positron emission tomography and colorectal cancer

18
Critical Reviews in Oncology/Hematology 77 (2011) 30–47 Positron emission tomography and colorectal cancer Michael Lin a,b,c,, Karen Wong b,c,d , Weng Leong Ng b,c,e , Ivan Ho Shon a,b , Matthew Morgan c,f a Department of Nuclear Medicine and PET, Liverpool Hospital, Sydney, Australia b University of New South Wales, Sydney, Australia c South Western Sydney Colorectal Tumour Group, Sydney South West Area Health Service, Sydney, Australia d Department of Radiation Oncology, Liverpool Hospital, Sydney, Australia e Department of Medical Oncology, Liverpool Hospital, Sydney, Australia f Department of Colorectal Surgery, Bankstown Hospital, Sydney, Australia Accepted 30 April 2010 Contents 1. Introduction ........................................................................................................... 31 2. Positron emission tomography .......................................................................................... 31 2.1. PET and PET-CT ................................................................................................ 31 2.2. PET-CT colonography ........................................................................................... 31 3. Clinical applications ................................................................................................... 31 3.1. Initial staging ................................................................................................... 31 3.1.1. Rectal cancer ........................................................................................... 31 3.1.2. Colon cancer ........................................................................................... 32 3.2. Staging of recurrent colorectal cancer ............................................................................. 32 3.2.1. Evaluation of hepatic metastases for liver resection ......................................................... 33 3.2.2. Assessment of local recurrence for salvage treatment ....................................................... 34 3.2.3. Early detection of biochemical recurrence (rising serum CEA levels) ......................................... 34 3.2.4. To identify peritoneal carcinomatosis ...................................................................... 34 3.3. Prognostication and evaluation of treatment response ............................................................... 34 3.3.1. Neoadjuvant therapy for rectal cancer ..................................................................... 35 3.3.2. Chemotherapy .......................................................................................... 36 3.3.3. Local ablative therapy ................................................................................... 36 3.4. Radiotherapy planning ........................................................................................... 38 3.5. Post-treatment surveillance ....................................................................................... 40 3.6. Screening ...................................................................................................... 40 3.7. Incidental colonic FDG uptake ................................................................................... 40 4. Future ................................................................................................................ 42 4.1. PET-MR ....................................................................................................... 42 4.2. Non-FDG tracers ................................................................................................ 42 5. Conclusions ........................................................................................................... 42 Reviewer ............................................................................................................. 43 References ............................................................................................................ 43 Biographies ........................................................................................................... 47 Corresponding author at: Department of Nuclear Medicine and PET, 1 Elizabeth Drive, Liverpool Hospital, Liverpool, New South Wales 2170, Australia. Tel.: +61 2 9828 3515; fax: +61 2 9828 3529. E-mail address: [email protected] (M. Lin). 1040-8428/$ – see front matter. Crown Copyright © 2010 Published by Elsevier Ireland Ltd. All rights reserved. doi:10.1016/j.critrevonc.2010.04.011

Upload: michael-lin

Post on 26-Jun-2016

218 views

Category:

Documents


3 download

TRANSCRIPT

Page 1: Positron emission tomography and colorectal cancer

C

12

3

4

5

T

1d

Critical Reviews in Oncology/Hematology 77 (2011) 30–47

Positron emission tomography and colorectal cancer

Michael Lin a,b,c,∗, Karen Wong b,c,d, Weng Leong Ng b,c,e,Ivan Ho Shon a,b, Matthew Morgan c,f

a Department of Nuclear Medicine and PET, Liverpool Hospital, Sydney, Australiab University of New South Wales, Sydney, Australia

c South Western Sydney Colorectal Tumour Group, Sydney South West Area Health Service, Sydney, Australiad Department of Radiation Oncology, Liverpool Hospital, Sydney, Australiae Department of Medical Oncology, Liverpool Hospital, Sydney, Australia

f Department of Colorectal Surgery, Bankstown Hospital, Sydney, Australia

Accepted 30 April 2010

ontents

. Introduction. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 31

. Positron emission tomography . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 312.1. PET and PET-CT. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 312.2. PET-CT colonography . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 31

. Clinical applications . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 313.1. Initial staging . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 31

3.1.1. Rectal cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 313.1.2. Colon cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 32

3.2. Staging of recurrent colorectal cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 323.2.1. Evaluation of hepatic metastases for liver resection . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 333.2.2. Assessment of local recurrence for salvage treatment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 343.2.3. Early detection of biochemical recurrence (rising serum CEA levels) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 343.2.4. To identify peritoneal carcinomatosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 34

3.3. Prognostication and evaluation of treatment response . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 343.3.1. Neoadjuvant therapy for rectal cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 353.3.2. Chemotherapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 363.3.3. Local ablative therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 36

3.4. Radiotherapy planning . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 383.5. Post-treatment surveillance . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 403.6. Screening . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 403.7. Incidental colonic FDG uptake . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 40

. Future . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 42

4.1. PET-MR . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 424.2. Non-FDG tracers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 42

. Conclusions. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 42Reviewer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 43References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 43Biographies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 47

∗ Corresponding author at: Department of Nuclear Medicine and PET, 1 Elizabeth Drive, Liverpool Hospital, Liverpool, New South Wales 2170, Australia.el.: +61 2 9828 3515; fax: +61 2 9828 3529.

E-mail address: [email protected] (M. Lin).

040-8428/$ – see front matter. Crown Copyright © 2010 Published by Elsevier Ireland Ltd. All rights reserved.oi:10.1016/j.critrevonc.2010.04.011

Page 2: Positron emission tomography and colorectal cancer

A

(iPmC

K

1

mwbt2mp[cl1mfia[cP

2

2

fPlPaaasn

rucmPu(b

M. Lin et al. / Critical Reviews in Oncology/Hematology 77 (2011) 30–47 31

bstract

Colorectal cancer (CRC) is a major cause of cancer-related morbidity and mortality. Molecular imaging using positron emission tomographyPET) is now an integral part of multidisciplinary cancer care. In this review, we discuss the role of PET in CRC including well establishedndications in the assessment of recurrent disease and emerging applications such as initial staging, monitoring therapy efficacy and usingET for radiotherapy planning. With rapid advancement in imaging technology, we also discuss the future potential of combining PET andagnetic resonance imaging and the use of novel radiotracers.rown Copyright © 2010 Published by Elsevier Ireland Ltd. All rights reserved.

ncolog

aFb

2

aiepscwi6tfwcCtppia

3

3

eimctcr

eywords: Positron emission tomography; PET; FDG; Colorectal cancer; O

. Introduction

Colorectal cancer (CRC) is a major cause of cancer-relatedorbidity and mortality in developed countries. In 2006 thereere 412,900 new cases of CRC in Europe and responsi-le for 217,400 deaths [1]. In the United States, CRC is thehird most common cause of cancer mortality estimated in009 [2]. Over the last two decades, there has been an enor-ous growth in the literature demonstrating the benefits of

ositron emission tomography (PET) in many malignancies3] and is now an integral part of multidisciplinary cancerare. The initial results from the United States National Onco-ogic PET Registry of 22,975 studies in 21,419 patients from178 centres showed that physicians changed their intendedanagement in more than a third of cases in light of PETndings [4]. Since the basic principles and various oncologicpplications of PET were reviewed in this Journal recently5], the general principles of PET scanning will only be dis-ussed in brief. In this review, we shall focus on the role ofET in CRC.

. Positron emission tomography

.1. PET and PET-CT

PET detects pairs of photons emitted in opposite directionsollowing the annihilation of positron emitting radioisotopes.ET cameras record data in three dimensions and allow the

ocalisation of this process in vivo. The advent of combinedET and computed tomography (CT) scanners (PET-CT)llows the fusion of functional and anatomical information insingle scan with an improvement in diagnostic confidence

nd accuracy compared to PET and CT data viewed side byide, PET alone or CT alone [6–8]. As a result, PET-CT hasow largely replaced PET-only systems.

[Fluorine-18]-2-fluoro-2-deoxy-d-glucose (FDG) is aadiolabelled glucose analogue and is by far the most widelysed positron emitting radiopharmaceutical for PET. Becauseancer cells exhibit enhanced glycolytic metabolism, thisolecular process is depicted by mapping FDG uptake using

ET cameras. The degree of metabolic activity or FDGptake by tissue, expressed as a standardised uptake valueSUV), can also be quantitatively assessed and comparedetween studies (e.g. pre- and post-treatment). PET allows

3

ra

y; Therapy response

unique insight into tumour biology and the correlation ofDG metabolism and tumour proliferative rates may in turne useful in determining prognosis in CRC [9].

.2. PET-CT colonography

There has been recent interest in combining PET-CTnd virtual colonography as an “all-in-one” staging modal-ty. By using pharmacologic bowel relaxation, rectal waternema to maintain bowel distension and PET imaging in therone position, Veit-Haibach and colleagues demonstrated alight improvement in the accuracy of staging using PET-CTolonography compared with PET and CT [10]. In 47 patientsith 50 lesions, the overall TNM stage was correctly assigned

n 74% of the patients using FDG PET-CT colonography and4% using CT and PET. Patients with incomplete conven-ional colonoscopy due to obstructing lesions may also benefitrom PET-CT colonography. In a small number of patientsith CRC (n = 13) that could not be traversed by optical

olonoscopy, PET-CT colonography detected 2 synchronousRC proximal to the stenosis [11]. The major advantage of

he procedure is that it could be performed without bowelreparation. Despite limited data showing its feasibility andotential utility, the precise role of PET-CT colonographys not well defined at this point in time and should not bedopted as standard protocol.

. Clinical applications

.1. Initial staging

The role of FDG PET in staging CRC is not wellstablished and is currently not recommended as a routinenvestigation [1]. While there is emerging evidence of incre-

ental benefit in addition to conventional modalities in rectalancer, its role in colon cancer remains controversial. Fur-hermore, the studies often comprised both colon and rectalancers from which subgroup analyses were not always car-ied out.

.1.1. Rectal cancerTumour stage is the strongest predictor of recurrence in

ectal cancer [12]. Accurate staging determines different ther-peutic options such as the type of surgery and the need for

Page 3: Positron emission tomography and colorectal cancer

3 Oncolo

plitsTitfflotttaPio

intli6mnbfsvbtrwo[cwfiooMtnpdba(Sbsbm

tfnrimt

3

t[ssrraoStb5oPov

ot[p[oocp

ubpsdfn

3

aqMa

2 M. Lin et al. / Critical Reviews in

reoperative neoadjuvant therapy for downstaging. Transanalocal excision or transanal endoscopic microsurgery (TEMS)s usually undertaken for early rectal cancers and rectal resec-ion with total mesorectal excision (TME) as the preferredurgical option for more advanced tumours (mainly T2 or3). TME achieves maximum radial clearance for cancers

n the middle or lower third of the rectum and preopera-ive evaluation of tumour extent in relation to the mesorectalascia has gained particular importance. A positive circum-erential resection margin (CRM) is strongly predictive ofocal recurrence following surgery [12–15]. Magnetic res-nance imaging (MRI) has exquisite soft tissue definitionhat allows a detailed evaluation of the rectal wall, mesorec-al fascia, extent of local invasion and the likelihood of aumour-free CRM [16,17]. Endoscopic ultrasound (EUS) islso useful in local staging of rectal cancers [18]. Currently,ET is unlikely to contribute significantly towards determin-

ng the T-category of the primary tumour in addition to MRIr EUS due to its limited spatial resolution.

Where PET appears promising in staging rectal cancers isn the detection of lymph node and distant metastases. Lymphode involvement in rectal cancer is prognostic [14], however,he assessment by various imaging modalities remains a chal-enge. A meta-analysis (n = 90) evaluating CT, MRI and EUSn lymph node staging revealed sensitivities of 55%, 66% and7% respectively [18]. Assessment of lymph node involve-ent based on size criteria is unreliable and in one study,

o significant difference in size was found on MRI betweenenign and malignant lymph nodes [19]. On the other hand,unctional imaging with FDG PET may be useful and recenttudies have shown the benefit of PET in addition to con-entional staging in primary rectal cancer. In a study (n = 37)y Gearhart and colleagues, the addition of FDG PET-CTo spiral CT and transrectal ultrasound (TRUS) or MRIevealed discordant findings in 38% of patients, 50% of whomere upstaged, and the commonest finding was the detectionf unsuspected inguino-femoral or pelvic lymphadenopathy20]. Discordant findings were found to be significantly moreommon in patients with low-lying rectal cancers than thoseith mid or upper cancers (P = 0.0027). These discordantndings resulted in a change in planned management in 27%f patients. The largest series so far involved 83 patients allf whom were staged with abdominal and pelvic CT scan,RI or TRUS or both, as well as PET-CT [21]. A change in

umour stage occurred in 31% of patients after PET-CT andearly half the patients were upstaged by identifying unsus-ected systemic and lymph node metastases. All the casesown-staged by PET-CT were also proven to be accurateased on resected specimens and clinical follow-up. Over-ll PET-CT led to a high impact change of managementchange in treatment modality or intent) in 14% of patients.imilar results were found in other studies with smaller num-

er of subjects that used PET alone or combined PET-CTcanners [22–25]. However, in one series, the incrementalenefit of PET over multi-detector CT scan was reportedlyuch smaller [26]. Early experience with the use of con-

tsrt

gy/Hematology 77 (2011) 30–47

rast enhancement in combined PET-CT studies suggestedurther diagnostic benefit may be gained when compared withon-enhanced PET-CT in depicting regional nodal status inectal cancer [27]. For detection of distant metastases, PETs highly accurate in detecting synchronous liver and lung

etastases and may reveal more extensive metastatic diseasehat precludes metastasectomy [23,28].

.1.2. Colon cancerPET has a high sensitivity for detecting the primary

umour in the colon ranging between 95% and 100%24,26,29–31]. In contrast, for lymph node staging, mosttudies have reported low sensitivity (29–43%) and highpecificity (56–96%) [23,24,29,31]. The high false negativeates are most likely due to a combination of limited spatialesolution of PET, high FDG activity in the primary tumournd physiologic tracer excretion in the bladder which canbscure detection of closely adjacent involved lymph nodes.ize of involved lymph nodes is also a factor. In a morphome-

ric study of colon cancer specimens, lymph nodes infiltratedy metastases averaged 5.9 mm in diameter and more than0% were less than 5 mm [32], which approaches the limitsf resolution of clinical PET systems. The accuracy of FDGET in nodal staging has not been shown to be superior to thatf multi-detector CT scan and may be of limited incrementalalue [24].

PET has been shown to be more sensitive in the detectionf distant metastases not recognized on conventional diagnos-ic modalities in a small number of patients with colon cancer23,29,31], in particular extra-hepatic distant metastases inatients who are potential candidates for metastasectomy23,24]. In a pre-selected group of patients (n = 100) with pre-perative serum CEA levels ≥10 ng/mL or equivocal findingsn CT scan for metastatic disease, the addition of PET-CThanged the treatment modality or extent of surgery in 27%atients with colorectal cancers [33].

Although the data is limited and consisted of a mixed pop-lation of colon and rectal cancer patients, there appeared toe potential incremental benefit with PET in only selectiveatients with colon cancer. PET is most useful in patients withuspected but inconclusive synchronous distant metastaticisease on conventional modalities. Routine PET scanningor the initial staging of colon cancer, currently, is probablyot justified.

.2. Staging of recurrent colorectal cancer

Recurrent CRC following curative resection is commonnd occurs in approximately 40% of patients; the most fre-uent sites are hepatic, pulmonary or loco-regional [34].etastasectomy of solitary hepatic or pulmonary lesions

nd resection of isolated local recurrence in CRC is poten-

ially curative. Previous reports have documented 5 yearurvival rates of 20–31% following resection of localecurrence [35,36] and 37–47% after hepatic metastasec-omy [37,38]. However a significant proportion of patients
Page 4: Positron emission tomography and colorectal cancer

M. Lin et al. / Critical Reviews in Oncolo

Fig. 1. (a) A 64-year-old man with previous right hemicolectomy for aT2N0M0 adenocarcinoma of the caecum 3 years ago presented with apresumed solitary hepatic metastasis in the right lobe of liver for potentiallycurative metastasectomy. Preoperative FDG PET scan for restagingdemonstrated intense focal uptake in segment VI/VII of the liver (arrow) aswell as a small focus of mildly increased FDG accumulation within a 9 mmlymph node in the medial left supraclavicular fossa (arrowhead) suspiciousfor extrahepatic metastases. Fine needle aspiration biopsy of the lymph nodeconfirmed metastatic adenocarcinoma thereby obviating futile surgery. In therest of the scan, there was normal FDG uptake in the base of brain, laryngeal

hypmCai[ia

3r

wtttmdeifdtMtePlssTPAandscCt[

tmrni

mTslfg

gy/Hematology 77 (2011) 30–47 33

ave unresectable disease at surgery or relapse within 1–2ears following resection, implying suboptimal preoperativeatient selection by conventional means [39]. The incre-ental benefit with the addition of FDG PET in restagingRC is well established, cost effective [40] and has mademajor impact on patient management mainly by identify-

ng further unexpected metastases or more extensive disease41–44]. FDG PET is an invaluable tool in recurrent CRC ands incorporated into management guidelines and diagnosticlgorithms [1,45,46].

.2.1. Evaluation of hepatic metastases for liveresection

FDG uptake in hepatic metastases from colorectal originas demonstrated on a single-slice PET scanner more than

wo and a half decades ago [47]. Since the initial observa-ion, there has been an abundance of literature examininghe use of PET in this group of patients. Two systematic

eta-analyses comprising 32 and 61 studies respectivelyemonstrated high diagnostic values of FDG PET in thevaluation of liver metastases [48,49]. The pooled sensitiv-ty and specificity of PET was 88% and 96.1% respectivelyor hepatic disease and 91.5% and 95.4% for extra-hepaticisease [48]. The sensitivity of PET for detecting liver metas-ases was superior to other imaging modalities including

RI on a per patient basis [49]. When compared with CT,he superiority of PET is in the detection of unsuspectedxtra-hepatic disease [44,48,50] (Fig. 1) and the yield ofET correlated with clinical risk scores [37,51]. For small

iver lesions (<1 cm), however, several recent studies havehown MRI with liver-specific contrast agents or dedicatedequences to be slightly more sensitive than PET-CT [50,52].he improved survival observed in patients screened withET prior to surgery reflects better patient selection [53,54].

retrospective analysis of 461 patients showed preoper-tive PET imaging was associated with a lower risk ofon-therapeutic laparotomy [55]. A recent phase III ran-omised, multicenter trial (n = 150) further underlined thetrength of PET by showing the addition of PET to CT-basedonventional work-up prior to hepatic resection in metastaticRC (mCRC) reduced unnecessary laparotomies from 45%

o 28% and avoided futile surgery in 1 out of every 6 patients56].

Several interesting observations have also been made inhe era of using PET for the assessment of CRC hepatic

etastases. In FDG PET-staged patients who undergo liveresection, early recurrence (<1yr) was observed predomi-antly in the liver whereas later recurrences (>1yr) occurredn extra-hepatic sites [57]. It has also been shown that SUV

uscles, rest of the hepatic parenchyma, spleen, and urinary tracer excretion.here was also prominent physiologic uptake in the lower abdomen in theigmoid colon. (b) Fused transaxial PET and CT images allowed preciseocalisation of FDG uptake to a small lymph node in the left supraclavicularossa providing the site for fine needle aspiration biopsy under ultrasounduidance.

Page 5: Positron emission tomography and colorectal cancer

3 Oncolo

ol[pSottwlba

3t

irmslsfiitpssdpcltdtmwpda

3s

posuetwoerrt

tsaass

3

nmtapsepo[he

3r

imsomuapoEhiricissboaPsnao

4 M. Lin et al. / Critical Reviews in

f hepatic metastases may be prognostic with significantlyonger survival for patients with lesions exhibiting a low SUV58,59]. This may have a potential for selection of high riskatients for more aggressive therapy based on PET. Finally,cheer and colleagues observed a significant increase in SUVf CRC hepatic metastases after resection of the primaryumour compared with patients without surgical or therapeu-ic intervention [60]. The change in SUV did not correlateith the size of lesions and a possible mechanism related to

oss of anti-angiogenic factors such as angiostatin producedy the primary tumour and a potential role for adjuvant anti-ngiogenesis therapy to prevent progression was suggested.

.2.2. Assessment of local recurrence for salvagereatment

Early studies have shown PET to be useful in identify-ng locally recurrent CRC [61] and to differentiate betweenecurrence and post operative inflammation/scar [62,63]. Aeta-analysis (n = 366) showed PET had an overall sen-

itivity of 94.5% and specificity of 97.7% for identifyingocal/pelvic recurrence [41]. PET is more sensitive than CTcans in detecting local recurrence, increases diagnostic con-dence in lesions deemed equivocal on anatomic imaging and

dentifies additional metastases [64–66]. However, inflamma-ory abdominal or pelvic lesions are known causes of falseositivity. Combined PET-CT results in an improvement inensitivity and specificity when compared with PET-alonetudies in the assessment of pelvic recurrences [67,68] andiagnostic accuracy may be further improved with PET-CTerformed with contrast enhancement [69]. In a recent studyomprising 93 patients with symptomatic residual structuralesions at various locations considered equivocal on conven-ional imaging, PET (97% of the patients underwent PET-CT)etected additional lesions in nearly 50% of subjects and ledo a high impact management change (i.e. a change in treat-

ent modality or intent) in 65% of patients [44]. Patientsith additional lesions on PET also had a significantly poorerrogression-free survival (P = 0.04). A previous surveyocumented similar results where there was a change in man-gement following PET in more than 60% of patients [42].

.2.3. Early detection of biochemical recurrence (risingerum CEA levels)

Serum carcinoembryonic antigen (CEA) elevation mayrecede detection of disease relapse by conventional meth-ds by a few months. Previously, serum CEA-directedecond-look laparotomies had low resectability rates due tonexpected metastatic disease especially when CEA levelsxceeded 11 ng/ml [70]. PET has been shown to be ableo identify the site of suspected recurrent CRC in patientsith an unexplained rise in CEA levels [65,71]. With normalr equivocal radiologic findings and rising serum CEA lev-

ls, the sensitivity and positive predictive value of PET areeportedly 87–100% and 89–96% respectively in detectingecurrent malignancy [72–74] (Fig. 2). A significant correla-ion was found between serum CEA levels and PET derived

(icc

gy/Hematology 77 (2011) 30–47

umour volumes reflecting tumour burden [75]. Some authorsuggest a cutoff value for unexplained serum CEA > 10 ng/mls an indication for FDG PET scanning [74]. PET may alsollow earlier disease detection preceding subsequent rise inerum CEA levels in patients with clinical or radiologicaluspicion for tumour recurrence [76].

.2.4. To identify peritoneal carcinomatosisPeritoneal recurrence in mCRC is a known cause for false

egative findings on FDG PET scans [43,64,65,72,74]. PETay also lead to an underestimation of the extent of peri-

oneal involvement [77] and lower sensitivities in mucinousdenocarcinomas due to hypocellularity [64]. When com-ared with CT scan, some studies have documented higherensitivity with PET in CRC [65,77] while others have shownqually poor performance [78]. Although certain diagnosticatterns of peritoneal carcinomatosis have been describedn PET [79] and superior accuracy with combined PET-CT80], the number of CRC patients studied so far using PETas been small. PET currently cannot replace laparoscopy forxclusion of peritoneal carcinomatosis.

.3. Prognostication and evaluation of treatmentesponse

Monitoring therapy response is essential to guide optimalndividualised cancer treatment. Early identification of treat-

ent failure allows a switch to an alternative managementtrategy with the hope of achieving better outcomes. On thether hand, low risk patients who demonstrate early responseay qualify for less aggressive regimen avoiding potential

nnecessary treatment related toxicities and complicationsnd may be cost saving. Currently, the assessment of thera-eutic efficacy in solid tumours is based on changes in sizef lesions measured on anatomical imaging (e.g. Responsevaluation Criteria in Solid Tumours [RECIST]) [81] whichave well recognized limitations. A tumour that has reducedn size may contain viable malignancy, and conversely aesidual mass may represent post-therapy changes such asnflammation, fibrosis or necrosis with no viable malignantells. Since metabolic changes precede volumetric changes,maging tumour bio-markers using PET as a surrogate mea-ure for therapy response should be more sensitive and morepecific. A favourable therapeutic response is often governedy the degree of reduction in SUV. However, the thresholdf reduction that gives the best predictive value is variablend mainly relies on post-hoc analyses. Regardless of theET techniques used in response evaluation (e.g. qualitative,emi-quantitative or quantitative), standardisation of scan-ing parameters, time points at which responses are measurednd interpretative criteria are paramount to minimise inter-bserver variability. The recent publication of PERCIST

PET Response Criteria in Solid Tumors) 1.0 criteria is anmportant starting point and represents a draft framework thatan be used for clinical studies [82]. In locally advanced rectalancer, there is mounting evidence of the value of FDG PET
Page 6: Positron emission tomography and colorectal cancer

M. Lin et al. / Critical Reviews in Oncology/Hematology 77 (2011) 30–47 35

Fig. 2. (a) A 55-year-old man with previous subtotal colectomy for an obstructed T4N2M0 moderately differentiated adenocarcinoma of the transverse colon.Post-operative carcinoembryonic antigen (CEA) level was 4.9 ng/ml. During adjuvant FOLFOX (fluorouracil, leucovorin and oxaliplatin) chemotherapy, theserum CEA level rose to 15 ng/ml. A diagnostic CT scan showed two ill-defined hypodense lesions in the spleen which were deemed equivocal for metastases.F (arrowT l FDG ur in the l

afiap

3

acpa

dpo1oats

DG PET scan demonstrated two foci of intense FDG uptake in the spleenransaxial CT, PET and fused PET-CT images demonstrating intense focaevealed a subtle hypodensity. There was physiologic urinary tracer activity

s a prognostic marker and in predicting pathologic responseollowing neoadjuvant treatment. PET is also useful in mon-toring therapy response following local ablative treatmentsnd PET may be able to tailor neoadjuvant chemotherapyrior to metastasectomy and in the palliative setting.

.3.1. Neoadjuvant therapy for rectal cancerPathologic complete response following neoadjuvant ther-

py is a good prognostic factor in locally advanced rectalancers. Where less aggressive surgery (such as sphincter-reserving operations) may be contemplated in responders,n escalation of treatment (such as increasing the radiation

laap

s) highly suspicious for metastases, which were confirmed at surgery. (b)ptake at the site of the splenic lesion which on concurrent CT scan, only

eft kidney.

ose) may potentially be appropriate in non-responders. Theotential of FDG PET in assessing the therapeutic effectf chemo-radiation for rectal cancer was raised in the early990s [63]. A number of studies have now shown the abilityf PET to predict pathologic response in the primary tumour,lthough most of them are post-hoc analyses and variable inheir methodologies in terms of time points at which PETcans are performed (mostly between 2 and 6 weeks fol-

owing neoadjuvant treatment), metabolic response criteriand reference standards [83–88]. SUV reduction as earlys 12 days after chemo-radiation has also been shown toredict tumour regression grade (P < 0.0001) [89]. On the
Page 7: Positron emission tomography and colorectal cancer

3 Oncolo

olifPawa[sratrtsi

3

oa(iatIlisebddh

imndlocmdbtairpcsmdt

FwwCt

e(t4c7iariaeaaepd(cifraTedbhTiwa

3

ioqisuiirf(

6 M. Lin et al. / Critical Reviews in

ther hand, the sensitivity for assessing response in regionalymph nodes is disappointing [85] and PET is not accuraten predicting tumour clearance from the mesorectal fasciaollowing neoadjuvant therapy [87]. Metabolic response onET can also predict survival [90,91]. In the study by Capircind colleagues, 5-year overall survival was 91% in patientsith a negative PET performed 7 weeks after chemoradi-

tion and 72% in those with a positive PET (P = 0.024)90]. Post-radiation effects did not seem to compromise PETcan interpretation for therapy response amongst experiencedeaders [92]. Despite the heterogeneity of the studies, PETppears promising and most likely has a role to play inhe assessment of response to neoadjuvant therapy. Whetherisk adapted strategies (either escalation or de-escalation ofreatment) based on metabolic response identified on PETcanning improves clinical outcomes can only be borne outn future randomised controlled trials.

.3.2. ChemotherapyThere has been significant progress in the management

f mCRC with the incorporation of novel targeted therapiesgainst angiogenesis and epidermal growth factor receptorEGFR). The vascular endothelial growth factor (VEGF)nhibitor bevacizumab and EGFR antagonist cetuximab inddition to chemotherapy are gradually being adopted inreatment regimens following promising results from phaseII trials [93]. Some of these agents are cytostatic and may notead to a substantial reduction in tumour size and responses often underestimated by anatomical methods [94]. Patientelection is important in order to avoid futile treatment. Forxample, K-ras mutation status determines who would notenefit from cetuximab. A recent study using dynamic PETata and FDG kinetics also demonstrated the potential to pre-ict angiogenesis related gene expression that may furtherelp individualize treatment [95].

Several small studies investigated the use of FDG PETn the setting of neoadjuvant therapy and colorectal liver

etastases prior to hepatic resection (Table 1). Followingeoadjuvant treatment, most studies demonstrate a significantecrease in the sensitivity and accuracy of PET in detectingiver metastases [96–99]. In one study [97], the sensitivityf PET-CT following neoadjuvant chemotherapy was 49%ompared to a sensitivity of 93.3% in patients without treat-ent (P < 0.0001). The reasons for these findings include a

ecrease in the size of metastases following chemotherapyeyond the spatial resolution of PET [100], downregula-ion of the glycolytic enzyme hexokinase of tumour cellsnd compounded by the high physiologic FDG activityn normal hepatic parenchyma [96]. Complete metabolicesponse is therefore an unreliable predictor for completeathologic response [98] and the decision whether to pro-eed to liver resection or to determine the extent of surgery

hould not solely rely on PET findings. However, PETay have a prognostic role. In a recent study (n = 40),

isease-free survival was predicted by metabolic responseo FOLFOX (fluorouracil, leucovorin and oxaliplatin) or

wpa

gy/Hematology 77 (2011) 30–47

OLFIRI (fluorouracil, leucovorin and irinotecan) with orithout bevacizumab prior to liver resection [101]. Patientsho did not demonstrate a response to chemotherapy on PET-T had a 3.8 times faster rate to disease recurrence compared

o patients with a response.There is limited data on the use of FDG PET in the

valuation of response to palliative chemotherapy in mCRCTable 1). In an early study by Findlay et al. [102], a reduc-ion in tumour to liver uptake (T/L) of ≥15% on PET at–5 weeks after the initiation of 5-fluorouracil (5-FU) basedhemotherapy had a sensitivity and specificity of 100% and5% respectively in predicting tumour response. Paradox-cally, they also observed a marked increase in T/L ratiost 1–2 weeks following treatment in lesions that showed aesponse later on and labelled it a “flare phenomenon”—annflammatory reaction due to infiltration of macrophagess a response to tumour cell kill. However, this was notvident when PET was performed very early at 72 h aftersingle infusion of chemotherapy [103]. The prognostic

spects of PET following chemotherapy have also beenxamined. Using quantitative techniques and FDG kineticarameters, Dimitrakopoulou-Strauss et al. were able to pre-ict survival and stratify patients into short (<1 yr) and long>1 yr) survival classes following second-line chemotherapyomprising 5-FU, folinic acid and oxaliplatin [104]. For clin-cal applicability, more simplified SUV measurements wereound to be reliable [106]. Although there was a strong cor-elation between metabolic and radiological responses, instudy using the European Organisation for Research andreatment of Cancer (EORTC) criteria [107] for assessingarly metabolic response, PET was not found to be pre-ictive of long-term outcomes [108]. However, this coulde a reflection of the response criteria used which may notave given the best predictive value specifically in mCRC.he incorporation of PET to assess chemotherapy response

n future clinical trials allow an early selection of patientsho are refractory to chemotherapy regimens and a switch to

lternative treatments with the aim of improving outcomes.

.3.3. Local ablative therapyIn patients where surgical resection of liver metastases

s not feasible such as unfavourable tumour location, extentr patient co-morbidities, local ablative therapy (radiofre-uency ablation [RFA], cryosurgery ablation [CSA], LASERnduced thermotherapy [LITT], transarterial chemoemboli-ation [TACE] or selective internal radiation therapy [SIRT]sing yttrium-90 [90Y] labelled resin or glass microspheres)s an alternative treatment option for local control. Anatom-cal modalities are often unable to differentiate betweenesidual tumour and peri-lesional post therapy changes andunctional imaging with FDG PET may be advantageousTable 2).

An early study [109] comprising 23 patients who under-ent RFA or CSA of colorectal liver metastases, PETerformed within three weeks post-procedure had a neg-tive predictive value of 100% (51 of 56 lesions became

Page 8: Positron emission tomography and colorectal cancer

M.L

inetal./C

riticalReview

sin

Oncology/H

ematology

77(2011)

30–4737

Table 1FDG PET and monitoring chemotherapy response.

Author (Ref) N METS PET Regimen Timing of PET Parameters Reference standard Criteria Main results

Goshen [94] 7 Liver PET-CT FOLFOX andbevacicumab

Baseline and after 4cycles

Visual Histopathology Complete or partialresponse, stable orprogressive disease

PET findings correlated betterthan CT with pathology andmore accurately at predictingtumour necrosis

Akhurst [96] 42 Liver PET Neoadjuvantfluorouracil-basedchemo

Preop Visual, SUV Histopathology Lesion detection 15/41(37%) and 16/69 (23%)lesions in patients with orwithout chemotherapy notdetected by PET.Phosphorylating activity ofhexokinase significantlylower after chemotherapy

Lubezky [97] 75 Liver PET-CT NeoadjuvantFOLFOX/FOLFIRI± Bevacicumab

Baseline and Preop Visual Histopathology reports Lesion detection Sensitivity = 49% withneoadjuvant therapySensitivity= 93.3% withoutneoadjuvant treatment(P < 0.0001)

Tan [98] 14 Liver (withCMR)

PET-CT Variouschemotherapy± bevacicumab/cetuximab

Baseline and Preop Visual Histopathology Complete pathologicresponse

Only 5/34 lesions (15%) andin 3/14 patients (21%) hadcomplete pathologic responsedespite PET showing CMR

Adie [99] 74 Liver PET-CT Various neoadjuvantchemotherapyregimens

Preop Visual Intraoperativeexamination andultrasoundHistopathology

Accurate PET-CT scans Accurate scans in 29%patients (n = 6) with chemo vs71% (n = 28) without chemo(p = 0.06). 1.7 vs 0.7 lesionsmissed (chemo vs no chemo)

Takahashi [100] 7 Liver PET andPET-CT

Neoadjuvantfluorouracil-basedchemo

Baseline and Preop Visual Histopathology Viable lesion detection Sensitivity = 100% and 17%in evaluating viability ofhepatic mets >2 cm and<2 cm respectively followingneoadjuvant therapy

Small [101] 40 Liver PET-CT NeoadjuvantFOLFOX/FOLFIRI± Bevacicumab

Before and afterchemo

Visual OS, DFS Complete or partialresponse, stable orprogressive disease

Response on PET-CT notpredictive of OS butpredictive of DFS

Findlay [102] 14 Liver (>3 cm) Non dedicatedPET

5-fluorouracil± interferon-�

Baseline, 1–2 weeks,4–5 weeks

Tumour to liver (T/L)ratio, SUV

Tumour dimensions onCT/MRI at 12 weeks.WHO criteria

Tumour dimensions T/L reduction of 15% at 4–5weeks: Sensitivity = 100%Specificity = 75%. SUV lessreliable.

Bender [103] 6 Liver and lung PET Single infusion 5-fluorouracil + folinicacid

Baseline and 72h SUV Tumour dimensions onCT or MRI at-6 weekintervals WHO criteria

Change in SUV Responding mets—22%reduction in SUV

Dimitrakopoulou-Strauss[104]

20 Liver (93%) Dynamic PET 2nd line5-fluorouracil, folinicacid, oxaliplatin

Baseline, after 1cycle, after 4 cycles

SUV, kinetics, fractaldimensions

Survival Short survival class(<1yr)

CCR:SUV after 1cycle = 62%, SUV after 4cycles = 69%, kinetic dataafter 1 and 4 cycles = 78%

Long survival class(>1yr)

Baseline SUV and fractaldimensions also prognostic

Page 9: Positron emission tomography and colorectal cancer

38 M. Lin et al. / Critical Reviews in Oncolo

Tabl

e1

(Con

tinu

ed)

Aut

hor

(Ref

)N

ME

TS

PET

Reg

imen

Tim

ing

ofPE

TPa

ram

eter

sR

efer

ence

stan

dard

Cri

teri

aM

ain

resu

lts

Dim

itrak

opou

lou-

Stra

uss

[105

]28

Liv

er(9

4%)

Dyn

amic

PET

2nd

line

5-flu

orou

raci

l,fo

linic

acid

,oxa

lipla

tin

Bas

elin

e,af

ter

1cy

cle,

3m

onth

sSU

V,f

ract

aldi

men

sion

sC

linic

alre

spon

se,W

HO

crite

ria

Prog

ress

ive

(PD

)or

stab

ledi

seas

e(S

D),

part

ialr

espo

nse

CC

Rfo

rba

selin

eSU

Van

dfr

acta

ldim

ensi

ons:

93%

for

PD,6

0%fo

rSD

.B

asel

ine

SUV

<4.

6pr

edic

tsno

n-re

spon

ders

deG

eus-

Oei

[106

]50

Liv

eran

dlu

ng(9

1%)

Dyn

amic

PET

Var

ious

chem

ore

gim

ens

Bas

elin

e,2

mon

ths,

6m

onth

sM

RG

lu,S

UV

OS,

PFS

Frac

tiona

lcha

nges

ingl

ucos

em

etab

olis

mSi

gnifi

cant

corr

elat

ion

ofM

RG

luan

dSU

V.

Are

duct

ion

inM

RG

luof

>65

%an

dSU

Vof

>20

%at

2m

onth

spr

edic

tive

ofO

S

Bys

trom

[108

]51

Liv

er,l

ung,

lym

phno

des

PET

Irin

otec

anba

sed

regi

men

Bas

elin

e,af

ter

2cy

cles

Vis

ual,

SUV

Rad

iolo

gica

lres

pons

e(R

EC

IST

crite

ria)

ever

y4c

ycle

s,T

TP,

OS

EO

RT

Ccr

iteri

afo

rm

etab

olic

resp

onse

PPV

=71

%,N

PV=

81%

com

pare

dw

ithra

diol

ogic

alre

spon

se.N

osi

gnifi

cant

corr

elat

ion

betw

een

met

abol

icre

spon

sean

dT

TP

orO

S

Met

s:si

tes

ofm

etas

tatic

dise

ase,

CM

R:c

ompl

ete

met

abol

icre

spon

se,C

CR

:cor

rect

clas

sific

atio

nra

tes,

MR

Glu

:met

abol

icra

teof

gluc

ose,

OS:

over

alls

urvi

val,

DFS

:dis

ease

free

surv

ival

,PPV

:pos

itive

pred

ictiv

eva

lue,

NPV

:ne

gativ

epr

edic

tive

valu

e,ch

emo:

chem

othe

rapy

,TT

P:tim

eto

tum

our

prog

ress

ion,

FOL

FOX

:5-

fluor

oura

cil,

leuc

ovor

in,o

xalip

latin

,FO

LFI

RI:

5-flu

orou

raci

l,le

ucov

orin

,iri

note

can,

and

Preo

p:pr

ior

tohe

patic

rese

ctio

n.

pmbr(spa(IiwAupph[e6

twlACimDFaisorMdets

3

(remaptlrMpt

gy/Hematology 77 (2011) 30–47

hotopaenic on FDG PET) during a follow up period of 16onths. Similar high specificities have also been achieved

y other investigators [110–112]. PET also detected recur-ence and residual tumour earlier than conventional follow-upmainly CT scan and serum CEA levels) in several smalltudies [109–115]. Hence PET following initial RFA mayrovide an early signal to repeat ablation and guide ther-py by pinpointing the focus of enhanced metabolic activityi.e. viable malignancy) in a previously treated area [116].n a recent study (n = 16), both PET-CT and MRI had sim-lar accuracies for detecting local tumour progression andere significantly better than PET alone (P < 0.008) [117].lthough an early assessment (within 2 days following RFA)sing PET seems feasible in a small pilot study [118] and hasotential advantages in identifying early treatment failure,ost-RFA inflammation/tissue regeneration at this time pointas been observed which can also confound interpretation114]. To get round this, some have suggested either a veryarly PET scan within 12–24 h post RFA or alternatively after–8 weeks as the best time points [120].

Liver metastases derive most of their blood supply fromhe hepatic artery. Selective internal radiation therapy (SIRT)ith intra-hepatic arterial administration of radioactive 90Y

abelled resin (SIRTEX Medical, North Ryde, Sydney,ustralia) or glass (TheraSphere, MDS Nordion, Ottawa,anada) microspheres aims to deliver high doses of ion-

sing beta-radiation to hepatic metastases selectively whileinimising radiation dose to normal hepatic parenchyma.espite a relatively small number of patients studied so far,DG PET has consistently been shown to be a more accuratend sensitive indicator of treatment response than anatomicmaging where there may not be a significant change in lesionize due to tumour necrosis, cystic degeneration or haem-rrhage following SIRT [121–125]. Using SUV, metabolicesponse can be measured semi-quantitatively [124–127].

ore recently, Flamen and colleagues were able to pre-ict metabolic response during pre-therapeutic work up bystimating the dose delivery to the tumours using a combina-ion of CT, Tc99m-labeled macroaggregated albumin (MAA)cintigraphy and FDG PET data [128].

.4. Radiotherapy planning

Incorporating PET data in radiation therapy planningRTP) seems intuitive. The advantages include more accu-ate and less variable active tumour volume determination,ncompassing unsuspected regional metastases in the treat-ent field, avoiding irradiation of non-malignant anatomical

bnormalities, and delineating tumour burden allowing doseainting (varying radiation doses to different regions of theumour). While benefits have been shown in non-small cellung and oesophageal cancers [129], there is a paucity of data

egarding the use of PET in RTP in rectal cancer (Table 3).

ost PET-CT scans performed for the purposes of RTP areerformed in the radiotherapy treatment position with posi-ion fixation devices such as a belly board. A few small studies

Page 10: Positron emission tomography and colorectal cancer

M.L

inetal./C

riticalReview

sin

Oncology/H

ematology

77(2011)

30–4739

Table 2FDG PET and local ablative therapy.

Author (Ref) N (CRC) PET Therapy Timing of PET PET criteria Follow-up Reference standard Main results

Lagenhoff [109] 23 (23) PET CSA (n = 17)RFA (n = 6)

≤3 weekspost-treatment

Visual 16 months PET, CT, CEA NPV = 100%, PPV = 80%. PET detectedrecurrent malignancy earlier than CT

Donckier [110] 17 (11) PET RFA Baseline, 1 week, 1,3 monthspost-treatment

Visual 11 months PET, CT, Histopathology NPV = 100% (13/13), PPV = 100% (4/4)for local recurrence. PET superior to CT inearly detection of residual tumour

Anderson [111] 13 (8) PET RFA Pre and post RFA Visual Not specified Clinical, PET, CT,MRI PPV = 100% (11/11 lesions). PET superiorto MRI/CT in detection ofrecurrence/residual tumour

Joosten [112] 43 (43) PET CSA (n = 17)RFA (n = 6)

≤3 weeks posttreatment

Visual CSA-26 months, RFA-25months

PET, CT, CEA NPV = 100%, PPV = 88%. PET detectedrecurrent malignancy earlier than CT

Blokhuis [113] 15 (11) PET RFA Post RFA Visual 16.8 months Not specified PET identified local recurrence earlier thanCT (6.8 vs 9.8 months) following RFA

Veit [114] 11 (11) PET-CT RFA Baseline, 2 days Visual, SUV 11 months Histopathology, allimaging, clinical, CEA

Sensitivity and accuracies for residualtumour = 65% and 68% for PET/PET-CT,44% and 47% for CT alone

Travaini [115] 9 (7) PET-CT RFA 1 week, 1, 3, 6, 9months

Visual 9 months PET-CT, multi-detectorCT

PET superior to CT in early detection oflocal treatment failure in all cases

Kuehl [117] 16 (16) PET-CT RFA Baseline, 24 h, 1, 3, 6months then every 6months

Visual, SUV 22 months Histopathology, CT,clinical

PET-CT significantly better than PETalone for detecting LTP

No significant differences betweenPET-CT and MRI

Khandani [118] 8 (5) PET-CT RFA 2–41 h Visual 3–16 months Follow-up PET Infrequent inflammatory uptake at RFAsite of liver metastases on early PET

Denecke [119] 21 PET LITT Suspected residual orrecurrent disease

Visual, SUV >12 months Histopathology, CT,MRI, clinical

SUVmax of ablated lesions of 4.2discriminates partial and completeablation-Accuracy = 94%Overall NPV = 96%

Wong [121] 8 (8) PET SIRT(glass) Baseline, 3 month Visual 3 months PET, CT or MRI, CEA PET more sensitive in detecting responsethan CT/MRICorrelated better with CEA

Properl [122] 23 (12) PET or PET-CT SIRT(resin) Baseline, 3 month Visual 3 months PET, CT, tumour markers PET more sensitive in detecting responsethan CT

Bienert [124] 5 (4) PET-CT SIRT(resin) Baseline, 1 and 3months

Visual, SUV 22 months Response = SUVreduction of 20%

Metabolic changes more accurate thananatomical changes

Szyszko [125] 21 (10) PET SIRT(resin) Baseline, 6–8 weeks Visual, SUV 6–8 weeks PET, CT(RECIST),tumour markers

PET more sensitive in detecting responsethan CT. Significant reduction in SUVfollowing SIRT

Wong [126] 27 (27) PET SIRT(glass) Baseline, 3 months Visual, SUV 3 months PET (Visual vssemi-quantitative)

SUV of total liver uptake correlates withvisual analyses for response (r = 0.75)

Wong [127] 19 (10) PET SIRT(resin) Baseline, 3 months Visual, SUV 3 months PET (Visual vssemi-quantitative)

SUV of total liver uptake correlates withvisual analyses for response (r = 0.72)

Flamen [128] 8 (8) PET-CT SIRT(resin) Baseline, 6 weeks TLG 6 weeks PET response Simulated median absorbed dose = 20 Gyand 46 Gy in poor (<50%TLG change) andgood (>50%TLG change) responders

N (CRC): total number of patients (patients with colorectal cancer), CSA: cryosurgery ablation, RFA: radiofrequency ablation, CEA: carcino-embryonic antigen, LTP: local tumour progression, PPV: positivepredictive value, NPV: negative predictive value, LITT: LASER induced thermotherapy, SIRT: selective internal radiation therapy, and TLG: total lesion glycolysis.

Page 11: Positron emission tomography and colorectal cancer

40 M. Lin et al. / Critical Reviews in Oncology/Hematology 77 (2011) 30–47

Table 3PET and radiotherapy planning (RTP) in rectal cancer.

Author (Ref) N Method Results and conclusions

Ciernik [130] 6 Prone or supine PET-CT GTV increased in 3 patientsMean increase of GTV of 50%

Ciernik [131] 11 Prone PET-CT with belly boardAutomated region growing with fixedthreshold

Good correlation between BTV and CT based GTVGood correlation between PET and CT derived PTV

Patel [132] 6 Prone PET-CT with belly board Greater inter-observer agreement of GTVQualitative. FDG and FLT. No difference between FDG and FLT derived tumour volumes

Anderson [133] 23 20 rectal cancer, 3 anal cancer PET detected distant metastases in 25% rectal cancer patients.Prone PET-CT with belly boardQualitative

Radiation treatment plan changed in 26%Change in PTV in 17% patientsMean PET-GTV < Mean CT-GTV (92 cm3 vs 100 cm3)

Bassi [134] 25 Prone or supine PET-CT with knee fixdevice

Change in stage or treatment intent in 16% (4/25)

Fixed threshold methodPET-CT GTV and PET-CT CTV significantly greater than CT-GTV(25% increase) and CT-CTV

Paskeviciute [135] 36 Prone PET-CT with belly boardVisual, SUV > 2.5

Mean PET-CT-GTV vs Mean CT-GTV (62 cm3 vs 163 cm3)In 46% (n = 16) patients, PET-CT resulted in modification ofCT-PTV due to geographic miss. 8% (n = 3) patients changed fromdefinitive to palliative management

Roels [136] 15 Prone PET-CT with belly board FDGand FLT or FMISO

FDG and FLT TV show good spatial correspondence

Pre and post chemoradiationLess reliable for FMISO

Roels [137] 15 Prone PET-CT with belly boardBefore, during, and after preoperativechemoradiation

Substantial change of GTVMean mismatch of 50% of PET and MRI tumour volume

G ic targefl

hbhtTPa

3

iaruifrfipsrp9watop

3

anCsJuiatgi(nacgwogf

3

PET and MRI

TV: gross tumour volume, PTV: planning target volume, BTV: biologuorodeoxythymidine, and FMISO:18F-fluoromisonidazole.

ave shown the detection of unsuspected metastatic diseasey PET and thereby altering management [133,134]. Othersave documented changes in tumour volume delineation withhe incorporation of metabolic information in RTP [134,135].he use of PET in RTP is experimental and whether usingET-CT data for RTP improves clinical outcomes is unknownnd larger studies are needed.

.5. Post-treatment surveillance

Surveillance in CRC in order to detect early relapses a contentious topic especially with regards to the mostppropriate imaging modality [138]. Earlier detection ofecurrences in asymptomatic patients by systematic follow-p allows potentially more curative resections [139] andmproved outcomes. This supports the idea of an intensiveollow-up programme, perhaps in selected patients at highisk of relapse. While there is emerging evidence of bene-ts using FDG PET in routine surveillance of asymptomaticatients with other cancers [140,141], there is very limitedupportive data in CRC [142,143]. In one study (n = 130),ecurrences were detected after a shorter time interval inatients who were followed-up with the addition of PET atand 15 months after initial surgery compared with patientsho underwent conventional follow-up [143]. The authors

lso reported a higher number of curative surgical interven-ions in the PET group. Currently, due to sparse data, the usef FDG PET in routine surveillance of CRC in asymptomaticatients is not recommended but may have a future role.

Wt

t volume, CTV: clinical target volume, TV: tumour volume, FLT: 18F-

.6. Screening

There is strong evidence that screening reduces mortalitynd incidence of CRC [144]. The ideal screening test is cheap,on-invasive and detects cancer early in high-risk individuals.RC and adenomatous polyps have been detected in various

creening programs incorporating FDG PET [145–148]. In aapanese nationwide survey involving 50,558 subjects whonderwent screening with PET or PET-CT, CRC was detectedn 102 (0.2%) and predominantly in men above 50 years ofge [149]. However, it is debatable whether PET is cost effec-ive when applied to unselected general population screeningiven the significant number of false negative and false pos-tive PET findings [148,150–152]. In one screening seriesn = 2911), PET detected 7 cases of CRC and was falselyegative in 28 asymptomatic individuals [148]. There werelso 104 false positive cases out of 111 in the colorectal regionomprising predominantly a misinterpretation of physiologicastrointestinal uptake, inflammatory lesions and 11 casesith benign adenomas. At this stage, screening with PET isf unproven efficacy. Whether it has a future role in more tar-eted populations, for example in first degree relatives with aamily history of CRC, using PET-CT, remains speculative.

.7. Incidental colonic FDG uptake

Physiologic colonic FDG uptake is common on PET scans.hile diffuse uptake can be seen as a normal variant or due

o a benign aetiology such as inflammation, focal colonic

Page 12: Positron emission tomography and colorectal cancer

Oncolo

uoboust[

dubH

Fdarsaa

M. Lin et al. / Critical Reviews in

ptake may indicate a more sinister pathology. The majorityf cases of incidental focal colonic FDG uptake are causedy a synchronous adenoma or carcinoma which is uncoveredn further evaluation when the bowel uptake was deemed an

nusual site for metastatic disease [154–156] (Fig. 3). Theensitivity of FDG PET to detect pre-malignant adenoma-ous polyps correlated with size [146] and grade of dysplasia157]. Non-pre-malignant lesions such as hyperplastic polyps

[fpi

ig. 3. (a) A 58-year-old man with newly diagnosed non-small cell lung cancer unemonstrated intense FDG uptake in the right pulmonary hilum encasing the bronlso mild ill-defined uptake surrounding this consistent with associated atelectasisectosigmoid junction (arrowhead). There was physiological urinary FDG accumulaagittal PET and CT images demonstrated intense FDG uptake corresponded to circsecond malignancy. Colonoscopy and biopsies revealed a synchronous adenocar

nterior and inferior to the colorectal tumour.

gy/Hematology 77 (2011) 30–47 41

o not tend to accumulate FDG [29]. The intensity of FDGptake in terms of maximal SUV was unable to discriminateetween malignant, pre-malignant and benign lesions [155].owever, the specificity is improved with combined PET-CT

7] (Table 4) and incidental focal colonic uptake deservesurther confirmatory investigations to exclude a malignantathology in appropriate patients whose management for thendex cancer is not palliative.

derwent a PET-CT scan for staging. Maximum intensity projection imagechus intermedius (arrow) at the site of the primary malignancy. There wasand inflammation. There was unsuspected focal intense FDG uptake at thetion in the bladder below this and tracer excretion in both kidneys. (b) Fusedumferential mucosal thickening at the rectosigmoid junction suspicious forcinoma. There was intense physiologic FDG excretion in the bladder just

Page 13: Positron emission tomography and colorectal cancer

42 M. Lin et al. / Critical Reviews in Oncology/Hematology 77 (2011) 30–47

Table 4PET-CT and incidental focal bowel uptake.

Author (Ref) N Population Incidental boweluptake (%)

Diagnosis confirmed (%) Pre-malignantpolyp (%)

Carcinoma (%) Other malignancy PPV

Kamel [153] 3281 Cancer (2836)non-oncologic(445)

98 patients (3%) 69/98 patients (70%) 27 (39%) 9 (13%) 52%

Israel [155] 4390 Cancer 58 patients (1.3%) 28/58 patients (48%) 9 (32%) 4 (14%) 2 (Mets) 54%Even-Sapir [156] 2360 Cancer 33 patients (1.4%) 29/39 lesions (74%) 7 (24%) 11 (38%) 2 (Mets, NHL) 69%Gutman [158] 1716 Cancer 45 patients (2.6%) 20/45 patients (44%) 10 (50%) 3 (15%) 65%Hemandas [159] 110 Cancer 10 patients (9%) 7/10 patients (70%) 7(100%) 100%Lee* [160] 2916 Cancer 85 patients (2.9%) 44/85 patients (52%) 32 patients with

true positivefindings

73%

Lee [161] 1665 Cancer 62 patients (3.7%) 40/67 lesions (60%) 12 (30%) 10 (25%) 1 (NHL) 58%

rapolateP metasta

4

4

bsasgsndmTetoapc[n

4

pmlsFuaiwtt

waa[up

ot1htbmapPn

5

wCtrpntfcai

* Number of patients with carcinoma and adenoma cannot reliably be extPV: positive predictive value, NHL: Non-Hodgkins lymphoma, and Mets:

. Future

.1. PET-MR

Technology is advancing at a tremendous pace. As CTecomes a constant neighbour of PET, new partnershipsuch as combined PET-MRI are making the transition fromresearch tool into clinical practice [162]. The notion of

uperimposing PET and MRI data in CRC was first sug-ested by Ito and colleagues in 1992 [63]. Where PET-CTcans are acquired sequentially, PET-MRI allows simulta-eous acquisition and temporal correlation of both sets ofata thereby improving spatial correlation by eliminatingotion artefact as a result of patient and organ movement.he strengths of such “one stop” multimodality imagingxtend beyond simply superimposing anatomical and func-ional images but also allowing simultaneous acquisitionf dynamic quantitative PET and functional MRI data inssessing tumour metabolism, perfusion and viability. Theotentially attractive clinical applications would include pre-ise therapy response assessment and treatment planning163]. The future may also see the combination of hybriduclear medicine and optical imaging systems [164].

.2. Non-FDG tracers

While FDG is the stalwart of PET, other molecularathways are being targeted using non-FDG tracers anday be complementary to FDG. 18F-fluoro-3′-deoxy-3′-

-fluorothymidine (FLT) has been investigated as a morepecific radioisotope for cellular proliferation. Imaging withLT potentially leads to fewer false positives since FDGptake can be due to local inflammation post-treatmentnd FLT specifically reflects cellular proliferation. However,

n a recent small study of rectal cancer patients (n = 10)ho underwent long course neoadjuvant chemoradiation,

he degree of change in FLT uptake at 2 weeks after initia-ion and completion of neoadjuvant therapy did not correlate

iuvc

d from the data presented.ses.

ith histopathological tumour regression [165]. Similar dis-ppointing results have been found with the radiolabelledmino acid tracer carbon-11 methyl-l-methionine (MET)166]. These reports suggested that the reduction in tracerptake following therapy may reflect impaired tracer trans-ort rather than correlate with tumour cell viability.

Tumour hypoxia is increasingly recognised as onef the negative factors which impacts on the effec-iveness of chemotherapy and radiotherapy. Although8F-fluoromisonidazole (FMISO) is the most studiedypoxic tracer for PET imaging, it has limitations dueo prominent non-tumoural bowel uptake and may note optimal for rectal cancer. 60Cu-diacetyl-bis (N4-ethylthiosemicarbazone)(60Cu-ATSM) may be a suitable

lternative. In a pilot study of 17 patients with rectal cancers,atients with hypoxic tumours as depicted by 60Cu-ATSMET, were less responsive to therapy and had a worse prog-osis [167].

. Conclusions

The role of PET in CRC continues to evolve. While it isell established in the assessment of recurrent and residualRC and in patients prior to potential curative metastasec-

omy, the benefits of FDG PET in particular for staging inectal cancer, in the assessment of therapy efficacy as well asroviding early prognostic information in patients receivingeoadjuvant treatment are emerging. PET also has the poten-ial to assist in planning radiotherapy. FDG PET performedor indications other than CRC can detect incidental focalolonic uptake which has a significant rate of pre-malignancynd malignancy that warrants further investigations. With thencreasing appreciation and understanding of the underly-

ng molecular processes in oncogenesis, functional imagingsing PET and different radiopharmaceuticals will enable inivo assessment of tumour biology that would allow clini-ians to target, individualise and monitor therapy.
Page 14: Positron emission tomography and colorectal cancer

Oncolo

R

M

R

M. Lin et al. / Critical Reviews in

eviewer

Dr. Jean-Pierre Papazyan, Clinique de Genolier, Nuclearedicine, 1 route du Muids, CH-1272 Genolier, Switzerland.

eferences

[1] Van Cutsem E, Oliveira J. Primary colon cancer: ESMO clinical rec-ommendations for diagnosis, adjuvant treatment and follow-up. AnnOncol 2009;20(Suppl. 4):49–50.

[2] Jemal A, Siegel R, Ward E, Hao Y, Xu J, Thun MJ. Cancer statistics2009. CA Cancer J Clin 2009;59:225–49.

[3] Lin M, Ho Shon I, Lin P. Positron emission tomography–current statusand future challenges. Intern Med J 2010;40:19–29.

[4] Hillner BE, Siegel BA, Liu D, et al. Impact of positron emissiontomography/computed tomography and positron emission tomogra-phy (PET) alone on expected management of patients with cancer:initial results from the National Oncologic PET Registry. J Clin Oncol2008;26:2155–61.

[5] Papathanassiou D, Bruna-Muraille C, Liehn J-C, Nguyen TD, CuréH. Positron Emission Tomography in oncology: Present and future ofPET and PET/CT. Crit Rev Oncol Hematol 2009;72:239–54.

[6] Antoch G, Saoudi N, Kuehl H, et al. Accuracy of whole-body dual-modality fluorine-18-2-fluoro-2-deoxy-D-glucose positron emissiontomography and computed tomography (FDG-PET/CT) for tumorstaging in solid tumors: comparison with CT and PET. J Clin Oncol2004;22:4357–68.

[7] Cohade C, Osman M, Leal J, Wahl R. Direct comparison of 18F-FDGPET and PET/CT in patients with colorectal carcinoma. J Nucl Med2003;44:1797–803.

[8] Czernin J, Allen-Auerbach M, Schelbert H. Improvements in cancerstaging with PET/CT: literature-based evidence as of September 2006.J Nucl Med 2007;48:78S–88S.

[9] Burt BM, Humm JL, Kooby DA, et al. Using positron emissiontomography with [18F]FDG to predict tumor behavior in experimentalcolorectal cancer. Neoplasia 2001;3:189–95.

[10] Veit-Haibach P, Kuehle CA, Beyer T, et al. Diagnostic accuracy ofcolorectal cancer staging with whole-body PET/CT colonography.JAMA 2006;296:2590–600.

[11] Nagata K, Ota Y, Okawa T, Endo S, Kudo SE. PET/CT colonographyfor the preoperative evaluation of the colon proximal to the obstructivecolorectal cancer. Dis Colon Rectum 2008;51(6):882–90.

[12] Heriot A, Kumar D. Rectal cancer recurrence: factors and mecha-nisms. Colorectal Dis 2000;2:126–37.

[13] Quirke P, Durdey P, Dixon MF, Williams NS. Local recurrenceof rectal adenocarcinoma due to inadequate surgical resection.Histopathological study of lateral tumour spread and surgical exci-sion. Lancet 1986;328(8514):996–9.

[14] Adam IJ, Mohamdee MO, Martin IG, et al. Role of circumferentialmargin involvement in the local recurrence of rectal cancer. Lancet1994;344(8924):707–11.

[15] Chan KW, Boey J, Wong SK. A method of reporting radial inva-sion and surgical clearance of rectal carcinoma. Histopathology1985;9(12):1319–27.

[16] MERCURY Study Group. Diagnostic accuracy of preoperative mag-netic resonance imaging in predicting curative resection of rectalcancer: prospective observational study. BMJ 2006;333(7572):779.

[17] Klessen C, Rogalla P, Taupitz M. Local staging of rectal cancer. Thecurrent role of MRI. Eur Radiol 2007;17:379–89.

[18] Bipat S, Glas AS, Slors FJM, Zwinderman AH, Bossuyt PMM, Stoker

J. Rectal cancer: local staging and assessment of lymph node involve-ment with endoluminal US, CT, and MR imaging-a meta-analysis.Radiology 2004;232:773–83.

[19] Brown G, Richards CJ, Bourne MW, et al. Morphologic predictorsof lymph node status in rectal cancer with use of high-spatial-

gy/Hematology 77 (2011) 30–47 43

resolution MR imaging with histopathologic comparison. Radiology2003;227:371–7.

[20] Gearhart SL, Frassica D, Rosen R, Choti M, Schulick R, WahlR. Improved staging with pretreatment positron emission tomogra-phy/computed tomography in low rectal cancer. Ann Surg Oncol2006;13:397–404.

[21] Davey K, Heriot AG, Mackay J, et al. The impact of18-fluorodeoxyglucose positron emission tomography-computedtomography on the staging and management of primary rectal cancer.Dis Colon Rectum 2008;51:997–1003.

[22] Heriot AG, Hicks RJ, Drummond EGP, et al. Does positron emis-sion tomography change management in primary rectal cancer? Aprospective assessment. Dis Colon Rectum 2004;47:451–8.

[23] Llamas-Elvira JM, Rodriguez-Fernandez A, Gutierrez-Sainz J, etal. Fluorine-18 fluorodeoxyglucose PET in the preoperative stag-ing of colorectal cancer. Eur J Nucl Med Mol Imaging 2007;34:859–67.

[24] Akiyoshi T, Oya M, Fujimoto Y, et al. Comparison of preoper-ative whole-body positron emission tomography with MDCT inpatients with primary colorectal cancer. Colorectal Dis 2009;11:464–9.

[25] Eglinton T, Luck A, Bartholomeusz D, Varghese R, Lawrence M.Positron-emission tomography/computed tomography in the initialstaging of primary rectal cancer. Colorectal Dis 2010;12:667–73.

[26] Furukawa H, Ikuma H, Seki A, et al. Positron emission tomographyscanning is not superior to whole body multidetector helical com-puted tomography in the preoperative staging of colorectal cancer.Gut 2006;55:1007–11.

[27] Tateishi U, Maeda T, Morimoto T, Miyake M, Arai Y, Kim EE.Non-enhanced CT versus contrast-enhanced CT in integrated PET/CTstudies for nodal staging of rectal cancer. Eur J Nucl Med Mol Imaging2007;34:1627–34.

[28] Nahas CSR, Akhurst T, Yeung H, et al. Positron emission tomographydetection of distant metastatic or synchronous disease in patients withlocally advanced rectal cancer receiving preoperative chemoradiation.Ann Surg Oncol 2008;15(3):704–11.

[29] Abdel-Nabi H, Doerr RJ, Lamonica DM, et al. Staging of primary col-orectal carcinomas with fluorine-18 fluorodeoxyglucose whole-bodyPET: correlation with histopathologic and CT findings. Radiology1998;206:755–60.

[30] Mukai M, Sadahiro S, Yasuda S, et al. Preoperative evaluation bywhole-body 18F-fluorodeoxyglucose positron emission tomographyin patients with primary colorectal cancer. Oncol Rep 2000;7:85–7.

[31] Kantorova I, Lipska L, Belohlavek O, Visokai V, Trubac M, Schnei-derova M. Routine (18)F-FDG PET preoperative staging of colorectalcancer: comparison with conventional staging and its impact on treat-ment decision making. J Nucl Med 2003;44:1784–8.

[32] Monig SP, Baldus SE, Zirbes TK, et al. Lymph node sizeand metastatic infiltration in colon cancer. Ann Surg Oncol1999;6(6):579–81.

[33] Park IJ, Kim HC, Yu CS, et al. Efficacy of PET/CT in the accu-rate evaluation of primary colorectal carcinoma. Eur J Surg Oncol2006;32(9):941–7.

[34] Galandiuk S, Wieand HS, Moertel CG, et al. Patterns of recurrenceafter curative resection of carcinoma of the colon and rectum. SurgGynecol Obstet 1992;174:27–32.

[35] Salo JC, Paty PB, Guillem J, Minsky BD, Harrison LB, Cohen AM.Surgical salvage of recurrent rectal carcinoma after curative resection:a 10 year experience. Ann Surg Oncol 1999;6:171–7.

[36] Tepper JE, O’Connell M, Hollis D, Niedzwiecki D, Cooke E, MayerRJ. Analysis of surgical salvage after failure of primary therapy in rec-tal cancer: results from Intergroup Study 0114. J Clin Oncol 2003;21:

3623–8.

[37] Fong Y, Fortner J, Sun RL, Brennan MF, Blumgart LH. Clinicalscore for predicting recurrence after hepatic resection for metastaticcolorectal cancer: analysis of 1001 consecutive cases. Ann Surg1999;230:309–18.

Page 15: Positron emission tomography and colorectal cancer

4 Oncolo

4 M. Lin et al. / Critical Reviews in

[38] Wei AC, Greig PD, Grant D, Taylor B, Langer B, Gallinger S. Survivalafter hepatic resection for colorectal metastases: a 10-year experience.Ann Surg Oncol 2006;13(5):668–76.

[39] Steele Jr G, Bleday R, Mayer RJ, Lindblad A, Petrelli N, Weaver D. Aprospective evaluation of hepatic resection for colorectal carcinomametastases to the liver: Gastrointestinal Tumor Study Group Protocol6584. J Clin Oncol 1991;9:1105–12.

[40] Lejeune C, Bismuth MJ, Conroy T, et al. Use of a decision analysismodel to assess the cost-effectiveness of 18F-FDG PET in the man-agement of metachronous liver metastases of colorectal cancer. J NuclMed 2005;46:2020–8.

[41] Huebner RH, Park KC, Shepherd JE, et al. A meta-analysis of theliterature for whole-body FDG PET detection of recurrent colorectalcancer. J Nucl Med 2000;41:1177–89.

[42] Meta J, Seltzer M, Schiepers C, et al. Impact of 18F-FDG PET onmanaging patients with colorectal cancer: the referring physician’sperspective. J Nucl Med 2001;42:586–90.

[43] Kalff V, Hicks RJ, Ware RE, Hogg A, Binns D, McKenzie AF. Theclinical impact of 18F-FDG PET in patients with suspected or con-firmed recurrence of colorectal cancer: a prospective study. J NuclMed 2002;43:492–9.

[44] Scott AM, Gunawardana DH, Kelley B, et al. PET changes manage-ment and improves prognostic stratification in patients with recurrentcolorectal cancer: results of a multicenter prospective study. J NuclMed 2008;49:1451–7.

[45] National Comprehensive Cancer Network-Clinical PracticeGuidelines in Oncology: Colon Cancer, v.1.2009, available atwww.nccn.org.

[46] Australian Cancer Network Colorectal Cancer Guidelines RevisionCommittee. Guidelines for the prevention, early detection and man-agement of colorectal cancer. Sydney: The Cancer Council Australiaand Australian Cancer Network; 2005.

[47] Yonekura Y, Benua RS, Brill AB, et al. Increased accumulation of2-deoxy-2-[18F]Fluoro-d-glucose in liver metastases from colon car-cinoma. J Nucl Med 1982;23:1133–7.

[48] Wiering B, Krabbe PFM, Jager GJ, Oyen WJG, Ruers TJM.The impact of fluor-18-deoxyglucose-positron emission tomogra-phy in the management of colorectal liver metastases. Cancer2005;104:2658–70.

[49] Bipat S, van Leeuwen MS, Comans EFI, et al. Colorectal livermetastases: CT, MR imaging, and PET for diagnosis—meta-analysis.Radiology 2005;237:123–31.

[50] Kong G, Jackson C, Koh DM, et al. The use of (18)F-FDG PET/CT incolorectal liver metastases-comparison with CT and liver MRI. Eur JNucl Med Mol Imaging 2008;35:1323–9.

[51] Schussler-Fiorenza CM, Mahvi DM, Niederhuber J, Rikkers LF,Weber SM. Clinical risk score correlates with yield of PET scanin patients with colorectal hepatic metastases. J Gastrointest Surg2004;8:150–8.

[52] Coenegrachts K, De Geeter F, ter Beek L, et al. Comparison ofMRI (including SS SE-EPI and SPIO-enhanced MRI) and FDG-PET/CT for the detection of colorectal liver metastases. Eur Radiol2009;19:370–9.

[53] Strasberg SM, Dehdashti F, Siegel BA, Drebin JA, Linehan D. Sur-vival of patients evaluated by FDG-PET before hepatic resection formetastatic colorectal carcinoma: a prospective database study. AnnSurg 2001;233:293–9.

[54] Fernandez FG, Drebin JA, Linehan DC, Dehdashti F, Siegel BA,Strasberg SM. Five-year survival after resection of hepatic metas-tases from colorectal cancer in patients screened by positron emissiontomography with F-18 fluorodeoxyglucose (FDG-PET). Ann Surg2004;240:438–47.

[55] Pawlik TM, Assumpcao L, Vossen JA, et al. Trends in nontherapeuticlaparotomy rates in patients undergoing surgical therapy for hepaticcolorectal metastases. Ann Surg Oncol 2009;16:371–8.

[56] Ruers TJM, Wiering B, van der Sijp JRM, et al. Improved selec-tion of patients for hepatic surgery of colorectal liver metastases

gy/Hematology 77 (2011) 30–47

with 18F-FDG PET: a randomized study. J Nucl Med 2009;50:1036–41.

[57] Finkelstein SE, Fernandez FG, Dehdashti F, et al. Unique site- andtime-specific patterns of recurrence following resection of colorec-tal carcinoma hepatic metastases in patients staged by FDG-PET. JHepatobiliary Pancreat Surg 2008;15:483–7.

[58] Riedl CC, Akhurst T, Larson S, et al. 18F-FDG PET scanning cor-relates with tissue markers of poor prognosis and predicts mortalityfor patients after liver resection for colorectal metastases. J Nucl Med2007;48:771–5.

[59] de Geus-Oei LF, Wiering B, Krabbe PF, Ruers TJM, Punt CJA, OyenWJG. FDG-PET for prediction of survival of patients with metastaticcolorectal carcinoma. Ann Oncol 2006;17:1650–5.

[60] Scheer MGW, Stollman TH, Vogel WV, Boerman OC, Oyen WJG,Ruers TJM. Increased metabolic activity of indolent liver metas-tases after resection of a primary colorectal tumor. J Nucl Med2008;49:887–91.

[61] Beets G, Penninckx F, Schiepers C, et al. Clinical value of whole-body positron emission tomography with [18F]fluorodeoxyglucosein recurrent colorectal cancer. Br J Surg 1994;81:1666–70.

[62] Strauss LG, Corius JH, Schlag P, et al. Recurrence of colorectaltumours: PET evaluation. Radiology 1989;170:329–32.

[63] Ito K, Kato T, Tadokoro M, et al. Recurrent rectal cancer and scardifferentiation with PET and MR imaging. Radiology 1992;182:549–52.

[64] Whiteford MH, Whiteford HM, Yee LF, et al. Usefulness of FDG-PETscan in the assessment of suspected metastatic or recurrent adenocar-cinoma of the colon and rectum. Dis Colon Rectum 2000;43:759–67.

[65] Flamen P, Stroobants S, Van Cutsem E, et al. Additional valueof whole-body positron emission tomography with fluorine-18-2-fluoro-2-deoxy-d-glucose in recurrent colorectal cancer. J Clin Oncol1999;17:894–901.

[66] Lonneux M, Reffad AM, Detry R, Kartheuser A, Gigot JF, Pauwels S.FDG-PET improves the staging and selection of patients with recur-rent colorectal cancer. Eur J Nucl Med Mol Imaging 2002;29:915–21.

[67] Even-Sapir E, Parag Y, Lerman H, et al. Detection of recurrencein patients with rectal cancer: PET/CT after abdominoperineal oranterior resection. Radiology 2004;232:815–22.

[68] Votrubova J, Belohlavek O, Jaruskova M, et al. The role of FDG-PET/CT in the detection of recurrent colorectal cancer. Eur J NuclMed Mol Imaging 2006;33:779–84.

[69] Kitajima K, Murakami K, Yamasaki E, et al. Performance of inte-grated FDG PET/contrast-enhanced CT in the diagnosis of recurrentcolorectal cancer: Comparison with integrated FDG PET/non-contrast-enhanced CT and enhanced CT. Eur J Nucl Med Mol Imaging2009;36:1388–96.

[70] Minton JP, Hoehn JL, Gerber DM, et al. Results of a 400-patient car-cinoembryonic antigen second-look colorectal cancer study. Cancer1985;55:1284–90.

[71] Ruhlmann J, Schomburg A, Bender H, et al. Fluorodeoxyglu-cose whole-body positron emission tomography in colorectal cancerpatients studied in routine daily practice. Dis Colon Rectum1997;40:1195–204.

[72] Zervos EE, Badgwell BD, Burak Jr WE, Arnold MW, Martin EW.Fluorodeoxyglucose positron emission tomography as an adjunctto carcinoembryonic antigen in the management of patients withpresumed recurrent colorectal cancer and nondiagnostic radiologicworkup. Surgery 2001;130:636–43.

[73] Flanagan FL, Dehdashti F, Ogunbiyi OA, Kodner IJ, Siegel BA. Utilityof FDG-PET for investigating unexplained plasma CEA elevation inpatients with colorectal cancer. Ann Surg 1998;227:319–23.

[74] Liu FY, Chen JS, Changchien CR, et al. Utility of 2-fluoro-2-deoxy-

D-glucose positron emission tomography in managing patients ofcolorectal cancer with unexplained carcinoembryonic antigen eleva-tion at different levels. Dis Colon Rectum 2005;48:1900–12.

[75] Choi MY, Lee KM, Chung JK, et al. Correlation between serum CEAlevel and metabolic volume as determined by FDG PET in post-

Page 16: Positron emission tomography and colorectal cancer

Oncolo

M. Lin et al. / Critical Reviews in

operative patients with recurrent colorectal cancer. Ann Nucl Med2005;19(2):123–9.

[76] Sarikaya I, Bloomston M, Povoski SP, et al. FDG-PET scan inpatients with clinically and/or radiologically suspicious colorectalcancer recurrence but normal CEA. World J Surg Oncol 2007;5:64.

[77] Tanaka T, Kawai Y, Kanai M, Taki Y, Nakamoto Y, Takabayashi A.Usefulness of FDG-positron emission tomography in diagnosing peri-toneal recurrence of colorectal cancer. Am J Surg 2002;184:433–6.

[78] Dromain C, Leboulleux S, Auperin A, et al. Staging of peri-toneal carcinomatosis: enhanced CT vs. PET/CT. Abdom Imaging2007;33:87–93.

[79] De Gaetano AM, Calcagni ML, Rufini V, Valenza V, Giordano A,Bonomo L. Imaging of peritoneal carcinomatosis with FDG PET-CT: diagnostic patterns, case examples and pitfalls. Abdom Imaging2009;34(3):391–402.

[80] Dirisamer A, Schima W, Heinisch M, et al. Detection of histologicallyproven peritoneal carcinomatosis with fused 18F-FDG-PET/MDCT.Eur J Radiol 2009;69:536–41.

[81] Eisenhauer EA, Therasse P, Bogaerts J, et al. New response evaluationcriteria in solid tumours: Revised RECIST guideline (version 1.1). EurJ Cancer 2009;45:228–47.

[82] Wahl RL, Jacene H, Kasamon Y, Lodge MA. From RECIST to PER-CIST: evolving considerations for PET response criteria in solidtumors. J Nucl Med 2009;50(Suppl. 1):122S–50S.

[83] Guillem JG, Puig-La Calle J, Akhurst T, et al. Prospective assess-ment of primary rectal cancer response to preoperative radiation andchemotherapy using 18-fluordeoxyglucose positron emission tomog-raphy. Dis Colon Rectum 2000;43:18–24.

[84] Amthauer H, Denecke T, Rau B, et al. Response prediction by FDG-PET after neoadjuvant radiochemotherapy and combined regionalhyperthermia of rectal cancer: correlation with endorectal ultrasoundand histopathology. Eur J Nucl Med Mol Imaging 2004;31(6):811–9.

[85] Denecke T, Rau B, Hoffmann KT, et al. Comparison of CT, MRI andFDG-PET in response prediction of patients with locally advancedrectal cancer after multimodal preoperative therapy: is there a benefitin using functional imaging? Eur Radiol 2005;15:1658–66.

[86] Melton GB, Lavely WC, Jacene HA, et al. Efficacy of preoperativecombined 18-fluorodeoxyglucose positron emission tomography andcomputed tomography for assessing primary rectal cancer responseto neoadjuvant therapy. J Gastrointest Surg 2007;11:961–9.

[87] Vliegen RF, Beets-Tan RG, Vanhauten B, et al. Can an FDG-PET/CTpredict tumor clearance of the mesorectal fascia after preoperativechemoradiation of locally advanced rectal cancer? Strahlenther Onkol2008;184:457–64.

[88] Konski A, Li T, Sigurdson E, et al. Use of molecular imaging topredict clinical outcome in patients with rectal cancer after preop-erative chemotherapy and radiation. Int J Radiat Oncol Biol Phys2009;74(1):55–9.

[89] Cascini GL, Avallone A, Delrio P, et al. 18 F-FDG PET isan early predictor of pathologic tumor response to preoperativeradiochemotherapy in locally advanced rectal cancer. J Nucl Med2006;47:1241–8.

[90] Capirci C, Rubello D, Chierichetti F, et al. Long-term prognostic valueof 18F-FDG PET in patients with locally advanced rectal cancer previ-ously treated with neoadjuvant radiochemotherapy. Am J Roentgenol2006;187:202–8.

[91] Kalff V, Duong C, Drummond EG, Matthews JP, Hicks RJ. Find-ings on 18F-FDG PET scans after neoadjuvant chemoradiationprovides prognostic stratification in patients with locally advancedrectal carcinoma subsequently treated by radical surgery. J Nucl Med2006;47:14–22.

[92] Kalff V, Ware R, Heriot A, Chao M, Drummond E, Hicks RJ. Radi-

ation changes do not interfere with postchemoradiation restaging ofpatients with rectal cancer by FDG PET/CT before curative surgicaltherapy. Int J Radiation Oncology Biol Phys 2009;74(1):60–6.

[93] Chau I, Cunningham D. Treatment in advanced colorectal cancer:what, when and how? Br J Cancer 2009;100:1704–19.

gy/Hematology 77 (2011) 30–47 45

[94] Goshen E, Davidson T, Zwas ST, Aderka D. PET/CT in the eval-uation of response to treatment of liver metastases from colorectalcancer with bevacizumab and irinotecan. Technol Cancer Res Treat2006;5:37–43.

[95] Strauss LG, Koczan D, Klippel S, et al. Impact of angiogenesis-related gene expression on the tracer kinetics of 18F-FDG in colorectaltumors. J Nucl Med 2008;49:1238–44.

[96] Akhurst T, Kates TJ, Mazumdar M, et al. Recent chemotherapyreduces the sensitivity of [18F]fluorodeoxyglucose positron emissiontomography in the detection of colorectal metastases. J Clin Oncol2005;23:8713–6.

[97] Lubezky N, Metser U, Geva R, et al. The role and limita-tions of 18-fluoro-2-deoxy-D-glucose positron emission tomography(FDG-PET) scan and computerized tomography (CT) in restagingpatients with hepatic colorectal metastases following neoadjuvantchemotherapy: comparison with operative and pathological findings.J Gastrointest Surg 2007;11:472–8.

[98] Tan MC, Linehan DC, Hawkins WG, Siegel BA, Strasberg SM.Chemotherapy-induced normalization of FDG uptake by colorec-tal liver metastases does not usually indicate complete pathologicresponse. J Gastrointest Surg 2007;11:1112–9.

[99] Adie S, Yip C, Chu F, Morris DL. Resection of liver metastases fromcolorectal cancer: does preoperative chemotherapy affect the accuracyof PET in preoperative planning? ANZ J Surg 2009;79:358–61.

[100] Takahashi S, Kuroki Y, Nasu K, et al. Positron emission tomog-raphy with F-18 fluorodeoxyglucose in evaluating colorectalhepatic metastasis down-staged by chemotherapy. Anticancer Res2006;26:4705–11.

[101] Small RM, Lubezky N, Shmueli E, et al. Response to chemotherapypredicts survival following resection of hepatic colorectal metas-tases in patients treated with neoadjuvant therapy. J Surg Oncol2009;99:93–8.

[102] Findlay M, Young H, Cunningham D, et al. Noninvasive monitoring oftumor metabolism using fluorodeoxyglucose and positron emissiontomography in colorectal cancer liver metastases: correlation withtumor response to fluorouracil. J Clin Oncol 1996;14:700–8.

[103] Bender H, Bangard N, Metten N, et al. Possible role of FDG-PETin the early prediction of therapy outcome in liver metastases ofcolorectal cancer. Hybridoma 1999;18:87–91.

[104] Dimitrakopoulou-Strauss A, Strauss LG, Burger C, et al. Prognosticaspects of 18F-FDG PET kinetics in patients with metastatic col-orectal carcinoma receiving FOLFOX chemotherapy. J Nucl Med2004;45:1480–7.

[105] Dimitrakopoulou-Strauss A, Strauss LG, Rudi J. PET-FDG as pre-dictor of therapy response in patients with colorectal carcinoma. Q JNucl Med 2003;47:8–13.

[106] de Geus-Oei LF, van Laarhoven HW, Visser EP, et al. Chemotherapyresponse evaluation with FDG PET in patients with colorectal cancer.Ann Oncol 2007;19:348–52.

[107] Young H, Baum R, Cremerius U, et al. Measurement of clinicaland subclinical tumour response using [18F]-fluorodeoxyglucose andpositron emission tomography: review and 1999 EORTC recom-mendations. European Organization for Research and Treatment ofCancer (EORTC) PET Study Group. Eur J Cancer 1999;35:1773–82.

[108] Bystrom P, Berglund A, Garske U, et al. Early prediction of responseto first-line chemotherapy by sequential [18F]-2-fluoro-2-deoxy-D-glucose positron emission tomography in patients with advancedcolorectal cancer. Ann Oncol 2009;20:1057–62.

[109] Langenhoff BS, Oyen WJ, Jager GJ, et al. Efficacy of fluorine-18-deoxyglucose positron emission tomography in detecting tumorrecurrence after local ablative therapy for liver metastases: a prospec-

tive study. J Clin Oncol 2002;20:4453–8.

[110] Donckier V, Van Laethem JL, Goldman S, et al. [F-18] fluo-rodeoxyglucose positron emission tomography as a tool for earlyrecognition of incomplete tumor destruction after radiofrequencyablation for liver metastases. J Surg Oncol 2003;84:215–23.

Page 17: Positron emission tomography and colorectal cancer

4 Oncolo

6 M. Lin et al. / Critical Reviews in

[111] Anderson GS, Brinkmann F, Soulen MC, Alavi A, Zhuang H. FDGpositron emission tomography in the surveillance of hepatic tumorstreated with radiofrequency ablation. Clin Nucl Med 2003;28:192–7.

[112] Joosten J, Jager G, Oyen W, Wobbes T, Ruers T. Cryosurgery andradiofrequency ablation for unresectable colorectal liver metastases.Eur J Surg Oncol 2005;31:1152–9.

[113] Blokhuis TJ, van der Schaaf MC, van den Tol MP, Comans EFI,Manoliu RA, van der Sijp JR. Results of radio frequency abla-tion of primary and secondary liver tumors: long-term follow-upwith computed tomography and positron emission tomography-18F-deoxyfluoroglucose scanning. Scand J Gastroenterol (Suppl)2004;241:93–7.

[114] Veit P, Antoch G, Stergar H, Bockisch A, Forsting M, Kuehl H. Detec-tion of residual tumor after radiofrequency ablation of liver metastasiswith dual-modality PET/CT: initial results. Eur Radiol 2006;16:80–7.

[115] Travaini L, Trifiro G, Ravasi L, et al. Role of [18F]FDG-PET/CT afterradiofrequency ablation of liver metastases: preliminary results. EurJ Nucl Med Mol Imaging 2008;35:1316–22.

[116] Barker DW, Zagoria RJ, Morton KA, Kavanagh PV, ShenP. Evaluation of liver metastases after radiofrequency abla-tion: Utility of 18F-FDG PET and PET/CT. Am J Roentgenol2005;184(4):1096–102.

[117] Kuehl H, Antoch G, Stergar H, et al. Comparison of FDG-PET,PET/CT and MRI for follow-up of colorectal liver metastasestreated with radiofrequency ablation: Initial results. Eur J Radiol2008;67:362–71.

[118] Khandani AH, Calvo BF, O’Neil BH, Jorgenson J, Mauro MA. Apilot study of early 18F-FDG PET to evaluate the effectivenessof radiofrequency ablation of liver metastases. Am J Roentgenol2007;189:1199–202.

[119] Denecke T, Steffen I, Hildebrandt B, et al. Assessment of local controlafter laser-induced thermotherapy of liver metastases from colorectalcancer: contribution of FDG-PET in patients with clinical suspicionof progressive disease. Acta Radiol 2007;48:821–30.

[120] Avril N. 18F-FDG PET after radiofrequency ablation: is timing every-thing? J Nucl Med 2006;47:1235–7.

[121] Wong CY, Salem R, Raman S, Gates VL, Dworkin HJ. Evaluating90Y-glass microsphere treatment response of unresectable colorectalliver metastases by [18F]FDG PET: a comparison with CT or MRI.Eur J Nucl Med Mol Imaging 2002;29:815–20.

[122] Popperl G, Helmberger T, Munzing W, Schmind R, Jacobs TF,Tasch K. Selective internal radiation therapy with SIR-spheres inpatients with nonresectable liver tumours. Cancer Biother Radio-pharm 2005;20:200–8.

[123] Lin M, Shon IH, Wilson R, D’Amours SK, Schlaphoff G, LinP. Treatment response in liver metastases following 90Y SIR-spheres: an evaluation with PET. Hepatogastroenterology 2007;54:910–2.

[124] Biernert M, McCook B, Carr BI, et al. 90Y microsphere treatment ofunresectable liver metastases: changes in the FDG uptake and tumorsize on PET–CT. Eur J Nucl Med Mol Imaging 2005;32:778–87.

[125] Szyszko T, Al-Nahhas A, Canelo R, et al. Assessment of responseto treatment of unresectable liver tumours with Y90 microspheres:Value of FDG–PET versus computed tomography. Nucl Med Com-mun 2007;28:15–20.

[126] Wong CY, Salem R, Qing F, et al. Metabolic response after intraarte-rial 90Y-glass microsphere treatment for colorectal liver metastases:comparison of quantitative and visual analyses by 18F-FDG PET. JNucl Med 2004;45:1892–7.

[127] Wong CY, Qing F, Savin M, et al. Reduction of metastatic load to liverafter intraarterial hepatic yttrium-90 radioembolization as evaluatedby [18F]fluorodeoxyglucose positron emission tomographic imaging.

J Vasc Interv Radiol 2005;16:1101–6.

[128] Flamen P, Vanderlinden B, Delatte P, et al. Multimodality imaging canpredict the metabolic response of unresectable colorectal liver metas-tases to radioembolization therapy with Yttrium-90 labeled resinmicrospheres. Phys Med Biol 2008;53:6591–603.

gy/Hematology 77 (2011) 30–47

[129] Gregoire V, Haustermans K, Geets X, Roels S, Lonneux M. PET-based treatment planning in radiotherapy: a new standard? J NuclMed 2007;48:68–77S.

[130] Ciernik IF, Dizendorf E, Baumert BG, et al. Radiation treatmentplanning with an integrated Positron Emission and Computer Tomog-raphy (PET/CT): a feasibility study. Int J Radiat Oncol Biol Phys2003;57:853–63.

[131] Ciernik IF, Huser M, Burger C, Davis JB, Szekely G. Automated func-tional image-guided radiation treatment planning for rectal cancer. IntJ Radiat Oncol Biol Phys 2005;62:893–900.

[132] Patel DA, Chang ST, Goodman KA, et al. Impact of integratedPET/CT on variability of target volume delineation in rectal cancer.Technol Cancer Res Treat 2007;6:31–6.

[133] Anderson C, Koshy M, Staley C, et al. PET-CT fusion in radiationmanagement of patients with anorectal tumors. Int J Radiat OncolBiol Phys 2007;69:155–62.

[134] Bassi MC, Turri L, Sacchetti G, et al. FDG-PET/CT imagingfor staging and target volume delineation in preoperative confor-mal radiotherapy of rectal cancer. Int J Radiat Oncol Biol Phys2008;70:1423–6.

[135] Paskeviciute B, Bölling T, Brinkmann M, et al. Impact of 18F-FDG-PET/CT on staging and irradiation of patients with locally advancedrectal cancer. Strahlenther Onkol 2009;185:260–5.

[136] Roels S, Slagmolen P, Nuyts J, et al. Biological image-guided radio-therapy in rectal cancer: Is there a role for FMISO or FLT, next toFDG? Acta Oncol 2008;47:1237–48.

[137] Roels S, Slagmolen P, Nuyts J, et al. Biological image-guided radio-therapy in rectal cancer: challenges and pitfalls. Int J Radiat OncolBiol Phys 2009;75:782–90.

[138] Li Destri G, Di Cataldo A, Puleo S. Colorectal cancer follow-up:useful or useless? Surg Oncol 2006;15:1–12.

[139] Goldberg RM, Fleming TR, Tangen CM, et al. Surgery for recurrentcolon cancer:strategies for identifying resectable recurrence and suc-cess rates after resection. Eastern Cooperative Oncology Group, theNorth Central Cancer Treatment Group, and the Southwest OncologyGroup. Ann Intern Med 1998;129:27–35.

[140] Abgral R, Querellou S, Potard G, et al. Does 18F-FDG PET/CTimprove the detection of posttreatment recurrence of head and necksquamous cell carcinoma in patients negative for disease on clinicalfollow-up? J Nucl Med 2009;50:24–9.

[141] Brooks RA, Rader JS, Dehdashti F, et al. Surveillance FDG-PETdetection of asymptomatic recurrences in patients with cervical can-cer. Gynecol Oncol 2009;112:104–9.

[142] Selvaggi F, Cuocolo A, Sciaudone G, Maurea S, Giuliani A, Main-olfi C. FGD-PET in the follow-up of recurrent colorectal cancer.Colorectal Dis 2003;5:496–500.

[143] Sobhani I, Tiret E, Lebtahi R, et al. Early detection of recurrence by18FDG-PET in the follow-up of patients with colorectal cancer. Br JCancer 2008;98(5):875–80.

[144] Lieberman D. Colon cancer screening and surveillance controversies.Curr Opin Gastroenterol 2009;25(5):422–7.

[145] Yasuda S, Shohtsu A. Cancer screening with whole-body18F-fluorodeoxyglucose positron-emission tomography. Lancet1997;350:1819.

[146] Yasuda S, Fujii H, Nakahara T, et al. 18F-FDG PET detection ofcolonic adenomas. J Nucl Med 2001;42:989–92.

[147] Kojima S, Zhou B, Teramukai S, et al. Cancer screening of healthy vol-unteers using whole-body 18F-FDG-PET scans: The Nishidai clinicstudy. Eur J Cancer 2007;43:1842–8.

[148] Terauchi T, Murano T, Daisaki H, et al. Evaluation of whole-bodycancer screening using 18F-2-deoxy-2-fluoro-d-glucose positronemission tomography: a preliminary report. Ann Nucl Med

2008;22:379–85.

[149] Minamimoto R, Senda M, Uno K, et al. Performance profileof FDG-PET and PET/CT for cancer screening on the basisof a Japanese Nationwide Survey. Ann Nucl Med 2007;21:481–91.

Page 18: Positron emission tomography and colorectal cancer

Oncolo

B

aLoAtSot

aFESSLW

aFESSLW

MCS

M. Lin et al. / Critical Reviews in

[150] Nishizawa S, Kojima S, Teramukai S, et al. Prospective eval-uation of whole-body cancer screening with multiple modalitiesincluding [18F]Fluorodeoxyglucose Positron Emission Tomogra-phy in a healthy population: a preliminary report. J Clin Oncol2009;27:1767–73.

[151] Beatty JS, Williams HT, Aldridge BA, et al. Incidental PET/CT find-ings in the cancer patient: how should they be managed? Surgery2009;146(2):274–81.

[152] Hillman BJ. Screening for cancer with high technology imaging tests.J Clin Oncol 2009;27:1740–1.

[153] Kamel EM, Thumshirn M, Truninger K, et al. Significance of inciden-tal 18F-FDG accumulations in the gastrointestinal tract in PET/CT:correlation with endoscopic and histopathologic results. J Nucl Med2004;45:1804–10.

[154] Agress Jr H, Cooper BZ. Detection of clinically unexpected malignantand premalignant tumors with whole-body FDG PET: histopathologiccomparison. Radiology 2004;230:417–22.

[155] Israel O, Yefremov N, Bar-Shalom R, et al. PET/CT detectionof unexpected gastrointestinal foci of 18F-FDG uptake: inci-dence, localization patterns, and clinical significance. J Nucl Med2005;46:758–62.

[156] Even-Sapir E, Lerman H, Gutman M, et al. The presenta-tion of malignant tumours and pre-malignant lesions incidentallyfound on PET-CT. Eur J Nucl Med Mol Imaging 2006;33:541–52.

[157] Van Kouwen MCA, Nagengast FM, Jansen JBMJ, Oyen WJG, DrenthJPH. 2-(18F)-fluoro-2-deoxy-d-glucose positron emission tomogra-phy detects clinical relevant adenomas of the colon: a prospectivestudy. J Clin Oncol 2005;23:3713–7.

[158] Gutman F, Alberini J-L, Wartski M, et al. Incidental colonicfocal lesions detected by FDG PET/CT. Am J Roentgenol2005;185:495–500.

[159] Hemandas AK, Robson NK, Hickish T, Talbot RW. Colorec-tal tubulovillous adenomas identified on fluoro-2-deoxy-d-glucosepositron emission tomography/computed tomography scans. Colorec-tal Dis 2008;10(4):386–9.

[160] Lee ST, Tan T, Poon AMT, et al. Role of low-dose, noncon-trast computed tomography from integrated positron emissiontomography/computed tomography in evaluating incidental 2-deoxy-2-[F-18]fluoro-d-glucose-avid colon lesions. Mol Imaging Biol2008;10(1):48–53.

[161] Lee JC, Hartnett GF, Hughes BGM, Ravi Kumar AS. The segmentaldistribution and clinical significance of colorectal fluorodeoxyglu-cose uptake incidentally detected on PET-CT. Nucl Med Commun2009;30:333–7.

[162] Schlemmer HPW, Pichler BJ, Schmand M, et al. SimultaneousMR/PET imaging of the human brain: feasibility study. Radiology2008;248(3):1028–35.

[163] Giesel FL, Mehndiratta A, Locklin J, et al. Exp Oncol2009;31:106–14.

[164] Cherry SR. Multimodality imaging: beyond PET/CT and SPECT/CT.

Semin Nucl Med 2009;39:348–53.

[165] Wieder HA, Geinitz H, Rosenberg R, et al. PET imaging with [18F]3′-deoxy-3′-fluorothymidine for prediction of response to neoadjuvanttreatment in patients with rectal cancer. Eur J Nucl Med Mol Imaging2007;34:878–83.

mATS

gy/Hematology 77 (2011) 30–47 47

[166] Wieder H, Ott K, Zimmermann F, et al. PET imaging with[11C]methyl-l-methionine for therapy monitoring in patients withrectal cancer. Eur J Nucl Med 2002;29:789–96.

[167] Dietz DW, Dehdashti F, Grigsby PW, et al. Tumor hypoxia detectedby positron emission tomography with 60Cu-ATSM as a predictor ofresponse and survival in patients undergoing neoadjuvant chemora-diotherapy for rectal carcinoma: a pilot study. Dis Col Rectum2008;51:1641–8.

iographies

Michael Lin is a Staff Specialist in Nuclear Medicinend PET at the Department of Nuclear Medicine and PET,iverpool Hospital, Sydney, Australia. Conjoint Lecturerf Faculty of Medicine, University of New South Wales,ustralia. Member of the South Western Sydney Colorec-

al Tumour Group, Sydney South West Area Health Service,ydney, Australia. Regional International Adviser and Fellowf the Royal College of Physicians of London and Fellow ofhe Royal Australasian College of Physicians.

Karen Wong is a Staff Specialist in Radiation Oncologyt Liverpool Hospital, Sydney, Australia. Clinical Researchellow at Collaboration for Cancer Outcomes Research andvaluation (CCORE), Liverpool Hospital; Member of theouth Western Sydney Colorectal Tumour Group, Sydneyouth West Area Health Service, Sydney, Australia. Conjointecturer of Faculty of Medicine, University of New Southales, Australia.

Weng Leong Ng is a Staff Specialist in Medical Oncologyt Liverpool Hospital, Sydney, Australia. Clinical Researchellow at Collaboration for Cancer Outcomes Research andvaluation (CCORE), Liverpool Hospital; Member of theouth Western Sydney Colorectal Tumour Group, Sydneyouth West Area Health Service, Sydney, Australia. Conjointecturer of Faculty of Medicine, University of New Southales, Australia.

Ivan Ho Shon is a Senior Staff Specialist in Nuclearedicine and PET at Liverpool Hospital, Sydney, Australia.onjoint Lecturer of Faculty of Medicine, University of Newouth Wales, Australia.

Matthew Morgan is a Visiting Medical Officer, Depart-

ent of Colorectal Surgery, Bankstown Hospital, Sydney,ustralia. Chair of the South Western Sydney Colorectalumour Group, Sydney South West Area Health Service,ydney, Australia.