09111912154317320.pdf

(319 KB) Pobierz
Beyond RECIST: Molecular and functional imaging techniques for evaluation of response to targeted therapy
Cancer Treatment Reviews 35 (2009) 309–321
Contents lists available at ScienceDirect
Cancer Treatment Reviews
HOT TOPIC
Beyond RECIST: Molecular and functional imaging techniques
for evaluation of response to targeted therapy
I.M.E. Desar a, * , C.M.L. van Herpen a,d , H.W.M. van Laarhoven a,d , J.O. Barentsz b,e ,
W.J.G. Oyen c,f , W.T.A. van der Graaf a,d
a Department of Medical Oncology, Radboud University Nijmegen Medical Centre, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
b Department of Radiology, Radboud University Nijmegen Medical Centre, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
c Department of Nuclear Medicine, Radboud University Nijmegen Medical Centre, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
article info
summary
Article history:
Received 19 August 2008
Received in revised form 21 November 2008
Accepted 3 December 2008
The development of targeted therapies is a major breakthrough in the treatment of cancer. By evoking
necrosis and cavitation, evaluation based on tumour size alone, as is done in the RECIST criteria, is no
longer an adequate method. New molecular and functional imaging techniques are developed. This
review focuses on the use of new imaging modalities for the evaluation of treatment response of pathway
based targeted therapies. First, the basic principles of functional and molecular imaging modalities are
briefly discussed. Thereafter, their clinical application in targeted therapies is correlated to the underlying
biological mechanism. In this way, the best method for response evaluation for a new agent can be
identified.
Keywords:
Cancer
Targeted therapy
Molecular imaging
Functional imaging
2008 Elsevier Ltd. All rights reserved.
Introduction
tial response (PR) as a reduction of at least 50% of the product of
these two diameters, progressive disease (PD) as an increase of
25% and stable disease as every response between PR and PD. In
2000, new evaluation criteria called RECIST (response evaluation
criteria in solid tumours) criteria were developed, to make tumour
evaluation easier. 2 RECIST is based on the sum of one-dimensional
measurements of the greatest diameter of the tumour and/ or
metastases. CR is defined by the complete absence of disease, PR
is defined as more than 30% decrease of the sum of these largest
diameters, PD as an increase of more than 20% of the sum of the
largest diameters and stable disease as all outcomes in between.
Thus, both methods are based on the assessment of the size of
the tumour or metastases. Although the RECIST criteria have some
limitations, for the evaluation of treatment response of solid tu-
mours to classic cytotoxic chemotherapy, they are widely applied
and well accepted. Several validation studies have been performed
for the classic cytotoxic tumour treatments. 3–8 However, the ques-
tion is whether these criteria are sufficient for the evaluation of re-
sponse to targeted therapies. The effects of the new therapeutic
modalities, such as angiogenesis inhibitors and anti-vascular ther-
apies, are more complex. Necrosis and cavitation without a change
in size are frequently observed. Thus, the effect of targeted therapy
is often underestimated by using tumour size based RECIST evalu-
ation. For example, single-agent treatment with sorafenib 9 and
bevacizumab 10 in metastatic renal cell cancer failed to achieve
significant objective response rates according to the RECIST crite-
ria, but did result in a significant increase in progression-free
The development of targeted therapies is a major breakthrough
in the treatment of cancer. Targeted therapies are designed to
interfere with specific aberrant biological pathways involved in
tumourigenesis. This is in contrast with the generalized cytotoxic
effects of standard chemotherapy. Various mechanisms relevant
in carcinogenesis are exploited by targeted therapies, such as angi-
ogenesis, cell growth signalling and apoptosis. Targeted therapy
has found its way in oncology, administered as, e.g. monoclonal
antibodies or tyrosine kinase inhibitors.
When evaluating response of tumours to anticancer treatment,
a reliable and standardised methodology is essential, not only in
clinical research but also in daily patient care. Therefore, more than
25 years ago, the World Health Organisation (WHO) criteria for tu-
mour evaluation were developed, based on tumour mass assess-
ment on CT or MRI. 1 According to these WHO criteria, the size of
the tumour was assessed by two perpendicular diameters. A com-
plete response (CR) was defined as total absence of disease, a par-
* Corresponding author. Tel.: +31 24 3610353; fax: +31 24 3540788.
E-mail addresses: i.desar@aig.umcn.nl (I.M.E. Desar), c.vanherpen@onco.umcn.nl
(C.M.L. van Herpen), h.vanlaarhoven@onco.umcn.nl (H.W.M. van Laarhoven),
J.barentsz@rad.umcn.nl (J.O. Barentsz), w.oyen@nucmed.umcn.nl (W.J.G. Oyen),
w.vandergraaf@onco.umcn.nl (W.T.A. van der Graaf).
d Tel.: +31 24 3610353; fax: +31 24 3540788.
e Tel.: +31 24 3617133; fax: +31 24 36540866.
f
0305-7372/$ - see front matter 2008 Elsevier Ltd. All rights reserved.
doi:10.1016/j.ctrv.2008.12.001
Tel.: +31 24 3610972; fax: +31 24 3618942.
207376045.002.png 207376045.003.png 207376045.004.png
310
I.M.E. Desar et al. / Cancer Treatment Reviews 35 (2009) 309–321
survival (PFS), demonstrating its clinical efficacy. A striking exam-
ple underlining the potential of molecular imaging is the use of
positron emission tomography (PET) with [F-18] fluorodeoxyglu-
cose (FDG) in gastrointestinal stromal tumours (GIST) treated with
imatinib, in which FDG–PET responses preceded the anatomical re-
sponse on CT by several weeks. 11 Response on FDG–PET was asso-
ciated with longer PFS. After comparing FDG–PET results and CT
results in patients with GIST treated with imatinib, Choi et al. pro-
posed modified CT criteria based on both tumour size as tumour
density. PR is defined as a decrease in size of P10% or a decrease
in tumour density (HU: Hounsfield units) of P15% on CT. PD is de-
fined as an increase in tumour size of P10% and no decrease in tu-
mour density as defined in the criteria for PR. 12 The addition of
tumour density makes it possible to assess tumour necrosis. Be-
sides the difficulty regarding the lack of volume changes, CT alone
is not able to differentiate between vital tumour tissue or, for
example, fibrotic tissue. Furthermore, metabolic changes can pre-
cede volume changes, which can be detected by different molecu-
lar and functional imaging techniques. Depicting these early
changes can help to choose the right treatment strategy, including
the ability to prevent unnecessary long treatment courses with
their inherent adverse events and, specifically in the case of expen-
sive targeted therapies, their costs.
To meet the need for better tools to assess the effects of targeted
therapy, several options can be envisioned. Besides the develop-
ment of new criteria for conventional imaging methods, such as
the Choi criteria for GIST, biomarkers and new functional and
molecular imaging technologies are being introduced. Functional
and molecular imaging implies the quantitative measurement of
physiologic (functional) and molecular events in tumours, which
is now possible utilizing non-invasive new imaging modalities,
radiopharmaceuticals and contrast agents. 13
This review focuses on the use of new imaging modalities for
the evaluation of response to treatment with targeted therapies.
First, the basic principles of functional and molecular imaging
modalities are briefly discussed. Thereafter, their clinical applica-
tion in the evaluation of treatments response to targeted therapies
is correlated with the underlying biological mechanism. In this
way, one can more easily identify the best methods for response
evaluation for a group of drugs. When drugs have multiple targets,
the most dominant pathway is chosen. Only human studies are
discussed.
Figure 1. Parameters as defined by Tofts et al.
a standard set of quantity names and symbols. These include: (1)
a volume transfer constant K trans (min 1 ) also known as wash-in
rate, (2) the volume of the extravascular space (EES) per unit vol-
ume of tissue m e and (3) the flux rate constant between EES and
plasma k ep (min 1 ), the wash-out rate. The rate constant is the ra-
tio of the transfer constant to the EES (k ep = K trans / m e ). Lower values
of k ep or K trans can indicate (1) lower perfusion, (2) lower perme-
ability and/or (3) a smaller blood vessel surface area ( Fig. 1 ).
DCE-MRI has been shown to have diagnostic value in various
tumours and correlates with therapy effect of standard chemother-
apy and radiotherapy.
Diffusion weighted imaging
Diffusion weighted MRI (DWI or DW-MRI) is a unique MRI
alone technique that depicts image contrast that is related to the
thermal motion of water molecules. In biologic tissues the move-
ment of water molecules is restricted because their motion is mod-
ified and limited by interactions with cell membranes and
macromolecules. There is an inverse correlation between the de-
gree of restriction of motion of the water and the tissue cellularity
and integrity of cell membranes. 19–22 This means that in a tumour
with high cellularity, the motion of water molecules is more re-
stricted. On the other hand, when a tumour has a high glandular
component or has significant necrosis, there is less restriction of
motion. The apparent diffusion coefficient (ADC; unit mm 2 /s) is
the quantitative parameter used for the assessment of tissue diffu-
sion. The word ‘‘apparent” in this notation is being used to indicate
that diffusion in tissues is not free. A low ADC reflects restricted
diffusion. In general, treatments inducing apoptosis (e.g. chemo-
therapy) results in increased ADC values due to cell swelling, tu-
mour lysis and necrosis. 23 Statistically significant increases in
ADC values can be seen within a few days. However, the duration
of raised ADC values tends to be short because tissue dehydration
following cell death subsequently decreases ADC value 24 this is un-
like K trans changes which are persistently reduced following suc-
cessful therapy.
Molecular and functional imaging techniques
Magnetic resonance imaging (MRI)
MRI is a non-invasive imaging technique, which makes use of
strong magnetic fields. It has a good depth penetration and is able
to provide high-resolution anatomical information, without radia-
tion safety issues. By using specific MR techniques functional infor-
mation on tumour perfusion, 14 vascular permeability, 15 vascular
volume and flow, extracellular space tortuosity 14 and hypoxia 16
can be obtained.
Dynamic contrast enhanced MRI
Dynamic contrast enhanced MRI (DCE-MRI) is performed after
injection of a contrast agent. Usually the low molecular weight
contrast agents (e.g. gadopentetate dimeglumine, Gd-DTPA) are
used. MR sequences can be designed to be sensitive to the vascular
phase of contrast medium delivery, so-called T2 methods. From
these images data on tissue perfusion and blood volume can be ex-
tracted. By using sequences sensitive to the presence of contrast
medium in the extravascular extracellular space, so-called T1
methods, information on microvessel perfusion, permeability and
extracellular leakage space is obtained. 17 Tofts et al. 18 described
Magnetic resonance spectroscopy
Magnetic resonance spectroscopy (MRS) provides chemical
information about tissue metabolites. 25 Just like MRI MRS makes
use of the properties of certain nuclear isotopes (e.g. hydrogen 1
( 1 H), phosphorus 31 ( 31 P), fluorine 19 ( 19 F), carbon 13 ( 13 C), deute-
rium ( 2 H), tritium ( 3 H) or sodium 23 ( 23 Na)). When placed in a
strong magnetic field these nuclei start to resonate at a specific fre-
quency, which can be measured by MRS. The application of such a
strong magnetic field induces electronic currents in atoms and
molecules producing further small magnetic fields. The size of
these small fields depends on the (electronic) environment of the
specific nucleus. Different chemical environments give rise to sig-
207376045.005.png
I.M.E. Desar et al. / Cancer Treatment Reviews 35 (2009) 309–321
311
nals at different frequencies. Thus, a MR spectrum is a kind of
molecular fingerprint and may contain many resonance frequen-
cies or peaks, one for each chemically distinguishable site or group
in a given metabolite or for specific sites in a variety of metabolites.
and an inefficient metabolism, which is the molecular basis for
FDG-positive cancer lesions on PET scans. However, high glucose
metabolism is not specific for cancer. Normal organs, such as the
brain, 26 and in some patients the heart 27 accumulate FDG, as do
activated white blood cells in inflammatory lesions. 28 Besides the
use of FDG–PET for characterizing, staging and restaging of tu-
mours, the application of FDG–PET as a prognostic marker and an
(early) predictor of therapy response after conventional chemo-
therapy was evaluated. Review of these data is beyond the scope
of this study. Excellent reviews have been published before (e.g.
[29,30] ).
One of the challenges of using FDG–PET in clinical studies and
daily practice is the need for standardization and validated re-
sponse criteria. In 1999, the EORTC proposed a set of criteria to
determine PET response. In 2006, a workshop of the National Can-
cer Institute made recommendations to develop a standard FDG–
PET protocol for NCI-sponsored clinical trials to assess treatment
efficacy. In these recommendations was stated that there is no
one best methodology for obtaining or analyzing FDG–PET scans,
nor is there one agreed-on standard for judging the significance
of a response seen on FDG–PET. The timing of the post treatment
imaging was recommended two weeks after the end of a specific
chemotherapy cycle, although the exact timing could depend on
the frequency and duration of therapy. 31 An ENASCO working
group is now working on the revision of these standardised re-
sponse criteria for FDG–PET in solid tumours. Driven by nation-
wide multicenter trials, a Dutch multidisciplinary group developed
extensive recommendations for further standardization of PET
acquisition and processing (Boellaard et al., EJNMMI 2008, Epub
ahead of print). Recently, an International Harmonization Project
(IHP) was convened to discuss standardization of clinical trial
parameters in lymphoma. 32 It was considered adequate to rely
on visual assessment alone for interpreting PET findings as positive
or negative when assessing response after therapy in lymphoma. In
solid tumours, however, only visual assessment is insufficient. At
present a wide range of quantitative criteria based on SUVs has
been proposed. The most often applied SUV reduction of 35% for
a minor response and 60% for a major response seem reasonable.
These criteria should also allow to combine the metabolic changes
with volumetric changes, as can be obtained with a integrated
FDG–PET-CT. Validated and generally accepted quantitative crite-
ria to assess metabolic response using SUV are urgently required.
This is the only way to make sure that future evaluations of (tar-
geted) therapy by FDG–PET will be reproducible and validated.
In summary, FDG–PET has proven to be an important improve-
ment in cancer imaging including its use for the evaluation of
treatment response to conventional chemotherapy. Its role in the
evaluation of targeted therapy is less clear. Available human data
will be discussed in the pathway based imaging section.
Computed tomography (CT)
Contrast enhanced CT is the most commonly used imaging
technique in oncology, as is it widely available, fast and conve-
nient. It provides a high anatomical resolution and by using the
Hounsfield units (HU) it gives information about tissue density.
However, it lacks the ability to provide biochemical and physiolog-
ical information.
Dynamic contrast enhanced perfusion CT
Dynamic contrast enhanced CT (CTP) is also called functional
CT, dynamic CT or perfusion CT. CTP can provide information about
blood flow, blood volume, capillary permeability and micro vessel
density. After an intravenous bolus of conventional iodinated con-
trast, a series of images is performed. There is a linear relation be-
tween the concentration of contrast agent and the attenuation
numbers (expressed in HU). The used parameter is the standard-
ised perfusion value (SPV), defined as the ratio of tumour perfusion
to whole body perfusion.
Radionuclide imaging
PET and single-photon emission computed tomography (SPECT)
are both radionuclide imaging techniques. PET is based on a unique
feature of positron emitting radionuclides: upon annihilation of a
positron with an electron two 512 keV gamma rays are emitted
with an angle of 180. A ring of detectors is used to detect these
gamma rays, allowing three-dimensional image reconstruction.
Standard acquisition protocols provide whole body images, which
allow convenient quantification of all visualized lesions, expressed
as standardised uptake values (SUV). More advanced, dynamic
imaging allows absolute quantization of tracer uptake and kinetics
over time. PET-CT enables the assessment of molecular character-
istics as depicted by PET with anatomical structures on CT. Cur-
rently, efforts are being made to construct combined PET–MRI
scanners to further enhance the potential of multimodality
imaging.
For PET, a wide variety of radiopharmaceuticals are available,
encompassing specific molecular features of tumours and normal
tissues, e.g. metabolism (FDG), proliferation (FLT), amino acid up-
take and protein synthesis (e.g. FET), hypoxia (e.g. F-MISO), perfu-
sion and blood volume ( 15 O-water, 11 C-carbon monoxide).
Furthermore, the recently introduced use of positron emitting
radionuclides for labelling of monoclonal antibodies (immunoPET),
peptides and other receptor-targeting compounds, has broadened
the horizon for mechanistic studies.
SPECT involves the use of radionuclides that emit single gamma
rays. In contrast to PET, SPECT generally has lower sensitivity and
quantification of SPECT is more challenging. Several biological
compounds such as antibodies and peptides can readily be labelled
with single photon-emitting radionuclides, such as indium-111,
radioiodine and Tc-99m.
Fluoro- L -thymidine PET (FLT-PET)
The thymidine analogue FLT accumulates in proliferating tis-
sues. FLT is transported across cell membranes by nucleoside
transporter proteins. Once intracellular, FLT is phosphorylated by
thymidine kinase I. Similarly to FDG, the phosphorylated FLT is
trapped intracellularly, but is not further incorporated into DNA.
Thereby, FLT-PET uptake is a marker for proliferation.
FDG–PET
FDG is by far the most commonly used radiopharmaceutical for
PET. FDG is transported into the cell by glucose transporters,
mainly glucose transporter-1 (GLUT-1). Thereafter, FDG is phos-
phorylated. As FDG-6-phosphate is not further metabolized it is
as such irreversibly trapped inside the cell. Thus, FDG uptake on
PET reflects the metabolic activity of cells. Cancer cells are known
to have higher rates of glucose uptake, due to increased demand
F-MISO-PET
This tracer has been used to image tissue hypoxia in human tu-
mours. F-MISO accumulates in tissue by binding macromolecules
when pO 2 < 10 mm Hg. F-MISO is only sensitive to hypoxia in via-
ble cells, not in necrosis.
In lung tumours, F-MISO was able to monitor the changing hy-
poxia during radiotherapy. 33
In head and neck cancer, 34 F-MISO
312
I.M.E. Desar et al. / Cancer Treatment Reviews 35 (2009) 309–321
uptake was correlated to poor outcome to chemotherapy and
radiotherapy.
DCE-MRI parameters (namely K ep and K trans ) was observed, indi-
cating a drop in tumour blood volume and/or perfusion and/or
permeability. However, these changes were not significantly cor-
related to clinical response in most cancers except perhaps renal
cell cancer. 41–43 Targeted therapies directed to VEGFR or down
stream signalling have also been evaluated by DCE-MRI. Unfortu-
nately, different MRI protocols and different methods for quanti-
fication have been used. Overall, in most studies dose dependent
decreases in tumour blood perfusion, or tumour blood flow are
found, even in phase I studies when the tumour microvasculature
would be expected relatively resistant. Although changes in DCE-
MRI parameters can occur as early as 24 h after initiation of the
therapy, such changes are not always reliable for all anti-VEGF
therapies because of the transient phenomenon of vascular nor-
malization when regional increases in blood flow can be detected
– this is not always observed. ( Table 1 ) Since vascular normaliza-
tion is of short duration, vascular collapse using anti-VEGF strat-
egies is most reliably detected after two weeks. From the two
registered multi kinase VEGFR inhibitors, sorafenib and sunitinib,
sorafenib is most extensively evaluated by molecular and func-
tional imaging techniques. In total more than 220 patients, in
nine studies 44–52 were treated with sorafenib (some of them com-
bined with bevacizumab 44 ). Four studies showed decreases in
DCE-MRI parameters corresponding with clinical treatment out-
come. 44–47 Remarkably, in the combined sorafenib–bevacizumab
study, no significant changes were found at first evaluation of sin-
gle-agent therapy (4 weeks after start of treatment) while after
the following combination therapy in the same patients DCE-
MRI parameters, especially K trans , decreased. 44 Suninitib has not
been evaluated by DCE-MRI so far. Of the other VEGFR inhibitors,
vatalanib, semaxanib and cediranib, have been most intensive
evaluated by DCE-MRI. Vatalanib and cediranib showed decreases
in DCE-MRI parameters as early as 2 days after start of treatment.
In case of vatalanib, this decrease was dose dependent and corre-
lated with clinical outcome. 53–56 However, vatalanib was unsuc-
cessful in phase III combination studies indicating that apparent
success as a single agent in early phase clinical studies may not
translate into ultimate clinical efficacy. The DCE-MRI results for
semaxanib in four studies are conflicting ( Table 1 ). For the other
VEGF/PDGF pathway agents only single DCE-MRI studies with
limited numbers of patients have been published. Disparate re-
sults with VEGF/PDGF inhibition on DCE-MRI maybe due to
antagonistic effects of anti-permeability due to VEGF inhibition
and pro-permeability effects of PDGF inhibition. This indicates
that anti-angiogenic therapies do not always result in reductions
of blood flow in the short term and that DCE-MRI kinetic re-
sponse relationships are not universally strong across all tissue
sites for all drugs.
CTP showed a decrease in tumour perfusion and blood volume
7–12 days of start of treatment with bevacizumab. 57,58 These stud-
ies did not correlate imaging findings to clinical outcome. In ad-
vanced carcinoids on stable octreotide doses, patients were
randomly assigned to bevacizumab or peg-IFN- a 2b. CTP showed
a significant decrease in tumour blood flow, respectively, 2 days
and 18 weeks after start of bevacizumab compared to interferon.
Only the day 2 CTP results of bevacizumab were related to a longer
PFS. 59 Cediranib, 60,61 SU6668, 62 sorafenib and sunitinib 63 have also
been studied by CTP and showed a drop in tumour blood flow and
tumour blood volume. The decrease in tumour blood flow was
predictive for treatment response in case of sorafenib and sunitinib
treatment. 63
Only four studies used ultrasound for the evaluation of treat-
ment response. The observed decrease in vascular density in pa-
tients treated with semaxanib did not correlate with clinical
outcome, 64 while responders to sorafenib as assessed by DCE-US
proved to have a better PFS. 48 Similar DCE-US results were found
15 O-water-PET
15 O is able to bind to hydrogen or carbogen, thereby providing
two important tracers: 15 O-water and C 15 O. 15 O-water-PET allows
determination of blood flow perfusable tissue fraction, and volume
of distribution with high spatial resolution.
Radiolabeled receptor-targeting ligands
Several radiopharmaceuticals have been developed for PET and
SPECT imaging of receptor and antigen expression on tumour cells.
Aims of these techniques are for example to monitor VEGF expres-
sion or to evaluate the tumour response to drugs as bevacizumab 35
and anti-angiogenic drugs. Examples are 123 I-VEGF and 111 In-hnTf-
VEGF (drug target: VEGFRs), 111 In-trastuzumab (drug target:
HER2), 111 In-EGF (drug target EGFRs), 18 F-fluoro-oestradiol and
radiolabelled somatostatin analogues. 36,37
Ultrasound (US)
Ultrasound uses waves to visualize body organs and blood-flow
velocity, based upon the fact that the speed of sound through tis-
sue varies with tissue density and that the accompanying echoes
can reveal these differences. The Doppler technique makes it pos-
sible to assess blood flow. US is frequently used in the oncologic
clinical practice.
Dynamic contrast enhanced US
US contrast agents, such as microbubbles, nanoparticles or per-
fluorcarbon gas, alter wave absorption and reflection. This en-
hances the intensity of the signals bouncing back from the
tissues. This provides morphologic and physiologic information,
without biochemical information.
Molecular and function imaging for response evaluation to
targeted therapy
Several molecular and functional imaging techniques have been
used for the evaluation of targeted therapy. Frequently, this has
been done within phase I or early phase II trials. As a result, num-
bers of patients are low, while the diseases and treatment or imag-
ing protocols are heterogeneous. Here we summarize the literature
on this.
VEGFR/PDGFR pathway
VEGF and its receptors play a pivotal role in both normal and
pathological malignant angiogenesis. Activation of the VEGFR
pathway leads to endothelial cell activation, proliferation and sur-
vival, degradation of the basement membrane which is necessary
for endothelial cell migration and invasion, increased vascular per-
meability and mobilization of endothelial progenitor cells (EPCs)
from the bone marrow into the peripheral circulation. 38 VEGF is
expressed in response to hypoxia, oncogenes or cytokines. The
VEGFR pathway is considered to be the most important and best
explored pathway in angiogenesis of tumours. The platelet derived
growth factor receptor (PDGFR) pathway is also important for angi-
ogenesis. However, the exact mechanism remains to be eluci-
dated. 39 PDGF is important for the recruitment of pericytes,
which are required for microvessel stability. 40
DCE-MRI is the most frequently used functional imaging tech-
nique for the evaluation of this pathway. Usually as a pharmaco-
dynamic biomarker in early phase, single-agent studies. After
start of VEGF antibody therapy, i.e. bevacizumab, a decrease of
I.M.E. Desar et al. / Cancer Treatment Reviews 35 (2009) 309–321
313
Table 1
VEGFR/PDGFR.
Reference Drug
Target
Tumour
Technique n Result
[98]
Axitinib
VEGFR, PDGFR, KIT Solid cancer DCE-MRI 17 Decrease in K trans and AUC on day 2, inversely proportional to axitinib
exposure
[99]
ABT869
VEGFR, PDGFR, FLT-3 Solid cancer DCE-MRI 15 Permeability-surface area product and distributed parameters model shows
better correlation with drug exposure than K trans
[42]
Bevacizumab
(+chemotherapy)
VEGF
Breast cancer DCE-MRI 18 Decrease in K trans , k ep and v e after 3, 12, 21 weeks. No correlation with
response
[41]
Bevacizumab
(+chemotherapy)
VEGF
Breast cancer DCE-MRI 19 Decrease in K trans , k ep and AUC
[100]
Bevacizumab
(+chemotherapy)
VEGF
GBM
DCE-MRI 20 Changes in K trans value were highly correlated with the % decline in tumour
volume after 1 cycle but not correlated to treatment outcome
[58]
Bevacizumab
VEGF
CRC
CTP
12 Decrease in HU values and perfusion score
[59]
Bevacizumab
VEGF
carcinoid
CTP
12 Significant decrease in tumour blood flow after 2 days and after 18 weeks of
treatment compared to IFN- a control group
[65]
Bevacizumab
VEGF
HCC
DCE-US
42 Decrease in AUC, peak intensity, AUC wash in and out. DCE-US on day 3 and 8
predicts the treatment response after 4 months
[57]
Bevacizumab (+
radiotherapy)
VEGF
Rectal cancer CTP, FDG–
PET
6 Decrease in tumour blood perfusion and blood volume. Decreased FDG
uptake after 6 weeks
[69]
Bevacizumab
(+chemotherapy)
VEGF
GBM
FLT-PET
19 Metabolic response in 47%, predictive for overall survival
[68]
Bevacizumab
(+chemotherapy)
VEGF
GBM
FDG–PET 35 6/35 patients completed one year treatment who had a hypometabolic FDG–
PET result
[66]
Bevacizumab
(+chemotherapy)
VEGF
CRC
FDG–PET 7 Better correlation with pathologic response for FDG–PET compared to CT
[67]
Bevacizumab
(+chemotherapy)
VEGF
Liver
metastasis of
CRC
FDG–PET-
CT
11 Decrease in median SUV max from 8 (baseline) to 4 (after 1 cycle). Decrease of
SUV max in responder group >50% compared to baseline
[101]
BIBF 1120
VEGFR, PDGFR, FGFR Hepatic
metastasis of
solid cancer
DCE-MRI 7 Median decrease of 15% in hepatic perfusion index after 28 days
[102]
CDP860
PDGFR
CRC, ovarian
cancer
DCE-MRI 8 No effects on iAUC and K trans . In 3/8 patients increased vascularised tumour
volume on day 1
[103]
Cediranib
VEGFR, PDGFR, c-KIT Solid cancer DCE-MRI 32 Reduction in iAUC60, less on day 2 compared to day 28 and 56
[104]
Cediranib
VEGFR, PDGFR, c-KIT Prostate cancer DCE-MRI 10 Decrease in K trans and k ep , consistent with response
[105]
Cediranib
VEGFR, PDGFR, c-KIT GBM
DCE-MRI,
DWI
16 Decrease in K trans and ADC, starting on day 1
[60]
Cediranib
VEGFR, PDGFR, c-KIT Solid cancer CTP
6 Reduction in perfusion and permeability surface product
[61]
Cediranib and
Gefetinib
VEGFR, PDGFR, c-
KIT, EGFR-1
Solid cancer CTP
13 Initial decrease in perfusion.
[43]
HuMV833
VEGF
Solid cancer DCE-MRI,
HuMV833-
PET
20 Decrease in k first pass after 48 h. No changes in HuMV833-PET
[106]
IMC-1C11
VEGFR
CRC
DCE-MRI 11 Decrease in K in and EF after 4 weeks
[107]
Semaxanib
VEGFR, PDGFR, KIT Solid cancer DCE-MRI 17 No significant changes in K trans and v e
[108]
Semaxanib
VEGFR, PDGFR, KIT Solid cancer DCE-MRI 11 Increase in k ep , wide variations
[109]
Semaxanib
VEGFR, PDGFR, KIT Melanoma
DCE-MRI 5 Decrease in tumour perfusion
[110]
Semaxanib
VEGFR, PDGFR, KIT Solid cancer DCE-MRI 11 Decrease in AUC60, AUC90 and AMX
[111]
Semaxanib
VEGFR, PDGFR, KIT NSCLC
DCE-MRI 35 Significant reduction in K trans of 35–38% after 2 and 28 days. No significant
correlation with clinical outcome
[112]
Semaxanib
VEGFR, PDGFR, KIT RCC
FDG–PET,
H 2 15 O-PET
5 No consistent findings
[64]
Semaxanib
VEGFR, PDGFR, KIT Head and neck
cancer
Power
doppler US
7 Decrease in vascular density, not corresponding with clinical outcome
[113]
Semaxanib
VEGFR, PDGFR, KIT CRC
DCE-US
2 Reduced tumour perfusion in stable disease, increased tumour perfusion in
progressive disease
[45]
Sorafenib
Raf-1, B-raf, VEGFR,
PDGFR, FLT-3, C-KIT
RCC
DCE-MRI 16 Decline in K trans and v e . High K trans at baseline and percentage decline of K trans
correlated with time to progression
[114]
Sorafenib
Raf-1, B-raf, VEGFR,
PDGFR, FLT-3, C-KIT
RCC
DCE-MRI 48 iAUC90 and K trans are markers for progression. High variability. Changes in
DCE-MRI parameters were not predictive for PFS
[47]
Sorafenib
Raf-1, B-raf, VEGFR,
PDGFR, FLT-3, C-KIT
RCC
DCE-MRI 17 Decrease in K trans . Association between PFS-baseline K trans and percentage
K trans decline
[48]
Sorafenib
Raf-1, B-raf, VEGFR,
PDGFR, FLT-3, C-KIT
RCC
DCE-US
9 4/9 good responders, correlated with better PFS
[49]
Sorafenib
Raf-1, B-raf, VEGFR,
PDGFR, FLT-3, C-KIT
RCC
FDG–PET-
CT
10 Decrease in mean glucose uptake after 1 month
[50]
Sorafenib
Raf-1, B-raf, VEGFR,
PDGFR, FLT-3, C-KIT
HCC
FDG–PET 6 Early dynamic changes in FDG uptake seem to be predictive for clinical
outcome
[44]
Sorafenib and
bevacizumab
Raf-1, B-raf, VEGFR,
PDGFR, FLT-3, C-KIT,
VEGF
Solid cancer DCE-MRI,
FDG–PET
38 Reduction of tumour vessel permeability and blood flow in patients with
stable disease or minor regression. No significant changes in PET-SUV
[51]
Sorafenib,
sunitinib,
interferon or
placebo
Raf-1, B-raf, VEGFR,
PDGFR, FLT-3, C-KIT
RCC
CTP
44 26/44 patients with sorafenib or sunitinib. In these patients significant drop
in tumour blood flow and tumour blood volume after 6 weeks. Tumour blood
flow predictive for response
[52]
Sorafenib or
sunitinib
Raf-1, B-raf, VEGFR,
PDGFR, FLT-3, C-KIT
RCC
DCE-US
36 DCE-US at day 15 is able to predict efficacy at 3 months and correlated to PFS
(continued on next page)
207376045.001.png
Zgłoś jeśli naruszono regulamin