Abstract

Background

Selection of suitable candidates for intraoperative tumour detection and cytoreductive surgery (CRS) combined with hyperthermic intraperitoneal chemotherapy (HIPEC) is important for improving outcomes for patients with colorectal peritoneal metastases. Previous research demonstrated the use of single-photon emission computed tomography (SPECT), intraoperative radiodetection, and near-infrared fluorescence (NIRF)-guided surgery with a dual-labelled 111In-labelled dodecane tetra-acetic acid (DOTA)–labetuzumab-IRDye800CW tracer to detect peritoneal metastases before operation. The aim of this study was to validate these results.

Methods

A single-centre phase II study was conducted to evaluate the safety and feasibility of 111In-labelled DOTA–labetuzumab-IRDye800CW in patients with colorectal peritoneal metastases undergoing CRS-HIPEC. SPECT/computed tomography (CT) was undertaken before surgery, after intravenous administration of 10 mg 111In-labelled DOTA–labetuzumab-IRDye800CW (mean 101.25 MBq). During surgery, radiodetection and NIRF imaging were used for tumour detection. Adverse events were assessed, and tumour-to-background ratios (TBRs) and peritoneal cancer index scores were analysed.

Results

Seven patients were included. No study-related severe adverse events were reported. Imaging before surgery revealed previously undetected metastases in one patient. The mean(standard deviation, s.d.) SPECT/CT peritoneal cancer index score was 3(2), and the intraoperative score was 14(7) (P = 0.032). A total of 52 lesions were removed during CRS, of which 37 were malignant. With NIRF imaging, 34 (92%) of 37 malignant lesions were detectable. Of 52 fluorescent lesions, 4 were false-positive. Mean(s.d.) fluorescence TBR was 3.4(1.8) and mean radiodetection TBR was 4.4(1.4).

Conclusion

This study confirmed the safety and feasibility of multimodal image-guided surgery in patients with peritoneal metastases.

Introduction

Colorectal cancer may spread to the peritoneal cavity, with peritoneal metastases developing in 3–20% of patients, depending on stage and location of the primary tumour1,2. Peritoneal metastatic disease is challenging to treat, as it is often an end-stage disease with a poor prognosis3,4. Survival rates began to improve with the introduction of cytoreductive surgery (CRS) combined with hyperthermic intraperitoneal chemotherapy (HIPEC)5–8. However, because of the significant morbidity associated with the procedure, it is important to select patients eligible for CRS-HIPEC before surgery9,10. CRS-HIPEC should only be conducted in patients with an acceptable peritoneal tumour burden relative to the anticipated improvement in prognosis, as estimated using the peritoneal cancer index (PCI) or other grading methods10,11. Conventional imaging techniques such as positron emission tomography or computed tomography (CT) generally underestimate the PCI, so surgeons rely on direct and tactile examinations during surgery to detect additional lesions and to distinguish between benign fibrosis and malignant lesions12–14. Enhanced tissue discrimination and higher sensitivity in detection through improved imaging potentially increases the likelihood of achieving complete cytoreduction, which is mandatory for patient survival.

Near-infrared fluorescence (NIRF) imaging techniques using tumour-targeting tracers, such as 111In-labelled dodecane tetra-acetic acid (DOTA)–labetuzumab-IRDye800CW15, a carcinoembryonic antigen (CEA)-specific tumour-targeted dual-labelled agent, may aid in the intraoperative identification of malignant tissue of colorectal origin16–18. Multimodal imaging after administration of a dual-labelled tracer combining a γ-emitting radionuclide and a near-infrared fluorophore on the same targeting molecule may potentially improve cytoreductive surgery19. The radionuclide not only enables single-photon emission CT (SPECT)/CT before surgery, but also intraoperative radio-guided differentiation between benign and malignant tissue using a handheld γ probe20. The dual-labelled tracer used in this research enables the use of both with a single agent.

In a proof-of-concept phase I dose escalation study, it was demonstrated that the use of 111In-labelled DOTA–labetuzumab-IRDye800CW for tumour detection both before and during surgery in 15 patients was safe and identified the optimal dose of tracer (10 mg). A higher dose (50 mg) increased the incidence of false-positive lesions, and detectability was too low with use of a lower dose (2 mg) in this study21.

It is important to validate the results from the phase I study with the optimized dosage22,23. The aims of this confirmatory trial were to assess the reproducibility of the previous results on safety and feasibility of SPECT/CT, intraoperative radio detection, and NIRF-guided surgery after intravenous administration of the optimal dose of 10 mg 111In-labelled DOTA–labetuzumab-IRDye800CW for the detection and treatment of peritoneal metastases of colorectal origin before surgery.

Methods

A single-centre, open-label confirmatory cohort phase II study (NCT03699332) was conducted to evaluate the safety and feasibility of image-guided surgery using 111In-labelled DOTA–labetuzumab-IRDye800CW.

The preparation of 111In-labelled DOTA–labetuzumab-IRDye800CW was described previously21. Briefly, labetuzumab (Research Resource Identifier AB_2910852) was first incubated with IRDye800CW-N-hydroxysuccinimide (NHS) and DOTA-NHS. After conjugation, the product was purified via dialysis, sterile filtrated, aliquoted, and stored. On the day of injection, DOTA–labetuzumab-IRDye800CW was radiolabelled with 111In (100 MBq).

Subject selection

Patients aged ≥ 18 years with confirmed peritoneal metastases of colorectal cancer scheduled for CRS-HIPEC or redo CRS-HIPEC were selected for inclusion. Exclusion criteria were: pregnant or breastfeeding women; patients with a serum CEA concentration > 500 ng/ml; a known CEA-negative malignancy; recent administration of a radionuclide no later than 10 physical half-lives before study enrolment; or the presence of other uncontrolled medical conditions potentially jeopardizing the patients’ well-being and/or the study. The trial was approved by the regional ethical review board MREC Oost-Nederland, formerly known as Committee on Research Involving Human Subjects region Arnhem–Nijmegen, adhering to the principles of the Declaration of Helsinki. Before participation, written informed consent was obtained from all patients.

Safety

Patients received a single intravenous dose of 111In-labelled DOTA–labetuzumab-IRDye800CW 5 or 6 days before surgery and were continuously monitored for adverse events (AEs) up to 3 hours after injection. AEs associated with the study procedures, not necessarily linked to 111In-labelled DOTA–labetuzumab-IRDye800CW administration, were considered treatment-related if they occurred within 10 days of surgery, in accordance with the National Cancer Institute Common Terminology Criteria for AEs (version 4.03).

SPECT/CT before surgery

This was conducted 4 or 5 days following the administration of the dual-labelled antibody to obtain images of the thorax and abdomen. A physician specializing in nuclear medicine assessed the accumulation of 111In-labelled DOTA–labetuzumab-IRDye800CW using the PCI score.

Image-guided surgery using NIRF camera and γ probe

Following surgical exposure and examination of the abdomen, the PCI was evaluated using standard methods. Subsequently, fluorescence-based PCI assessment was conducted using a Spectrum® NIRF camera (Quest Medical Imaging, Middenmeer, the Netherlands). Radiosignal counts for background measurements were taken from the liver, abdominal wall, and colon. Cytoreductive surgery was then carried out in accordance with usual practice, including the excision of any additional lesions detected with NIRF imaging or increased radiosignal.

After complete cytoreduction, exploration of the abdominal cavity was carried out using both the Europrobe© 3.2 SOE 3216-7 (Eurorad SA, Eckbolsheim, France) and the Spectrum® NIRF camera for remnant tumour tissue. Radiodetection and NIRF imaging were conducted on all resected tissue specimens. Quantification of tracer accumulation was undertaken through back-table γ probe detection for 5 seconds on all suspected malignant lesions, with the highest result being registered for analysis. For each lesion, one corresponding benign region (background tissue) was used as a negative control.

Tissue analysis

Tissue slides (4 µm) from all formalin-fixed paraffin-embedded blocks were scanned using an Odyssey® CLx flatbed fluorescence imaging system (LI-COR Biosciences, Lincoln, NE, USA) at 800 nm for fluorescent signal detection. The images were then exported in tagged image file format using Image Studio™ Lite 5.2.5 (LI-COR Biosciences). After scanning, the sections were stained with haematoxylin and eosin and for CEA to confirm the presence of colorectal tumour cells. Following this confirmation, specific regions of interest (ROIs), delineating tumours and adjacent normal tissues, were identified on the haematoxylin and eosin slides using CaseViewer™ 2.4 (3Dhistech, Budapest, Hungary). These ROIs were subsequently imported into Fiji-ImageJ (Version 1.51) for quantifying mean fluorescence intensity in both tumour and benign tissues24.

Study outcomes

The primary outcomes of this study were the safety and feasibility of multimodal image-guided surgery following the intravenous administration of 111In-labelled DOTA–labetuzumabIRDye800CW in patients with CRS-HIPEC with colorectal peritoneal carcinomatosis. The safety of image-guided surgery relied on the absence of severe AEs related to 111In-labelled DOTA–labetuzumab-IRDye800CW administration or study procedures. Feasibility was determined by detecting specific accumulation of 111In-labelled DOTA–labetuzumab-IRDye800CW in CEA-expressing tumour tissue using available technologies (radiodetection, optical imaging, and (immuno)histological analyses).

Secondary outcomes were identifying additional lesions via NIRF imaging after standard CRS, evaluating tumour-to-background ratios (TBRs) through ex vivo γ measurements of surgical specimens and flatbed fluorescence measurements of 4-µm tissue sections, and assessing the relationship between tracer accumulation and CEA expression. This study did not repeat dose assessments or pharmacokinetic analyses from the previous proof-of-concept study21.

Statistical analysis

Descriptive statistics, including mean radiosignal and fluorescence-based TBRs, are depicted as mean(standard deviation, s.d.). SPECT/CT PCI and surgical PCI scores were depicted as mean(s.d.) and compared using a paired-samples Student’s t test. Statistical analyses were undertaken using GraphPad™ Prism version 5.03 (GraphPad Software, San Diego, CA, USA) and SPSS® version 25.0 (IBM, Armonk, NY, USA). P < 0.050 was considered statistically significant.

Results

Patient characteristics and safety

A total of 41 patients were scheduled for cytoreductive surgery, of whom 9 had an unsuitable tumour on histological assessment. Eleven patients provided written informed consent for the study; four patients were excluded because of technical issues with tracer or equipment, leaving seven for assessment in this study (Fig. 1). Median age of the participants was 71 (range 48–82) years. Temperature, blood pressure, and haematological markers for liver and renal function remained stable in the hours after injection (day 0), at time of the SPECT/CT (day 4/5), and until the day of surgery (day 5/6). The mean 111In dose received was 101.25 (range 94.72–104.5) MBq. Median length of hospital stay was 14 (range 5–47) days (Table 1). No study-related AEs occurred according to National Cancer Institute Common Terminology Criteria; however, five severe AEs were reported, though none were related to study procedures. All reported AEs are summarized in Table 2.

Flow diagram showing patient inclusion
Fig. 1

Flow diagram showing patient inclusion

CRS, cytoreductive surgery; HIPEC, hyperthermic intraperitoneal chemotherapy.

Table 1

Clinical characteristics

PatientAge (years), sexTreatment before surgeryPattern of metastasisHistologyCompleteness of cytoreduction scoreClinical PCI scoreSPECT PCI
score
Pathological PCI scoreLength of hospital stay (days)
171, FNoneSMACCC01421443
270, FNoneSMMACCC0143314
348, FNoneMMACCC02142111
478, MNoneMMACCC01541514
582, MNoneSMACCC032324
673, MSystemic therapySMACCC060447
750, FNoneSMMACCC2> 236NA5
PatientAge (years), sexTreatment before surgeryPattern of metastasisHistologyCompleteness of cytoreduction scoreClinical PCI scoreSPECT PCI
score
Pathological PCI scoreLength of hospital stay (days)
171, FNoneSMACCC01421443
270, FNoneSMMACCC0143314
348, FNoneMMACCC02142111
478, MNoneMMACCC01541514
582, MNoneSMACCC032324
673, MSystemic therapySMACCC060447
750, FNoneSMMACCC2> 236NA5

PCI, peritoneal cancer index; SPECT, single-photon emission computed tomography; F, female; SM, synchronous metastases; AC, adenocarcinoma; MAC, mucinous adenocarcinoma; MM, metachronous metastases; M, male.

Table 1

Clinical characteristics

PatientAge (years), sexTreatment before surgeryPattern of metastasisHistologyCompleteness of cytoreduction scoreClinical PCI scoreSPECT PCI
score
Pathological PCI scoreLength of hospital stay (days)
171, FNoneSMACCC01421443
270, FNoneSMMACCC0143314
348, FNoneMMACCC02142111
478, MNoneMMACCC01541514
582, MNoneSMACCC032324
673, MSystemic therapySMACCC060447
750, FNoneSMMACCC2> 236NA5
PatientAge (years), sexTreatment before surgeryPattern of metastasisHistologyCompleteness of cytoreduction scoreClinical PCI scoreSPECT PCI
score
Pathological PCI scoreLength of hospital stay (days)
171, FNoneSMACCC01421443
270, FNoneSMMACCC0143314
348, FNoneMMACCC02142111
478, MNoneMMACCC01541514
582, MNoneSMACCC032324
673, MSystemic therapySMACCC060447
750, FNoneSMMACCC2> 236NA5

PCI, peritoneal cancer index; SPECT, single-photon emission computed tomography; F, female; SM, synchronous metastases; AC, adenocarcinoma; MAC, mucinous adenocarcinoma; MM, metachronous metastases; M, male.

PatientTime after injection (days)AEInterventionRelated to studyCTCAE gradeSevere AE
111
12
13
16
Fever (without focus)
Chyle leakage
Gastroparesis
Aspiration pneumonia
Antibiotics
MCT diet
Nasogastric tube
Antibiotics
No
No
No
No
1
1
3
3
No
No
No
No
27CVC line infectionAntibioticsNo3No
411Subsegmental pulmonary embolismNon-invasive ventilationNo2Yes
511
11
14
18
Multiple pulmonary embolism
Ileus
Jugular vein thrombus
Fascial dehiscence
Enema
Relaparotomy
No
No
No
No
1
3
2
3
No
No
No
Yes
614
18
25
27
Anastomotic leakage
Wound infection
Parastomal abscess
Fascial dehiscence
Relaparotomy
Antibiotics
Drainage
Relaparotomy
No
No
No
No
3
3
1
3
Yes
Yes
Yes
No
PatientTime after injection (days)AEInterventionRelated to studyCTCAE gradeSevere AE
111
12
13
16
Fever (without focus)
Chyle leakage
Gastroparesis
Aspiration pneumonia
Antibiotics
MCT diet
Nasogastric tube
Antibiotics
No
No
No
No
1
1
3
3
No
No
No
No
27CVC line infectionAntibioticsNo3No
411Subsegmental pulmonary embolismNon-invasive ventilationNo2Yes
511
11
14
18
Multiple pulmonary embolism
Ileus
Jugular vein thrombus
Fascial dehiscence
Enema
Relaparotomy
No
No
No
No
1
3
2
3
No
No
No
Yes
614
18
25
27
Anastomotic leakage
Wound infection
Parastomal abscess
Fascial dehiscence
Relaparotomy
Antibiotics
Drainage
Relaparotomy
No
No
No
No
3
3
1
3
Yes
Yes
Yes
No

AE, adverse event; CTCAE, common terminology criteria for adverse events; MCT, medium chain triglycerides; CVC, central venous catheter.

PatientTime after injection (days)AEInterventionRelated to studyCTCAE gradeSevere AE
111
12
13
16
Fever (without focus)
Chyle leakage
Gastroparesis
Aspiration pneumonia
Antibiotics
MCT diet
Nasogastric tube
Antibiotics
No
No
No
No
1
1
3
3
No
No
No
No
27CVC line infectionAntibioticsNo3No
411Subsegmental pulmonary embolismNon-invasive ventilationNo2Yes
511
11
14
18
Multiple pulmonary embolism
Ileus
Jugular vein thrombus
Fascial dehiscence
Enema
Relaparotomy
No
No
No
No
1
3
2
3
No
No
No
Yes
614
18
25
27
Anastomotic leakage
Wound infection
Parastomal abscess
Fascial dehiscence
Relaparotomy
Antibiotics
Drainage
Relaparotomy
No
No
No
No
3
3
1
3
Yes
Yes
Yes
No
PatientTime after injection (days)AEInterventionRelated to studyCTCAE gradeSevere AE
111
12
13
16
Fever (without focus)
Chyle leakage
Gastroparesis
Aspiration pneumonia
Antibiotics
MCT diet
Nasogastric tube
Antibiotics
No
No
No
No
1
1
3
3
No
No
No
No
27CVC line infectionAntibioticsNo3No
411Subsegmental pulmonary embolismNon-invasive ventilationNo2Yes
511
11
14
18
Multiple pulmonary embolism
Ileus
Jugular vein thrombus
Fascial dehiscence
Enema
Relaparotomy
No
No
No
No
1
3
2
3
No
No
No
Yes
614
18
25
27
Anastomotic leakage
Wound infection
Parastomal abscess
Fascial dehiscence
Relaparotomy
Antibiotics
Drainage
Relaparotomy
No
No
No
No
3
3
1
3
Yes
Yes
Yes
No

AE, adverse event; CTCAE, common terminology criteria for adverse events; MCT, medium chain triglycerides; CVC, central venous catheter.

SPECT/CT before surgery

Six of seven patients underwent SPECT/CT 4 or 5 days after tracer injection. One patient did not undergo SPECT/CT because of suspected COVID-19 infection. SPECT/CT revealed peritoneal metastases in five of six patients. The mean(s.d.) SPECT/CT PCI score was 3(2). CEA-targeted SPECT/CT revealed lesions such as primary tumours, ovarian metastases, lymph node metastases, and some peritoneal metastases, all > 10 mm. In one patient, there were additional findings outside the abdominal cavity during SPECT/CT, which showed accumulation of tracer in retroclavicular and paraoesophageal nodes (Fig. 2). This patient also had extensive peritoneal disease during laparotomy with a PCI score > 23.

Multimodal imaging of previously undetected lymph node metastases (patient 7)
Fig. 2

Multimodal imaging of previously undetected lymph node metastases (patient 7)

a Retroclavicular node, b para-oesophageal node. SPECT, single-photon emission computed tomography; CT, computed tomography.

Surgical procedure and histopathological analysis

All patients were primary candidates for CRS-HIPEC. Six patients underwent complete cytoreduction of peritoneal metastases. Two of these had synchronous metastasis and their primary tumours were resected in the same procedure. One patient did not undergo CRS-HIPEC because of a high clinical PCI score during exploratory laparotomy, resulting in termination of the procedure. The mean(s.d.) surgical PCI was 14(7), which was significantly higher than the mean SPECT/CT PCI score of 3(2) (P =0.032). The mean pathological PCI score was 9(8).

In total, 52 lesions were removed for pathological assessment and measured by NIRF imaging. Final pathology demonstrated that 37 of these were malignant, of which 34 (92%) were detected with NIRF imaging. Four tumour-positive lesions were not detected by NIRF imaging. Four of the 52 lesions (8%) were found to be histopathologically benign but were falsely assessed as positive for fluorescence by the surgeon. All these false-positive lesions were found in one patient, and pathology revealed three granulomas and a diverticulum.

Back-table γ counting was undertaken in five of seven patients because of technical issues with the probe in two patients. In 48 of the 52 resected lesions, γ counting was undertaken. This revealed a mean(s.d.) TBR of 4.4(1.4) in all malignant lesions (Fig. 3). Three of the four false-positive fluorescent lesions emitted a low radiosignal, with a mean TBR of 1.3(0.5). One of the false-positive fluorescent lesions had a radiosignal TBR of 2.7.

Mean fluorescence and mean radiosignal TBR per patient
Fig. 3

Mean fluorescence and mean radiosignal TBR per patient

Black dots indicate values for individual patients. Median value (bold line), interquartile range (box), and range (error bars) are also shown. TBR, tumour-to-background ratio.

A total of 65 histological slides from the 52 lesions were included in the final analysis, and all slides exhibited clear overlap of the NIRF signal and localization of CEA-expressing tumour cells. Fluorescence TBR was 3.4(1.8) (Fig. 3). Further detailed fluorescent and radioactive measurements are provided in the supplementary material.

Multimodal image-guided surgery resulted in alteration of the clinical strategy in three of seven patients. A total of five lesions were missed initially during surgical inspection, but were identified by NIRF imaging, resulting in additional resections of NIRF-positive lesions (Fig. 4).

In vivo and ex vivo fluorescence detection in peritoneal metastases (patient 1)
Fig. 4

In vivo and ex vivo fluorescence detection in peritoneal metastases (patient 1)

a Intraoperative near-infrared fluorescence (NIRF) imaging of a metastatic node near the medial colic artery (MCA), b  ex vivo NIRF imaging of suspicious node near the superior mesenteric vein (SMV). From top to bottom: NIR-fluorescence colour overlay, NIR-fluorescence, brightfield.

Discussion

In this confirmatory trial, the optimal dose of 10 mg 111In-labelled DOTA–labetuzumab-IRDye800CW, as established in a previous phase I dose-finding study21, was administered to seven patients. Intravenous administration of the compound was safe and well tolerated, with no AEs reported during the study period. Tumour-targeted intraoperative NIRF imaging and radioguidance enabled successful identification, localization, and delineation of colorectal tumours, lymph node metastases, and peritoneal metastases. These findings were supported by quantitative analyses of TBRs for both fluorescence and radiosignals conducted ex vivo. Five additional lesions that were initially missed by routine abdominal evaluation during surgery were detected with intraoperative NIRF imaging. In three patients, this resulted in additional surgical resections. This further supports the value of intraoperative NIRF imaging and radiodetection as shown in the previous study21.

During surgery, NIRF imaging confirmed 92% of the malignant lesions, which was comparable to the 16 malignant lesions (95%) found in the previously described five patients with the 10-mg dose21. A similar study25 including patients undergoing HIPEC was undertaken, using a single-labelled fluorescent tracer, that showed a sensitivity of 98.5%. This small discrepancy in sensitivity can perhaps be explained by the experience of the surgical team with the fluorescence camera or different tumour locations. Of the 52 excised lesions, four (8%) were false-positive on NIRF imaging; this was also comparable to the previous 10-mg cohort, which showed three lesions (16%) as false-positive21. A possible explanation could be that differentiating genuine positive fluorescence from autofluorescence or non-tumour-related causes is not consistently feasible, as shown in previous studies16,26,27. Three lesions showed low tracer uptake (mean TBR 1.3), confirming the additional significance of the radiosignal. Multimodal imaging using a dual-labelled tracer is therefore a promising technique whereby radionuclide detection is complementary to NIRF imaging. Fluorescence signals can be detected with high spatial accuracy, but only superficially, in contrast to radionuclide detection, which makes it possible to examine deeper tissues.

Imaging with CEA-targeted SPECT/CT before surgery did not contribute to a more reliable PCI assessment, but revealed a contraindication to CRS-HIPEC by detecting retroclavicular and paraoesophageal lymph nodes in one patient that were missed on standard imaging. This is clinically significant because patients with peritoneal metastases and extraperitoneal/systemic disease are usually not eligible for CRS-HIPEC. CEA-targeted SPECT/CT could be used to avoid futile laparotomies and associated morbidity in the future, serving as a potentially valuable diagnostic tool with an impact on treatment policies. In the future, it is likely that CT scanners will have increased resolution and speed that enable visualization of even smaller anatomical structures and abnormalities, resulting in a higher sensitivity for this tracer and maximizing the impact of dual-labelled tracers28. This could also be applicable to other CEA-expressing malignancies, such as oesophageal and pancreatic cancers26,29–31.

The authors are currently studying whether multimodal imaging can be applied to patients with rectal and pancreatic cancer, using another multimodal 111In-labelled DOTA–anti-CEA antibody (NCT06395337). The principle of multimodal imaging using dual-labelled antibodies is a promising research development that extends beyond gastrointestinal malignancies. For example, 111In-labelled DOTA–girentuximab-IRDye800CW, a carbonic anhydrase IX-targeting antibody previously used in clear cell renal cell carcinoma19, and the monoclonal antibody D2B32 named 111In-labelled prostate-specific membrane antigen-IRDye800CW, which has been used in prostate carcinoma, have both seen promising results32. Studies with fluorescently labelled antibodies (without a radiolabel), such as SGM-101, have also been shown to be feasible18,26,33. Currently, a multicentre phase II–III study is ongoing in the Netherlands using SGM-101 in locally advanced and recurrent rectal cancer (NCT04642924).

Complete surgical resection remains fundamental in CRS-HIPEC treatment in patients with metastatic colorectal disease34; therefore, clinicians will persistently encounter significant procedure-related morbidity while undertaking extensive resections35,36. Intraoperative imaging appears to be a promising technique in supporting the surgeon in targeted resection. Previous studies have already demonstrated its potential value to patients with colorectal metastases. Boogerd et al.16 used a fluorescent anti-CEA tracer in patients with both primary and metastatic colorectal cancer and demonstrated alterations of surgical strategy based on intraoperative fluorescence imaging in 35% of patients. Combining the study results of de Gooyer et al.21 and this present study, 12 patients in total have received 10 mg 111In-labelled DOTA–labetuzumab-IRDye800CW. This resulted in a change of clinical management in six patients by additional resection of suspected lesions based on intraoperative imaging. Furthermore, extraperitoneal disease was demonstrated in two patients; this had a potentially significant impact, as a major procedure with futile advantage for the patient could have been avoided.

Four false-negative non-fluorescent lesions were detected, of which three were in one patient. These lesions were deemed malignant after γ counting, with a mean TBR of 2.5(0.49), and pathological analysis. Of all 46 clinically suspicious lesions 11 (24%) did not contain malignant cells, supporting the overestimation of clinical PCI37. The specificity and negative predictive value of multimodal imaging needs confirmation in a broader study population. Nevertheless, these findings suggest that, in the future, surgeons might opt to leave non-fluorescent lesions in situ if no raised radiosignal is detected, potentially resulting in more precise cytoreduction and reduced surgical trauma.

Funding

The authors have no funding to declare.

Author contributions

Aaya Darai (Data curation, Formal analysis, Methodology, Writing—original draft preparation), Jan Marie de Gooyer (Conceptualization, Data curation, Formal analysis, Methodology, Writing—review & editing), Erik H. J. G. Aarntzen (Data curation, Formal analysis, Writing—review & editing), Sander Ubels (Data curation, Investigation, Writing—review & editing), Andre Bremers (Data curation, Writing—review & editing), Philip de Reuver (Data curation, Writing—review & editing), Iris Nagtegaal (Formal analysis, Writing—review & editing), Mark Rijpkema (Conceptualization, Methodology, Supervision, Writing—review & editing), and Johannes De Wilt (Conceptualization, Data curation, Methodology, Supervision, Writing—original draft, Writing—review & editing)

Disclosure

The authors declare no conflict of interest.

Supplementary material

Supplementary material is available at BJS Open online.

Data availability

This study is a replication of a previously published study. Because of patient privacy constraints, the data produced during this trial are not publicly accessible. However, they can be obtained from the corresponding author upon reasonable request and approval from the trial sponsor, following the applicable guidelines at the time of the request.

References

1

Rijken
 
A
,
van de Vlasakker
 
VCJ
,
Simkens
 
GA
,
Rovers
 
KP
,
van Erning
 
FN
,
Koopman
 
M
 et al.  
Primary tumor resection or systemic treatment as palliative treatment for patients with isolated synchronous colorectal cancer peritoneal metastases in a nationwide cohort study
.
Clin Exp Metastasis
 
2023
;
40
:
289
298

2

van Gestel
 
YRBM
,
Thomassen
 
I
,
Lemmens
 
VEPP
,
Pruijt
 
JFM
,
van Herk-Sukel
 
MPP
,
Rutten
 
HJT
 et al.  
Metachronous peritoneal carcinomatosis after curative treatment of colorectal cancer
.
Eur J Surg Oncol
 
2014
;
40
:
963
969

3

Razenberg
 
LG
,
Lemmens
 
VEPP
,
Verwaal
 
VJ
,
Punt
 
CJA
,
Tanis
 
PJ
,
Creemers
 
G-J
 et al.  
Challenging the dogma of colorectal peritoneal metastases as an untreatable condition: results of a population-based study
.
Eur J Cancer
 
2016
;
65
:
113
120

4

Quere
 
P
,
Facy
 
O
,
Manfredi
 
S
,
Jooste
 
V
,
Faivre
 
J
,
Lepage
 
C
 et al.  
Epidemiology, management, and survival of peritoneal carcinomatosis from colorectal cancer: a population-based study
.
Dis Colon Rectum
 
2015
;
58
:
743
752

5

Verwaal
 
VJ
,
Bruin
 
S
,
Boot
 
H
,
van Slooten
 
G
,
van Tinteren
 
H
.
8-year follow-up of randomized trial: cytoreduction and hyperthermic intraperitoneal chemotherapy versus systemic chemotherapy in patients with peritoneal carcinomatosis of colorectal cancer
.
Ann Surg Oncol
 
2008
;
15
:
2426
2432

6

Kuijpers
 
AM
,
Mirck
 
B
,
Aalbers
 
AGJ
,
Nienhuijs
 
SW
,
de Hingh
 
IHJT
,
Wiezer
 
MJ
 et al.  
Cytoreduction and HIPEC in the Netherlands: nationwide long-term outcome following the Dutch protocol
.
Ann Surg Oncol
 
2013
;
20
:
4224
4230

7

Elias
 
D
,
Gilly
 
F
,
Boutitie
 
F
,
Quenet
 
F
,
Bereder
 
J-M
,
Mansvelt
 
B
 et al.  
Peritoneal colorectal carcinomatosis treated with surgery and perioperative intraperitoneal chemotherapy: retrospective analysis of 523 patients from a multicentric French study
.
J Clin Oncol
 
2010
;
28
:
63
68

8

van der Ven
 
RGFM
,
van den Heuvel
 
TBM
,
Rovers
 
KPB
,
Nienhuijs
 
SW
,
Boerma
 
D
,
van Grevenstein
 
WMU
 et al.  
Towards equal access to cytoreductive surgery with hyperthermic intraperitoneal chemotherapy and survival in patients with isolated colorectal peritoneal metastases: a nationwide population-based study
.
Ann Surg Oncol
 
2024
;
31
:
3758
3768

9

Hallam
 
S
,
Tyler
 
R
,
Price
 
M
,
Beggs
 
A
,
Youssef
 
H
.
Meta-analysis of prognostic factors for patients with colorectal peritoneal metastasis undergoing cytoreductive surgery and heated intraperitoneal chemotherapy
.
BJS Open
 
2019
;
3
:
585
594

10

van Oudheusden
 
TR
,
Braam
 
HJ
,
Luyer
 
MDP
,
Wiezer
 
MJ
,
van Ramshorst
 
B
,
Nienhuijs
 
SW
 et al.  
Peritoneal cancer patients not suitable for cytoreductive surgery and HIPEC during explorative surgery: risk factors, treatment options, and prognosis
.
Ann Surg Oncol
 
2015
;
22
:
1236
1242

11

Jacquet
 
P
,
Sugarbaker
 
PH
.
Clinical research methodologies in diagnosis and staging of patients with peritoneal carcinomatosis
.
Cancer Treat Res
 
1996
;
82
:
359
374

12

Rivard
 
JD
,
Temple
 
WJ
,
McConnell
 
YJ
,
Sultan
 
H
,
Mack
 
LA
.
Preoperative computed tomography does not predict resectability in peritoneal carcinomatosis
.
Am J Surg
 
2014
;
207
:
760
764
.

13

Laghi
 
A
,
Bellini
 
D
,
Rengo
 
M
,
Accarpio
 
F
,
Caruso
 
D
,
Biacchi
 
D
 et al.  
Diagnostic performance of computed tomography and magnetic resonance imaging for detecting peritoneal metastases: systematic review and meta-analysis
.
Radiol Med
 
2017
;
122
:
1
15

14

Elekonawo
 
FMK
,
Starremans
 
B
,
Laurens
 
ST
,
Bremers
 
AJA
,
de Wilt
 
JHW
,
Heijmen
 
L
 et al.  
Can [18F]F-FDG PET/CT be used to assess the pre-operative extent of peritoneal carcinomatosis in patients with colorectal cancer?
 
Abdominal Radiology
 
2020
;
45
:
301
306

15

Rijpkema
 
M
,
Oyen
 
WJ
,
Bos
 
D
,
Franssen
 
GM
,
Goldenberg
 
DM
,
Boerman
 
OC
.
SPECT- and fluorescence image-guided surgery using a dual-labeled carcinoembryonic antigen-targeting antibody
.
J Nucl Med
 
2014
;
55
:
1519
1524

16

Boogerd
 
LSF
,
Hoogstins
 
CES
,
Schaap
 
DP
,
Kusters
 
M
,
Handgraaf
 
HJM
,
van der Valk
 
MJM
 et al.  
Safety and effectiveness of SGM-101, a fluorescent antibody targeting carcinoembryonic antigen, for intraoperative detection of colorectal cancer: a dose-escalation pilot study
.
Lancet Gastroenterol Hepatol
 
2018
;
3
:
181
191

17

Harlaar
 
NJ
,
Koller
 
M
,
de Jongh
 
SJ
,
van Leeuwen
 
BL
,
Hemmer
 
PH
,
Kruijff
 
S
 et al.  
Molecular fluorescence-guided surgery of peritoneal carcinomatosis of colorectal origin: a single-centre feasibility study
.
Lancet Gastroenterol Hepatol
 
2016
;
1
:
283
290

18

de Gooyer
 
JM
,
Elekonawo
 
FMK
,
Bos
 
DL
,
van der Post
 
RS
,
Pèlegrin
 
A
,
Framery
 
B
 et al.  
Multimodal CEA-targeted image-guided colorectal cancer surgery using 111)In-labeled SGM-101
.
Clin Cancer Res
 
2020
;
26
:
5934
5942

19

Hekman
 
MC
,
Rijpkema
 
M
,
Muselaers
 
CH
,
Oosterwijk
 
E
,
Hulsbergen-Van de Kaa
 
CA
,
Boerman
 
OC
 et al.  
Tumor-targeted dual-modality imaging to improve intraoperative visualization of clear cell renal cell carcinoma: a first in man study
.
Theranostics
 
2018
;
8
:
2161
2170

20

van Dam
 
GM
,
Themelis
 
G
,
Crane
 
LMA
,
Harlaar
 
NJ
,
Pleijhuis
 
RG
,
Kelder
 
W
 et al.  
Intraoperative tumor-specific fluorescence imaging in ovarian cancer by folate receptor-α targeting: first in-human results
.
Nat Med
 
2011
;
17
:
1315
1319

21

de Gooyer
 
JM
,
Elekonawo
 
FMK
,
Bremers
 
AJA
,
Boerman
 
OC
,
Aarntzen
 
EHJG
,
de Reuver
 
PR
 et al.  
Multimodal CEA-targeted fluorescence and radioguided cytoreductive surgery for peritoneal metastases of colorectal origin
.
Nat Commun
 
2022
;
13
:
2621

22

Viera
 
C
.
Why is replication in research important?
 
American Journal of Experts
 
2023
. https://www.aje.com/arc/why-is-replication-in-research-important/ (accessed 19 March 2025)

23

Raphael
 
MP
,
Sheehan
 
PE
,
Vora
 
GJ
.
A controlled trial for reproducibility
.
Nature
 
2020
;
579
:
190
192

24

Fiji: an open-source platform for biological-image analysis. Nature Methods.

25

Schaap
 
DP
,
de Valk
 
KS
,
Deken
 
MM
,
Meijer
 
RPJ
,
Burggraaf
 
J
,
Vahrmeijer
 
AL
 et al.  
Carcinoembryonic antigen-specific, fluorescent image-guided cytoreductive surgery with hyperthermic intraperitoneal chemotherapy for metastatic colorectal cancer
.
Br J Surg
 
2020
;
107
:
334
337

26

Meijer
 
RPJ
,
de Valk
 
KS
,
Deken
 
MM
,
Boogerd
 
LSF
,
Hoogstins
 
CES
,
Bhairosingh
 
SS
 et al.  
Intraoperative detection of colorectal and pancreatic liver metastases using SGM-101, a fluorescent antibody targeting CEA
.
Eur J Surg Oncol
 
2021
;
47
:
667
673

27

Zhou
 
Q
,
van den Berg
 
NS
,
Kang
 
W
,
Pei
 
J
,
Nishio
 
N
,
van Keulen
 
S
 et al.  
Factors for differential outcome across cancers in clinical molecule-targeted fluorescence imaging
.
J Nucl Med
 
2022
;
63
:
1693
1700

29

Shi
 
NQ
,
Cui
 
X-Y
,
Zhou
 
C
,
Tang
 
N
,
Cui
 
D-X
.
Application of near-infrared fluorescence imaging in theranostics of gastrointestinal tumors
.
Gastroenterol Rep (Oxf)
 
2023
;
11
:
goad055

30

Wong
 
P
,
Li
 
L
,
Chea
 
J
,
Hu
 
W
,
Poku
 
E
,
Ebner
 
T
 et al.  
Antibody targeted PET imaging of 64Cu-DOTA-anti-CEA PEGylated lipid nanodiscs in CEA positive tumors
.
Bioconjug Chem
 
2020
;
31
:
743
753

31

van Dam
 
MA
,
Vuijk
 
FA
,
Stibbe
 
JA
,
Houvast
 
RD
,
Luelmo
 
SAC
,
Crobach
 
S
 et al.  
Overview and future perspectives on tumor-targeted positron emission tomography and fluorescence imaging of pancreatic cancer in the era of neoadjuvant therapy
.
Cancers (Basel)
 
2021
;
13
:
6088

32

Lütje
 
S
,
Rijpkema
 
M
,
Franssen
 
GM
,
Fracasso
 
G
,
Helfrich
 
W
,
Eek
 
A
 et al.  
Dual-modality image-guided surgery of prostate cancer with a radiolabeled fluorescent anti-PSMA monoclonal antibody
.
J Nucl Med
 
2014
;
55
:
995
1001

33

de Valk
 
KS
,
Deken
 
MM
,
Schaap
 
DP
,
Meijer
 
RP
,
Boogerd
 
LS
,
Hoogstins
 
CE
 et al.  
Dose-finding study of a CEA-targeting agent, SGM-101, for intraoperative fluorescence imaging of colorectal cancer
.
Ann Surg Oncol
 
2021
;
28
:
1832
1844

34

Quénet
 
F
,
Elias
 
D
,
Roca
 
L
,
Goéré
 
D
,
Ghouti
 
L
,
Pocard
 
M
 et al.  
Cytoreductive surgery plus hyperthermic intraperitoneal chemotherapy versus cytoreductive surgery alone for colorectal peritoneal metastases (PRODIGE 7): a multicentre, randomised, open-label, phase 3 trial
.
Lancet Oncol
 
2021
;
22
:
256
266

35

Kuijpers
 
AM
,
Aalbers
 
AGJ
,
Nienhuijs
 
SW
,
de Hingh
 
IHJT
,
Wiezer
 
MJ
,
van Ramshorst
 
B
 et al.  
Implementation of a standardized HIPEC protocol improves outcome for peritoneal malignancy
.
World J Surg
 
2015
;
39
:
453
460

36

Ammerata
 
G
,
Filippo
 
R
,
Laface
 
C
,
Memeo
 
R
,
Solaini
 
L
,
Cavaliere
 
D
 et al.  
Hyperthermic intraperitoneal chemotherapy and colorectal cancer: from physiology to surgery
.
World J Clin Cases
 
2022
;
10
:
10 852
10 861

37

de Boer
 
NL
,
Brandt-Kerkhof
 
ARM
,
Madsen
 
EVE
,
Doukas
 
M
,
Verhoef
 
C
,
Burger
 
JWA
.
The accuracy of the surgical peritoneal cancer index in patients with peritoneal metastases of colorectal cancer
.
Dig Surg
 
2021
;
38
:
205
211

This is an Open Access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0/), which permits unrestricted reuse, distribution, and reproduction in any medium, provided the original work is properly cited.

Supplementary data