Download as pdf or txt
Download as pdf or txt
You are on page 1of 17

Review Article

Immunotherapy and the Role of Imaging


Brett W. Carter, MD ; Priya R. Bhosale, MD; and Wei T. Yang, MD

Significant advances in the genetic and molecular characterization of cancer have led to the development of effective immunothera-
pies. These therapeutics help the host immune system recognize cancer as foreign, promote the immune system, and relieve the inhi-
bition that allows growth and spread of tumors. Experience with various immunotherapies, particularly the immunomodulatory
monoclonal antibody ipilimumab, has demonstrated that unique patterns of response may be encountered that cannot be adequately
captured by traditional response criteria, such as the World Health Organization (WHO) criteria and Response Evaluation Criteria in
Solid Tumors (RECIST), which have been used primarily with cytotoxic chemotherapies. In response to these observations, several
novel response criteria have been developed to evaluate patients who receive immunotherapy, including immune-related response
criteria (irRC), immune-related RECIST (irRECIST), and immune RECIST (iRECIST). These criteria are typically used in conjunction
with RECIST version 1.1 in the clinical trial setting, because approval of new therapeutics by the US Food and Drug Administration
relies on the responses derived from RECIST version 1.1. Finally, a wide variety of immune-related adverse events may affect patients
who receive immunotherapy, many of which can be identified on imaging studies such as computed tomography, magnetic resonance
imaging, and 2-deoxy-2-(fluorine-18)fluoro-D-glucose–positron emission tomography/computed tomography. In this review, the
authors present the role of imaging in the evaluation of patients treated with immunotherapy, including the background and applica-
tion of irRC, irRECIST, and iRECIST; the imaging of immune-related adverse events; and future directions in advanced imaging of
immunotherapy. Cancer 2018;124:2906-22. V C 2018 American Cancer Society.

KEYWORDS: imaging, immune Response Evaluation Criteria in Solid Tumors (iRECIST), immune-related response criteria (irRC),
immune-related Response Evaluation Criteria in Solid Tumors (irRECIST), immunotherapy, response criteria.

INTRODUCTION
Significant advances in the genetic and molecular characterization of cancer have led to the development of effective
immunotherapies, which assist the host immune system in recognizing cancer as foreign, stimulate the immune system,
and relieve the inhibition that allows growth and spread of tumors. In contrast to traditional chemotherapy, which mainly
targets rapidly dividing cells, and targeted therapies, which interfere with important molecular events in cancer cells that
drive growth and invasion, immunotherapy attempts to assist in the recognition of cancer as foreign by the host immune
system, stimulate the immune system, and relieve the inhibition that allows tumor growth and spread. Immunotherapy is
described as either active or passive, depending on its interaction with the host immune system and the type of response
elicited. For instance, the active immune response involves humoral and/or cell-mediated immunity, whereas the passive
immune response requires no activation of the immune system and involves passively infused, preformed antitumor
immunoglobulins to tumor-associated antigens.
As the treatment options for cancer continue to advance, the development of imaging criteria for appropriately
assessing response to therapy is critical. Increased use of immunotherapy has demonstrated a wide variety of imaging fea-
tures and patterns of disease that are imperative to understand when assessing patients.1-3 Extensive experience with ipili-
mumab, an immunomodulatory monoclonal antibody, has demonstrated that several unique responses may be observed
during the course of treatment with immunotherapies that are not adequately captured by traditional response criteria like
the World Health Organization (WHO) criteria and Response Evaluation Criteria in Solid Tumors (RECIST).4 For
instance, in contrast to conventional response criteria, a transient increase followed by a subsequent decrease may be
encountered as treatment response (so called pseudoprogression). However, delayed or subsequent regression has been
demonstrated in only about 10% of patients with melanoma.4 Because of these observations, several response criteria have
been developed to evaluate patients who receive immunotherapy, including immune-related response criteria (irRC),
immune-related RECIST (irRECIST), and immune RECIST (iRECIST).4,5 With the increasing use of immunotherapy,
it is becoming easier to recognize numerous immune-related adverse events, such as colitis, hepatitis, pneumonitis, and
various endocrine toxicities.2,6 It is important for radiologists and other providers to be familiar with the imaging
Corresponding author: Brett W. Carter, MD, Department of Diagnostic Radiology, Section of Thoracic Imaging, The University of Texas MD Anderson Cancer
Center, 1515 Holcombe Boulevard, Unit 1478, Houston, TX 77030; bcarter2@mdanderson.org

Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, Texas

DOI: 10.1002/cncr.31349, Received: November 16, 2017; Revised: January 30, 2018; Accepted: February 1, 2018, Published online April 19, 2018 in Wiley
Online Library (wileyonlinelibrary.com)

2906 Cancer July 15, 2018


Immunotherapy and the Role of Imaging/Carter et al

manifestations of immune-related adverse events in between patients who are and are not responding to a par-
patients who are receiving immunotherapy and to distin- ticular treatment as early as possible.
guish these findings from progression of disease.
In this review, we present the role of imaging in the Immune-Related Response Criteria
Background and development
evaluation of patients treated with immunotherapy,
In 2004 and 2005, a multidisciplinary group of experts
including specific response criteria, such as irRC, irRE-
presented and analyzed their experiences with immuno-
CIST, and iRECIST, and the imaging of immune-related
adverse events. therapy and treatment response. This group outlined sev-
eral key points: 1) Measurable antitumor activity may
EVALUATION OF TREATMENT RESPONSE take longer to manifest for immunotherapies than for tra-
Investigators traditionally have relied on response criteria ditional chemotherapy; 2) responses to immunotherapy
to evaluate the effectiveness of chemotherapy, the most may occur after the identification of PD by traditional
widely used of which have been the WHO criteria and response criteria, such as RECIST 1.1; 3) continuation of
RECIST guidelines. The WHO criteria were the first immunotherapy may be appropriate in clinically stable
standardized response criteria but were limited by several patients who have PD; 4) consideration of clinically insig-
factors.7 The original RECIST guidelines (RECIST 1.0), nificant PD is recommended (such as the development of
published in 2000, were developed by a large, interna- new, small lesions in a patient with lesions that are
tional group of investigators to standardize the characteri- responding to therapy); and 5) prolonged and durable sta-
zation of treatment efficacy using specific response ble disease (SD) may represent effective therapy.4 An anal-
definitions.8 This was followed by the release of revised ysis of 487 patients with unresectable stage III or IV
RECIST criteria (RECIST 1.1) in in 2009.9 These criteria melanoma who received treatment with ipilimumab (a
are optimized to evaluate the cytotoxic effects of chemo- human monoclonal antibody that blocks cytotoxic T-
therapeutic agents on solid tumors, and specific effects lymphocyte antigen-4 [CTLA-4]) in 3 multicenter, phase
can be identified on imaging studies usually within a few 2 clinical trials demonstrated 4 patterns of clinical
weeks of therapy. These guidelines depend on a decrease response, including: 1) a decrease in lesion size without
in the size of tumors and the absence of new lesions.1,4 new disease, 2) SD after the completion of therapy, 3) a
Therefore, it is assumed that a significant increase in delay in response after an initial increase in total tumor
tumor growth and/or the appearance of new lesions indi- burden (TTB), and 4) the appearance of new lesions
cates progressive disease (PD). Typically, chemotherapy is before a decrease in tumor size. The first 2 response pat-
discontinued once progressive disease (PD) is docu- terns were appropriately captured with conventional
mented. A significant limitation of these traditional response criteria, including responses in baseline lesions
response criteria is that several specific patterns of without new lesions and, in some patients, SD followed
response known to occur in patients who receive treat- by a slow, steady decline in disease. The other 2 response
ment with other agents, such as antiangiogenic therapies patterns were novel and included responses after an initial
or immune therapies, are not appropriately captured. For increase in TTB (so-called pseudoprogression) and a
instance, new agents, such as cytokines, cancer vaccines, reduction in TTB during or after the appearance of new
and immunomodulatory monoclonal antibodies, may lesion(s).4 The immune-related response criteria (irRC)
produce a delayed response, a transient increase in size fol- published by Wolchok et al in 2009 were based on these
lowed by tumor shrinkage, and/or the appearance of new observed patterns.
lesions, which become stable, decrease, or resolve without
further treatment.4 However, only about 10% of patients Principles and application
who receive immunotherapy demonstrate regression after The irRC were adapted from the WHO criteria and
initial progression.4 designed to address treatment response and recommenda-
To reflect these observations and overcome existing tions for imaging follow-up of patients who received
limitations, many modifications have been made to tradi- immunotherapy. There are many differences between
tional response criteria; however, in many instances, these conventional response criteria, such as RECIST 1.1, and
criteria have not been standardized and incorporated into irRC (Table 1). For instance, the irRC do not specifically
clinical trials in an organized fashion. Thus, new criteria, address which imaging modalities should be used in the
such as irRC, irRECIST, and iRECIST, have been devel- evaluation of treatment response, and anatomic and com-
oped to assist investigators and clinicians in distinguishing bined anatomic and metabolic methods of assessment are

Cancer July 15, 2018 2907


TABLE 1. Comparison of Response Evaluation Criteria in Solid Tumors (RECIST), Immune-Related Response Criteria, Immune-Related RECIST,
and Immune RECIST

2908
Variable RECIST 1.1 irRC irRECIST iRECIST

Imaging modality CT, MRI, FDG-PET, chest radiography;a Not specifically mentioned; CT, MRI, CT, MRI, FDG-PET, chest radiography;a CT, MRI, FDG-PET, chest radiography;a
clinical measurements documented FDG-PET may be used; clinical clinical measurements documented by clinical measurements documented by
Review Article

by photography with rulerb measurements documented by photography with rulerb photography with rulerb
photography with rulerb
No. of target Two per organ, 5 total Five per organ, up to 10 visceral and 5 Two per organ, 5 total Two per organ, 5 total
lesions cutaneous
Measurable size Solid-organ lesions 10 mm in long axis, 5 3 5 mm Solid-organ lesions 10 mm in long axis; Solid-organ lesions 10 mm in long axis, 15
15 mm in short axis for lymph nodesc 15 mm in short axis for lymph nodesc mm in short axis for lymph nodesc
Measurement Unidimensional measurement Bidimensional measurements Unidimensional measurement Unidimensional measurement
parameters
Tumor burden Sum of the greatest dimensions of all Sum of the products of the 2 greatest Sum of the greatest dimensions of all Sum of the greatest dimensions of all
target lesions perpendicular dimensions of target lesions target lesions
all target lesions
Unmeasurable 1. Measurable lesions not selected as Not specifically defined; nontarget lesions Follow definitions from RECIST 1.1 Follow definitions from RECIST 1.1
lesions target lesions derived from irRC include: lymphangitic
carcinomatosis, skin involvement
in breast cancer, abdominal masses
that can be palpated but not measured
2. Unmeasurable disease, including lesions
that are too small to measure (<10 mm
in long axis for solid-organ lesions or <15
mm in short axis for lymph nodes)
3. Other types of lesions that are difficult to
measure in a reproducible manner include:
bone metastases, leptomeningeal metas-
tases, malignant ascites, pleural or peri-
cardial effusions, ascites, inflammatory
breast disease, lymphangitic
carcinomatosis, ill-defined abdominal
masses, skin lesions, etc
New lesions Constitutes PD Does not constitute PD automatically; Does not constitute PD automatically; Results in iUPD; iCPD only achieved if
incorporated into total tumor burden incorporated into total measured additional new lesions are present on
tumor burden the subsequent time point or there is an
increase in the size of previous new lesions
(5 mm for the sum of new
target lesions or any increase in new
nontarget lesions)

Abbreviations CT, computed tomography; iCPD, immune RECIST-confirmed progressive disease; irRC, immune-related response criteria; iUPD, immune RECIST-unconfirmed progressive disease; MRI, magnetic
resonance imaging; PD, progressive disease; PET-FDG, positron emission tomography–2-deoxy-2-(fluorine-18)fluoro-D-glucose; RECIST 1.1, Response Evaluation Criteria in Solid Tumors, version 1.1.
a
Target lesions require a size of 20 mm on chest radiography according to RECIST.

Cancer
b
Cutaneous lesions are included in potential tumor burden; therefore, clinical measurements theoretically may be allowed.
c
Sizes are confirmed on newer generation multidetector imaging. Other parameters are listed for nonhelical CT (10 mm slice thickness) and chest radiography.

July 15, 2018


Immunotherapy and the Role of Imaging/Carter et al

TABLE 2. Response Assessment and New Lesions in Immune-Related Response Criteria

irRC Category Description

Complete response (irCR) Complete disappearance of all lesions; confirmation by repeat, consecutive assessment 4 wks from first
documentation
Partial response (irPR) Decrease in tumor burden 50% relative to baseline; confirmation by repeat, consecutive assessment 4 wks from
first documentation
Stable disease (irSD) Does not meet criteria for irCR or irPR, in the absence of irPD
Progressive disease (irPD) Increase in tumor burden 25% relative to nadir (minimum recorded tumor burden); confirmation by repeat,
consecutive assessment 4 wks from first documentation
New measureable lesions Incorporated into tumor burden
New nonmeasurable lesions New, nonmeasurable lesions at follow-up time points are not added to total tumor burden and do not define
progression but preclude irCR; nontarget, unequivocal progression contributes to irCR but not irPD

Abbreviations: irRC, immune-related response criteria.

Figure 1. Images of pseudoprogression from a woman aged 64 years who had adenocarcinoma of the right lower lobe and
received radiation therapy and immunotherapy are shown. (A) A contrast-enhanced, axial computed tomography (CT) scan
obtained at baseline reveals a right lower lobe nodule (arrow). Note the small right pleural effusion (asterisk). (B) A follow-up
contrast-enhanced, axial CT scan obtained 6 weeks later shows an interval increase in the size of the nodule (arrow) and an
increase in the size of the right pleural effusion (asterisk). (C) Another follow-up contrast-enhanced, axial CT obtained 6 weeks
later reveals an interval decrease in the size of the nodule (arrow) and a slight increase in the size of the right pleural effusion
(asterisk). With novel response criteria designed for use with immunotherapy, new lesions or an increase in tumor burden must
be confirmed by repeat imaging at a minimum of 4 weeks later to constitute progression.

Cancer July 15, 2018 2909


Review Article

Figure 2. Images of pseudoprogression from a woman aged 66 years who received immunotherapy for lung cancer are shown.
(A) A contrast-enhanced, axial computed tomography (CT) scan at baseline demonstrates a normal-sized, right paratracheal
lymph node (arrow). (B) A follow-up contrast-enhanced, axial CT scan obtained 6 weeks later reveals a marked interval increase
in the size of the right paratracheal lymph node (arrow) and the presence of additional, smaller lymph nodes in the right prevas-
cular mediastinum (arrowhead). (C) Another follow-up contrast-enhanced, axial CT scan obtained 6 weeks later shows a marked
interval decrease in the size of the right paratracheal lymph node (arrow) and disappearance of the other lymph nodes in the
right prevascular mediastinum.

frequently used interchangeably in clinical trials. How- Once the TTB is calculated, an overall response cate-
ever, in contrast to RECIST 1.1, only anatomic measure- gory is assigned to each case based on changes between
ments are considered. For all cases, the TTB, defined as time point assessments. These response categories include
the sum of the products of the 2 greatest perpendicular a complete response (irCR), a partial response (irPR), SD
dimensions (SPD) of all index lesions (measured in (irSD), and PD (irPD) (Table 2). The TTB must increase
mm2), is calculated at baseline and at all subsequent time by 25% and must be confirmed by repeat imaging at a
points. Target lesions must measure 5 3 5 mm, and up minimum of 4 weeks later to constitute definitive irPD.
to 5 may be selected per organ (up to 10 lesions involving New lesions or a perceived increase in TTB because of
visceral organs and 5 cutaneous lesions). At subsequent pseudoprogression can result from immune cell recruit-
time points, the TTB is calculated by the addition of the ment to sites of microscopic disease (Figs. 1 and 2). In
SPD of all target lesions and the SPD of any new measur- irRC, all patients who have an irCR, irPR, and irPD must
able lesions (TTB 5 SPDindex lesions 1 SPDnew measur- have repeat imaging performed at a minimum of 4 weeks
able lesions). This differs significantly from conventional later for confirmation if they are clinically stable; however,
response criteria, which consider all new lesions as PD. follow-up with observation alone may not be appropriate

2910 Cancer July 15, 2018


Immunotherapy and the Role of Imaging/Carter et al

TABLE 3. Response Assessment and New Lesions in Immune-Related Response Evaluation Criteria in Solid
Tumors

irRC Category Description

Complete response (irCR) Complete disappearance of all measurable and nonmeasurable lesions; lymph nodes must decrease to <10 mm
in the short axis; confirmation is not mandatory
Partial response (irPR) Decrease 30% in TMTB relative to baseline; nontarget lesions are irNN; no unequivocal progression of new
nonmeasurable lesions
Stable disease (irSD) Failure to meet criteria for irCR or irPR in the absence of irPD
Progressive disease (irPD) Minimum 20% increase and minimum 5-mm absolute increase in TMTB compared with nadir or irPD for
nontarget or new nonmeasurable lesions; confirmation of progression is recommended a minimum of 4 wks
after the first irPD assessment
New measureable lesions Greatest dimensions of new measurable, nonlymph node lesions and short axes of new measurable lymph nodes
will be recorded and added to the TMTB at follow-up
New nonmeasurable lesions All new lesions not selected as new measurable lesions are considered new nonmeasurable lesions and are
followed qualitatively; only a massive and unequivocal progression of new nonmeasurable lesions leads to
an overall assessment of irPD for the time point; persisting new unmeasurable lesions prevent irCR

Abbreviations: irRC, immune-related response criteria; irNN, irNon-CR/Non-PD; TMTB, total measured tumor burden.

for those who have a rapid decline in performance status. ipilimumab for melanoma. Those authors demonstrated
The response of patients who have irSD, particularly those that unidimensional measurements simulating RECIST
with a slowly declining tumor burden (25% from base- 1.1 were more reproducible compared with the bidimen-
line at the last tumor assessment), is considered clinically sional measurements of irRC.5 The group recommended
meaningful, because they exhibit an objectively measur- that the number of targets could be reduced to up to 2
able reduction in TTB without reaching the 50% thresh- per organ and up to 5 in total, as defined in RECIST
old that defines irPR. In addition, it is recommended 1.1, and proposed the use of unidimensional measure-
that, after the completion of therapy, an evaluation of ments to assess response to immunotherapy in solid
treatment response should be made with 2 consecutive tumors given their relative simplicity, higher reproduc-
follow-up imaging studies at least 4 weeks apart because ibility, and high concordance with the bidimensional
of the potential for a delayed response. measurements of irRC.5

Limitations Principles and application


Although the irRC introduced several advancements in The irRECIST are very similar to RECIST 1.1 in terms
terms of assessing response in patients treated with immu- of guidance regarding the selection of imaging modalities,
notherapy, several limitations have been described. For definitions of measureable and unmeasurable disease, and
instance, the irRC provide no guidance regarding the num- criteria for selecting target and nontarget lesions.11 For
ber of lesions that should be included, and assessing a rela- example, target lesions may include nonlymph node
tively large number of lesions per organ could be relatively lesions measuring 10 mm in the long axis or lymph
time consuming in patients with extensive disease.2,10 In nodes measuring 15 mm in the short axis. As in
addition, there is no specific guidance regarding the assess- RECIST 1.1, up to 5 total target lesions may be selected
ment of lymph node disease. The reproducibility of bidi- with a maximum of 2 per organ, and the measurements of
mensional assessment also is lower than that of these lesions are recorded as the total measured tumor bur-
unidimensional assessment. Finally, although nontarget den (TMTB). Nontarget lesions may include measurable
unequivocal progression contributes to irCR based on the lesions not selected as target lesions, sites of nonmeasur-
initial description by Wolchok and colleagues,4 it does not able disease, and lesions that may be difficult to measure
do so with respect to irPD; however, this was not used in in a reproducible manner, such as bone lesions, leptome-
the majority of studies that applied the irRC. ningeal metastases, inflammatory breast disease, malignant
ascites, pleural or pericardial effusions, and lymphangitic
Immune-Related RECIST carcinomatosis. It is generally recommended that previ-
Background and development ously treated lesions should not be selected as target lesions
Nishino and colleagues evaluated the impact of reducing unless local progression has been documented.
the number of target lesions and using unidimensional A significant difference between irRECIST and
measurements to assess patients who received RECIST 1.1 and other response criteria is the method by

Cancer July 15, 2018 2911


Review Article

TABLE 4. Response Assessment and New Lesions in Immune Response Evaluation Criteria in Solid Tumors

iRECIST Category Description

Complete response (iCR) Resolution of all lesions, confirmed 4 wks


Partial response (iPR) Decrease 30% in tumor burden compared with baseline in the absence of any new lesion or progression of nontarget
lesions
Stable disease (iSD) Neither PR or PD in the absence of any new lesion or progression of nontarget lesions
Unconfirmed progressive Increase 20% in tumor burden from nadir, progression of nontarget lesions, or new lesions; iUPD requires confirmation,
disease (iUPD) which is done on the basis of observing either a further increase in size (or in the number of new lesions) in the lesion
category in which progression was first identified (target or nontarget disease) or progression (defined by RECIST 1.1)
in lesion categories that had not previously met RECIST 1.1 progression criteria; progression is confirmed in the target
lesion category if the next imaging assessment after iUPD (4-8 wks later) confirms a further increase in sum of
measures of target disease from iUPD, with an increase of at least 5 mm
Confirmed progressive Progressive disease is confirmed (iCPD) if the next imaging assessment, done 4-8 wks after iUPD, confirms additional
disease (iCPD) new lesions, further increase in previous new lesion size (sum of measures increase in new target lesion 5 mm or
any increase for new nontarget lesions), or further increase in existing target or nontarget lesions from iUPD
New lesions Assessed according to RECIST 1.1 guidelines but recorded separately and not included in the sum of lesions for target
lesions identified at baseline; new lesions constitute iUPD; iCPD is only achieved if additional new lesions appear or
there is an increase in size of new lesions (5 mm for the sum of new target lesions or any increase in new nontarget
lesions); presence of new lesions when none have previously been recorded can also confirm iCPD

Abbreviations: iRECIST, Immune Response Evaluation Criteria in Solid Tumors; PD, progressive disease; PR, partial response; RECIST 1.1, Response Evalua-
tion Criteria in Solid Tumors, version 1.1.

which new lesions are incorporated into the assessment of Limitations


treatment response. In irRECIST, new lesions may be One of the most significant limitations of irRECIST is
characterized as measurable or unmeasurable, and those that these recommendations have not always been consis-
selected as new target lesions must meet the same criteria tently applied, raising concerns among clinicians and
for inclusion as the lesions that were present at baseline. researchers regarding the comparability of data and results
When multiple qualifying lesions are present, it is recom- across various clinical trials. In addition, studies that dem-
mended that they be prioritized according to size, with the onstrated the efficacy of irRECIST were performed on rel-
largest lesions selected as new target lesions. In total, 5 atively small cohorts of patients, and more detailed
new target lesions may be selected, with a maximum of 2 evaluation of irRECIST 1.1 is still required. Finally, in
per organ. New measurable lesions that are not selected as contrast to iRECIST, with irRECIST, a delayed response
target lesions may be classified as new nonmeasurable after pseudoprogression that does not reach irSD will
lesions and can be qualitatively followed. When new mea- result in a confirmation of irPD.
surable lesions are selected as target lesions, the greatest
dimensions of both existing and new nonlymph node tar- Immune RECIST
get lesions and the short axis dimensions of both existing Background and development
and new lymph node target lesions constitute the TMTB. In an effort to standardize and validate response criteria,
This represents a significant difference from RECIST 1.1, the RECIST working group evaluated data collected from
in which the appearance of new lesions is always consid- prospective clinical trials using immunotherapy and
ered PD, and therapy typically is discontinued, without a RECIST 1.1.12 That group reported that most clinical tri-
method for following these new lesions. als involving immunotherapy used RECIST 1.1 to define
Similar to irRC, the overall response categories for primary and secondary efficacy-based endpoints and used
irRECIST include irCR, irSD, irPR, and irPD and are the irRC or modified definitions of RECIST for explor-
based on changes in the TMTB of measured target atory endpoints.13,14 There was significant variability in
lesions (baseline and new), nontarget lesion assessment, which criteria were incorporated clinically within differ-
and new nonmeasurable lesions (Table 3). These ent groups, raising concerns about the interpretation of
response categories have defined thresholds for irPR and pooled data sets. In addition, most clinical trials that
irPD that are aligned with RECIST 1.1. For patients applied immune-modified criteria used independent
with irPD and a minimal TMTB percentage increase imaging review by a commercial entity for those criteria
>20%, particularly early on in therapy (such as during rather than investigator assessments. In response to the
the first 12 weeks of treatment), confirmatory evaluation limitations of existing response criteria and these observa-
may be considered. tions, the RECIST working group developed guidelines

2912 Cancer July 15, 2018


Immunotherapy and the Role of Imaging/Carter et al

Figure 4. Images of pneumonitis are shown. (A) An unen-


hanced, axial computed tomography (CT) scan from a woman
aged 65 years with lung cancer who received immunotherapy
Figure 3. Images of pneumonitis are shown. Contrast- reveals diffuse opacities in the lungs. (B) An unenhanced, axial
enhanced (A) axial and (B) coronal computed tomography CT scan from a woman aged 53 years with melanoma who
images from a man aged 21 years with melanoma who received immunotherapy demonstrates diffuse ground-glass
received immunotherapy demonstrate a cryptogenic organiz- opacities superimposed on a background of interlobular septal
ing pneumonia-like appearance of immune-related pneumoni- thickening in a crazy-paving configuration. Immune-related
tis with opacities in the lower lobes, appearing as central pneumonitis may manifest in several ways, 1 of the most com-
ground-glass opacity and surrounding consolidation. mon of which is ground-glass opacity.

for the use of modified RECIST termed iRECIST to separately and are not included in the sum of the greatest
ensure consistent design and data collection and to facili- dimensions of all target lesions, in contrast to irRECIST.
tate the ongoing collection of data from clinical trials and The overall response categories for iRECIST
ultimate validation of the guidelines. include iCR, iSD, iPR, unconfirmed progressive disease
(iUPD), and confirmed progressive disease (iCPD),
Principles and application which are designed to allow for the identification and
The tenets of iRECIST are very similar to those of improved characterization of atypical responses12 (Table
RECIST 1.1 and irRECIST regarding the recommended 4). For target lesions, iCR, iPR, and iSD all can be
imaging modalities, definitions of measureable and assigned after iUPD has been documented, as long as
unmeasurable disease, and criteria for selecting target and iCPD has not been achieved. For nontarget lesions,
nontarget lesions.12 New lesions may be classified as target iUPD may be documented before iCR or when the crite-
or nontarget lesions and are evaluated as in RECIST 1.1. ria for neither iCR nor iPD have been met (referred to as
However, target and nontarget lesions are recorded non-iCPD/non-iUPD) and can be assigned several

Cancer July 15, 2018 2913


Review Article

Figure 5. Images of sarcoid-like reaction are shown. (A) A contrast-enhanced, axial computed tomography (CT) scan and (B) a fused,
axial 2-deoxy-2-(fluorine-18)fluoro-D-glucose–positron emission tomography (FDG-PET)/CT image from a man aged 64 years with
melanoma who received immunotherapy reveal new, FDG-avid, mediastinal and hilar lymphadenopathy (arrows). These lymph nodes
were subsequently biopsied without evidence of malignancy, and a clinical diagnosis of sarcoid-like reaction was made. (C) An unen-
hanced, axial CT scan and (D) and a fused, axial FDG-PET/CT study from the same patient 6 weeks later after discontinuation of ther-
apy demonstrates a marked interval decrease in the size and FDG uptake of the lymph nodes (arrows).

times, as long as iCPD has not been achieved. The change, if there is a decrease in tumor burden compared
appearance of new lesions results in iUPD; however, this with the baseline study that meets the criteria for iCR,
requires confirmation by a further increase in size (or in iPR, or iSD, then iUPD must be reached again and con-
the number of new lesions) in the lesion category in firmed at the next time point for iCPD to have been
which progression was first identified (target or nontar- achieved. If there is no change in tumor size or extent,
get) or by progression in lesion categories that did not then the time point response remains iUPD.
previously meet RECIST 1.1 progression criteria on an The RECIST working group recommends that
examination performed 4 to 8 weeks after iUPD. For treatment beyond initial RECIST 1.1-defined progression
instance, iCPD may be assigned if additional new lesions (iUPD) should/could only allow patients who are clini-
appear or if there is a further increase in the size of previ- cally stable to continue on treatment until the next assess-
ous new lesions (5 mm for the sum of target lesions or ment. This should be performed 4 weeks later but no
any increase in nontarget lesions) on the subsequent longer than 8 weeks later, to ensure that patients remain
examination. However, in the case of iUPD and no fit for salvage therapies. All decisions regarding

2914 Cancer July 15, 2018


Immunotherapy and the Role of Imaging/Carter et al

continuation or discontinuation of therapy should be


made by the oncologist and patient.

IMPLEMENTATION OF RESPONSE
CRITERIA IN CLINICAL TRIALS
Although these new response criteria have been developed
with principles of immunotherapy in mind, it should be
noted that the current recommendation is for them to be
used alongside RECIST 1.1 in clinical trials, because these
conventional criteria are used by the US Food and Drug
Administration for decisions regarding the approval of
therapeutics. For instance, the irRC have been used in sev-
eral clinical trials alongside RECIST 1.1 in which the
patient continues therapy beyond initial PD provided
there is investigator-assessed clinical benefit and the
patient tolerates the therapy. In this instance, the use of
the irRC is beneficial to avoid premature termination of
an effective immunotherapy with pseudoprogression when
evaluating treatment response with the imaging modalities
that are currently available. Hodi and colleagues compared
irRC and RECIST 1.1 in patients with advanced mela-
noma who received pembrolizumab and observed that
RECIST 1.1 underestimated the therapy benefit, as deter-
mined by overall survival, in 15% of patients.14 Those
authors suggested that the use of modified criteria that
permit treatment beyond initial PD determined by
RECIST 1.1 may prevent premature cessation of treat-
ment. However, atypical response rates for other tumor
entities (such as lung cancer and renal cancer) seem to be
inferior, and real progression might be more likely.
As a general limitation, all of these response criteria
are defined for anticancer response evaluation in clinical tri-
als of solid tumors with a distinct start point (baseline) and
endpoint (tumor progression). Many oncologists currently Figure 6. Images of sarcoid-like reaction are shown. (A) An
use RECIST 1.1, irRECIST, and iRECIST in their daily unenhanced, coronal, reformatted computed tomography (CT)
scan from a man aged 41 years with melanoma who received
clinical practice to follow their patients with cancer by immunotherapy reveals that numerous, small pulmonary nod-
repeated imaging studies and make decisions about contin- ules are present along the bronchovascular bundles and in the
subpleural regions, compatible with a perilymphatic pattern
ued therapy on the basis of both objective and symptomatic (arrows). (B) A contrast-enhanced, axial CT scan from a woman
criteria. However, these criteria may be limited and are not aged 37 years with lung cancer who received immunotherapy
demonstrates multiple, ill-defined pulmonary nodules in a peril-
always appropriate for routine clinical use in normal clinical ymphatic distribution (arrows) and thickening of bronchovascu-
therapy schemas, which frequently involve pauses of treat- lar bundles and interlobular septa in the left lung. In both of
these patients, the pulmonary findings resolved after discontin-
ment, modification of drug dosage, combined systemic and uation of therapy, and sarcoid-like reaction was diagnosed.
local tumor therapy, and treatment beyond progression, Sarcoid-like reaction is a rare complication that can manifest
which are not addressed by these criteria. with multiple micronodules with or without ground-glass opaci-
ties and/or mediastinal/hilar lymphadenopathy.

The Role of Positron Emission Tomography–2-


Deoxy-2-(Fluorine-18)Fluoro-D-Glucose/
Computed Tomography PET/CT) in the evaluation of patients who receive immuno-
The role of positron emission tomography–2-deoxy-2-(fluo- therapy is uncertain. One of the significant challenges with
rine-18)fluoro-D-glucose/computed tomography (FDG- FDG-PET/CT is the false-positive finding that can result

Cancer July 15, 2018 2915


Review Article

Figure 7. Imaging studies of thyroiditis are shown. (A,B) Contrast-enhanced, axial computed tomography (CT) scans from a
woman aged 49 years (A) before and (B) after the initiation of immunotherapy reveal a normal appearance of the thyroid gland
(A), which subsequently demonstrates enlargement (B). The patient’s thyroid function tests were abnormal, and a subsequent
ultrasound study confirmed a heterogenous and hypervascular thyroid, consistent with thyroiditis. (C) A fused, axial 2-deoxy-2-
(fluorine-18)fluoro-D-glucose (FDG)–positron emission tomography/CT image and (D) and a contrast-enhanced, axial CT scan
from a man aged 64 years with lung cancer who received immunotherapy demonstrate diffuse, increased FDG uptake through-
out the thyroid gland with mild enlargement and heterogeneous attenuation.

from increased FDG uptake by inflammatory processes, response was assessed based on RECIST 1.1, irRC, Positron
which is an issue, because immunotherapy elicits a natural Emission Tomography Response Criteria in Solid Tumors
inflammatory response, and immune-related adverse events (PERCIST 1.0), and European Organization for Research
also may demonstrate increased FDG uptake. Two studies and Treatment of Cancer criteria. Those authors developed
in patients with melanoma demonstrated the inability of criteria comprised of anatomic and functional information
FDG-PET/CT to differentiate between patients who had that predicted eventual response with 100% sensitivity, 93%
pseudoprogression because of inflammatory infiltrate and specificity, and 95% accuracy. FDG-PET/CT has demon-
those who had actual PD.15,16 Cho et al evaluated the ability strated benefit in evaluating the response of patients with
of FDG-PET/CT in conjunction with other imaging melanoma to treatment with BRAF and MEK inhibitors18
modalities to predict early response to therapy in patients but has proven to be inadequate in examining responses to
with advanced melanoma who receive treatment with several immunotherapy for other cancers, such as renal cell
checkpoint inhibitors.17 At each post-treatment time point, carcinoma.19

2916 Cancer July 15, 2018


Immunotherapy and the Role of Imaging/Carter et al

suggesting that they are not true autoimmune processes


but are the result of general immunologic enhancement.6
Analysis of clinical trial data has demonstrated significant
variability in the reporting quality of adverse events,21,22
which is typically inadequate.23-25 The most common
immune-related adverse event is dermatologic toxicity,
which may result in skin rash, and is typically low grade;
however, toxic necrotizing epidermolysis mucositis has
been reported.2,6 Additional effects include enterocolitis,
hepatitis, pneumonitis, and endocrinopathies, such as
hypophysitis, thyroiditis, and adrenal insufficiency. Other
complications of immunotherapy include sarcoid-like
reaction, acute kidney injury, pancreatitis, neurotoxicity,
cardiotoxicity, and ophthalmologic toxicity. Severe colitis
has the highest mortality and worst outcome associated
with immune-related adverse events.26

Imaging of Immune-Related Adverse Events


In evaluating patients who receive immunotherapy, it is
imperative for radiologists and other providers to recog-
nize unique adverse events to guide appropriate manage-
ment and prevent misinterpretation of studies. Side
effects often result in abnormalities that may be identified
on CT, MR imaging, or FDG-PET/CT performed for
restaging and/or surveillance. Immune-related adverse
effects may produce findings and metabolic changes
before the appearance of clinical symptoms, allowing early
change of therapy.1
Pneumonitis is the most common immune-related
adverse event in the chest. Chest radiography may be the
first imaging modality to demonstrate abnormalities
Figure 8. Imaging studies of colitis are shown. (A) A attributable to pneumonitis because of its widespread
contrast-enhanced, coronal computed tomography (CT) scan
from a woman aged 61 years with lung cancer who received
availability and use; however, these findings are typically
immunotherapy demonstrates segmental wall thickening incompletely characterized and nonspecific and are opti-
involving the descending colon (arrow), consistent with coli- mally evaluated on chest CT. Naidoo and colleagues
tis. (B) A contrast-enhanced, axial CT scan from a man aged
23 years with melanoma who received immunotherapy demonstrated that only 67% of cases resulted in identifi-
reveals diffuse thickening of the colon (arrows) and a small able findings.27 In their study, several different patterns of
amount of ascites (arrowheads). Segmental and diffuse colitis
have been described; the former has been associated with disease were identified, including: 1) a cryptogenic orga-
watery or bloody diarrhea, and the latter resulted in watery nizing pneumonia (COP)-like pattern (Fig. 3), 2)
diarrhea.
ground-glass opacities (Fig. 4), 3) an interstitial pattern,
4) a hypersensitivity pneumonitis-like pattern, and 5)
pneumonitis not otherwise specified. The imaging find-
Immune-Related Adverse Events ings present in pneumonitis may evolve over time. For
Over the course of therapy, unintended autoimmune- instance, cases that are characterized by a COP-like pat-
mediated complications may develop because of altera- tern may develop extensive ground-glass opacities. Others
tions in the host immune system. Complications related with a predominantly ground-glass pattern may develop
to immunotherapy are referred to as immune-related interstitial abnormalities. A study of patients with
adverse events and may be the result of the induction of nivolumab-induced pneumonitis described 4 patterns on
either autoimmunity or a proinflammatory state.20 These chest CT: COP, nonspecific interstitial pneumonia,
events tend to resolve with discontinuation of therapy, hypersensitivity pneumonitis, and acute interstitial

Cancer July 15, 2018 2917


Review Article

Figure 9. Imaging studies of pancreatitis are shown. (A) A contrast-enhanced, axial computed tomography (CT) scan from a
man aged 57 years demonstrates focal, low attenuation in the body of the pancreas (arrow) and inflammatory changes in the
adjacent peripancreatic fat (arrowhead). (B) A fused, axial 2-deoxy-2-(fluorine-18)fluoro-D-glucose (FDG)–positron emission
tomography/CT study from a woman aged 48 years reveals focal, increased FDG uptake in the body of the pancreas that corre-
sponds to a region of focal parenchymal enlargement on the unenhanced CT image (arrows, C.).

pneumonia/acute respiratory distress syndrome.28 The ground-glass opacities with consolidation in the depen-
COP-like pattern manifests with patchy ground-glass dent lung.30 Pneumonitis occasionally may result in the
opacities and consolidation in a subpleural, peribronchial, appearance of nodular opacities in the lungs; however,
or band pattern, which may be observed with the reversed none of these findings should be misinterpreted as recur-
halo sign. The nonspecific interstitial pneumonia pattern rent disease.
includes bilateral ground-glass opacities with reticular Sarcoid-like reaction is a rare immune-related
opacities, traction bronchiectasis or bronchiolectasis, and adverse event that can result in numerous small pulmo-
minimal or absent honeycombing in a basal distribution nary nodules in a perilymphatic distribution (along the
with subpleural sparing.29,30 The hypersensitivity pneu- bronchovascular bundles and in the subpleural regions)
monitis pattern results in centrilobular nodules and with or without ground-glass opacities and/or mediasti-
mosaic attenuation of the lung parenchyma because of air nal/hilar lymphadenopathy31 (Figs. 5 and 6). Involve-
trapping with an upper lobe-predominant distribution.30 ment of the airways is an uncommon complication of
Finally, the acute interstitial pneumonia/acute respiratory immunotherapy that tends to affect the trachea. Cardio-
distress syndrome pattern includes patchy, bilateral vascular abnormalities may be encountered, the most

2918 Cancer July 15, 2018


Immunotherapy and the Role of Imaging/Carter et al

on echocardiography and MR imaging.32-34 The most


common complication involving the thyroid gland is
thyroiditis, which may be identified on iodine-123 thy-
roid scintigraphy and/or ultrasound. On CT, the thyroid
gland is typically enlarged and demonstrates heteroge-
neously low attenuation (Fig. 7). Immune-related thy-
roiditis may be identified on FDG-PET/CT as diffusely
increased FDG uptake (Fig. 7).
Several immune-related adverse events may affect
the abdomen and pelvis, the most common of which
include colitis and hepatitis. Colitis is a significant clinical
complication that has the highest mortality of all
immune-related adverse events, and prolonged time to
diagnosis and management is associated with poor out-
comes.35 On CT and MR imaging, immune-related coli-
tis may manifest as segmental or diffuse wall thickening,
mucosal enhancement, submucosal edema, air-fluid lev-
els, infiltration of the pericolonic fat, and ascites (Fig. 8).
Segmental and diffuse patterns of colitis were observed
with ipilimumab treatment: the former manifested clini-
cally with watery or bloody diarrhea, and the latter
resulted in watery diarrhea36. Other adverse events that
may affect the gastrointestinal tract include perforation
and bleeding. Immune-related pancreatitis results in
imaging findings similar to those from acute pancreatitis
unrelated to immunotherapy. On CT, enlargement and
an edematous appearance of the pancreas with adjacent
fat stranding, edema, and free fluid are most common37
(Fig. 9). Increased FDG uptake may be present on FDG-
PET/CT38 (Fig. 9). Autoimmune hepatitis may result in
periportal or portal vein hyperechogenicity on ultrasound
and periportal edema and hypoattenuation of the edema-
tous liver parenchyma on CT.39 Involvement of the bili-
ary system (cholangiopathy) may manifest as thickening
Figure 10. Imaging studies of cholangiopathy are shown. (A) of the bile duct wall and/or biliary ductal dilatation (Fig.
A contrast-enhanced, coronal, reformatted computed tomog- 10). Other abnormalities, including endocrinopathies like
raphy (CT) scan from a woman aged 76 years reveals mild
thickening and enhancement of the common bile duct wall hypophysitis (Fig. 11) and adrenal insufficiency, also may
(arrow), with more extensive abnormality distally (arrow- be present. On MR imaging, hypophysitis manifests as
head). (B) A contrast-enhanced, coronal, reformatted CT
scan from a man aged 58 years reveals more extensive thick- mild-to-moderate, diffuse pituitary enlargement without
ening and enhancement of the common bile duct (arrow) as compression of the optic chiasm. Thickening of the pitui-
well as some involvement of the left intrahepatic bile ducts
(arrowhead). Immune-related cholangiopathy may manifest
tary gland is often present. After the administration of
as thickening of the bile duct wall and/or biliary ductal intravenous gadolinium contrast material, homogeneous
dilatation.
and heterogenous pituitary enhancement may be
observed.40

common of which is pericarditis, which can produce a FUTURE DIRECTIONS


pericardial effusion and/or pericardial thickening on CT. Although irRC, irRECIST, and iRECIST represent con-
Imaging findings suggestive of ventricular interdepen- tinual improvement over the conventional WHO and
dence and constrictive, effusive physiology (including RECIST criteria for evaluating response to immunother-
septal bounce and respiratory septal shift) may be present apy, limitations and challenges remain, and further

Cancer July 15, 2018 2919


Review Article

Figure 11. Imaging studies of hypophysitis are shown. (A) An axial 2-deoxy-2-(fluorine-18)fluoro-D-glucose (FDG)–positron emis-
sion tomographic study from a man aged 65 years who had melanoma reveals focal increased FDG uptake in the region of the
pituitary gland (arrow). (B,C) Contrast-enhanced, coronal and sagittal, T1-weighted magnetic resonance images from the same
patient demonstrate enlarged and enhancing infundibulum (arrow in B) and pituitary gland (arrow in C).

refinements are warranted. In this effort, there is ongoing In that study, there was a marked differential in granzyme
work to develop volumetric imaging, dynamic contrast- B expression between treated responders and nonrespond-
enhanced imaging, and molecular imaging for optimal ers. The study suggests that granzyme B PET imaging can
evaluation of immunotherapy response and immune- serve as a quantitatively useful predictive biomarker for
related adverse events. Several novel PET radiotracers efficacious responses to cancer immunotherapy. In
involving amino acids, nucleotides, choline, and S recep- another preclinical study, 89Zr-desferrioxamine–labeled
tor to detect cell proliferation or cell death are currently anti-CD8 cys-diabody (89Zr-malDFO-169 cDb)
being investigated.41 In addition, investigational use of immuno-PET was developed for the tracking of endoge-
MR imaging for depicting apoptosis and cell lysis to mon- nous CD8-positive T cells, and the investigators observed
itor tumor response to immunotherapy also has been that the technique could detect changes in systemic CD8-
reported.42 Other noninvasive imaging strategies are in positive tumor-infiltrating lymphocytes in murine tumor
various stages of development and implementation. immunotherapy models.46 In a different study involving
Immuno-PET imaging is a noninvasive, quantitative 64Cu-radiolabeled atezolizumab, specific binding to
method of imaging the whole body using radiolabeled tumor cells in vitro based on PET/CT imaging correlated
monoclonal antibodies that can provide a means to evalu- with PD-L1 expression levels.47 Other PET studies have
ate antibody drug pharmacokinetics and the expression of reported the feasibility of using peptide or small-molecule
cell surface markers of disease.43 Currently, immuno- compounds radiolabeled with gallium-68 or 111In to
PET imaging is performed using copper-64 (64Cu)- assess PD-L1 expression.48,49
labeled and zirconium-89 (89Zr)-labeled antibodies. The term radiomics has been used to describe the
Work has demonstrated that the detection of pro- extraction of advanced quantitative imaging features
grammed death 1/programmed death ligand 1 (PD-1/ related to the underlying molecular features and pheno-
PD-L1) and/or CTLA4 with immuno-PET, as well as type of tumors from imaging modalities like CT, PET/
single-photon emission CT (SPECT) radiolabeled with CT, and MR imaging. In contrast to the standard qualita-
indium-111 (In111), may improve patient identification, tive imaging characteristics of neoplasms that are rou-
stratification, and the early assessment of response.44 tinely evaluated on radiologic studies, advanced analytics
Recently, a preclinical study using granzyme B, a down- like texture analysis have the potential to uncover tumor
stream effector of tumoral cytotoxic T cells, distinguished features that cannot be identified by visual assessment and
treated responders from nonresponders with excellent can provide detailed characterization of the specific tissue
predictive ability.45 In addition, the group evaluated the or lesion of interest and the associated microenvironment.
clinical value of a granzyme B imaging paradigm in which A recent proof-of-concept study reported that radiomics
they analyzed biopsy specimens from patients with mela- predicted pneumonitis in patients who received immuno-
noma who were receiving checkpoint inhibitor therapy. therapy with an accuracy of 100%.50 Finally,

2920 Cancer July 15, 2018


Immunotherapy and the Role of Imaging/Carter et al

radiogenomics, the linkage between imaging phenotypes 11. Henze J, Maintz D, Persigehl T. RECIST 1.1, irRECIST 1.1, and
mRECIST: how to do [serial online]. Curr Radiol Rep. 2016;4:48.
and tumor genomics, may assist in the development of 12. Seymour L, Bogaerts J, Perrone A, et al. iRECIST: guidelines for
more robust stratification and endpoint imaging bio- response criteria for use in trials testing immunotherapeutics. Lancet
Oncol. 2017;18:e143-e152.
markers for molecular-targeted clinical trials. 13. Bohnsack O, Ludajic K, Hoos A. Adaptation of the immune-related
response criteria: irRECIST. Ann Oncol. 2014;25(suppl 4):iv361-
CONCLUSIONS iv372.
14. Hodi FS, Hwu WJ, Kefford R, et al. Evaluation of immune-related
The role of immunotherapy in treating patients with response criteria and RECIST v1.1 in patients with advanced mela-
cancer continues to expand. Therefore, it is essential that noma treated with pembrolizumab. J Clin Oncol. 2016;34:1510-
1517.
radiologists and other providers have a thorough under- 15. Bier G, Hoffmann V, Kloth C, et al. CT imaging of bone and bone
standing of the novel response criteria developed to eval- marrow infiltration in malignant melanoma—challenges and limita-
uate these patients, such as irRC, irRECIST, and tions for clinical staging in comparison to 18FDG-PET/CT. Eur J
Radiol. 2016;85:732-738.
iRECIST. In addition, because a wide variety of 16. Kong BY, Menzies AM, Saunders CA, et al. Residual FDG-PET
immune-related adverse events may affect patients who metabolic activity in metastatic melanoma patients with prolonged
response to anti-PD-1 therapy. Pigment Cell Melanoma Res. 2016;29:
receive immunotherapy, the prompt identification and 572-577.
reporting of such side effects is imperative. 17. Cho SY, Lipson EJ, Im HJ, et al. Prediction of response to immune
checkpoint inhibitor therapy using early-time-point F-FDG PET/CT
imaging in patients with advanced melanoma. J Nucl Med. 2017;58:
FUNDING SUPPORT 1421-1428.
No specific funding was disclosed. 18. Wong ANM, McArthur GA, Hofman MS, Hicks RJ. The advan-
tages and challenges of using FDG PET/CT for response assessment
in melanoma in the era of targeted agents and immunotherapy. Eur
CONFLICT OF INTEREST DISCLOSURES J Nucl Med Mol Imaging. 2017;44(suppl 1):67-77.
Brett W. Carter reports royalties from Elsevier, Inc. Wei T. Yang 19. Gilles R, de Geus-Oei LF, Mulders PFA, Oyen WJG. Immunother-
apy response evaluation with F-FDG-PET in patients with advanced
reports personal fees from Seno Medical outside the submitted
stage renal cell carcinoma. World J Urol. 2013;31:841-846.
work and royalties from Elsevier, Inc. Priya R. Bhosale made no 20. Kaufman HL, Kirkwood JM, Hodi FS, et al. The Society for Immu-
disclosures. notherapy of Cancer consensus statement on tumour immunother-
apy for the treatment of cutaneous melanoma. Nat Rev Clin Oncol.
2013;10:588-598.
REFERENCES 21. Ioannidis JP, Lau J. Completeness of safety reporting in randomized
1. Kwak JJ, Tirumani SH, Van den Abbeele AD, Koo PJ, Jacene HA. trials: an evaluation of 7 medical areas. JAMA. 2001;285:437-443.
Cancer immunotherapy: imaging assessment of novel treatment 22. Pitrou I, Boutron I, Ahmad N, Ravaud P. Reporting of safety results
response patterns and immune-related adverse events. Radiographics. in published reports of randomized controlled trials. Arch Intern
2015;35:424-437. Med. 2009;169:1756-1761.
2. Nishino M, Tirumani SH, Ramaiya NH, Hodi FS. Cancer immu- 23. Paoletti X, Le Tourneau C, Verweij J, et al. Defining dose-limiting
notherapy and immune-related response assessment: the role of radi- toxicity for phase 1 trials of molecularly targeted agents: results of a
ologists in the new arena of cancer treatment. Eur J Radiol. 2015;84: DLT-TARGETT international survey. Eur J Cancer. 2014;50:2050-
1259-1268. 2056.
3. Okada H, Weller M, Huang R, et al. Immunotherapy response 24. Peron J, Maillet D, Gan HK, et al. Adherence to CONSORT
assessment in neuro-oncology: a report of the RANO Working adverse event reporting guidelines in randomized clinical trials evalu-
Group. Lancet Oncol. 2015;16:e534-e542. ating systemic cancer therapy: a systematic review. J Clin Oncol.
4. Wolchok JD, Hoos A, Bohnsack O, et al. Guidelines for the evalua- 2013;31:3957-3963.
tion of immune therapy activity in solid tumors: immune-related 25. Sivendran S, Latif A, McBride RB, et al. Adverse event reporting in
response criteria. Clin Cancer Res. 2009;15:7412-7420. cancer clinical trial publications. J Clin Oncol. 2014;32:83-89.
5. Nishino M, Giobbie-Hurder A, Gargano M, Suda M, Ramaiya NH, 26. Weber JS, Dummer R, de Pril V, Lebbe C, Hodi FS; MDX010-20
Hodi FS. Developing a common language for tumor response to Investigators. Patterns of onset and resolution of immune-related
immunotherapy: immune-related response criteria using unidimen- adverse events of special interest with ipilimumab: detailed safety
sional measurements. Clin Cancer Res. 2013;19:3936-3943. analysis from a phase 3 trial in patients with advanced melanoma.
6. Postow MA, Wolchok JD. Ipilimumab: developmental history, clini- Cancer. 2013;119:1675-1682.
cal considerations and future perspectives. Melanoma Lett. 2012;30: 27. Naidoo J, Wang X, Woo KM, et al. Pneumonitis in patients treated
1-4. with anti-programmed death-1/programmed death ligand 1 therapy.
7. World Health Organization (WHO). WHO Handbook for Report- J Clin Oncol. 2017;35:709-717.
ing Results of Cancer Treatment. Offset Publication No. 48. 28. Nishino M, Ramaiya NH, Awad MM, et al. PD-1 inhibitor-related
Geneva, Switzerland: WHO; 1979. pneumonitis in advanced cancer patients: radiographic patterns and
8. Therasse P, Arbuck SG, Eisenhauer EA, et al. New guidelines to clinical course. Clin Cancer Res. 2016;22:6051-6060.
evaluate the response to treatment in solid tumors. J Natl Cancer 29. Hansell DM, Bankier AA, MacMahon H, McLoud TC, Muller NL,
Inst. 2000;92:205-216. Remy J. Fleischner Society: glossary of terms for thoracic imaging.
9. Eisenhauer EA, Therasse P, Bogaerts J, et al. New Response Evalua- Radiology. 2008;246:697-722.
tion Criteria in Solid Tumours: revised RECIST guideline (version 30. Travis WD, Costabel U, Hansell DM, et al. An official American Tho-
1.1). Eur J Cancer. 2009;45:228-247. racic Society/European Respiratory Society statement: update of the
10. Nishino M, Gargano M, Suda M, Ramaiya NH, Hodi FS. Optimiz- international multidisciplinary classification of the idiopathic intersti-
ing immune-related tumor response assessment: does reducing the tial pneumonias. Am J Respir Crit Care Med. 2013;188:733-748.
number of lesions impact response assessment in melanoma patients 31. Prieto PA, Yang JC, Sherry RM, et al. CTLA-4 blockade with ipili-
treated with ipilimumab [serial online]? J Immunother Cancer. 2014; mumab: long-term follow-up of 177 patients with metastatic mela-
2:17. noma. Clin Cancer Res. 2012;18:2039-2047.

Cancer July 15, 2018 2921


Review Article

32. Yun S, Vincelette ND, Mansour I, Hariri D, Motamed S. Late onset 41. Aarntzen EH, Srinivas M, Radu GC, et al. In vivo imaging of
ipilimumab-induced pericarditis and pericardial effusion: a rare but therapy-induced anti-cancer immune responses in humans. Cell Mol
life threatening complication [serial online]. Case Rep Oncol Med Life Sci. 2013;70:2237-2257.
2015:794842, 2015. 42. Lazovic J, Jensen MC, Ferkassian E, Aguilar B, Raubitschek A,
33. Kolla BC, Patel MR. Recurrent pleural effusions and cardiac tampo- Jacobs RE. Imaging immune response in vivo: cytolytic action of
nade as possible manifestations of pseudoprogression associated with genetically altered T cells directed to glioblastoma multiforme. Clin
nivolumab therapy—a report of 2 cases [serial online]. J Immunother Cancer Res. 2008;14:3832-3839.
Cancer. 2016;4:80. 43. Wu AM, Senter PD. Arming antibodies: prospects and challenges
34. Johnson DB, Balko JM, Compton ML, et al. Fulminant myocarditis for immunoconjugates. Nat Biotechnol. 2005;23:1137-1146.
with combination immune checkpoint blockade. N Engl J Med. 44. Bauckneht M, Piva R, Sambuceti G, Grossi F, Morbelli S. Evalua-
2016;375:1749-1755. tion of response to immune checkpoint inhibitors: is there a role for
35. Kim KW, Ramaiya NH, Krajewski KM, et al. Ipilimumab associated positron emission tomography? World J Radiol. 2017;9:27-33.
colitis: CT findings. AJR Am J Roentgenol. 2013;200:W468-W474. 45. Larimer BM, Wehrenberg-Klee E, Dubois F, et al. Granzyme B
36. O’Regan KN, Jagannathan JP, Ramaiya N, Hodi FS. Radiologic PET imaging as a predictive biomarker of immunotherapy response.
aspects of immune-related tumor response criteria and patterns of Cancer Res. 2017;77:2318-2327.
immune-related adverse events in patients undergoing ipilimumab 46. Tavare R, Escuin-Ordinas H, Mok S, et al. An effective immuno-
therapy. AJR Am J Roentgenol. 2011;197:W241-W246. PET imaging method to monitor CD8-dependent responses to
37. Bronstein Y, Ng CS, Hwu P, Hwu WJ. Radiologic manifestations of immunotherapy. Cancer Res. 2016;76:73-82.
immune-related adverse events in patients with metastatic melanoma 47. Lesniak WG, Chatterjee S, Gabrielson M, et al. PD-L1 detection in
undergoing anti-CTLA-4 antibody therapy. AJR Am J Roentgenol. tumors using [Cu]atezolizumab with PET. Bioconj Chem. 2016;27:
2011;197:W992-W1000. 2103-2110.
38. Alabed YZ, Aghayev A, Sakellis C, Van den Abbeele AD. Pancreati- 48. Mayer AT, Natarajan A, Gordon SR, et al. Practical immuno-PET
tis secondary to anti-programmed death receptor 1 immunotherapy radiotracer design considerations for human immune checkpoint
diagnosed by FDG PET/CT. Clin Nucl Med. 2015;40:e528-e529. imaging. J Nucl Med. 2017;58:538-546.
39. Kim KW, Ramaiya NH, Krajewski KM, et al. Ipilimumab associated 49. Chatterjee S, Lesniak WG, Miller ME, et al. Rapid PD-L1 detection
hepatitis: imaging and clinicopathologic findings. Invest New Drugs. in tumors with PET using a highly specific peptide. Biochem Biophys
2013;31:1071-1077. Res Commun. 2017;483:258-263.
40. Faje AT, Sullivan R, Lawrence D, et al. Ipilimumab-induced hypo- 50. Colen RR, Fujii T, Bilen MA, et al. Radiomics to predict
physitis: a detailed longitudinal analysis in a large cohort of patients immunotherapy-induced pneumonitis: proof of concept [published
with metastatic melanoma. J Clin Endocrinol Metab. 2014;99:4078- online ahead of print October 27, 2017]. Invest New Drugs. doi:
4085. 10.1007/s10637-017-0524-2.

2922 Cancer July 15, 2018

You might also like