ArticlePDF Available

Chemotherapy-induced Cardiac 18 F-FDG Uptake in Patients with Lymphoma: An Early Metabolic Index of Cardiotoxicity?

Authors:
  • Universidade Federal do Pernambuco

Abstract

Background: It is uncertain whether myocardial fluorodeoxyglucose uptake occurs solely due to physiological features or if it represents a metabolic disarrangement under chemotherapy. Objective: To investigate the chemotherapy effects on the heart of patients with lymphoma by positron emission tomography associated with computed tomography scans (PET/CT) with 2-deoxy-2[18F] fluoro-D-glucose (18F-FDG PET/CT) before, during and/or after chemotherapy. Methods: Seventy patients with lymphoma submitted to 18F-FDG PET/CT were retrospectively analyzed. The level of significance was 5%. 18F-FDG cardiac uptake was assessed by three measurements: left ventricular maximum standardized uptake value (SUVmax), heart to blood pool (aorta) ratio, and heart to liver ratio in all the exams. Body weight, fasting blood sugar, post-injection time, and the injected dose of 18F-FDG between the scans were also compared. Results: Mean age was 50.4 ± 20.1 years and 50% was female. The analysis was carried out in two groups: baseline vs. interim PET/CT, and baseline vs. post-therapy PET/CT. There was no significant difference in clinical variables or protocol scans variables. We observed an increase in left ventricular (LV) SUVmax from 3.5±1.9 (baseline) to 5.6±4.0 (interim), p=0.01, and from 4.0±2.2 (baseline) to 6.1±4.2 (post-therapy), p<0.001. A percentage increase ≥30% of LV SUVmax occurred in more than half of the sample. The rise of cardiac SUV was accompanied by an increase in LV SUVmax/Aorta SUVmax and LV SUVmean/Liver SUVmean ratios. Conclusion: This study showed a clear increase in cardiac 18F-FDG uptake in patients with lymphoma during and/or after chemotherapy. The literature corroborates with these findings and suggests that 18F-FDG PET/CT is a sensitive and reliable imaging exam to detect early metabolic signs of cardiotoxicity.
Arq Bras Cardiol. 2021; [online].ahead print, PP.0-0
Original Article
Chemotherapy-induced Cardiac 18F-FDG Uptake in Patients with
Lymphoma: An Early Metabolic Index of Cardiotoxicity?
Mayara L. C. Dourado,1 Luca T. Dompieri,2 Glauber M. Leitão,³ Felipe A. Mourato,4 Renata G. G. Santos,4
Paulo J. Almeida Filho,4 Brivaldo Markman Filho,1 Marcelo D. T. Melo,5 Simone C. S. Brandão1
Departamento de Pós-Graduação em Ciências da Saúde, Universidade Federal de Pernambuco,1 Recife, PE – Brazil
Faculdade de Medicina, Universidade Federal de Pernambuco,2 Recife, PE – Brazil
Serviço de Oncologia, Hospital das Clínicas/Universidade Federal de Pernambuco,3 Recife, PE – Brazil
Real Nuclear, Real Hospital Português,4 Recife, PE – Brazil
Departamento de Medicina Interna, Universidade Federal da Paraíba,5 João Pessoa, PB – Brazil
Mailing Address: Simone Cristina Soares Brandão
Departamento de Medicina Nuclear – Hospital das Clínicas – Universidade
Federal de Pernambuco – Rua Professor Moraes Rego, 1235.
Postal Code 50670-901, Recife, PE – Brasil
E-mail: sbrandaonuclearufpe@gmail.com
Manuscript received May 27, 2021, revised manuscript August 04, 2021,
accepted September 01, 2021
DOI: https://doi.org/10.36660/abc.20210463
Abstract
Background: It is uncertain whether myocardial fluorodeoxyglucose uptake occurs solely due to physiological features
or if it represents a metabolic disarrangement under chemotherapy.
Objective: To investigate the chemotherapy effects on the heart of patients with lymphoma by positron emission
tomography associated with computed tomography scans (PET/CT) with 2-deoxy-2[18F] fluoro-D-glucose (18F-FDG
PET/CT) before, during and/or after chemotherapy.
Methods: Seventy patients with lymphoma submitted to 18F-FDG PET/CT were retrospectively analyzed. The level
of significance was 5%. 18F-FDG cardiac uptake was assessed by three measurements: left ventricular maximum
standardized uptake value (SUVmax), heart to blood pool (aorta) ratio, and heart to liver ratio in all the exams.
Body weight, fasting blood sugar, post-injection time, and the injected dose of 18F-FDG between the scans were also
compared.
Results: Mean age was 50.4 ± 20.1 years and 50% was female. The analysis was carried out in two groups: baseline
vs. interim PET/CT, and baseline vs. post-therapy PET/CT. There was no significant difference in clinical variables or
protocol scans variables. We observed an increase in left ventricular (LV) SUVmax from 3.5±1.9 (baseline) to 5.6±4.0
(interim), p=0.01, and from 4.0±2.2 (baseline) to 6.1±4.2 (post-therapy), p<0.001. A percentage increase ≥30% of
LV SUVmax occurred in more than half of the sample. The rise of cardiac SUV was accompanied by an increase in LV
SUVmax/Aorta SUVmax and LV SUVmean/Liver SUVmean ratios.
Conclusion: This study showed a clear increase in cardiac 18F-FDG uptake in patients with lymphoma during and/or
after chemotherapy. The literature corroborates with these findings and suggests that 18F-FDG PET/CT is a sensitive
and reliable imaging exam to detect early metabolic signs of cardiotoxicity.
Keywords: Cardiotoxicity; Chemotherapy; Lymphoma.
Introduction
Chemotherapy and radiotherapy-induced cardiotoxicity
(CTX) encompasses various forms of injury to the cardiovascular
system, that trigger an increased production of reactive
oxygen (ROS) and nitrogen species, lipid peroxidation and
inflammation. This leads to cardiomyocyte apoptosis and
interstitial fibrosis, increasing the risk for impaired coronary
endothelial function, left ventricular (LV) dysfunction and
heart failure.1-3
Today, CTX is monitored by periodic imaging with
echocardiography for assessment of left ventricular
ejection fraction (LVEF) reduction and/or decreased global
longitudinal strain.4 However, the diagnosis of CTX based
on these cardiac function parameters is late, and can be
an indication of a significant and irreversible myocardial
injury.5,6 Therefore, it is necessary to evaluate myocardial
abnormalities at subcellular level for an early and sensitive
assessment of drug-induced CTX.7,8
Cardiac imaging techniques of nuclear medicine have
proved extremely useful to identify subclinical disease in
the context of cancer therapy-induced organ damage.9–11
Positron emission tomography associated with computed
tomography scans (PET/CT) with 2-deoxy-2[18F] fluoro-
1
Arq Bras Cardiol. 2021; [online].ahead print, PP.0-0
Original Article
Dourado et al.
18F-FDG Uptake and Cardiotoxicity
D-glucose (18F-FDG) is widely used in oncology, especially
in patients with lymphoma.12,13 Tissue 18F-FDG uptake and
tissue distribution is variable and depend on several factors
such as glucose level, fasting period and drugs.14 Furthermore,
recent data suggest that myocardial 18F-FDG accumulation
is not entirely due to glucose consumption.15 The tracer
retention was found to be dependent upon the enzymatic
activity of hexose-6-phosphate-dehydrogenase (H6PD) in
the endoplasmic reticulum (ER).15 This enzyme can process
many hexoses, including FDG,16 to trigger a pentose phosphate
pathway and preserve NADPH levels in response to oxidative
stress conditions, such as CTX.17
This study aimed to identify potential early signs of
metabolic cardiac injury by assessing changes in cardiac
18F-FDG uptake by PET/CT in patients with lymphoma before,
during and/or after chemotherapy.
Material and Methods
Patients
Seventy patients diagnosed with lymphoma and submitted
to 18F-FDG PET/CT in the Division of Nuclear Medicine of Real
Hospital Português in Recife, Pernambuco, Brazil, between
January 1, 2012 and August 28, 2017 were retrospectively
analyzed in this study. The study was approved by the Research
Ethics Board of the Federal University of Pernambuco Health
Sciences Center, which granted a waiver of written consent
due to the retrospective nature of the study.
Inclusion criteria were primary diagnosis of lymphoma,
aged 10 years or older and, at least two 18F-FDG PET/CT scans
before, during and/or after chemotherapy. Exclusion criteria
were no baseline or control tests, unavailability and/or inability
to assess clinical data and imaging tests, and insulin therapy
on the day of the scan.
Patients’ clinical features, medical history and variables
related to the 18F-FDG PET/CT protocol recorded in their
medical records were collected, such as, weight, injected
dose of 18F-FDG, fasting blood sugar (FBS) and time after
injection. For imaging exams, 18F-FDG uptake was quantified
by measuring the mean and the maximum standardized
uptake value (SUVmean and SUVmax, respectively).
Four patients had only baseline and interim PET/CT scans,
40 had only baseline and post-therapy and 26 had all three.
For analysis, the patients were then divided into two groups,
group 1, patients with baseline and interim PET/CT scan data
(n = 30); and group 2, patients with baseline and post-therapy
PET/CT data (n = 66). Thus, some patients participated in
both analyses.
Each group was then divided in two subgroups according
to the change in the LV 18F-FDG SUVmax between baseline
and control tests: a percentage increase above or equal to
30% (Group ≥ 30%), and a less than 30% 18F-FDG uptake
change (Group <30%). The choice of a 30% cutoff was
based on PERCIST18 (PET Response Criteria in Solid Tumors),
which is a set of criteria for assessment of tumor response to
chemotherapy and radiotherapy, through metabolic changes
verified by 18F-FDG PET/CT scans.18
18F-FDG PET/CT Protocol
For the 18F-FDG PET/CT, patients were instructed to fast
at least six hours prior to the test, not to discontinue any
medication or exercise for 24 hours before the scan. On the
day of the scan, body weight (kg) and FBS were measured
and, venous puncture was used to administer 18F-FDG. Blood
sugar levels should be below 180 mg/dL. The 18F-FDG was
administered at an activity dose of 3.7 to 4.8MBq/kg and
after 60 minutes, the images were obtained by the PET/CT
(Biograph 16, Siemens Healthcare, USA), extending from the
base of the skull to the proximal-middle third of the femur,
three minutes per bed position. The acquisition parameters
of the CT scan included: 5mm slices, 120kV voltage, and no
intravenous contrast administration.
Imaging processing was done with iterative reconstruction
(two iteractions, eight subsets with Gaussian filter) by a
nuclear physician, who performed a quantitative analysis
with SUVmax and SUVmean. Both SUVs were measured at
the left ventricle on fused PET/CT images and determined
semi-automatically with the aid of the syngo via software
version 5.1 (Siemens Healthcare) through the demarcation of
a volume of interest (VOI) including the entire left ventricle.
SUVmax and SUVmean for blood pool were measured by
reconstruction of a region of interest (ROI) in the descendent
aorta just after the aortic arch. SUVmax and SUVmean for
liver were measured by reconstruction of a ROI of 4.0 cm
diameter in the VI segment.
Statistical analysis
Data was analyzed with Stata 12.1 statistical software.
Continuous variables were expressed as mean ± standard
deviation (SD); and categorical variables were summarized
by frequency and percentage. Percentage comparisons
between two independent groups were performed using the
Pearson’s chi-square test or, when it was not applicable, the
Fisher’s exact test. The Student’s t-test was used to compare
two means for both independent and paired samples. In
all tests, a significance level of 5% was used to reject the
null hypothesis.
Results
The mean age of the 70 patients studied was 50.4 ± 20.1
years (16-88 years) and 50% were female. Twenty patients
(28.6%) had hypertension and 10 (14.3%) had diabetes. About
67% (n= 47) had non-Hodgkin’s lymphoma (nHL) and the
remainder (n=23) had Hodgkin’s lymphoma (HL). Only three
patients (4.3%) underwent mediastinal radiotherapy between
the end of chemotherapy and the control 18F-FDG PET/CT
scan. It was possible to define the chemotherapy regimen
in 33 patients (47.1%) and all regimens included known
cardiotoxic drugs (Table 1).
Group 1: baseline and interim 18F-FDG PET/CT
There was standardization of the 18F-FDG PET/CT protocol
between the baseline and interim scans. There was no
difference in the injected dose of 18F-FDG, FBS and time
post-injection between baseline and interim exams. Mean
body weight of patients also did not change significantly,
2
Arq Bras Cardiol. 2021; [online].ahead print, PP.0-0
Original Article
Dourado et al.
18F-FDG Uptake and Cardiotoxicity
Table 1 – Clinical and therapeutic characteristics of the patients (n=70)
Variable N (%)
Female sex 35 (50.0)
Hypertension 20 (28.6)
Diabetes 10 (14.3)
Dyslipidemia 14 (20.0)
Smoking
Non-smoker 49 (70.0)
Former smoker 20 (28.6)
Current smoker 1 (1.4)
Alcoholism 0 (0)
Coronary artery disease 5 (7.1)
Hemodialysis 1 (1.4)
Medication
No 10 (14.3)
Non-cardioprotective medication a40 (57.1)
Cardioprotective medication a20 (28.6)
Cancer
Hodgkin’s Lymphoma 23 (32.9)
Non-Hodgkin’s Lymphoma 47 (67.1)
Chemotherapy b
RCHOP 11 (33.3)
RCHOP + alternative 6 (18.2)
ABVD 11 (33.3)
ABVD + alternative 2 (6.1)
DA-EPOCH-R 1 (3.0)
BEACOPP 1 (3.0)
RCOP 1 (3.0)
Mediastinal
Radiotherapy After Baseline Pet 3 (4.3)
a Cardioprotective medication: angiotensin II receptor blocker, beta-blocker, angiotensin-converting enzyme inhibitor. b Available for 33 patients. ABVD:
Adriamycin or Doxorubicin + Bleomycin + Vinblastine + Dacarbazine; BEACOPP: Bleomycin + Etoposide + Adriamycin or Doxorubicin + Cyclophosphamide
+ Vincristine + Procarbazine + Prednisolone; DA-EPOCH-R: Dose-Adjusted Etoposide + Prednisolone + Vincristine + Cyclophosphamide + Doxorubicin
or Hydroxydaunorubicin + Rituximab, RCHOP: Rituximab + Cyclophosphamide + Doxorubicin or Hydroxydaunorubicin + Vincristine + Prednisolone,
RCOP: Rituximab + Cyclophosphamide + Vincristine + Prednisolone.
making it possible to compare the 18F-FDG uptake in the
target organs (Table 2).
On the other hand, 18F-FDG LV SUVmax increased at
the interim scan compared to baseline. Similarly, there was
a significant increase in the LV SUVmax/aorta SUVmax and
LV SUVmean/liver SUVmean ratios from baseline to interim
scans (Figure 1A). The mean time interval between baseline
and interim scans was 95.4 ± 32.2 days.
Of the 30 patients who underwent baseline and interim
18F-FDG PET/CT scans,16 (53.3%) presented an increase
≥30% (Group ≥ 30%) in 18F-FDG LV SUVmax. Regarding
clinical variables, such as cardiovascular risk factors and drugs
in use, no differences were observed.
The values of the LV SUVmax/aorta SUVmax and LV
SUVmean/liver SUVmean ratios also increased significantly at
the interim evaluation compared to the baseline in the group
≥30% (Figure 1B). In the group<30% (n=14), there was no
statistically significant increase in these ratios from baseline
to interim scans (Figure 1C).
Group 2: baseline and post-therapy 18F-FDG PET/CT
Sixty-six patients underwent baseline and post-therapy
18F-FDG PET/CT scans. No statistically significant differences
were seen in FBS, 18F-FDG injected activity and time post-
injection were found between the two evaluations. Patients’
mean body weight was slightly higher in the post-therapy scan
compared with baseline (Table 3).
3
Arq Bras Cardiol. 2021; [online].ahead print, PP.0-0
Original Article
Dourado et al.
18F-FDG Uptake and Cardiotoxicity
Table 2 – Comparison of body weight, fasting blood sugar, injected dose of 18F-fluorodeoxy glucose (18FDG), and mean
post-injection time of patients between baseline and interim positron emission tomography associated with computed tomography
(PET/CT) scans
Variable (N=30) Baseline Interim p*
Mean ± SD Mean ± SD
Weight (Kg) 75.3 ± 14.3 74.7 ± 13.5 0.551
FBS (mg/dL) 92.6 ± 19.5 93.4 ± 19.9 0.816
Dose of 18FDG mCi 9.1 ± 2.7 9.1 ± 2.0 0.971
Post-injection time (min) 68.8 ± 10.0 65.9 ± 9.9 0.308
*Student’s t-test. FBS: Fasting Blood Sugar
Figure 1 – Group 01 – A) Comparison of maximum left ventricular (LV) standardized uptake value (SUVmax), LV SUVmax/aorta SUVmax and mean LV SUV
(SUV mean)/liver SUVmean ratios, between baseline and interim positron emission tomography (PET). B) Comparison of LV SUVmax/aorta SUVmax and LV
SUVmean/liver SUVmean ratios between baseline and interim PET in the Group with increase of LV SUVmax 30%. C) Comparison of LV SUVmax/Aorta
SUVmax and LV SUVmean/Liver SUVmean ratios, between Baseline and Interim PET in the Group with increase of LV SUVmax < 30%; LVmaxAOmax: LV
SUVmax/Aorta SUVmax, LVmean LIVER mean: LV SUVmean/Liver SUVmean.
The mean value of the LV SUVmax was significantly higher
in the post-therapy PET. We observed an absolute increase
in the 18F-FDG cardiac uptake value of 2.1 (95% CI:1.3
to 3.0), which represents a percentage increase of 66.5%
(95%CI:43.3% to 89.7%) over the baseline scan.
The values of the LV SUV max/aorta SUV max and the LV SUV
mean/liver SUV mean ratios also increased significantly in the post-
therapy PET as compared with baseline, Figure 2A. The mean time
between baseline and post-therapy exams was 231.8±125.7 days.
Of the 66 patients, 38 (57.6%) presented ≥30% increase
in 18F-FDG cardiac uptake (Group 30%). There were no
differences between the groups regarding the clinical variables,
such as cardiovascular risk factors and medications in use.
The values of the LV SUVmax/aorta SUVmax and LV
SUVmean/liver SUVmean ratios increased significantly in
the post-therapy evaluation compared to the baseline in the
≥30% group (Figure 2B). In the Group<30% (n=28), there
was no statistically significant increase in the ratios (Figure 2C).
Figure 3 illustrates a case example of the 18F-FDG LV SUV
max behavior before, during and after chemotherapy.
Discussion
The present study showed that chemotherapy in patients
with lymphoma caused an unbalance in cardiac metabolism,
evidenced by a higher myocardial 18F-FDG uptake. These
4
Arq Bras Cardiol. 2021; [online].ahead print, PP.0-0
Original Article
Dourado et al.
18F-FDG Uptake and Cardiotoxicity
Table 3 – Comparison of body weight, fasting blood sugar, injected dose of 18F-fluorodeoxy-glucose (18FDG), and mean post-injection
timel of patients between baseline and post-therapy positron emission tomography associated with computed tomography scans
(PET/CT)
Variable (N=66) Baseline Pet Post-Therapy Pet p*
Mean ± SD Mean ± SD
Weight (Kg) 72.7 ± 14.8 75.2 ± 15.2 0.014
FBS (mg/dL) 91.6 ± 15.6 91.6 ± 16.7 >0.99
Dose of 18FDG mCi 9.2 ± 2.3 9.5 ± 2.2 0.308
Post-injection time (min) 68.6 ± 9.1 70.4 ± 5.8 0.606
*Student’s t-test. FBS: Fasting Blood Sugar
Figure 2 – Group 02 – A) Comparison of LV SU Vmax, LV SUVmax/Aorta SUVmax and LV SUVmean/Liver SUVmean ratios, between Baseline and Post-therapy
PET. B) Comparison of LV SUVmax/Aorta SUVmax and LV SUVmean/Liver SUVmean ratios, between Baseline and Post-therapy PET in the Group with
increase of LV SUVmax 30%. C) Comparison of LV SUVmax/Aorta SUVmax and LV SUVmean/Liver SUVmean ratios, between Baseline and Post-therapy
PET in the Group with increase of LV SUVmax < 30%; LVmaxAOmax: LV SUVmax/Aor ta SUVmax, LVmean LIVER mean: LV SUVmean/Liver SUVmean.
Figure 3 – Case example - LV SUVmax in Baseline (5.86), Interim (8.95 / 52.73% percentage increase from baseline) and Post-therapy PET/CT (9.67 /
65.02% percentage increase from baseline). LV: Left Ventricle; PET/CT: Positron emission tomography associated with computed tomography scans; SUV:
Standard Uptake Value; SUVmax: Maximum SUV.
5
Arq Bras Cardiol. 2021; [online].ahead print, PP.0-0
Original Article
Dourado et al.
18F-FDG Uptake and Cardiotoxicity
results are supported by recent evidence suggesting that it may
be an early sign of CTX in response to the redox stress. The
cardiac 18F-FDG increase occurred in more than 50% of the
patients and was observed in the interim PET and in the post-
therapy scan. These results suffered no interference regarding
the18F-FDG injected activity or any possible differences in
exam preparation and timing.
The 18F-FDG PET/CT is a well-established method in
the diagnosis and staging of oncologic patients, especially
with lymphoma, with a potential capacity to assess early
manifestations of CTX in a way analogue to the ischemic
cascade, as postulated in Figure 4.
Antineoplastic therapies have improved overall survival
rates in oncologic patients. However, their cytotoxic
effects have shown a wide spectrum of acute and chronic
alterations to the cardiovascular system.19 The cellular and
molecular mechanisms of CTX are known to disrupt the redox
homeostasis mostly in the myocardium and endothelium,
significantly impairing cardiovascular health.20
CTX affects the cardiovascular system first by the inhibition
of topoisomerase II and the formation of ROS. The intrinsic
mitochondria-dependent and extrinsic death receptor
pathways of apoptosis are then triggered. The cascade
continues with the activation of caspase 3, phosphatidylserine
expression, DNA fragmentation, chromatin condensation,
and phospholipid membrane metabolization.21 The final
stage is characterized by membrane blebbing and cell
shrinkage.22 This is the mechanism underlying subclinical
CTX and it provides various opportunities to assess early
signs of this entity.
The current recommendations and guidelines rely on
imaging techniques focused on anatomy-based parameters,
such as echocardiography, multigated radionuclide angiography
(MUGA), and cardiac magnetic resonance imaging (CMRI).23
However, these approaches detect late manifestations of CTX
with low sensitivity for subclinical alterations.24
Nuclear medicine techniques may be a tool to assess
specific points of the CTX pathway. The 18F-FDG PET/CT,
commonly used to detect tumoral glycolytic metabolism, has
presented itself as an early marker of CTX. Initially, several
studies pointed out that doxorubicin (DXR), one of the most
utilized anthracyclines, can specifically affect myocardial
metabolism, as showed by experimental study.25
Several experimental and clinical studies have shown that
cardiotoxic therapy, such as sunitinib and anthracyclines,
increases the cardiac 18F-FDG uptake over time and is related
to echocardiographic alterations.26-33
Although 18F-FDG uptake has been commonly associated
with glucose consumption, more recent data have shown
otherwise. The redox stress and its antioxidant response
have been characterized as a possible mechanism behind
the progression of cardiac contractile impairment in CTX
and in the 18F-FDG uptake independently of the glycolytic
metabolism.34
Redox stress to the endoplasmic reticulum (ER) environment
might activate the local H6PD-triggered pentose phosphate
pathway to fuel the NADPH levels needed for the antioxidant
response, and is related to an increased 18F-FDG uptake.35
In situations of oxidative stress, NADPH is a major source of
electrons for reductive reactions.36 It is generated intraluminally
by H6PD, a bifunctional enzyme that catalyzes the first two
steps of the pentose phosphate pathway, converting glucose-
6-phosphate to 6-phosphogluconate with the concomitant
production of NADPH.37 H6PD has as substrate several
hexoses such as 2-deoxyglucose and FDG.38
In the heart, there is a direct link between ER oxidative
stress and myocardial uptake of 2-deoxyglucose,39 that may be
considered an early metabolic phase of contractile dysfunction
by pressure overload.40 Furthermore, Hrelia et al.41 showed
that the increase of 2-deoxyglucose uptake induced by DXR
in cardiomyocytes can be reverted by the antioxidant effect
of alpha-tocopherol.41
Bauckneht et al.,33 in 2019, analyzed the effect of DXR
-induced oxidative damage on the correlation between
myocardial 18F-FDG uptake, overall glucose consumption and
the H6PD-triggered metabolic response in mice. The study
showed that myocardial redox stress persisted and directly
correlated with the enhancement in 18F-FDG uptake (SUV
increase), and the activation of physiological antioxidant
pathways such as the catalytic function of H6PD.33 The study
also showed that the metabolic alteration persisted after the
disappearance of DXR, and it preceded the manifestation
of contractile impairment.33 Previous reports also showed a
positive loop connecting ROS generation and 18F-FDG uptake
in cancer.42
In agreement with these findings, recent studies showed an
increased 18F-FDG uptake on PET/CT independent of glycolytic
metabolism and linked to the enzymatic activity of H6PD in
the brain.43,44 Another analysis showed the link between 18F-
FDG uptake and ROS generation in hyperglycemia-induced
redox stress involving H6PD activation.45
Despite its interesting results and background of the
present study, its retrospective nature makes the assessment
of the mechanisms underlying the increased myocardial 18F-
FDG uptake difficult. However, no other cardiotoxic factors,
besides CTX, were identified between baseline and control
exams in the largest sample of patients with lymphoma
evaluated during and after chemotherapy. In addition, unlike
the other studies, we measured not only the LV SUVmax, but
also the LV uptake values corrected for liver and blood pool,
as control, confirming the increase of the cardiac uptake.
Furthermore, the 18F-FDG PET/CT protocol and the possible
factors of SUV variability were the same in all baseline and
control scans.
More studies are necessary to correlate increased cardiac
18F-FDG uptake with clinical outcomes, the class and dose
of chemotherapy, troponin and NT-proBNP levels, and with
other imaging methods such as echocardiography and CMRI.
Conclusion
The present study showed a clear increase in cardiac
18F-FDG uptake in patients with lymphoma, verified by
18F-FDG PET/CT during and/or after chemotherapy. The
literature corroborates with these findings and suggests that it
may be an important and early sign of CTX that can be easily
assessed by a widely available method. With the progressive
6
Arq Bras Cardiol. 2021; [online].ahead print, PP.0-0
Original Article
Dourado et al.
18F-FDG Uptake and Cardiotoxicity
Figure 4 – Cardiotoxicity cascade – Cardiotoxic injury triggers series of metabolic alterations in response to the oxidative stress, it is detectable by 18F-FDG
PET/CT. The sustained injury and the failure of the myocyte self-healing contribute to cell dysfunction and mechanic alterations detected by strain rate
imaging. Furthermore, the process continues with a decrease in the cardiac overall performance assessed by the LVEF. Signs of heart failure are then
noticeable, suggesting that the heart no longer meet the body’s demands, or do it at the expense of high ventricular filling pressures (ROS: reactive oxygen
species; ER: endoplasmic reticulum; PPP: pentose phosphate pathway; H6PD: hexose-6-phosphate dehydrogenase; FDG: 18F-fluorodeoxy-glucose; LVEF:
Left Ventricle Ejection Fraction).
7
Arq Bras Cardiol. 2021; [online].ahead print, PP.0-0
Original Article
Dourado et al.
18F-FDG Uptake and Cardiotoxicity
improvement in anticancer therapies, CTX is still a concern that
requires further investigation and new diagnostic approaches.
Acknowledgements
We are pleased to acknowledge the support provided by
all technicians and nuclear physicians of the Real Nuclear at
the Real Hospital Português in hosting our study.
Author Contributions
Conception and design of the research: Dourado MLC,
Leitão GM, Mourato FA, Almeida Filho PJ, Markman Filho
B, Melo MDT, Brandão SCS; Acquisition of data, Analysis
and interpretation of the data and Critical revision of the
manuscript for intellectual content: Dourado MLC, Dompieri
LT, Leitão GM, Mourato FA, Santos RGG, Almeida Filho PJ,
Markman Filho B, Melo MDT, Brandão SCS; Statistical analysis:
Dourado MLC, Brandão SCS; Obtaining financing: Dourado
MLC; Writing of the manuscript: Dourado MLC, Dompieri
LT, Brandão SCS.
Potential Conflict of Interest
No potential conflict of interest relevant to this article was
reported.
Sources of Funding
There were no external funding sources for this study.
Study Association
This article is part of the thesis of master submitted
by Mayara L. C. Dourado, from Universidade Federal de
Pernambuco - UFPE.
1. Awadalla M, Hassan MZO, Alvi RM, Neilan TG. Advanced Imaging
Modalities to Detect Cardiotoxicity. Curr Probl Cancer. 2018;42(4):386-96.
doi: 10.1016/j.currproblcancer.2018.05.005.
2. Kalil Filho R, Hajjar LA, Bacal F, Hoff PM, Diz MP, Galas FR, et al. I Brazilian
Guideline for Cardio-Oncology from Sociedade Brasileira de Cardiologia.
Arq Bras Cardiol. 2011;96(2 Suppl 1):1-52.
3. Jain D, Russell RR, Schwartz RG, Panjrath GS, Aronow W. Cardiac
Complications of Cancer Therapy: Pathophysiology, Identification,
Prevention, Treatment, and Future Directions. Curr Cardiol Rep.
2017;19(5):36. doi: 10.1007/s11886-017-0846-x.
4. Seidman A, Hudis C, Pierri MK, Shak S, Paton V, Ashby M, et al. Cardiac
Dysfunction in the Trastuzumab Clinical Trials Experience. J Clin Oncol.
2002;20(5):1215-21. doi: 10.1200/JCO.2002.20.5.1215.
5. Curigliano G, Cardinale D, Suter T, Plataniotis G, Azambuja E, Sandri
MT, et al. Cardiovascular Toxicity Induced by Chemotherapy, Targeted
Agents and Radiotherapy: ESMO Clinical Practice Guidelines. Ann Oncol.
2012;23(Suppl 7):155-66. doi: 10.1093/annonc/mds293.
6. Negishi T, Negishi K. Echocardiographic Evaluation of Cardiac Function After
Cancer Chemotherapy. J Echocardiogr. 2018;16(1):20-7. doi: 10.1007/
s12574-017-0344-6.
7. Barros-Gomes S, Herrmann J, Mulvagh SL, Lerman A, Lin G, Villarraga HR.
Rationale for Setting up a Cardio-Oncology Unit: Our Experience at Mayo
Clinic. Cardiooncology. 2016;2(1):5. doi: 10.1186/s40959-016-0014-2.
8. Felker GM, Thompson RE, Hare JM, Hruban RH, Clemetson DE, Howard
DL, et al. Underlying Causes and Long-Term Survival in Patients with Initially
Unexplained Cardiomyopathy. N Engl J Med. 2000;342(15):1077-84. doi:
10.1056/NEJM200004133421502.
9. Simoni LJC, Brandão SCS. New Imaging Methods for Detection of Drug-
Induced Cardiotoxicity in Cancer Patients. Curr Cardiovasc Imaging Rep.
2017;10(18):1-11. doi: 10.1007/s12410-017-9415-3.
10. Rix A, Drude NI, Mrugalla A, Baskaya F, Pak KY, Gray B, et al. Assessment of
Chemotherapy-Induced Organ Damage with Ga-68 Labeled Duramycin.
Mol Imaging Biol. 2020;22(3):623-33. doi: 10.1007/s11307-019-01417-3.
11. Kahanda MG, Hanson CA, Patterson B, Bourque JM. Nuclear Cardio-
Oncology: From its Foundation to its Future. J Nucl Cardiol. 2020;27(2):511-
8. doi: 10.1007/s12350-019-01655-6.
12. Wu X, Bhattarai A, Korkola P, Pertovaara H, Eskola H, Kellokumpu-Lehtinen
PL. The Association Between Liver and Tumor [18F]FDG Uptake in Patients
with Diffuse Large B Cell Lymphoma During Chemotherapy. Mol Imaging
Biol. 2017;19(5):787-94. doi: 10.1007/s11307-017-1044-3.
13. Zhou Y, Zhao Z, Li J, Zhang B, Sang S, Wu Y, et al. Prognostic Values of
Baseline, Interim and End-of Therapy 18F-FDG PET/CT in Patients with
Follicular Lymphoma. Cancer Manag Res. 2019;11:6871-85. doi: 10.2147/
CMAR.S216445.
14. Bascuñana P, Thackeray JT, Bankstahl M, Bengel FM, Bankstahl JP. Anesthesia
and Preconditioning Induced Changes in Mouse Brain [18F] FDG Uptake
and Kinetics. Mol Imaging Biol. 2019;21(6):1089-96. doi: 10.1007/s11307-
019-01314-9.
15. Marini C, Ravera S, Buschiazzo A, Bianchi G, Orengo AM, Bruno S, et
al. Discovery of a Novel Glucose Metabolism in Cancer: The Role of
Endoplasmic Reticulum Beyond Glycolysis and Pentose Phosphate Shunt.
Sci Rep. 2016;6:25092. doi: 10.1038/srep25092.
16. Clarke JL, Mason PJ. Murine Hexose-6-Phosphate Dehydrogenase:
A Bifunctional Enzyme with Broad Substrate Specificity and
6-Phosphogluconolactonase Activity. Arch Biochem Biophys.
2003;415(2):229-34. doi: 10.1016/s0003-9861(03)00229-7.
17. Rogoff D, Black K, McMillan DR, White PC. Contribution of Hexose-6-
Phosphate Dehydrogenase to NADPH Content and Redox Environment in
the Endoplasmic reticulum. Redox Rep. 2010;15(2):64-70. doi: 10.1179/
174329210X12650506623249.
18. Pinker K, Riedl C, Weber WA. Evaluating Tumor Response with FDG PET:
Updates on PERCIST, Comparison with EORTC Criteria and Clues to Future
Developments. Eur J Nucl Med Mol Imaging. 2017;44(Suppl 1):55-66. doi:
10.1007/s00259-017-3687-3.
19. Aggarwal S, Kamboj J, Arora R. Chemotherapy-related
Cardiotoxicity. Ther Adv Cardiovasc Dis. 2013;7(2):87-98. doi:
10.1177/1753944712474332.
20. Vincent DT, Ibrahim YF, Espey MG, Suzuki YJ. The Role of Antioxidants in the
Era of Cardio-Oncology. Cancer Chemother Pharmacol. 2013;72(6):1157-
68. doi: 10.1007/s00280-013-2260-4.
21. Zhang S, Liu X, Bawa-Khalfe T, Lu LS, Lyu YL, Liu LF, et al. Identification
of the Molecular Basis of Doxorubicin-Induced Cardiotoxicity. Nat Med.
2012;18(11):1639-42. doi: 10.1038/nm.2919.
22. Vangestel C, Peeters M, Mees G, Oltenfreiter R, Boersma HH, Elsinga PH,
et al. In Vivo Imaging of Apoptosis in Oncology: An Update. Mol Imaging.
2011;10(5):340-58. doi: 10.2310/7290.2010.00058.
References
8
Arq Bras Cardiol. 2021; [online].ahead print, PP.0-0
Original Article
Dourado et al.
18F-FDG Uptake and Cardiotoxicity
23. Markman TM, Markman M. Cardiotoxicity of Antineoplastic Agents: What is
the Present and Future Role for Imaging? Curr Oncol Rep. 2014;16(8):396.
doi: 10.1007/s11912-014-0396-y.
24. Wood PW, Choy JB, Nanda NC, Becher H. Left Ventricular Ejection Fraction
and Volumes: It Depends on the Imaging Method. Echocardiography.
2014;31(1):87-100. doi: 10.1111/echo.12331.
25. Yang Y, Zhang H, Li X, Yang T, Jiang Q. Effects of PPARα/PGC-1α on the
Energy Metabolism Remodeling and Apoptosis in the Doxorubicin Induced
Mice Cardiomyocytes in Vitro. Int J Clin Exp Pathol. 2015;8(10):12216-24.
26. Borde C, Kand P, Basu S. Enhanced Myocardial Fluorodeoxyglucose Uptake
Following Adriamycin-Based Therapy: Evidence of Early Chemotherapeutic
Cardiotoxicity? World J Radiol. 2012;4(5):220-3. doi: 10.4329/wjr.
v4.i5.220.
27. O’Farrell AC, Evans R, Silvola JM, Miller IS, Conroy E, Hector S, et al.
A Novel Positron Emission Tomography (PET) Approach to Monitor
Cardiac Metabolic Pathway Remodeling in Response to Sunitinib
Malate. PLoS One. 2017;12(1):e0169964. doi: 10.1371/journal.
pone.0169964.
28. Sourdon J, Lager F, Viel T, Balvay D, Moorhouse R, Bennana E, et al. Cardiac
Metabolic Deregulation Induced by the Tyrosine Kinase Receptor Inhibitor
Sunitinib is Rescued by Endothelin Receptor Antagonism. Theranostics.
2017;7(11):2757-74. doi: 10.7150/thno.19551.
29. Kim J, Cho SG, Kang SR, Yoo SW, Kwon SY, Min JJ, et al. Association Between
FDG Uptake in the Right Ventricular Myocardium and Cancer Therapy-
Induced Cardiotoxicity. J Nucl Cardiol. 2020;27(6):2154-63. doi: 10.1007/
s12350-019-01617-y.
30. Sarocchi M, Bauckneht M, Arboscello E, Capitanio S, Marini C, Morbelli S, et
al. An Increase in Myocardial 18-Fluorodeoxyglucose Uptake is Associated
with Left Ventricular Ejection Fraction Decline in Hodgkin Lymphoma
Patients Treated with Anthracycline. J Transl Med. 2018;16(1):295. doi:
10.1186/s12967-018-1670-9.
31. Bauckneht M, Ferrarazzo G, Fiz F, Morbelli S, Sarocchi M, Pastorino F, et
al. Doxorubicin Effect on Myocardial Metabolism as a Prerequisite for
Subsequent Development of Cardiac Toxicity: A Translational 18F-FDG
PET/CT Observation. J Nucl Med. 2017;58(10):1638-45. doi: 10.2967/
jnumed.117.191122.
32. Bauckneht M, Morbelli S, Fiz F, Ferrarazzo G, Piva R, Nieri A, et al. A
Score-Based Approach to 18F-FDG PET Images as a Tool to Describe
Metabolic Predictors of Myocardial Doxorubicin Susceptibility. Diagnostics.
2017;7(4):57 doi: 10.2967/jnumed.117.191122.
33. Bauckneht M, Pastorino F, Castellani P, Cossu V, Orengo AM, Piccioli P, et al.
Increased Myocardial 18F-FDG Uptake as a Marker of Doxorubicin-Induced
Oxidative Stress. J Nucl Cardiol. 2020;27(6):2183-94. doi: 10.1007/
s12350-019-01618-x.
34. Octavia Y, Tocchetti CG, Gabrielson KL, Janssens S, Crijns HJ, Moens AL.
Doxorubicin-Induced Cardiomyopathy: from Molecular Mechanisms
to Therapeutic Strategies. J Mol Cell Cardiol. 2012;52(6):1213-25. doi:
10.1016/j.yjmcc.2012.03.006.
35. Bánhegyi G, Benedetti A, Fulceri R, Senesi S. Cooperativity Between
11beta-Hydroxysteroid Dehydrogenase type 1 and Hexose-6-Phosphate
Dehydrogenase in the Lumen of the Endoplasmic Reticulum. J Biol Chem.
2004;279(26):27017-21. doi: 10.1074/jbc.M404159200.
36. Fico A, Paglialunga F, Cigliano L, Abrescia P, Verde P, Martini G, et al. Glucose-
6-Phosphate Dehydrogenase Plays a Crucial Role in Protection from
Redox-Stress-Induced Apoptosis. Cell Death Differ. 2004;11(8):823-31.
doi: 10.1038/sj.cdd.4401420.
37. Mason PJ, Stevens D, Diez A, Knight SW, Scopes DA, Vulliamy TJ. Human
Hexose-6-Phosphate Dehydrogenase (glucose 1-dehydrogenase)
Encoded at 1p36: Coding Sequence and Expression. Blood Cells Mol Dis.
1999;25(1):30-7. doi: 10.1006/bcmd.1999.0224.
38. Clarke JL, Mason PJ. Murine Hexose-6-Phosphate Dehydrogenase:
A Bifunctional Enzyme with Broad Substrate Specificity and
6-Phosphogluconolactonase Activity. Arch Biochem Biophys.
2003;415(2):229-34. doi: 10.1016/s0003-9861(03)00229-7.
39. Sen S, Kundu BK, Wu HC, Hashmi SS, Guthrie P, Locke LW, et al.
Glucose Regulation of Load-Induced mTOR Signaling and ER Stress in
Mammalian Heart. J Am Heart Assoc. 2013;2(3):e004796. doi: 10.1161/
JAHA.113.004796.
40. Zhong M, Alonso CE, Taegtmeyer H, Kundu BK. Quantitative PET Imaging
Detects Early Metabolic Remodeling in a Mouse Model of Pressure-
Overload Left Ventricular Hypertrophy in Vivo. J Nucl Med. 2013;54(4):609-
15. doi: 10.2967/jnumed.112.108092.
41. Hrelia S, Fiorentini D, Maraldi T, Angeloni C, Bordoni A, Biagi PL, et al.
Doxorubicin Induces Early Lipid Peroxidation Associated with Changes
in Glucose Transport in Cultured Cardiomyocytes. Biochim Biophys Acta.
2002;1567(1-2):150-6. doi: 10.1016/s0005-2736(02)00612-0.
42. Chen L, Zhou Y, Tang X, Yang C, Tian Y, Xie R, et al. EGFR Mutation
Decreases FDG Uptake in Non-Small Cell Lung Cancer via the NOX4/ROS/
GLUT1 Axis. Int J Oncol. 2019;54(1):370-80. doi: 10.3892/ijo.2018.4626.
43. Cossu V, Marini C, Piccioli P, Rocchi A, Bruno S, Orengo AM, et al. Obligatory
Role of Endoplasmic Reticulum in Brain FDG Uptake. Eur J Nucl Med Mol
Imaging. 2019;46(5):1184-96. doi: 10.1007/s00259-018-4254-2.
44. Buschiazzo A, Cossu V, Bauckneht M, Orengo A, Piccioli P, Emionite L, et
al. Effect of Starvation on Brain Glucose Metabolism and 18F-2-fluoro-2-
deoxyglucose Uptake: An Experimental In-vivo and Ex-vivo Study. EJNMMI
Res. 2018;8(1):44. doi: 10.1186/s13550-018-0398-0.
45. Bauckneht M, Cossu V, Castellani P, Piccioli P, Orengo AM, Emionite L, et
al. FDG Uptake Tracks the Oxidative Damage in Diabetic Skeletal Muscle:
An Experimental Study. Mol Metab. 2020;31:98-108. doi: 10.1016/j.
molmet.2019.11.007.
9
Arq Bras Cardiol. 2021; [online].ahead print, PP.0-0
Original Article
Dourado et al.
18F-FDG Uptake and Cardiotoxicity
This is an open-access article distributed under the terms of the Creative Commons Attribution License
10
... A few of these retrospective studies [21,34,77] have demonstrated the possible contribution of 18 F-FDG toward CTX assessment, as summarized in Table 1. Borde et al. [21] analyzed the myocardial uptake of 18 F-FDG in patients with lymphoma who were treated with doxorubicin. ...
... Dourado et al. [77] retrospectively evaluated 70 patients with lymphoma submitted to 18 F-FDG PET/CT. They observed an increase in LV maximum SUV (SUVmax) from 3,5 ± 1,9 (baseline) to 5,6 ± 4,0 (interim), p = 0.01, and from 4,0 ± 2,2 (baseline) to 6,1 ± 4,2 (post-therapy), p < 0,001. ...
... They observed an increase in LV maximum SUV (SUVmax) from 3,5 ± 1,9 (baseline) to 5,6 ± 4,0 (interim), p = 0.01, and from 4,0 ± 2,2 (baseline) to 6,1 ± 4,2 (post-therapy), p < 0,001. A percentage increase ≥ 30% of LV SUVmax occurred in more than half of the sample [77]. The rise of cardiac SUV was accompanied by an increase in LV SUVmax/Aorta SUVmax and LV SUVmax/Liver SUVmean (mean standard uptake value) ratios. ...
Article
Full-text available
Despite advances in chemotherapy, the drugs used in cancer treatment remain rather harmful to the cardiovascular system, causing structural and functional cardiotoxic changes. Positron-emission tomography associated with computed tomography (PET/CT) has emerged like a promising technique in the early diagnosis of these adverse drug effects as the myocardial tissue uptake of fluorodeoxyglucose labeled with fluorine-18 (¹⁸F-FDG), a glucose analog, is increased after their use. Among these drugs, anthracyclines are the most frequently associated with cardiotoxicity because they promote heart damage through DNA breaks, and induction of an oxidative, proinflammatory, and toxic environment. This review aimed to present the scientific evidence available so far regarding the use of ¹⁸F-FDG PET/CT as an early biomarker of anthracycline-related cardiotoxicity. Thus, it discusses the physiological basis for its uptake, hypotheses to justify its increase in the myocardium affected by anthracyclines, importance of ¹⁸F-FDG PET/CT findings for cardio-oncology, and primary challenges of incorporating this technique in standard clinical oncology practice.
... Although there is no definite answer in the literature, studies have suggested that this increase may be an early indicator of cardiotoxicity. [4][5][6][7][8][9] Borde et al. 4 reported that, in lymphoma patients treated with doxorubicin (doses >250mg/m²), enhanced myocardial 18 F-FDG uptake may be an early marker of cardiotoxicity. Another study with 69 patients with Hodgkin disease showed a progressive increase in myocardial 18 F-FDG uptake during treatment that persisted six months after the end of chemotherapy. ...
... This study also showed a significant association between right ventricular 18 F-FDG uptake and cardiotoxicity. 7 Dourado et al. 8 observed a significant increase in myocardial 18 F-FDG uptake in patients undergoing chemotherapy for lymphoma. In more than half of patients, a percentage increase greater than 30% of left ventricular maximal SUV occurred after chemotherapy as compared with before chemotherapy. ...
... Although there is no definite answer in the literature, studies have suggested that this increase may be an early indicator of cardiotoxicity. [4][5][6][7][8][9] Borde et al. 4 reported that, in lymphoma patients treated with doxorubicin (doses >250mg/m²), enhanced myocardial 18 F-FDG uptake may be an early marker of cardiotoxicity. Another study with 69 patients with Hodgkin disease showed a progressive increase in myocardial 18 F-FDG uptake during treatment that persisted six months after the end of chemotherapy. ...
... This study also showed a significant association between right ventricular 18 F-FDG uptake and cardiotoxicity. 7 Dourado et al. 8 observed a significant increase in myocardial 18 F-FDG uptake in patients undergoing chemotherapy for lymphoma. In more than half of patients, a percentage increase greater than 30% of left ventricular maximal SUV occurred after chemotherapy as compared with before chemotherapy. ...
Article
Cancer treatment has the potential to cause cardiovascular issues and can encourage the appearance of all aspects of cardiac disease, including coronary heart disease, myocardial disease, heart failure, structural heart disease, and rhythm problems. Imaging is required for both diagnostic workup and therapy monitoring for all possible cardiovascular side effects of cancer therapy. Echocardiography is the cardiac imaging gold standard in cardio-oncology. Despite advancements in its use, this method is often not sensitive to early-stage or subclinical impairment. The use of molecular imaging technologies for diagnosing, assessing, and tracking cardiovascular illness as well as for treating, it is fast growing. Molecular imaging techniques using biologically targeted markers are gradually replacing the traditional anatomical or physiological approaches. They offer unique insight into patho-biological processes at the molecular and cellular levels and enable the evaluation and treatment of cardiovascular disease. This review paper will describe molecular-based single-photon emission computed tomography (SPECT) and positron emission tomography (PET) imaging techniques that are now available and in development to assess post-infarction cardiac remodeling. These methods could be used to evaluate important biological processes such as inflammation, angiogenesis, and scar formation.
Article
Full-text available
Objectives: The present study aims to verify the relationship between glucose consumption and uptake of 18F-2-deoxy-glucose (FDG) in the skeletal muscle (SM) of experimental models of streptozotocin-induced diabetes mellitus (STZ-DM). Methods: The study included 36 Balb/c mice. Two weeks after intraperitoneal administration of saline (control group, n = 18) or 150 mg streptozotocin (STZ-DM group, n = 18), the two cohorts were submitted to an oral glucose tolerance test and were further subdivided into three groups (n = 6 each): untreated and treated with metformin (MTF) at low or high doses (10 or 750 mg/kg daily, respectively). Two weeks thereafter, all mice were submitted to dynamic micro-positron emission tomography (PET) imaging after prolonged fasting. After sacrifice, enzymatic pathways and response to oxidative stress were evaluated in harvested SM. Results: On PET imaging, the FDG uptake rate in hindlimb SM was significantly lower in nondiabetic mice as compared with STZ-DM-untreated mice. MTF had no significant effect on SM FDG uptake in untreated mice; however, its high dose induced a significant decrease in STZ-DM animals. Upon conventional analysis, the SM standard uptake value was higher in STZ-DM mice, while MTF was virtually ineffective in either control or STZ-DM models. This metabolic reprogramming was not explained by any change in cytosolic glucose metabolism. By contrast, it closely agreed with the catalytic function of hexose-6P-dehydrogenase (H6PD; i.e., the trigger of a specific pentose phosphate pathway selectively located within the endoplasmic reticulum). In agreement with this role, the H6PD enzymatic response to both STZ-DM and MTF matched the activation of the NADPH-dependent antioxidant responses to the increased generation of reactive oxygen species caused by chronic hyperglycemia. Ex vivo analysis of tracer kinetics confirmed that the enhanced SM avidity for FDG occurred despite a significant reduction in glucose consumption, while it was associated with increased radioactivity transfer to the endoplasmic reticulum. Conclusions: These data challenge the current dogma linking FDG uptake to the glycolytic rate. They instead introduce a new model considering a strict link between the uptake of this glucose analog, H6PD reticular activity, and oxidative damage in diabetes, at least under fasting condition.
Article
Full-text available
Purpose In the present study, we aimed to investigate the role of baseline, interim and end-of treatment positron emission tomography/computed tomography (PET/CT) in assessing the prognosis of follicular lymphoma (FL). Methods A total of 84 FL patients were retrospectively analyzed in this study. Baseline (n=59), interim (n=24, after 2–4 cycles) and end-of treatment (n=43) PET/CT images were re-evaluated, and baseline maximum standardized uptake value (SUVmax), total metabolic tumor volume (tMTV) and total lesion glycolysis (TLG) were recorded. Interim (I-PET) and end-of treatment (E-PET) PET/CT responses were interpreted by Deauville five-point scale (D-5PS) and International Harmonization Project criteria (IHP). Survival curves were calculated by Kaplan-Meier curves, and differences between groups were compared by log-rank test. Results The 2-year progression-free survival (PFS) of the high- and low-TLG groups was 57.14% and 95.56%, respectively (p=0.0001). The 2-year overall survival (OS) of the high- and low-TLG groups was 62.50% and 100%, respectively (p<0.0001). Multivariate analysis showed that TLG was an independent prognostic factor for PFS (p=0.001, HR=6.577, 95% CI=2.167–19.960) and OS (p=0.030, HR=19.291, 95% CI =2.689–137.947). Besides, Eastern Cooperative Oncology Group (ECOG) was the independent prognostic factor for OS (HR=8.924, 95% CI=1.273–62.559, p=0.028). Interim PET results based on D-5PS or IHP criteria were not significantly correlated with PFS (all p>0.05). However, E-PET results using D-5PS and IHP criteria were statistically significant (p=0.0001 and p=0.006). The D-5PS showed stronger prognostic value compared with IHP criteria. The optimal cutoff value of ΔSUVmax% was 66.95% according to I-PET and 68.97% according to E-PET. However, only the ΔSUVmax% from the baseline to the end-of therapy yielded statistically significant results in the prediction of PFS (p=0.0002). Conclusion Our findings indicated that the baseline TLG and E-PET results were significantly associated with prognosis in patients with FL.
Article
Full-text available
Purpose The endoplasmic reticulum (ER) contains hexose-6P-dehydrogenase (H6PD). This enzyme competes with glucose-6P-phosphatase for processing a variety of phosphorylated hexoses including 2DG-6P. The present study aimed to verify whether this ER glucose-processing machinery contributes to brain FDG uptake. Methods Effect of the H6PD inhibitor metformin on brain 18F-FDG accumulation was studied, in vivo, by microPET imaging. These data were complemented with the in vitro estimation of the lumped constant (LC). Finally, reticular accumulation of the fluorescent 2DG analogue 2-[N-(7-nitrobenz-2-oxa-1,3-diazol-4-yl)amino]-2-deoxyglucose (2NBDG) and its response to metformin was studied by confocal microscopy in cultured neurons and astrocytes. Results Metformin halved brain 18F-FDG accumulation without altering whole body tracer clearance. Ex vivo, this same response faced the doubling of both glucose consumption and lactate release. The consequent fall in LC was not explained by any change in expression or activity of its theoretical determinants (GLUTs, hexokinases, glucose-6P-phosphatase), while it agreed with the drug-induced inhibition of H6PD function. In vitro, 2NBDG accumulation selectively involved the ER lumen and correlated with H6PD activity being higher in neurons than in astrocytes, despite a lower glucose consumption. Conclusions The activity of the reticular enzyme H6PD profoundly contributes to brain 18F-FDG uptake. These data challenge the current dogma linking 2DG/FDG uptake to the glycolytic rate and introduce a new model to explain the link between 18-FDG uptake and neuronal activity.
Article
Full-text available
Abstract Background Doxorubicin (DOX)-based chemotherapy for Hodgkin lymphoma (HL) yields excellent disease-free survival, but poses a substantial risk of subsequent left ventricular (LV) dysfunction and heart failure, typically with delayed onset. At the cellular level, this cardiotoxicity includes deranged cardiac glucose metabolism. Methods By reviewing the hospital records from January 2008 through December 2016, we selected HL patients meeting the following criteria: ≥ 18 year-old; first-line DOX-containing chemotherapy; no diabetes and apparent cardiovascular disease; 18-fluoro-deoxyglucose positron emission tomography (18FDG-PET) scans before treatment (PETSTAGING), after 2 cycles (PETINTERIM) and at the end of treatment (PETEOT); at least one echocardiography ≥ 6 months after chemotherapy completion (ECHOPOST). We then evaluated the changes in LV 18FDG standardized uptake values (SUV) during the course of DOX therapy, and the relationship between LV-SUV and LV ejection fraction (LVEF), as calculated from the LV diameters in the echocardiography reports with the Teicholz formula. Results Forty-three patients (35 ± 13 year-old, 58% males) were included in the study, with 26 (60%) also having a baseline echocardiography available (ECHOPRE). LV-SUV gradually increased from PETSTAGING (log-transformed mean 0.20 ± 0.27) to PETINTERIM (0.27 ± 0.35) to PETEOT (0.30 ± 0.41; P for trend
Article
Purpose Evaluation of [⁶⁸Ga]NODAGA-duramycin as a positron emission tomography (PET) tracer of cell death for whole-body detection of chemotherapy-induced organ toxicity. Procedures Tracer specificity of Ga-68 labeled NODAGA-duramycin was determined in vitro using competitive binding experiments. Organ uptake was analyzed in untreated and doxorubicin, busulfan, and cisplatin-treated mice 2 h after intravenous injection of [⁶⁸Ga]NODAGA-duramycin. In vivo data were validated by immunohistology and blood parameters. Results In vitro experiments confirmed specific binding of [⁶⁸Ga]NODAGA-duramycin. Organ toxicities were detected successfully using [⁶⁸Ga]NODAGA-duramycin PET/X-ray computed tomography (CT) and confirmed by immunohistochemistry and blood parameter analysis. Organ toxicities in livers and kidneys showed similar trends in PET/CT and immunohistology. Busulfan and cisplatin-related organ toxicities in heart, liver, and lungs were detected earlier by PET/CT than by blood parameters and immunohistology. Conclusion [⁶⁸Ga]NODAGA-duramycin PET/CT was successfully applied to non-invasively detect chemotherapy-induced organ toxicity with high sensitivity in mice. It, therefore, represents a promising alternative to standard toxicological analyses with a high translational potential.
Article
Purpose 2-Deoxy-2-[¹⁸F]fluoro-D-glucose ([¹⁸F]FDG) has been widely used for imaging brain metabolism. Tracer injection in anesthetized animals is a prerequisite for performing dynamic positron emission tomography (PET) scanning. Since preconditioning, as well as anesthesia, has been described to potentially influence brain [¹⁸F] FDG levels, this study evaluated how these variables globally and regionally affect both [¹⁸F] FDG uptake and kinetics in murine brain. Procedures Sixty-minute dynamic [¹⁸F] FDG PET scans were performed in adult male C57BL/6 mice anesthetized with isoflurane [control (in 100 % O2), in medical air, in 100 % O2 + insulin pre-treatment, and in 100 % O2 after 18 h fasting], ketamine/xylazine, sevoflurane, and chloral hydrate. An additional group was scanned after awake uptake. Blood glucose levels were determined, and data was analyzed by comparing percent injected dose per cc tissue (%ID/cc) and glucose influx rate and metabolic rate (MRGlu) calculated by Patlak plot. Results Ketamine/xylazine and chloral hydrate anesthesia induced a lower whole-brain uptake of [¹⁸F] FDG (2.86 ± 0.67 %ID/cc, p < 0.001; 4.25 ± 0.28 %ID/cc, p = 0.0179, respectively) compared to isoflurane anesthesia (5.04 ± 0.19 %ID/cc). In addition, protocols affected differently distribution of [¹⁸F] FDG uptake in brain regions. Ketamine/xylazine reduced [¹⁸F] FDG influx rate in murine brain (0.0135 ± 0.0009 vs 0.0247 ± 0.0014 ml/g/min; p < 0.005) and chloral hydrate increased MRGlu (66.72 ± 3.75 vs 41.55 ± 3.06 μmol/min/100 ml; p < 0.01) compared to isoflurane. Insulin-pretreated animals showed a higher influx rate (0.0477 ± 0.0101 ml/min/g; p < 0.05) but a reduced MRGlu (21.92 ± 3.12 μmol/min/100 ml; p < 0.01). Blood glucose levels were negatively correlated to [¹⁸F] FDG uptake and influx rate, but positively correlated to MRGlu. Conclusions Choice of anesthesia and pre-conditioning affect not only [¹⁸F] FDG uptake but also kinetics and regional distribution in the mouse brain. Both anesthesia and pre-conditioning should be carefully considered in the interpretation of [¹⁸F] FDG studies due to its great influence on the uptake and distribution of the tracer along the brain regions.
Article
Cardio-oncology is a growing field focused on the prevention and treatment of cardiovascular disease in oncologic patients. While a major focus of chemotherapy-related cardiac dysfunction has been on left ventricular ejection fraction, oncologic treatment can lead to cardiovascular pathology in a variety of ways. The use of multimodality imaging is essential to the care of these patients, with nuclear cardiology playing an important role. We will review nuclear cardiology’s history, its current role, and its promising future in cardio-oncology and the management of these patients.
Article
Background Oxidative stress and its interference on myocardial metabolism play a major role in Doxorubicin (DXR) cardiotoxic cascade. Methods Mice models of neuroblastoma (NB) were treated with 5 mg DXR/kg, either free (Free-DXR) or encapsulated in untargeted (SL[DXR]) or in NB-targeting Stealth Liposomes (pep-SL[DXR] and TP-pep-SL[DXR]). Control mice received saline. FDG-PET was performed at baseline (PET1) and 7 days after therapy (PET2). At PET2 Troponin-I and NT-proBNP were assessed. Explanted hearts underwent biochemical, histological, and immunohistochemical analyses. Finally, FDG uptake and glucose consumption were simultaneously measured in cultured H9c2 in the presence/absence of Free-DXR (1 μM). Results Free-DXR significantly enhanced the myocardial oxidative stress. Myocardial-SUV remained relatively stable in controls and mice treated with liposomal formulations, while it significantly increased at PET2 with respect to baseline in Free-DXR. At this timepoint, myocardial-SUV was directly correlated with both myocardial redox stress and hexose-6-phosphate-dehydrogenase (H6PD) enzymatic activity, which selectively sustain cellular anti-oxidant mechanisms. Intriguingly, in vitro, Free-DXR selectively increased FDG extraction fraction without altering the corresponding value for glucose. Conclusion The direct correlation between cardiac FDG uptake and oxidative stress indexes supports the potential role of FDG-PET as an early biomarker of DXR oxidative damage.
Article
Background The aim of this study was to investigate changes in myocardial uptake evaluated by oncologic ¹⁸F-fluorodeoxyglucose (FDG) PET/CT scans and to determine the relationship between myocardial FDG uptake and cancer therapy-induced cardiotoxicity in breast cancer patients who underwent anthracycline or trastuzumab. Methods We reviewed 121 consecutive patients who underwent oncologic FDG PET/CT and echocardiography at baseline and post-therapy with anthracyclines or trastuzumab for breast cancer. Grade in LV wall, uptake pattern in LV wall, and the presence of RV wall uptake were assessed by visual analysis, and the mean SUV in the LV and RV walls and the change of SUV (ΔSUV) between baseline and post-therapy PET/CT were measured by quantitative analysis. Multiple logistic regression analyses were performed to evaluate the association between PET parameters and cardiotoxicity. Results Fifteen patients (12%) showed cardiotoxicity after therapy. The cardiotoxic group tended to show more diffuse LV uptake, higher SUV, and ΔSUV of RV wall than the non-cardiotoxic group following therapy with anthracyclines or trastuzumab. Logistic regression analysis showed that the presence of RV wall uptake, SUV of RV wall (> 1.8), and ΔSUV of RV wall (> 0.4) were significantly associated with cardiotoxicity after controlling for age, radiotherapy, and treatment. Conclusions The presence of RV wall uptake and the increase of SUV of RV wall on post-therapy PET/CT were associated with cardiotoxicity in breast cancer patients who underwent anthracycline or trastuzumab. Oncologic FDG PET/CT scans can provide information regarding cancer therapy-induced cardiotoxicity as well as tumor response.
Article
[18F]fluoro‑2‑deoxyglucose (FDG) positron emission tomography (PET)‑computed tomography (CT) is a functional imaging modality based on glucose metabolism. The association between the maximum standardized uptake value (SUVmax) from 18F‑FDG PET‑CT scanning and epidermal growth factor receptor (EGFR) mutation status has, to the best of our knowledge, not previously been fully elucidated, and the potential mechanisms by which EGFR mutations alter FDG uptake are largely unknown. A total of 157 patients who were pathologically diagnosed with non‑small cell lung cancer (NSCLC) who underwent EGFR mutation testing and PET‑CT pretreatment between June 2015 and October 2017 were retrospectively analyzed. χ2 and univariate analyses were performed to identify the contributors to EGFR mutation. The receiver operating characteristic (ROC) curve was analyzed, and the area under the curve (AUC) was calculated. Glucose transporter 1 (GLUT1) and NADPH oxidase 4 (NOX4) expression, and reactive oxygen species (ROS) activity were detected in the A549 (wild‑type), PC‑9 (EGFR mutation‑positive, EGFR exon 19del) and NCI‑H1975 (EGFR mutation‑positive, combined with L858R and T790M substitution) cell lines. A total of 109 patients who met the criteria were enrolled, and 63 of those tested as EGFR mutation‑positive. The SUVmax values were significantly lower in patients with EGFR mutations (mean, 6.52±0.38) compared with in patients with wild‑type EGFR (mean, 9.37±0.31; P<0.001). Using univariate analysis, EGFR mutation status was significantly associated with sex, smoking status, tumor histology and SUVmax of the primary tumor. In the multivariate analysis, smoking status (never‑smoking), histopathology (adenocarcinoma) and SUVmax (≤9.91) were the statistically significant predictors of EGFR mutations. ROC curve analysis identified that the SUVmax cut‑off point was 9.92, for which the AUC was 0.75 (95% confidence interval, 0.68‑0.83). Reverse transcription‑polymerase chain reaction indicated that the GLUT1 mRNA decreased in the PC‑9 and NCI‑H1975 cell lines compared with the A549 cell line (0.82±0.07 and 0.72±0.04 vs. 0.98±0.04, respectively; P<0.05) and decreased ROS activity was observed in the PC‑9 cell line. Furthermore, the expression of NOX4 mRNA decreased by 20% in PC‑9 (P<0.01) and by 14% (P<0.05) in NCI‑H1975 cells. In addition, NOX4 protein expression decreased by 13% in PC‑9 and by 16% in NCI‑H1975 cells (both P<0.05) compared with the A549 cell line. The SUVmax could be considered to effectively predict EGFR mutation status of patients with NSCLC, and the EGFR mutation status may alter FDG uptake partially via the NOX4/ROS/GLUT1 axis.