Skip to main content
Erschienen in: Journal of Medical Case Reports 1/2021

Open Access 01.12.2021 | Case report

Pancreatic ductal adenocarcinoma concomitant with pancreatic metastases of clear-cell renal cell carcinoma: a case report

verfasst von: Lena Haeberle, Melanie Busch, Julian Kirchner, Georg Fluegen, Gerald Antoch, Wolfram Trudo Knoefel, Irene Esposito

Erschienen in: Journal of Medical Case Reports | Ausgabe 1/2021

Abstract

Background

Metastatic spread to the pancreas is a rare event. Renal cell carcinoma represents one possible site of origin of pancreatic metastases. Renal cell carcinoma often metastasizes late and exclusively to the pancreas, suggesting a special role of renal cell carcinoma among primaries metastasizing to the pancreas. Even rarer, renal cell carcinoma may occur simultaneously with pancreatic ductal adenocarcinoma.

Case presentation

We present the case of a 78-year-old male Caucasian patient with a history of clear-cell renal cell carcinoma treated with oncological left nephrectomy 20 years before. The patient was diagnosed with pancreatic ductal adenocarcinoma by fine-needle aspiration cytology. At our institution, he received neoadjuvant therapy with folic acid, fluorouracil, irinotecan, oxaliplatin for borderline-resectable pancreatic ductal adenocarcinoma, and subsequently underwent total pancreatectomy. Upon resection, pancreatic ductal adenocarcinoma as well as two metachronous metastases of clear-cell renal cell carcinoma occurring simultaneously and cospatially with pancreatic ductal adenocarcinoma were diagnosed in the pancreatic body.

Conclusions

Renal cell carcinoma metastases of the pancreas are rare and often occur decades after the initial diagnosis of renal cell carcinoma. The combination of renal cell carcinoma metastases and pancreatic ductal adenocarcinoma is even rarer. However, the possibility should be considered by clinicians, radiologists, and pathologists. The special role of renal cell carcinoma as a site of origin of pancreatic metastasis should be further elucidated.
Hinweise

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
Ca 19-9
Carbohydrate antigen 19-9
(cc)RCC
(Clear-cell) renal cell carcinoma
CK7
Cytokeratin 7
CPR
Cardiopulmonary resuscitation
(PET-)CT
(Positron emission tomography-)computed tomography
ECOG
Eastern Cooperative Oncology Group
ERCP
Endoscopic retrograde cholangiopancreatography
(EUS-)FNA
(Endoscopic ultrasound-guided) fine-needle aspiration
FOLFIRINOX
Folic acid, fluorouracil, irinotecan, oxaliplatin
GI
Gastrointestinal
IPMN
Intraductal papillary-mucinous neoplasm
NSTEMI
Non-ST-segment elevating myocardial infarction
Pax-8
Paired box protein 8
pNEN
Pancreatic neuroendocrine neoplasm
PDAC
Pancreatic ductal adenocarcinoma
SCN
Serous cystic neoplasm
VHL
Von Hippel–Lindau

Background

Pancreatic ductal adenocarcinoma (PDAC) is the most frequent form of pancreatic neoplasm, accounting for approximately 85% of pancreatic tumors, and is most commonly localized in the pancreatic head [1]. Neoadjuvant therapy is emerging in PDAC treatment, especially in the context of borderline-resectable PDAC [2].
Pancreatic metastases are generally uncommon, with the most frequent sites of origin being lung cancer and gastrointestinal (GI) cancers [3]. More rarely, renal cell carcinoma (RCC) can spread to the pancreas, accounting for approximately 5% of all pancreatic metastases [3].
Here, we report the rare case of concomitant neoadjuvant-treated PDAC and two clear-cell RCC (ccRCC) metastases in the pancreas of a 78-year-old male patient and present a brief review of literature.

Case presentation

A 78-year-old male Caucasian patient presented at our hospital with PDAC of the pancreatic body that had been diagnosed cytologically at a different institution. Due to newly occurring jaundice, he had received stenting of the distal bile duct and pancreatic fine needle aspiration (FNA) cytology. The patient had a history of oncological left nephrectomy for ccRCC 20 years prior. Relevant secondary diagnoses included permanent atrial fibrillation, hypertension, coronary artery disease, and chronic kidney failure. When the patient initially presented at our institution, he reported unintentional weight loss of 3–4 kg and a lack of appetite. Physical examination showed no remarkable findings.
During staging, abdominal sonography, computed tomography (CT), and chest x-ray demonstrated no distant metastases of PDAC. Multiple space-occupying masses in the pancreas were found on CT scans (Fig. 1). Due to radiologically suspected infiltration of the portal vein and possible abutment of the celiac trunk, the PDAC was classified as borderline resectable.
The patient received neoadjuvant chemotherapy with folic acid, fluorouracil, irinotecan, oxaliplatin (FOLFIRINOX) regimen because of good Eastern Cooperative Oncology Group (ECOG) performance status and borderline resectability of the PDAC. However, after the second FOLFIRINOX cycle, he developed a non-ST-segment elevating myocardial infarction (NSTEMI). Consequently, treating physicians decided to stop chemotherapy and proceed to surgical resection.
Subsequent positron emission tomography-computed tomography (PET-CT) scans showed the already known space-occupying masses in the body and tail of the pancreas. An additional, morphologically distinct area of high metabolic activity was seen in the pancreatic head.
The patient underwent total pancreatectomy with splenectomy and segmental portal/superior mesenteric vein resection and reconstruction, hemigastrectomy, and cholecystectomy.
Upon pathological examination of the resection specimen, two well-defined tumors (1.8 cm in diameter and 0.4 cm in diameter, respectively) with inhomogeneous yellow and brown cut surface were found in the pancreatic body, encompassed by another ill-defined solid white-yellowish tumor with a diameter of 2.8 cm, extending to the pancreatic tail (Fig. 2).
Upon microscopic examination, tumor cell formations with different histomorphology were found in the pancreatic body/tail (Fig. 3a). The two well-defined tumors shared an identical histomorphology displaying nodular growth of uniform tumor cells with transparent cytoplasm and small nuclei (Fig. 3b). Upon immunohistochemistry, the tumor cells stained positive for vimentin and Pax-8 (Fig. 3c, d), whereas they were negative for CK7 and Ca19-9, and were therefore diagnosed as ccRCC metastases. The larger, ill-defined tumor extending to the pancreatic tail showed a more heterogeneous histomorphology. While some areas displayed small- to medium-sized irregular tubular glands embedded in a desmoplastic stroma, other areas showed a cribriform to solid growth pattern and consisted of tumor cells with prominent eosinophilic cytoplasm, sometimes containing cytoplasmic vacuoles and highly pleomorphic enlarged nuclei (Fig. 3e). Upon immunohistochemistry, these tumor cells expressed Ca19-9 and CK7 (Fig. 3f, g), while staining negative for vimentin and Pax-8, thereby representing PDAC. Adenosquamous differentiation was excluded by p40 immunohistochemistry (not shown). Hypereosinophilic cytoplasm and cytoplasmic vacuoles were interpreted as regressive cytopathic changes following preoperative chemotherapy. The tumor regression was classified as grade 3 (poor response) according to the College of American Pathologists (CAP) tumor regression grading system [4]. Moreover, numerous venous and perineural infiltrations, six regional lymph node metastases, and peritoneal carcinomatosis were found. The final TNM stage of the PDAC was ypT2 ypN2 (6/45) ypM1 (PER) L0 V1 Pn1 R1.
The patient initially recovered steadily after surgery. However, approximately 2 weeks after surgery, he suffered from pulmonary aspiration and had to receive CPR and bronchoscopic suctioning. In a subsequent abdominal CT scan, free intraabdominal gas was detected, prompting surgery including atypical partial resection of the stomach due to ischemic perforation. Although surgery was successful, the patient developed septic shock with disseminated intravascular coagulation the following day. Despite further surgery, including subtotal colectomy due to ischemia, the patient passed away one day later as a result of refractory shock and multiple organ failure.
An overview of the clinical course is given in Table 1.
Table 1
Overview of the clinical course in the present case
Time point
Event
T = 0
ERCP with stenting of distal bile duct and EUS-FNA cytology of the pancreatic body due to painless jaundice (ex domo)
T = 5 days
Diagnosis of G2 PDAC of the pancreatic body in EUS-FNA cytology material (ex domo)
T = 5 weeks
Begin of FOLFIRINOX chemotherapy with neoadjuvant intent due to borderline resectability of PDAC and patient’s good general condition
T = 7 weeks
NSTEMI with subsequent coronary artery stenting and stop of FOLFIRINOX therapy
T = 8 weeks
Interdisciplinary decision for surgical therapy
T = 10 weeks
Total pancreatectomy with splenectomy and segmental portal/superior mesenteric vein resection and reconstruction, hemigastrectomy and cholecystectomy
T = 12 weeks
Worsening postoperative condition, including ischemic perforation of the stomach, colon ischemia, refractory shock, and multiple organ failure, leading to the patient’s death
ERCP endoscopic retrograde cholangiopancreatography, EUS-FNA endoscopic ultrasound-guided fine-needle aspiration, FOLFIRINOX folinic acid, fluorouracil, irinotecan, oxaliplatin, NSTEMI non-ST-segment elevating myocardial infarction, PDAC pancreatic ductal adenocarcinoma

Discussion

Metastases to the pancreas are rare and are often not clinically apparent. In a study considering surgical specimens and autopsy data, most resected pancreatic tumors were primary pancreatic tumors (such as PDAC), while only 3.9% of pancreatic surgical specimens harbored tumors of extrapancreatic origin, involving the pancreas secondarily [3]. However, 43% of the pancreatic tumors found in autopsy cases were found to be secondary tumors, suggesting that secondary tumors in the pancreas are often clinically inapparent [3]. While in the autopsy series, lung cancer (42%) and GI cancers (25%) were the most frequent sites of origin for metastases in the pancreas, the surgical series contained mainly lymphomas (29%) and carcinomas of the stomach (18%) and kidney (16%) as sites of origin of pancreatic metastases [3]. A systematic literature review found that, among patients who received pancreatectomy for metastases, RCC metastases were the most frequent (63%) [5].
The median interval between primary RCC and metastasis to the pancreas is approximately 7 years, and pancreatic metastasis has been shown to occur as late as 20 years after the primary diagnosis of RCC [6]. The fact that pancreatic metastases of RCC often occur late, taken together with the fact that the pancreas is frequently the only organ to be affected by metachronous RCC metastases, has prompted authors to suggest a “seed and soil” theory in the context of RCC metastases to the pancreas. Accordingly, disseminated tumor cells spread through the vascular system and may only develop into metastases at locations where specific interactions between tumor cells and host tissue allow for maturation to metastases, for example, facilitated by the formation of a premetastatic niche, specific chemokine interactions between tumor cells and host tissue, and tumor-cell-favoring immunoediting in/of the host tissue [7]. However, the exact reasons for the “pancreatotropism” of RCC cells remains unknown. Patients resected for pancreas metastases from RCC seem to have a significantly more favorable prognosis compared with patients resected for pancreas metastases of other origin [5]. This is in line with findings indicating that ccRCC with pancreatic metastasis is characterized by high vascularization, homogeneous growth pattern, and low-grade nuclei in histopathology as well as a less aggressive molecular profile compared with ccRCC with nonpancreatic metastases [8]. This suggests that ccRCC with (exclusive) metastases to the pancreas represent a specific rather indolent subset of ccRCC. The above-described histomorphological aspects can also be found in the present case (Fig. 3).
In the present case, late metachronous RCC metastasis were found concomitant with PDAC. Patients with RCC generally seem to have a significantly higher risk of developing various other primary malignancies [9]. As chemotherapy and/or radiation are usually not part of the initial therapy of RCC, it can be hypothesized that these additional primary malignancies in RCC patients are not therapy induced, but rather linked to exogenous and/or genetic factors. For example, tobacco smoking is a risk factor shared by various cancers, including RCC and PDAC. Pathogenic variants of VHL (Von Hippel–Lindau tumor suppressor) are a major genetic parameter linked to ccRCC. Patients with VHL disease harbor an increased risk of developing ccRCC, among other tumors [10]. In addition, 46–82% of sporadic ccRCC contain pathogenic mutations of VHL [10]. Patients with VHL disease are also prone to develop serous cystic neoplasms (SCN) of the pancreas and pancreatic neuroendocrine neoplasms (pNEN); however, neither VHL disease and nor sporadic VHL variants are linked to PDAC [11].
Cases of synchronous or metachronous RCC and PDAC in the pancreas have rarely been reported in the literature. In 2012, a retrospective study of 1178 patients with pancreatic cancer found 16 cases of PDAC or intraductal papillary mucinous neoplasm (IPMN) in the pancreas with synchronous or metachronous RCC. Among 12 PDAC patients, 9 patients with metachronous RCC, but only 3 patients with synchronous RCC, were identified [12]. In Table 2, an overview is given of the available cases in the English-speaking literature, with detailed information on simultaneously occurring PDAC and RCC.
Table 2
Overview of literature on simultaneously occurring pancreatic ductal adenocarcinoma (PDAC) and renal cell carcinoma (RCC)
Age
Sex
Subtype and localization of RCC
Procedure for RCC
Localization of PDAC
Procedure for PDAC
Follow-up
Source
78
M
ccRCC, two pancreas body metastases
Total pancreatectomy
Pancreatic body/tail
Total pancreatectomy
Perioperative death*
Present case
70
M
ccRCC, kidney (exact site unknown)
Partial nephrectomy
Proximal pancreas
ppWhipple
35 months, alive
[12]
70
M
ccRCC, kidney (exact site unknown)
Partial nephrectomy
Proximal pancreas
ppWhipple
5.25 months, dead
[12]
71
M
Papillary RCC, kidney (exact site unknown)
Partial nephrectomy
Proximal pancreas
ppWhipple
3.25 months, dead
[12]
67
M
ccRCC, left kidney
Left nephrectomy
Pancreas head
ppWhipple
15 months, dead
[13]
62
M
Papillary RCC, right kidney
Radical right nephrectomy
Pancreas body
Unknown; not resected
Unknown
[14]
70
F
ccRCC, right kidney
Chemotherapy
Pancreas body
Chemotherapy
13 months, dead
[15]
PDAC pancreatic ductal adenocarcinoma, (cc)RCC (clear-cell) renal cell carcinoma, F female, M male, pp pylorus-preserving
*Death 15 days after surgery
The strength of our case report resides in the detailed radiological and pathological evaluation of the simultaneously and cospatially occurring PDAC and pancreatic ccRCC metastases. However, its limitation is the short follow-up period due to the patient’s postoperative death. Of course, reasons for and against surgery must be carefully weighed in a case with extensive involvement of the pancreas by multiple masses in a patient with relevant comorbidities and reduced general health condition. Decision for surgery was made in the present case in an interdisciplinary tumor board despite the patient’s advanced age and recent myocardial infarction during FOLFIRINOX therapy, as PDAC was classified as borderline resectable, he was in stable general condition, and he expressed an explicit wish to undergo surgery.
In the present case, the primary RCC occurred antecedent to the primary PDAC; however, RCC metastases and PDAC occurred simultaneously and in the same location. It is speculative whether in this case the presence of PDAC might have played a role in the development of pancreatic RCC metastasis, for example, by creating a favorable local immune environment, or vice versa.
Generally, the case presented here illustrates that, although preoperative diagnostic tools have reached a high standard of quality, the validity of a preoperative diagnosis can still be limited, underlining the importance of pathological examination in the diagnostic process.

Conclusions

Although RCC metastases in the pancreas are rare, and their combination with PDAC even more so, clinicians, radiologists, and pathologists should be aware of this possibility. Interpretation of imaging in such cases may be highly challenging, especially after preoperative treatment. Hence, close interdisciplinary collaboration is essential in the diagnostic process.

Acknowledgements

Not applicable.

Declarations

The patient received standard-of-care diagnostics and therapy. No experimental treatment was applied.
Written informed consent was obtained from the next of kin of the patient for publication of this case report and any accompanying images. A copy of the written consent is available for review by the Editor-in-Chief of this journal.

Competing interests

The authors declare no competing interests.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

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 Allgemeinmedizin

Kombi-Abonnement

Mit e.Med Allgemeinmedizin erhalten Sie Zugang zu allen CME-Fortbildungen und Premium-Inhalten der allgemeinmedizinischen Zeitschriften, inklusive einer gedruckten Allgemeinmedizin-Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Hruban RH, Pitman MB, Klimstra DS. Tumors of the pancreas: In: AFIP Atlas of Tumor Pathology, series 4, fascicle 6. Washington, DC; 2007. Hruban RH, Pitman MB, Klimstra DS. Tumors of the pancreas: In: AFIP Atlas of Tumor Pathology, series 4, fascicle 6. Washington, DC; 2007.
2.
Zurück zum Zitat Verbeke CS, Haeberle L, Lenggenhager D, Esposito I. Pathology assessment of pancreatic cancer following neoadjuvant treatment: time to move on. Pancreatology. 2018;18:467–76.CrossRef Verbeke CS, Haeberle L, Lenggenhager D, Esposito I. Pathology assessment of pancreatic cancer following neoadjuvant treatment: time to move on. Pancreatology. 2018;18:467–76.CrossRef
3.
Zurück zum Zitat Adsay NV, Andrea A, Basturk O, Kilinc N, Nassar H, Cheng JD. Secondary tumors of the pancreas: an analysis of a surgical and autopsy database and review of the literature. Virchows Arch. 2004;444:527–35.CrossRef Adsay NV, Andrea A, Basturk O, Kilinc N, Nassar H, Cheng JD. Secondary tumors of the pancreas: an analysis of a surgical and autopsy database and review of the literature. Virchows Arch. 2004;444:527–35.CrossRef
5.
Zurück zum Zitat Adler H, Redmond CE, Heneghan HM, Swan N, Maguire D, Traynor O, et al. Pancreatectomy for metastatic disease: a systematic review. Eur J Surg Oncol. 2014;40:379–86.CrossRef Adler H, Redmond CE, Heneghan HM, Swan N, Maguire D, Traynor O, et al. Pancreatectomy for metastatic disease: a systematic review. Eur J Surg Oncol. 2014;40:379–86.CrossRef
6.
Zurück zum Zitat Wente MN, Kleeff J, Esposito I, Hartel M, Müller MW, Fröhlich BE, et al. Renal cancer cell metastasis into the pancreas: a single-center experience and overview of the literature. Pancreas. 2005;30:218–22.CrossRef Wente MN, Kleeff J, Esposito I, Hartel M, Müller MW, Fröhlich BE, et al. Renal cancer cell metastasis into the pancreas: a single-center experience and overview of the literature. Pancreas. 2005;30:218–22.CrossRef
7.
Zurück zum Zitat Sellner F. Observations on solitary versus multiple isolated pancreatic metastases of renal cell carcinoma: another indication of a seed and soil mechanism? Cancers (Basel). 2019;11:1379.CrossRef Sellner F. Observations on solitary versus multiple isolated pancreatic metastases of renal cell carcinoma: another indication of a seed and soil mechanism? Cancers (Basel). 2019;11:1379.CrossRef
8.
Zurück zum Zitat Singla N, Xie Z, Zhang Z, Gao M, Yousuf Q, Onabolu O, et al. Pancreatic tropism of metastatic renal cell carcinoma. JCI Insight. 2020;5:e134564. Singla N, Xie Z, Zhang Z, Gao M, Yousuf Q, Onabolu O, et al. Pancreatic tropism of metastatic renal cell carcinoma. JCI Insight. 2020;5:e134564.
9.
Zurück zum Zitat Beisland C, Talleraas O, Bakke A, Norstein J. Multiple primary malignancies in patients with renal cell carcinoma: a national population-based cohort study. BJU Int. 2006;97:698–702.CrossRef Beisland C, Talleraas O, Bakke A, Norstein J. Multiple primary malignancies in patients with renal cell carcinoma: a national population-based cohort study. BJU Int. 2006;97:698–702.CrossRef
10.
Zurück zum Zitat Cowey CL, Rathmell WK. VHL gene mutations in renal cell carcinoma: role as biomarker of disease outcome and drug efficacy. Curr Oncol Rep. 2010;11:94–101.CrossRef Cowey CL, Rathmell WK. VHL gene mutations in renal cell carcinoma: role as biomarker of disease outcome and drug efficacy. Curr Oncol Rep. 2010;11:94–101.CrossRef
11.
Zurück zum Zitat Lonser RR, Glenn GM, Walther M, Chew EY, Libutti SK, Linehan WM, et al. Von Hippel-Lindau disease. Lancet. 2003;361:2059–67.CrossRef Lonser RR, Glenn GM, Walther M, Chew EY, Libutti SK, Linehan WM, et al. Von Hippel-Lindau disease. Lancet. 2003;361:2059–67.CrossRef
12.
Zurück zum Zitat Müller SA, Pahernik S, Hinz U, Martin DJ, Wente MN, Hackert T, et al. Renal tumors and second primary pancreatic tumors: a relationship with clinical impact? Patient Saf Surg. 2012;6:18.CrossRef Müller SA, Pahernik S, Hinz U, Martin DJ, Wente MN, Hackert T, et al. Renal tumors and second primary pancreatic tumors: a relationship with clinical impact? Patient Saf Surg. 2012;6:18.CrossRef
13.
Zurück zum Zitat Alexakis N, Bosonnet L, Connor S, Ellis I, Sutton R, Campbell F, et al. Double resection for patients with pancreatic cancer and a second primary renal cell cancer. Dig Surg. 2003;20:428–32.CrossRef Alexakis N, Bosonnet L, Connor S, Ellis I, Sutton R, Campbell F, et al. Double resection for patients with pancreatic cancer and a second primary renal cell cancer. Dig Surg. 2003;20:428–32.CrossRef
14.
Zurück zum Zitat Ismail TO, Janane A, Hajji F, Dekkak Y, Bekkali S, Sossa J, et al. Synchronous primary tumor of kidney and pancreas: case report. Afr J Urol. 2010;16:128–31. Ismail TO, Janane A, Hajji F, Dekkak Y, Bekkali S, Sossa J, et al. Synchronous primary tumor of kidney and pancreas: case report. Afr J Urol. 2010;16:128–31.
15.
Zurück zum Zitat Mahfoud T, Tanz R, Khmamouche MR, Allaoui M, Belbaraka R, Khouchani M, Ichou M. Synchronous primary renal cell carcinoma and pancreatic ductal adenocarcinoma: case report and literature review. Case Rep Oncol. 2017;10:1050–6.CrossRef Mahfoud T, Tanz R, Khmamouche MR, Allaoui M, Belbaraka R, Khouchani M, Ichou M. Synchronous primary renal cell carcinoma and pancreatic ductal adenocarcinoma: case report and literature review. Case Rep Oncol. 2017;10:1050–6.CrossRef
Metadaten
Titel
Pancreatic ductal adenocarcinoma concomitant with pancreatic metastases of clear-cell renal cell carcinoma: a case report
verfasst von
Lena Haeberle
Melanie Busch
Julian Kirchner
Georg Fluegen
Gerald Antoch
Wolfram Trudo Knoefel
Irene Esposito
Publikationsdatum
01.12.2021
Verlag
BioMed Central
Erschienen in
Journal of Medical Case Reports / Ausgabe 1/2021
Elektronische ISSN: 1752-1947
DOI
https://doi.org/10.1186/s13256-021-02768-8

Weitere Artikel der Ausgabe 1/2021

Journal of Medical Case Reports 1/2021 Zur Ausgabe