Skip to main content
Current Oncology logoLink to Current Oncology
. 2012 Dec;19(6):e468–e477. doi: 10.3747/co.19.1167

Report from the 13th Annual Western Canadian Gastrointestinal Cancer Consensus Conference; Calgary, Alberta; September 8–10, 2011

MM Vickers *,, J Pasieka , E Dixon , S McEwan , A McKay §, D Renouf ||, D Schellenberg #, D Ruether *
PMCID: PMC3503677  PMID: 23300370

Abstract

The 13th annual Western Canadian Gastrointestinal Cancer Consensus Conference was held in Calgary, Alberta, September 8–10, 2011. Health care professionals involved in the care of patients with gastrointestinal cancers participated in presentation and discussion sessions for the purposes of developing the recommendations presented here. This consensus statement addresses current issues in the management neuroendocrine tumours and locally advanced pancreatic cancer.

Keywords: Neuroendocrine tumours, pancreatic cancer, systemic therapy, radiation

1. TERMS OF REFERENCE

1.1. Purpose

To develop the consensus opinion of oncologists and allied health professionals from across western Canada in an attempt to define best care practices and to improve care and outcomes for patients with gastrointestinal cancers.

1.2. Participants

Medical, radiation, and surgical oncologists and allied health professionals from western Canada involved in the care of patients with gastrointestinal malignancies.

1.3. Target audience

Health care professionals involved in the care of patients with gastrointestinal malignancies.

1.4. Basis of Recommendations

The recommendations reported here are based on presentation and discussion of the best available evidence (Table i). Where applicable, references are cited.

TABLE I.

Levels of evidence1

Level Meaning
i Evidence from one or more randomized controlled trials
ii-1 Evidence from one or more controlled trials without randomization
ii-2 Evidence from cohort or case–control analytic studies, preferably from more than one centre or research group
ii-3 Evidence from comparisons between times or places with or without the intervention; dramatic results in uncontrolled experiments could be included here
iii Opinions of respected authorities, based on clinical experience; descriptive studies or reports of expert committees

2. NEUROENDOCRINE TUMOURS

Question: What is the role of octreotide lar (long-acting release) in the treatment of asymptomatic advanced low- and intermediate-grade midgut neuroendocrine tumours (nets)?

  • The promid study2 showed a time-to-progression benefit for patients with metastatic low- and intermediate-grade midgut nets (level i evidence).

  • The data do not present a strong case for the immediate introduction of octreotide lar therapy in those patients (level iii evidence).

  • Patients who become symptomatic, demonstrate disease progression, or have a biochemical indication for therapy could be considered for octreotide lar therapy. Such patients should also be considered for multidisciplinary evaluation (level iii evidence).

Summary of Evidence: Neuroendocrine tumours are a group of rare neoplasms arising from neuroendocrine cells located throughout the body. The incidence of these tumours has increased significantly since the early 1970s3. More than 80% of nets express a high density of somatostatin receptors, of which 5 subtypes have been identified4.

The introduction of synthetic somatostatin analogs (ssas) in the 1980s revolutionized the treatment of patients with functional nets. These agents are effective in managing the symptoms associated with functional tumours in more than 50% of patients5 and are used worldwide today for their palliative benefit. They are generally well tolerated.

A subject of controversy has been the treatment of nonfunctioning nets with ssas. In vitro studies suggest that ssas may have an antiproliferative effect6,7, but nonrandomized clinical studies have produced disappointing results, with tumour shrinkage according to World Health Organization or the Response Evaluation Criteria in Solid Tumors seen in fewer than 5% of patients. Stabilization of tumour growth is described in up to 50% of patients whose disease was progressing before treatment with ssas8.

The promid study is the first randomized prospective trial with data supporting the possible antineoplastic effect of octreotide lar in a relatively homogeneous population of patients with midgut nets. The primary endpoint of the study, time to progression, was significantly longer (15.6 months) in the experimental arm of the trial than in the control (placebo) arm (5.9 months)2. Patients benefitted equally, regardless of whether their tumour was functional or nonfunctional. The benefit of treatment as defined by the study on other important clinical outcomes such as overall survival (os) is unknown.

Patients with advanced midgut nets can be relatively asymptomatic from their disease, particularly with nonfunctional tumours. Patients may remain well or minimally symptomatic for months to years. The promid data do not present sufficiently strong evidence to suggest that all patients with advanced midgut nets should be treated with octreotide lar.

Patients with midgut nets who become symptomatic, who have a biochemical indication for therapy, or who demonstrate disease progression should be considered for treatment with octreotide lar based on the growing body of evidence that supports the use of that agent for the palliation of disease-related symptoms and for the potential antiproliferative benefits of treatment in the setting of progressive disease.

Question: What is the role of inhibitors of the mammalian target of rapamycin (mtor) combined with octreotide in the treatment of advanced low- and intermediate-grade nets?

  • Treatment with everolimus and octreotide lar may be considered in patients with a history of symptoms consistent with carcinoid and documented progressive disease (level i evidence)9.

  • Correcting for informative censoring bias, a 5.5-month progression-free survival (pfs) benefit was seen (level i evidence)9.

Summary of Evidence: A serine/threonine kinase, mtor plays a central role in cellular growth, proliferation, and metabolism and in angiogenesis1012. Neuroendocrine tumours are linked to genetic alterations in inherited syndromes, including tuberous sclerosis, neurofibromatosis, and von Hippel–Lindau disease, in which upregulated mtor inhibition is associated with the development of islet cell and carcinoid tumours1315. Sporadic carcinoid tumours have been shown to co-express insulin-like growth factor 1 (igf-1), an upstream activator of the mtor pathway, and the igf receptor16. Octreotide downregulates igf-1 and reduces serum igf-1 levels in patients with solid tumours17, and everolimus inhibits mtor12, which suppresses the growth of nets16,18. Combination therapy with octreotide and mtor inhibitors is therefore a rational approach to the treatment of advanced nets that has been investigated in clinical trials with interesting results.

In 60 patients with low- to intermediate-grade advanced nets, Yao and colleagues demonstrated antitumour activity with the combination of octreotide lar (30 mg every 28 days) with two dose schedules of everolimus (5 mg and 10 mg daily). This phase ii study reported a response rate of 20% and disease stabilization in 70% of patients19. Combination therapy was associated with grade 3 and 4 toxicities in 10% of patients in the study.

The phase iii radiant 2 study20 randomized 429 patients with low- to intermediate-grade advanced nets and carcinoid syndrome to octreotide lar 30 mg every 28 days with either placebo or everolimus 10 mg daily. The heterogeneous study population included patients with small intestine, colon, lung, and other primary tumour sites. The primary outcome in this selected population of patients with documented radiographic progression of disease in the 12 months before commencement of therapy was pfs by central radiographic review. According to that review, pfs was prolonged by 5.1 months in patients treated with combination therapy. The trial failed to meet pre-specified statistical significance. Assessment of pfs by local investigators favoured the combination therapy arm with an improvement in pfs of 3.4 months, and a pre-specified statistical analysis of inverse probability of censoring weights supported a meaningful improvement in pfs. Treatment was associated with manageable toxicity and side effects consistent with those seen in other trials involving these agents.

In the phase iii radiant 3 study, which was designed to assess the efficacy of everolimus in patients with advanced pancreatic nets (pnets), concomitant use of octreotide was permitted as part of best supportive care. Of patients treated on this trial, 40% received treatment with octreotide, providing additional evidence that the combination of octreotide and everolimus can be administered safely with a tolerable side effect and toxicity profile9. Ongoing trials, including the radiant 4 study, will help to further elucidate the potential benefits of combining ssa therapy and mtor inhibition in selected patient populations with nets other than pnets.

At the present time, based on a sound preclinical rationale and data from phase ii and iii clinical trials, it is reasonable to consider treatment with a combination of everolimus and octreotide lar in net patients with a history of carcinoid symptoms and documented disease progression.

Question: What is the role of everolimus and sunitinib in the treatment of advanced pancreatic nets?

  • Both agents have demonstrated a pfs benefit compared with placebo in a randomized phase iii trial setting. The magnitude of the benefit is similar for the two agents (level i evidence)9,21.

  • The optimal sequencing of these agents in reference to other treatment modalities has not been established (level iii evidence).

Summary of Evidence: The study of pnets and carcinoid tumours in patients with tuberous sclerosis and neurofibromatosis is in part responsible for the current understanding of the importance of the mtor pathway in the pathogenesis of those malignancies14,15. The tuberous sclerosis complex (TSC1/2) and the neurofibromatosis (NF1) gene are regulators of the mtor pathway in normal neuroendocrine cells2225.

Neuroendocrine tumours are highly vascular tumours26 that overexpress vascular endothelial growth factor and its receptor. Autocrine activation of the vascular endothelial growth factor pathway may also potentiate tumour growth2729. Signalling pathways that express platelet-derived growth factor and its receptor, igf-1 and its receptor, and others have also been implicated in the pathogenesis of nets3034.

Patients with pnets often present with locally advanced (20%) or metastatic disease (60%)9,35. Median survival in patients with metastatic pnets is 24–28 months3,9,35, and 65% of patients with advanced disease will die within 5 years35.

Systemic treatment with streptozocin-based doublets results in response rates of 10%–45%36. Treatment is inconvenient for patients, often necessitating travel to specialized facilities familiar with the administration of these agents and with their associated side effects and toxicities, which can be considerable. Not all patients with advanced pnets are appropriate for treatment with streptozocin-based chemotherapy, and streptozocin is now available in Canada only through a special-access program. There remains a significant unmet need for effective, convenient, and tolerable systemic therapy for patients with advanced pnets.

Based on the growing understanding of the potential role of mtor and the vascular endothelial growth factor receptor in the pathogenesis of pnets, clinical trials designed to test agents targeting those pathways have been completed or are ongoing worldwide. In February 2011, the published results of two randomized phase iii trials in patients with advanced pnets raised hope for patients living with those cancers. The trials were conducted in a selected population of patients whose disease had progressed in the 12 months before study entry.

In a trial involving 410 patients, Yao and colleagues9 demonstrated a 6-month pfs improvement in patients treated with everolimus, an orally administered mtor inhibitor, compared with patients receiving placebo. Treatment with everolimus was well tolerated, and the side effects and toxicities reported were consistent with those reported in clinical trials of that agent in other cancers.

Raymond and colleagues21 reported the results of a phase iii trial of orally administered sunitinib compared with placebo in patients with advanced pnets. Because of a higher number of serious adverse events and deaths in the placebo arm of the study, the trial was discontinued after 171 of 340 patients had been accrued. The study demonstrated a clinically meaningful pfs prolongation of 6 months in patients treated with sunitinib.

Treatment with everolimus or sunitinib is appropriate for consideration in patients with advanced pnets whose disease is progressing. At the present time, the selection of one agent over the other will likely be determined by the patient’s comorbidities and the differing toxicity profiles of these drugs. The optimal sequencing of everolimus and sunitinib is not established and should be the subject of clinical trials.

Question: What is the role of resection of the primary tumour in the setting of advanced nets?

  • These patients should be referred to an experienced surgical team and reviewed in a multidisciplinary setting to define the most appropriate treatment options (level iii evidence).

  • Resection of the primary tumour may provide clinical benefit (level ii-2 evidence).

Summary of Evidence: Patients having small-bowel nets frequently present with metastatic mesenteric and liver disease. The primary is usually small and undetectable with standard imaging modalities. In the past, asymptomatic patients with advanced disease were not referred for surgical resection. However, many centres of excellence in nets have developed an aggressive surgical approach for these patients. Retrospective data have demonstrated improved survival in patients undergoing resection of the mesenteric disease compared with patients that did not (median survival: 11 years vs. 2.6 years)3739. The suggestion is that resection of regional disease delayed onset of the obstruction and ischemia frequently seen in later stages of the disease. Although these data are biased because of surgical selection, it does appear that aggressive surgical resection of regional disease may improve palliation in these patients, who are often experiencing ischemic symptoms38,40.

Furthermore, some investigators have retrospectively shown an improved median pfs of 56 months compared with 25 months in patients whose primary tumour was resected in the face of advanced liver disease41. The authors postulated that the primary tumour may be producing growth factors or peptides that promote the growth of the liver metastases. An ongoing prospective study is now looking at that very question. Further evidence to support the theory that the primary may play a significant role in survival was found in a recent multivariate analysis of 360 small-bowel nets in the United Kingdom. Resection of the primary, age, and Ki67 index proved to be independent predictors of survival42.

The ability to accurately predict the extent of disease both regionally and distantly on convention anatomic and functional imaging has been shown to be limited43. Imaging underestimated the disease found at laparotomy in 35% of the cases and failed to detect liver metastases in 15% of patients with clinical carcinoid syndrome. Surgical exploration by an experienced surgical team is the most accurate way to stage these patients. Thus, all patients with small-bowel nets should be assessed early in the course of their disease within a multidisciplinary clinic where the appropriate surgical expertise is available44.

Question: What is the role of debulking surgery in the management of advanced nets?

  • Debulking, performed by a specialized surgical team, should be considered in a multidisciplinary setting for patients with metastatic disease. Resection may offer benefits in terms of palliation, reduction of serum serotonin metabolite levels, outcomes, and response to subsequent therapies (level ii-2 evidence).

Summary of Evidence: The management of metastatic nets is largely a discussion concerning the therapy of metastatic lesions in the liver. Previous publications examining this topic gave the impression that a complete or R0 resection of metastatic disease was possible (similar to surgical resection of metastatic colorectal cancer). Recent work in this area has demonstrated that the disease pattern in these patients almost always involves multiple bilateral tumour deposits, often in a miliary pattern38,45,46. Consequently, it is almost always impossible to achieve an R0 resection. That having been said, strong evidence appears to demonstrate that aggressive maximal debulking of the hepatic disease is beneficial. Potential mechanisms of benefit include improved survival related to near complete extirpation of tumour, improved symptom control related to decreased endocrinopathy, less damage to the heart as a result of decreased systemic hormone levels, and facilitation of the delivery of other adjuvant therapies (radiolabelled and yttrium therapies). The goals of therapy therefore aim at improving survival or quality of life, or both. To accomplish those goals, surgical therapy needs to be delivered by a team with experience in both hepatobiliary and endocrine surgery. The treatment decisions should be made in the context of a multidisciplinary team and the need to include a full complement of therapies and adjuncts to surgical therapy so that residual disease not removed by surgery can be treated. The other therapies include (but are not limited to) systemic therapies; regional therapies, including radiolabelled and yttrium interventions; bland and chemoembolization; and ablative techniques. All patients should be assessed and considered for disease debulking. Surgical resection and debulking need to be done with low perioperative morbidity and mortality to be justified in the context of palliative surgery.

Question: What are the indications for radioisotope therapy in the management of nets?

  • Baseline octreotide and metaiodobenzylguanidine scans are required if radioisotope therapy is being considered (level iii evidence).

  • Indications for therapy include progressive disease or progressive symptoms, regardless of whether tumours are functional or nonfunctional (level iii evidence).

  • Prospective multicentre trials are required to better elucidate the indications for radioisotope therapy in this patient group (level iii evidence).

3. LOCALLY ADVANCED PANCREATIC CANCER

Question: What is the definition of borderline resectable pancreatic cancer?

  • To determine resectability status, these patients should be referred to an experienced surgeon with expertise in the surgical management of pancreatic cancers.

  • We are in agreement with the consensus definition of borderline resectable pancreatic cancer sponsored by the American Hepato-Pancreato-Biliary Association, the Society of Surgical Oncology, and the Society for Surgery of the Alimentary Tract47, which includes these features:
    • No distant metastases
    • Venous involvement of the superior mesenteric (smv) or portal vein demonstrating tumour abutment with or without impingement and narrowing of the lumen, encasement of the smv or portal vein but without encasement of the nearby arteries, or short segment venous occlusion resulting from either tumour thrombus or encasement but with suitable vessel proximal and distal to the area of vessel involvement, allowing for safe resection and reconstruction
    • Gastroduodenal artery encasement up to the hepatic artery with either short segment encasement or direct abutment of the hepatic artery, without extension to the celiac axis
    • Tumour abutment of the superior mesenteric artery not to exceed 180 degrees of the circumference of the vessel wall

Summary of Evidence: The safety of surgical resection of pancreatic tumours has greatly improved in recent decades. In the 1970s, some authors suggested that it should be abandoned because of prohibitively high morbidity and mortality48. In recent years, pancreatic surgery has been associated with mortality approaching 1% in high-volume centres49. The improved safety profile of pancreatic surgery has led to more aggressive and complicated resections being undertaken. Consequently, the definition of what is technically resectable has also evolved. Large series of pancreaticoduodenectomies with venous resection have demonstrated low mortality and long-term survival equivalent to those without vascular resection50. Resection of the portal vein or smv is now widely accepted. Resection and reconstruction of the celiac axis or superior mesenteric artery during pancreatic resection for adenocarcinoma remains much more controversial51.

The definition of resectable disease varies, which creates difficulties in comparing results across studies and selecting appropriate surgical, neoadjuvant, and adjuvant therapies. To address those problems, considerable effort has been made to standardize the definition of resectable pancreatic cancer47. The concept of a category of “borderline resectable” pancreatic cancer has recently been advocated52,53. A consensus conference on the topic was recently jointly organized by the American Hepato-Pancreato-Biliary Association, the Society of Surgical Oncology, and the Society for Surgery of the Alimentary Tract47. From that conference54, localized and resectable pancreatic cancer was defined as no distant metastases; no radiographic evidence of smv and portal vein abutment, distortion, tumour thrombus, or venous encasement; and clear fat planes around the celiac axis, hepatic artery, and superior mesenteric artery. Tumours considered borderline resectable were also defined as noted earlier.

This distinction from resectable pancreatic cancer is important, because these patients are at higher risk of complications from the increased complexity of the surgery, at high risk of margin positivity, and at high risk of early systemic failure because of the advanced nature of the tumour. Consideration should there be given to neoadjuvant therapy for borderline resectable tumours55. Other authors have shown that a neoadjuvant approach may improve patient selection and lead to low rates of margin positivity and high survival rates despite the high-risk nature of the tumours52.

Although ongoing study is likely to lead to further modification and refinement, the establishment of these standard definitions is an important step in the development of future studies of the treatment of pancreatic cancer. Standardizing these definitions based on objective radiologic features will improve the ability to make comparisons across future studies and represents an important advance in the research into treating pancreatic cancer.

Question: What is the role of systemic therapy in the management of locally advanced pancreatic cancer?

  • In the absence of a clinical trial, patients with a good performance status can be considered for gemcitabine with or without erlotinib (level i evidence)56,57.

  • We encourage further investigation of folfirinox (folinic acid, 5-fluorouracil, irinotecan, oxaliplatin) in this patient population

Summary of Evidence: Historically, patients with locally advanced pancreatic cancer (lapc) have been included in trials with patients having metastatic disease, and thus the bulk of the chemotherapy knowledge for lapc comes from trials that combined locally advanced and metastatic patients. Gemcitabine has been the standard treatment since the landmark trial by Burris and colleagues56 that demonstrated an improvement in os and clinical benefit (a composite measurement of performance status, analgesic use, and pain) with the use of gemcitabine compared with fluorouracil. Since then, numerous trials have attempted to assess the efficacy of various chemotherapy combinations compared with gemcitabine5865. No individual phase iii trial has demonstrated improved survival for combination chemotherapy compared with gemcitabine alone, although a mild improvement in survival was noted in a combined analysis of results from several trials with the combination of gemcitabine and a platinum agent66 and with the combination of gemcitabine and capecitabine63.

More recently, there has been significant interest in assessing the efficacy of molecularly targeted therapies given in combination with gemcitabine. A study by Moore and colleagues57 demonstrated a statistically significant improvement in os from the combination of gemcitabine and erlotinib (an epidermal growth factor receptor tyrosine kinase inhibitor) compared with gemcitabine alone. That clinical trial was, at the time, the only one to demonstrate an improvement in os with combination therapy versus gemcitabine alone. The improvement in os was statistically significant, but only in the range of several weeks, and therefore its clinical significance has been questioned. Despite the questions, combination therapy was adopted as the new standard of care in some centres. Numerous other studies assessing the efficacy of biologic agents, given either in combination with chemotherapy or alone, have been performed, but none has been able to demonstrate an improvement in os compared with gemcitabine alone6770. At the American Society of Clinical Oncology general meeting in 2010, the results of a phase iii study comparing gemcitabine with folfirinox—a chemotherapy regimen combining fluorouracil, leucovorin, irinotecan, and oxaliplatin—were first presented71. The trial included only patients with good performance status, and it should be noted that a high percentage of patients had primary tumours in the body or tail of the pancreas, and were thus less likely than patients with head-of-the-pancreas tumours (the most common primary location seen clinically) to have biliary obstruction72. The trial results were impressive, with a response rate to folfirinox of 31.6% compared with 9.4% for gemcitabine and significant improvements in pfs (6.4 months vs. 3.3 months; hazard ratio: 0.47; p < 0.001) and os (11.1 months vs. 6.8 months; hazard ratio: 0.57; p < 0.001) for folfirinox compared with gemcitabine. Given those results, folfirinox is now an option for first-line therapy in metastatic patients with good performance status. It should be noted that folfirinox was also associated with significant toxicity, including high rates of grade 3 or 4 neutropenia (45.7%).

The design of clinical trials in pancreatic cancer is evolving, and in keeping with recommendations from an expert consensus group73, patients with lapc are, for the most part, no longer included with metastatic patients in clinical trials of advanced pancreatic cancer. Patients with locally advanced disease were not included in the folfirinox trial, and therefore the results of that trial cannot be extrapolated to the lapc population.

Data for the use of folfirinox in the lapc population are limited. A small pilot study of neoadjuvant folfirinox in patients with lapc was presented at the American Society of Clinical Oncology gastrointestinal cancer symposium in 201174. A retrospective series of 12 patients with lapc had been treated with neoadjuvant folfinox. Of 10 evaluable patients, 6 were felt to have been converted to resectability, and 4 had R0 resections. The estimated median survival for that group was 20.7 months. These data are promising, but given the small size of the study, further research is needed.

At the current time, gemcitabine and the combination of gemcitabine and erlotinib remain the standard chemotherapy options for lapc. The data on folfinox in the metastatic setting are encouraging, but given that patients with lapc were not included in the phase iii trial and that only minimal data are available for that population, further investigation of the regimen in locally advanced disease is necessary. Several clinical trials investigating folfirinox either alone, in combination with a biologic agent, or given sequentially with chemoradiotherapy in the lapc setting are planned or are currently ongoing (search for NCT01359007, NCT01413022, and NCT01397019 at http://Clinicaltrials.gov).

Chemoradiotherapy is another option for the treatment of lapc75, but controversy remains concerning whether it adds to chemotherapy alone and what the optimal timing and type of chemotherapy and radiotherapy are. More details are discussed in the evidence summary for the next question. An ongoing phase iii study assessing whether the addition of chemoradiotherapy improves survival compared with systemic therapy alone will help to clarify the role of chemoradiotherapy in the management of lapc76.

Question: What is the role of combined chemoradiation in the management of lapc?

  • There is no consistent evidence of benefit with chemoradiation compared with chemotherapy in patients with lapc (level iii evidence).

  • Chemoradiation could be considered in selected patients after discussion in a multidisciplinary setting (level iii evidence).

Summary of Evidence: The addition of radiation to chemotherapy for lapc continues to be a controversial issue. The support for radiation comes from older Radiation Therapy Oncology Group literature77 as well as from the high local relapse rates reported in modern chemotherapy trials78. The rapid distant spread of pancreatic cancer and the lack of benefit from radiation in the European Study Group for Pancreatic Cancer and the European Organisation for Research and Treatment of Cancer 40891 trials (both of which examined chemoradiation only in the postoperative rather than the locally advanced setting) have adversely influenced the use of radiation for locally advanced tumours79,80. Furthermore, given the disappointing survival in lapc, ongoing attempts to add increasing amounts of chemotherapy as opposed radiation continue to be pursued with very limited success57,67.

With os being closely correlated with distant disease spread, a pragmatic approach of beginning with 2–6 cycles of chemotherapy before delivering radiation is generally endorsed in current trials and by the conference78,81,82. That approach removes patients with early distant disease from the radiation-treated cohort, given that they would likely derive little benefit from radiation treatment. Theoretically, it also allows those who are at greater risk of local relapse (because of absence of disease spread) to receive further targeted local therapy to their site of greatest disease. Gemcitabine and fluorouracil– capecitabine both remain the standard chemoradiation agents, with many trials examining the role of other chemotherapeutics concurrent or sequential with chemoradiation.

Question: What is the role of stereotactic radiation in the management of lapc?

  • At present, no available evidence supports the use of stereotactic radiation in the management of lapc.

Summary of Evidence: Stereotactic radiation has been demonstrated to be acutely well tolerated and convenient for patients. It allows for an increased radiation dose to be delivered to the pancreatic tumour in a reduced number of patient visits. It has also shown benefit in pain control. The increased local control of pancreatic cancer provided by stereotactic radiation has now been demonstrated by phase ii trials from several institutions8387, but no head-to-head trials between stereotactic radiation and conventional three-dimensional conformal radiation have been conducted, and there is no evidence from randomized controlled trial of improved os with stereotactic radiation. We recommend that stereotactic radiation continue to be studied in research settings sequential with chemotherapy.

4. ACKNOWLEDGMENTS

The Western Canadian Gastrointestinal Cancer Consensus Conference organizing committee thanks all meeting participants who contributed to the development of this consensus statement.

5. CONFLICT OF INTEREST DISCLOSURES

DRu has acted in an advisory role for Novartis and Pfizer. The other authors have no financial conflicts of interest to declare.

6. REFERENCES

  • 1.Canadian Task Force on Preventive Health Care (ctfphc) Table 2. Levels of Evidence—Research Design Rating [Web page] London, ON: CTFPHC; 2003. [Available at: http://www.canadiantaskforce.ca/_archive/ctfphc&methods.htm#Table_2; cited June 8, 2012] [Google Scholar]
  • 2.Rinke A, Müller HH, Schade–Brittinger C, et al. on behalf of the promid Study Group Placebo-controlled, double-blind, prospective, randomized study on the effect of octreotide lar in the control of tumor growth in patients with metastatic neuroendocrine midgut tumors: a report from the promid Study Group. J Clin Oncol. 2009;27:4656–63. doi: 10.1200/JCO.2009.22.8510. [DOI] [PubMed] [Google Scholar]
  • 3.Yao JC, Hassan M, Phan A, et al. One hundred years after “carcinoid”: epidemiology of and prognostic factors for neuroendocrine tumors in 35,825 cases in the United States. J Clin Oncol. 2008;26:3063–72. doi: 10.1200/JCO.2007.15.4377. [DOI] [PubMed] [Google Scholar]
  • 4.Reubi JC, Waser B, Schaer JC, Laissue JA. Somatostatin receptor sst1–sst5 expression in normal and neoplastic human tissues using receptor autoradiography with subtype-selective ligands. Eur J Nucl Med. 2001;28:836–46. doi: 10.1007/s002590100541. [DOI] [PubMed] [Google Scholar]
  • 5.Eriksson B, Oberg K. Summing up 15 years of somatostatin analog therapy in neuroendocrine tumors: future outlook. Ann Oncol. 1999;10(suppl 2):S31–8. doi: 10.1093/annonc/10.suppl_2.S31. [DOI] [PubMed] [Google Scholar]
  • 6.Theodoropoulou M, Zhang J, Laupheimer S, et al. Octreotide, a somatostatin analogue, mediates its antiproliferative action in pituitary tumor cells by altering phosphatidylinositol 3-kinase signaling and inducing Zac1 expression. Cancer Res. 2006;66:1576–82. doi: 10.1158/0008-5472.CAN-05-1189. [DOI] [PubMed] [Google Scholar]
  • 7.Florio T. Molecular mechanisms of the antiproliferative activity of somatostatin receptors (sstrs) in neuroendocrine tumors. Front Biosci. 2008;13:822–40. doi: 10.2741/2722. [DOI] [PubMed] [Google Scholar]
  • 8.Plöckinger U, Wiedenmann B. Neuroendocrine tumors. Biotherapy. Best Pract Res Clin Endocrinol Metab. 2007;21:145–62. doi: 10.1016/j.beem.2007.01.002. [DOI] [PubMed] [Google Scholar]
  • 9.Yao JC, Shah MH, Ito T, et al. on behalf of the RAD001 in Advanced Neuroendocrine Tumors, Third Trial (radiant-3) Study Group Everolimus for advanced pancreatic neuroendocrine tumors. N Engl J Med. 2011;364:514–23. doi: 10.1056/NEJMoa1009290. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.O’Reilly T, McSheehy PM. Biomarker development for the clinical activity of the mtor inhibitor everolimus (RAD001): processes, limitations, and further proposals. Transl Oncol. 2010;3:65–79. doi: 10.1593/tlo.09277. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Meric–Bernstam F, Gonzalez–Angulo AM. Targeting the mtor signaling network for cancer therapy. J Clin Oncol. 2009;27:2278–87. doi: 10.1200/JCO.2008.20.0766. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Faivre S, Kroemer G, Raymond E. Current development of mtor inhibitors as anticancer agents. Nat Rev Drug Discov. 2006;5:671–88. doi: 10.1038/nrd2062. [DOI] [PubMed] [Google Scholar]
  • 13.Plank TL, Logginidou H, Klein–Szanto A, Henske EP. The expression of hamartin, the product of the TSC1 gene, in normal human tissues and in TSC1- and TSC2-linked angiomyolipomas. Mod Pathol. 1999;12:539–45. [PubMed] [Google Scholar]
  • 14.Verhoef S, van Diemen–Steenvoorde R, Akkersdijk WL, et al. Malignant pancreatic tumour within the spectrum of tuberous sclerosis complex in childhood. Eur J Pediatr. 1999;158:284–7. doi: 10.1007/s004310051073. [DOI] [PubMed] [Google Scholar]
  • 15.Eledrisi MS, Stuart CA, Alshanti M. Insulinoma in a patient with tuberous sclerosis: is there an association? Endocr Pract. 2002;8:109–12. doi: 10.4158/EP.8.2.109. [DOI] [PubMed] [Google Scholar]
  • 16.von Wichert G, Jehle PM, Hoeflich A, et al. Insulin-like growth factor-i is an autocrine regulator of chromogranin A secretion and growth in human neuroendocrine tumor cells. Cancer Res. 2000;60:4573–81. [PubMed] [Google Scholar]
  • 17.Pollak MN, Polychronakos C, Guyda H. Somatostatin analogue sms 201–995 reduces serum igf-i levels in patients with neoplasms potentially dependent on igf-i. Anticancer Res. 1989;9:889–91. [PubMed] [Google Scholar]
  • 18.Moreno A, Akcakanat A, Munsell MF, Soni A, Yao JC, Meric–Bernstam F. Antitumor activity of rapamycin and octreotide as single agents or in combination in neuroendocrine tumors. Endocr Relat Cancer. 2008;15:257–66. doi: 10.1677/ERC-07-0202. [DOI] [PubMed] [Google Scholar]
  • 19.Yao JC, Phan AT, Chang DZ, et al. Efficacy of RAD001 (everolimus) and octreotide lar in advanced low- to intermediate-grade neuroendocrine tumors: results of a phase ii study. J Clin Oncol. 2008;26:4311–18. doi: 10.1200/JCO.2008.16.7858. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Pavel ME, Hainsworth JD, Baudin E, et al. Everolimus plus octreotide long-acting repeatable for the treatment of advanced neuroendocrine tumours associated with carcinoid syndrome (radiant-2): a randomised, placebo-controlled, phase 3 study. Lancet. 2011;378:2005–12. doi: 10.1016/S0140-6736(11)61742-X. [DOI] [PubMed] [Google Scholar]
  • 21.Raymond E, Dahan L, Raoul JL, et al. Sunitinib malate for the treatment of pancreatic neuroendocrine tumors. N Engl J Med. 2011;364:501–13. doi: 10.1056/NEJMoa1003825. [DOI] [PubMed] [Google Scholar]
  • 22.Johannessen CM, Reczek EE, James MF, Brems H, Legius E, Cichowski K. The NF1 tumor suppressor critically regulates tsc2 and mtor. Proc Natl Acad Sci U S A. 2005;102:8573–8. doi: 10.1073/pnas.0503224102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Tan CC, Hall RI, Semeraro D, Irons RP, Freeman JG. Ampullary somatostatinoma associated with von Recklinghausen’s neurofibromatosis presenting as obstructive jaundice. Eur J Surg Oncol. 1996;22:298–301. doi: 10.1016/S0748-7983(96)80022-9. [DOI] [PubMed] [Google Scholar]
  • 24.van Basten JP, van Hoek B, de Bruïne A, Arends JW, Stockbrügger RW. Ampullary carcinoid and neurofibromatosis: case report and review of the literature. Neth J Med. 1994;44:202–6. [PubMed] [Google Scholar]
  • 25.Yoshida A, Hatanaka S, Ohi Y, Umekita Y, Yoshida H. von Recklinghausen’s disease associated with somatostatin-rich duodenal carcinoid (somatostatinoma), medullary thyroid carcinoma and diffuse adrenal medullary hyperplasia. Acta Pathol Jpn. 1991;41:847–56. doi: 10.1111/j.1440-1827.1991.tb01629.x. [DOI] [PubMed] [Google Scholar]
  • 26.Turner HE, Harris AL, Melmed S, Wass JA. Angiogenesis in endocrine tumors. Endocr Rev. 2003;24:600–32. doi: 10.1210/er.2002-0008. [DOI] [PubMed] [Google Scholar]
  • 27.Terris B, Scoazec JY, Rubbia L, et al. Expression of vascular endothelial growth factor in digestive neuroendocrine tumours. Histopathology. 1998;32:133–8. doi: 10.1046/j.1365-2559.1998.00321.x. [DOI] [PubMed] [Google Scholar]
  • 28.La Rosa S, Uccella S, Finzi G, Albarello L, Sessa F, Capella C. Localization of vascular endothelial growth factor and its receptors in digestive endocrine tumors: correlation with microvessel density and clinicopathologic features. Hum Pathol. 2003;34:18–27. doi: 10.1053/hupa.2003.56. [DOI] [PubMed] [Google Scholar]
  • 29.Christofori G, Naik P, Hanahan D. Vascular endothelial growth factor and its receptors, Flt-1 and Flk-1, are expressed in normal pancreatic islets and throughout islet cell tumorigenesis. Mol Endocrinol. 1995;9:1760–70. doi: 10.1210/me.9.12.1760. [DOI] [PubMed] [Google Scholar]
  • 30.Chaudhry A, Papanicolaou V, Oberg K, Heldin CH, Funa K. Expression of platelet-derived growth factor and its receptors in neuroendocrine tumors of the digestive system. Cancer Res. 1992;52:1006–12. [PubMed] [Google Scholar]
  • 31.Van Gompel JJ, Chen H. Insulin-like growth factor 1 signaling in human gastrointestinal carcinoid tumor cells. Surgery. 2004;136:1297–302. doi: 10.1016/j.surg.2004.06.061. [DOI] [PubMed] [Google Scholar]
  • 32.Zhang PJ, Furth EE, Cai X, Goldblum JR, Pasha TL, Min KW. The role of beta-catenin, tgf beta 3, ngf2, fgf2, igfr2, and bmp4 in the pathogenesis of mesenteric sclerosis and angiopathy in midgut carcinoids. Hum Pathol. 2004;35:670–4. doi: 10.1016/j.humpath.2003.12.010. [DOI] [PubMed] [Google Scholar]
  • 33.Koch CA, Gimm O, Vortmeyer AO, et al. Does the expression of c-Kit (CD117) in neuroendocrine tumors represent a target for therapy? Ann N Y Acad Sci. 2006;1073:517–26. doi: 10.1196/annals.1353.055. [DOI] [PubMed] [Google Scholar]
  • 34.Krishnamurthy S, Dayal Y. Immunohistochemical expression of transforming growth factor alpha and epidermal growth factor receptor in gastrointestinal carcinoids. Am J Surg Pathol. 1997;21:327–33. doi: 10.1097/00000478-199703000-00009. [DOI] [PubMed] [Google Scholar]
  • 35.Halfdanarson TR, Rabe KG, Rubin J, Petersen GM. Pancreatic neuroendocrine tumors (pnets): incidence, prognosis and recent trend toward improved survival. Ann Oncol. 2008;19:1727–33. doi: 10.1093/annonc/mdn351. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Jensen RT, Delle Fave G. Promising advances in the treatment of malignant pancreatic endocrine tumors. N Engl J Med. 2011;364:564–5. doi: 10.1056/NEJMe1013903. [DOI] [PubMed] [Google Scholar]
  • 37.Hellman P, Lundström T, Ohrvall U, et al. Effect of surgery on the outcome of midgut carcinoid disease with lymph node and liver metastases. World J Surg. 2002;26:991–7. doi: 10.1007/s00268-002-6630-z. [DOI] [PubMed] [Google Scholar]
  • 38.Chambers AJ, Pasieka JL, Dixon E, Rorstad O. The palliative benefit of aggressive surgical intervention for both hepatic and mesenteric metastases from neuroendocrine tumors. Surgery. 2008;144:645–51. doi: 10.1016/j.surg.2008.06.008. [DOI] [PubMed] [Google Scholar]
  • 39.Norlén O, Stålberg P, Öberg K, et al. Long-term results of surgery for small intestinal neuroendocrine tumors at a tertiary referral center. World J Surg. 2012;36:1419–31. doi: 10.1007/s00268-011-1296-z. [DOI] [PubMed] [Google Scholar]
  • 40.Akerström G, Hellman P, Hessman O, Osmak L. Management of midgut carcinoids. J Surg Oncol. 2005;89:161–9. doi: 10.1002/jso.20188. [DOI] [PubMed] [Google Scholar]
  • 41.Givi B, Pommier SJ, Thompson AK, Diggs BS, Pommier RF. Operative resection of primary carcinoid neoplasms in patients with liver metastases yields significantly better survival. Surgery. 2006;140:891–7. doi: 10.1016/j.surg.2006.07.033. [DOI] [PubMed] [Google Scholar]
  • 42.Ahmed A, Turner G, King B, et al. Midgut neuroendocrine tumours with liver metastases: results of the ukinets study. Endocr Relat Cancer. 2009;16:885–94. doi: 10.1677/ERC-09-0042. [DOI] [PubMed] [Google Scholar]
  • 43.Chambers AJ, Pasieka JL, Dixon E, Rorstad O. Role of imaging in the preoperative staging of small bowel neuroendocrine tumors. J Am Coll Surg. 2010;211:620–7. doi: 10.1016/j.jamcollsurg.2010.07.016. [DOI] [PubMed] [Google Scholar]
  • 44.Singh S, Law C. Multidisciplinary reference centers: the care of neuroendocrine tumors. J Oncol Pract. 2010;6:e11–16. doi: 10.1200/JOP.2010.000098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Mayo SC, de Jong MC, Pulitano C, et al. Surgical management of hepatic neuroendocrine tumor metastasis: results from an international multi-institutional analysis. Ann Surg Oncol. 2010;17:3129–36. doi: 10.1245/s10434-010-1154-5. [DOI] [PubMed] [Google Scholar]
  • 46.Sarmiento JM, Heywood G, Rubin J, Ilstrup DM, Nagorney DM, Que FG. Surgical treatment of neuroendocrine metastases to the liver: a plea for resection to increase survival. J Am Coll Surg. 2003;197:29–37. doi: 10.1016/S1072-7515(03)00230-8. [DOI] [PubMed] [Google Scholar]
  • 47.Vauthey JN, Dixon E. ahpba/sso/ssat Consensus Conference on Resectable and Borderline Resectable Pancreatic Cancer: rationale and overview of the conference. Ann Surg Oncol. 2009;16:1725–6. doi: 10.1245/s10434-009-0409-5. [DOI] [PubMed] [Google Scholar]
  • 48.Crile G., Jr The advantages of bypass operations over radical pancreatoduodenectomy in the treatment of pancreatic carcinoma. Surg Gynecol Obstet. 1970;130:1049–53. [PubMed] [Google Scholar]
  • 49.Cameron JL, Riall TS, Coleman J, Belcher KA. One thousand consecutive pancreaticoduodenectomies. Ann Surg. 2006;244:10–15. doi: 10.1097/01.sla.0000217673.04165.ea. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Tseng JF, Raut CP, Lee JE, et al. Pancreaticoduodenectomy with vascular resection: margin status and survival duration. J Gastrointest Surg. 2004;8:935–49. doi: 10.1016/j.gassur.2004.09.046. [DOI] [PubMed] [Google Scholar]
  • 51.Bockhorn M, Burdelski C, Bogoevski D, Sgourakis G, Yekebas EF, Izbicki JR. Arterial en bloc resection for pancreatic carcinoma. Br J Surg. 2011;98:86–92. doi: 10.1002/bjs.7270. [DOI] [PubMed] [Google Scholar]
  • 52.Katz MH, Pisters PW, Evans DB, et al. Borderline resectable pancreatic cancer: the importance of this emerging stage of disease. J Am Coll Surg. 2008;206:833–46. doi: 10.1016/j.jamcollsurg.2007.12.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Evans DB, Erickson BA, Ritch P. Borderline resectable pancreatic cancer: definitions and the importance of multimodality therapy. Ann Surg Oncol. 2010;17:2803–5. doi: 10.1245/s10434-010-1285-8. [DOI] [PubMed] [Google Scholar]
  • 54.Callery MP, Chang KJ, Fishman EK, et al. Pretreatment assessment of resectable and borderline resectable pancreatic cancer: expert consensus statement. Ann Surg Oncol. 2009;16:1727–33. doi: 10.1245/s10434-009-0408-6. [DOI] [PubMed] [Google Scholar]
  • 55.Abrams RA, Lowy AM, O’Reilly EM, Wolff RA, Picozzi VJ, Pisters PW. Combined modality treatment of resectable and borderline resectable pancreas cancer: expert consensus statement. Ann Surg Oncol. 2009;16:1751–6. doi: 10.1245/s10434-009-0413-9. [DOI] [PubMed] [Google Scholar]
  • 56.Burris HA, 3rd, Moore MJ, Andersen J, et al. Improvements in survival and clinical benefit with gemcitabine as first-line therapy for patients with advanced pancreas cancer: a randomized trial. J Clin Oncol. 1997;15:2403–13. doi: 10.1200/JCO.1997.15.6.2403. [DOI] [PubMed] [Google Scholar]
  • 57.Moore MJ, Goldstein D, Hamm J, et al. on behalf of the ncic Clinical Trials Group Erlotinib plus gemcitabine compared with gemcitabine alone in patients with advanced pancreatic cancer: a phase iii trial of the National Cancer Institute of Canada Clinical Trials Group. J Clin Oncol. 2007;25:1960–6. doi: 10.1200/JCO.2006.07.9525. [DOI] [PubMed] [Google Scholar]
  • 58.Heinemann V, Quietzsch D, Gieseler F, et al. Randomized phase iii trial of gemcitabine plus cisplatin compared with gemcitabine alone in advanced pancreatic cancer. J Clin Oncol. 2006;24:3946–52. doi: 10.1200/JCO.2005.05.1490. [DOI] [PubMed] [Google Scholar]
  • 59.Colucci G, Giuliani F, Gebbia V, et al. Gemcitabine alone or with cisplatin for the treatment of patients with locally advanced and/or metastatic pancreatic carcinoma: a prospective, randomized phase iii study of the Gruppo Oncologia dell’Italia Meridionale. Cancer. 2002;94:902–10. doi: 10.1002/cncr.10323. [DOI] [PubMed] [Google Scholar]
  • 60.Colucci G, Labianca R, Di Costanzo F, et al. on behalf of the Gruppo Oncologico Italia Meridionale (goim) the Gruppo Italiano per lo Studio dei Carcinomi dell’Apparato Digerente (giscad) the Gruppo Oncologico Italiano di Ricerca Clinica (goirc) Randomized phase iii trial of gemcitabine plus cisplatin compared with single-agent gemcitabine as first-line treatment of patients with advanced pancreatic cancer: the gip-1 study. J Clin Oncol. 2010;28:1645–51. doi: 10.1200/JCO.2009.25.4433. [DOI] [PubMed] [Google Scholar]
  • 61.Louvet C, Labianca R, Hammel P, et al. Gemcitabine in combination with oxaliplatin compared with gemcitabine alone in locally advanced or metastatic pancreatic cancer: results of a gercor and giscad phase iii trial. J Clin Oncol. 2005;23:3509–16. doi: 10.1200/JCO.2005.06.023. [DOI] [PubMed] [Google Scholar]
  • 62.Herrmann R, Bodoky G, Ruhstaller T, et al. Gemcitabine plus capecitabine compared with gemcitabine alone in advanced pancreatic cancer: a randomized, multicenter, phase iii trial of the Swiss Group for Clinical Cancer Research and the Central European Cooperative Oncology Group. J Clin Oncol. 2007;25:2212–17. doi: 10.1200/JCO.2006.09.0886. [DOI] [PubMed] [Google Scholar]
  • 63.Cunningham D, Chau I, Stocken DD, et al. Phase iii randomized comparison of gemcitabine versus gemcitabine plus capecitabine in patients with advanced pancreatic cancer. J Clin Oncol. 2009;27:5513–18. doi: 10.1200/JCO.2009.24.2446. [DOI] [PubMed] [Google Scholar]
  • 64.Rocha Lima CM, Green MR, Rotche R, et al. Irinotecan plus gemcitabine results in no survival advantage compared with gemcitabine monotherapy in patients with locally advanced or metastatic pancreatic cancer despite increased tumor response rate. J Clin Oncol. 2004;22:3776–83. doi: 10.1200/JCO.2004.12.082. [DOI] [PubMed] [Google Scholar]
  • 65.Oettle H, Richards D, Ramanathan RK, et al. A phase iii trial of pemetrexed plus gemcitabine versus gemcitabine in patients with unresectable or metastatic pancreatic cancer. Ann Oncol. 2005;16:1639–45. doi: 10.1093/annonc/mdi309. [DOI] [PubMed] [Google Scholar]
  • 66.Heinemann V, Labianca R, Hinke A, Louvet C. Increased survival using platinum analog combined with gemcitabine as compared to single-agent gemcitabine in advanced pancreatic cancer: pooled analysis of two randomized trials, the gercor/giscad intergroup study and a German multicenter study. Ann Oncol. 2007;18:1652–9. doi: 10.1093/annonc/mdm283. [DOI] [PubMed] [Google Scholar]
  • 67.Philip PA, Benedetti J, Corless CL, et al. Phase iii study comparing gemcitabine plus cetuximab versus gemcitabine in patients with advanced pancreatic adenocarcinoma: Southwest Oncology Group–directed Intergroup Trial S0205. J Clin Oncol. 2010;28:3605–10. doi: 10.1200/JCO.2009.25.7550. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Van Cutsem E, Vervenne WL, Bennouna J, et al. Phase iii trial of bevacizumab in combination with gemcitabine and erlotinib in patients with metastatic pancreatic cancer. J Clin Oncol. 2009;27:2231–7. doi: 10.1200/JCO.2008.20.0238. [DOI] [PubMed] [Google Scholar]
  • 69.Moore MJ, Hamm J, Dancey J, et al. on behalf of ncic Clinical Trials Group Comparison of gemcitabine versus the matrix metalloproteinase inhibitor bay 12-9566 in patients with advanced or metastatic adenocarcinoma of the pancreas: a phase iii trial of the National Cancer Institute of Canada Clinical Trials Group. J Clin Oncol. 2003;21:3296–302. doi: 10.1200/JCO.2003.02.098. [DOI] [PubMed] [Google Scholar]
  • 70.Kindler HL, Niedzwiecki D, Hollis D, et al. Gemcitabine plus bevacizumab compared with gemcitabine plus placebo in patients with advanced pancreatic cancer: phase iii trial of the Cancer and Leukemia Group B (calgb 80303) J Clin Oncol. 2010;28:3617–22. doi: 10.1200/JCO.2010.28.1386. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Conroy T, Desseigne F, Ychou M, et al. on behalf of the Groupe Tumeurs Digestives of Unicancer and the prodige Intergroup folfirinox versus gemcitabine for metastatic pancreatic cancer. N Engl J Med. 2011;364:1817–25. doi: 10.1056/NEJMoa1011923. [DOI] [PubMed] [Google Scholar]
  • 72.Kim R. folfirinox: a new standard treatment for advanced pancreatic cancer? Lancet Oncol. 2011;12:8–9. doi: 10.1016/S1470-2045(10)70237-0. [DOI] [PubMed] [Google Scholar]
  • 73.Philip PA, Mooney M, Jaffe D, et al. Consensus report of the national cancer institute clinical trials planning meeting on pancreas cancer treatment. J Clin Oncol. 2009;27:5660–9. doi: 10.1200/JCO.2009.21.9022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Hosein PJ, Kawamura C, Macintyre J, et al. Pilot study of neoadjuvant folfirinox in unresectable locally advanced pancreatic carcinoma [abstract 234] J Clin Oncol. 2011;29(suppl 4) [Available online at http://www.asco.org/ASCOv2/Meetings/Abstracts?&vmview=abst_detail_view&confID=103&abstractID=70687; cited September 13, 2012] [Google Scholar]
  • 75.Huguet F, Girard N, Guerche CS, Hennequin C, Mornex F, Azria D. Chemoradiotherapy in the management of locally advanced pancreatic carcinoma: a qualitative systematic review. J Clin Oncol. 2009;27:2269–77. doi: 10.1200/JCO.2008.19.7921. [DOI] [PubMed] [Google Scholar]
  • 76.Hammel P, Huguet F, Van Laethem JL, et al. Randomized multicenter phase iii study in patients with locally advanced adenocarcinoma of the pancreas: gemcitabine with or without chemoradiotherapy and with or without erlotinib—lap 07 study [abstract e14619] J Clin Oncol. 2011. p. 29. [Available online at http://www.asco.org/ASCOv2/Meetings/Abstracts?&vmview=abst_detail_view&confID=102&abstractID=83116; cited September 13, 2012]
  • 77.Treatment of locally unresectable carcinoma of the pancreas: comparison of combined-modality therapy (chemotherapy plus radiotherapy) to chemotherapy alone. Gastrointestinal Tumor Study Group J Natl Cancer Inst. 1988;80:751–5. [PubMed] [Google Scholar]
  • 78.Wilkowski R, Boeck S, Ostermaier S, et al. Chemoradiotherapy with concurrent gemcitabine and cisplatin with or without sequential chemotherapy with gemcitabine/cisplatin vs chemoradiotherapy with concurrent 5-fluorouracil in patients with locally advanced pancreatic cancer—a multi-centre randomised phase ii study. Br J Cancer. 2009;101:1853–9. doi: 10.1038/sj.bjc.6605420. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Neoptolemos JP, Dunn JA, Stocken DD, et al. on behalf of the European Study Group for Pancreatic Cancer Adjuvant chemoradiotherapy and chemotherapy in resectable pancreatic cancer: a randomised controlled trial. Lancet. 2001;358:1576–85. doi: 10.1016/S0140-6736(01)06651-X. [DOI] [PubMed] [Google Scholar]
  • 80.Smeenk HG, van Eijck CH, Hop WC, et al. Long-term survival and metastatic pattern of pancreatic and periampullary cancer after adjuvant chemoradiation or observation: long-term results of eortc trial 40891. Ann Surg. 2007;246:734–40. doi: 10.1097/SLA.0b013e318156eef3. [DOI] [PubMed] [Google Scholar]
  • 81.Patel M, Hoffe S, Malafa M, et al. Neoadjuvant gtx chemotherapy and imrt-based chemoradiation for borderline resectable pancreatic cancer. J Surg Oncol. 2011;104:155–61. doi: 10.1002/jso.21954. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Gillmore R, Laurence V, Raouf S, et al. Chemoradiotherapy with or without induction chemotherapy for locally advanced pancreatic cancer: a U.K. multi-institutional experience. Clin Oncol (R Coll Radiol) 2010;22:564–9. doi: 10.1016/j.clon.2010.05.007. [DOI] [PubMed] [Google Scholar]
  • 83.Schellenberg D, Kim J, Christman–Skieller C, et al. Single-fraction stereotactic body radiation therapy and sequential gemcitabine for the treatment of locally advanced pancreatic cancer. Int J Radiat Oncol Biol Phys. 2011;81:181–8. doi: 10.1016/j.ijrobp.2010.05.006. [DOI] [PubMed] [Google Scholar]
  • 84.Mahadevan A, Jain S, Goldstein M, et al. Stereotactic body radiotherapy and gemcitabine for locally advanced pancreatic cancer. Int J Radiat Oncol Biol Phys. 2010;78:735–42. doi: 10.1016/j.ijrobp.2009.08.046. [DOI] [PubMed] [Google Scholar]
  • 85.Schellenberg D, Goodman KA, Lee F, et al. Gemcitabine chemotherapy and single-fraction stereotactic body radiotherapy for locally advanced pancreatic cancer. Int J Radiat Oncol Biol Phys. 2008;72:678–86. doi: 10.1016/j.ijrobp.2008.01.051. [DOI] [PubMed] [Google Scholar]
  • 86.Didolkar MS, Coleman CW, Brenner MJ, et al. Image-guided stereotactic radiosurgery for locally advanced pancreatic adenocarcinoma results of first 85 patients. J Gastrointest Surg. 2010;14:1547–59. doi: 10.1007/s11605-010-1323-7. [DOI] [PubMed] [Google Scholar]
  • 87.Rwigema JC, Parikh SD, Heron DE, et al. Stereotactic body radiotherapy in the treatment of advanced adenocarcinoma of the pancreas. Am J Clin Oncol. 2011;34:63–9. doi: 10.1097/COC.0b013e3181d270b4. [DOI] [PubMed] [Google Scholar]

Articles from Current Oncology are provided here courtesy of Multidisciplinary Digital Publishing Institute (MDPI)

RESOURCES