Notes
Article history
The research reported in this issue of the journal was funded by the EME programme as project number 11/100/24. The contractual start date was in October 2013. The final report began editorial review in December 2015 and was accepted for publication in May 2016. The authors have been wholly responsible for all data collection, analysis and interpretation, and for writing up their work. The EME editors and production house have tried to ensure the accuracy of the authors’ report and would like to thank the reviewers for their constructive comments on the final report document. However, they do not accept liability for damages or losses arising from material published in this report.
Declared competing interests of authors
David Jayne is a member of the Medical Research Council/Efficacy and Mechanism Evaluation (EME) Strategy Group. This group has responsibility for setting EME programme funding policy. This position does not have an impact on the performance or results of this study. Nicholas West received grants outside this submitted work from Yorkshire Cancer Research, The Pathological Society of Great Britain and Ireland as well as the Academy of Medical Sciences in the last 3 years. Philip Quirke also received grants outside this submitted work from Yorkshire Cancer Research during the 36 months prior to submission of this report.
Permissions
Copyright statement
© Queen’s Printer and Controller of HMSO 2016. This work was produced by Andrew et al. under the terms of a commissioning contract issued by the Secretary of State for Health. This issue may be freely reproduced for the purposes of private research and study and extracts (or indeed, the full report) may be included in professional journals provided that suitable acknowledgement is made and the reproduction is not associated with any form of advertising. Applications for commercial reproduction should be addressed to: NIHR Journals Library, National Institute for Health Research, Evaluation, Trials and Studies Coordinating Centre, Alpha House, University of Southampton Science Park, Southampton SO16 7NS, UK.
Chapter 1 Background
Colorectal cancer
Colorectal cancer is the fourth most common cancer in the UK with an incidence of > 41,581 new cases per annum and almost two-thirds occurring in the colon;1,2 as such it represents a substantial burden on health-care resources. In comparison with rectal cancer, which has seen improvement in survival over the past decade, the survival from colon cancer has remained largely unchanged with 5-year overall survival of 58.7%. In terms of segmental distribution, the right colon is the most common site, harbouring ≈25% of colorectal cancers, followed by the sigmoid colon with ≈20%. 1
Curative resection involves segmental colectomy to resect the primary cancer and the draining lymphatic field. It has been shown that the standard of segmental colectomy as performed in the UK is of variable quality and that improvement in technique may help to improve outcomes with a survival advantage of up to 27% in patients with lymph node (LN) involvement. 2 It has also been suggested that there is a difference in the oncological quality of the resected specimen between surgeons who routinely perform complete mesocolic resection and those who do not. 3,4 If this is correct, then standardisation of surgical technique to complete mesocolic resection with D3 lymphadenectomy could make a substantial contribution to improving the prognosis of patients with colon cancer. 5–8
The corollary is that only ≈25% of patients have LN involvement, meaning that for the other 75% of patients D3 lymphadenopathy potentially represents overtreatment and exposes patients to unnecessary additional morbidity. Ideally, one would want to tailor the radicality of surgery to the disease biology, such that patients with LN disease had the option of radical lymphadenectomy to maximally eradicate cancer spread, while patients without LN disease could have limited resection of their cancer with similar oncological outcomes to radical surgery. Presently, however, it is not possible to determine pre- or intraoperatively which patients have LN disease and, therefore, as a default all patients undergo varying degrees of lymphadenectomy regardless of the stage of their disease.
There is an unmet clinical need to develop strategies that accurately predict LN disease prior to or at surgery and so allow colorectal cancer surgery to be tailored to the oncological needs of the patient.
Strategies to improve colon cancer surgery
Segmental colectomy is the current standard of surgery for colon cancer based on resection of the primary cancer along with the draining lymphatic field. In this way, the primary cancer is removed along with any regional LN metastases so as to minimise the chance of local tumour recurrence. Emerging evidence suggests that survival outcomes following colon cancer surgery can be improved by increasing the radicality of lymphadenectomy and respecting oncological planes of resection. 6,9 The technique of complete mesocolic resection with extended lymphadenectomy, as described by Hohenberger et al. ,5 has reported local recurrence rates of 3.6% and 5-year disease-free survival of 89.1%, which has been attributed to removal of more tissue and LNs in the correct surgical planes. These figures compare favourably to accepted local recurrence rates of 8–10% reported elsewhere. 10 Further, West et al. 2 showed a survival advantage from mesocolic plane surgery of 15% across all disease stages and up to 27% in patients with LN involvement. In the series reported by West et al.,2 only 32% of the specimens appeared optimal in terms of pathological grade of mesocolic completeness, suggesting substantial room for improvement in surgical technique. 8
Currently, the standard surgery for colon cancer in the UK differs from that described by Hohenberger5 in the extent of the lymphadenectomy performed. Standard surgery would generally involve a ‘D2 lymphadenectomy’, whereby the second tier of draining LNs are removed but the central high ligation required for ‘D3 lymphadenectomy’ is not routinely practised. Attempts to reproduce Hohenberger’s5 results have been encouraging with a retrospective study from Copenhagen, Denmark, reporting that complete mesocolic excision was a significant independent factor for improved disease-free survival using multivariate Cox regression. 6 This study compared 4-year disease-free survival between complete mesocolic excision and standard resection in patients with stage I–III disease and showed survival rates of 85.8% versus 75.9%, respectively. Recently, two systematic reviews have examined the evidence supporting complete mesocolic excision. 3,9 They agree that radical resection removes more tissue and produces better-quality specimens; however, they stress that the available literature has fundamental limitations that make it difficult to recommend widespread implementation of the technique outside of expert centres.
However, this ‘one-size-fits-all’ approach fails to take into account the biological variation of colon cancer or the fitness and expectations of the patient. Only 25% of cancers have metastatic disease to the LNs, suggesting that D3 lymphadenectomy is overtreatment in the majority. There is the added concern that the majority of colorectal cancer patients are elderly with significant comorbidity and that a universal policy of radical resection will lead to unnecessary morbidity. Concerns regarding an increased complication rate owing to the technical demands of complete mesocolic resection have been raised,11 although there is evidence in the literature to the contrary. 4,9,12 Other series, such as that from Hillerød Hospital, Denmark, have reported improved oncological resection without any increase in morbidity. 7 Another factor that needs to be taken into account when determining future surgical strategy is the changing pattern of disease presentation with the introduction of screening programmes. In the UK, the introduction of the National Bowel Cancer Screening Programme has seen a shift in incidence of early cancers (Dukes’ stage A) from 10.1% prior to screening to 45.3% following implementation. 13 As the incidence of LN metastases in Dukes’ A cancer is < 10%, a policy of D3 lymphadenectomy for all cannot be justified and it is unlikely to produce any survival benefit. Therefore, it seems sensible to adopt a more selective approach whereby patients with LN involvement are offered D3 lymphadenectomy, while those without nodal involvement undergo a more conventional D2 lymphadenectomy. Currently, there is no reliable method for determining LN status either pre- or intraoperatively. The decision to carry out D2 or D3 lymphadenectomy therefore remains at the discretion of the operating surgeon. It is only with an accurate and objective system for LN staging that the level of resection can be tailored to individual patients. The proposed research will evaluate the merits of 5-aminolevulinic acid (5-ALA) fluorescence for this purpose.
Preoperative lymph node staging
The difficulty in implementing a selective strategy for surgical resection is in accurately defining pre-operative LN status. No clear radiological definition of a malignant LN is agreed. A common definition is any node > 1 cm or a cluster of three or more nodes < 1 cm. Some studies have used a cut-off size of 1.5 cm or have used contrast enhancement to distinguish positive nodes. 14 The presence of micrometastases within normal sized LNs and benign enlargement of nodes due to inflammation are known to contribute to inaccuracies of size-based criteria. In a prospective audit of 84 patients with colon cancer undergoing pre-operative multidetector computerised tomography (CT) scanning, the accuracy, sensitivity and specificity for detection of LN disease was 58% [95% confidence interval (CI) 48% to 68%], 64% (95% CI 48% to 77%) and 53% (95% CI 39% to 67%), respectively, with poor interobserver agreement for node status. 15 Further, when nodes were assessed according to tumour node metastasis classification by separating into N0, N1 and N2 disease, the accuracy, sensitivity and specificity fell to 50% for all values. A meta-analysis of 19 studies has reported an overall sensitivity and specificity for CT-detected malignant LNs to be 70% (95% CI 59% to 80%) and 78% (95% CI 66% to 86%), respectively, with a diagnostic odds ratio of 8.1 (95% CI 4.7 to 14.1). 14 Attempts to improve LN staging by combining functional imaging, such as positron emission tomography, with CT or using functional diffusion weighted magnetic resonance imaging, have shown some promise but are not universally available and, therefore, as yet have limited application.
The Japanese Society for Cancer of the Colon and Rectum has circumvented this dilemma to some extent by recommending surgical resection based on primary tumour T stage, with D3 lymphadenectomy recommended for T3/4 disease. 16 A similar strategy was adopted in the National Cancer Research Institute the Fluoropyrimidine, Oxaliplatin and Targeted-Receptor pre-Operative Therapy for patients with high-risk, operable colon cancer (FOxTROT) study, with T3/4 cancers randomised to either surgery or pre-operative chemotherapy with our without antiepidermal growth factor receptor monoclonal antibody therapy. 17 Although still not perfect, CT imaging is more accurate in determining T stage than it is for nodal status and within the context of FOxTROT has been shown to accurately select patients for pre-operative chemotherapy. 18 The accuracy, sensitivity and specificity of CT scans in selecting patients with ‘bad’ T3/4-stage cancers was reported to be 74% (95% CI 64% to 82%), 78% (95% CI 65% to 87%) and 67% (95% CI 49% to 81%), respectively. 15 Although this, to some extent, gets around the problem of inaccurate pre-operative staging of local disease, there is no absolute correlation between ‘bad’ T stage and LN status; approximately 50% of T4 cancers will not have metastatic disease to the LNs and in ‘good’ T-stage patients, approximately 30% will have node-positive disease. 15
Strategies for intraoperative lymph node staging
Sentinel lymph node mapping
The ideal solution to the problem of accurate pre-operative LN staging is to develop a reliable means of intraoperative staging, such as sentinel lymph node (SLN) biopsy in breast cancer surgery. This would enable a real-time assessment of LN status. The concept of intraoperative LN staging is not new but has received much interest with the introduction of SLN mapping techniques. The SLN was defined by Morton et al. 19 as the first regional LN encountered by metastasising tumour cells in a study using SLN mapping in melanoma. It was proposed that if the SLN was free of tumour, then it could be assumed that the remaining LNs would also be tumour free and, hence, unnecessarily extensive resections could be avoided in patients with early-stage disease. Early studies of SLN mapping were performed by injecting a dye, such as isosulphan blue or patent blue-V into the primary lesion,19,20 which was taken up into the lymphatics so that the LNs could be visualised at operation. Subsequently, lymphoscintigraphy was added to blue dye, whereby a locally injected radionucleotide was detected intraoperatively using a gamma probe, enabling higher detection rates. 21,22 This concept has proven validity in some cancers, most notably breast cancer, for which the absence of an involved sentinel node spares the patient more radical axillary clearance and associated morbidity. The Association of Breast Surgery Guidelines23 now recommend SLN biopsy for the majority of patients with early invasive breast cancer. SLN mapping was first reported in colorectal cancer in 1999 with blue dye injected around the tumour prior to resection and histological examination of the LNs for cytokeratin expression. The results from early series were not encouraging with SLN detection rates of 58–98%, sensitivity rates of 40–100% and false-negative rates between 0% and 60%. 24 Further studies failed to improve a great deal on these results with large variations in the reported detection rates, sensitivity and false-negative rates, which have been variously attributed to heterogeneity in detection techniques, definition of the SLN, time interval between injection and SLN detection, histopathological techniques, and patient characteristics, including tumour stage and body mass index. 25 A recent meta-analysis showed a SLN identification rate of 92% with a pooled sensitivity rate for detecting LN metastases of 69.6% (range 33.3–100%) and a false-negative rate of 30.4%. 26
Importantly, concerns have been expressed regarding the high false-negative rates and whether metastases from colon cancer follow an orderly spread through tiers of LNs, or rather metastasise as skip lesions. The variability in anatomical site of the first metastatic LN was highlighted by Tan et al. ,27 who found that in 48% of cases the first metastatic LN was not adjacent to the tumour or was 5 cm beyond the longitudinal tumour margin in 18% of cases. Further, Park et al. 28 reported that in 6% of caecal cancers LN metastases occurred along the right branch of the middle colic artery, which might not be included in a standard D2 segmental resection.
Currently, therefore, SLN mapping is not in routine use in colon cancer, although it may have a role in focused histological ultra-staging, whereby positive nodes are subjected to detailed step sectioning in order to increase the detection of micrometastatic disease. 29 The main disadvantage of SLN mapping techniques is that, although they identify the tumour-draining lymphatic basin, they do not discriminate LNs with or without metastatic disease. Therefore, SLN cannot be used for guiding the extent of surgical resection in colon cancer.
Photodynamic diagnosis with 5-aminolevulinic acid
A potential solution to intraoperative LN staging involves drugs used in photodynamic diagnosis and therapy. Probably the most studied drug in this respect is 5-ALA, which is a pro-drug, taken up into the mitochondria of cells, where it serves as a precursor of protoporphyrin IX (PpIX), which, in turn, is the direct precursor of haem. 30 PpIX is a fluorescent molecule which, when exposed to blue–violet light of excitation wavelength 405 nm, emits a characteristic red fluorescence at a wavelength of 630–700 nm.
The 5-ALA is preferentially taken up by cancer cells, which exhibit altered levels of transporter molecules and catalytic enzymes, and metabolised to PpIX. The tendency for cancer cells to accumulate PpIX is enhanced by exogenous administration of 5-ALA. When administered in high doses and irradiated with blue–violet light, 5-ALA is cytotoxic to cancer cells (photodynamic effect), while in lower doses blue–violet light causes the cancers to emit a pink–red fluorescence (photodiagnosis effect).
The reasons for the selective uptake of 5-ALA in tumour cells have not been fully elucidated. In normal cells, 5-ALA synthesis is regulated by a feedback control system that is driven by high intracellular concentrations of free haem. This system is overridden when exogenous 5-ALA is administered. An accumulation of PpIX in a tumour relative to normal cells has been attributed to alterations in the activity of the rate-limiting enzymes porphobilinogen deaminase and ferrochelatase. 30 The former is increased and the latter decreased in tumour cells. Alterations in enzymatic activity may also be due to reduced availability of intracellular Fe2+ as a result of rapid cell division. In addition, several biochemical and structural changes in tumour cells have been linked to their avidity for PpIX, including reduced pH, up-regulation of low-density lipoprotein receptors and stromal abnormalities including large interstitial space, leaky vasculature and large amounts of newly synthesised collagens and lipids. 31
Following systemic administration, 5-ALA is metabolised in the liver and excreted in the bile and urine. 30 Much of the 5-ALA is used by the liver to synthesise PpIX and the remaining 5-ALA circulates to other body sites where all other cell types (except those without mitochondria) convert it to PpIX. Intracellular PpIX returns to normal levels up to 48 hours post administration.
A substantial body of work has accumulated on the use of 5-ALA and its derivatives in the fluorescence detection of solid cancers. In humans, 5-ALA fluorescence has been extensively used as a diagnostic aid in transitional cell carcinoma of the bladder,32–35 in neurosurgery to guide malignant glioma resection,36–38 and in gynaecology to detect endometriosis,39,40 peritoneal metastases from ovarian cancer41 and cervical squamous intraepithelial lesions. 42 In urology, several studies have shown the superior detection of transitional cell carcinoma and carcinoma in situ of the bladder using photodynamic diagnosis compared with white-light cystoscopy. 32 Both 5-ALA and its hexyl-derivative, hexaminolaevulinate [(HEX) Hexvix®, GE Healthcare], have been evaluated in bladder cancer. Burger et al. 33 reported reduced residual tumour and increased recurrence-free survival in patients undergoing photodynamic diagnosis with 5-ALA or HEX than white light alone. Residual tumour was present in 33% of patients with white-light cystoscopy, 15% with 5-ALA and 9% with HEX. The 3-year recurrence-free survival was 67% with white light, 80% with 5-ALA and 82% with HEX and differences between 5-ALA and HEX were non-significant. Similar promising results were reported by Daniltchenko et al. 34 in 115 patients with superficial bladder cancer, but a larger randomised study of 300 patients failed to demonstrate a difference in tumour recurrence. 35 A 2013 systematic review32 showed strong evidence that using 5-ALA or HEX improves tumour detection rates and reduces the residual tumour rate. It also found that, although the majority of the randomised controlled studies demonstrated a reduced local recurrence rate, not all studies confirmed this finding and, therefore, the evidence for recurrence is less convincing.
In a study examining the clinical pharmacokinetics of 5-ALA in normal healthy volunteers and patients at high risk of recurrent bladder cancer, the peak plasma concentration of 5-ALA following oral administration was achieved at 0.83 ± 0.20 hours. Plasma 5-ALA concentrations declined with a terminal half-life of approximately 45 minutes. 43
In neurosurgery, a randomised controlled Phase III multicentre trial36 was performed to investigate the safety and efficacy of fluorescence-guided resection in malignant gliomas. The study involved oral administration of 5-ALA (20 mg/kg) 3 hours (range 2–4 hours) before induction of anaesthesia. A modified neurosurgical microscope was used that allowed switching between conventional white xenon illumination and blue–violet excitation light. Follow-up (median 35.4 months) was available for 139 fluorescence-guided resections and 131 resections carried out with conventional white-light microsurgery. It was shown that 5-ALA fluorescence-guided surgery improved rates of complete resections36 and progression-free survival. 37 It has also been demonstrated in a recent study that there was no difference in the number of adverse events between patients given 5-ALA and the controls. 37 However, a Cochrane review38 of image-guided surgery for brain tumours found that there was low-quality evidence that 5-ALA-guided surgery improves complete tumour resection and the effect on survival is unclear.
In colorectal cancer and its metastases, PpIX has been identified as the predominant endogenous fluorophore and even in the absence of exogenous 5-ALA, it can distinguish involved from uninvolved LNs with a sensitivity and specificity of 62% and 78%, respectively. 44 A total of 33 patients were included in this study, which did not examine intraoperative fluorescence, and the primary tumours and LNs were examined ex vivo. In a study using a murine model of colon cancer, 5-ALA-induced PpIX fluorescence successfully detected LN metastases, whereas benign LNs did not exhibit any apparent fluorescence. 45
In a pilot study involving a mixed group of 18 patients with inoperable adenocarcinomas or adenomas of the oesophagus, duodenum and colon, patients were administered 30–60 mg/kg of 5-ALA orally. 46 In colorectal cancer patients, flexible sigmoidoscopy was performed with serial biopsies. Peak fluorescence in the large bowel tumours was achieved within 6 hours, with good selectivity between tumour and normal mucosa at a ratio of 5 : 1. Similarly effective results were seen in oesophageal and duodenal lesions, but with a lower dose of 30 mg/kg of 5-ALA. In a more recent pilot study, 5-ALA was administered orally at a dose of 20 mg/kg around 4 hours prior to patients undergoing endoscopic resection of early gastric or colorectal tumours. 47 Fluorescence was observed in 6 out of 10 gastric lesions and in one out of the three colorectal lesions. The study noted that fluorescence was observed only in areas of malignant tissue. In this series, four of the patients developed derangement of their liver function tests (LFTs), all of which spontaneously normalised.
A 2013 study examined the ability of 5-ALA to detect LN metastases in freshly excised LNs after en bloc resection of colorectal tumours. 48 The fluorescent images were analysed using a spectral unmixing method to specifically detect PpIX fluorescence. The accuracy, sensitivity and specificity of this method to detect LN metastases were 87.4%, 88.3% and 92.0%, respectively.
An important issue with the clinical use of any photosensitising agent is the potential for side effects. This problem appears to be confined to systemic administration when high doses are used for photodynamic therapy as opposed to lower doses used in fluorescence diagnosis (FD). When used systemically, it is important to establish a safe dose of photosensitiser according to the interval drug to light time to avoid photosensitivity side effects. Normal precautions include keeping the patient out of direct sunlight for 24–48 hours following drug administration. Occasionally reported side effects following systemic administration include nausea, vomiting, tachycardia, hypotension, photosensitivity for up to 48 hours, and elevated liver enzymes. 30 No safety concerns or serious adverse reactions have been highlighted in the literature following the use of 5-ALA in humans.
Proposed strategy for 5-aminolevulinic acid intraoperative lymph node detection
We aimed to combine existing techniques in SLN mapping using colorimetric dyes to provide an overall lymphatic map with the tumour-specific properties of 5-ALA FD. To our knowledge, this approach had never before been tried. This combination was particularly attractive in the context of laparoscopic surgery with both agents visible with the Storz D-Light Laparoscopic System (KARL STORZ GmbH & Co. KG; Tuttlingen, Germany), which combines white-light (colorimetric dyes) and blue-light (5-ALA) modes together with enhanced stereoscopic magnification. We focused on cancers of the right and sigmoid colon and performed segmental colectomy with D3 lymphadenectomy when appropriate. Resection specimens were scrutinised using routine and enhanced histopathological methods to determine the sensitivity, specificity and positive and negative predictive values for 5-ALA FD as compared with histological analysis.
We chose to use 5-ALA systemically via oral administration as ranges for the optimal dose and time of pre-operative administration in humans have been previously determined. The supplier of 5-ALA for oral administration was Photonamic GmbH & Co. KG, Wedel, Germany.
Chapter 2 Trial objectives
Purpose
The purpose of this study was to evaluate and optimise the use of 5-ALA for intraoperative FD of metastatic LNs in colon cancer.
Primary objective
To optimise the dose of oral 5-ALA administration for intraoperative FD of metastatic LNs in colon cancer.
Secondary objectives
To establish a reliable and repeatable methodology for FD of LN metastasis by standardisation of:
-
pre-operative CT LN reporting
-
intraoperative fluorescence detection system
-
surgical technique for laparoscopic segmental colonic resection with D3 lymphadenectomy
-
histopathological examination of resected specimens.
Chapter 3 Methods
Trial design
The GLiSten study was a feasibility study to assess the practicality of using fluorescence-assisted surgery and the ability of 5-ALA to accurately detect involved LNs intraoperatively. The study was designed in two parts:
-
a preliminary developmental phase
-
a larger evaluation phase.
The sites participating in the developmental phase were St James’s University Hospital, Leeds, UK, and The Mater Misericordiae University Hospital, Dublin, Ireland.
Developmental phase
This would involve a small number of centres to optimise the use and dosage of 5-ALA for intraoperative LN staging in colon cancer and to determine if 5-ALA provided an accurate means for detecting LN metastasis. Two cohorts of 10 patients with positive LNs, as verified on post-operative histology, were treated with different doses of 5-ALA to determine the optimum dose. The end point for this phase was the ability of 5-ALA to detect positive nodes in at least 2 out of the 10 patients with involved LNs in a single cohort. This corresponded to the upper boundary of the 99% exact CI of the sensitivity being at least 60%, that is, a good indication of activity given that the technical protocols and techniques may not have been fully optimised. A 99% CI was chosen to reflect the additional uncertainty in 10 patients. In patients without obvious LN disease at surgery, fluorescence of the primary cancer was used as a positive control for the efficacy of 5-ALA FD in colon cancer. If the initial preliminary phase achieved this end point then the trial would proceed to the evaluation phase.
The most common dose of 5-ALA in the literature is 20 mg/kg and this was chosen as the starting dose given to patients within cohort 1. If 5-ALA produced good fluorescence at this dose, the second cohort was to be treated with a lower dose of 10 mg/kg to reduce the potential of 5-ALA-related side effects. If there was inadequate fluorescence in the first cohort then the dose of 5-ALA was to be increased to 30 mg/kg. The dosing schedule is illustrated below (Figure 1).
By assessing the first 20 patients with positive LNs with this regimen it was hoped that the dose of 5-ALA would be optimised.
For each cohort, particular attention was given to detecting and recording:
-
any side effects related to 5-ALA
-
intraoperative LN fluorescence
-
histopathological confirmation of LN status.
If 5-ALA had detected positive LNs in at least 2 out of 10 patients in either cohort then the optimal dose of 5-ALA would have been identified. At least a further 10 patients with confirmed LN involvement would then be recruited and treated with this optimal dose, with flexibility to include further patients to confirm the validity of the technique before proceeding to the evaluation phase.
Evaluation phase
In this phase, several centres would recruit patients with colon cancer to determine sensitivity, specificity and diagnostic accuracy of 5-ALA intraoperative LN staging compared with in-depth histopathology, creating a much larger cohort.
Participant recruitment
Any adult patient undergoing elective surgery for right or sigmoid colon cancer amenable for laparoscopic resection, including those with metastatic disease, was eligible for the trial. The trial attempted to enrich the study population to contain patients with LN disease using the FOxTROT radiology criteria for locally advanced disease to obtain as much information as possible on 5-ALA for LN metastases. This included patients with radiological stage more than or equal to radiological stage T3 disease with either three or more visible LNs, at least one node of ≥ 10 mm in size or one irregularly enhancing node. Because of the inaccuracy of CT staging, > 10 patients were recruited to each cohort to achieve the required 10 with positive LN disease (Figure 2).
Participant eligibility
The predefined inclusion and exclusion criteria for the trial are described below.
Inclusion criteria
-
Able to give informed consent and willing to follow trial protocol.
-
Aged > 18 years.
-
Patients with cancers of the right and sigmoid colon amenable to laparoscopic resection incorporating D3 lymphadenectomy, as agreed by multidisciplinary team discussion following histopathological confirmation of cancer diagnosis and radiological staging.
-
Patients with distant metastatic disease will be eligible, provided laparoscopic resection of the cancer is part of routine clinical care.
-
Fit for standard laparoscopic resection.
-
American Society of Anaesthesiologists (ASA) classification of ≤ 3.
-
Normal hepatic and renal function on most recent blood tests (to be within 30 days prior to surgery). For the purposes of the trial, normal hepatic and renal function were defined as:
-
total bilirubin within normal institutional limits
-
aspartate transaminase/alanine transaminase concentrations of < 2.5 × institutional upper limit of normal
-
glomerular filtration rate of ≥ 60 ml/minute/1.73m2 or creatinine concentration within 10% of upper value for normal institutional limits.
-
Exclusion criteria
-
Patients with cancers of the transverse and left colon, owing to difficulty in defining D3 lymphadenectomy in these anatomical locations.
-
Past history of hypersensitivity reactions to 5-ALA or colorimetric dye.
-
Acute or chronic porphyria or a family history of the same.
-
Patients with synchronous colonic or rectal cancer (but patients with synchronous benign polyps are eligible).
-
Patients with coexistent inflammatory bowel disease, such as Crohn’s disease, ulcerative colitis or active diverticulitis, which may influence the lymphatic uptake of 5-ALA.
-
Pregnant (positive pregnancy test) or breastfeeding.
-
Received an Investigational Medicinal Product (IMP) at any dose within 28 days before registration.
-
Poorly controlled or serious medical or psychiatric illness that, in the investigator’s opinion, was likely to interfere with participation and/or compliance in this clinical trial.
Potential risks
Participants for this study were selected on the basis that they had a diagnosis of colon cancer that required surgical resection and were suitable for segmental colectomy with D3 lymphadenectomy. There were the normal risks of surgical complications associated with general anaesthesia and surgical resection. There was a small risk of photosensitivity reactions related to 5-ALA administration. This included transient derangement of LFTs (expected to return to baseline at 24–72 hours post administration) and skin hypersensitivity reactions owing to exposure to ultraviolet light. Other mild and transient side effects associated with 5-ALA administration in the literature include nausea, vomiting, tachycardia and hypotension. When 5-ALA is administered in conjunction with surgical resection of malignant brain tumours, a potential side effect is brain oedema and, although this was felt to be an unlikely occurrence in our trial patients, this information was included in the trial patient information leaflet.
In response to these potential adverse effects, all patients were kept out of direct sunlight for at least the first 48 hours following surgery and monitored for changes in vital signs and liver function (which is part of the standard post-operative care). During the operation, patient’s eyes and skin were protected from the operating lights using standard methods such as sterile drapes and tape to keep their eyes closed.
Concurrent clinical trials
Patients were screened for participation in other clinical trials. Providing there was no conflict, then patients could be included in both GLiSten and other trials. Notable exceptions were patients recruited to FOxTROT in which pre-operative chemotherapy is given. This can downstage the disease and, therefore, may have reduced the probability of detecting LN metastases.
Duration of participant participation
Patients were followed up for the first 30 post-operative days and this completed their participation in the trial. After this point, patients continued with standard follow-up for their colon cancer as per their institution’s normal clinical practice.
Withdrawals or removal of patient criteria
The right of the patient to refuse consent without giving reason was respected and all patients were free to withdraw from the study at any time without giving reasons and without prejudicing any further treatment.
Intervention details
Pre-operative care
All participants underwent a pre-operative staging CT of the thorax, abdomen and pelvis as standard practice within 8 weeks of surgery. All other pre-operative assessment was as per routine cancer practice including colorimetric tattooing of the tumour at colonoscopy. Oral 5-ALA was prepared as described in Appendix 3 and administered to the patient 1–6 hours prior to surgery.
Perioperative care
Participants were initially assessed with blue-light laparoscopy to detect any fluorescence within the tumour and draining lymphatic field within the mesentery. Fluorescent LNs were marked with Ligaclips [Ethicon Endo-Surgery (Europe) GmbH; Norderstedt, Germany] to facilitate subsequent pathological identification. Patients then underwent a segmental colectomy with D3 lymphadenectomy, if appropriate. For cancers of the right colon, segmental colectomy with D3 lymphadenectomy aimed to include central dissection at the origin of the ileocolic, right colic (when present) and middle colic vessels, with high vascular ligation and division. For cancers of the sigmoid colon, segmental colectomy with D3 lymphadenectomy included high ligation and division of the inferior mesenteric vessels proximal to the origin of the left colic vessels. Specimen extraction, anastomosis and formation of stoma were left to the discretion of the operating surgeon.
The sites of any fluorescent nodes were marked at operation with small surgical clips to facilitate the pathologists’ assessment. Resection specimens were subjected to in-depth histopathology, as per the Royal College of Pathologists guidance49 as well as additional assessment of fluorescent LNs in the form of step sectioning. This included:
-
specimen photography and assessment of the completeness of mesocolic resection of the fresh and the fixed specimen
-
distance of tumour and bowel wall to vascular ligation and length of vessels
-
area of lymphadenectomy
-
level of lymphadenectomy based on vascular anatomy
-
histological analysis and mapping of all LNs.
Post-operative care
Participants received standard post-operative care with additional monitoring for side effects to 5-ALA, including measurement of liver and kidney function tests. Patients were reviewed at 30 days postoperatively, as per standard clinical practice. This marked the end of their participation within the trial but they continued to receive appropriate follow-up care as per guidelines. 50
5-aminolevulinic acid
5-aminolevulinic acid in the form of its hydrochloride, 5-ALA HCl, has been used in humans for FD and photodynamic therapy in a variety of cancers. It preferentially accumulates in cancer cells where it is metabolised to PpIX, a component of the haem pathway and an endogenous fluorophore. 22
5-aminolevulinic acid is safe for human administration and it is used topically in dermatological malignancies, intravesically to detect bladder cancers and orally in premalignant oesophageal lesions and malignant gliomas. 23 It has also been administered intradermally, intraperitoneally and intravenously in experimental studies without adverse effects. 5-ALA is available in various forms depending on the intended mode of application. 5-ALA is cleared by the liver and kidneys with a short half-life and return to baseline levels by 24 hours. Much smaller doses are required for FD – typically 20 mg/kg.
Investigational medicinal product supply, labelling, handling and investigational medicinal product preparation
5-aminolevulinic acid was supplied by photonamic GmbH & Co. KG with a trial-specific label in line with Directive 2001/20/EC and the Medicines for Human Use (Clinical Trials) Regulations 2004 (amended 2006). 5-ALA was prepared and handled in line with manufacturers’ recommendations.
5-aminolevulinic acid administration
5-aminolevulinic acid was supplied in a sterile powder format within individual vials. Each vial contained 1.5 g of 5-ALA and was reconstituted in the vial by adding 50 ml of tap water using a needle and syringe, giving a concentration of 30 mg/ml. This was done immediately prior to oral administration.
The patient’s weight was used to calculate the amount of 5-ALA to be given, which was checked and verified by the hospital pharmacy department prior to them dispensing the drug the day prior to surgery. On the morning of surgery, the drug was reconstituted as above by the clinical research fellow and witnessed by nursing staff who countersigned the prescription. The correct volume of the reconstituted 5-ALA was calculated and then given to the patient.
Concomitant medications
The only absolute contraindications to the use of 5-ALA are:
-
a previous history of hypersensitivity to photosensitising drugs
-
a previous history of acute or chronic porphyria
-
hepatic or renal dysfunction
-
pregnancy.
5-aminolevulinic acid should be used with caution:
-
with other medications known to have a photosensitising effect, for example tetracylines, sulphonamides, quinolones. Patients should not be exposed to any photosensitising agents up to 2 weeks after administration of 5 ALA
-
with other medications associated with acute porphyria, for example diclofenac, barbiturates, carbamazepine, phenytoin
-
with other medication associated with hepatic or renal dysfunction, for example non-steroidal anti-inflammatory drugs, angiotensin-converting enzyme inhibitors, loop diuretics, phenytoin.
Patient and public involvement
The study protocol was designed with patient and public input provided by the Leeds National Institute for Health Research Healthcare Technology Co-operative in Colorectal Therapies. The ability to identify metastatic LNs and, thus, tailor the radicality of surgery had been identified as an important unmet clinical need in Healthcare Technology Co-operative patient and public workshops. Specific input into the study protocol, in particular focusing on patient safety related to the interventional drug, was provided by Jean Gallagher, Richard Boards, David Wilkinson and Gillian Ivey. All four provided valuable feedback on the patient information leaflets and the plain English summary for the study. Jean Gallagher and David Wilkinson were also integral members of the Trial Management Group (TMG) with access to all data on trial progress. The TMG found the involvement of both Jean Gallagher and David Wilkinson particularly beneficial in commenting on patient recruitment issues and in providing solutions to help recruitment. Gillian Ivey and Richard Boards were major contributing members to the Trial Steering Committee (TSC) providing valuable insights during the temporary halt to the trial.
The decision to conclude the study after the developmental phase, in light of inadequate sensitivity and specificity of 5-ALA for LN detection meaning the end point was not achieved, was endorsed by the trial patient and public involvement (PPI) at the final TMG meeting.
Chapter 4 Data analysis
Primary objective
The primary objective for this study was to optimise the dose of oral 5-ALA administration for intraoperative FD of metastatic LNs in colon cancer.
The presence of any fluorescent LNs was documented at the time of surgery and marked with surgical clips to aid pathological identification. The pathological outcome (positive or negative for metastatic disease) for all LNs was documented. Patients were considered as node positive if metastatic disease was detected in at least one LN on histopathological examination. Similarly, if no metastatic disease was found in any of the LNs on standard histopathological examination, then the patient was considered node negative.
Assessment of diagnostic accuracy was performed on a patient level rather than node level for disease and fluorescence LN status. The sensitivity and specificity of 5-ALA at different doses within the two cohorts was calculated based on whether or not 5-ALA fluorescence detected the pathological outcome of a LN containing metastatic disease. The diagnostic test result was categorised as positive (‘fluorescent LNs’) if at least one of the positive nodes fluoresced.
The ability of 5-ALA to detect positive nodes in at least 2 out of 10 patients with involved LNs in either cohort was required to progress to the evaluation phase. A 99% CI was used to reflect the additional uncertainty in a relatively small number of patients.
Secondary objectives
Pre-operative staging was compared with the histopathological findings of the resected specimen to determine the sensitivity and specificity of pre-operative CT reporting. Histopathological analysis of the extent of lymphadenectomy, the plane and completeness of mesocolic resection and the resection margin clearance were used to demonstrate the oncological quality of the surgical specimen.
Any potential side effects secondary to 5-ALA were recorded together with conversion rates and any intraoperative or post-operative complications up to 30 days post surgery. Conversion refers to the changeover from the intended laparoscopic surgery to an open laparotomy, which can occur for a variety of reasons.
Chapter 5 Results
Trial recruitment
A total of 44 patients were recruited to the trial with no participants withdrawing during the study period. A total of 37 patients were recruited at St James’s University Hospital, Leeds, UK, and seven were recruited from The Mater Misericordiae University Hospital, Dublin, Ireland. Cohort 1 patients were recruited between October 2013 and October 2014 and received an oral dose of 20 mg/kg of 5-ALA. Eighteen patients were required to achieve the target of 10 patients with metastatic LN disease as detected on standard histopathology. Cohort 2 patients were recruited between October 2014 and June 2015 and received an oral dose of 30 mg/kg of 5-ALA, in line with the dose escalation defined in the trial protocol. Twenty-six patients were treated within cohort 2 and nine of these patients had node-positive disease on standard histopathology.
Of the 44 trial patients, four did not undergo blue-light laparoscopy with subsequent histopathological examination. Two patients (one in each cohort) received their dose of 5-ALA and the operation was then cancelled. Another patient received the trial drug but did not have blue-light laparoscopy, as one of the components of the Storz D-light system had not returned from the sterilisation facility. The fourth patient received the trial drug and underwent blue-light laparoscopy, which demonstrated fluorescence of the tumour but no LN fluorescence. However, the tumour was determined to be unresectable and no histopathological examination of the tumour was possible. Therefore, 41 patients underwent blue-light laparoscopy, but 40 patients in the trial (17 in cohort 1 and 23 in cohort 2) underwent an operation with the aid of fluorescence detection and histopathological examination of their specimen was performed.
Patient demographics
Twenty-six male and 18 female patients participated in the trial (Table 1), with a mean age of 71 years (range 52–88 years). The mean body mass index was 27.3 kg/m2 (range 19.1–37.8 kg/m2), with a median ASA grade of 2. The ratio of right-sided to left-sided cancers was 30 : 14. There were six (14.3%) conversions among the 42 patients who underwent surgery. The majority of patients had pT3, pN0/1 disease. Three patients had metastatic disease involving either the liver or lungs: one in cohort 1 and two in cohort 2. There were no significant differences in the baseline characteristics between the two cohorts.
Demographics | Cohort 1 (n = 18) | Cohort 2 (n = 26) |
---|---|---|
Sex, male : female | 10 : 8 | 16 : 10 |
Age (years), mean (range) | 69.3 (52–85) | 71.8 (53–88) |
BMI (kg/m2), mean (range) | 26.1 (21–35) | 28 (19–38) |
ASA grade | ||
I | 9 | 5 |
II | 8 | 13 |
III | 1 | 8 |
Tumour site, right : left colon | 12 : 6 | 18 : 8 |
Conversion, yes | 4 | 2 |
5-ALA-related complication, yes | 0 | 8 |
Primary objective: diagnostic accuracy of 5-aminolevulinic acid
Of the 41 patients who underwent blue-light laparoscopy, 14 had fluorescent primary tumours (34.1%) and seven of these 14 patients also had fluorescent LNs (Table 2). None of the patients had fluorescent LNs without having fluorescence of the primary tumour in either cohort.
Observed fluorescence | Cohort 1 (n = 17) | Cohort 2 (n = 24) | Total |
---|---|---|---|
Patients with fluorescent primary tumour | 6 | 8 | 14 |
Patients with ≥ 1 fluorescent LN | 3 | 4 | 7 |
Cohort 1
In the first cohort, 18 patients were treated with 20 mg/kg of 5-ALA but only 17 were operated on with blue-light laparoscopy, as one patient’s operation was cancelled. Six out of the 17 patients had a fluorescent primary tumour (35.3%) and three of these patients had fluorescent LNs (see Table 2).
One out of the three patients with fluorescent LNs had confirmed metastatic nodal disease within the fluorescent node on standard histopathology. Therefore, there was only one confirmed case of a fluorescent positive node out of 10 patients with positive LN disease. The success detection criteria (the ability of 5-ALA to detect positive LNs in at least 2 out of 10 node-positive patients) for cohort 1 was therefore not met.
In cohort 1, 5-ALA has a sensitivity of 11.1% and specificity of 75% for determining the pathological outcome of LN assessment using fluorescence (Table 3).
Cohort 1 | Node positive on histology | Node negative on histology | Total |
---|---|---|---|
Patients with fluorescent LNs | 1 | 2 | 3 |
Patients with non-fluorescent LNs | 8 | 6 | 14 |
Total | 9 | 8 | 17 |
Two other patients in cohort 1 had fluorescent nodes; however, none of these fluorescent nodes contain any metastatic disease on standard histopathological examination. One patient had no detectable metastatic disease in any of their LNs including the fluorescent node on standard histopathological examination. Therefore, this patient is counted within the fluorescent node-negative group. The third patient had metastatic disease present in 6 out of 23 nodes found within the pathological specimen; however, no metastatic disease was found within the fluorescent node on standard histopathological examination. This patient could occupy two groups in Table 3: the fluorescent node-negative group or the non-fluorescent node-positive group. As we are primarily interested in the outcome of the fluorescent nodes, this patient was counted in the fluorescent node-negative group as they are considered to be false positive for the purposes of statistical analysis. However, this does mean that the total number of node-positive patients in Table 3 will appear to be nine instead of 10.
In summary, in cohort 1, 18 patients were treated with a dose of 20 mg/kg of 5-ALA and only one-third of patients had a fluorescent primary tumour. A total of 10 out of the 18 patients were found to be node positive. Only 1 out of these 10 patients had correctly identified node-positive disease via fluorescence, that is, a positive diagnosis. Therefore, cohort 1 did not meet the predefined criteria for acceptable 5-ALA sensitivity (2 out of 10) and the dosage was increased to 30 mg/kg, as per the protocol.
Cohort 2
Twenty-six patients were recruited to cohort 2 and nine of these patients had confirmed nodal disease on standard histopathology. Only 24 patients underwent blue-light laparoscopy because one patient had their operation cancelled and the equipment was not available on the day of surgery for another. A third patient was found to have unresectable disease but blue-light laparoscopy was performed. The tumour exhibited fluorescence, however none of the LNs fluoresced in this patient; therefore, only 23 patientsunderwent blue-light laparoscopy and had histopathological examination of the tumour performed.
Eight out of the 24 patients who underwent blue-light laparoscopy had a primary fluorescent tumour (33.3%) and four of these cases also had fluorescent LNs. None of the patients with fluorescent LNs was found to contain metastatic disease on standard histopathological examination (Table 4).
Cohort 2 | Node positive on histology | Node negative on histology | Total |
---|---|---|---|
Patients with fluorescent LNs | 0 | 4 | 4 |
Patients with non-fluorescent LNs | 7 | 12 | 19 |
Total | 7 | 16 | 23 |
In cohort 2, 5-ALA has a sensitivity of 0% and specificity of 75% for determining the pathological outcome of LN assessment using fluorescence (see Table 4).
Out of the four patients in cohort 2 with fluorescent LNs, two had no evidence of metastatic disease in any of the LNs within the specimens and are, therefore, counted in the fluorescent node-negative group. The other two patients had non-fluorescent nodes containing metastatic disease but the fluorescent nodes were benign on standard histopathological examination; therefore, similarly to the patient in cohort 1, they have been counted in the fluorescent node-negative group instead of the non-fluorescent node-positive group. This means that the total number of patients with node-positive disease in cohort 2 will appear to be seven instead of nine, according to Table 4.
In summary, in cohort 2, 26 patients were treated with a dose of 30 mg/kg of 5-ALA. Nine out of the 26 patients were found to be node positive. None of the nine patients had correctly identified node-positive disease via fluorescence, that is, a positive diagnosis. Recruitment to the trial ceased prior to reaching the target of 10 patients with node-positive disease in cohort 2; however, it is clear that the predefined criteria of at least two correct diagnoses in the 10 node-positive patients could not be reached as there had not been one correct diagnosis in nine node-positive patients.
Additional histopathological studies of all fluorescent benign LNs in both cohorts are continuing. Further staining of the LNs has not shown any evidence of metastatic disease but the results of immunohistochemistry are still awaited.
Summary
In conclusion, 7 out of 19 patients with node-positive disease exhibited LN fluorescence (36.8%). Neither dose of 5-ALA had adequate sensitivity to meet the defined end point and allow progression into the developmental phase of the trial.
Secondary objectives: pre-operative staging
Although 42 patients had surgery performed, one patient had unresectable disease and, therefore, comparison between pre-operative CT imaging and pathology staging is only possible in 41 patients, 19 of whom had nodal metastases.
In 23 out of the 41 patients, pre-operative staging of LN status correctly detected non-involved (N0) and involved (N1/2) nodes compared with histopathological findings, an accuracy of 56.1%. In 11 cases, the LN status was over staged and understaged in seven cases (Table 5).
LN stage on pathology | ||
---|---|---|
N1/2 | N0 | |
LN stage on CT | ||
N1/2 | 13 | 11 |
N0 | 7 | 10 |
The sensitivity and specificity for CT detection of LN stage in this study were 65% and 47.6%, respectively. This compares to reported sensitivity and specificity of 64–70% and 53–78% in the literature. 14,15
Pre-operative CT imaging successfully detected the presence of extramural vascular invasion (EMVI) in 14 out of 41 cases, showing an accuracy of EMVI detection across the two sites of 65.9% (Table 6). The sensitivity and specificity for CT detection of EMVI were 60.9% and 72.2%, respectively.
EMVI on pathology | ||
---|---|---|
Positive | Negative | |
EMVI on CT | ||
Present | 14 | 5 |
Absent | 9 | 13 |
Pre-operative CT accurately predicted whether or not the tumour had invaded beyond the muscularis propria in 33 out of 41 cases (Table 7). It is important to remember that for the purposes of this study, patients were selected on the basis of having suspected locally advanced disease on CT imaging.
T stage on pathology | |||||
---|---|---|---|---|---|
Pre-operative CT T stage | T0/1 | T2 | T3 | T4 | Total |
T2 | 1 | 0 | 0 | 1 | 2 |
T3 | 0 | 6 | 16 | 9 | 31 |
T4a | 0 | 1 | 2 | 4 | 7 |
T4b | 0 | 0 | 0 | 1 | 1 |
Total | 1 | 7 | 18 | 15 | 41 |
Secondary objectives: standardisation of surgical technique and histopathological examination
Quality of pathology specimens
The oncological quality of the resected specimen was judged on the plane of resection, which was reported in 40 pathology specimens. A total of 26 out of the 40 specimens were resected in the mesocolic plane (65%). There was a higher proportion of mesocolic resections in cohort 2 than cohort 1: 78.2% versus 47.1%, respectively (Table 8). These figures should be interpreted in light of the enriched cohort of locally advanced colon cancers. Although the 65% for complete mesocolic excision is lower than reported elsewhere in the literature, the advanced nature of the cancers in this study will have directly impacted on the ability to perform complete mesocolic plane surgery.
Resection plane | Cohort 1 (n = 17) | Cohort 2 (n = 24) |
---|---|---|
Mesocolic | 8 | 18 |
Intramesocolic | 8 | 5 |
Muscularis propria | 1 | 0 |
Missing data | 0 | 1 |
Resection margins
A total of 10 cases have been reported as having positive microscopic resection margins (R1), resulting in a positive resection margin rate of 23.8%, which is below the rate deemed to be excessive in the protocol (> 30%). Although this is higher than the R1 resection rate normally reported in colon cancer surgery, it probably reflects the enrichment of the cohorts with advanced stage disease and that some pathologists classify T4 disease with serosal involvement as a positive resection margin and would not be a cause for concern. There were no R2 resections in either cohort.
Complications
The overall anastomotic leak rate was 4.5%, which is below the rate deemed to be excessive in the trial protocol, but in keeping with the literature for this type of surgery. In the first cohort, four laparoscopic cases were converted to open laparotomies, resulting in a conversion rate of 22%, which is below the rate deemed to be excessive in the protocol (> 50%). There were two conversions in the second cohort giving a conversion rate of 8.3%. Although a conversion rate of 22% in cohort 1 would ordinarily be viewed as high, this cohort was enriched with advanced cancers and within this context a 22% conversion rate is probably acceptable.
There have been three intraoperative complications: one visceral perforation and two cases of intraoperative haemorrhage. In the first case, there was a perforation to the greater curve of the stomach following introduction of the Veress needle – this was repaired laparoscopically and the patient made an uneventful recovery. In the first case of intraoperative haemorrhage, there was bleeding from the inferior vena cava, which was clipped laparoscopically and no conversion was necessary; the patient made an uneventful recovery. In the second case of intraoperative haemorrhage, blood was seen in the abdominal cavity during closure of the laparoscopic port sites. The surgeon converted to an open procedure and found that the source of bleeding was from a peripancreatic vessel. This was oversewn and haemostasis achieved. The patient was admitted to the high-dependency unit (HDU) postoperatively for observation and made a good recovery. All three of these intraoperative complications are recognised within the type of surgery performed.
Two other patients were managed for post-operative ileus in addition to a patient who had to be readmitted, all of whom had CT scans during the initial few days of their post-operative course. One of these patients was treated for a chest infection with intravenous antibiotics. The other patient with a post-operative ileus underwent CT and the scan did not show any pathology. However, the patient became seriously ill with septic shock in the intensive care unit (ICU). A decision was made to undertake exploratory laparotomy, despite the normal CT scan, and the patient was found to have a sigmoid volvulus causing ischaemic perforation of the bowel. A completion colectomy with end ileostomy was performed. The patient subsequently had a prolonged stay in the ICU, during which he sustained a left internal jugular vein thrombosis secondary to central line placement requiring treatment with low-molecular-weight heparin.
Thirty-day mortality
Unfortunately, one patient died within the first 30 days of surgery. This patient had significant comorbidities and was identified as a high-risk surgical candidate on pre-operative cardiopulmonary exercise testing. The risks were discussed with the patient and their family and the patient was keen to proceed with surgery, including participation within the GLiSten study. The patient developed a lower respiratory tract infection in the early post-operative period but responded well to conservative management. Unfortunately, the patient was found to be acutely peritonitic > 10 days post surgery and findings at relaparotomy were consistent with an anastomotic leak. The anastomosis was resected and a defunctioning stoma was created. Subsequently, the patient suffered a non-ST elevation myocardial infarction and went on to develop multiorgan failure, to which he succumbed. This case was reviewed by the Data Monitoring and Ethics Committee (DMEC) and TSC and 5-ALA was not felt to have been implicated in the patient’s development of complications and demise.
Readmissions
Three patients had to be readmitted to hospital shortly following their discharge from the surgical ward. The first patient developed a lower respiratory tract infection that required treatment with intravenous antibiotics. This is a well-recognised post-operative complication and was not thought to be related to 5-ALA. The patient responded well to antibiotic treatment. The second patient requiring readmission presented with abdominal pain and acute kidney injury. The patient had urea and electrolyte levels checked for the first 5 post-operative days and, apart from a slight increase in creatinine on day 4 which had normalised by day 5, no other abnormalities were detected. The kidney injury was believed to be secondary to concomitant medications, which can cause renal impairment, and not as a consequence of receiving 5-ALA. The patient’s renal function recovered with intravenous fluids and no other intervention was required. The third patient was managed for a post-operative ileus and a CT scan showed a small amount of intraperitoneal gas, with no demonstrable intra-abdominal collection. It is unclear whether or not this patient had a small anastomotic leak, but the patient was managed conservatively with oral antibiotics for 7 days and was readmitted to hospital for 2 days following their initial discharge.
Drug safety
There have been no drug-related serious adverse events (SAEs) but we did see some minor 5-ALA side effects in eight patients. There were two patients, both male in cohort 2, who developed mild self-limiting photosensitivity reactions that affected the head and neck region. Both of these patients were admitted to the HDU postoperatively for observation and it must be noted that HDU has a much brighter ambient environment than the normal surgical ward. In both cases, the reaction was noted within the first 48 hours following surgery and had resolved by the time of discharge (around post-operative day 5). It is proposed that the brighter ambient environment in the HDU is likely to be a contributing factor. No intervention was required in either patient other than a topical emollient cream for associated skin dryness.
Four patients were noted to have elevated LFTs in the immediate post-operative period. No intervention was required, apart from monitoring, and the LFTs normalised within 3 days post surgery. Two patients had a slight transient deterioration in their renal function but it is not clear whether this was related to the surgery or the 5-ALA. Either way, renal function returned to normal in both patients and without any intervention.
Chapter 6 Discussion
Recruitment to the trial was of a longer duration than originally anticipated. This was mainly due to a temporary halt to the trial from December 2013 to February 2014 as there had been concerns regarding the efficacy of the IMP. Initially, there was good fluorescence seen of the primary tumour but then in consecutive cases no fluorescence was apparent. This prompted a temporary suspension of the trial while the IMP and laparoscopic camera equipment were investigated. No fault was found with either product and the lack of fluorescence was felt to be due to tumour biology. There were no subsequent concerns during the remainder of the trial.
A 2014 study attempted to evaluate the use of 5-ALA FD of peritoneal metastases in patients with colon cancer. 51 A total of 12 patients were selected on the basis of suspected peritoneal disease on CT scans and, in eight patients (66.7%), fluorescence of peritoneal metastases was shown. Unfortunately, this study did not comment on fluorescence of the primary tumour, LNs or any other metastatic deposits that would allow comparison with our study findings. In another recent study, in 2015, the ability of 5-ALA to assist with endoscopic resection of early-stage gastric and colorectal tumours was evaluated. 47 Only three patients in the study had colorectal lesions, of which one showed fluorescence (33.3%). This proportion fits with the pattern seen in this study, accepting that the numbers studied are very small. There are no other studies evaluating the use of 5-ALA FD for patients with colonic cancer for comparison. However, as discussed previously, several other surgical specialties utilise 5-ALA fluorescence for the detection of solid tumours. A systematic review of the use of 5-ALA in non-muscle-invasive bladder cancer surgery found that the sensitivity of tumour detection ranged from 76% to 97% with a specificity of 56%32 and with good evidence that FD reduced the rate of residual disease.
A Cochrane review38 of image-guided surgery for brain tumours found that the evidence for the use of 5-ALA fluorescence-assisted surgery is possibly not as strong as expected. However, there was a significant improvement in complete resection of the tumour when using 5-ALA fluorescence compared with standard resection: 65% and 36%, respectively.
5-aminolevulinic acid appears to be more efficient in detecting urological and neurological malignancies than colorectal tumours, and other factors for the decreased sensitivity seen in our study must be considered. Timing and dose of 5-ALA administration were similar to other published studies with similar patient characteristics. Other possibilities include autofluorescence of the surrounding tissue rendering PpIX fluorescence of the tumour imperceptible and natural variability of fluorescence intensity depending on tumour biology. Another possibility is photobleaching of the tumour by white-light laparoscopy at the beginning of the procedure, diminishing the effect of fluorescence over time. Neither of these explanations is thought to have materially contributed to the low fluorescence rates (primary cancer and LNs) observed in our study.
In our series of 41 patients, 19 had nodal disease and, of these, seven exhibited intraoperative fluorescence of the LNs (36.8%). 5-ALA fluorescence studies in other malignancies have not focused on detection of LN metastases and, therefore, the only data for comparison are within animal studies of colorectal cancer.
In a small study using a murine model of colorectal cancer,45 10 mice were administered intraperitoneal 5-ALA. Of these, three had fluorescent mesocolic LNs present and all fluorescent LNs had metastatic disease present on histopathological examination. Unfortunately, the study did not perform histopathological examination of all LNs; therefore, sensitivity and specificity are not known. There has been a small study recently examining 5-ALA fluorescence and its ability to detect LNs metastases in freshly excised colonic tumour resections. 48 Fourteen patients, the majority of whom had stage III disease (n = 12), were included in the study and nine had node-positive disease. The study used a spectral unmixing method to remove the possibility of masking true PpIX fluorescence from collagen autofluorescence. The sensitivity and specificity for detecting LN metastatic disease in this study were 88.3% and 92%, respectively, which are much higher than our findings. The study also states that there were no fluorescent benign LNs; however, it must be stressed that fluorescence was tested ex vivo and further details of LN fluorescence, including any non-fluorescent LNs containing metastatic disease, were not provided.
Our results show that 5-ALA is not sensitive enough to detect LN metastasis and, therefore, cannot be recommended for use in intraoperative staging of colonic cancer. This is disappointing, but valuable knowledge has been gained because there is no other study that has used 5-ALA for intraoperative fluorescent diagnosis of colonic LN metastases.
Sensitivity and specificity of CT imaging to detect LN metastases preoperatively in the literature are 64–70% and 53–78%, respectively, bearing in mind that different studies use slightly different definitions of malignant LNs. In our study, CT imaging had a sensitivity of 68.4% and a specificity of 47.6%, which is comparable to published work in the literature14,15 (see Table 5). 5-ALA FD has not demonstrated an improved ability to detect LN metastases compared with pre-operative imaging; therefore, other strategies for improving the staging of LN involvement in colorectal cancer need to be investigated.
Patients with suspected locally advanced disease on CT imaging were selected in an attempt to enrich the study with patients who had node-positive disease. Thirty-three patients (80.5%) in the study were confirmed to have locally advanced disease on histopathological examination. As the literature suggests that around 75% of patients with ‘bad’ T-stage disease would have positive LNs, we would have expected to find approximately 30 patients with node-positive disease across the two cohorts. Instead, the total number of patients with LN disease was 19 (46.3%). None of our patients received pre-operative chemotherapy and so this lower rate of node-positive disease probably reflects inaccurate patient selection based on CT imaging.
Chapter 7 Conclusions
This is the first human clinical trial of a photosensitiser used for intraoperative fluorescence LN staging in colon cancer. There have been no safety concerns identified using 5-ALA or with regard to surgical procedure. All SAEs were recognised post-operative complications and not related to 5-ALA.
A distinct difference has been observed in the fluorescence of the primary colon cancers, with around 30% of cancers exhibiting fluorescence and 70% showing no fluorescence. This might indicate an underlying difference in the uptake and metabolism of 5-ALA, which could possibly be of prognostic benefit. Further work is planned to investigate the difference in fluorescence at the molecular level and to extend follow-up to include cancer recurrence and overall survival, which will be funded separately to the GLiSten study.
Another area of interest is the non-specific LN fluorescence (false-positive cases) seen in some patients. It is possible that this could be related to the host inflammatory/stromal response. Again, this will be investigated further.
5-aminolevulinic acid has been shown to have poor sensitivity for detecting LN metastases and cannot be recommended for intraoperative staging on the basis of this trial. However, the study design was acceptable to patients and it has demonstrated that fluorescence-assisted surgery using the Storz D-light system is feasible, which is important for the design of future studies using alternative fluorescent probes. Although our findings do not support the use of 5-ALA for intraoperative detection of malignant LNs, useful experience in fluorescence-assisted surgery has been gained and interesting areas for further research have been identified. Stratified surgery for colorectal cancer remains a ‘holy grail’ and will become increasingly important as the population ages and elderly and frail patients become less able to tolerate radical surgery. Ultimately, more sensitive and specific fluorescent probes are required if we are going to succeed in the challenge of providing personalised surgery for patients with colon cancer.
Recommendations for future research
An interesting finding of this trial is that only one-third of the primary tumours fluoresced and we feel that this warrants further investigation, as the existing literature would lead us to believe there should have been a higher rate of fluorescence. As discussed previously, the fluorescence from 5-ALA is due to its metabolite, PpIX. The most important element to ascertain is whether or not the 5-ALA and its metabolite were present within the tumour. We plan to perform an extraction procedure on each tumour specimen to determine if PpIX is present to see if this explains the low rate of fluorescence.
Another possibility is that the natural variability in tumour biology has had some effect on the fluorescence seen in the primary tumours. Other factors that could have had an impact are the effect of the inflammatory response incited by the tumour and potential underlying differences in cell metabolism. Further research investigating whether or not differences in tumour cell density, T-cell tumour infiltration and metabolic pathways could explain the low levels of fluorescence seen in the primary tumours is being conducted. This includes histological assessment of cellular and stromal components of the cancers with specific reference to immune profile and deoxyribonucleic acid gene array profiling.
The GLiSten study is the first human clinical trial using 5-ALA in laparoscopic colorectal cancer resection, and the majority of the work that has informed the existing literature on this area is based on laboratory animal work. It is difficult to know how representative the animal models were that demonstrated these significant fluorescence results. There is a need to develop better small-animal models for pre-clinical testing of molecular probes to facilitate translation to first-in-man translation.
The need to improve LN staging in colorectal cancer still exists and the use of fluorescence to achieve this aim is still a viable option. However, identification of better fluorescent probes for intraoperative visualisation of primary cancers and LN disease is required.
Nanomedicine is an exciting expanding area of research and there is particular interest in using nanoparticles in the imaging and treatment of tumours. Specific targeting of tumour tissue by nanoparticles is possible by combining them with tumour-specific cell surface receptor antibodies. Work on using antibodies to target fluorescent nanoparticles to create novel probes against colorectal cancer cell lines is currently ongoing at the University of Leeds (further information can be obtained from the corresponding author).
Acknowledgements
Contributions of authors
Miss Helen Andrew contributed by fulfilling duties of the clinical research fellow responsible for the trial, active participation in data collection analysis and interpretation of the data as well as contributing to the writing of the report with final approval of the manuscript.
Miss Gemma Gossedge contributed by fulfilling duties of the clinical research fellow responsible for the trial as well as final approval of the manuscript.
Mrs Julie Croft provided support throughout the running of the trial as well as final approval of the manuscript.
Mr Neil Corrigan contributed to the conception and design of the trial, provided support throughout the running of the trial and assisted with analysis and interpretation of the data as well as final approval of the manuscript.
Professor Julia M Brown contributed to the conception and design of the trial as well as having final approval of the manuscript.
Dr Nicholas West contributed to the study by active participation in data collection, pathological assessment of the patient specimens as well as having final approval of the manuscript.
Professor Philip Quirke contributed to the conception and design of the trial and pathological assessment of the patient specimens as well as having final approval of the manuscript.
Dr Damien Tolan contributed to the trial by assessing all pre-operative CT imaging and providing radiological support.
Professor Ronan Cahill contributed to the conception and design of the trial, along with active participation of patient recruitment, execution of the study as well as having final approval of the manuscript.
Professor David G Jayne contributed to the conception and design of the trial, along with active participation of patient recruitment, execution of the study, analysis and interpretation of the data as well as contributing to the writing of the report with final approval of the manuscript.
Trial personnel
A special thank you to the following people who made a substantial contribution to the study, but are not named in the authorship.
Dr Niall Mulligan: Pathologist at Mater Misericordiae University Hospital, Dublin, Ireland.
Professor Helen Fenlon: Radiologist at Mater Misericordiae University Hospital, Dublin, Ireland.
Mrs Aoife Kelly: Research Nurse at Mater Misericordiae University Hospital, Dublin, Ireland.
Mr Greg Taylor: Academic Clinical Lecturer in Surgery at University of Leeds, Leeds, UK.
Mrs Catherine Moriarty: Senior Research Nurse at St James’s University Hospital, Leeds, UK.
Mrs Catherine Lowe: Clinical Trials Co-ordinator, University of Leeds, Leeds, UK.
Mrs Emma Fulton: Clinical Trials Co-ordinator, University of Leeds, Leeds, UK.
Mrs Vicky Napp: Clinical Trials Research Unit Principal Investigator at University of Leeds, Leeds, UK.
Mr Glenn Webb: regulatory approvals co-ordinated by All Ireland Cooperative Oncology Research Group, Dublin, Ireland.
Dr Colin Watts: Higher Education Funding Council for England Clinical Senior Lecturer and Chairperson of the TSC.
Dr Anton Krige: Consultant in Intensive Care Medicine & Anaesthesia and member of the TSC at Lancashire Hospitals NHS Trust, Blackburn.
Mr Mike Bradburn: Senior Statistician and member of the TSC at University of Sheffield, Sheffield.
Mrs Gillian Ivey: PPI representative on the TSC.
Mr Richard Boards: PPI representative on the TSC.
Mrs Jean Gallagher: PPI representative on the TMG.
Mr David Wilkinson: PPI representative on the TMG.
Professor Chris Roberts: Professor of Biostatistics and Chairperson of the DMEC.
Dr John Hartley: Senior Lecturer in Surgery and member of the DMEC.
Mrs Christina Wong: Consultant Pharmacist and member of the DMEC.
Publications
Gossedge G, Lowe C, West N, Tolan D, Brown J, Quirke P, et al. Next generation intraoperative lymph node staging for stratified colon cancer surgery (GLiSten). Belgrade: special issue – Abstracts of the ESCP (European Society of Coloproctology) 8th Scientific and Annual Meeting; 25–27 September 2013. Colorectal Dis 2013;15(Suppl. 3):1–129.
Data sharing statement
Patient information was prospectively collected and recorded on specifically designed case report forms (CRFs). The data from the CRFs were transferred onto a secure database managed through the Clinical Trials Research Unit at the University of Leeds. The paper CRFs are stored securely at St James’s University Hospital. All trial documentation and data will be securely archived in accordance with the 1998 Data Protection Act and the principals of Good Clinical Practice and will be kept for a minimum of 15 years from the end of the trial as per the Sponsor’s policy. Clinical trial summary results have been posted in the EudraCT. All available anonymised data can be obtained by contacting the corresponding author.
Disclaimers
This report presents independent research. The views and opinions expressed by authors in this publication are those of the authors and do not necessarily reflect those of the NHS, the NIHR, the MRC, NETSCC, the EME programme or the Department of Health. If there are verbatim quotations included in this publication the views and opinions expressed by the interviewees are those of the interviewees and do not necessarily reflect those of the authors, those of the NHS, the NIHR, NETSCC, the EME programme or the Department of Health.
References
- Cancer Research UK . Bowel Cancer Incidence Statistics 2015. www.cancerresearchuk.org/cancer-info/cancerstats/types/bowel/incidence/ (accessed November 2015).
- West NP, Morris EJ, Rotimi O, Cairns A, Finan PJ, Quirke P. Pathology grading of colon cancer surgical resection and its association with survival: a retrospective observational study. Lancet Oncol 2008;9:857-65. http://dx.doi.org/10.1016/S1470-2045(08)70181-5.
- Kontovounisios C, Kinross J, Tan E, Brown G, Rasheed S, Tekkis P. Complete mesocolic excision in colorectal cancer: a systematic review. Colorectal Dis 2015;17:7-16. http://dx.doi.org/10.1111/codi.12793.
- West NP, Kobayashi H, Takahashi K, Perrakis A, Weber K, Hohenberger W, et al. Understanding optimal colonic cancer surgery: comparison of Japanese D3 resection and European complete mesocolic excision with central vascular ligation. J Clin Oncol 2012;30:1763-9. http://dx.doi.org/10.1200/JCO.2011.38.3992.
- Hohenberger W, Weber K, Matzel K, Papadopoulos T, Merkel S. Standardized surgery for colonic cancer: complete mesocolic excision and central ligation – technical notes and outcome. Colorectal Dis 2009;11:354-64. http://dx.doi.org/10.1111/j.1463-1318.2008.01735.x.
- Bertelsen CA, Neuenschwander AU, Jansen JE, Wilhelmsen M, Kirkegaard-Klitbo A, Tenma JR, et al. Disease-free survival after complete mesocolic excision compared with conventional colon cancer surgery: a retrospective, population-based study. Lancet Oncol 2015;16:161-8. http://dx.doi.org/10.1016/S1470-2045(14)71168-4.
- Bertelsen CA, Bols B, Ingeholm P, Jansen JE, Neuenschwander AU, Vilandt J. Can the quality of colonic surgery be improved by standardization of surgical technique with complete mesocolic excision?. Colorectal Dis 2011;13:1123-9. http://dx.doi.org/10.1111/j.1463-1318.2010.02474.x.
- West NP, Hohenberger W, Weber K, Perrakis A, Finan PJ, Quirke P. Complete mesocolic excision with central vascular ligation produces an oncologically superior specimen compared with standard surgery for carcinoma of the colon. J Clin Oncol 2010;28:272-8. http://dx.doi.org/10.1200/JCO.2009.24.1448.
- Killeen S, Mannion M, Devaney A, Winter DC. Complete mesocolic resection and extended lymphadenectomy for colon cancer: a systematic review. Colorectal Dis 2014;16:577-94. http://dx.doi.org/10.1111/codi.12616.
- Jayne DG, Thorpe HC, Copeland J, Quirke P, Brown JM, Guillou PJ. Five-year follow-up of the Medical Research Council CLASICC trial of laparoscopically assisted versus open surgery for colorectal cancer. Br J Surg 2010;97:1638-45. http://dx.doi.org/10.1002/bjs.7160.
- Rosenberg J, Fischer A, Haglind E. Scandinavian Surgical Outcomes Research Group. Current controversies in colorectal surgery: the way to resolve uncertainty and move forward. Colorectal Dis 2012;14:266-9. http://dx.doi.org/10.1111/j.1463-1318.2011.02896.x.
- Gao ZD, Ye YJ, Wang S, Yang XD, Yin MJ, Liang B, et al. [Complete mesocolic excision versus traditional radical resection in colon cancer. Zhonghua Wei Chang Wai Ke Za Zhi 2012;15:19-23.
- Ellul P, Fogden E, Simpson CL, Nickerson CL, McKaig BC, Swarbrick ET, et al. Downstaging of colorectal cancer by the National Bowel Cancer Screening programme in England: first round data from the first centre. Colorectal Dis 2010;12:420-2. http://dx.doi.org/10.1111/j.1463-1318.2009.02069.x.
- Dighe S, Purkayastha S, Swift I, Tekkis PP, Darzi A, A’Hern R, et al. Diagnostic precision of CT in local staging of colon cancers: a meta-analysis. Clin Radiol 2010;65:708-19. http://dx.doi.org/10.1016/j.crad.2010.01.024.
- Dighe S, Blake H, Koh MD, Swift I, Arnaout A, Temple L, et al. Accuracy of multidetector computed tomography in identifying poor prognostic factors in colonic cancer. Br J Surg 2010;97:1407-15. http://dx.doi.org/10.1002/bjs.7096.
- Okuno K. Surgical treatment for digestive cancer. Current issues – colon cancer. Dig Surg 2007;24:108-14. http://dx.doi.org/10.1159/000101897.
- University of Birmingham . FOxTROT: Fluoropyrimidine, Oxaliplatin &Amp; Targted Receptor Pre-Operative Therapy for Colon Cancer. UK Clinical Research Network: Portfoio Database n.d. www.birmingham.ac.uk/research/activity/mds/trials/bctu/trials/coloproctology/foxtrot/index.aspx (accessed 2015).
- Dighe S, Swift I, Magill L, Handley K, Gray R, Quirke P, et al. Accuracy of radiological staging in identifying high-risk colon cancer patients suitable for neoadjuvant chemotherapy: a multicentre experience. Colorectal Dis 2012;14:438-44. http://dx.doi.org/10.1111/j.1463-1318.2011.02638.x.
- Morton DL, Wen DR, Wong JH, Economou JS, Cagle LA, Storm FK, et al. Technical details of intraoperative lymphatic mapping for early stage melanoma. Arch Surg 1992;127:392-9. http://dx.doi.org/10.1001/archsurg.1992.01420040034005.
- Reintgen D, Cruse CW, Wells K, Berman C, Fenske N, Glass F, et al. The orderly progression of melanoma nodal metastases. Ann Surg 1994;220:759-67. http://dx.doi.org/10.1097/00000658-199412000-00009.
- Albertini JJ, Cruse CW, Rapaport D, Wells K, Ross M, DeConti R, et al. Intraoperative radio-lympho-scintigraphy improves sentinel lymph node identification for patients with melanoma. Ann Surg 1996;223:217-24. http://dx.doi.org/10.1097/00000658-199602000-00016.
- Pijpers R, Collet GJ, Meijer S, Hoekstra OS. The impact of dynamic lymphoscintigraphy and gamma probe guidance on sentinel node biopsy in melanoma. Eur J Nucl Med 1995;22:1238-41. http://dx.doi.org/10.1007/BF00801606.
- Association of Breast Surgery at Baso 2009 . Surgical guidelines for the management of breast cancer. Eur J Surg Oncol 2009;35:1-22. http://dx.doi.org/10.1016/j.ejso.2009.01.008.
- Mulsow J, Winter DC, O’Keane JC, O’Connell PR. Sentinel lymph node mapping in colorectal cancer. Br J Surg 2003;90:659-67. http://dx.doi.org/10.1002/bjs.4217.
- Oh SY, Kim DY, Kim YB, Suh KW. Clinical application of sentinel lymph node mapping in colon cancer: in vivo vs. ex vivo techniques. Ann Surg Treat Res 2014;87:118-22. http://dx.doi.org/10.4174/astr.2014.87.3.118.
- van der Zaag ES, Bouma WH, Tanis PJ, Ubbink DT, Bemelman WA, Buskens CJ. Systematic review of sentinel lymph node mapping procedure in colorectal cancer. Ann Surg Oncol 2012;19:3449-59. http://dx.doi.org/10.1245/s10434-012-2417-0.
- Tan KY, Kawamura YJ, Mizokami K, Sasaki J, Tsujinaka S, Maeda T, et al. Distribution of the first metastatic lymph node in colon cancer and its clinical significance. Colorectal Dis 2010;12:44-7. http://dx.doi.org/10.1111/j.1463-1318.2009.01924.x.
- Park IJ, Choi GS, Kang BM, Lim KH, Jun SH. Lymph node metastasis patterns in right-sided colon cancers: is segmental resection of these tumors oncologically safe?. Ann Surg Oncol 2009;16:1501-6. http://dx.doi.org/10.1245/s10434-009-0368-x.
- Saha S, Monson KM, Bilchik A, Beutler T, Dan AG, Schochet E, et al. Comparative analysis of nodal upstaging between colon and rectal cancers by sentinel lymph node mapping: a prospective trial. Dis Colon Rectum 2004;47:1767-72. http://dx.doi.org/10.1007/s10350-004-0661-5.
- Krammer B, Plaetzer K. ALA and its clinical impact, from bench to bedside. Photochem Photobiol Sci 2008;7:283-9. http://dx.doi.org/10.1039/B712847A.
- Kennedy JC, Pottier RH. Endogenous protoporphyrin IX, a clinically useful photosensitizer for photodynamic therapy. J Photochem Photobiol Biol 1992;14:275-92.
- Rink M, Babjuk M, Catto JW, Jichlinski P, Shariat SF, Stenzl A, et al. Hexyl aminolevulinate-guided fluorescence cystoscopy in the diagnosis and follow-up of patients with non-muscle-invasive bladder cancer: a critical review of the current literature. Eur Urol 2013;64:624-38. http://dx.doi.org/10.1016/j.eururo.2013.07.007.
- Burger M, Stief CG, Zaak D, Stenzl A, Wieland WF, Jocham D, et al. Hexaminolevulinate is equal to 5-aminolevulinic acid concerning residual tumor and recurrence rate following photodynamic diagnostic assisted transurethral resection of bladder tumors. Urology 2009;74:1282-6. http://dx.doi.org/10.1016/j.urology.2009.06.088.
- Daniltchenko DI, Riedl CR, Sachs MD, Koenig F, Daha KL, Pflueger H, et al. Long-term benefit of 5-aminolevulinic acid fluorescence assisted transurethral resection of superficial bladder cancer: 5-year results of a prospective randomized study. J Urol 2005;174:2129-33. http://dx.doi.org/10.1097/01.ju.0000181814.73466.14.
- Schumacher MC, Holmang S, Davidsson T, Friedrich B, Pedersen J, Wiklund NP. Transurethral resection of non-muscle-invasive bladder transitional cell cancers with or without 5-aminolevulinic Acid under visible and fluorescent light: results of a prospective, randomised, multicentre study. Eur Urol 2010;57:293-9. http://dx.doi.org/10.1016/j.eururo.2009.10.030.
- Stummer W, Pichlmeier U, Meinel T, Wiestler OD, Zanella F, Reulen HJ. ALA-Glioma Study Group . Fluorescence-guided surgery with 5-aminolevulinic acid for resection of malignant glioma: a randomised controlled multicentre phase III trial. Lancet Oncol 2006;7:392-401. http://dx.doi.org/10.1016/S1470-2045(06)70665-9.
- Stummer W, Tonn JC, Mehdorn HM, Nestler U, Franz K, Goetz C, et al. Counterbalancing risks and gains from extended resections in malignant glioma surgery: a supplemental analysis from the randomized 5-aminolevulinic acid glioma resection study. Clinical article. J Neurosurg 2011;114:613-23. http://dx.doi.org/10.3171/2010.3.JNS097.
- Barone DG, Lawrie TA, Hart MG. Image Guided Surgery for the Resection of Brain Tumours (Review) 2015. http://onlinelibrary.wiley.com/doi/10.1002/14651858.CD009685.pub2/abstract;jsessionid=493817DE73CD007861D8C95ABE1C22D0.f04t01 (accessed May 2016).
- Hillemanns P, Weingandt H, Stepp H, Baumgartner R, Xiang W, Korell M. Assessment of 5-aminolevulinic acid-induced porphyrin fluorescence in patients with peritoneal endometriosis. Am J Obstet Gynecol 2000;183:52-7.
- Malik E, Berg C, Meyhöfer-Malik A, Buchweitz O, Moubayed P, Diedrich K. Fluorescence diagnosis of endometriosis using 5-aminolevulinic acid. Surg Endosc 2000;14:452-5. http://dx.doi.org/10.1007/s004640000160.
- Löning M, Diddens H, Küpker W, Diedrich K, Hüttmann G. Laparoscopic fluorescence detection of ovarian carcinoma metastases using 5-aminolevulinic acid-induced protoporphyrin IX. Cancer 2004;100:1650-6. http://dx.doi.org/10.1002/cncr.20155.
- Collinet P, Delemer M, Jouve E, Regis C, Farine MO, Vinatier D, et al. Fluorescence diagnosis of cervical squamous intraepithelial lesions: A clinical feasibility study. Photodiagnosis Photodyn Ther 2007;4:112-16. http://dx.doi.org/10.1016/j.pdpdt.2006.10.003.
- Dalton JT, Yates CR, Yin D, Straughn A, Marcus SL, Golub AL, et al. Clinical pharmacokinetics of 5-aminolevulinic acid in healthy volunteers and patients at high risk for recurrent bladder cancer. J Pharmacol Exp Ther 2002;301:507-12. http://dx.doi.org/10.1124/jpet.301.2.507.
- Moesta KT, Ebert B, Handke T, Nolte D, Nowak C, Haensch WE, et al. Protoporphyrin IX occurs naturally in colorectal cancers and their metastases. Cancer Res 2001;61:991-9.
- Kato S, Kawamura J, Kawada K, Hasegawa S, Sakai Y. Fluorescence diagnosis of metastatic lymph nodes using 5-aminolevulinic acid (5-ALA) in a mouse model of colon cancer. J Surg Res 2012;176:430-6. http://dx.doi.org/10.1016/j.jss.2011.10.031.
- Regula J, MacRobert AJ, Gorchein A, Buonaccorsi GA, Thorpe SM, Spencer GM, et al. Photosensitisation and photodynamic therapy of oesophageal, duodenal, and colorectal tumours using 5 aminolaevulinic acid induced protoporphyrin IX – a pilot study. Gut 1995;36:67-75. http://dx.doi.org/10.1136/gut.36.1.67.
- Nakamura M, Nishikawa J, Hamabe K, Goto A, Nishimura J, Shibata H, et al. Preliminary study of photodynamic diagnosis using 5-aminolevulinic acid in gastric and colorectal tumors. World J Gastroenterol 2015;21:6706-12. http://dx.doi.org/10.3748/wjg.v21.i21.6706.
- Harada K, Harada Y, Beika M, Koizumi N, Inoue K, Murayama Y, et al. Detection of lymph node metastases in human colorectal cancer by using 5-aminolevulinic acid-induced protoporphyrin IX fluorescence with spectral unmixing. Int J Mol Sci 2013;14:23140-52. http://dx.doi.org/10.3390/ijms141123140.
- Loughrey MB, Quirke P, Shepherd N. Standards and Datasets for Reporting Cancers n.d. www.rcpath.org/resourceLibrary/dataset-for-colorectal-cancer-histopathology-reports–3rd-edition-.html (accessed July 2016).
- National Institute for Health and Care Excellence . Colorectal Cancer: Diagnosis And Management (2011 Updated 2014) NICE Guideline CG131 n.d. www.nice.org.uk/guidance/cg131/evidence/full-guideline-183509677 (accessed July 2016).
- Kondo Y, Murayama Y, Konishi H, Morimura R, Komatsu S, Shiozaki A, et al. Fluorescent detection of peritoneal metastasis in human colorectal cancer using 5-aminolevulinic acid. Int J Oncol 2014;45:41-6. http://dx.doi.org/10.3892/ijo.2014.2417.
- European Medicines Agency . EMEA/H/C/000744-IAIN/0008/G/–/Gliolan:/EPAR/–/Product/Information/March/2014 n.d. www.ema.europa.eu/ema/index.jsp?curl=pages/medicines/human/medicines/000744/human_med_000807.jsp&mid=WC0b01ac058001d124 (accessed November 2015).
Appendix 1 Literature review search strategies
Included in this appendix are the full electronic search strategies used to review the existing literature. All literature searches were conducted through the Ovid MEDLINE database and were last reviewed in November 2015.
Research question: has the National Bowel Cancer Screening Programme caused a downstaging effect in colorectal cancer? | ||
---|---|---|
Search string number | Search string (including any limits) | Total number of results found |
1 | national adj1 bowel adj1 cancer adj1 screening adj1 program$ | 53 |
2 | downstag$ | 1997 |
3 | 1 and 2 | 1 |
Research question: does complete mesocolic excision/resection affect patient outcomes in colorectal cancer surgery? | ||
---|---|---|
Search string number | Search string (including any limits) | Total number of results found |
1 | exp Colorectal Neoplasms | 163,580 |
2 | colo$ | 770,305 |
3 | rect$ | 168,199 |
4 | 2 or 3 | 889,325 |
5 | cancer | 1,114,158 |
6 | malig$ | 424,628 |
7 | adeno$ | 596,493 |
8 | carcin$ | 795,956 |
9 | 4 and 5 | 164,695 |
10 | 4 and 6 | 38,496 |
11 | 4 and 7 | 68,630 |
12 | 4 and 8 | 89,968 |
13 | 1 or 9 or 10 or 11 or 12 | 269,904 |
14 | complete adj1 mesocolic adj1 excision | 77 |
15 | complete adj1 mesocolic adj1 resection | 5 |
16 | 14 or 15 | 78 |
17 | exp Recurrence or exp neoplasm Recurrence, Local | 248,743 |
18 | recurrence | 361,374 |
19 | local adj1 recurrence | 102,965 |
20 | 17 or 18 or 19 | 361,374 |
21 | exp disease-free survival | 51,202 |
22 | disease adj1 free adj1 survival | 65,748 |
23 | 21 or 22 | 65,748 |
24 | 20 or 23 | 404,424 |
25 | 13 and 16 and 24 | 26 |
26 | limit 25 to English language | 22 |
Research question: does an extended D3 lymphadenectomy improve outcomes in patients with colorectal cancer? | ||
---|---|---|
Search string number | Search string (including any limits) | Total number of results found |
1 | exp Colorectal Neoplasms | 163,580 |
2 | colo$ | 770,305 |
3 | rect$ | 168,199 |
4 | 2 or 3 | 889,325 |
5 | cancer | 1,114,158 |
6 | malig$ | 424,628 |
7 | adeno$ | 596,493 |
8 | carcin$ | 795,956 |
9 | 4 and 5 | 164,695 |
10 | 4 and 6 | 38,496 |
11 | 4 and 7 | 68,630 |
12 | 4 and 8 | 89,968 |
13 | 1 or 9 or 10 or 11 or 12 | 269,904 |
14 | exp Recurrence or exp Neoplasm Recurrence, Local | 248,743 |
15 | recurrence | 361,374 |
16 | local adj1 recurrence | 102,965 |
17 | 14 or 15 or 16 | 361,374 |
18 | exp disease-free survival | 51,202 |
19 | disease adj1 free adj1 survival | 65,748 |
20 | exp Lymph Node Excision | 38,894 |
21 | lymphadenectomy | 12,229 |
22 | D3 adj3 lymphadenectomy | 95 |
23 | 20 or 21 or 22 | 43,257 |
24 | 18 or 19 | 65,748 |
23 | 17 or 24 | 404,424 |
26 | 13 and 23 and 25 | 831 |
27 | limit 26 to English language | 629 |
28 | 13 and 22 and 25 | 22 |
Research question: does complete mesocolic excision/resection increase the surgical complication rate? | ||
---|---|---|
Search string number | Search string (including any limits) | Total number of results found |
1 | exp Colorectal Neoplasms | 163,580 |
2 | colo$ | 770,305 |
3 | rect$ | 168,199 |
4 | 2 or 3 | 889,325 |
5 | cancer | 1,114,158 |
6 | malig$ | 424,628 |
7 | adeno$ | 596,493 |
8 | carcin$ | 795,956 |
9 | 4 and 5 | 164,695 |
10 | 4 and 6 | 38,496 |
11 | 4 and 7 | 68,630 |
12 | 4 and 8 | 89,968 |
13 | 1 or 9 or 10 or 11 or 12 | 269,904 |
14 | complete adj1 mesocolic adj1 excision | 77 |
15 | complete adj1 mesocolic adj1 resection | 5 |
16 | 14 or 15 | 78 |
17 | exp Intraoperative Complications or exp Postoperative Complications | 478,358 |
18 | complica$ | 1,161,022 |
19 | intraoper$ adj3 complica$ | 34,562 |
20 | postoper$ adj3 complica$ | 334,633 |
21 | 17 or 18 or 19 or 20 | 1,288,161 |
22 | 13 and 16 and 21 | 27 |
Research question: does extended D3 lymphadenectomy increase the surgical complication rate? | ||
---|---|---|
Search string number | Search string (including any limits) | Total number of results found |
1 | exp Colorectal Neoplasms | 163,580 |
2 | colo$ | 770,305 |
3 | rect$ | 168,199 |
4 | 2 or 3 | 889,325 |
5 | cancer | 1,114,158 |
6 | malig$ | 424,628 |
7 | adeno$ | 596,493 |
8 | carcin$ | 795,956 |
9 | 4 and 5 | 164,695 |
10 | 4 and 6 | 38,496 |
11 | 4 and 7 | 68,630 |
12 | 4 and 8 | 89,968 |
13 | 1 or 9 or 10 or 11 or 12 | 269,904 |
14 | exp Intraoperative Complications or exp Postoperative Complications | 478,358 |
15 | complica$ | 1,161,022 |
16 | intraoper$ adj3 complica$ | 34,562 |
17 | postoper$ adj3 complica$ | 334,633 |
18 | 14 or 15 or 16 or 17 | 1,288,161 |
19 | exp Lymph Node Excision | 38,894 |
20 | lymphadenectomy | 12,229 |
21 | D3 adj3 lymphadenectomy | 95 |
22 | 19 or 20 or 21 | 43,257 |
23 | 13 and 18 and 22 | 674 |
24 | limit 23 to English language | 491 |
Research question: how accurate is pre-operative staging in detecting LN metastases in colorectal cancer? | ||
---|---|---|
Search string number | Search string (including any limits) | Total number of results found |
1 | exp Colorectal Neoplasms | 163,580 |
2 | colo$ | 770,305 |
3 | rect$ | 168,199 |
4 | 2 or 3 | 889,325 |
5 | cancer | 1,114,158 |
6 | malig$ | 424,628 |
7 | adeno$ | 596,493 |
8 | carcin$ | 795,956 |
9 | 4 and 5 | 164,695 |
10 | 4 and 6 | 38,496 |
11 | 4 and 7 | 68,630 |
12 | 4 and 8 | 89,968 |
13 | 1 or 9 or 10 or 11 or 12 | 269,904 |
14 | lymph node metas$ | 32,188 |
15 | nod$ meta$ | 40,469 |
16 | exp neoplasm metastasis or exp lymphatic metastasis | 169,914 |
17 | lymp$ met$ | 77,565 |
18 | nod$ adj3 met$ | 52,525 |
19 | lym adj3 nod$ adj3 met$ | 43,104 |
20 | lym$ adj3 met$ | 101,830 |
21 | 14 or 15 or 16 or 17 or 18 or 19 or 20 | 200,829 |
22 | exp Neoplasm Staging | 136,750 |
23 | stag$ | 849,852 |
24 | preopera$ adj3 stag$ | 4254 |
23 | 22 or 23 or 24 | 849,852 |
26 | 13 and 21 and 25 | 8091 |
27 | accuracy | 227,249 |
28 | accur$ | 474,923 |
29 | detection$ | 611,530 |
30 | detec$ | 1,653,696 |
31 | 27 or 28 or 29 or 30 | 2,021,880 |
32 | 26 and 31 | 2811 |
33 | limit 32 to English language | 2335 |
Research question: is CT accurate in detecting LN metastases in colorectal cancer? | ||
---|---|---|
Search string number | Search string (including any limits) | Total number of results found |
1 | exp Colorectal Neoplasms | 163,580 |
2 | colo$ | 770,305 |
3 | rect$ | 168,199 |
4 | 2 or 3 | 889,325 |
5 | cancer | 1,114,158 |
6 | malig$ | 424,628 |
7 | adeno$ | 596,493 |
8 | carcin$ | 795,956 |
9 | 4 and 5 | 164,695 |
10 | 4 and 6 | 38,496 |
11 | 4 and 7 | 68,630 |
12 | 4 and 8 | 89,968 |
13 | 1 or 9 or 10 or 11 or 12 | 269,904 |
14 | lymph node metas$ | 32,188 |
15 | nod$ meta$ | 40,469 |
16 | exp neoplasm metastasis or exp lymphatic metastasis | 169,914 |
17 | lymp$ met$ | 77,565 |
18 | nod$ adj3 met$ | 52,525 |
19 | lym adj3 nod$ adj3 met$ | 43,104 |
20 | lym$ adj3 met$ | 101,830 |
21 | 14 or 15 or 16 or 17 or 18 or 19 or 20 | 200,829 |
22 | accuracy | 227,249 |
23 | accur$ | 474,923 |
24 | detection$ | 611,530 |
23 | detec$ | 1,653,696 |
26 | 22 or 23 or 24 or 25 | 2,021,880 |
27 | exp Tomography, X-Ray Computed | 334,528 |
28 | CT adj3 imag$ | 30,260 |
29 | CT adj3 stag$ | 2322 |
30 | 27 or 28 or 29 | 341,239 |
31 | 13 and 21 and 26 and 30 | 503 |
32 | limit 31 to English language | 399 |
Research question: can LN metastases be detected intraoperatively in colorectal cancer? | ||
---|---|---|
Search string number | Search string (including any limits) | Total number of results found |
1 | lymph node metas$ | 32,188 |
2 | nod$ meta$ | 40,469 |
3 | exp neoplasm metastasis or exp lymphatic metastasis | 169,914 |
4 | lymp$ met$ | 77,565 |
5 | nod$ adj3 met$ | 52,525 |
6 | lym adj3 nod$ adj3 met$ | 43,104 |
7 | lym$ adj3 met$ | 101,830 |
8 | 1 or 2 or 3 or 4 or 5 or 6 or 7 | 200,829 |
9 | exp Colorectal Neoplasms | 163,580 |
10 | colo$ | 770,305 |
11 | rect$ | 168,199 |
12 | 10 or 11 | 889,325 |
13 | cancer | 1,114,158 |
14 | malig$ | 424,628 |
15 | adeno$ | 596,493 |
16 | carcin$ | 795,956 |
17 | 12 and 13 | 164,695 |
18 | 12 and 14 | 38,496 |
19 | 12 and 15 | 68,630 |
20 | 12 and 16 | 89,968 |
21 | 9 or 17 or 18 or 19 or 20 | 269,904 |
22 | intraoperative staging | 119 |
23 | intraop$ adj3 stag$ | 411 |
24 | intraop$ | 130,252 |
25 | stag$ | 849,852 |
26 | detec$ | 1,653,696 |
27 | 24 and 25 | 9958 |
28 | 24 and 26 | 11,875 |
29 | intraop$ adj3 detec$ | 1471 |
30 | intraoperative detec$ | 419 |
31 | 22 or 23 or 27 or 28 or 29 or 30 | 20,186 |
32 | 8 and 21 and 31 | 431 |
33 | limit 32 to English language | 342 |
Research question: how accurate is SLN mapping in detecting LN metastases in colorectal cancer? | ||
---|---|---|
Search string number | Search string (including any limits) | Total number of results found |
1 | exp Colorectal Neoplasms | 163,580 |
2 | colo$ | 770,305 |
3 | rect$ | 168,199 |
4 | 2 or 3 | 889,325 |
5 | cancer | 1,114,158 |
6 | malig$ | 424,628 |
7 | adeno$ | 596,493 |
8 | carcin$ | 795,956 |
9 | 4 and 5 | 164,695 |
10 | 4 and 6 | 38,496 |
11 | 4 and 7 | 68,630 |
12 | 4 and 8 | 89,968 |
13 | 1 or 9 or 10 or 11 or 12 | 269,904 |
14 | exp Lymph Nodes or exp Sentinel Lymph Node Biopsy or exp Melanoma or exp Lymphatic Metastasis | 211,417 |
15 | sentinel lymph node | 10,299 |
16 | sentinel adj1 lymph | 10,584 |
17 | sentinel adj1 lymph adj1 node | 10,300 |
18 | sentinel adj3 mapping | 1155 |
19 | 14 or 15 or 16 or 17 or 18 | 211,876 |
20 | 13 and 19 | 14,765 |
21 | accuracy | 227,249 |
22 | accur$ | 474,923 |
23 | detection$ | 611,530 |
24 | detec$ | 1,653,696 |
23 | detection | 610,414 |
26 | accuracy$ | 227,321 |
27 | 21 or 22 or 23 or 24 or 25 or 26 | 2,021,880 |
28 | 20 and 27 | 3724 |
29 | limit 28 to English language | 3152 |
30 | exp Neoplasm Metastasis | 169,914 |
31 | exp Lymphatic Metastasis | 75,941 |
32 | lymphatic met$ | 76,590 |
33 | lymp$ met$ | 77,565 |
34 | lym$ adj3 met$ | 101,830 |
35 | lymph node | 123,910 |
36 | lymph node metas$ | 32,188 |
37 | lym$ adj3 nod$ adj3 met$ | 43,104 |
38 | 30 or 31 or 32 or 33 or 34 or 35 or 36 or 37 | 260,653 |
39 | 28 and 38 | 2960 |
40 | limit 39 to English language | 2441 |
Research question: can photodynamic diagnosis be used to detect LN metastases intraoperatively in colorectal cancer? | ||
---|---|---|
Search string number | Search string (including any limits) | Total number of results found |
1 | lymph node metas$ | 32,188 |
2 | nod$ meta$ | 40,469 |
3 | exp neoplasm metastasis or exp lymphatic metastasis | 169,914 |
4 | lymp$ met$ | 77,565 |
5 | nod$ adj3 met$ | 52,525 |
6 | lym adj3 nod$ adj3 met$ | 43,104 |
7 | lym$ adj3 met$ | 101,830 |
8 | 1 or 2 or 3 or 4 or 5 or 6 or 7 | 200,829 |
9 | exp Colorectal Neoplasms | 163,580 |
10 | colo$ | 770,305 |
11 | rect$ | 168,199 |
12 | 10 or 11 | 889,325 |
13 | cancer | 1,114,158 |
14 | malig$ | 424,628 |
15 | adeno$ | 596,493 |
16 | carcin$ | 795,956 |
17 | 12 and 13 | 164,695 |
18 | 12 and 14 | 38,496 |
19 | 12 and 15 | 68,630 |
20 | 12 and 16 | 89,968 |
21 | 9 or 17 or 18 or 19 or 20 | 269,904 |
22 | intraoperative staging | 119 |
23 | intraop$ adj3 stag$ | 411 |
24 | intraop$ | 130,252 |
25 | stag$ | 849,852 |
26 | detec$ | 1,653,696 |
27 | 24 and 25 | 9958 |
28 | 24 and 26 | 11875 |
29 | intraop$ adj3 detec$ | 1471 |
30 | intraoperative detec$ | 419 |
31 | 22 or 23 or 27 or 28 or 29 or 30 | 20,186 |
32 | 8 and 21 and 31 | 431 |
33 | photodynamic diagnosis | 294 |
34 | photody$ adj3 diagno$ | 395 |
35 | exp Photosensitizing Agents | 26,476 |
36 | 33 or 34 or 35 | 26,595 |
37 | 32 and 36 | 4 |
Research question: use of 5-ALA in photodynamic diagnosis of malignancy | ||
---|---|---|
Search string number | Search string (including any limits) | Total number of results found |
1 | exp Aminolevulinic acid | 4496 |
2 | aminolevulinic adj1 acid | 6780 |
3 | photodynamic diagnosis | 294 |
4 | photodyna$ adj3 diagnos$ | 395 |
5 | 1 or 2 | 6780 |
6 | 3 or 4 | 395 |
7 | 5 and 6 | 199 |
8 | exp Neoplasms | 2,805,544 |
9 | 7 and 8 | 179 |
10 | limit 9 to English language | 141 |
Research question: can 5-ALA detect LN metastases in colorectal cancer? | ||
---|---|---|
Search string number | Search string (including any limits) | Total number of results found |
1 | lymph node metas$ | 32,188 |
2 | nod$ meta$ | 40,469 |
3 | exp neoplasm metastasis or exp lymphatic metastasis | 169,914 |
4 | lymp$ met$ | 77,565 |
5 | nod$ adj3 met$ | 52,525 |
6 | lym adj3 nod$ adj3 met$ | 43,104 |
7 | lym$ adj3 met$ | 101,830 |
8 | 1 or 2 or 3 or 4 or 5 or 6 or 7 | 200,829 |
9 | exp Colorectal Neoplasms | 163,580 |
10 | colo$ | 770,305 |
11 | rect$ | 168,199 |
12 | 10 or 11 | 889,325 |
13 | cancer | 1,114,158 |
14 | malig$ | 424,628 |
15 | adeno$ | 596,493 |
16 | carcin$ | 795,956 |
17 | 12 and 13 | 164,695 |
18 | 12 and 14 | 38,496 |
19 | 12 and 15 | 68,630 |
20 | 12 and 16 | 89,968 |
21 | 9 or 17 or 18 or 19 or 20 | 269,904 |
22 | intraoperative staging | 119 |
23 | intraop$ adj3 stag$ | 411 |
24 | intraop$ | 130,252 |
25 | stag$ | 849,852 |
26 | detec$ | 1,653,696 |
27 | 24 and 25 | 9958 |
28 | 24 and 26 | 11,875 |
29 | intraop$ adj3 detec$ | 1471 |
30 | intraoperative detec$ | 419 |
31 | 22 or 23 or 27 or 28 or 29 or 30 | 20,186 |
32 | 8 and 21 and 31 | 431 |
33 | photodynamic diagnosis | 294 |
34 | photody$ adj3 diagno$ | 395 |
35 | exp Photosensitizing Agents | 26,476 |
36 | 33 or 34 or 35 | 26,595 |
37 | 32 and 36 | 4 |
38 | exp Aminolevulinic acid | 4496 |
39 | aminolevulinic acid | 6780 |
40 | nod$ adj3 met$ adj1 aminolevulinic adj1 acid | 0 |
41 | 38 or 39 or 40 | 6780 |
42 | 32 and 41 | 3 |
37 | 37 and 41 | 3 |
Appendix 2 Trial case report forms
Appendix 3 5-aminolevulinic acid prescription and guidance for reconstitution
Appendix 4 Summary of product characteristics
Information reproduced from European Medicines Agency: EMEA/H/C/000744-IAIN/0008/G – Gliolan: EPAR – Product Information March 2014. www.ema.europa.eu/ema/index.jsp?curl=pages/medicines/human/medicines/000744/human_med_000807.jsp&mid=WC0b01ac058001d12452 (accessed November 2015). © EMA [1995-2016].
© EMA [1995-2016]
Appendix 5 End-of-trial documentation
List of abbreviations
- 5-ALA
- 5-aminolevulinic acid
- ASA
- American Society of Anaesthesiologists
- CI
- confidence interval
- CRF
- case report form
- CT
- computerised tomography
- DMEC
- Data Monitoring and Ethics Committee
- EME
- Efficacy and Mechanism Evaluation
- EMVI
- extramural vascular invasion
- FD
- fluorescence diagnosis
- FOxTROT
- Fluoropyrimidine, Oxaliplatin and Targeted-Receptor pre-Operative Therapy for patients with high-risk, operable colon cancer
- GLiSten
- Next Generation intraoperative Lymph node staging for Stratified colon cancer surgery
- HDU
- high-dependency unit
- HEX
- hexaminolaevulinate
- ICU
- intensive care unit
- IMP
- Investigational Medicinal Product
- LFT
- liver function test
- LN
- lymph node
- PPI
- patient and public involvement
- PpIX
- protoporphyrin IX
- SAE
- serious adverse event
- SLN
- sentinel lymph node
- TMG
- Trial Management Group
- TSC
- Trial Steering Committee