Panitumumab is an anti-EGF receptor (EGFR) antibody approved for use in treatment of chemotherapy-refractory colorectal cancers lacking K-RAS mutations. Despite overall response rates approximating 10%, no marker predictive of clinical benefit has been identified. We describe a chemotherapy-refractory patient whose clinical condition necessitated rapid identification of an effective agent in whom we used 18F-fluorodeoxyglucose-positron emission tomography (FDG-PET)/computed tomographic scanning 48 hours after an initial dose of panitumumab to document a pharmacodynamic response to the antibody. The initial 46% ± 2.7% drop in SUVmax of four target lesions correlated with a partial response by Response Evaluation Criteria in Solid Tumors and a >90% drop in serum carcinoembryonic antigen at 8 weeks, indicating that an early decrease in FDG uptake may predict subsequent clinical benefit in response to anti-EGFR antibody therapy in colorectal cancer. Mol Cancer Ther; 11(7); 1385–8. ©2012 AACR.

Panitumumab and cetuximab are monoclonal anti-EGF receptor (EGFR) antibodies approved for use in chemotherapy-refractory colorectal cancer (CRC). Single agent overall response rates are 9% to 12% and no clinically useful positive predictors of response have been identified as yet, although tumors with K-RAS mutations are unlikely to respond (1). Studies of patients with absent or low tumor EGFR expression by immunohistochemistry have found that response rates are statistically indistinguishable from those patients with higher EGFR expression, indicating that immunohistochemical staining may not always detect physiologically relevant receptor expression (2, 3). An important consequence of EGFR activation is increased glucose uptake and retention, which supports tumor energetic and biosynthetic requirements (4, 5, 6), suggesting that 18F-fluorodeoxyglucose-positron emission tomography (FDG-PET) may be a sensitive assay for detection of changes in EGFR activity (7, 8). Here, we describe a chemotherapy-refractory patient whose tumor was negative for EGFR expression by immunohistochemistry in whom we used FDG-PET to show a panitumumab-mediated decrease in FDG accumulation over a period of 48 hours, leading to a progressive decline in metabolic activity and partial response by Response Evaluation Criteria in Solid Tumors (RECIST; ref. 9) criteria.

Case report

The patient is a 31-year-old female referred for adjuvant chemotherapy following laparoscopic resection of a T3N2MO stage IIIC sigmoid adenocarcinoma. She had no family history of CRC, no other neoplastic lesions on colonoscopy, and genetic analyses were inconsistent with hereditary nonpolyposis CRC. Six months of adjuvant FOLFOX chemotherapy was administered and follow-up FDG-PET/computed tomography (CT) was negative.

Four months later she presented with abdominal pain and CT revealed a 14-cm complex right ovarian mass with 3 hepatic lesions. The patient underwent laparotomy with hysterectomy and bilateral salpingo-oophorectomy revealing metastatic colon cancer. Nine cycles of FOLFIRI chemotherapy with bevacizumab were administered, with abdominal CT and FDG-PET/CT showing partial responses of all hepatic lesions without evidence of other disease. The patient was taken to laparotomy for radiofrequency ablation however multiple small peritoneal implants were noted. The largest central metastasis was left as measurable disease but the 2 superficial hepatic metastases were ablated following biopsy. The biopsy was negative for K-RAS mutation and for EGFR expression (immunohistochemistry) by 2 independent laboratories. The patient was then treated with 6 cycles of single-agent irinotecan with bevacizumab and filgrastim on an every 3-week schedule, with stability of the central hepatic lesion on serial CT scans, after which she declined further therapy.

Nine weeks later she presented with symptomatic and radiologic evidence of bowel obstruction. A FDG-PET/CT revealed multiple peritoneal and mesenteric metastases, multiple new liver metastases, and a left supraclavicular lymph node metastasis. She underwent laparotomy with bypass of obstructing lesions in the pelvis and presented to the medical oncology clinic for further therapy 4 weeks later. Given evidence that patients with EGFR immunohistochemical-negative tumors can respond to anti-EGFR antibodies (2, 3), it was decided to proceed with panitumumab treatment. However, to avoid continuation of ineffective therapy, we attempted to determine whether physiologically relevant levels of EGFR, undetectable by immunohistochemistry, were targeted by the antibody using early FDG-PET scanning.

Treatment and evaluation

Panitumumab (Amgen) was administered i.v. at a dose of 6 mg/kg every 2 weeks with carcinoembryonic antigen (CEA) determination before each treatment. PET imaging was conducted with integrated CT on a Phillips Gemini large bore TOF PET/CT 90 minutes following injection of 10 to 12 mCi of FDG (Cardinal Health). Scanning was carried out craniocaudal at 1.75 minutes per bed position, FOV 576 mm in 144 × 144 matrix in 3-D TOF mode generating 4-mm slices. For each study the subject had been fasting greater than 6 hours and blood glucose determinations were below 120 mg/dL at the time of FDG administration. FDG-PET/CT scans were obtained immediately before the first dose, 48 hours later, and immediately before the fifth dose of therapy.

Imaging analysis

Four target lesions were identified on the baseline PET/CT scan based on the ability to accurately measure them on the noncontrast CT portion of the study. These were a left supraclavicular node, 2 hepatic lesions and a preaortic lymph node. Volume of interest to encompass the entire lesion was defined on the baseline image and reproduced on subsequent scans. SUVmax and longest diameter were determined at sequential time points and percentage of reduction was calculated for each lesion.

FDG-PET imaging showed a mean 46% ± 2.7% drop in the SUVmax of the 4 target lesions over 48 hours following the initial panitumumab dose, without meaningful change in size (Fig. 1; Table 1). Following the completion of 8 weeks of treatment, the mean SUVmax of the target lesions dropped by 74.4% ± 8.4%, which correlated with a 47.5% decrease in the sum of the longest diameters, consistent with a partial response by RECIST criteria. This response also correlated with a decline in CEA from122 ng/mL before treatment to 11 ng/mL after 8 weeks of treatment (Fig. 2).

Figure 1.

Representative noncontrast CT and fused FDG-PET/CT cuts. A, upper thorax illustrating the enlarged left supraclavicular node (arrow). B, superior right hepatic lobe illustrating multiple liver metastases with progressively decreasing FDG uptake. C, mid-liver illustrating the large central hepatic lesion. Note the increased calcification after 8 weeks of therapy.

Figure 1.

Representative noncontrast CT and fused FDG-PET/CT cuts. A, upper thorax illustrating the enlarged left supraclavicular node (arrow). B, superior right hepatic lobe illustrating multiple liver metastases with progressively decreasing FDG uptake. C, mid-liver illustrating the large central hepatic lesion. Note the increased calcification after 8 weeks of therapy.

Close modal
Figure 2.

Progressive decline in CEA after initiation of panitumumab (arrow). Institutional normal value 0 to 5 ng/mL.

Figure 2.

Progressive decline in CEA after initiation of panitumumab (arrow). Institutional normal value 0 to 5 ng/mL.

Close modal
Table 1.

Change in FDG uptake and size of target lesions following panitumumab therapy

Target lesionsBaselineDay 2Day 58D2 decreaseD58 decrease
L supraclavicular node 
 SUVmax 7.1 3.9 45.07% 71.83% 
 Diameter,a mm 20.3 21.1 10.5 −3.94% 48.28% 
R hepatic lobe 
 SUVmax 12.9 6.6 1.8 48.84% 86.05% 
 Diameter,a mm 26.5 25.5 13.6 3.77% 48.68% 
Central hepatic 
 SUVmax 12.1 6.9 4.1 42.98% 66.12% 
 Diameter,a mm 45.0 40.4 24.6 10.22% 45.33% 
Preaortic node 
 SUVmax 7.9 4.1 2.1 48.10% 73.42% 
 Diameter,a mm 19.4 20.1 9.7 −3.61% 50.00% 
Target lesionsBaselineDay 2Day 58D2 decreaseD58 decrease
L supraclavicular node 
 SUVmax 7.1 3.9 45.07% 71.83% 
 Diameter,a mm 20.3 21.1 10.5 −3.94% 48.28% 
R hepatic lobe 
 SUVmax 12.9 6.6 1.8 48.84% 86.05% 
 Diameter,a mm 26.5 25.5 13.6 3.77% 48.68% 
Central hepatic 
 SUVmax 12.1 6.9 4.1 42.98% 66.12% 
 Diameter,a mm 45.0 40.4 24.6 10.22% 45.33% 
Preaortic node 
 SUVmax 7.9 4.1 2.1 48.10% 73.42% 
 Diameter,a mm 19.4 20.1 9.7 −3.61% 50.00% 

aLongest diameter.

Both the chimeric anti-EGFR monoclonal antibody cetuximab and the fully human monoclonal panitumumab have been approved for the treatment of chemotherapy-refractory CRC. By binding to EGFR in the inactive state, these antibodies inhibit activation of intracellular signaling, including the RAS-RAF-MAPK and phosphoinositide 3-kinase (PI3K)-Akt pathways that play critical roles in regulating growth, apoptosis, and cellular glucose transport and retention (6, 10, 11). Retrospective analyses have showed that the 40% of patients with K-RAS mutations are unlikely to respond but no clinically applicable biomarkers predictive of benefit have been identified despite 10% overall response rates, which are only modestly improved to 17% in the K-RAS wild-type population (1, 12). Thus therapeutic trials lasting 6 to 12 weeks are required to identify the minority of patients with wild-type K-RAS who will derive clinical benefit. For the majority of patients who do not benefit, these therapeutic trials can delay the start of alternative therapies including clinical trials of investigational agents and come at great expense, given the high price of these biologic agents.

In the case described here, rapid identification of a potentially effective therapy was necessitated by the patient's progressive clinical deterioration from her abdominal tumor burden. Given her prior therapies, panitumumab seemed the agent with the best therapeutic index despite her tumor being negative for EGFR expression by immunohistochemistry. While it might be expected that tumors lacking EGFR expression would not respond to this highly specific agent, tumors that express EGFR below the limit of detectability of the immunohistochemical assay might. Knowing that this patient's tumor was FDG-avid, we reasoned that acute changes in FDG uptake by PET could indicate modulation of EGFR function. Preclinical studies using lung and breast cancer cell lines showed marked inhibition of glucose uptake and retention over a period of hours following treatment with the kinase inhibitor gefitinib (7) and cetuximab (8), respectively. In addition, small clinical studies of gefitinib treatment of lung adenocarcinomas enriched for activating EGFR mutations have showed significant decreases in FDG uptake by PET after 2 days (13, 14). On the basis of these studies and those of radiolabeled panitumumab pharmacokinetics (J.L. Tatum, unpublished data) we chose a 48-hour time point to reassess FDG uptake by PET/CT.

At 48 hours after the initial panitumumab treatment we noted uniform nearly 50% declines in target lesion FDG uptake (Table 1). This correlated well with a progressive decline in CEA (Fig. 2) and subsequent tumor shrinkage consistent with partial response by RECIST criteria at 8 weeks, with grade II acneiform rash as the only adverse event. This case shows the feasibility of using FDG/PET scanning as an early downstream pharmacodynamic marker of anti-EGFR monoclonal antibody activity. While FDG-PET has been routinely used to assess response in clinical trials and increasingly in clinical practice, when used to evaluate the efficacy of nontargeted cytotoxic therapy a sufficient time interval must be allowed for the acute tumor adaptive and host immunologic responses to subside, as well as a modest reduction in viable cell mass to occur, for interpretable results (15). However, when used to assess activity of agents targeting receptor tyrosine kinases regulating glucose metabolism, such as imatinib for gastrointestinal stromal tumors and gefitinib for lung adenocarcinomas, early changes in FDG uptake can be predictive of clinical response (14, 16). In these tumors, activating mutations in KIT and EGFR, respectively, are potent predictors of clinical response but in CRC such positive predictors of response to anti-EGFR antibodies do not exist. Use of FDG-PET in this setting may be a particularly sensitive assay owing to the ability of these antibodies to not only disrupt downstream signaling affecting glucose metabolism but also to potentially disrupt EGFR-mediated stabilization of the sodium/glucose cotransporter-1 (5).

While this case illustrates the feasibility of using early FDG-PET to show a pharmacodynamic response to anti-EGFR antibodies, which correlated with a clear partial response, this correlation may not always hold. The flexibility of tumors to use alternative receptors to activate downstream signaling pathways regulating glucose metabolism has been well-established (17, 18, 19), so that some tumors may only experience a transient inconvenience following inhibition of EGFR, and those patients may not experience clinical benefit despite an early pharmacodynamic response. Conversely, in some cases a late immune-mediated response to antibody treatment (1, 20) may be the most important factor in producing clinical benefit and thus lack of an early decrease in FDG uptake may not predict lack of benefit. Therefore, a prospective clinical trial will be required to establish the power of early FDG-PET to predict clinical benefit from anti-EGFR monoclonal antibody therapy. If early FDG-PET is proven to be predictive of clinical response, the costly practice of a protracted therapeutic trial in the majority of patients destined not to benefit can be eliminated, allowing those patients to be quickly shifted to alternative therapies, especially clinical trials of novel agents. In addition, such a trial should enhance our understanding of the mechanisms by which these biologic agents induce clinical response and provide insight as to why they are efficacious in only a minority of patients.

No potential conflicts of interest were disclosed.

Conception and design: G.W. Krystal, J.L. Tatum

Development of methodology: G.W. Krystal, J.L. Tatum

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): G.W. Krystal, J.L. Tatum

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): G.W. Krystal, J.L. Tatum

Writing, review, and/or revision of the manuscript: G.W. Krystal, E. Alesi, J.L. Tatum

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): G.W. Krystal, J.L. Tatum

Study supervision: J.L. Tatum

1.
Ciardiello
F
,
Tortora
G
. 
EGFR antagonists in cancer treatment
.
N Engl J Med
2008
;
358
:
1160
74
.
2.
Chung
KY
,
Shia
J
,
Kemeny
NE
,
Shah
M
,
Schwartz
GK
,
Tse
A
, et al
Cetuximab shows activity in colorectal cancer patients with tumors that do not express the epidermal growth factor receptor by immunohistochemistry
.
J Clin Oncol
2005
;
23
:
1803
10
.
3.
Hecht
JR
,
Mitchell
E
,
Neubauer
MA
,
Burris
HA
 III
,
Swanson
P
,
Lopez
T
, et al
Lack of correlation between epidermal growth factor receptor status and response to panitumumab monotherapy in metastatic colorectal cancer
.
Clin Cancer Res
2010
;
16
:
2205
13
.
4.
Vander
Heiden MG
,
Cantley
LC
,
Thompson
CB
. 
Understanding the Warburg effect: the metabolic requirements of cell proliferation
.
Science
2009
;
324
:
1029
33
.
5.
Weihua
Z
,
Tsan
R
,
Huang
WC
,
Wu
Q
,
Chiu
CH
,
Fidler
IJ
, et al
Survival of cancer cells is maintained by EGFR independent of its kinase activity
.
Cancer Cell
2008
;
13
:
385
93
.
6.
Engelman
JA
,
Cantley
LC
. 
A sweet role for EGFR in cancer
.
Cancer Cell
2008
;
13
:
375
6
.
7.
Su
H
,
Bodenstein
C
,
Dumont
RA
,
Seimbille
Y
,
Dubinett
S
,
Phelps
ME
, et al
Monitoring tumor glucose utilization by positron emission tomography for the prediction of treatment response to epidermal growth factor receptor kinase inhibitors
.
Clin Cancer Res
2006
;
12
:
5659
67
.
8.
Smith
TA
,
Cheyne
RW
. 
Predicting tumour response to anti-HER1 therapy using medical imaging: a literature review and in vitro study of [18F]-FDG incorporation by breast cancer cells responding to cetuximab
.
Br J Biomed Sci
2011
;
68
:
158
66
.
9.
Eisenhauer
EA
,
Therasse
P
,
Bogaerts
J
,
Schwartz
LH
,
Sargent
D
,
Ford
R
, et al
New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1)
.
Eur J Cancer
2009
;
45
:
228
47
.
10.
Scaltriti
M
,
Baselga
J
. 
The epidermal growth factor receptor pathway: a model for targeted therapy
.
Clin Cancer Res
2006
;
12
:
5268
72
.
11.
Yun
J
,
Rago
C
,
Cheong
I
,
Pagliarini
R
,
Angenendt
P
,
Rajagopalan
H
, et al
Glucose deprivation contributes to the development of KRAS pathway mutations in tumor cells
.
Science
2009
;
325
:
1555
9
.
12.
Amado
RG
,
Wolf
M
,
Peeters
M
,
Van Cutsem
E
,
Siena
S
,
Freeman
DJ
, et al
Wild-type KRAS is required for panitumumab efficacy in patients with metastatic colorectal cancer
.
J Clin Oncol
2008
;
26
:
1626
34
.
13.
Sunaga
N
,
Oriuchi
N
,
Kaira
K
,
Yanagitani
N
,
Tomizawa
Y
,
Hisada
T
, et al
Usefulness of FDG-PET for early prediction of the response to gefitinib in non-small cell lung cancer
.
Lung Cancer
2008
;
59
:
203
10
.
14.
Takahashi
R
,
Hirata
H
,
Tachibana
I
,
Shimosegawa
E
,
Inoue
A
,
Nagatomo
I
, et al
Early [18F]fluorodeoxyglucose positron emission tomography at 2 days of gefitinib treatment predicts clinical outcome in patients with adenocarcinoma of the lung
.
Clin Cancer Res
2012
;
18
:
220
8
.
15.
Ben-Haim
S
,
Ell
P
. 
18F-FDG PET and PET/CT in the evaluation of cancer treatment response
.
J Nucl Med
2009
;
50
:
88
99
.
16.
Stroobants
S
,
Goeminne
J
,
Seegers
M
,
Dimitrijevic
S
,
Dupont
P
,
Nuyts
J
, et al
18FDG-Positron emission tomography for the early prediction of response in advanced soft tissue sarcoma treated with imatinib mesylate (Glivec)
.
Eur J Cancer
2003
;
39
:
2012
20
.
17.
Warshamana-Greene
GS
,
Litz
J
,
Buchdunger
E
,
Hofmann
F
,
García-Echeverría
C
,
Krystal
GW
. 
The insulin-like growth factor-I (IGF-I) receptor kinase inhibitor NVP-ADW742, in combination with STI571, delineates a spectrum of dependence of small cell lung cancer on IGF-I and stem cell factor signaling
.
Mol Cancer Ther
2004
;
3
:
527
35
.
18.
Stommel
JM
,
Kimmelman
AC
,
Ying
H
,
Nabioullin
R
,
Ponugoti
AH
,
Wiedemeyer
R
, et al
Coactivation of receptor tyrosine kinases affects the response of tumor cells to targeted therapies
.
Science
2007
;
318
:
287
90
.
19.
Engelman
JA
,
Zejnullahu
K
,
Mitsudomi
T
,
Song
Y
,
Hyland
C
,
Park
JO
, et al
MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling
.
Science
2007
;
316
:
1039
43
.
20.
Schneider-Merck
T
,
Lammerts van Bueren
JJ
,
Berger
S
,
Rossen
K
,
van Berkel
PH
,
Derer
S
, et al
Human IgG2 antibodies against epidermal growth factor receptor effectively trigger antibody-dependent cellular cytotoxicity but, in contrast to IgG1, only by cells of myeloid lineage
.
J Immunol
2010
;
184
:
512
20
.