Skip to main content
Erschienen in: Clinical and Translational Oncology 6/2017

Open Access 19.12.2016 | Special Article

Consensus guidelines for diagnosis, treatment and follow-up of patients with pancreatic cancer in Spain

verfasst von: M. Hidalgo, R. Álvarez, J. Gallego, C. Guillén-Ponce, B. Laquente, T. Macarulla, A. Muñoz, M. Salgado, R. Vera, J. Adeva, I. Alés, S. Arévalo, J. Blázquez, A. Calsina, A. Carmona, E. de Madaria, R. Díaz, L. Díez, T. Fernández, B. G. de Paredes, M. E. Gallardo, I. González, O. Hernando, P. Jiménez, A. López, C. López, F. López-Ríos, E. Martín, J. Martínez, A. Martínez, J. Montans, R. Pazo, J. C. Plaza, I. Peiró, J. J. Reina, A. Sanjuanbenito, R. Yaya, Alfredo Carrato

Erschienen in: Clinical and Translational Oncology | Ausgabe 6/2017

Abstract

The management of patients with pancreatic cancer has advanced over the last few years. We convey a multidisciplinary group of experts in an attempt to stablish practical guidelines for the diagnoses, staging and management of these patients. This paper summarizes the main conclusions of the working group. Patients with suspected pancreatic ductal adenocarcinoma should be rapidly evaluated and referred to high-volume centers. Multidisciplinary supervision is critical for proper diagnoses, staging and to frame a treatment plan. Surgical resection together with chemotherapy offers the highest chance for cure in early stage disease. Patients with advanced disease should be classified in treatment groups to guide systemic treatment. New chemotherapeutic regimens have resulted in improved survival. Symptomatic management is critical in this disease. Enrollment in a clinical trial is, in general, recommended.

Introduction

Pancreatic ductal adenocarcinoma (PDAC), the most common form of pancreatic cancer, currently stands as the third most common cause of cancer related deaths [1]. In 2008 a total of 70,000 of were diagnosed in Europe [2, 3]. Rates of new cases of pancreatic cancer have increased on average 0.8% annually over the last ten years. The current 5- and 10-year survival rates at 7.2% and below 4%, respectively [4].
The aim of the present consensus guidelines is to provide a general overview of the diagnosis, a more global patient´s classification, not just based on performance status, treatment, and management of associated complications of patients with PDAC. These guidelines are the result of expert consensus meetings that took place during the months of September to December 2015 sponsored by Fundación ECO, where a total of forty-two medical oncologists, radiotherapists, surgeons, radiologists, pathologists, endocrinologists, gastrointestinal specialists and palliative care specialists shared their opinions. These recommendations are based on the results of clinical trials, retrospective, observational studies, as well as the group of experts´ opinion (levels of evidence: quality of evidence: I–III; strength of recommendation: A–E) [5].

PDAC: signs and symptoms

The absence of specific manifestations, together with its biological aggressiveness, results in delayed diagnosis in more than 80% of cases. The most common symptoms include fatigue, anorexia, weight loss, abdominal pain and dark urine [6]. Sixty to 70% of tumors originate in the pancreatic head, 20–25% in the body and tail and in 10–20% there is a diffuse involvement of the gland [7]. While tumors in the pancreatic head tend to be diagnosed at earlier stages because of the jaundice associated with bile duct obstruction, tumors in the body and tail are usually detected in advanced stages. Tumor with obstructive jaundice may be associated with palpable gallbladder (Courvoisier sign). Head tumors may also be associated with steatorrhea as a consequence of exocrine pancreatic insufficiency and obstruction of the pancreatic duct. Although with low specificity, the combination of diabetes mellitus (DM) of recent onset associated with weight loss should lead to suspicion PDAC [8]. About one in 125 (0.85%) patients with new-onset DM presents PDAC, eight times more than expected, compared to the general population [9]. Patients with newly diagnosed DM without metabolic syndrome and difficult glycemic control should be evaluated to rule out PDAC [10] (IIB). Special attention should be paid to patients that present the following symptoms: weight loss, abdominal pain, DM of recent onset or with poor disease control. Patients with advanced stages of PDAC may have epigastric palpable mass, hepatomegaly secondary to liver metastases, ascites caused by peritoneal carcinomatosis, pain, vascular and nerve infiltration and/or gastric outlet obstruction. The PDAC is associated with arterial and venous (hypercoagulable state) thrombosis such as Trousseau syndrome (migratory superficial vein thrombosis. PDAC may also be associated with palpable supraclavicular lymph nodes (Virchow), anterior axillary (Irish) or periumbilical mass (Sister Mary Joseph).

Familial PDAC and hereditary PDAC

The International Cancer of the Pancreas Consortium Screening (CAPS) recommends screening of PDAC in high-risk individuals (HRI) or families with familial pancreatic cancer (FPC), although this is not yet included in routine clinical practice [11] (IIIC). PDAC screening programs should be performed in high-volume centers that have multidisciplinary teams with experience in the screening and treatment of the disease. HRI are considered as those who have at least 5–10 times increased risk of PDAC [11] and include families with at least two first-degree relatives affected, or ≥3 individuals regardless of the degree of kinship. In addition, subjects with Peutz–Jeghers syndrome, hereditary pancreatitis, or those affected by other hereditary cancer syndromes (germline mutations in the BRCA genes, familial melanoma or colorectal cancer hereditary) with a case of PDAC in the family [12].
The best screening techniques for HRI is magnetic resonance imaging (MRI) with magnetic resonance cholangiopancreatography (MRCP) and upper gastrointestinal endoscopic ultrasound (EUS) [1115] (Algorithm 1). https://static-content.springer.com/image/art%3A10.1007%2Fs12094-016-1594-x/MediaObjects/12094_2016_1594_Figa_HTML.gif Screening tests aim to detect small pancreatic solid tumors (≤1 cm), irregular pancreatic duct, and pancreatic precursor lesions such as intraductal papillary mucinous neoplasm (IPMN), which are usually present as cysts [16]. The prevalence of suspected cystic pancreatic lesions is 33–45% in HRI [11, 15]. The CAPS recommend starting screening at age 50 years, or ten years before the age of the youngest case in the family [11] with annual exams if no pancreatic lesions are observed [11] (IIIB). These intervals could be modified at the discretion of the multidisciplinary tumor committee. If suspicious abnormalities are detected, surgery is indicated although the current available evidence is limited [1116].

Diagnosis

In patients with suspected PDAC based on medical history and/or physical exam, the primary diagnostic approach to the patient with PDAC is radiological. An abdominal ultrasound is often the first test performed in patients with abdominal pain and/or jaundice. US can detect dilatation of the bile duct or pancreatic duct and the presence of a pancreatic mass. However, the sensitivity for detecting pancreatic tumors is low and ranges between 50 and 70% [17]. Computer tomography (CT) scan is the gold standard technique in the evaluation of patients with suspected PDAC (IIA). The study of a pancreatic mass must include a biphasic CT performed with an arterial phase (40–50 s) and portal venous phase (65–70 s) [7, 1719]. In most cases, PDAC is seen in the arterial phase as a hypodense pancreatic lesion, with poorly defined margins [7]. Furthermore, CT scan detects the vascular involvement (arterial and venous), extra-pancreatic local extension, presence of lymph nodes, and detection of liver or peritoneal nodules [7]. The availability of a high-quality multidetector CT (preferably ≥16 detectors) combined with experience in the interpretation of these studies has shown a more accurate preoperative staging and better patients´ management [18]. Magnetic resonance imaging (MRI) has equal sensitivity and specificity than CT for staging PDAC, but its use is not widespread because of its high cost and reduced availability and is usually reserved for difficult cases, cystic neoplasms of the pancreas and to explore biliary anatomy as well [18]. Radiology report should follow a standard format [18, 19].
Upper gastrointestinal endoscopy is useful to take biopsies of tumors infiltrating the duodenum as well as for palliative decompression of the duodenum and/or bile duct [7]. Endoscopic retrograde cholangiopancreatography (ERCP) is restricted to cases with obstruction of the bile duct because is associated with significant adverse effects and has low profitability of achieving a histological diagnosis (20%) [7]. Endoscopic ultrasound (EUS) is an important complementary examination in the diagnosis and staging of PDAC as it permits examining the primary tumors, relationship with neighboring structures as well as obtaining tissue for pathological diagnoses [19]. The sensitivity and specificity of EUS-guided tumor fine needle aspiration (FNA) are 90 and 98%, respectively [20]. It should be noted, however, that patients with clinical and radiological suspected PDAC with resectable do not need a pathological diagnoses before surgical resection (IIIB). Pathological diagnoses are needed in patients with atypical presentation and in those with locally advanced disease that are managed with chemotherapy and/or radiation therapy (IIIB).

Pathological diagnoses and classification

In the presence of suspicious lesions that are resectable, a tumor biopsy prior surgery is not required. Likewise, because of the complexity to obtain it as well as the limitations on its interpretation an intraoperative biopsy is not required [7, 21] (IIIB). A pathological diagnosis is always required in patients with unresectable or borderline lesions to be managed with chemotherapy and/or radiation therapy as well as in patients with amenable to treatment with metastatic disease [21] (IIIB).
The technique of choice to obtain tissue for pathological diagnoses depends on the location of the lesion and the stage of the disease. For primary tumors, a EUS-FNA is the safest procedure with highest sensitivity and specificity that can also provide additional staging information [22]. In patients with metastatic liver disease, a percutaneous US- or CT-guided biopsy is the procedure of choice [7, 21] (IIIB). If a biopsy does not confirm malignancy, it should be repeated atleast once. If during surgery the tumor is unresectable, histological diagnosis should be made (IIIB).
Table 1 summarizes the histopathologic classification of PDAC tumors. The most common type (95%) is ductal adenocarcinoma of the pancreas, referred to as pancreatic cancer. These tumors originate in ductal epithelium, have glandular differentiation, may produce mucin, and are associated with a significant desmoplastic reaction. There are no specific diagnostic markers to differentiate PDAC from other adenocarcinomas though reactive glands, a CK7+/CK20− profile, while not specific, supports a pancreatic origin.
Table 1
Pathological diagnosis
Classification of pancreatic tumors (WHO 2010 classification)
Benign
Acinar cell cystadenoma
Serous cystadenoma
Premalignant lesions
Pancreatic intraepithelial neoplasia type 3 (PanIN-3)
Intraductal papillary mucinous neoplasm with low or intermediate grade dysplasia
Intraductal papillary mucinous neoplasms
High-grade dysplasia
Tubulo-papillary intraductal neoplasia
Mucinous cystic dysplasia neoplasia with low or intermediate grade
Mucinous cystic neoplasm with high-grade dysplasia
Malignant
Ductal adenocarcinoma
Adenosquamous carcinoma
Colloid carcinoma (non-cystic mucinous carcinoma)
Hepatoid carcinoma
Medullary carcinoma
Cell carcinoma signet ring
Undifferentiated carcinoma
Undifferentiated carcinoma with osteoclast-like giant cells
Acinar cell carcinoma
Cystoadenocarcinoma acinar cells
Intraductal papillary mucinous neoplasm or associated with invasive carcinoma
Mixed acinar-ductal carcinoma
Mixed acinar-neuroendocrine carcinoma
Mixed-neuroendocrine carcinoma acinar-ductal
Mixed ductal-neuroendocrine carcinoma
Mucinous cystic neoplasm associated with invasive carcinoma
Pancreatoblastoma
Serous cystoadenocarcinoma
Pseudopapillary or solid neoplasia
Neuroendocrine neoplasms
 Pancreatic neuroendocrine microadenoma
 Neuroendocrine tumor (NET)
Pancreatic, not functioning G1, G2 NET
G1 NET
G2 NET
 Neuroendocrine carcinoma (NEC)
Large cell NEC
Small cell NEC
 NET serotonin producer (carcinoid)
 Gastrinoma
 Glucagonoma
 Insulinoma
 Somatostatinoma
 VIPoma
Mature teratoma
Mesenchymal tumors
Lymphomas
Metastasis

Biomarkers

Although many biomarkers have been studied for PDAC, only carbohydrate antigen 19.9 (CA 19.9) has proven useful and therefore, it is the only biomarker routinely used [23] (IA). For diagnoses, it has a sensitivity ranging between 70 and 92% and a specificity of 68–92% depending on tumor size. False positive results are associated with benign diseases such as pancreatitis, cirrhosis, acute cholangitis, and other diseases causing of cholestasis. Determining CA 19.9 is a complementary test in the diagnoses and management of PDAC [24]. In patients with resectable disease, plasma levels >100 U/mL values predict the presence of occult metastatic disease. In advanced disease, elevated CA 19.9 is considered as unfavorable prognostic factor [25, 26]. However, given its low positive predictive value, particularly in asymptomatic individuals, is not recommended as a screening marker [27].

Timeline for diagnosis and management

Because PDAC is very aggressive, it is recommended to reduce the diagnostic time as much as possible to rapidly initiate treatment [28]. In a study in patients with advanced PDAC it was estimated that the average doubling time of pancreatic tumor was 40–60 days [29]. A month from the onset of the first symptoms or suspicious signs is a reasonable goal (IIIB).
The National Cancer Strategy Health System (2010) states that the time from therapeutic decision until the actual start of treatment should be less than 2 weeks for surgical treatment, one week for chemotherapy and 4 weeks (including treatment planning) for radiation therapy [30] (IIIB). Strategies to minimize time to diagnoses include raising awareness in the general population and among health professionals, rapid diagnostic protocols for patients with suspected lesion in primary care settings as well as preferential referral pathways to specialize care and treatment centers. PDAC, like any other complex tumor, should be managed in high referral centers by multidisciplinary teams: Medical oncologists, pathologists, radiotherapist, GI specialist, surgeons and radiologists [30] (IIIB).

Staging and disease classification

Pancreatic cancer can be staged, based on imaging and pathological studies, in stages as per the TNM classification [31, 32]. However, from a management perspective, patients are better classified based on the extension of disease in resectable, borderline resectable, locally advanced unresectable and metastatic (IIIA).
In addition, from a treatment perspective, patients with locally advanced/metastatic disease are further classified as: (a) candidate to chemotherapy treatment without limitations; (b) candidate for chemotherapy with limitations and (c) not candidate for treatment with chemotherapy (Table 2) [6, 3337] (IIIB).
Table 2
Patients’ classification, according to treatment perspective (IIIB)
Patients’ classification
Factors
Patient suitable for chemotherapy treatment without limitations
The presence of ALL the following factors
 ECOG 0–1
 Age ≤75 years
 Bilirubin ≤1.5 ULN
 Good nutritional status (serum albumin >2.5 mg/dl, weight lost <10% over the last 3–6 months and BMI >20 kg/m2)
 Lack of co-morbidities
Patient suitable for chemotherapy with limitations
The presence of AT LEAST ONE of the following factors
 ECOG 2 (which can lead to KPS 70%)
 Age >75 years
 Mild to moderate neurological or endocrine-metabolic organ dysfunction; in case of liver dysfunction, hyperbilirubinemia >1.5 × ULN (once optimized if obstructive causes are present, for example with biliary stent) marks the degree of dysfunction. It is considered appropriate to adjust the dose, for example, using GEM at 600-800 mg/m2 and nab paclitaxel 75–100 mg/m2) [37]
 Cardiac dysfunction, especially a recent ischemic event; acute, symptomatic, severe TED such as PE with hemodynamic instability or DVT with risk and limb amputation [38]
BMI <20 kg/m2 or >10% weight loss in 3–6 months
Patient not suitable for chemotherapy treatment
The presence of AT LEAST ONE of the following factors
 ECOG 3-4 (which may result in KPS ≤ 60%). Active treatment will be initiated in patients with ECOG 3 secondary to the disease (not to their previous comorbidities) without any severe organ dysfunction, thus moving this subgroup of patients to the “candidate for chemotherapy treatment with limitations” group
 Severe organ dysfunction: neurological (e.g., severe cognitive impairment, Alzheimer’s type); endocrine-metabolic, infectious (uncontrolled HIV), renal, hepatic dysfunctions, etc
ECOG Eastern Cooperative Oncology Group, ULN upper normal limit, BMI body mass index, GEM gemcitabine, KPS Karnofsky performance status, TED thromboembolic disease, PE pulmonary embolism, DVT deep venous thrombosis

Practical considerations in treatment decision process

The treatment plans for patients with PDAC patients should be made individually. A complete staging process is essential to determine the extent of the tumor that drives treatment plan and prognosis. In parallel, patient´s status, which is linked to its ability to tolerate an aggressive treatment, should be defined. This includes the functional status as determine by the Karnofsky Performance Scale (KPS) and/or the Eastern Cooperative Oncology Group (ECOG). Patients with KPS of less than 60–70% or ECOG less than 0–1 are limited to receive aggressive chemotherapy. For elderly patients, it is also advisable to use geriatric scales such as the Barthel scale that assesses the degree of autonomy in basic activities of daily living [37]. The assessment of nutritional status as measured by physical exam (weight, body mass index, presence of edema), recent weight lost (>10% over 6 months); plasma protein levels (albumin, prealbumin, transferrin) is crucial [38]. Validated nutritional scales such as Mini Nutritional Assessment are useful in this regard. In addition, a life expectancy of >3 months is usually needed to administer cancer treatment. Mechanical problems caused by tumor masses such as bile duct and bowel obstruction need to be assessed and corrected prior to treatment commencement. Finally, patient priorities and preferences need to be considered (IIIA).

Treatment approaches

Resectable disease/borderline resectable disease

Neoadjuvant treatment

Neoadjuvant treatment, which is the treatment with chemotherapy and/or radiotherapy administered before surgical resections, aims to increase overall survival by increasing the rate of R0 resection and early treatment of micrometastatic disease. In addition, preoperative treatment may lead to avoiding unnecessary surgical resection in patients with aggressive tumors that develop early progression.
It should be noted, however, that there are no randomized phase 3 studies to support any of these assumptions. Prior studies suggest an increment in the rate of R0 resections [3941]. Most studies reported thus far were conducted with old, less effective chemotherapy regiments and the data available with modern regimens [gemcitabine (GEM)/nab paclitaxel, FOLFIRINOX], came from single-center trials [35, 4247].
Here, we discuss preoperative management of patients with resectable or borderline resectable disease. Prior to treatment initiation, it is important to have pathological diagnosis as well as normalized bile duct drainage. Endoscopically placement of a metal stent is the procedure of choice to palliate obstructive jaundice (IIIB).
For patients with resectable disease neoadjuvant treatment cannot be recommended outside a clinical trial. However, preoperative treatment is one of the available approaches in patients with borderline resectable disease (IIB). The chemotherapy treatments used should be those associated with higher response rate in patients with metastatic disease (GEM/nab paclitaxel, FOLFIRINOX) [35, 46] (IIB). Currently there is no evidence to recommend one versus the other and the decision should be based on patients´ characteristics and center experience. In general, treatment should be administered for 3–4 months with a reassessment and multidisciplinary discussion afterwards (IIB). Patients with responding tumors by either radiological criteria or CA 19.9 could proceed to surgical resection [48, 49] (IIB). Radiotherapy the alone is not recommended and should be combined with either fluoropyrimidines or GEM (IIB). IMRT is associated with reduced toxicity and should be used when available. Patients who receive chemo-radiation should wait four to eight weeks before attempting surgical resection (IIB).
Radiological evaluation must be conducted after neoadjuvant treatment. Lack of objective radiological response should not be a criterion to rule out surgical resection [52] (IIB). Those patients with suspected disease progression by either elevated CA 19.9 without radiological evidence of disease progression should be carefully evaluated and PET scan and laparoscopy should be considered (IIB). Patients with documented metastatic progression are not candidates for surgery and should be managed as such (IIB).

Surgical treatment

An R0 surgical resection is the only curative treatment for patients with pancreas cancer and should always be attempted. Prior to considering surgery, patients need to be assessed by a multidisciplinary team and classified as resectable, borderline resectable or unresectable locally advanced being the multidetector CT scan the radiological procedure of choice for this matter [18, 50] (IIA). Based on the extent of the tumor, involvement of blood vessels [portal vein, superior mesenteric vein (SMV); superior mesenteric artery (SMA); celiac trunk and hepatic artery] patients are classified in one of the above-mentioned group [31, 5155]. Table 3 provides the specific criteria [57]. More recent classifications also include changes induced by preoperative treatments. It should be noted that extension to adjacent organs, if resectable, is not a contraindication for surgery.
Table 3
Resectability criteria [57]
Category
Arterial
Venous
Resectable
Absence of tumoral contact with CT, MSA or CHA
Absence of tumoral contact with SMV or PV or contact ≤180° without irregularities in the venous contour
Borderline resectable
Head of the pancreas and uncinate process
 Solid tumoral contact with CHA, without extension to CT or HA bifurcation, that allows resection and complete and safe vascular reconstruction
 Solid tumoral contact with SMA ≤180°
 The presence of arterial anatomic variants should be evaluated (i.e., right accessory HA, replacement of right HA, replacement of CHA as well as source of replaced or accessory artery) and the presence and degree of tumoral contact due to their influence when planning the surgical procedure
Body and tail of the pancreas
 Solid tumoral contact with CT ≤ 180°
 Solid tumoral contact with CT > 180° without aorta involvement and with intact GDA (there is no consensus on this criteria and can be included in the non-resectable category)
Solid tumoral contact with SMV or PV >180°, contact ≤180° with irregularities in the venous contour or venous thrombosis but with adequate proximal and distal ends that allow safe vascular resection and replacement
 Solid tumor contact with IVC
Non-resectable
Distant metastasis (including metastasis in non-regional lymph nodes)
Head of the pancreas and uncinate process
 Solid tumoral contact with SMA or CT >180°
 Solid tumoral contact with the first jejunal branch of SMA
Body and tail of the pancreas
 Solid tumoral contact with SMA or CTC > 180°
 Solid tumoral contact with CT and aortic infiltration
Head of the pancreas and uncinate process
 Tumoral infiltration or thrombosis (thrombosis may not be tumoral) in SMV or PV, that does not allow reconstruction
 Contact with the most proximal jejunal vein that drains in SMV
Body and tail of the pancreas
 Tumoral infiltration or thrombosis (the thrombosis may not be tumoral) in SMV or PV, that does not allow reconstruction
CT celiac trunk, SMA superior mesenteric artery, CHA common hepatic artery, SMV superior mesenteric vein, PV portal vein, IVC inferior vena cava inferior, GDA gastroduodenal artery
In addition to stage classifications, a complete assessment of operative risk should be performed. Considering the high morbimortality of pancreatic cancer resection, its assessment is of great importance. Classic surgical risk scales, such as Apache, ASA and POSSUM, do not predict accurately the morbidity after pancreatic surgery. Other more recent classification such as the one published by Braga, as well as the Preoperative Pancreatic Resection (PREPARE) and SOAR (Surgical Outcomes Analysis and Research) scores are based on the integration of multiple parameters and appear more accurate [5659].
Prior to surgical resection, it is critical to gain an adequate nutritional status and either nutritional supplements or even parenteral nutrition should be considered for 1–2 weeks prior to surgery in malnourished patients. In patients with large tumors, particularly of the tail of the pancreas, borderline resection and high tumor marker, a diagnostic laparoscopy should be considered prior to laparotomy.
For patients with tumors in the head of the pancreas, the procedure of choice is the duodenal pancreatectomy (Whipple procedure), which includes en bloc resection of the head of the pancreas, duodenum, gall bladder and bile duct, together with regional lymphadenectomy [57] (IA). Pylorus preserving pancreatectomy is equivalent to classic Whipple with regards to morbimortality and outcome and the selection of surgery type depends on surgeon preference. Other procedures such as extended pancreatectomy, total pancreatectomy, and extended lymphadenectomy are reserved for selected cases [60, 61].
Patients with tumors of the body or tail of the pancreas are treated with distal pancreatectomy.
As mentioned above, laparoscopy can detect small peritoneal implants or liver metastasis not visible by CT scan and is often used in patients with high risk of metastatic disease [62] (IIB). In addition, for patients with tumors in the body and tail of the pan, laparoscopy resection with or without robot assistance, is gaining acceptance [63]. Finally, vascular involvement has been traditionally considered a formal contraindication for resection [64, 65]. More recently, however, venous resection and reconstruction is accepted as an optimal surgical procedure and is not associated with worse prognosis. However, arterial resection and reconstruction is still considered an investigational approach [51, 57, 65].

Adjuvant treatment

Adjuvant treatment is recommended in patients who undergo an R0/R1 resection with a PT1-4/N0-1M0, with an ECOG PS 0–1 and proper nutritional status [66, 67]. Treatment of patients with ECOG 2 needs to be individualized [66, 67].
It is recommended that adjuvant treatment is initiated within the next 12 weeks after surgery in patients who do not have any active infection, any serious postsurgical complication or presents with signs or symptoms of recurrent disease. There is no consensus on the adjuvant treatment in patients who have received neoadjuvant treatment [7, 6870]. Those patients need to be evaluated in a multidisciplinary board. In general, adjuvant treatment in this population is still considered investigational. As a general rule, patients who have received neoadjuvant treatment should receive adjuvant treatment to complete a total of six months of treatment (IIIB).
With regards to the role of radiation therapy, there is even less information and could be considered in those patients with positive margin providing was not administered in the preoperative period (IIC). Prior to adjuvant chemotherapy commencement patient needs to be evaluated with a CBC, chemistry, renal function test, albumin, LDH and CA 19.9 levels [7, 7173]. A CT of the chest, abdomen and pelvis is required to document lack of disease progression [18, 71, 73]. Currently, and until the results of ongoing studies are available, the recommended treatment is single agent GEM, or 5-FU and leucovorin (LV), for a total of six months [66, 67, 7476] (IA). The results of the recent clinical trial ESPAC-4 support the use of GEM + capecitabine in this setting [76] (IA). The role of radiation therapy is less defined and should be considered for patients with positive margins and in selected cases with lymph node positive disease [7582] (IC).
Those patients with positive margins and neoadjuvant treatment that did not include radiation therapy are a particularly suitable group for postoperative radiotherapy. Currently, there is no biomarker predictive of outcome in this patient population and CA 19.9 is the only prognostic indicator [83]. Once treatment is completed, patient should be followed every three months with measurement of CA 19.9 levels and a physical exam [7, 8385] (IIIB). A CT scan should be performed every 6 months during the first 2–3 years after surgery and yearly thereafter [7, 8385].

Unresectable disease

Management of patients with locally advanced disease

The management of patients with locally advanced disease is one of the most controversial areas in the treatment of PDAC due to the paucity of well controlled, randomized clinical trials. The goal in treating these patients is to improve survival which is better achieved if a complete surgical resection is feasible. Patients with locally advanced disease need to be evaluated like any other patient with PDAC with special attention to nutritional status, ECOG performance status, symptoms related to tumor local growth (pain, bowel and/or bile duct obstruction). The presence of bile duct or bowel obstruction need to be corrected before treatment is initiated. For patients who are candidates of chemotherapy treatment without limitations, this is in general the recommended approach (Table 2). While there are no data with regards to the most efficient regimen in this particular setting, current trend is to use either GEM-nab paclitaxel or FOLFILRINOX based on the data available for patients with advanced disease [35, 46, 86, 87] (IIB). Chemotherapy is usually administered for 3–4 months followed by assessment of tumor response. In patients with partial response that allows surgical resection, it could be a treatment option. In the remaining patients with partial response and those with stable disease, chemotherapy treatment as well as consolidation with chemotherapy and radiotherapy are valid options.
The recent data from the LAP007 study (Phase III study that compared chemoradiotherapy and chemotherapy in patients with a locally advanced pancreatic cancer controlled after four months of GEM with or without erlotinib) indicates that there are no survival benefits for chemo-radiation as compared to continuing chemotherapy alone, although it decreased the risk of local progression and improved PFS [88] (IA).
It should be noted, however, that this study used conventional external beam radiation and gemcitabine-based chemotherapy and the results cannot be extrapolated to those obtained with new chemotherapy regimens and more modern radiation techniques (IMRT). Data from the SCALOP trial (Phase II study of induction chemotherapy followed by GEM or capecitabine-based chemoradiation in locally advanced pancreatic cancer) suggest that the radiotherapy combined with fluoropyrimidines achieves better results than when combined with GEM [89] (IA). Patients who are candidates for chemotherapy with limitations have a very poor prognosis and should be managed with either single agent chemotherapy (GEM alone), combination therapy (GEM + nab paclitaxel) or radiation therapy alone (Table 2).

Management of patients with metastatic disease

First line treatment The management of patients with advanced PDAC is based on systemic chemotherapy. In those patients who have received prior adjuvant or neadjuvant treatment, rechallenge should be considered when the disease-free interval is ≥6 months. Prior to treatment initiation, patients should be classified based on performance status, nutritional status, age and comorbidities according to Table 2. For patients able to receive chemotherapy without limitations, the current standard of care is either GEM-nab paclitaxel or FOLFIRINOX [35, 46] (IA). In the lack of randomized studies comparing these two regiments no one can be recommended [90, 91]. FOLFIRINOX should not be administered to patients >75 years old. In addition, this regimen is associated with a higher incidence of toxicity and thromboembolic complications and often requires growth factor support. FOLFIRINOX should be used with caution in patients with biliary stents who have increased risk of biliary tract infections and sepsis. In addition, it is recommended to administer antiemetic prophylaxis at least for moderately emetogenic chemotherapy. Patients who are candidates to chemotherapy with limitations are best managed with GEM-nab paclitaxel, and should be administered until progression or unacceptable toxicity; this is particularly the case for patients with ECOG 2 secondary to high tumor volume in whom tumor reduction may result in symptomatic improvement (IIB). Patients with ECOG 2 secondary to comorbidities or those with severe peripheral neuropathy can be treated with GEM alone (IIIB). Patients who are not candidates for chemotherapy should receive palliative treatment. The optimal management should always be reassessed and modified if the condition of the patient changes. Thus, subjects whose condition improves should be considered for more aggressive approaches. In any group, enrollment in a clinical trial should always be the preferred option (Algorithm 2). https://static-content.springer.com/image/art%3A10.1007%2Fs12094-016-1594-x/MediaObjects/12094_2016_1594_Figb_HTML.gif
Second line treatment Second line treatment is in general recommended after progression to first line treatment [92] (IA). Treatment decision should be based on the general status of the patient as well as the first line treatment. For patients who have been treated with GEM based regimen FOLFOX chemotherapy has demonstrated improvement in survival as compared to 5-FU in the CONKO-003 study [93]. These results, however, have not been confirmed in the PANCREOX trial [94]. More recently, the NAPOLI-1 showed that MM-398 (liposomal formulation of irinotecan) in combination with 5-FU/LV is better than 5-FU/LV alone [95]. For patients who have received 5-FU/LV based chemotherapy on the first lie sett, there is very little data to base second line choices. In general, either GEM alone or GEM combination is recommended [96].
Treatment monitoring The response to treatment should be monitored every 8–12 weeks by a CT scan of the chest, abdomen and pelvis (IIIB). Other imaging modalities such as MRI and/or PET are not routinely recommended to be used in a serial basis (IIIB). The tumor marker CA 19.9 should be measured before treatment and every 4–8 weeks thereafter (IIIB). Tumor progression in patients with rising CA 19.9 should be confirmed radiologically [7, 35, 46, 97] (IIIB).

Supportive care

Supportive care aims to improve symptoms, reduce hospital admission and preserve quality of life. Proper symptomatic management is critical to allow administration of chemotherapy and radiotherapy. Symptomatic management should be accomplished in a multidisciplinary fashion. In this section, we will describe the most common approach to diagnosis and management of the most common symptoms of PADC.
1.
Bile duct obstruction.
 
Up to 75% of patients with tumors in the head of the pancreas develop bile duct obstruction which results in jaundice, itching cholangitis and hepatic dysfunction. If untreated, hepatic failure may ensure [98]. While surgical management had been the preferred approach in the past, particularly coledocoenterostomy, the high mortality of these techniques together with the excellent results obtained with endoscopic approaches has resulted in the preference of endoscopic management [99]. The preferred approach is endoscopic stent (via ERCP) [100]. Percutaneous transhepatic colangiopancreatography with stent placement is associated with higher risk of infectious complications and bleeding, being only recommended for patients in whom the endoscopic approach is not feasible [101]. Plastic stents have a life span of about four months and are only recommended for patients with expected short survival. Patients with longer expected survival should be treated with metal stents that have a longer functionality [102]. Surgical management is only recommended for patients who undergo a laparotomy for other reasons. Bile duct drainage is clearly recommended in patients who are scheduled for preoperative chemotherapy, those with cholangitis, or those in whom surgical resection is expected to be delayed. However, patients with moderate bilirubin elevation scheduled to undergo surgery can be safely operated without drainage [103105].
2.
Duodenal obstruction.
 
Ten to twenty-five per cent of patients develop duodenal obstruction which is associated with severe symptoms and deterioration of quality of life. The most common approach nowadays is the endoscopic placement of an expansible metallic stent. This approach results in over 90% success with very few complications. The preferred surgical treatment is a gastric jejunonostomy which is only recommended in very selected patients because of high morbidity and mortality when performed as a treatment modality [106]. Prophylactic gastrojejunostomy should be considered in patients with non-resectable tumors who undergo an exploratory laparotomy. Endoscopic stents are associated with rapid recovery of oral intake, less morbimortality and a shorter hospital stay [107, 108]. In contrast, surgical treatment is associated with better long term outcome. For this reason surgery is only considered for patients with expected long survival. All these recommendations achieve a level of evidence IIB.
3.
Pain.
 
Fifty to sixty per cent of patients with PADC develop some short of pain. These patients need intensive treatment, with both pharmacological and non-pharmacological approaches [109]. It is important to consider the precise cause of the pain, such as for example bowel obstruction, liver or bone metastasis, or secondary to chemotherapy (neuropathy, mucositis, enteritis) [110113]. Table 4 summarizes the most important approaches for pain management (IIA).
Table 4
Pain management strategies (IIA)
Mild pain
NSAIDs: Taking into account maximum doses and side effects (gastrointestinal bleeding, nephrotoxicity [110] acetaminophen
Moderate/severe pain
Opioids: any opioid as first choice, except for methadone (secondary choice). Methadone has great benefit in neuropathic pain due to its anti NMDA effect. Should be administered by trained personnel
Other treatments
 Adjuvant treatment/co-analgesics
Corticosteroids
Gabapentine (if neuropathic pain)
 Intrathecal catheters
To manage moderate to severe pain
Lower frequency of secondary adverse events, with better pain control [111]
First choice: hydromorphone, ziconotide, local anesthetic
Severe neuropathic pain: baclofen, clonidine
Miscellaneous (little evidence)
Other therapies: phentolamine, capsaicin, cryoablation, acupuncture
Radiotherapy
Indicated for management of refractory pain, especially in patients with good performance status and localized pain caused by isolated metastases or pancreas and adjacent structures involvement
Celiac plexus block [112]
Provides better analgesic control (benefit in >80% of the patients) and/or decreases the opioid dose when compared to standard analgesic treatment
Cause a disruption in the pain signaling by an average of 3 months
It can be performed via percutaneous under ultrasound control, surgical or endoscopic by ultrasound. In terms of technique, there isn’t enough evidence to make any recommendations Side effects are rare (transient hypotension, constipation or diarrhea)
There is no evidence to recommend the timing for the blockage (early, at diagnosis, or late when there is poor pain control)
There is no evidence that increases survival
In the clinical practice celiac plexus block is reserved for patients with poor pain control despite escalation with opioids or for those with opioid related secondary adverse events [113]
There are limited data regarding the repeated use of celiac plexus block (pain relief is achieved in 29% of the patients)
4.
Nutritional support.
 
Malnourishment is very common in patients with PADC secondary to problems with intake as well as cancer-associated cachexia. Frequent assessment of nutritional status is recommended, being Patient-Generated Subjective Global Assessment (PG-SGA) the most common scale used in oncology [114]. Intervention ranges from dietary counseling, dietary supplements and enteral feeding. Parenteral nutrition is only recommended as a temporary approach in patients with transient inability for enteral feeding with good general status. For patients scheduled to undergo surgery who present with severe malnutrition (weight loss >10% in 6 months, BMI <18.5 kg/m2 and serum albumin <3 g/dl) it is recommended to provide nutritional support by dietary supplements, enteral feeding or parenteral feeding for 7–14 days. More than 50% with PADC have pancreatic exocrine insufficiency. This problem usually presents as esteatorrhea. Optimal substitutive treatment with substitute pancreatic enzymes is recommended (25,000–150,000 units per meal intake) [115].
Cachexia is a multifactorial syndrome characterized by permanent loss of lean body mass. It does not respond to conventional nutritional support and leads to progressive functional deterioration. 20 to 80% of patients with PADC have cachexia, being more common in advance disease [116]. It is a poor prognosis factor and unfortunately, there is no effective treatment once established. Identification of patients in a pre-cachexia stage were multimodality approach may reverse the symptoms is critical [117]. Megestrol acetate and high dose steroids are approved for this condition. However, in case of megestrol acetate, side effects such as thrombotic episodes limit its universal recommendation [118]. All these nutritional support recommendations achieve a level of evidence of IIB.
5.
Thromboembolic disease.
 
Thromboembolic disease (TED) is one of the most common complications, with an incidence of 20–35% [119]. Its etiology is multifactorial, being associated with poor prognosis, particularly in patients with early thrombosis. The risk of TED increase in the perioperative period in patients with advanced disease and in those treated with chemotherapy. An elevated D-dymer, poor performance status, central catheter and absence of prophylaxis increases the risk. The Khorana index is useful to identify high risk patients [120]. In randomized clinical trials, prophylaxis of deep venous thrombosis (DVT) in patients with PDAC resulted in significant decrease in the rate of thrombosis, with no impact on survival [121, 122]. Routine prophylaxis of thrombosis is not recommended in the ambulatory setting [123] (IA). In patients with a Khorana index ≥3 and no risk of bleeding, prophylaxis with low molecular weight heparin could be considered (IIB). Hospitalized patients and those who underwent surgery treatment with low molecular weight heparin are recommended for at least 6 months (IIA).

Acknowledgements

The authors thank Sofía Perea, Pharm D, PhD, for her support in writing the manuscript. Funding was provided by Fundación ECO.

Compliance with ethical standards

Conflict of interest

The authors declare to have no conflict of interest.

Funding

The support for medical writing was supported by Fundación ECO.
NA.

Research involving human participants and/or animals

NA.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Literatur
2.
Zurück zum Zitat GLOBOCAN. European age-standardised rates calculated by the statistical information team at cancer research UK, 2011 using data from GLOBOCAN 2008 v1.2, IARC, version 1.2. http://globocan.iarc.fr. Accessed May 2016. GLOBOCAN. European age-standardised rates calculated by the statistical information team at cancer research UK, 2011 using data from GLOBOCAN 2008 v1.2, IARC, version 1.2. http://​globocan.​iarc.​fr. Accessed May 2016.
3.
Zurück zum Zitat Hidalgo M, Cascinu S, Kleeff J, Labianca R, Löhr JM, Neoptolemos J, et al. Addressing the challenges of pancreatic cancer: future directions for improving outcomes. Pancreatology. 2015;15:8–18.PubMedCrossRef Hidalgo M, Cascinu S, Kleeff J, Labianca R, Löhr JM, Neoptolemos J, et al. Addressing the challenges of pancreatic cancer: future directions for improving outcomes. Pancreatology. 2015;15:8–18.PubMedCrossRef
4.
Zurück zum Zitat Ko AH. Pancreatic cancer and the possibility of long-term survival. A glimmer of hope. JAMA Oncol. 2016;2:380–1.PubMedCrossRef Ko AH. Pancreatic cancer and the possibility of long-term survival. A glimmer of hope. JAMA Oncol. 2016;2:380–1.PubMedCrossRef
5.
Zurück zum Zitat Dykewicz CA. Summary of the guidelines for preventing opportunistic infections among hematopoietic stem cell transplant recipients. Clin Infect Dis. 2001;33:139–44.PubMedCrossRef Dykewicz CA. Summary of the guidelines for preventing opportunistic infections among hematopoietic stem cell transplant recipients. Clin Infect Dis. 2001;33:139–44.PubMedCrossRef
6.
Zurück zum Zitat Porta M, Fabregat X, Malats N, Guarner L, Carrato A, de Miguel A, et al. Exocrine pancreatic cancer: symptoms at presentation and their relation to tumour site and stage. Clin Trans Oncol. 2005;7:189–97.CrossRef Porta M, Fabregat X, Malats N, Guarner L, Carrato A, de Miguel A, et al. Exocrine pancreatic cancer: symptoms at presentation and their relation to tumour site and stage. Clin Trans Oncol. 2005;7:189–97.CrossRef
7.
Zurück zum Zitat Ducreux M, Cuhna AS, Caramella C, Hollebecque A, Burtin P, Goere D, et al. Cancer of the pancreas: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2015;26:56–68.CrossRef Ducreux M, Cuhna AS, Caramella C, Hollebecque A, Burtin P, Goere D, et al. Cancer of the pancreas: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2015;26:56–68.CrossRef
8.
Zurück zum Zitat Hart PA, Kamada P, Rabe KG, Srinivasan S, Basu A, Aggarwal G, et al. Weight loss precedes cancer-specific symptoms in pancreatic cancer-associated diabetes mellitus. Pancreas. 2011;40:768–72.PubMedPubMedCentralCrossRef Hart PA, Kamada P, Rabe KG, Srinivasan S, Basu A, Aggarwal G, et al. Weight loss precedes cancer-specific symptoms in pancreatic cancer-associated diabetes mellitus. Pancreas. 2011;40:768–72.PubMedPubMedCentralCrossRef
9.
Zurück zum Zitat Chari ST, Leibson CL, Rabe KG, Ransom J, de Andrade M, Petersen GM. Probability of pancreatic cancer following diabetes: a population-based study. Gastroenterology. 2005;129:504–11.PubMedPubMedCentralCrossRef Chari ST, Leibson CL, Rabe KG, Ransom J, de Andrade M, Petersen GM. Probability of pancreatic cancer following diabetes: a population-based study. Gastroenterology. 2005;129:504–11.PubMedPubMedCentralCrossRef
10.
Zurück zum Zitat Ben Q, Xu M, Ning X, Liu J, Hong S, Huang W, et al. Diabetes mellitus and risk of pancreatic cancer: a meta-analysis of cohort studies. Eur J Cancer. 2011;47:1928–37.PubMedCrossRef Ben Q, Xu M, Ning X, Liu J, Hong S, Huang W, et al. Diabetes mellitus and risk of pancreatic cancer: a meta-analysis of cohort studies. Eur J Cancer. 2011;47:1928–37.PubMedCrossRef
11.
Zurück zum Zitat Canto MI, Harink F, Hruban RH, Offerhaus GJ, Poley JW, Kamel I, et al. International Cancer of the pancreas screening consortium summit on the management of patients with increased risk for familial pancreatic cancer. Gut. 2013;62:339–47.PubMedCrossRef Canto MI, Harink F, Hruban RH, Offerhaus GJ, Poley JW, Kamel I, et al. International Cancer of the pancreas screening consortium summit on the management of patients with increased risk for familial pancreatic cancer. Gut. 2013;62:339–47.PubMedCrossRef
12.
Zurück zum Zitat Guillen-Ponce C, Mocci E, Earl J, Márquez M, Solera J, Salazar-López MT, et al. PANGEN-FAM: Spanish Registry of Hereditary Pancreatic Cancer. Eur J Cancer. 2015;51:1911–7.PubMedCrossRef Guillen-Ponce C, Mocci E, Earl J, Márquez M, Solera J, Salazar-López MT, et al. PANGEN-FAM: Spanish Registry of Hereditary Pancreatic Cancer. Eur J Cancer. 2015;51:1911–7.PubMedCrossRef
13.
Zurück zum Zitat Canto MI, Goggins M, Hruban RH, Petersen GM, Giardiello FM, Yeo C, et al. Screening for early pancreatic neoplasia in high-risk individuals: a prospective controlled study. Clin Gastroenterol Hepatol. 2006;4:766–81.PubMedCrossRef Canto MI, Goggins M, Hruban RH, Petersen GM, Giardiello FM, Yeo C, et al. Screening for early pancreatic neoplasia in high-risk individuals: a prospective controlled study. Clin Gastroenterol Hepatol. 2006;4:766–81.PubMedCrossRef
14.
Zurück zum Zitat Poley JW, Kluijt I, Gouma DJ, Harinck F, Wagner A, Aalfs C, et al. The yield of first-time endoscopic ultrasonography in screening individuals at a high risk of developing pancreatic cancer. Am J Gastroenterol. 2009;104:2175–81.PubMedCrossRef Poley JW, Kluijt I, Gouma DJ, Harinck F, Wagner A, Aalfs C, et al. The yield of first-time endoscopic ultrasonography in screening individuals at a high risk of developing pancreatic cancer. Am J Gastroenterol. 2009;104:2175–81.PubMedCrossRef
15.
Zurück zum Zitat Canto MI, Hruban RH, Fishman EK, Kamel IR, Schulick R, Zhang Z, et al. American Cancer of the Pancreas Screening (CAPS) Consortium. Frequent detection of pancreatic lesions in asymptomatic high-risk individuals. Gastroenterology. 2012;142:796–804.PubMedPubMedCentralCrossRef Canto MI, Hruban RH, Fishman EK, Kamel IR, Schulick R, Zhang Z, et al. American Cancer of the Pancreas Screening (CAPS) Consortium. Frequent detection of pancreatic lesions in asymptomatic high-risk individuals. Gastroenterology. 2012;142:796–804.PubMedPubMedCentralCrossRef
16.
Zurück zum Zitat Humphris JL, Johns A, Simpson H, Cowley MJ, Pajic M, Chang DK, et al. Clinical and pathologic features of familial pancreatic cancer. Cancer. 2014;120:3669–75.PubMedCrossRef Humphris JL, Johns A, Simpson H, Cowley MJ, Pajic M, Chang DK, et al. Clinical and pathologic features of familial pancreatic cancer. Cancer. 2014;120:3669–75.PubMedCrossRef
17.
Zurück zum Zitat Miura F, Takada T, Amano H, Yoshida M, Furui S, Takeshita K, et al. Diagnosis of pancreatic cancer. HPB (Oxford). 2006;8:337–42.CrossRef Miura F, Takada T, Amano H, Yoshida M, Furui S, Takeshita K, et al. Diagnosis of pancreatic cancer. HPB (Oxford). 2006;8:337–42.CrossRef
18.
Zurück zum Zitat Al-Hawary MM, Francis IR, Chari ST, Fishman EK, Hough DM, Lu DS, et al. Pancreatic ductal adenocarcinoma radiology reporting template: consensus statement of the Society of Abdominal Radiology and the American Pancreatic Association. Radiology. 2014;270:248–60.PubMedCrossRef Al-Hawary MM, Francis IR, Chari ST, Fishman EK, Hough DM, Lu DS, et al. Pancreatic ductal adenocarcinoma radiology reporting template: consensus statement of the Society of Abdominal Radiology and the American Pancreatic Association. Radiology. 2014;270:248–60.PubMedCrossRef
19.
Zurück zum Zitat Fernández-del Castill. Clinical manifestations, diagnosis, and staging of exocrine pancreatic cancer. 2015. (Up To Date: August journal). Fernández-del Castill. Clinical manifestations, diagnosis, and staging of exocrine pancreatic cancer. 2015. (Up To Date: August journal).
20.
Zurück zum Zitat Nawaz H, Fan CY, Kloke J, Khalid A, McGrath K, Landsittel D, Papachristou GI, et al. Performance characteristics of endoscopic ultrasound in the staging of pancreatic cancer: a meta-analysis. JOP. 2013;14:484–97.PubMedPubMedCentral Nawaz H, Fan CY, Kloke J, Khalid A, McGrath K, Landsittel D, Papachristou GI, et al. Performance characteristics of endoscopic ultrasound in the staging of pancreatic cancer: a meta-analysis. JOP. 2013;14:484–97.PubMedPubMedCentral
21.
Zurück zum Zitat Asbun HJ, Conlon K, Fernandez-Cruz L, Friess H, Shrikhande SV, Adham M, et al. When to perform a pancreatoduodenectomy in the absence of positive histology? A consensus statement by the International Study Group of Pancreatic Surgery. Surgery. 2014;155:887–92.PubMedCrossRef Asbun HJ, Conlon K, Fernandez-Cruz L, Friess H, Shrikhande SV, Adham M, et al. When to perform a pancreatoduodenectomy in the absence of positive histology? A consensus statement by the International Study Group of Pancreatic Surgery. Surgery. 2014;155:887–92.PubMedCrossRef
22.
Zurück zum Zitat Munroe CA, Fehmi SM, Savides TJ. Endoscopic ultrasound in the diagnosis of pancreatic cancer. Expert Opin Med Diagn. 2013;7:25–35.PubMedCrossRef Munroe CA, Fehmi SM, Savides TJ. Endoscopic ultrasound in the diagnosis of pancreatic cancer. Expert Opin Med Diagn. 2013;7:25–35.PubMedCrossRef
23.
Zurück zum Zitat Morris-Stiff G, Taylor MA. CA 19.9 and pancreatic cancer: is it really that good? J Gastrointest Oncol. 2012;3:88–389.PubMedPubMedCentral Morris-Stiff G, Taylor MA. CA 19.9 and pancreatic cancer: is it really that good? J Gastrointest Oncol. 2012;3:88–389.PubMedPubMedCentral
24.
Zurück zum Zitat Cwik G, Wallner G, Skoczylas T, Ciechanski A, Zinkiewicz K. Cancer antigens 19-9 and 125 in the differential diagnosis of pancreatic mass lesions. Arch Surg. 2006;141:968.PubMedCrossRef Cwik G, Wallner G, Skoczylas T, Ciechanski A, Zinkiewicz K. Cancer antigens 19-9 and 125 in the differential diagnosis of pancreatic mass lesions. Arch Surg. 2006;141:968.PubMedCrossRef
25.
Zurück zum Zitat Kondo N, Murakami Y, Uemura K, Hayashidani Y, Sudo T, Hashimoto Y, et al. Prognostic impact of perioperatory serum CA 19.9 levels in patients with resectable pancreatic cancer. Ann Surg Oncol. 2010;17:2321.PubMedCrossRef Kondo N, Murakami Y, Uemura K, Hayashidani Y, Sudo T, Hashimoto Y, et al. Prognostic impact of perioperatory serum CA 19.9 levels in patients with resectable pancreatic cancer. Ann Surg Oncol. 2010;17:2321.PubMedCrossRef
26.
Zurück zum Zitat Humphris JL, Chang DK, Al Johns, Scarlett CJ, Pajic M, Jones MD, et al. The prognostic and predictive value of serum CA 19.9 in pancreatic cancer. Ann Oncol. 2012;23:1713.PubMedPubMedCentralCrossRef Humphris JL, Chang DK, Al Johns, Scarlett CJ, Pajic M, Jones MD, et al. The prognostic and predictive value of serum CA 19.9 in pancreatic cancer. Ann Oncol. 2012;23:1713.PubMedPubMedCentralCrossRef
27.
Zurück zum Zitat Kim JE, Lee KT, Lee JK, Paik SW, Rhee JC, Choi KW, et al. Clinical usefulness of carbohydrate antigen 19.9 as a screening test for pancreatic cancer in an asymptomatic population. J Gastroenterol Hepatol. 2004;19:182.PubMedCrossRef Kim JE, Lee KT, Lee JK, Paik SW, Rhee JC, Choi KW, et al. Clinical usefulness of carbohydrate antigen 19.9 as a screening test for pancreatic cancer in an asymptomatic population. J Gastroenterol Hepatol. 2004;19:182.PubMedCrossRef
28.
Zurück zum Zitat Yu J, Blackford AL, Molin M, Wolfgang CL, Goggins M. Time to progression of pancreatic ductal adenocarcinoma from low-to-high tumour stages. Gut. 2015;0:1–7. Yu J, Blackford AL, Molin M, Wolfgang CL, Goggins M. Time to progression of pancreatic ductal adenocarcinoma from low-to-high tumour stages. Gut. 2015;0:1–7.
29.
Zurück zum Zitat Nishida K, Kaneko T, Yoneda M, Nakagawa S, Ishikawa T, Yamane E, et al. Doubling time of serum CA 19-9 in the clinical course of patients with pancreatic cancer and its significant association with prognosis. J Surg Oncol. 1999;71:140–6.PubMedCrossRef Nishida K, Kaneko T, Yoneda M, Nakagawa S, Ishikawa T, Yamane E, et al. Doubling time of serum CA 19-9 in the clinical course of patients with pancreatic cancer and its significant association with prognosis. J Surg Oncol. 1999;71:140–6.PubMedCrossRef
33.
Zurück zum Zitat Søgaard M, Thomsen RW, Bossen KS, Sørensen HT, Nørgaard M. The impact of comorbidity on cancer survival: a review. Clin Epidemiol. 2013;5:3–29.PubMedPubMedCentralCrossRef Søgaard M, Thomsen RW, Bossen KS, Sørensen HT, Nørgaard M. The impact of comorbidity on cancer survival: a review. Clin Epidemiol. 2013;5:3–29.PubMedPubMedCentralCrossRef
34.
Zurück zum Zitat Chen YG, Pan HH, Dai MS, Lin C, Lu CS, Su SL, et al. Impact of comorbidity and age on determinants therapeutic strategies in advanced pancreatic head cancer patients with obstructive jaundices. Medicine (Baltimore). 2015;94:e1298.CrossRef Chen YG, Pan HH, Dai MS, Lin C, Lu CS, Su SL, et al. Impact of comorbidity and age on determinants therapeutic strategies in advanced pancreatic head cancer patients with obstructive jaundices. Medicine (Baltimore). 2015;94:e1298.CrossRef
35.
Zurück zum Zitat Von Hoff DD, Ervin T, Arena FP, Chiorean G, Infante J, Moore M, et al. Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N Engl J Med. 2013;369:1691–703.CrossRef Von Hoff DD, Ervin T, Arena FP, Chiorean G, Infante J, Moore M, et al. Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N Engl J Med. 2013;369:1691–703.CrossRef
36.
Zurück zum Zitat Gillen S, Schuster T, Meyer Zum Büschenfelde C, Friess H, Kleef J. Preoperative/neoadjuvant therapy in pancreatic cancer: a systematic review and meta-analysis of response and resection percentages. PLoS Med. 2010;7:e1000267.PubMedPubMedCentralCrossRef Gillen S, Schuster T, Meyer Zum Büschenfelde C, Friess H, Kleef J. Preoperative/neoadjuvant therapy in pancreatic cancer: a systematic review and meta-analysis of response and resection percentages. PLoS Med. 2010;7:e1000267.PubMedPubMedCentralCrossRef
37.
Zurück zum Zitat Assifi MM, Lu X, Eibl G, Reber HA, Li G, Hines OJ. Neoadjuvant therapy in pancreatic adenocarcinoma: a meta-analysis of phase II trials. Surgery. 2011;150:466–73.PubMedPubMedCentralCrossRef Assifi MM, Lu X, Eibl G, Reber HA, Li G, Hines OJ. Neoadjuvant therapy in pancreatic adenocarcinoma: a meta-analysis of phase II trials. Surgery. 2011;150:466–73.PubMedPubMedCentralCrossRef
38.
Zurück zum Zitat Andriulli A, Festa V, Botteri E, Valvano MR, Koch M, Bassi C, Maisonneuve P, et al. Neoadjuvant/preoperative gemcitabine for patients with localized pancreatic cancer: a meta-analysis of prospective studies. Ann Surg Oncol. 2012;19:1644–62.PubMedCrossRef Andriulli A, Festa V, Botteri E, Valvano MR, Koch M, Bassi C, Maisonneuve P, et al. Neoadjuvant/preoperative gemcitabine for patients with localized pancreatic cancer: a meta-analysis of prospective studies. Ann Surg Oncol. 2012;19:1644–62.PubMedCrossRef
39.
Zurück zum Zitat Christians K, Tsai S, Mahmoud A, Ritch P, Thomas JP, Wiebe L, et al. Neoadjuvant FOLFIRINOX for borderline resectable pancreas cancer: a new treatment paradigm? Oncologist. 2014;19:266–74.PubMedPubMedCentralCrossRef Christians K, Tsai S, Mahmoud A, Ritch P, Thomas JP, Wiebe L, et al. Neoadjuvant FOLFIRINOX for borderline resectable pancreas cancer: a new treatment paradigm? Oncologist. 2014;19:266–74.PubMedPubMedCentralCrossRef
40.
Zurück zum Zitat Mahaseth H, Brutcher E, Kauh J, Hawk N, Kim S, Chen Z, et al. Modified FOLFIRINOX regimen with improved safety and maintained efficacy in pancreatic adenocarcinoma. Pancreas. 2013;42:1311–5.PubMedCrossRef Mahaseth H, Brutcher E, Kauh J, Hawk N, Kim S, Chen Z, et al. Modified FOLFIRINOX regimen with improved safety and maintained efficacy in pancreatic adenocarcinoma. Pancreas. 2013;42:1311–5.PubMedCrossRef
41.
Zurück zum Zitat Peddi PF, Lubner S, McWilliams R, Tan BR, Picus J, Sorscher SM, et al. Multi-institutional experience with FOLFIRINOX in pancreatic adenocarcinoma. JOP. 2012;13:497–501.PubMed Peddi PF, Lubner S, McWilliams R, Tan BR, Picus J, Sorscher SM, et al. Multi-institutional experience with FOLFIRINOX in pancreatic adenocarcinoma. JOP. 2012;13:497–501.PubMed
42.
Zurück zum Zitat Hosein PJ, Macintyre J, Kawamura C, Maldonado JC, Ernani V, Loaiza-Bonilla A, et al. A retrospective study of neoadjuvant FOLFIRINOX in unresectable or borderline-resectable locally advanced pancreatic adenocarcinoma. BMC Cancer. 2012;12:199.PubMedPubMedCentralCrossRef Hosein PJ, Macintyre J, Kawamura C, Maldonado JC, Ernani V, Loaiza-Bonilla A, et al. A retrospective study of neoadjuvant FOLFIRINOX in unresectable or borderline-resectable locally advanced pancreatic adenocarcinoma. BMC Cancer. 2012;12:199.PubMedPubMedCentralCrossRef
43.
Zurück zum Zitat Mellon EA, Hoffe SE, Springett GM, Frakes JM, Strom TJ, Hodul PJ, et al. Long-term outcomes of induction chemotherapy and neoadjuvant stereotactic body radiotherapy for borderline resectable and locally advanced pancreatic adenocarcinoma. Acta Oncol. 2015;54:585–979.CrossRef Mellon EA, Hoffe SE, Springett GM, Frakes JM, Strom TJ, Hodul PJ, et al. Long-term outcomes of induction chemotherapy and neoadjuvant stereotactic body radiotherapy for borderline resectable and locally advanced pancreatic adenocarcinoma. Acta Oncol. 2015;54:585–979.CrossRef
44.
Zurück zum Zitat Marthey L, Sa-Cunha A, Blanc JF, Gauthier M, Cueff A, Francois E, et al. FOLFIRINOX for locally advanced pancreatic adenocarcinoma: results of an AGEO multicenter prospective observational cohort. Ann Surg Oncol. 2015;22:295–301.PubMedCrossRef Marthey L, Sa-Cunha A, Blanc JF, Gauthier M, Cueff A, Francois E, et al. FOLFIRINOX for locally advanced pancreatic adenocarcinoma: results of an AGEO multicenter prospective observational cohort. Ann Surg Oncol. 2015;22:295–301.PubMedCrossRef
45.
Zurück zum Zitat Alvarez R, Musteanu M, Garcia-Garcia E, López-Casas PP, Megias D, Guerra C, et al. Stromal disrupting effects of nab-paclitaxel in pancreatic cancer. Br J Cancer. 2013;109:926–33.PubMedPubMedCentralCrossRef Alvarez R, Musteanu M, Garcia-Garcia E, López-Casas PP, Megias D, Guerra C, et al. Stromal disrupting effects of nab-paclitaxel in pancreatic cancer. Br J Cancer. 2013;109:926–33.PubMedPubMedCentralCrossRef
46.
Zurück zum Zitat Conroy T, Desseigne F, Ychou M, Bouché O, Guimbaud R, Bécouarn Y, et al. FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. N Engl J Med. 2011;364:1817–25.PubMedCrossRef Conroy T, Desseigne F, Ychou M, Bouché O, Guimbaud R, Bécouarn Y, et al. FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. N Engl J Med. 2011;364:1817–25.PubMedCrossRef
47.
Zurück zum Zitat Katz MH, Fleming JB, Bhosale P, Varadhachary G, Je Lee, Wolff R, et al. Response of borderline resectable pancreatic cancer to neoadjuvant therapy is not reflected by radiographic indicators. Cancer. 2012;118:5749–56.PubMedCrossRef Katz MH, Fleming JB, Bhosale P, Varadhachary G, Je Lee, Wolff R, et al. Response of borderline resectable pancreatic cancer to neoadjuvant therapy is not reflected by radiographic indicators. Cancer. 2012;118:5749–56.PubMedCrossRef
48.
Zurück zum Zitat Tzeng CW, Balachandran A, Ahmad M, et al. Serum carbohydrate antigen 19-9 represents a marker of response to neoadjuvant therapy in patients with borderline resectable pancreatic cancer. HPB (Oxford). 2014;16:430–8.CrossRef Tzeng CW, Balachandran A, Ahmad M, et al. Serum carbohydrate antigen 19-9 represents a marker of response to neoadjuvant therapy in patients with borderline resectable pancreatic cancer. HPB (Oxford). 2014;16:430–8.CrossRef
49.
Zurück zum Zitat Katz MH, Varadhachary GR, Fleming JB, Wolff RA, Lee JE, Pisters PWT, et al. Serum CA 19-9 as a marker of resectability and survival in patients with potentially resectable pancreatic cancer treated with neoadjuvant chemoradiation. Ann Surg Oncol. 2010;17:1794–801.PubMedPubMedCentralCrossRef Katz MH, Varadhachary GR, Fleming JB, Wolff RA, Lee JE, Pisters PWT, et al. Serum CA 19-9 as a marker of resectability and survival in patients with potentially resectable pancreatic cancer treated with neoadjuvant chemoradiation. Ann Surg Oncol. 2010;17:1794–801.PubMedPubMedCentralCrossRef
50.
Zurück zum Zitat Tamm EP, Balachandran A, Bhosale PR, Katz MH, Fleming JB, Lee JH, et al. Imaging of pancreatic adenocarcinoma: update on staging/resectability. Radiol Clin North Am. 2012;50:407–28.PubMedCrossRef Tamm EP, Balachandran A, Bhosale PR, Katz MH, Fleming JB, Lee JH, et al. Imaging of pancreatic adenocarcinoma: update on staging/resectability. Radiol Clin North Am. 2012;50:407–28.PubMedCrossRef
51.
Zurück zum Zitat Bockhorn M, Uzunoglu FG, Adham M, Imrie C, Milicevic M, Sandberg AA, et al. Borderline resectable pancreatic cancer: a consensus statement by the International Study Group of Pancreatic Surgery (ISGPS). Surgery. 2014;155:977–88.PubMedCrossRef Bockhorn M, Uzunoglu FG, Adham M, Imrie C, Milicevic M, Sandberg AA, et al. Borderline resectable pancreatic cancer: a consensus statement by the International Study Group of Pancreatic Surgery (ISGPS). Surgery. 2014;155:977–88.PubMedCrossRef
52.
Zurück zum Zitat Callery MP, Chang KJ, Fishman EK, Talamonti MS, Traverso W, Linehan DC, et al. Pretreatment assessment of resectable and borderline resectable pancreatic cancer: expert consensus statement. Ann Surg Oncol. 2009;16:1727–33.PubMedCrossRef Callery MP, Chang KJ, Fishman EK, Talamonti MS, Traverso W, Linehan DC, et al. Pretreatment assessment of resectable and borderline resectable pancreatic cancer: expert consensus statement. Ann Surg Oncol. 2009;16:1727–33.PubMedCrossRef
53.
Zurück zum Zitat Varadhachary GR, Tamm EP, Abbruzzese JL, Xiong HQ, Crane CH, Wang H, et al. Borderline resectable pancreatic cancer: definitions, management, and role of preoperative therapy. Ann Surg Oncol. 2006;13:1035–46.PubMedCrossRef Varadhachary GR, Tamm EP, Abbruzzese JL, Xiong HQ, Crane CH, Wang H, et al. Borderline resectable pancreatic cancer: definitions, management, and role of preoperative therapy. Ann Surg Oncol. 2006;13:1035–46.PubMedCrossRef
54.
Zurück zum Zitat Katz MH, Marsh R, Herman JM, Shi Q, Collison E, Venook AP, et al. Borderline resectable pancreatic cancer: need for standardization and methods for optimal clinical trial design. Ann Surg Oncol. 2013;20:2787–95.PubMedCrossRef Katz MH, Marsh R, Herman JM, Shi Q, Collison E, Venook AP, et al. Borderline resectable pancreatic cancer: need for standardization and methods for optimal clinical trial design. Ann Surg Oncol. 2013;20:2787–95.PubMedCrossRef
55.
Zurück zum Zitat Evans DB, Farnell MB, Lillemoe KD, Vollmer C Jr, Strasberg SM, Schulick RD. Surgical treatment of resectable and borderline resectable pancreas cancer: expert consensus statement. Ann Surg Oncol. 2009;16:1736–44.PubMedCrossRef Evans DB, Farnell MB, Lillemoe KD, Vollmer C Jr, Strasberg SM, Schulick RD. Surgical treatment of resectable and borderline resectable pancreas cancer: expert consensus statement. Ann Surg Oncol. 2009;16:1736–44.PubMedCrossRef
56.
Zurück zum Zitat Braga M, Capretti G, Pecorelli N, Balzano G, Doglioni C, Ariotti R, et al. A prognostic score to predict major complications after pancreaticoduodenectomy. Ann Surg. 2011;254:702–8.PubMedCrossRef Braga M, Capretti G, Pecorelli N, Balzano G, Doglioni C, Ariotti R, et al. A prognostic score to predict major complications after pancreaticoduodenectomy. Ann Surg. 2011;254:702–8.PubMedCrossRef
57.
Zurück zum Zitat Uzunoglu FG, Reeh M, Vettorazzi E, Ruschke T, Hannah P, Nentwich MF, et al. Preoperative Pancreatic Resection (PREPARE) score: a prospective multicenter-based morbidity risk score. Ann Surg. 2014;260:857–64.PubMedCrossRef Uzunoglu FG, Reeh M, Vettorazzi E, Ruschke T, Hannah P, Nentwich MF, et al. Preoperative Pancreatic Resection (PREPARE) score: a prospective multicenter-based morbidity risk score. Ann Surg. 2014;260:857–64.PubMedCrossRef
58.
Zurück zum Zitat Ragulin-Coyne E, Carroll JE, Smith JK, Witkowski ER, Ng SC, Shah SA, et al. Perioperative mortality after pancreatectomy: a risk score to aid decision-making. Surgery. 2012;152(3 Suppl. 1):S120–7.PubMedCrossRef Ragulin-Coyne E, Carroll JE, Smith JK, Witkowski ER, Ng SC, Shah SA, et al. Perioperative mortality after pancreatectomy: a risk score to aid decision-making. Surgery. 2012;152(3 Suppl. 1):S120–7.PubMedCrossRef
60.
Zurück zum Zitat Hartwig W, Vollmer CM, Fingerhut A, Yeo CJ, Neoptolemos JP, Adham M, et al. Extended pancreatectomy in pancreatic ductal adenocarcinoma: definition and consensus of the International Study Group for Pancreatic Surgery (ISGPS). Surgery. 2014;156:1–14.PubMedCrossRef Hartwig W, Vollmer CM, Fingerhut A, Yeo CJ, Neoptolemos JP, Adham M, et al. Extended pancreatectomy in pancreatic ductal adenocarcinoma: definition and consensus of the International Study Group for Pancreatic Surgery (ISGPS). Surgery. 2014;156:1–14.PubMedCrossRef
61.
Zurück zum Zitat Hartwig W, Gluth A, Hinz U, Bergmann F, Spronk PE, Hackert T, et al. Total pancreatectomy for primary pancreatic neoplasms: renaissance of an unpopular operation. Ann Surg. 2015;261:537–46.PubMedCrossRef Hartwig W, Gluth A, Hinz U, Bergmann F, Spronk PE, Hackert T, et al. Total pancreatectomy for primary pancreatic neoplasms: renaissance of an unpopular operation. Ann Surg. 2015;261:537–46.PubMedCrossRef
62.
Zurück zum Zitat Boggi U, Amorese G, Vistoli F, Caniglia F, De Lio N, Perrone V, et al. Laparoscopic pancreaticoduodenectomy: a systematic literature review. Surg Endosc. 2015;29:9–23.PubMedCrossRef Boggi U, Amorese G, Vistoli F, Caniglia F, De Lio N, Perrone V, et al. Laparoscopic pancreaticoduodenectomy: a systematic literature review. Surg Endosc. 2015;29:9–23.PubMedCrossRef
63.
Zurück zum Zitat Stafford AT, Walsh M. Robotic surgery of the pancreas: the current state of the art. J Surg Oncol. 2015;112:289–94.PubMedCrossRef Stafford AT, Walsh M. Robotic surgery of the pancreas: the current state of the art. J Surg Oncol. 2015;112:289–94.PubMedCrossRef
64.
Zurück zum Zitat Roder JD, Stein HJ, Siewert JR. Carcinoma of the periampullary region: who benefits from portal vein resection. Am J Surg. 1996;171:170–4.PubMedCrossRef Roder JD, Stein HJ, Siewert JR. Carcinoma of the periampullary region: who benefits from portal vein resection. Am J Surg. 1996;171:170–4.PubMedCrossRef
65.
Zurück zum Zitat Siriwardana HP, Siriwardena AK. Systematic review of outcome of synchronous portal-superior mesenteric vein resection during pancreatectomy for cancer. Br J Surg. 2006;93:662–73.PubMedCrossRef Siriwardana HP, Siriwardena AK. Systematic review of outcome of synchronous portal-superior mesenteric vein resection during pancreatectomy for cancer. Br J Surg. 2006;93:662–73.PubMedCrossRef
66.
Zurück zum Zitat Oettle H, Neuhaus P, Hochhaus A, Hartmann JT, Gellert K, Ridwelski K, et al. Adjuvant chemotherapy with gemcitabine and longterm outcomes among patients with resected pancreatic cancer: the CONKO-001 randomized trial. JAMA. 2013;310:1473–81.PubMedCrossRef Oettle H, Neuhaus P, Hochhaus A, Hartmann JT, Gellert K, Ridwelski K, et al. Adjuvant chemotherapy with gemcitabine and longterm outcomes among patients with resected pancreatic cancer: the CONKO-001 randomized trial. JAMA. 2013;310:1473–81.PubMedCrossRef
67.
Zurück zum Zitat Neoptolemos JP, Stocken DD, Bassi C, Ghaneh P, Cunningham D, Goldstein D, et al. Adjuvant chemotherapy with fluorouracil plus folinic acid vs gemcitabine following pancreatic cancer resection. JAMA. 2010;304:1073–112.PubMedCrossRef Neoptolemos JP, Stocken DD, Bassi C, Ghaneh P, Cunningham D, Goldstein D, et al. Adjuvant chemotherapy with fluorouracil plus folinic acid vs gemcitabine following pancreatic cancer resection. JAMA. 2010;304:1073–112.PubMedCrossRef
68.
Zurück zum Zitat Alamo JM, Marín LM, Suárez G, et al. Improving outcomes in pancreatic cancer: key points in perioperative management. World J Gastroenterol. 2014;20:14237–45.PubMedPubMedCentralCrossRef Alamo JM, Marín LM, Suárez G, et al. Improving outcomes in pancreatic cancer: key points in perioperative management. World J Gastroenterol. 2014;20:14237–45.PubMedPubMedCentralCrossRef
69.
Zurück zum Zitat Duelge K, Krepline AN, Mahmoud A (2014) Survival benefit of adjuvant therapy for resectable pancreatic cancer (RPC) patients treated with neoadjuvant Therapy. In: Oncology 67th annual Cancer Symposium of the Society of Surgical Oncology Phoenix. Annals of Surgical Oncology; Poster P173. Duelge K, Krepline AN, Mahmoud A (2014) Survival benefit of adjuvant therapy for resectable pancreatic cancer (RPC) patients treated with neoadjuvant Therapy. In: Oncology 67th annual Cancer Symposium of the Society of Surgical Oncology Phoenix. Annals of Surgical Oncology; Poster P173.
70.
Zurück zum Zitat Roland CL, Katz MH, Tzeng CD, Lin H, Varadhachary GR, Shroff R, et al. The addition of postoperative chemotherapy is associated with improved survival in patients with pancreatic cancer treated with preoperative therapy. Ann Surg Oncol. 2015. [Epub ahead of print]. Roland CL, Katz MH, Tzeng CD, Lin H, Varadhachary GR, Shroff R, et al. The addition of postoperative chemotherapy is associated with improved survival in patients with pancreatic cancer treated with preoperative therapy. Ann Surg Oncol. 2015. [Epub ahead of print].
71.
Zurück zum Zitat Cascinu S, Falconi M, Valentini V, Jelic S, on behalf of the ESMO Guidelines Working Group. Pancreatic cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2010;21:v55–8.PubMedCrossRef Cascinu S, Falconi M, Valentini V, Jelic S, on behalf of the ESMO Guidelines Working Group. Pancreatic cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2010;21:v55–8.PubMedCrossRef
72.
Zurück zum Zitat Berger AC, Garcia M, Hoffman JP, Regine WF, Abrams RA, Safran H, et al. Postresection CA 19-9 predicts overall survival in patients with pancreatic cancer treated with adjuvant chemoradiation: a prospective validation by RTOG 9704. J Clin Oncol. 2008;26:5918–22.PubMedPubMedCentralCrossRef Berger AC, Garcia M, Hoffman JP, Regine WF, Abrams RA, Safran H, et al. Postresection CA 19-9 predicts overall survival in patients with pancreatic cancer treated with adjuvant chemoradiation: a prospective validation by RTOG 9704. J Clin Oncol. 2008;26:5918–22.PubMedPubMedCentralCrossRef
73.
Zurück zum Zitat Yang GY, Malik NK, Chandrasekhar R, Ma WW, Flaherty L, Iyer R, et al. Change in CA 19-9 levels after chemoradiotherapy predicts survival in patients with locally advanced unresectable pancreatic cancer. J Gastrointest Oncol. 2013;4:361–9.PubMedPubMedCentral Yang GY, Malik NK, Chandrasekhar R, Ma WW, Flaherty L, Iyer R, et al. Change in CA 19-9 levels after chemoradiotherapy predicts survival in patients with locally advanced unresectable pancreatic cancer. J Gastrointest Oncol. 2013;4:361–9.PubMedPubMedCentral
74.
Zurück zum Zitat Neoptolemos JP, Stocken DD, Friess H, Bassic C, Dunn JA, Hickey H, et al. A randomized trial of chemoradiotherapy and chemotherapy after resection of pancreatic cancer. N Engl J Med. 2004;350:1200–10.PubMedCrossRef Neoptolemos JP, Stocken DD, Friess H, Bassic C, Dunn JA, Hickey H, et al. A randomized trial of chemoradiotherapy and chemotherapy after resection of pancreatic cancer. N Engl J Med. 2004;350:1200–10.PubMedCrossRef
75.
Zurück zum Zitat Liao WC, Chien KL, Lin YL, Wu MS, Lin JT, Wang HP, et al. Adjuvant treatments for resected pancreatic adenocarcinoma: a systematic review and network meta-analysis. Lancet Oncol. 2013;14:1095–103.PubMedCrossRef Liao WC, Chien KL, Lin YL, Wu MS, Lin JT, Wang HP, et al. Adjuvant treatments for resected pancreatic adenocarcinoma: a systematic review and network meta-analysis. Lancet Oncol. 2013;14:1095–103.PubMedCrossRef
76.
Zurück zum Zitat Neoptolemos JP, Palmer D, Ghaneh P, Valle JW, Cunningham D, Wadsley J, et al. ESPAC-4: A multicenter, international, open-label randomized controlled phase III trial of adjuvant combination chemotherapy of gemcitabine (GEM) and capecitabine (CAP) versus monotherapy gemcitabine in patients with resected pancreatic ductal adenocarcinoma. J Clin Oncol. 2016;34 (abstr LBA4006). Neoptolemos JP, Palmer D, Ghaneh P, Valle JW, Cunningham D, Wadsley J, et al. ESPAC-4: A multicenter, international, open-label randomized controlled phase III trial of adjuvant combination chemotherapy of gemcitabine (GEM) and capecitabine (CAP) versus monotherapy gemcitabine in patients with resected pancreatic ductal adenocarcinoma. J Clin Oncol. 2016;34 (abstr LBA4006).
77.
Zurück zum Zitat Gastrointestinal Tumor Study Group. Further evidence of effective adjuvant combined radiation and chemotherapy following curative resection of pancreatic cancer. Cancer. 1987;59:2006–10.CrossRef Gastrointestinal Tumor Study Group. Further evidence of effective adjuvant combined radiation and chemotherapy following curative resection of pancreatic cancer. Cancer. 1987;59:2006–10.CrossRef
78.
Zurück zum Zitat Klinkenbijil JH, Jeekel J, Sahmoud T. Adjuvant radiotherapy and 5-fluoruracil after curative resection of cancer of the pancreas and periampullary region: phase III trial of the EORTC gastrointestinal tract cancer cooperative group. Ann Surg. 1999;230:776–82.CrossRef Klinkenbijil JH, Jeekel J, Sahmoud T. Adjuvant radiotherapy and 5-fluoruracil after curative resection of cancer of the pancreas and periampullary region: phase III trial of the EORTC gastrointestinal tract cancer cooperative group. Ann Surg. 1999;230:776–82.CrossRef
79.
Zurück zum Zitat Regine WF, Winter KA, Abrams RA. Fluoruracil vs gemcitabine chemotherapy before and after fluoruracil-based chemoradiation following resection of pancreatic adenocarcinoma: a randomized controlled trial. JAMA. 2008;299:1019–26.PubMedCrossRef Regine WF, Winter KA, Abrams RA. Fluoruracil vs gemcitabine chemotherapy before and after fluoruracil-based chemoradiation following resection of pancreatic adenocarcinoma: a randomized controlled trial. JAMA. 2008;299:1019–26.PubMedCrossRef
80.
Zurück zum Zitat Corsini MM, Miller RC, Haddock MG, Donohue JH. Adjuvant radiotherapy and chemotherapy for pancreatic carcinoma: the Mayo Clinic experience (1975–2005). J Clin Oncol. 2008;26:3511–6.PubMedCrossRef Corsini MM, Miller RC, Haddock MG, Donohue JH. Adjuvant radiotherapy and chemotherapy for pancreatic carcinoma: the Mayo Clinic experience (1975–2005). J Clin Oncol. 2008;26:3511–6.PubMedCrossRef
81.
Zurück zum Zitat Herman JM, Swartz MJ, Hsu CC. Analysis of fluoruracil based adjuvant chemotherapy and radiation after pancreaticoduodenectomy for ductal adenocarcinoma of the pancreas: results of a large prospectively collected database at the Johns Hopkins Hospital. J Clin Oncol. 2008;26:3503–10.PubMedPubMedCentralCrossRef Herman JM, Swartz MJ, Hsu CC. Analysis of fluoruracil based adjuvant chemotherapy and radiation after pancreaticoduodenectomy for ductal adenocarcinoma of the pancreas: results of a large prospectively collected database at the Johns Hopkins Hospital. J Clin Oncol. 2008;26:3503–10.PubMedPubMedCentralCrossRef
82.
Zurück zum Zitat Hazard L, Tward JD, Szabo A. Radiation Therapy is associated with improved survival in patients pancreatic adenocarcinoma: results of a study form the Surveillance, Epidemiology and End Results (SEER) registry data. Cancer. 2007;110:2191–201.PubMedCrossRef Hazard L, Tward JD, Szabo A. Radiation Therapy is associated with improved survival in patients pancreatic adenocarcinoma: results of a study form the Surveillance, Epidemiology and End Results (SEER) registry data. Cancer. 2007;110:2191–201.PubMedCrossRef
83.
Zurück zum Zitat Witkowski ER, Smith JK, Ragulin-Coyne E, Ng SC, Shah SA, Tseing JF. Is it worth looking? Abdominal imaging after pancreatic cancer resection: a national study. J Gastrointest Surg. 2012;16:121–8.PubMedCrossRef Witkowski ER, Smith JK, Ragulin-Coyne E, Ng SC, Shah SA, Tseing JF. Is it worth looking? Abdominal imaging after pancreatic cancer resection: a national study. J Gastrointest Surg. 2012;16:121–8.PubMedCrossRef
84.
Zurück zum Zitat Sheffield KM, Crowell KT, Lin YL, Djukom C, Goodwin JS, Riall TS. Surveillance of pancreatic cancer patients after surgical resection. Ann Surg Oncol. 2012;19:1670–7.PubMedCrossRef Sheffield KM, Crowell KT, Lin YL, Djukom C, Goodwin JS, Riall TS. Surveillance of pancreatic cancer patients after surgical resection. Ann Surg Oncol. 2012;19:1670–7.PubMedCrossRef
85.
Zurück zum Zitat Benavides M, Abad A, Ales I, Carrato A, Díaz Rubio E, Gallego J, et al. TTD consensus document on the diagnosis and management of exocrine pancreatic cancer. Clin Transl Oncol. 2014;16:865–78.PubMedCrossRef Benavides M, Abad A, Ales I, Carrato A, Díaz Rubio E, Gallego J, et al. TTD consensus document on the diagnosis and management of exocrine pancreatic cancer. Clin Transl Oncol. 2014;16:865–78.PubMedCrossRef
86.
Zurück zum Zitat Faris JE, Blaszkowsky LS, McDermott S, Guimaraes AR, Szymonifka J, Huynh MA, et al. FOLFIRINOX in locally advanced pancreatic cancer: the Massachusetts General Hospital Cancer Center experience. Oncologist. 2013;18:543–8.PubMedPubMedCentralCrossRef Faris JE, Blaszkowsky LS, McDermott S, Guimaraes AR, Szymonifka J, Huynh MA, et al. FOLFIRINOX in locally advanced pancreatic cancer: the Massachusetts General Hospital Cancer Center experience. Oncologist. 2013;18:543–8.PubMedPubMedCentralCrossRef
87.
Zurück zum Zitat Burris HA, Moore MJ, Andersen J, Green MR, Rothenberg ML, Modiano MR, 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.PubMedCrossRef Burris HA, Moore MJ, Andersen J, Green MR, Rothenberg ML, Modiano MR, 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.PubMedCrossRef
88.
Zurück zum Zitat Hammel P, Huguet F, Van Laethem JL, Goldstein D, Glimelius B, Artru P, et al. Comparison of Comparison of chemoradiotherapy (CRT) and chemotherapy (CT) in patients with a locally advanced pancreatic cancer (LAPC) controlled after 4 months of gemcitabine with or without erlotinib: final results of the international phase III LAP 07 study. J Clin Oncol. 2013;31:LBA4003.CrossRef Hammel P, Huguet F, Van Laethem JL, Goldstein D, Glimelius B, Artru P, et al. Comparison of Comparison of chemoradiotherapy (CRT) and chemotherapy (CT) in patients with a locally advanced pancreatic cancer (LAPC) controlled after 4 months of gemcitabine with or without erlotinib: final results of the international phase III LAP 07 study. J Clin Oncol. 2013;31:LBA4003.CrossRef
89.
Zurück zum Zitat Mukherjee S, Hurt C, Griffiths G, Bridgewater JA, Crosby T, Falk S, et al. SCALOP: Results of a randomized phase II study of induction chemotherapy followed by gemcitabine (G) or capecitabine (Cap) based chemoradiation (CRT) in locally advanced pancreatic cancer (LANPC). J Clin Oncol. 2013;31:LBA146.CrossRef Mukherjee S, Hurt C, Griffiths G, Bridgewater JA, Crosby T, Falk S, et al. SCALOP: Results of a randomized phase II study of induction chemotherapy followed by gemcitabine (G) or capecitabine (Cap) based chemoradiation (CRT) in locally advanced pancreatic cancer (LANPC). J Clin Oncol. 2013;31:LBA146.CrossRef
90.
Zurück zum Zitat Peixoto RD, Ho M, Renouf DJ, Lim HJ, Gill S, Ruan JY, et al. Eligibility of Metastatic Pancreatic Cancer Patients for First-Line Palliative Intent nab-Paclitaxel Plus Gemcitabine Versus FOLFIRINOX. Am J Clin Oncol. 2015. [Epub ahead of print]. Peixoto RD, Ho M, Renouf DJ, Lim HJ, Gill S, Ruan JY, et al. Eligibility of Metastatic Pancreatic Cancer Patients for First-Line Palliative Intent nab-Paclitaxel Plus Gemcitabine Versus FOLFIRINOX. Am J Clin Oncol. 2015. [Epub ahead of print].
91.
Zurück zum Zitat Thota R, Pauff JM, Berlin JD. Treatment of metastatic pancreatic adenocarcinoma: a review. Oncology (Williston Park). 2014;28(1):70–4. Thota R, Pauff JM, Berlin JD. Treatment of metastatic pancreatic adenocarcinoma: a review. Oncology (Williston Park). 2014;28(1):70–4.
92.
Zurück zum Zitat Rahma OE, Duffy A, Liewehr DJ, Steinberg SM, Greten TF, et al. Second-line treatment in advanced pancreatic cancer: a comprehensive analysis of published clinical trials. Ann Oncol. 2013;24:1972–9.PubMedPubMedCentralCrossRef Rahma OE, Duffy A, Liewehr DJ, Steinberg SM, Greten TF, et al. Second-line treatment in advanced pancreatic cancer: a comprehensive analysis of published clinical trials. Ann Oncol. 2013;24:1972–9.PubMedPubMedCentralCrossRef
93.
Zurück zum Zitat Oettle H, Riess H, Stieler JM, Heil G, Schwaner I, Seraphin J, et al. Second-line oxaliplatin, folinic acid, and fluorouracil versus folinic acid and fluorouracil alone for gemcitabine-refractory pancreatic cancer: outcomes from the CONKO-003 Trial. J Clin Oncol. 2014;32:2423–9.PubMedCrossRef Oettle H, Riess H, Stieler JM, Heil G, Schwaner I, Seraphin J, et al. Second-line oxaliplatin, folinic acid, and fluorouracil versus folinic acid and fluorouracil alone for gemcitabine-refractory pancreatic cancer: outcomes from the CONKO-003 Trial. J Clin Oncol. 2014;32:2423–9.PubMedCrossRef
94.
Zurück zum Zitat Gill S, Ko YJ, Cripps C, Beaudoin A, Dhesy-Thind SK, Zulfigar M, et al. PANCREOX: a randomized phase 3 study of 5FU/LV with or without oxaliplatin for second-line advanced pancreatic cancer (APC) in patients (pts) who have received gemcitabine (GEM)-based chemotherapy (CT). J Clin Oncol. 2014;32(5s):A4022. Gill S, Ko YJ, Cripps C, Beaudoin A, Dhesy-Thind SK, Zulfigar M, et al. PANCREOX: a randomized phase 3 study of 5FU/LV with or without oxaliplatin for second-line advanced pancreatic cancer (APC) in patients (pts) who have received gemcitabine (GEM)-based chemotherapy (CT). J Clin Oncol. 2014;32(5s):A4022.
95.
Zurück zum Zitat Wang-Gillam A, Li CP, Bodoky G, Dean A, Shan YS, Jameson G, et al. Nanoliposomal irinotecan with fluorouracil and folinic acid in metastatic pancreatic cancer after previous gemcitabine-based therapy (NAPOLI-1): a global, randomised, open-label, phase 3 trial. Lancet. 2016;387:545–57.PubMedCrossRef Wang-Gillam A, Li CP, Bodoky G, Dean A, Shan YS, Jameson G, et al. Nanoliposomal irinotecan with fluorouracil and folinic acid in metastatic pancreatic cancer after previous gemcitabine-based therapy (NAPOLI-1): a global, randomised, open-label, phase 3 trial. Lancet. 2016;387:545–57.PubMedCrossRef
96.
Zurück zum Zitat Bertocchi P, Abeni C, Meriggi F, Rota L, Rizzi A, Di Biasi B, et al. Gemcitabine plus nab-paclitaxel as second-line and beyond treatment for metastatic pancreatic cancer: a single institution retrospective analysis. Rev Recent Clin Trials. 2015;10:142–5.PubMedCrossRef Bertocchi P, Abeni C, Meriggi F, Rota L, Rizzi A, Di Biasi B, et al. Gemcitabine plus nab-paclitaxel as second-line and beyond treatment for metastatic pancreatic cancer: a single institution retrospective analysis. Rev Recent Clin Trials. 2015;10:142–5.PubMedCrossRef
97.
Zurück zum Zitat Locker GY, Hamilton S, Harris J, Jessup JM, Kemeny N, Macdonald JS, et al. ASCO 2006 update of recommendations for the use of tumor markers in gastrointestinal cancer. J Clin Oncol. 2006;24:5313–27.PubMedCrossRef Locker GY, Hamilton S, Harris J, Jessup JM, Kemeny N, Macdonald JS, et al. ASCO 2006 update of recommendations for the use of tumor markers in gastrointestinal cancer. J Clin Oncol. 2006;24:5313–27.PubMedCrossRef
98.
Zurück zum Zitat Stark A, Hines OJ. Endoscopic and operative palliation strategies for pancreatic ductal adenocarcinoma. Semin Oncol. 2015;42:163–76.PubMedCrossRef Stark A, Hines OJ. Endoscopic and operative palliation strategies for pancreatic ductal adenocarcinoma. Semin Oncol. 2015;42:163–76.PubMedCrossRef
99.
Zurück zum Zitat Sarfeh IJ, Rypins EB, Jakowatz JG. A prospective, randomized clinical investigation of cholecystoenterostomy and choledochoenterostomy. Am Surg. 1989;55:55–60. Sarfeh IJ, Rypins EB, Jakowatz JG. A prospective, randomized clinical investigation of cholecystoenterostomy and choledochoenterostomy. Am Surg. 1989;55:55–60.
100.
Zurück zum Zitat Glazer ES, Hornbrook MC. A meta-analysis of randomized trials: immediate stent replacement vs surgical bypass in the palliative management of malignant biliary obstruction. J Pain Symptom Manag. 2014;47:307–14.CrossRef Glazer ES, Hornbrook MC. A meta-analysis of randomized trials: immediate stent replacement vs surgical bypass in the palliative management of malignant biliary obstruction. J Pain Symptom Manag. 2014;47:307–14.CrossRef
101.
Zurück zum Zitat Pinol V, Castells A. Percutaneous self-expanding metal stents versus endoscopic polyethylene endoprosthesis for treating malignant biliary obstruction: randomized clinical trial. Radiology. 2002;225:27–34.PubMedCrossRef Pinol V, Castells A. Percutaneous self-expanding metal stents versus endoscopic polyethylene endoprosthesis for treating malignant biliary obstruction: randomized clinical trial. Radiology. 2002;225:27–34.PubMedCrossRef
102.
Zurück zum Zitat Moss AC, Morris E, Leyden J, MacMathuna P. Do the benefits of metal stents justify the costs? A systematic review and meta-analysis of trials comparing endoscopic stents for malignant biliary obstruction. Eur J Gastroenterol Hepatol. 2007;19:1119–24.PubMedCrossRef Moss AC, Morris E, Leyden J, MacMathuna P. Do the benefits of metal stents justify the costs? A systematic review and meta-analysis of trials comparing endoscopic stents for malignant biliary obstruction. Eur J Gastroenterol Hepatol. 2007;19:1119–24.PubMedCrossRef
103.
Zurück zum Zitat Arkadopoulos N, Kyriazi MA, PapaniKolau IS, et al. Preoperative biliary drainage of severely jaundiced patients increases morbidity of pancreaticoduodenectomy: results of a case control study. World J Surg. 2014;38:2967–72.PubMedCrossRef Arkadopoulos N, Kyriazi MA, PapaniKolau IS, et al. Preoperative biliary drainage of severely jaundiced patients increases morbidity of pancreaticoduodenectomy: results of a case control study. World J Surg. 2014;38:2967–72.PubMedCrossRef
104.
Zurück zum Zitat Tol JAMG, van Hooft JE, Timmer R, Kubben FJ, van der Harst E, de Hingh IH, et al. Metal or plastic stents for preoperative biliary drainage in respectable pancreatic cancer. Gut Aug. 2015. (25 Published Online First). Tol JAMG, van Hooft JE, Timmer R, Kubben FJ, van der Harst E, de Hingh IH, et al. Metal or plastic stents for preoperative biliary drainage in respectable pancreatic cancer. Gut Aug. 2015. (25 Published Online First).
105.
Zurück zum Zitat Van der Gaag NA, Rauws EAJ, van Eijck CHJ, Bruno MJ, van der Harst E, Kubben FJGM, et al. Preoperative biliary drainage for cancer of the head of the pancreas. N Engl J Med. 2010;362:129–37.PubMedCrossRef Van der Gaag NA, Rauws EAJ, van Eijck CHJ, Bruno MJ, van der Harst E, Kubben FJGM, et al. Preoperative biliary drainage for cancer of the head of the pancreas. N Engl J Med. 2010;362:129–37.PubMedCrossRef
106.
Zurück zum Zitat Lillemoe KD, Cameron JL, Hardacre JM, Sohn TA, Sauter PK, Coleman J, et al. Is prophylactic gastrojejunostomy indicated for unresectable periampullary cancer? A prospective randomized trial. Ann Surg. 1999;230:322–30.PubMedPubMedCentralCrossRef Lillemoe KD, Cameron JL, Hardacre JM, Sohn TA, Sauter PK, Coleman J, et al. Is prophylactic gastrojejunostomy indicated for unresectable periampullary cancer? A prospective randomized trial. Ann Surg. 1999;230:322–30.PubMedPubMedCentralCrossRef
107.
Zurück zum Zitat Rothlin MA, Schob O, Weber M. Laparoscopic gastro- and hepatico-jejunostomy for palliation of pancreatic cancer: a case controlled study. Surg Endosc. 1999;13:1065–9.PubMedCrossRef Rothlin MA, Schob O, Weber M. Laparoscopic gastro- and hepatico-jejunostomy for palliation of pancreatic cancer: a case controlled study. Surg Endosc. 1999;13:1065–9.PubMedCrossRef
108.
Zurück zum Zitat Van Hooft JE, Uitdehaag MJ, Bruno MJ, Timmer R, Siersema PD, Dijkgraaf MG, et al. Efficacy and safety of the new WallFlex enteral stent in palliative treatment of malignant gastric outlet obstruction (DUOFLEX study): a prospective multicenter study. Gastrointest Endosc. 2009;69:1059–66.PubMedCrossRef Van Hooft JE, Uitdehaag MJ, Bruno MJ, Timmer R, Siersema PD, Dijkgraaf MG, et al. Efficacy and safety of the new WallFlex enteral stent in palliative treatment of malignant gastric outlet obstruction (DUOFLEX study): a prospective multicenter study. Gastrointest Endosc. 2009;69:1059–66.PubMedCrossRef
109.
Zurück zum Zitat Hameed M, Hammeed H, Erdek M. Pain management in pancreatic cancer. Cancer. 2011;3:43–60.CrossRef Hameed M, Hammeed H, Erdek M. Pain management in pancreatic cancer. Cancer. 2011;3:43–60.CrossRef
110.
Zurück zum Zitat Foley K. Pain syndromes and pharmacological management of pancreatic cancer pain. J Pain Symptom Manag. 1988;3:176–87.CrossRef Foley K. Pain syndromes and pharmacological management of pancreatic cancer pain. J Pain Symptom Manag. 1988;3:176–87.CrossRef
111.
Zurück zum Zitat Smith TJ, Staats PS, Deer T, Steams LJ, Rauck RL, Boortz-Mary RL, et al. Implantable drug delivery systems study group. Randomized clinical trial of an implantable drug delivery system compared with comprehensive medical management for refractory cancer pain: impact on pain, drug-related toxicity, and survival. J Clin Oncol. 2002;20:4040–9.PubMedCrossRef Smith TJ, Staats PS, Deer T, Steams LJ, Rauck RL, Boortz-Mary RL, et al. Implantable drug delivery systems study group. Randomized clinical trial of an implantable drug delivery system compared with comprehensive medical management for refractory cancer pain: impact on pain, drug-related toxicity, and survival. J Clin Oncol. 2002;20:4040–9.PubMedCrossRef
112.
Zurück zum Zitat Mercadante S, Klepstad P, Kurita GP, Sjogren P, Giarrato A, European Palliative Care Research Collaborative (EPCRC). Sympathetic blocks for visceral cancer pain management: a systematic review and EAPC recommendations. Crit Rev Oncol Hematol. 2015;96:577–83.PubMedCrossRef Mercadante S, Klepstad P, Kurita GP, Sjogren P, Giarrato A, European Palliative Care Research Collaborative (EPCRC). Sympathetic blocks for visceral cancer pain management: a systematic review and EAPC recommendations. Crit Rev Oncol Hematol. 2015;96:577–83.PubMedCrossRef
113.
Zurück zum Zitat Wyse JM, Carone M, Paquin SC. Randomized, double-blind, controlled trial of early endoscopic ultrasound-guided celiac plexus neurolysis to prevent pain progression in patients with newly diagnosed, painful, inoperable pancreatic cancer. J Clin Oncol. 2011;26:3541–6.CrossRef Wyse JM, Carone M, Paquin SC. Randomized, double-blind, controlled trial of early endoscopic ultrasound-guided celiac plexus neurolysis to prevent pain progression in patients with newly diagnosed, painful, inoperable pancreatic cancer. J Clin Oncol. 2011;26:3541–6.CrossRef
114.
Zurück zum Zitat Arends J, Bodoky G, Bozzetti F, Fearon K, Muscaritoli M, Selga G, et al. ESPEN Guidelines on enteral nutrition: non-surgical oncology. Clin Nutr. 2006;25:245–59.PubMedCrossRef Arends J, Bodoky G, Bozzetti F, Fearon K, Muscaritoli M, Selga G, et al. ESPEN Guidelines on enteral nutrition: non-surgical oncology. Clin Nutr. 2006;25:245–59.PubMedCrossRef
115.
Zurück zum Zitat Bartel MJ, Asbun H, Stauffer J, Raimondo M, Bosaeus I, Bruera E, Faisinger RL, et al. Pancreatic exocrine insufficiency in pancreatic cancer: a review of the literature. Dig Liver Dis. 2015;47:1013–20.PubMedCrossRef Bartel MJ, Asbun H, Stauffer J, Raimondo M, Bosaeus I, Bruera E, Faisinger RL, et al. Pancreatic exocrine insufficiency in pancreatic cancer: a review of the literature. Dig Liver Dis. 2015;47:1013–20.PubMedCrossRef
116.
Zurück zum Zitat Fearon K, Strasser F, Anker SD, Bosaeus I, Bruera E, Faisinger RL, et al. Definition and classification of cancer cachexia: an international consensus. Lancet Oncol. 2011;12(5):489–95.PubMedCrossRef Fearon K, Strasser F, Anker SD, Bosaeus I, Bruera E, Faisinger RL, et al. Definition and classification of cancer cachexia: an international consensus. Lancet Oncol. 2011;12(5):489–95.PubMedCrossRef
117.
Zurück zum Zitat Ozola Zalite I, Zykus R, Francisco González M, Saygili F, Pukitis A, Gaujoux S, et al. Influence of cachexia and sarcopenia on survival in pancreatic ductal adenocarcinoma: a systematic review. Pancreatology. 2015;15:19–24.PubMedCrossRef Ozola Zalite I, Zykus R, Francisco González M, Saygili F, Pukitis A, Gaujoux S, et al. Influence of cachexia and sarcopenia on survival in pancreatic ductal adenocarcinoma: a systematic review. Pancreatology. 2015;15:19–24.PubMedCrossRef
118.
Zurück zum Zitat Loprinzi CL, Kugler JW, Sloan JA, Mailliard JA, Krook JE, Wilwerding MB, et al. Randomized comparison of megestrol acetate versus dexamethasone versus fluoxymesterone for the treatment of cancer anorexia/cachexia. J Clin Oncol. 1999;17:3299–306.PubMedCrossRef Loprinzi CL, Kugler JW, Sloan JA, Mailliard JA, Krook JE, Wilwerding MB, et al. Randomized comparison of megestrol acetate versus dexamethasone versus fluoxymesterone for the treatment of cancer anorexia/cachexia. J Clin Oncol. 1999;17:3299–306.PubMedCrossRef
119.
Zurück zum Zitat Lyman GH, Eckert L, Wang Y, Wang H, Cohen A. Venous thromboembolism risk in patients with cancer receiving chemotherapy: a real-world analysis. Oncologist. 2013;18:1321–9.PubMedPubMedCentralCrossRef Lyman GH, Eckert L, Wang Y, Wang H, Cohen A. Venous thromboembolism risk in patients with cancer receiving chemotherapy: a real-world analysis. Oncologist. 2013;18:1321–9.PubMedPubMedCentralCrossRef
120.
Zurück zum Zitat Khorana AA, Kuderer NM, Culakova E, Lyman GH, Francis CW. Development and validation of a predictive model for chemotherapy-associated thrombosis. Blood. 2008;111:4902–7.PubMedPubMedCentralCrossRef Khorana AA, Kuderer NM, Culakova E, Lyman GH, Francis CW. Development and validation of a predictive model for chemotherapy-associated thrombosis. Blood. 2008;111:4902–7.PubMedPubMedCentralCrossRef
121.
Zurück zum Zitat Pelzer U, Opitz B, Deutschinoff G, Stauch M, Reitzig PC, Hahnfeld S, et al. Efficacy of prophylactic low-molecular weight heparin for ambulatory patients with advanced pancreatic cancer: outcomes from the CONKO-004 Trial. J Clin Oncol. 2015;33:2028–34.PubMedCrossRef Pelzer U, Opitz B, Deutschinoff G, Stauch M, Reitzig PC, Hahnfeld S, et al. Efficacy of prophylactic low-molecular weight heparin for ambulatory patients with advanced pancreatic cancer: outcomes from the CONKO-004 Trial. J Clin Oncol. 2015;33:2028–34.PubMedCrossRef
122.
Zurück zum Zitat Maraveyas A, Waters J, Roy R, Fyfe D, Propper D, Lofts S, et al. Gemcitabine versus gemcitabine plus dalteparin thromboprophylaxis in pancreatic cancer. Eur J Cancer. 2012;48:1283–92.PubMedCrossRef Maraveyas A, Waters J, Roy R, Fyfe D, Propper D, Lofts S, et al. Gemcitabine versus gemcitabine plus dalteparin thromboprophylaxis in pancreatic cancer. Eur J Cancer. 2012;48:1283–92.PubMedCrossRef
123.
Zurück zum Zitat Lyman GH, Bohlke K, Falanga A. Venous thromboembolism prophylaxis and treatment in patients with cancer: American Society of Clinical Oncology Clinical Practice Guideline Update. J Oncol Pract. 2015;11:e442–4.PubMedCrossRef Lyman GH, Bohlke K, Falanga A. Venous thromboembolism prophylaxis and treatment in patients with cancer: American Society of Clinical Oncology Clinical Practice Guideline Update. J Oncol Pract. 2015;11:e442–4.PubMedCrossRef
Metadaten
Titel
Consensus guidelines for diagnosis, treatment and follow-up of patients with pancreatic cancer in Spain
verfasst von
M. Hidalgo
R. Álvarez
J. Gallego
C. Guillén-Ponce
B. Laquente
T. Macarulla
A. Muñoz
M. Salgado
R. Vera
J. Adeva
I. Alés
S. Arévalo
J. Blázquez
A. Calsina
A. Carmona
E. de Madaria
R. Díaz
L. Díez
T. Fernández
B. G. de Paredes
M. E. Gallardo
I. González
O. Hernando
P. Jiménez
A. López
C. López
F. López-Ríos
E. Martín
J. Martínez
A. Martínez
J. Montans
R. Pazo
J. C. Plaza
I. Peiró
J. J. Reina
A. Sanjuanbenito
R. Yaya
Alfredo Carrato
Publikationsdatum
19.12.2016
Verlag
Springer International Publishing
Erschienen in
Clinical and Translational Oncology / Ausgabe 6/2017
Print ISSN: 1699-048X
Elektronische ISSN: 1699-3055
DOI
https://doi.org/10.1007/s12094-016-1594-x

Weitere Artikel der Ausgabe 6/2017

Clinical and Translational Oncology 6/2017 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.