Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 Jul 1.
Published in final edited form as: Pancreas. 2012 Jul;41(5):678–684. doi: 10.1097/MPA.0b013e318249955a

Expanding Surgical Treatment of Pancreatic Cancer: The Role of Regional Chemotherapy

Jeremy L Davis 1, Prakash K Pandalai 1, R Taylor Ripley 1, Russell C Langan 1, Itzhak Avital 1
PMCID: PMC3375496  NIHMSID: NIHMS355056  PMID: 22695088

Abstract

Objectives

Pancreatic cancer is a lethal disease that offers little chance of long-term survival for patients with unresectable tumors. Surgery remains the most effective means of attaining prolonged survival, yet its role remains limited. Regional chemotherapy has been described for patients with pancreatic cancer, including reports of objective tumor regression allowing for tumor resection in previously unresectable cases, however comprehensive data have not been reviewed to date.

Methods

A review of the literature from 1995 to 2010 was performed to analyze the results of regional chemotherapy administered to patients with advanced pancreatic cancer. Reports of individual cases, post-operative regional therapy and treatment of mixed tumor types were excluded.

Results

Twenty-one reports of 895 total patients with pancreatic cancer were reviewed. Greater than 95% of patients had stage III or IV adenocarcinoma. Objective response rates ranged from nil to 58%, with associated median survivals of 4 to 22 months. Low grade gastrointestinal and hematologic toxicities were not uncommon.

Conclusions

Regional chemotherapy can be administered safely to patients with pancreatic cancer, but with unclear benefit. Advanced pancreatic tumors converted to resectable status by the use of regional chemotherapy may improve patient survival.

Keywords: Pancreatic cancer, chemotherapy, cancer, regional perfusion, infusion, intra-arterial

Introduction

Every year in the United States alone an estimated 43,140 cases of pancreatic cancer are diagnosed and 36,800 patients die of the disease, making it one of the most lethal forms of cancer in adults.[1] Overall survival is poor, with approximately 23% of patients living 12 months after diagnosis.[2] Patients who undergo complete tumor resection survive 18 to 20 months in most series, with or without the addition of single-agent chemotherapy.[3] Unfortunately, less than 20% of patients with pancreatic cancer have resectable tumors at the time of diagnosis, most often due to invasion of adjacent vasculature or metastatic disease. In cases which tumors are detected early enough to allow resection, the choice of adjuvant chemotherapy is based on a randomized clinical trial that demonstrated significant improvement in median disease-free survival favoring gemcitabine over observation alone.[3] The addition of targeted molecular agents or cytotoxic drugs to gemcitabine adds little or no clinical benefit to patients with this disease.[4, 5] Likewise, current data are equivocal regarding the benefit of adjuvant chemoradiotherapy. For patients with inoperable pancreatic cancer systemic chemotherapy may prolong survival and improve quality of life, yet it can only be considered truly palliative in patients without a surgical treatment option.[6]

It can be estimated from population-based data that approximately 20 - 30% of patients with pancreatic cancer present with locally advanced disease, defined generally by tumor extension beyond the pancreas with or without regional lymph node metastases.[7] The term “locally advanced” has been applied liberally to patients with regional disease most often ascribed to advanced tumor stage (T3 and T4) that corresponds with American Joint Committee on Cancer (AJCC) stage II or III.[8] Advanced tumor stage that involves adjacent mesenteric vasculature is subdivided further into three categories based on pre-operative computed tomography: resectable, borderline resectable, and unresectable (Supplemental Table A). The accepted definitions of resectability are based on the type and degree of arterial and venous involvement and the presence of peritoneal or distant metastases.[9, 10]

Increasing the rate of complete tumor resection among patients with pancreatic cancer represents a practical approach to improving survival for those patients who present with advanced stage disease and currently have no surgical treatment option. Achieving this goal requires neoadjuvant therapy that mediates substantial tumor regression, allowing for complete resection in previously unresectable patients. Administration of regional chemotherapy is used currently to treat local-regional and metastatic disease for many cancer histologies. The pharmacologic rationale for regional drug delivery is to increase drug concentrations at tumor sites and limit systemic drug exposure and its toxicity.[11] The role of regional chemotherapy as an adjunctive therapy in patients with local-regional disease has been well documented since Creech et al. employed an extracorporeal circuit for regional isolation perfusion of nitrogen mustard compounds in the treatment of 24 patients with a variety of cancers.[12] Examples of effective contemporary regional therapy include isolated limb perfusion for cutaneous melanoma and hyperthermic intraperitoneal chemotherapy for primary peritoneal mesothelioma and carcinoma of ovarian, appendiceal and colorectal origin.[1317]

The purpose of this review is to evaluate the available evidence for regional chemotherapy in advanced stage pancreatic cancer. We use these data to understand the potential benefits of tumor response and patient survival as well as the toxicity of this treatment strategy. We also explore the rationale for prospective investigation of this strategy with the intent of minimizing toxicity and increasing treatment efficacy, as measured by tumor response and increased rates of tumor resection.

Methods

A search of the Medline database was performed to identify published reports of regional chemotherapy for pancreatic cancer in the English language literature from January 1995 to January 2010. Medical subject heading (MeSH) terms used included (1) pancreatic neoplasm, (2) infusion, intra-arterial, and (3) chemotherapy, cancer, regional perfusion. We focused the search by excluding the following: case reports, dose-escalation (phase I) trials, combined chemotherapy and radiation trials, and studies of perioperative or adjuvant regional chemotherapy. Reports that included multiple gastrointestinal histologies were also excluded. I n instances which institutions published updated patient data or combined analyses, we used the most recent publications. Data collected included the year of publication, patient sample size, age and gender, tumor histopathology and stage, type of regional therapy, toxicity and complications, response rate, and survival rate when available.

Patients

Twenty-one different studies published between 1995 and 2010 reported results of regional chemotherapy administered to 895 patients with advanced stage pancreatic cancer (Table 1).[1838] Included were 62% (389/630) males and 38% (241/630) females in 20 studies reporting gender data. The mean age was 60 years in ten reports, and the median age ranged from 55 to 66 years (median 62) in eight other reports; three studies did not provide patients’ age. Median time to follow-up was not reported in any series.

Table 1.

Trials of Regional Chemotherapy for Pancreatic Cancer: Patient Demographics, Technical Approach, and Chemotherapy

Author (Year) Country Patients Mean Age1 Gender (M:F) Procedure Chemotherapeutic agents
Fiorentini (1996) Italy 20 NR 16:4 HAP MMC
Muchmore (1996) USA 12 56 3:9 CAI MMC, 5-FU, Folinic acid
Link (1997) Gemany 32 60 19:13 CAI CDDP, Mitoxantrone, 5-FU, Folinic acid
Lorenz (1998) Germany 17 61* 12:5 HAP MMC
Maurer (1998) Switzerland 12 59.9 9:3 CAI CDDP, Mitoxantrone, 5-FU, Folinic acid
Klapdor (1999) Germany 28 NR 17:11 CAI Gemcitabine, MMC
Homma (2000) Japan 31 61.5 18:13 SAI CDDP, 5-FU, Folinic acid
Bayar (2003) Turkey 14 55* 8:6 CAI CDDP, MMC, 5-FU, Folinic acid
Ohigashi (2003) Japan 32 60 20:12 SAI Angiotensin-II, Methotrexate
van Ijken (2004) Netherlands 21 59 12:9 CAI2 / HAP Melphalan, MMC
Aigner (2005) Germany 265 NR NR CAI / HAP CDDP, Mitoxantrone, MMC, degradable startch microspheres
Takamori (2005) Japan 24 62.6* 16:8 CAI3 Gemcitabine, 5-FU, Folinic acid
Barletta (2006) Italy 32 62* 22:10 SAI4 Carboplatin, Epirubicin, 5-FU, Folinic acid
Mambrini (2006) Italy 211 61* 130:81 CAI Carboplatin, Epirubicin, 5-FU, Folinic acid
Meyer (2006) Germany 17 54.5 11:6 HAP MMC
Guadagni (2007) Italy 22 66* 12:10 HAP CDDP, MMC
Ikeda (2007) Japan 33 60 22:11 SAI5 Gemcitabine, 5-FU, Folinic acid
Ishikawa (2007) Japan 20 63.7* 9:11 CAI Angiotensin-II, CDDP, Gemcitabine, 5-FU, Folinic acid
Milandri (2007) Italy 19 62* 12:7 SAI4 Carboplatin, Epirubicin, 5-FU, Folinic acid
Nakchbandi (2008) Germany 17 65.7 11:6 CAI Gemcitabine, MMC, Warfarin
Sasada (2008) Japan 16 61.2 10:6 SAI5 CDDP, 5-FU, Folinic acid

HAP, hypoxic abdominal perfusion; CAI, celiac axis infusion; SAI, selective arterial infusion; MMC, mitomycin C; 5-FU, 5-fluorouracil; CDDP, cis-platinum;

1

NR, not reported;

*

Studies reporting median age.

2

CAI performed in 9 of 21 patients that underwent HAP.

3

CAI performed after selective peri-pancreatic artery embolization.

4

Patients with liver metastases received half of chemotherapy dose via hepatic artery.

5

SAI preceded by selective arterial embolization.

Clinicopathologic features of the primary pancreatic cancer

Eleven studies provided data on tumor type for all patients (n=425) whereas three studies reported histopathology for only a fraction (n=39); tumor pathology was either not confirmed or not reported in 431 of 895 (48%) patients (Table 2). Among 464 patients with histopathologic data, 96% (447/464) had adenocarcinoma, 2% (7/464) mucinous carcinoma, 1% (3/464) cystadenocarcioma, <1% (1/464) anaplastic carcinoma, and 1% (6/464) were classified as undifferentiated or “other”. Pancreatic cancer staging was clearly specified in nine studies encompassing 633 patients (71%, 633/ 895). Eight reports utilized UICC (International Union Against Cancer) staging, and one study cited the Japan Pancreas Society staging system while also providing the TNM (tumor, node, metastasis) classification. Of the nine studies reporting tumor stage using a defined system, 46% (290 of 633) were stage III, 54% (339 of 633) stage IV, and less than 1% (4 of 633) stage I–II. Reports that did not specify a staging system simply described patients as having “locally advanced”, “unresectable” or “inoperable” pancreatic cancer with or without metastases. Other reports stated that patients had stage III or IV disease without providing additional information. The term “locally advanced” often was used interchangeably with stage III disease, while metastatic disease was synonymous with stage IV. Therefore, despite the deficiencies in reporting, if ancillary information was used to decipher tumor stage based on current AJCC staging (e.g. “T4 tumor without metastases”), 41% (363 of 875) were stage III and 58% (508 of 875) were stage IV. Only one study, by Sasada et al., provided criteria for declaring tumors unresectable based on invasion of the superior mesenteric or celiac artery and/or occlusion or stenosis of the portal or superior mesenteric vein. The remaining studies in this review provided no information regarding criteria used for determining the resectability of non-metastatic tumors.

Table 2.

Clinicopathologic Details

Histology
Stage
Author Patients Adenocarcinoma Other Unknown1 Stage I/II Stage III Stage IV Unknown1 Staging System
Fiorentini 20 20 - - - 12 8 - NR
Muchmore 12 11 1 - - - - 12 NR
Link 32 28 4 - - 17 15 - UICC
Lorenz 17 17 - - 2 10 5 - UICC
Maurer 12 12 - - - 6 6 - UICC
Klapdor 28 - - 28 - - 20 8 NR
Homma 31 31 - - - - 31 - UICC
Bayar 14 - - 14 - 5 9 - NR
Ohigashi 32 - - 32 - 32 - - UICC
van Ijken 21 - - 21 - 15 6 - NR
Aigner 265 - - 265 - 112 153 - UICC
Takamori 24 - - 24 - 3 21 - NR
Barletta 32 32 - - - 7 25 - NR
Mambrini 211 203 8 - - 99 112 - UICC
Meyer 17 - - 17 - 7 10 - NR
Guadagni 22 20 2 - - 14 8 - NR
Ikeda 33 19 - 14 - - 33 - NR
Ishikawa 20 - - 20 - - 20 - NR
Milandri 19 17 2 - - 10 9 - NR
Nakchbandi 17 17 - - 2 2 13 - UICC
Sasada 16 9 - 7 - 12 4 - JPS3/TNM
1

Unreported or not confirmed prior to therapy; not discernible from manuscript

2

UICC, International Union Against Cancer

3

Japan Pancreas Society; TNM, tumor, node, metastasis.

NR, not reported.

Prior therapy

Seventeen studies provided information on 277 patients receiving treatment prior to regional chemotherapy. Fourteen studies described 196 patients as having had prior surgery; 14 patients were described as having a prior curative resection, 122 had a bypass or other palliative procedure, and the remaining 60 patients were not further specified. Eighty-one of the 277 patients received systemic chemotherapy and/or radiation treatment prior to regional chemotherapy. Three authors disclosed that no patients received systemic chemotherapy prior to regional chemotherapy.[29, 35, 38]

Regional chemotherapy

Catheter-based arterial infusion and perfusion techniques, with and without hemofiltration, were used to deliver regional chemotherapy in these studies. Briefly, arterial perfusion generally referred to the use of a closed circuit incorporating arterial and venous catheters, whereas infusion techniques did not utilize venous return catheters and therefore the chemotherapeutic agents were open to the effects of systemic circulation. Abdominal perfusion and arterial infusion were performed commonly via access to the femoral vessels whereby arterial catheters were positioned with fluoroscopic guidance in the celiac trunk for celiac axis infusion (CAI) and into peri-pancreatic arteries for selective arterial infusion (SAI). In three reports, arterial catheters were placed at the time of laparotomy to obtain more selective pancreatic infusion. For abdominal perfusion with or without isolation of the abdominal compartment, also referred to as hypoxic abdominal perfusion (HAP) or aortic stop-flow infusion, balloon catheters were inserted into the femoral artery and vein and advanced into the aorta and vena cava, respectively, with occluding balloon cuffs placed at the level of the diaphragm. Arterial and venous catheters were attached to a roller pump or extracorporeal circuit with or without a filtration device. In order to isolate the abdomen tourniquets were applied to the thighs to exclude perfusion of the legs.

Celiac axis infusion (CAI) was used in a majority of studies (52%, 11/21) whereas selective arterial infusion (SAI; 29%, 6/21) and hypoxic abdominal perfusion (HAP: 29%, 6/21) were used less often. Two series reported using hemofiltration in the extracorporeal circuit, whereas four studies of HAP did not use hemofiltration. In two studies, CAI and HAP were utilized sequentially.[27, 28] In an attempt to direct blood flow to tumor or pancreas only, four studies (19%, 4/21) utilized selective arterial embolization prior to arterial chemotherapy infusion.[24, 29, 34, 38] Technical variations in arterial catheterization, including percutaneous versus open surgical approach, appeared to reflect changes in procedural experience over time and the method of patient selection. A variety of chemotherapeutic agents were used alone or in combination (Table 1). 5-fluorouracil (5-FU) was used most often (57%) followed by mitomycin-C (MMC; 48%), cisplatinum (CDDP; 38%), gemcitabine (24%), mitoxantrone (19%), epirubicin and carboplatin (14%), methotrexate (5%) and melphalan (5%). Three studies also included adjuncts to chemotherapy: warfarin, angiotensin-II, and degradable starch microspheres.[26, 28, 37]

Response Rates

Tumor response rate and patient survival were reported as study endpoints; 19 studies reported tumor responses, 10 reported survival, and 9 reported both response and survival rates. Objective response criteria were defined in 17 of 21 (81%) studies (Table 3). World Health Organization (WHO) response criteria were used by 71% (15/21) of studies; nine authors (43%, 9/21) specifically referenced WHO, whereas six (29%, 6/21) provided definitions for partial and complete responses and progressive disease that were synonymous with WHO criteria. One study utilized Southwest Oncology Group (SWOG) criteria, while another used Response Evaluation Criteria In Solid Tumors (RECIST) criteria. The average response rate reported was 26% (range 0 to 58%) for 19 of 21 studies reporting response data, regardless of criteria. For 17 of 21 studies (564/895 patients) utilizing defined response criteria, the response rate was 25%. Two studies reported tumor response rate without defining response criteria, and two additional studies neither defined nor reported response rate. Studies by van Ijken, Milandri and Sasada et al. provided response data for only a subset of total patients treated (16 of 21, 16 of 19 and 12 of 16 patients, respectively), and these are included in the average response rates reported above.

Table 3.

Response Rate and Survival

Author Response
Criteria
Response Rate1
(%)
1-yr Survival
(%)
Median Survival
(months)
Fiorentini NR 50 NR NR
Muchmore WHO* 46 NR NR
Link WHO 19 NR 12 (Stage III)
4 (Stage IV)
Lorenz WHO 0 NR 4.2
Maurer WHO 8 NR 6
Klapdor WHO 46 NR 9
Homma WHO 58 67 NR
Bayar WHO* 36 NR 8
Ohigashi WHO* 6 56 13
van Ijken WHO 5^ NR 6
Aigner NR NR NR 9
Takamori WHO 21 51 14
Barletta WHO* 22 50 6.1
Mambrini SWOG 8 NR 9.2
Meyer NR 18 6 4.1
Guadagni WHO* 18 9 6
Ikeda WHO* 24 NR 13
Ishikawa WHO 25 45 12
Milandri WHO 25^ 16 6
Nakchbandi NR NR 6 6.8
Sasada RECIST 58^ 83 22
1

Response rate reflects partial and complete responses; mixed or minor responses not included

*

WHO criteria as described in methods, however WHO criteria reference not cited.

^

response rate based on fraction of total number of patients treated

WHO, World Health Organization; SWOG, Southwest Ongology Group; RECIST, Response Evaluation Criteria In Solid Tumors.

Toxicity

General toxicity was reported in either a graded format or simply with descriptive text. WHO and National Cancer Institute (NCI, Common Terminology Criteria for Adverse Events) criteria were at least noted in the text of 17 reports, however only 14 of 21 (67%) studies provided comprehensible toxicity data amenable to interpretation (Supplemental Table B). Of 895 total patients, toxicity data were available for 288 (32%). Grade I to II gastrointestinal toxicity occurred in 48% (139 of 288), and grade III to IV in 13% (38 of 288). The incidence of hematologic toxicity was similar with 46% (133 of 288) grade I to II, and 22% (62 of 288) grade III to IV. Link et al. reported toxicity as percentage of total cycles of infusion performed; the incidence of gastrointestinal toxicity was 54% grade I to II and 10% grade III to IV, and 9% for grade I to II and 1% for grade III to IV hematologic toxicity. Specific examples of procedure-related morbidity included arterial dissection (n=3), catheter dislocation (n=6), duodenal or gastric ulcer (n=4), hepatic abscess (n=2), port site dysfunction (n=2), groin lymphatic fistula (n=6), and deep vein thrombosis (n=6). Meyer et al. reported a 30-day mortality of 17.6 % (3 of 17 patients) and van Ijken et al. described a treatment related mortality attributed to acute mesenteric ischemia; total treatment-related mortality was 0.04% (4/895).

Survival

The average 1-year survival was 38.8% (range 6% to 83%, n=10 studies reporting), with a median survival of 4 to 22 months (median 8.5 months, n=17 studies reporting). Few studies analyzed survival by tumor stage. Link et al. reported 12 months median survival for patients with stage III disease compared to 4 months median survival for patients with stage IV disease.[20] Mambrini et al. reported 10.5 and 6.6 months median survival and Maurer et al. reported 8.5 and 5 months median survival for patients with stage III and IV disease, respectively.[22, 31] In a subsequent report by Mambrini et al., prognostic factors associated with improved survival were sought for the original cohort of 211 patients. Factors associated with improved survival were pain reduction during treatment greater than 30% of baseline, non-metastatic disease, and greater than 3 cycles of regional chemotherapy received.[39] Meyer et al. reported median survival of 3.2 months for patients without metastases and 4.7 months for those without.[32]

Progression to surgery following chemotherapy

Eighty-five patients (9%, 85/895) underwent exploratory surgery following regional chemotherapy. One patient was explored despite having progressive disease and underwent resection.[21] Nakbachandi et al. reported “downstaging” of tumor in three patients, resulting in resection and prolonged survival (8.2, 10.7 and 17.5 months).[37] In three additional studies, 7 patients were re-explored, one of which underwent a pancreaticoduodenectomy, one was unresectable, and the remaining five had a procedure not otherwise specified.[19, 22, 36] In the largest series to report re-exploration of patients following regional chemotherapy, Aigner et al. provided data on 80 patients surviving greater than 12 months beyond treatment, of which 74 underwent surgical exploration; 31 of 74 (42%) underwent resection via pancreaticoduodenectomy, partial pancreatectomy, or evacuation of tumor necrosis.[28]

Discussion

Approximately 80% of pancreatic cancer patients have no option for curative resection at the time of diagnosis. Half of all patients are diagnosed with metastatic disease, while approximately 25% present with advanced tumor stage considered unresectable due to involvement of adjacent mesenteric vasculature.[7, 40] Patients with unresectable tumors have only a 20% chance of surviving one year following diagnosis and treatment with single agent chemotherapy.[41] For the relatively small number of patients with tumors amenable to resection, the most common sites of recurrence are the local resection bed, liver and peritoneum. For these reasons the application of regional chemotherapy for advanced pancreatic cancer has been posited not only as a method for treating unresectable disease, but also as prevention of local and hepatic recurrences in the adjuvant setting. In this review of 21 published reports representing 895 patients with both locally invasive and metastatic pancreatic cancer, regional chemotherapy offers an average one-year survival rate of 39% and median survival of 8.5 months, which approximates survival for advanced (unresectable) pancreatic cancer patients receiving systemic chemotherapy with or without radiation therapy.[4244]

Although previous classification systems have described the extent of vascular involvement in order to guide pre-operative decision making, only within the last decade have patients benefited from consensus criteria used to determine tumor resectability.[9, 45, 46] In this review, the often used description of locally advanced (non-metastatic) pancreatic cancers as “inoperable” provides inadequate information regarding patient selection, thus weakening the conclusions taken from these studies. Different tumor resectability criteria used among investigators and increasing allowance for venous resection over time accounts for a heterogeneous population of patients from which these conclusions are drawn. Going forward, clinical trials must utilize current preoperative cross-sectional imaging criteria that allow for standardized classification of resectable and unresectable pancreatic tumors.

Even so, a category of borderline resectable tumors remains a topic of controversy with regard to best treatment strategy. Patients with borderline resectable tumors are at high risk for margin-positive (R1/R2) resections and may benefit from a neoadjuvant treatment strategy designed to achieve objective tumor regression and subsequent complete resection. Pre-operative radiographic criteria for determining resectability have been accepted as a means of identifying patients who might benefit from neoadjuvant therapy.[10] The rationale offered for a neoadjuvant treatment strategy in patients with borderline respectable or unresectable disease is (a) the so-called “biologic test” to gauge the aggressiveness of the tumor, (b) treatment of micrometastatic disease, (c) administration of chemotherapy in the neoadjuvant setting may be better tolerated than postoperative therapy and (d) the potential for tumor destruction to maximize the potential for a complete (R0) resection. This determined approach to patients with advanced disease may yield improved survival due to an effective neoadjuvant strategy and the application of well-defined pre-operative resection criteria.

Morganti and colleagues performed a review of thirteen studies evaluating patients undergoing surgery following neoadjuvant chemoradiotherapy for unresectable pancreatic cancer.[47] The resection rates in these studies ranged from 8.3% to 64.2%, while the median survival ranged from 9 to 23 months. The rate of surgical exploration following neoadjuvant therapy ranged from 25% to 67%, and included patients without a radiographic response to treatment. Comparatively, the rate of re-exploration in the largest series of regional chemotherapy by Aigner et al. was 28% (74/265).[28] The rate of resection was 36% (27/74) for those undergoing re-exploration, and 10% (27/265) for the entire cohort. Without resectability clearly defined prior to regional chemotherapy, it is unclear if patients that ultimately underwent re-exploration and resection were more likely to have had borderline resectable versus unresectable tumors on pre-treatment imaging. While this potentially confounds the interpretation of rates of conversion to resectable status, these results reinforce the concept that regional chemotherapy, similar to neoadjuvant chemoradiotherapy, may provide benefit to a subset of patients currently not offered tumor resection.

Progressive surgical techniques combined with current neoadjuvant chemoradiotherapy strategies have already yielded emerging support for a multimodality approach to treatment. The treatment algorithm advocated by Katz et al. has resulted in the highest reported 5-year survival rates likely due to tumor response to neoadjuvant therapy and improved patient selection afforded by precise pre-operative selection criteria.[48] Furthermore, the belief that vascular invasion is a harbinger of poorer survival has been countered with mounting evidence that en bloc vascular resection for locally advanced pancreatic cancer matches outcomes for standard pancreaticoduodenectomy. In a study by Yekebas and colleagues, patients undergoing pancreatectomy with en bloc vascular resection compared to pancreatectomy without vascular resection had equivalent perioperative mortality and median overall survival.[49] This convergence of data supporting multimodality adjuvant therapy and progressive surgical technique highlights the attributes of patient selection, standard operative approach and routine use of multimodality therapy. Thus, the addition of radiation therapy to regional chemotherapy has the potential to improve further tumor response and resectability conversion rates and warrants consideration.

This review is encumbered by additional factors such as the heterogeneity of reports, inconsistent use of standard chemotherapeutic agents (i.e. gemcitabine), ill-defined inclusion and exclusion criteria, and non-standardized chemotherapy administration techniques. The inclusion of data for patients receiving regional chemotherapy after pancreatic resection, although representing less than one quarter of all patients in this review, potentially confounds the interpretation of results by introducing a subset of patients for whom the trials were not necessarily meant to address. Future studies should address separately neoadjuvant and adjuvant regional chemotherapy, as these represent two distinct strategies. Similarly, the inclusion of patients with stage IV pancreatic cancer deserves special consideration. Although the future application of this treatment strategy will likely be directed to patients with unresectable disease due solely to advanced T stage, the data related to toxicity, survival and tumor response gathered from patients with metastases has worth. Therefore these patients were not considered for exclusion from this review because of the value they provide to developing future investigation of regional chemotherapy.

Conclusion

The unacceptably high death rate due to pancreatic cancer is attributed to aggressive tumor biology, late stage diagnosis and relatively in-effective systemic therapy. Without the ability to affect tumor biology, the current approach to improving patient survival is through timely diagnosis and discovery of meaningful targeted therapy. Until then, complete tumor resection offers patients the only chance at long-term survival. Regional chemotherapy may provide patients currently without the option of resection that chance. Within the past two decades patients have been treated safely with regional chemotherapy in the setting of advanced, unresectable pancreatic cancer. In the modern era, novel treatment strategies for pancreatic cancer should be pursued in the setting of prospective clinical trials. Therefore, further investigation of regional chemotherapy is warranted with the goals of minimizing toxicity through the use of relevant agents and increasing treatment efficacy as measured by tumor response and increased rates of tumor resection.

Supplementary Material

1
2

Acknowledgments

Source of Funding

Intramural funding through Center for Cancer Research, National Cancer Institute, NIH.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Conflicts of Interest

No conflicting interests to report.

References

  • 1.Jemal A, Siegel R, Xu J, Ward E. Cancer statistics, 2010. CA: a cancer journal for clinicians. 2010;60:277–300. doi: 10.3322/caac.20073. [DOI] [PubMed] [Google Scholar]
  • 2.Sener SF, Fremgen A, Menck HR, Winchester DP. Pancreatic cancer: a report of treatment and survival trends for 100,313 patients diagnosed from 1985–1995, using the National Cancer Database. J Am Coll Surg. 1999;189:1–7. doi: 10.1016/s1072-7515(99)00075-7. [DOI] [PubMed] [Google Scholar]
  • 3.Oettle H, Post S, Neuhaus P, et al. Adjuvant chemotherapy with gemcitabine vs observation in patients undergoing curative-intent resection of pancreatic cancer: a randomized controlled trial. Jama. 2007;297:267–277. doi: 10.1001/jama.297.3.267. [DOI] [PubMed] [Google Scholar]
  • 4.Moore MJ, Goldstein D, Hamm J, et al. 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;25:1960–1966. doi: 10.1200/JCO.2006.07.9525. [DOI] [PubMed] [Google Scholar]
  • 5.Poplin E, Feng Y, Berlin J, et al. Phase III, randomized study of gemcitabine and oxaliplatin versus gemcitabine (fixed-dose rate infusion) compared with gemcitabine (30-minute infusion) in patients with pancreatic carcinoma E6201: a trial of the Eastern Cooperative Oncology Group. J Clin Oncol. 2009;27:3778–3785. doi: 10.1200/JCO.2008.20.9007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Yip D, Karapetis C, Strickland A, et al. Chemotherapy and radiotherapy for inoperable advanced pancreatic cancer. Cochrane Database Syst Rev. 2006;3:CD002093. doi: 10.1002/14651858.CD002093.pub2. [DOI] [PubMed] [Google Scholar]
  • 7.Key C. Cancer of the Pancreas. SEER Survival Monograph: Cancer Survival Among Adults: US, SEER Program, 1988–2001. In: Ries LAG YJ, Keel GE, Eisner MP, Lin YD, Horner M-J, editors. Patient and Tumor Characteristics. Bethesda, MD: Edition National Cancer Institute; 2007. SEER Program, NIH Pub. No. 07-6215. [Google Scholar]
  • 8.Edge SB, Byrd DR, Compton CC, et al. AJCC Cancer Staging Manual. New York: Springer; 2010. [Google Scholar]
  • 9.Callery MP, Chang KJ, Fishman EK, et al. Pretreatment assessment of resectable and borderline resectable pancreatic cancer: expert consensus statement. Ann Surg Oncol. 2009;16:1727–1733. doi: 10.1245/s10434-009-0408-6. [DOI] [PubMed] [Google Scholar]
  • 10.Varadhachary GR, Tamm EP, Abbruzzese JL, et al. Borderline resectable pancreatic cancer: definitions, management, and role of preoperative therapy. Ann Surg Oncol. 2006;13:1035–1046. doi: 10.1245/ASO.2006.08.011. [DOI] [PubMed] [Google Scholar]
  • 11.Collins JM. Pharmacologic rationale for regional drug delivery. J Clin Oncol. 1984;2:498–504. doi: 10.1200/JCO.1984.2.5.498. [DOI] [PubMed] [Google Scholar]
  • 12.Creech O, Jr, Krementz ET, Ryan RF, Winblad JN. Chemotherapy of cancer: regional perfusion utilizing an extracorporeal circuit. Ann Surg. 1958;148:616–632. doi: 10.1097/00000658-195810000-00009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Cornett WR, McCall LM, Petersen RP, et al. Randomized multicenter trial of hyperthermic isolated limb perfusion with melphalan alone compared with melphalan plus tumor necrosis factor: American College of Surgeons Oncology Group Trial Z0020. J Clin Oncol. 2006;24:4196–4201. doi: 10.1200/JCO.2005.05.5152. [DOI] [PubMed] [Google Scholar]
  • 14.Park BJ, Alexander HR, Libutti SK, et al. Treatment of primary peritoneal mesothelioma by continuous hyperthermic peritoneal perfusion (CHPP) Ann Surg Oncol. 1999;6:582–590. doi: 10.1007/s10434-999-0582-6. [DOI] [PubMed] [Google Scholar]
  • 15.Markman M, Bundy BN, Alberts DS, et al. Phase III trial of standard-dose intravenous cisplatin plus paclitaxel versus moderately high-dose carboplatin followed by intravenous paclitaxel and intraperitoneal cisplatin in small-volume stage III ovarian carcinoma: an intergroup study of the Gynecologic Oncology Group, Southwestern Oncology Group, and Eastern Cooperative Oncology Group. J Clin Oncol. 2001;19:1001–1007. doi: 10.1200/JCO.2001.19.4.1001. [DOI] [PubMed] [Google Scholar]
  • 16.Yan TD, Black D, Savady R, Sugarbaker PH. Systematic review on the efficacy of cytoreductive surgery combined with perioperative intraperitoneal chemotherapy for peritoneal carcinomatosis from colorectal carcinoma. J Clin Oncol. 2006;24:4011–4019. doi: 10.1200/JCO.2006.07.1142. [DOI] [PubMed] [Google Scholar]
  • 17.Sugarbaker PH, Alderman R, Edwards G, et al. Prospective morbidity and mortality assessment of cytoreductive surgery plus perioperative intraperitoneal chemotherapy to treat peritoneal dissemination of appendiceal mucinous malignancy. Ann Surg Oncol. 2006;13:635–644. doi: 10.1245/ASO.2006.03.079. [DOI] [PubMed] [Google Scholar]
  • 18.Fiorentini G, Poddie D, Ricci S, et al. Intra-aortic stop-flow infusion (IASFI) with hypoxic abdominal perfusion (HAP) in UICC stage III/IV pancreatic carcinoma (PC): report of a phase II study. Regional Cancer Treatment. 1996;9:88–91. [Google Scholar]
  • 19.Muchmore JH, Preslan JE, George WJ. Regional chemotherapy for inoperable pancreatic carcinoma. Cancer. 1996;78:664–673. doi: 10.1002/(SICI)1097-0142(19960801)78:3<664::AID-CNCR47>3.0.CO;2-U. [DOI] [PubMed] [Google Scholar]
  • 20.Link KH, Gansauge F, Gorich J, et al. Palliative and adjuvant regional chemotherapy in pancreatic cancer. European journal of surgical oncology : the journal of the European Society of Surgical Oncology and the British Association of Surgical Oncology. 1997;23:409–414. doi: 10.1016/s0748-7983(97)93720-3. [DOI] [PubMed] [Google Scholar]
  • 21.Lorenz M, Petrowsky H, Heinrich S, et al. Isolated hypoxic perfusion with mitomycin C in patients with advanced pancreatic cancer. European journal of surgical oncology : the journal of the European Society of Surgical Oncology and the British Association of Surgical Oncology. 1998;24:542–547. doi: 10.1016/s0748-7983(98)93572-7. [DOI] [PubMed] [Google Scholar]
  • 22.Maurer CA, Borner MM, Lauffer J, et al. Celiac axis infusion chemotherapy in advanced nonresectable pancreatic cancer. International journal of pancreatology : official journal of the International Association of Pancreatology. 1998;23:181–186. doi: 10.1007/BF02788395. [DOI] [PubMed] [Google Scholar]
  • 23.Klapdor R, Seutter E, Lang-Polckow EM, et al. Locoregional/systemic chemotherapy of locally advanced/metastasized pancreatic cancer with a combination of mitomycin-C and gemcitabine and simultaneous follow-up by imaging methods and tumor markers. Anticancer research. 1999;19:2459–2469. [PubMed] [Google Scholar]
  • 24.Homma H, Doi T, Mezawa S, et al. A novel arterial infusion chemotherapy for the treatment of patients with advanced pancreatic carcinoma after vascular supply distribution via superselective embolization. Cancer. 2000;89:303–313. [PubMed] [Google Scholar]
  • 25.Bayar S, Unal E, Tez M, et al. Regional chemotherapy for advanced pancreatic carcinoma. Hepato-gastroenterology. 2003;50:550–552. [PubMed] [Google Scholar]
  • 26.Ohigashi H, Ishikawa O, Yokayama S, et al. Intra-arterial infusion chemotherapy with angiotensin-II for locally advanced and nonresectable pancreatic adenocarcinoma: further evaluation and prognostic implications. Annals of surgical oncology. 2003;10:927–934. doi: 10.1245/aso.2003.10.021. [DOI] [PubMed] [Google Scholar]
  • 27.van IJken MG, van Etten B, Guetens G, et al. Balloon catheter hypoxic abdominal perfusion with Mitomycin C and Melphalan for locally advanced pancreatic cancer: a phase I–II trial. European journal of surgical oncology : the journal of the European Society of Surgical Oncology and the British Association of Surgical Oncology. 2004;30:671–680. doi: 10.1016/j.ejso.2004.03.016. [DOI] [PubMed] [Google Scholar]
  • 28.Aigner KR, Gailhofer S. Celiac axis infusion and microembolization for advanced stage III/IV pancreatic cancer--a phase II study on 265 cases. Anticancer Res. 2005;25:4407–4412. [PubMed] [Google Scholar]
  • 29.Takamori H, Kanemitsu K, Tsuji T, et al. 5-fluorouracil intra-arterial infusion combined with systemic gemcitabine for unresectable pancreatic cancer. Pancreas. 2005;30:223–226. doi: 10.1097/01.mpa.0000158025.46172.ae. [DOI] [PubMed] [Google Scholar]
  • 30.Barletta E, Fiore F, Daniele B, et al. Second-line intra-arterial chemotherapy in advanced pancreatic adenocarcinoma. Frontiers in bioscience : a journal and virtual library. 2006;11:782–787. doi: 10.2741/1836. [DOI] [PubMed] [Google Scholar]
  • 31.Mambrini A, Sanguinetti F, Pacetti P, et al. Intra-arterial infusion of 5-fluorouracil, leucovorin, epirubicin and carboplatin (FLEC regimen) in unresectable pancreatic cancer: results of a ten-year experience. In Vivo. 2006;20:751–755. [PubMed] [Google Scholar]
  • 32.Meyer F, Gebauer T, Grote R, et al. Results of regional chemotherapy using the aortic stop-flow technique in advanced pancreatic carcinoma. Surgery today. 2006;36:155–161. doi: 10.1007/s00595-005-3119-z. [DOI] [PubMed] [Google Scholar]
  • 33.Guadagni S, Clementi M, Valenti M, et al. Hypoxic abdominal stop-flow perfusion in the treatment of advanced pancreatic cancer: a phase II evaluation/trial. European journal of surgical oncology : the journal of the European Society of Surgical Oncology and the British Association of Surgical Oncology. 2007;33:72–78. doi: 10.1016/j.ejso.2006.10.042. [DOI] [PubMed] [Google Scholar]
  • 34.Ikeda O, Tamura Y, Nakasone Y, et al. Comparison of intrahepatic and pancreatic perfusion on fusion images using a combined SPECT/CT system and assessment of efficacy of combined continuous arterial infusion and systemic chemotherapy in advanced pancreatic carcinoma. Cardiovascular and interventional radiology. 2007;30:912–921. doi: 10.1007/s00270-007-9134-2. [DOI] [PubMed] [Google Scholar]
  • 35.Ishikawa T, Kamimura H, Tsuchiya A, et al. Clinical efficacy of intra-arterial pharmacokinetic chemotherapy with 5-fluorouracil, CDDP, gemcitabine, and angiotensin-II in patients with advanced pancreatic cancer. Hepato-gastroenterology. 2007;54:2378–2382. [PubMed] [Google Scholar]
  • 36.Milandri C, Calzolari F, Passardi A, et al. Intra-arterial chemotherapy of advanced pancreatic cancer: a single center experience. Hepato-gastroenterology. 2007;54:2373–2377. [PubMed] [Google Scholar]
  • 37.Nakchbandi W, Muller H, Singer MV, et al. Prospective study on warfarin and regional chemotherapy in patients with pancreatic carcinoma. Journal of gastrointestinal and liver diseases : JGLD. 2008;17:285–290. [PubMed] [Google Scholar]
  • 38.Sasada T, Denno R, Tanaka T, et al. Intra-arterial infusion chemotherapy with 5-fluorouracil and cisplatin in advanced pancreatic cancer: a feasibility study. American journal of clinical oncology. 2008;31:71–78. doi: 10.1097/COC.0b013e31807a328c. [DOI] [PubMed] [Google Scholar]
  • 39.Mambrini A, Bassi C, Pacetti P, et al. Prognostic factors in patients with advanced pancreatic adenocarcinoma treated with intra-arterial chemotherapy. Pancreas. 2008;36:56–60. doi: 10.1097/mpa.0b013e31812e9672. [DOI] [PubMed] [Google Scholar]
  • 40.Saif MW. Pancreatic neoplasm in 2011: an update. JOP. 2011;12:316–321. [PubMed] [Google Scholar]
  • 41.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;15:2403–2413. doi: 10.1200/JCO.1997.15.6.2403. [DOI] [PubMed] [Google Scholar]
  • 42.Conroy T, Desseigne F, Ychou M, et al. FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. N Engl J Med. 2011;364:1817–1825. doi: 10.1056/NEJMoa1011923. [DOI] [PubMed] [Google Scholar]
  • 43.Crane CH, Winter K, Regine WF, et al. Phase II study of bevacizumab with concurrent capecitabine and radiation followed by maintenance gemcitabine and bevacizumab for locally advanced pancreatic cancer: Radiation Therapy Oncology Group RTOG 0411. J Clin Oncol. 2009;27:4096–4102. doi: 10.1200/JCO.2009.21.8529. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Willett CG, Czito BG, Bendell JC, Ryan DP. Locally advanced pancreatic cancer. J Clin Oncol. 2005;23:4538–4544. doi: 10.1200/JCO.2005.23.911. [DOI] [PubMed] [Google Scholar]
  • 45.Loyer EM, David CL, Dubrow RA, et al. Vascular involvement in pancreatic adenocarcinoma: reassessment by thin-section CT. Abdom Imaging. 1996;21:202–206. doi: 10.1007/s002619900046. [DOI] [PubMed] [Google Scholar]
  • 46.Lu DS, Reber HA, Krasny RM, et al. Local staging of pancreatic cancer: criteria for unresectability of major vessels as revealed by pancreatic-phase, thin-section helical CT. AJR Am J Roentgenol. 1997;168:1439–1443. doi: 10.2214/ajr.168.6.9168704. [DOI] [PubMed] [Google Scholar]
  • 47.Morganti AG, Massaccesi M, La Torre G, et al. A systematic review of resectability and survival after concurrent chemoradiation in primarily unresectable pancreatic cancer. Ann Surg Oncol. 2010;17:194–205. doi: 10.1245/s10434-009-0762-4. [DOI] [PubMed] [Google Scholar]
  • 48.Katz MH, Wang H, Fleming JB, et al. Long-term survival after multidisciplinary management of resected pancreatic adenocarcinoma. Ann Surg Oncol. 2009;16:836–847. doi: 10.1245/s10434-008-0295-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Yekebas EF, Bogoevski D, Cataldegirmen G, et al. En bloc vascular resection for locally advanced pancreatic malignancies infiltrating major blood vessels: perioperative outcome and long-term survival in 136 patients. Ann Surg. 2008;247:300–309. doi: 10.1097/SLA.0b013e31815aab22. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

1
2

RESOURCES