Elsevier

Magnetic Resonance Imaging

Volume 30, Issue 9, November 2012, Pages 1301-1312
Magnetic Resonance Imaging

Original contribution
Promise and pitfalls of quantitative imaging in oncology clinical trials

https://doi.org/10.1016/j.mri.2012.06.009Get rights and content

Abstract

Quantitative imaging using computed tomography, magnetic resonance imaging and positron emission tomography modalities will play an increasingly important role in the design of oncology trials addressing molecularly targeted, personalized therapies. The advent of molecularly targeted therapies, exemplified by antiangiogenic drugs, creates new complexities in the assessment of response. The Quantitative Imaging Network addresses the need for imaging modalities which can accurately and reproducibly measure not just change in tumor size but changes in relevant metabolic parameters, modulation of relevant signaling pathways, drug delivery to tumor and differentiation of apoptotic cell death from other changes in tumor volume. This article provides an overview of the applications of quantitative imaging to phase 0 through phase 3 oncology trials. We describe the use of a range of quantitative imaging modalities in specific tumor types including malignant gliomas, lung cancer, head and neck cancer, lymphoma, breast cancer, prostate cancer and sarcoma. In the concluding section, we discuss potential constraints on clinical trials using quantitative imaging, including complexity of trial conduct, impact on subject recruitment, incremental costs and institutional barriers. Strategies for overcoming these constraints are presented.

Introduction

The conduct of oncology clinical trials in the 21st century must address the paradigm-shifting recognition that effective cancer therapies will require individualized molecularly targeted drugs, either singly or in combination. Attempts to evaluate an array of molecularly targeted drugs with novel effects on virtually every aspect of tumor physiology have highlighted the limitations of response criteria based on anatomic imaging. Trials involving molecularly targeted drugs provide opportunities to develop equally novel functional imaging criteria for several important aspects of clinical design. These include noninvasive imaging approaches to pretreatment tumor phenotyping; early, often quantitative, imaging criteria for treatment response; pharmacodynamic measurements of drug delivery and target pathway modulation; and differentiation of tumor treatment response from other physiologic effects of the experimental therapy.

The optimal application of quantitative imaging (QI) to oncology clinical trials requires systemic evaluation of matching of modalities to the most appropriate physiologic parameters, the ability to standardize modalities and algorithms across different platforms and among multiple institutions, awareness of incremental cost of the imaging techniques and estimates of potential cost savings by enrichment of responders. The members of the Quantitative Imaging Network (QIN) contend that the investment of clinical trial funding resources by both industry and granting agencies into the innovation and development of validated QI tools will be necessary for the development of molecularly targeted therapeutics that are therapeutically effective and delivered at minimal cost. This article provides an overview of applications of QI to phase 0 through phase 3 oncology trials. We describe the most common uses of QI for several specific tumor types and identify challenges to standardization posed by the characteristics of these tumors. In the concluding section, we discuss potential constraints on QI, including complexity of trial design and conduct, impact on subject recruitment, incremental costs and institutional barriers. Strategies for overcoming these constraints are presented.

Section snippets

Clinical trial design and QI

In phase 1 clinical trials of molecularly targeted agents, maximal tolerated dose may not be relevant, and biomarkers for identifying the minimal dose required for biologically active drug delivery at the target site are needed. QI may be used to demonstrate actual drug delivery to tumor or physiologic changes associated with target modulation. Phase 0 “proof of mechanism” trials to demonstrate activity of novel compounds may use QI endpoints to measure target modulation or more downstream

PET

PET imaging produces a quantitative, three-dimensional (3D) image of the distribution of PET radioisotopes in the body. The PET radiotracer chosen is designed to reflect relevant biochemical processes in the body. A PET camera detects 511-keV gamma photons emitted by positron annihilation events due to positron emission by radioisotopes (e.g., F-18), which are linked to a molecule of interest (e.g., 2-deoxy-2-[18F]fluoro-d-glucose or FDG), which traces the early aspects of glucose metabolism in

Gliomas

Imaging has always played a critical role in assessing glioma response to treatment [17]. Repeated tumor biopsies are challenging in brain tumor patients, and imaging has the advantage of noninvasively capturing the heterogeneity of gliomas that may be missed through sampling error in only partially resectable gliomas. Unfortunately, though, as opposed to tissue-based makers such as MGMT methylation or IDH1 mutation, no validated imaging biomarkers have been discovered, and even seemingly

Lessons learned and future directions

While QI has promise for contributions to the conduct of clinical trials and oncology practice, QIN investigators have encountered several challenges when integrating QI into clinical trials. This section discusses and describes these challenges and the strategies developed to overcome them.

Uncertainty about measurement characteristics of QI biomarkers is a barrier to the planning of large-scale multicenter trials involving QI. Randomized phase 3 clinical trials may fail if they are

Acknowledgments

This work is supported by the National Institutes of Health, Quantitative Imaging Network (U01-CA148131). The authors are grateful to Robert Nordstrom, Larry Clarke, Hannah Linden, David Mankoff, Mark Muzi, Savannah Partridge, Mia Levy and Daniel Rubin for helpful discussions and to Alicia DePastino for administrative support.

References (102)

  • M.J. Morris et al.

    Fluorinated deoxyglucose positron emission tomography imaging in progressive metastatic prostate cancer

    Urology

    (2002)
  • G. Grignani et al.

    A phase II trial of sorafenib in relapsed and unresectable high-grade osteosarcoma after failure of standard multimodal therapy: an Italian Sarcoma Group study

    Ann Oncol

    (2012)
  • R.K. Doot et al.

    Design considerations for using PET as a response measure in single site and multicenter clinical trials

    Acad Radiol

    (2012)
  • D.J. Sargent et al.

    Validation of novel imaging methodologies for use as cancer clinical trial end-points

    Eur J Cancer

    (2009)
  • J.E. Dancey et al.

    Guidelines for the development and incorporation of biomarker studies in early clinical trials of novel agents

    Clin Cancer Res

    (2010)
  • M. Buyse et al.

    Biomarkers and surrogate end points — the challenge of statistical validation

    Nat Rev Clin Oncol

    (2010)
  • R.L. Wahl et al.

    From RECIST to PERCIST: evolving considerations for PET response criteria in solid tumors

    J Nucl Med

    (2009)
  • A. Hoering et al.

    Randomized phase III clinical trial designs for targeted agents

    Clin Cancer Res

    (2008)
  • R. Simon

    Clinical trial designs for evaluating the medical utility of prognostic and predictive biomarkers in oncology

    Per Med

    (2010)
  • L.L. Carr et al.

    Phase II study of daily sunitinib in FDG-PET-positive, iodine-refractory differentiated thyroid cancer and metastatic medullary carcinoma of the thyroid with functional imaging correlation

    Clin Cancer Res

    (2010)
  • G.D. Demetri et al.

    Efficacy and safety of imatinib mesylate in advanced gastrointestinal stromal tumors

    N Engl J Med

    (2002)
  • L. Sokoloff et al.

    The [14C]deoxyglucose method for the measurement of local cerebral glucose utilization: theory, procedure, and normal values in the conscious and anesthetized albino rat

    J Neurochem

    (1977)
  • R. Boellaard

    Need for standardization of 18F-FDG PET/CT for treatment response assessments

    J Nucl Med

    (2011)
  • M. Atri

    New technologies and directed agents for applications of cancer imaging

    J Clin Oncol

    (2006)
  • D.R. Macdonald et al.

    Response criteria for phase II studies of supratentorial malignant glioma

    J Clin Oncol

    (1990)
  • P.Y. Wen et al.

    Updated response assessment criteria for high-grade gliomas: response assessment in neuro-oncology working group

    J Clin Oncol

    (2010)
  • L.C. Hygino da Cruz et al.

    Diffusion MR imaging: an important tool in the assessment of brain tumors

    Neuroimaging Clin N Am

    (2011)
  • W.B. Pope et al.

    Recurrent glioblastoma multiforme: ADC histogram analysis predicts response to bevacizumab treatment

    Radiology

    (2009)
  • D.A. Hamstra et al.

    Functional diffusion map as an early imaging biomarker for high-grade glioma: correlation with conventional radiologic response and overall survival

    J Clin Oncol

    (2008)
  • C. Galbán et al.

    The parametric response map is an imaging biomarker for early cancer treatment outcome

    Nat Med

    (2009)
  • K. Herholz et al.

    Monitoring the effect of chemotherapy in a mixed glioma by C-11-methionine PET

    J Neuroimaging

    (2003)
  • M. Wyss et al.

    Early metabolic responses in temozolomide treated low-grade glioma patients

    J Neurooncol

    (2009)
  • J. Schwarzenberg et al.

    3′-deoxy-3′-18F-fluorothymidine PET and MRI for early survival predictions in patients with recurrent malignant glioma treated with bevacizumab

    J Nucl Med

    (2012)
  • A.M. Spence et al.

    Regional hypoxia in glioblastoma multiforme quantified with [18F]fluoromisonidazole positron emission tomography before radiotherapy: correlation with time to progression and survival

    Clin Cancer Res

    (2008)
  • A.M. Spence et al.

    NCI-sponsored trial for the evaluation of safety and preliminary efficacy of 3′-deoxy-3′-[18F]fluorothymidine (FLT) as a marker of proliferation in patients with recurrent gliomas: preliminary efficacy studies

    Mol Imaging Biol

    (2009)
  • D.R. Aberle et al.

    Reduced lung-cancer mortality with low-dose computed tomographic screening

    N Engl J Med

    (2011)
  • M.K. Gould et al.

    Test performance of positron emission tomography and computed tomography for mediastinal staging in patients with non-small-cell lung cancer: a meta-analysis

    Ann Intern Med

    (2003)
  • W.A. Weber et al.

    Positron emission tomography in non-small-cell lung cancer: prediction of response to chemotherapy by quantitative assessment of glucose use

    J Clin Oncol

    (2003)
  • R.J. Hicks

    Role of 18F-FDG PET in assessment of response in non-small cell lung cancer

    J Nucl Med

    (2009)
  • B. Zhao et al.

    A pilot study of volume measurement as a method of tumor response evaluation to aid biomarker development

    Clin Cancer Res

    (2010)
  • K. Alkhawaldeh et al.

    Impact of dual-time-point (18)F-FDG PET imaging and partial volume correction in the assessment of solitary pulmonary nodules

    Eur J Nucl Med Mol Imaging

    (2008)
  • S. Obrzut et al.

    Comparison of lesion-to-cerebellum uptake ratios and standardized uptake values in the evaluation of lung nodules with 18F-FDG PET

    Nucl Med Commun

    (2007)
  • M.E. Juweid et al.

    Use of positron emission tomography for response assessment of lymphoma: consensus of the Imaging Subcommittee of International Harmonization Project in Lymphoma

    J Clin Oncol

    (2007)
  • P. Castellucci et al.

    Potential pitfalls of 18F-FDG PET in a large series of patients treated for malignant lymphoma: prevalence and scan interpretation

    Nucl Med Commun

    (2005)
  • B.D. Cheson et al.

    Revised response criteria for malignant lymphoma

    J Clin Oncol

    (2007)
  • Y. Kasamon et al.

    Phase II study of risk-adapted therapy of newly diagnosed, aggressive non-Hodgkin lymphoma based on midtreatment FDG-PET scanning

    Biol Blood Marrow Transplant

    (2009)
  • C. Lin et al.

    Whole-body diffusion-weighted magnetic resonance imaging with apparent diffusion coefficient mapping for staging patients with diffuse large B-cell lymphoma

    Eur Radiol

    (2010)
  • R.L. Wahl et al.

    Metabolic monitoring of breast cancer chemohormonotherapy using positron emission tomography: initial evaluation

    J Clin Oncol

    (1993)
  • N.C. Krak et al.

    Measuring response to chemotherapy in locally advanced breast cancer: methodological considerations

    Eur J Nucl Med Mol Imaging

    (2004)
  • L.K. Dunnwald et al.

    Tumor metabolism and blood flow changes by positron emission tomography: relation to survival in patients treated with neoadjuvant chemotherapy for locally advanced breast cancer

    J Clin Oncol

    (2008)
  • Cited by (0)

    View full text