Assessing Response to Therapy

Chapter 5 Assessing Response to Therapy




Introduction


This chapter is intended to illustrate evolving strategies in the use of imaging to assess response to therapy. Both anatomic Response Evaluation Criteria In Solid Tumors (RECIST) 1.1 and functional PET Response Evaluation Criteria In Solid Tumors (PERCIST) 1.0 imaging response criteria are discussed.


Cancer continues to be a major health problem as one in four deaths in the United States is due to cancer. However, we continue to see incremental improvements over time with the relative 5-year survival rate for cancer in the United States at 68%, up from 50% in the mid 1970s. Cancer death rates fell 21.0% among men and 12.3% among women during the 1991 to 2006 period in the United States. The American Cancer Society estimates that the cancer incidence decreased 1.3%/yr among men from 2000 to 2006 and 0.5%/yr from 1998 to 2006 among women. This decline is attributed mainly to falling smoking rates, improved cancer treatments, and earlier detection of cancer.1


Oncologic imaging is recognized as an integral part of the management of cancer patients. Continued improvement in survival and the introduction of novel and multimodality therapies demand greater contributions from imaging to assess the presence of tumor, its extent, and response to therapy.


The improved understanding of the basic mechanisms of tumor biology, immunology, carcinogenesis, and genetics provides a rich foundation for translating these findings into enhancing efforts to reduce the impact of cancer. Some of these areas include the understanding of inherited or acquired genetic mutations or malfunctions; elucidating the molecular pathways of cell proliferation; acknowledging the effects of immune response and vascular proliferation; plus more effective clinical cancer detection including magnetic resonance imaging (MRI), computed tomography (CT), and molecular imaging techniques paired with gene screening arrays to identify molecular abnormalities in individual patient’s cancer cells.


The challenges to imaging are continuously evolving as novel personalized therapies and multimodality regimens are developed. However, the scientific limitations and economic realities burden us with the need to provide proof of principle, of which imaging is an integral part of the daily care and the design of various clinical trials to treat cancer patients.


The ability of imaging to provide indices to response such as tumor size, perfusion, and more recently, functional imaging makes it a standard component of clinical practice and assessment of novel therapies. This central role is best exemplified by the multidisciplinary approach to the management of cancer patients. The integration of surgery, pathology, imaging, medical oncology, radiation oncology, and medical physics to cancer patient care attests to the complex nature of the disease and the need to bring together the expertise of a group in lieu of the traditional models on which singular patient-physician relationships are developed followed by subspecialist referrals.


The traditional subspecialty designations in diagnostic imaging have and continue to be anatomic regions—for example, neuroradiology (head and/or neck), thoracic (chest), body (abdomen/pelvis), and others. However, cancer imaging demands expertise not only of specific anatomic areas but also in other modalities such as ultrasound (US), MRI, CT, x-ray plain films, and nuclear medicine, including positron-emission tomography (PET)/CT. This multimodality ability is now supported by the ready availability of images via picture archiving and communication system (PACS) and electronic medical records and, when necessary, ready access to other imaging specialists because it may be difficult to manage expertise in so many modalities. Easier access to referring physicians for consultation is also aided by fast communications via smartphones, the web, or the traditional page and phone system. Finally, the availability and use of voice-recognition systems and web access allows rapid turnaround of report results to both referring physicians and patients themselves. The transparency of these imaging reports should remind us all to avoid causing unnecessary anxiety in the proper use of language that is accurate and concise and hopefully answers the clinical question being posed.


For both the individual patient and clinical trial patients, close communication between the interpreting doctor and the referring physician is necessary for deciding the most appropriate imaging technique to use and when to perform a follow-up study to assess response. Appropriate care in planning the imaging component of clinical trials is essential, which may include proper imaging techniques, analysis, reporting, image transfer, and designing forms that may need to be filled out for these studies. Ideally, these imaging modalities and measurements are identical in both individual and trial patients, which may make clinical imaging research easier to perform or even to incorporate an individual patient into a clinical trial. Such planning will avoid added costs of repeat imaging or the need to go back and reanalyze images. Many of these imaging strategies could be made easier by accreditation of the imaging facility by the ACR (American College of Radiology) because it will ensure that the imaging equipment and qualifications of staff and physicians are registered, which then makes it easier to participate in clinical imaging trials such as ACRIN (American College of Radiology Imaging Network). Ensuring the high quality of imaging benefits primarily our patients but also allows easy participation in clinical research, which is the foundation of continuing improvement in our various specialties.




Why do We Need to Monitor Tumor Response?


The need for monitoring response became apparent in the early days of chemotherapy, particularly for conducting clinical comparative trials for various experimental chemotherapeutic agents in multiple cancer types. The typical development pathway for cancer therapeutic drugs is an evolution from phase I to phase II and to phase III clinical trials. In phase I trials, toxicity of the agent is assessed to determine what dose is appropriate for subsequent trials. In phase II trials, evidence of antitumor activity is obtained. Phase II trials can be done in several ways. One design is to examine tumor response rate versus a historical control population treated with an established drug. New drugs with a low response rate are typically not moved forward to advanced clinical testing under such a design. In such trials, tumor response has traditionally been determined with anatomic imaging techniques. An alternative approach is to use a larger sample size and have a randomized phase II trial, in which the new treatment is given in one treatment arm and compared with a standard treatment. Once drug activity is shown in phase II, phase III trials are then performed. Phase III trials are larger and usually have a control arm treated with a standard therapy. Therefore, imaging is expected to have a major role not only in the individual patient care but in designing clinical trials to select which therapies should be advanced to progressively larger trials and become standard of care.



History of an Evolving Imaging-based Response Assessment


An early study to assess response was done by Moertel and Hanley,2 in which 16 experienced oncologists were asked to measure 12 simulated tumors, placed underneath foam, using their clinical methods, which entailed physical examination with a ruler or caliper. Although seemingly crude, this was an appropriate simulation of the clinical setting in which a physician will palpate a tumor and then estimate its size before and after administering the treatment. This paper suggested that a 50% reduction in the perpendicular diameters of the tumors done at approximately 2 months is an acceptable objective response rate. This 50% reduction in bidimensional measurement of a single lesion was adopted in the World Health Organization (WHO) guidelines in 1979. Miller3 and coworkers recommended that a partial response be identified if there is a 50% reduction in the bidimensional measure of tumor area or, if multiple tumors are present, the sum of the product of the diameters. This study also described unidimensional measurements for “measurable” disease, bone metastases, and criteria for “nonmeasurable” disease. Tumor volume estimates were based on conventional radiography techniques by measuring the two longest perpendicular diameters and their product. Although widely used, obvious shortcomings of the WHO guidelines were the clinical foundation of the criteria without accounting for the improvements in imaging to determine tumor volumes. Tumors are rarely round or symmetrical, thus making these measurements difficult to implement, particularly by using a ruler or calipers. The lack of distinction between a complete versus a partial response in 50% to 90% decrease in tumor volume was an obvious flaw.


The European Organization of Research and Treatment of Cancer (EORTC) and the National Cancer Institute (NCI) of the United States and Canada set up a study group (RECIST)4 to standardize assessment criteria in cancer treatment trials. The objective was to simplify and standardize the methods to assess tumor response by more precisely defining tumor targets with proposed guidelines on imaging methods. Revisions on complete response, partial response, stable disease, and progressive disease were done. Unidimensional measurements were established for lesions of 2 cm or larger for CT, MRI, plain film, and physical examination and 1 cm or larger for spiral CT scan. The sum of the unidimensional tumor measurements was used for evaluation of response, which may decrease the sources of error.


RECIST criteria were adopted by multiple investigators, cooperative groups, and industry and government entities in assessing the treatment outcomes. However, a number of questions and issues have arisen that have led to the development of revised RECIST 1.1 guidelines.



RECIST 1.1: The Current Standard


The major changes of RECIST 1.1 include the number of lesions required to assess tumor burden for response determination has been reduced to a maximum of five total (and two per organ, maximum). Assessments of pathologic lymph nodes with a short axis of 15 mm are considered measurable and assessable as target lesions. The short-axis measurement should be included in the sum of lesions in calculation of tumor response. Nodes that shrink to less than 10 mm on the short axis are considered normal. Confirmation of response is required for trials with response primary endpoint but is no longer required in randomized studies because the control arm serves as appropriate means of interpretation of data. Disease progression is clarified in several aspects: in addition to the previous definition of progression in target disease of 20% increase in sum, a 5-mm absolute increase is now required as well to guard against overcalling progressive disease when the total sum is very small. Furthermore, guidance is offered on what constitutes “unequivocal progression” of nonmeasurable/nontarget disease, a source of confusion in the original RECIST guidelines. Finally, a section on detection of new lesions, including the interpretation of 2-[18F] fluoro-2-deoxy-D-glucose (FDG)–PET scan assessment, is included. Finally, the revised RECIST 1.1 includes a new imaging appendix with updated recommendations on the optimal anatomical assessment of lesions.5


The RECIST Working Group, in developing RECIST 1.1 concluded that, at present, there is not sufficient standardization or evidence to abandon unidimensional anatomic (vs. volumetric) assessment of tumor burden. The only exception to this is in the use of FDG-PET imaging as an adjunct to determination of progression.


Although these anatomic criteria have been evolving to affect better response criteria, the RECIST criteria and now, quite likely, the RECIST 1.1 criteria are or will be used in virtually every clinical trial of new solid tumor therapeutics, because response is essentially always measured. Regulatory agencies have accepted RECIST as the standard in response assessment for clinical trials in most countries. Familiarity with the implications of trials in which response is measured using the WHO, RECIST, and RECIST 1.1 criteria is essential because they are not identical and do not produce identical results. Table 5-1 presents RECIST 1.1 overall response criteria for both measurable and nonmeasurable lesions.


Table 5-1 RECIST 1.1 Target Lesions Response Criteria


















Objective response RECIST 1.1 target lesions* change in sum of LDs, maximum of two per organ up to five total.
Complete response
Partial response Decrease in target LD sum ≥ 30%, confirmed at 4 wk.
Progressive disease
Stable disease Does not meet other criteria.

CT, computed tomography; FDG, fluoro-2-deoxy-D-glucose; LD, lesion diameter; RECIST, Response Evaluation Criteria in Solid Tumors.


* Measurable lesion, unidimensional (LD only: size with conventional techniques ≥ 20 mm, with spiral CT ≥ 10 mm; nodes: target short axis ± 15 mm, nontarget 10- to 15-mm nodes, normal < 10 mm). Nonmeasurable: all other lesions, including small lesions; evaluable is not recommended.


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


Mar 6, 2016 | Posted by in GENERAL RADIOLOGY | Comments Off on Assessing Response to Therapy

Full access? Get Clinical Tree

Get Clinical Tree app for offline access