Feeds:
Posts
Comments

Posts Tagged ‘imaging-bio-markers’

Imaging-biomarkers is Imaging-based tissue characterization

Author – Writer: Dror Nir, PhD

For everyone who is skeptical about the future role of imaging-based tissue chracterisation in the management of cancer, the following “Statement paper” ESR statement on the stepwise development of imaging biomarkers published online: 9 February 2013, by the European Society of Radiology (ESR), should provide substantial reassurance that this kind of technology will become a must! In support of this claim I quote the following information:

The European Society of Radiology and its related European Institute for Biomedical Imaging Research (EIBIR) should have a relevant role in coordinating future developments of biomarkers and in the assessment and validation of imaging biomarkers as surrogate end points.

Acknowledgements

This paper was kindly prepared by the ESR Subcommittee on Imaging Biomarkers (Chairperson: Bernard Van Beers. Research Committee Chairperson: Luis Martí-Bonmatí. Members: Marco Essig, Thomas Helbich, Celso Matos, Wiro Niessen, Anwar Padhani, Harriet C. Thoeny, Siegfried Trattnig, Jean-Paul Vallée. Co-opted members: Peter Brader, Nicolas Grenier) on behalf of the European Society of Radiology (ESR) and with the help of Sabrina Doblas, INSERM U773, Paris, France.

It was approved by the ESR Executive Council in December 2012..

According to ESR: “There is increasing interest in developing the quantitative imaging of biomarkers in personalised medicine”. In this perspective, “Biomarkers” are tissue properties that can be quantitatively and reproducibly measured by imaging devices. One example for a major unmet need, which I found to be most interesting is the imaging-based detection of tumor invasiveness.

Quoting from the paper: ” Biomarkers are defined as “characteristics that are objectively measured and evaluated as indicators of normal biological processes, pathological processes, or pharmaceutical responses to a therapeutic intervention” [1]. Broadly, biomarkers fall into two categories: bio-specimen biomarkers, including molecular biomarkers and genetic biomarkers, and bio-signal biomarkers or imaging biomarkers. Bio-specimen biomarkers are obtained by removing a sample from a patient. Examples of these molecular biomarkers are genes and proteins detected from fluids or tissue samples. Bio-signal biomarkers remove no material from the patient, but rather detect and analyse an electromagnetic, photonic or acoustic signal emitted by the patient [2]. These imaging biomarkers have the advantage of being non-invasive, spatially resolved and repeatable [3]. They are of particular interest if they can overcome the limitations of the established histological “gold standards”. Indeed, invasive reference examinations, such as biopsy, can be inconclusive, are non-representative of the whole tissue (which is a tremendous limitation when assessing malignant tumours, which are known to be heterogeneous) and possess non-negligible levels of mortality and morbidity.

Genetic biomarkers indicate whether a disease may occur, but they are usually inefficient to assess the presence and stage of a disease. Similar to molecular biomarkers, imaging biomarkers can be used for early detection of diseases, staging and grading, and predicting or assessing the response to treatment [3]. Accordingly, because of their relative lower cost compared with imaging, molecular biomarkers may be more appropriate for disease screening and early detection than imaging biomarkers. With their high sensitivity, molecular biomarkers could also detect subclinical stages of disease before any morphological or functional change is detectable on imaging. In contrast, imaging biomarkers are often more useful than molecular biomarkers for disease staging, and also grading and for assessing tumour response, because localised information is crucial.

The main messages ESR wishes to deliver in this paper are that:

• Using imaging-biomarkers to streamline drug discovery and disease progression will drive a huge advancement in healthcare.

• The clinical qualification and validation of imaging biomarkers technology pose challenges, mainly in establishing the accuracy and reproducibility of such techniques. In that respect, agreements on standards and evaluation methods (e.g. clinical studies design) is imperative.

• There should be high motivation to pursue the development of imaging-biomarkers as the “clinical value of new biomarkers is of the highest priority in terms of patient management, assessing risk factors and disease prognosis.”

The paper deals to a great extent with the requirements on accuracy, reproducibility, standardization and quality control from the process of developing imaging-biomarkers:

Accuracy: Before being routinely used in the clinic, imaging biomarkers must be validated. Determining the accuracy implies calculating the sensitivity and specificity of the biomarker when compared with a biological process, such as tumour necrosis, which can be assessed at histopathological examination… [69]  [10, 11]

Reproducibility: Repeatability (measurements at short intervals on the same subjects using the same equipment in the same centres) and reproducibility (measurements at short intervals on the same subjects using different facilities in the same and different centres) studies must be conducted for image acquisition and image analysis…. Reproducibility studies are now very often included in scientific papers, as advised by the “standards for reporting of diagnostic accuracy” (STARD) criteria and should ideally include Bland-Altman plots and results of coefficients of repeatability [1617].

Standardisation: Standardisation relates to the establishment of norms or requirements about technical aspects. In the development of imaging biomarkers, two main aspects should be considered: Standardisation of image acquisition and Standardisation of image analysis…  [18] [1921]  [22] [27, 28] [3133]

Quality control: Adequate phantoms could be used to validate, on a day-to-day basis, that the biomarker stays robust and to avoid any drift in the machine, acquisition or processing protocol….  [34] [3035] [36] [37] [23].

The proposed development workflow:

“Similar to new drugs, the development of biomarkers has to pass along a pipeline going from discovery, through verification in different laboratories, validation and qualification before they can be used in clinical routine. Validation includes the determination of the accuracy and the precision (reproducibility) of the biomarker and standardisation concerns both acquisition and analysis. Qualification, defined as a “graded, fit-for-purpose evidentiary process linking a biomarker with biological processes and clinical end-points”, is a validation process in large cohorts of patients involving multiple centres, similar to phase III clinical trials, to obtain regulatory approval as surrogate endpoints [4]. A more extensive path to biomarker development has been reported [5]. The first step is the proof of concept, which defines any specific change relevant to the disease that can be studied using the available imaging and computational techniques. The relationship between this change and the presence, grading and response to treatment of the disease constitutes the proof of mechanism. The images needed to extract the biomarker must be appropriate (in terms of resolution, signal and contrast behaviour). Preparation of images relates to improving the data before the analysis (such as segmentation, filtering, interpolation or registration). The analysis and modelling of the signal by computational numerical adjustment of a mathematical model allow extracting the needed information (such as structural, physical, chemical, biological and functional properties). After this voxel-by-voxel computation, the spatial distribution of the biomarker can be depicted by parametric images, defined as derived secondary images which pixels represent the distribution values of a given parameter. Multivariate parametric images obtained by statistical modelling of the relevant parameters allow the reduction of data and a clear definition of the defined disease target. The abnormal values should be defined and measured through histogram analysis. A pilot test on a small sample of subjects, with and without the disease, has to be performed to validate the process—also called proof of principle—and to evaluate the influence of potential variations related to age, sex or any other source of biases. Finally, proofs of efficacy and effectiveness on larger and well-defined series of patients will show the ability of a biomarker to measure the clinical endpoint (Fig. 1).

Steps for the development of imaging biomarkers (adapted from [5])

Steps for the development of imaging biomarkers (adapted from [5])

The authors admit that the requirement posed on development of imaging-biomarkers represents a huge challenge and they try to offer ideas, mainly taken from the “MRI experience” to overcome certain hurdles. There is one important point on which they do not discuss: the definition of appropriate reference test. It is my own experience, based on many study protocols I developed in the past decade, that without reaching an agreement on that point, the development of imaging-biomarkers will just move in circles. Note, that today’s most “acceptable” reference test is histopathology, which everyone admits (as well mentioned in this paper); suffers many limitations. When it comes to validating imaging-biomarkers, the need to accurately match imaging products with histopathology is an additional major hurdle.

This is why, I see as a necessary step, to develop “real-time” imaging based tissue characterization combined with in-situ imaging-based histology.

 

References

1.

Biomarkers Definitions Working Group (2001) Biomarkers and surrogate endpoints: preferred definitions and conceptual framework. Clin Pharmacol Ther 69(3):89–95CrossRef

2.

Waterton JC, Pylkkanen L (2012) Qualification of imaging biomarkers for oncology drug development. Eur J Cancer 48(4):409–415PubMedCrossRef

3.

European Society of Radiology (2010) White paper on imaging biomarkers. Insights Imaging 1(2):42–45CrossRef

4.

Wagner JA, Williams SA, Webster CJ (2007) Biomarkers and surrogate end points for fit-for-purpose development and regulatory evaluation of new drugs. Clin Pharmacol Ther 81(1):104–107PubMedCrossRef

5.

Marti Bonmati L, Alberich-Bayarri A, Garcia-Marti G, Sanz Requena R, Pérez Castillo C, Carot Sierra JM, Herrera M (2012) Imaging biomarkers, quantitative imaging, and bioengineering. Radiol 54(3):269–278CrossRef

6.

Lewin M, Poujol-Robert A, Boelle PY et al (2007) Diffusion-weighted magnetic resonance imaging for the assessment of fibrosis in chronic hepatitis C. Hepatology 46(3):658–665PubMedCrossRef

7.

Luciani A, Vignaud A, Cavet M et al (2008) Liver cirrhosis: intravoxel incoherent motion MR imaging–pilot study. Radiology 249(3):891–899PubMedCrossRef

8.

Bonekamp S, Torbenson MS, Kamel IR (2011) Diffusion-weighted magnetic resonance imaging for the staging of liver fibrosis. J Clin Gastroenterol 45(10):885–892PubMedCrossRef

9.

Leitao HS, Doblas S, d’Assignies G, Garteiser P, Daire JL, Paradis V, Geraldes CF, Vilgrain V, Van Beers BE (2012) Fat deposition decreases diffusion parameters at MRI: a study in phantoms and patients with liver steatosis. Eur Radiol 23(2):461-467

10.

Le Bihan D, Urayama S, Aso T, Hanakawa T, Fukuyama H (2006) Direct and fast detection of neuronal activation in the human brain with diffusion MRI. PNAS 103(21):8263–8268PubMedCrossRef

11.

Xu J, Does MD, Gore JC (2011) Dependence of temporal diffusion spectra on microstructural properties of biological tissues. Magn Reson Imaging 29(3):380–390PubMedCrossRef

12.

Sinkus R, Van Beers BE, Vilgrain V, DeSouza N, Waterton JC (2012) Apparent diffusion coefficient from magnetic resonance imaging as a biomarker in oncology drug development. Eur J Cancer 48(4):425–431PubMedCrossRef

13.

Yablonskiy DA, Sukstanskii AL (2010) Theoretical models of the diffusion weighted MR signal. NMR Biomed 23(7):661–681PubMedCrossRef

14.

Eisenhauer EA, Therasse P, Bogaerts J et al (2009) New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer 45(2):228–247PubMedCrossRef

15.

Padhani AR, Khan AA (2010) Diffusion-weighted (DW) and dynamic contrast-enhanced (DCE) magnetic resonance imaging (MRI) for monitoring anticancer therapy. Target Oncol 5(1):39–52PubMedCrossRef

16.

Bossuyt PM, Reitsma JB, Bruns DE et al (2003) Towards complete and accurate reporting of studies of diagnostic accuracy: the STARD initiative. Radiology 226(1):24–28PubMedCrossRef

17.

Barnhart HX, Barboriak DP (2009) Applications of the repeatability of quantitative imaging biomarkers: a review of statistical analysis of repeat data sets. Transl Oncol 2(4):231–235PubMed

18.

Padhani AR, Liu G, Koh DM et al (2009) Diffusion-weighted magnetic resonance imaging as a cancer biomarker: consensus and recommendations. Neoplasia 11(2):102–125PubMed

19.

Taouli B, Koh DM (2010) Diffusion-weighted MR imaging of the liver. Radiology 254(1):47–66PubMedCrossRef

20.

Kwee TC, Takahara T, Koh DM, Nievelstein RA, Luijten PR (2008) Comparison and reproducibility of ADC measurements in breathhold, respiratory triggered, and free-breathing diffusion-weighted MR imaging of the liver. J Magn Reson Imaging 28(5):1141–1148PubMedCrossRef

21.

Ivancevic MK, Kwee TC, Takahara T et al (2009) Diffusion-weighted MR imaging of the liver at 3.0 Tesla using tracking only navigator echo (TRON): a feasibility study. J Magn Reson Imaging 30(5):1027–1033PubMedCrossRef

22.

Zussman B, Jabbour P, Talekar K, Gorniak R, Flanders AE (2011) Sources of variability in computed tomography perfusion: implications for acute stroke management. Neurosurg Focus 30(6):E8PubMedCrossRef

23.

Rajaraman S, Rodriguez JJ, Graff C et al (2011) Automated registration of sequential breath-hold dynamic contrast-enhanced MR images: a comparison of three techniques. Magn Reson Imaging 29(5):668–682PubMedCrossRef

24.

Wagner M, Doblas S, Daire JL, Paradis V, Haddad N, Leitao H, Garteiser P, Vilgrain V, Sinkus R, Van Beers BE (2012) Diffusion-weighted MR imaging for the regional characterization of liver tumors. Radiology 264(2):464–472PubMedCrossRef

25.

Moffat BA, Chenevert TL, Lawrence TS et al (2005) Functional diffusion map: a noninvasive MRI biomarker for early stratification of clinical brain tumor response. PNAS 102(15):5524–5529PubMedCrossRef

26.

Yang X, Knopp MV (2011) Quantifying tumor vascular heterogeneity with dynamic contrast-enhanced magnetic resonance imaging: a review. J Biomed Biotechnol 732848:1–12

27.

Buckley DL (2002) Uncertainty in the analysis of tracer kinetics using dynamic contrast-enhanced T1-weighted MRI. Magn Reson Med 47(3):601–606PubMedCrossRef

28.

Michoux N, Huwart L, Abarca-Quinones J et al (2008) Transvascular and interstitial transport in rat hepatocellular carcinomas: dynamic contrast-enhanced MRI assessment with low- and high-molecular weight agents. J Magn Reson Imaging 28(4):906–914PubMedCrossRef

29.

Leach MO, Brindle KM, Evelhoch JL et al (2005) The assessment of antiangiogenic and antivascular therapies in early-stage clinical trials using magnetic resonance imaging: issues and recommendations. Br J Cancer 92(9):1599–1610PubMedCrossRef

30.

Buckler AJ, Schwartz LH, Petrick N et al (2010) Data sets for the qualification of volumetric CT as a quantitative imaging biomarker in lung cancer. Opt Express 18(14):15267–15282PubMedCrossRef

31.

Huwart L, Sempoux C, Vicaut E et al (2008) Magnetic resonance elastography for the noninvasive staging of liver fibrosis. Gastroenterology 135(1):32–40PubMedCrossRef

32.

Friedrich-Rust M, Nierhoff J, Lupsor M et al (2012) Performance of Acoustic Radiation Force Impulse imaging for the staging of liver fibrosis: a pooled meta-analysis. J Viral Hepat 19(2):e212–e219PubMedCrossRef

33.

Degos F, Perez P, Roche B et al (2010) Diagnostic accuracy of FibroScan and comparison to liver fibrosis biomarkers in chronic viral hepatitis: a multicenter prospective study (the FIBROSTIC study). J Hepatol 53(6):1013–1021PubMedCrossRef

34.

Chenevert TL, Galban CJ, Ivancevic MK et al (2011) Diffusion coefficient measurement using a temperature-controlled fluid for quality control in multicenter studies. J Magn Reson Imaging 34(4):983–987PubMedCrossRef

35.

Lee YC, Fullerton GD, Baiu C, Lescrenier MG, Goins BA (2011) Preclinical multimodality phantom design for quality assurance of tumor size measurement. BMC Med Phys 11:1PubMedCrossRef

36.

Szegedi M, Rassiah-Szegedi P, Fullerton G, Wang B, Salter B (2010) A proto-type design of a real-tissue phantom for the validation of deformation algorithms and 4D dose calculations. Phys Med Biol 55(13):3685–3699PubMedCrossRef

37.

Wilhjelm JE, Jespersen SK, Falk E, Sillesen H (2006) The challenges in creating reference maps for verification of ultrasound images. Ultrasonics 4(Suppl 1):e141–e146CrossRef

38.

Wang TJ (2011) Assessing the role of circulating, genetic, and imaging biomarkers in cardiovascular risk prediction. Circulation 123(5):551–565PubMedCrossRef

39.

Polonsky TS, McClelland RL, Jorgensen NW et al (2010) Coronary artery calcium score and risk classification for coronary heart disease prediction. JAMA 303(16):1610–1616PubMedCrossRef

40.

Wahl RL, Jacene H, Kasamon Y, Lodge MA (2009) From RECIST to PERCIST: Evolving Considerations for PET response criteria in solid tumors. J Nucl Med 50(Suppl 1):122S–150SPubMedCrossRef

41.

Cummings J, Ward TH, Dive C (2010) Fit-for-purpose biomarker method validation in anticancer drug development. Drug Discov Today 15(19–20):816–825PubMedCrossRef

42.

Richter WS (2006) Imaging biomarkers as surrogate endpoints for drug development. Eur J Nucl Med Mol Imaging 33(Suppl 1):6–10PubMedCrossRef

43.

Woodcock J, Woosley R (2008) The FDA critical path initiative and its influence on new drug development. Annu Rev Med 59:1–12PubMedCrossRef

44.

Hanahan D, Weinberg RA (2011) Hallmarks of cancer: the next generation. Cell 144(5):646–674PubMedCrossRef

45.

Soloviev D, Lewis D, Honess D, Aboagye E (2012) [(18)F]FLT: an imaging biomarker of tumour proliferation for assessment of tumour response to treatment. Eur J Cancer 48(4):416–424PubMedCrossRef

46.

Nguyen QD, Challapalli A, Smith G, Fortt R, Aboagye EO (2012) Imaging apoptosis with positron emission tomography: ‘bench to bedside’ development of the caspase-3/7 specific radiotracer [(18)F]ICMT-11. Eur J Cancer 48(4):432–440

 

Read Full Post »