRI), and positron emission tomography (PET) now provide an unprecedented level of spatial detail and functional information . As medical imaging has progressed, older analog techniques have been steadily replaced with newer digital methods of image acquisition, processing, archiving, and display. This evolution has occurred in parallel with advancements in our understanding of the molecular underpinnings of disease and the rise of a more statistical and evidence-based approach to diagnosis and treatment. Medical imaging is now poised to leverage quantitative techniques in support of a wide range of clinical and research goals .

In a broad sense, quantitative imaging (QI) refers to the extraction and use of numerical/statistical features from medical images (see Box 1 for definitions of key terms). As a research field, QI includes the development, standardization, optimization, and application of anatomic, functional, and molecular imaging acquisition protocols, data analyses, display methods, and reporting structures, as well as the validation of QI results against relevant biological and clinical data . The QI concept is closely tied to that of a biomarker, defined as a characteristic that is objectively measured and evaluated as an indicator of a normal biological process, pathologic process, or response to a therapeutic intervention . A QI biomarker is therefore an objectively measured characteristic, derived from a medical image, that can be correlated with anatomically and physiologically relevant parameters including disease presence, disease severity, disease characterization (particularly at a molecular level), predicted disease course (both with and without treatment), and treatment response. The Quantitative Imaging Biomarkers Alliance (QIBA), organized by the Radiological Society of North America, has formally defined a QI biomarker as “an objective characteristic derived from an in vivo image measured on a ratio or interval scale as indicators [ sic ] of normal biological processes, pathogenic processes, or a response to a therapeutic intervention.” The emphasis of this definition on a ratio or interval variables would imply that tumor volumes or PET standardized uptake values would be considered QI biomarkers, because the difference or ratio between two values is meaningful, whereas ordinal variables such as Breast Imaging Reporting and Data System assessment categories would not. This strict definition is meant to guide QI research toward biomarkers that may be assessed and compared using robust statistical calculations including frequency distributions, medians, means, standard deviations, and standard errors of the mean .

Box 1:

Definitions related to quantitative imaging biomarker development

Analytical validation —Demonstration of the accuracy, precision, and feasibility of biomarker measurement

Biomarker —A characteristic that is objectively measured and evaluated as an indicator of a normal biological process, a pathogenic process, or a response to a therapeutic intervention

Predictive biomarker —A biomarker intended to forecast disease course in the presence of a specific treatment

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<

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<

Motivations for QI biomarker development

Get Radiology Tree app to read full this article<

Get Radiology Tree app to read full this article<

Figure 1, DCE-MRI as a QI technique for assessing breast cancer response to neoadjuvant therapy (color overlay = tumor). The top row illustrates an early reduction in the quantitative DCE-MRI parameter K trans in a patient who had a documented pCR at surgery (a) before therapy, (b) after one cycle of neoadjuvant therapy, and (c) at the conclusion of neoadjuvant therapy. The bottom row illustrates an early increase in K trans in a patient who had residual disease (non-pCR) at surgery (d) before therapy, (e) after one cycle of neoadjuvant therapy, and (f) at the conclusion of neoadjuvant therapy. ( Image courtesy of Lisa Li, PhD, Vanderbilt University .) DCE-MRI, dynamic contrast-enhanced magnetic resonance imaging; pCR, pathologic complete response; QI, quantitative imaging.

Get Radiology Tree app to read full this article<

Get Radiology Tree app to read full this article<

Challenges in QI biomarker development

Get Radiology Tree app to read full this article<

Analytical validation

Get Radiology Tree app to read full this article<

Figure 2, Repeatability of ADC measurements from DW-MRI in a breast cancer patient (color overlay = tumor). (a) The distributions of the ADC values from the tumor obtained on two separate scans within 1 week of each other. (b) Visit 1 and (c) visit 2 show the spatial variations at the voxel level. The mean with 95% confidence intervals for the two visits were 1.06 ± 0.01 and 1.03 ± 0.01 mm 2ms, respectively. The lack of overlap in the confidence intervals, despite the apparent similarity in the histograms, illustrates the importance of analytical validation studies to establish ranges of measurement error before deploying quantitative techniques to interrogate changes in underlying biology. ( Image courtesy of Lori Arlinghaus, PhD, Vanderbilt University. ) ADC, apparent diffusion coefficient; DW-MRI, diffusion-weighted magnetic resonance imaging.

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<

Qualification

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<

Utilization

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<

Figure 3, Volume-rendered computed tomography of the abdomen and pelvis with overlaid three-dimensional surface rendering of the spleen, segmented by a fully automated multiatlas content labeling algorithm. This technology is under investigation as a means of efficiently and accurately extracting spleen volume data for biomarker analyses.

Get Radiology Tree app to read full this article<

Where QI research is taking place

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<

The role of academic radiology in QI biomarker research

Get Radiology Tree app to read full this article<

Get Radiology Tree app to read full this article<

Conclusions

Get Radiology Tree app to read full this article<

Acknowledgments

Get Radiology Tree app to read full this article<

Get Radiology Tree app to read full this article<

References

  • 1. DiSantis D.J.: Early American Radiology: the pioneer years. AJR Am J Roentgenol 1986; 147: pp. 850-853.

  • 2. Buckler A.J., Bresolin L., Dunnick N.R., et. al., Group: A collaborative enterprise for multi-stakeholder participation in the advancement of quantitative imaging. Radiology 2011; 258: pp. 906-914.

  • 3. Buckler A.J., Paik D., Ouellette M., et. al.: A novel knowledge representation framework for the statistical validation of quantitative imaging biomarkers. J Digit Imaging 2013; 26: pp. 614-629.

  • 4. Jaffer F.A., Weissleder R.: Molecular imaging in the clinical arena. JAMA 2005; 293: pp. 855-862.

  • 5. Buckler A.J., Bresolin L., Dunnick N.R., et. al.: Quantitative imaging test approval and biomarker qualification: interrelated but distinct activities. Radiology 2011; 259: pp. 875-884.

  • 6. Meyer C.R., Armato S.G., Fenimore C.P., et. al.: Quantitative imaging to assess tumor response to therapy: common themes of measurement, truth data, and error sources. Transl Oncol 2009; 2: pp. 198-210.

  • 7. Biomarkers Definitions Working Group: Biomarkers and surrogate endpoints: preferred definitions and conceptual framework. Clin Pharmacol Ther 2001; 69: pp. 89-95.

  • 8. Kessler L.G., Barnhart H.X., Buckler A.J., et. al.: The emerging science of quantitative imaging biomarkers terminology and definitions for scientific studies and regulatory submissions. Stat Methods Med Res 2014;

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

  • 10. Arksey H., O’Malley L.: Scoping studies: towards a methodological framework. Int J Soc Res Methodol 2005; 8: pp. 19-32.

  • 11. Sullivan D.C.: Imaging as a quantitative science. Radiology 2008; 248: pp. 328-332.

  • 12. Boone J.M.: Radiological interpretation 2020: toward quantitative image assessment. Med Phys 2007; 34: pp. 4173-4179.

  • 13. Choong M.K., Tsafnat G.: The implications of biomarker evidence for systematic reviews. BMC Med Res Methodol 2012; 12: pp. 176.

  • 14. Eisenhauer E.A., Therasse P., Bogaerts J., et. al.: New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer 2009; 45: pp. 228-247.

  • 15. Vidaurre T., Wilkerson J., Simon R., et. al.: Stable disease is not preferentially observed with targeted therapies and as currently defined has limited value in drug development. Cancer J 2009; 15: pp. 366-373.

  • 16. Buyse M., Thirion P., Carlson R.W., et. al.: Relation between tumour response to first-line chemotherapy and survival in advanced colorectal cancer: a meta-analysis. Meta-Analysis Group in Cancer. Lancet 2000; 356: pp. 373-378.

  • 17. Slamon D.J., Leyland-Jones B., Shak S., et. al.: Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N Engl J Med 2001; 344: pp. 783-792.

  • 18. Hurwitz H., Fehrenbacher L., Novotny W., et. al.: Bevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancer. N Engl J Med 2004; 350: pp. 2335-2342.

  • 19. Johnson D.H., Fehrenbacher L., Novotny W.F., et. al.: Randomized phase II trial comparing bevacizumab plus carboplatin and paclitaxel with carboplatin and paclitaxel alone in previously untreated locally advanced or metastatic non-small-cell lung cancer. J Clin Oncol 2004; 22: pp. 2184-2191.

  • 20. Jubb A.M., Oates A.J., Holden S., et. al.: Predicting benefit from anti-angiogenic agents in malignancy. Nat Rev Cancer 2006; 6: pp. 626-635.

  • 21. Workman P., Aboagye E.O., Chung Y.L., et. al.: Minimally invasive pharmacokinetic and pharmacodynamic technologies in hypothesis-testing clinical trials of innovative therapies. J Natl Cancer Inst 2006; 98: pp. 580-598.

  • 22. Desar I.M., van Herpen C.M., van Laarhoven H.W., et. al.: Beyond RECIST: molecular and functional imaging techniques for evaluation of response to targeted therapy. Cancer Treat Rev 2009; 35: pp. 309-321.

  • 23. Abramson R.G., Yankeelov T.E.: Imaging biomarkers and surrogate endpoints in oncology clinical trials.Luna A.Vilanova J.C.Hygino Da Cruz L.C. et. al.Functional imaging in oncology.2014.SpringerHeidelberg:

  • 24. O’Connor J.P., Jackson A., Asselin M.C., et. al.: Quantitative imaging biomarkers in the clinical development of targeted therapeutics: current and future perspectives. Lancet Oncol 2008; 9: pp. 766-776.

  • 25. Garcia-Donas J., Rodriguez-Antona C., Jonasch E.: Molecular markers to predict response to therapy. Semin Oncol 2013; 40: pp. 444-458.

  • 26. Bhooshan N., Giger M.L., Jansen S.A., et. al.: Cancerous breast lesions on dynamic contrast-enhanced MR images: computerized characterization for image-based prognostic markers. Radiology 2010; 254: pp. 680-690.

  • 27. Fuleihan G.E., Testa M.A., Angell J.E., et. al.: Reproducibility of DXA absorptiometry: a model for bone loss estimates. J Bone Miner Res 1995; 10: pp. 1004-1014.

  • 28. Hua X., Lee S., Yanovsky I., et. al.: Optimizing power to track brain degeneration in Alzheimer’s disease and mild cognitive impairment with tensor-based morphometry: an ADNI study of 515 subjects. Neuroimage 2009; 48: pp. 668-681.

  • 29. van Klaveren R.J., Oudkerk M., Prokop M., et. al.: Management of lung nodules detected by volume CT scanning. N Engl J Med 2009; 361: pp. 2221-2229.

  • 30. Wahl R.L., Jacene H., Kasamon Y., et. al.: From RECIST to PERCIST: evolving considerations for PET response criteria in solid tumors. J Nucl Med 2009; 50: pp. 122S-150S.

  • 31. Wintermark M., Albers G.W., Alexandrov A.V., et. al.: Acute stroke imaging research roadmap. AJNR Am J Neuroradiol 2008; 29: pp. e23-e30.

  • 32. Merckel L.G., Verkooijen H.M., Peters N.H., et. al.: The added diagnostic value of dynamic contrast-enhanced MRI at 3.0 T in nonpalpable breast lesions. PLoS One 2014; 9: pp. e94233.

  • 33. Wang J., Liu H., Sun J., et. al.: Varying correlation between 18F-fluorodeoxyglucose positron emission tomography and dynamic contrast-enhanced MRI in carotid atherosclerosis: implications for plaque inflammation. Stroke 2014; 45: pp. 1842-1845.

  • 34. Kickingereder P., Sahm F., Wiestler B., et. al.: Evaluation of microvascular permeability with dynamic contrast-enhanced MRI for the differentiation of primary CNS lymphoma and glioblastoma: radiologic-pathologic correlation. AJNR Am J Neuroradiol 2014; 35: pp. 1503-1508.

  • 35. Lim I., Noh W.C., Park J., et. al.: The combination of FDG PET and dynamic contrast-enhanced MRI improves the prediction of disease-free survival in patients with advanced breast cancer after the first cycle of neoadjuvant chemotherapy. Eur J Nucl Med Mol Imaging 2014; 41: pp. 1852-1860.

  • 36. Kurland B.F., Gerstner E.R., Mountz J.M., et. al.: Promise and pitfalls of quantitative imaging in oncology clinical trials. Magn Reson Imaging 2012; 30: pp. 1301-1312.

  • 37. Institute of Medicine: Evaluation of biomarkers and surrogate endpoints in chronic disease.2010.National Academies PressWashington, DC

  • 38. Obuchowski N.A., Reeves A.P., Huang E.P., et. al.: Quantitative imaging biomarkers: a review of statistical methods for computer algorithm comparisons. Stat Methods Med Res 2014;

  • 39. Raunig D.L., McShane L.M., Pennello G., et. al.: Quantitative imaging biomarkers: a review of statistical methods for technical performance assessment. Stat Methods Med Res 2014;

  • 40. Padhani A.R., Hayes C., Landau S., et. al.: Reproducibility of quantitative dynamic MRI of normal human tissues. NMR Biomed 2002; 15: pp. 143-153.

  • 41. Yankeelov T.E., DeBusk L.M., Billheimer D.D., et. al.: Repeatability of a reference region model for analysis of murine DCE-MRI data at 7T. J Magn Reson Imaging 2006; 24: pp. 1140-1147.

  • 42. Barnes S.L., Whisenant J.G., Loveless M.E., et. al.: Assessing the reproducibility of dynamic contrast enhanced magnetic resonance imaging in a murine model of breast cancer. Magn Reson Med 2013; 69: pp. 1721-1734.

  • 43. Dula A.N., Arlinghaus L.R., Dortch R.D., et. al.: Amide proton transfer imaging of the breast at 3 T: establishing reproducibility and possible feasibility assessing chemotherapy response. Magn Reson Med 2013; 70: pp. 216-224.

  • 44. Alonzi R., Taylor N.J., Stirling J.J., et. al.: Reproducibility and correlation between quantitative and semiquantitative dynamic and intrinsic susceptibility-weighted MRI parameters in the benign and malignant human prostate. J Magn Reson Imaging 2010; 32: pp. 155-164.

  • 45. Whisenant J.G., Peterson T.E., Fluckiger J.U., et. al.: Reproducibility of static and dynamic (18)F-FDG, (18)F-FLT, and (18)F-FMISO microPET studies in a murine model of HER2+ breast cancer. Mol Imaging Biol 2013; 15: pp. 87-96.

  • 46. Tseng J.R., Dandekar M., Subbarayan M., et. al.: Reproducibility of 3′-deoxy-3′-(18)F-fluorothymidine microPET studies in tumor xenografts in mice. J Nucl Med 2005; 46: pp. 1851-1857.

  • 47. Dandekar M., Tseng J.R., Gambhir S.S.: Reproducibility of 18F-FDG microPET studies in mouse tumor xenografts. J Nucl Med 2007; 48: pp. 602-607.

  • 48. Sim J., Wright C.C.: The kappa statistic in reliability studies: use, interpretation, and sample size requirements. Phys Ther 2005; 85: pp. 257-268.

  • 49. Beaton D.E., Katz J.N., Fossel A.H., et. al.: Measuring the whole or the parts? Validity, reliability, and responsiveness of the disabilities of the arm, shoulder and hand outcome measure in different regions of the upper extremity. J Hand Ther 2001; 14: pp. 128-146.

  • 50. Bruton A., Conway J.H., Holgate S.T.: Reliability: what is it, and how is it measured?. Physiotherapy 2000; 86: pp. 94-99.

  • 51. Katz R.: Biomarkers and surrogate markers: an FDA perspective. NeuroRx 2004; 1: pp. 189-195.

  • 52. Bruns A., Kunnecke B., Risterucci C., et. al.: Validation of cerebral blood perfusion imaging as a modality for quantitative pharmacological MRI in rats. Magn Reson Med 2009; 61: pp. 1451-1458.

  • 53. Yoo A.J., Sheth K.N., Kimberly W.T., et. al.: Validating imaging biomarkers of cerebral edema in patients with severe ischemic stroke. J Stroke Cerebrovasc Dis 2013; 22: pp. 742-749.

  • 54. Wintermark M., Albers G.W., Broderick J.P., et. al.: Acute stroke imaging research roadmap II. Stroke 2013; 44: pp. 2628-2639.

  • 55. Huynh T.J., Flaherty M.L., Gladstone D.J., et. al.: Multicenter accuracy and interobserver agreement of spot sign identification in acute intracerebral hemorrhage. Stroke 2014; 45: pp. 107-112.

  • 56. Buyse M., Michiels S., Sargent D.J., et. al.: Integrating biomarkers in clinical trials. Expert Rev Mol Diagn 2011; 11: pp. 171-182.

  • 57. Puntmann V.O.: How-to guide on biomarkers: biomarker definitions, validation and applications with examples from cardiovascular disease. Postgrad Med J 2009; 85: pp. 538-545.

  • 58. Mandrekar S.J., Sargent D.J.: Clinical trial designs for predictive biomarker validation: theoretical considerations and practical challenges. J Clin Oncol 2009; 27: pp. 4027-4034.

  • 59. Buyse M., Sargent D.J., Grothey A., et. al.: Biomarkers and surrogate end points—the challenge of statistical validation. Nat Rev Clin Oncol 2010; 7: pp. 309-317.

  • 60. Sargent D.J., Rubinstein L., Schwartz L., et. al.: Validation of novel imaging methodologies for use as cancer clinical trial end-points. Eur J Cancer 2009; 45: pp. 290-299.

  • 61. Sargent D.J., Hayes D.F.: Assessing the measure of a new drug: is survival the only thing that matters?. J Clin Oncol 2008; 26: pp. 1922-1923.

  • 62. Hoering A., Leblanc M., Crowley J.J.: Randomized phase III clinical trial designs for targeted agents. Clin Cancer Res 2008; 14: pp. 4358-4367.

  • 63. Freidlin B., McShane L.M., Korn E.L.: Randomized clinical trials with biomarkers: design issues. J Natl Cancer Inst 2010; 102: pp. 152-160.

  • 64. Villaruz L.C., Socinski M.A.: The clinical viewpoint: definitions, limitations of RECIST, practical considerations of measurement. Clin Cancer Res 2013; 19: pp. 2629-2636.

  • 65. Green E., Yothers G., Sargent D.J.: Surrogate endpoint validation: statistical elegance versus clinical relevance. Stat Methods Med Res 2008; 17: pp. 477-486.

  • 66. Smith J.J., Sorensen A.G., Thrall J.H.: Biomarkers in imaging: realizing radiology’s future. Radiology 2003; 227: pp. 633-638.

  • 67. Jha S.: The allure of quantification. Magn Reson Imaging 2013; 31: pp. 1035-1036.

  • 68. Mackenzie R., Dixon A.K.: Measuring the effects of imaging: an evaluative framework. Clin Radiol 1995; 50: pp. 513-518.

  • 69. Abramson R.G., Su P.F., Shyr Y.: Quantitative metrics in clinical radiology reporting: a snapshot perspective from a single mixed academic-community practice. Magn Reson Imaging 2012; 30: pp. 1357-1366.

  • 70. Garcia-Lorenzo D., Francis S., Narayanan S., et. al.: Review of automatic segmentation methods of multiple sclerosis white matter lesions on conventional magnetic resonance imaging. Med Image Anal 2013; 17: pp. 1-18.

  • 71. Pomar-Nadal A., Perez-Castillo C., Alberich-Bayarri A., et. al.: Integrating information about imaging biomarkers into structured radiology reports. Radiologia 2013; 55: pp. 188-194.

  • 72. Travis A.R., Sevenster M., Ganesh R., et. al.: Preferences for structured reporting of measurement data: an institutional survey of medical oncologists, oncology registrars, and radiologists. Acad Radiol 2014; 21: pp. 785-796.

  • 73. Abajian A.C., Levy M., Rubin D.L.: Informatics in radiology: improving clinical work flow through an AIM database: a sample web-based lesion tracking application. Radiographics 2012; 32: pp. 1543-1552.

  • 74. Bresnahan B.W.: Economic evaluation in radiology: reviewing the literature and examples in oncology. Acad Radiol 2010; 17: pp. 1090-1095.

  • 75. Rubin D.L., Willrett D., O’Connor M.J., et. al.: Automated tracking of quantitative assessments of tumor burden in clinical trials. Transl Oncol 2014; 7: pp. 23-35.

  • 76. Enzmann D.R.: Radiology’s value chain. Radiology 2012; 263: pp. 243-252.

  • 77. Institute of Medicine: Developing biomarker-based tools for cancer screening, diagnosis and treatment: workshop summary.2007.National Academies PressWashington, DC

  • 78. Clarke L.P., Nordstrom R.J., Zhang H., et. al.: The Quantitative Imaging Network: NCI’s historical perspective and planned goals. Transl Oncol 2014; 7: pp. 1-4.

  • 79. Radiological Society of North America. QIBA protocols and profiles. Available at: https://www.rsna.org/QIBA_Protocols_and_Profiles.aspx . Accessed May 20, 2014.

  • 80. Chan E, Arlinghaus LR, Cardin DB, et al. Phase I trial of chemoradiation with capecitabine and vorinostat in pancreatic cancer (scientific abstract). Gastrointestinal cancers symposium, San Francisco, CA, 2013.

  • 81. Stadler WM. Clinical needs/values of imaging biomarkers (oral presentation). Chicago, IL: Radiological Society of North America (RSNA), 2010.

  • 82. Doot R.K., Thompson T., Greer B.E., et. al.: Early experiences in establishing a regional quantitative imaging network for PET/CT clinical trials. Magn Reson Imaging 2012; 30: pp. 1291-1300.

  • 83. Krishnaraj A., Weinreb J.C., Ellenbogen P.H., et. al.: The Future of imaging biomarkers in radiologic practice: Proceedings of the thirteenth annual ACR forum. J Am Coll Radiol 2014; 11: pp. 20-23.

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