Skip to main content
Erschienen in: Pathology & Oncology Research 2/2020

Open Access 17.06.2019 | Review

Solid-pseudopapillary Neoplasms of the Pancreas is still an Enigma: a Clinicopathological Review

verfasst von: Attila Zalatnai, Viktória Kis-Orha

Erschienen in: Pathology & Oncology Research | Ausgabe 2/2020

Abstract

The solid-pseudopapillary neoplasm of the pancreas is a rare but enigmatic entity occurring mainly in young women. Since the first description by V. Frantz in 1959 the terminology of this tumor has continuously changed but it has remained simply descriptive, because the exact histogenesis is still obscure. Although in majority of cases the survival is excellent, nevertheless, the expected prognosis is not exactly predictable. In this review the authors aim to summarize its clinico-pathological features, the expected biological behavior, the molecular alterations, the immune phenotype and discuss the putative histogenesis. From diagnostic point of view, the salient histological characteristic findings are analyzed that would help to differentiate it from other, look-alike pancreatic tumors, and suggestions are made about the desirable content of the histological report.
Hinweise
Attila Zalatnai and Viktória Kis-Orha contributed equally to this work.

Publisher’s Note

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

Introduction

Since the first description of a peculiar pancreatic tumor in 1959 [1], its terminology has been changed many times indicating that the exact nature and origin of this entity has remained speculative. In the original description the tumor was classified as a benign exocrine glandular lesion, most probably a papillary cystadenoma, although it was stated that ″accurate interpretation of these neoplasms is difficult”. It has long been disregarded since the 1978 WHO did not even mention. In the former literature it could be found under various names (solid-cystic tumor, solid-cystic acinar tumor, papillary-cystic tumor, solid-papillary tumor), but indeed, the actually used” solid- pseudopapillary neoplasm” (SPN) is similarly merely a descriptive designation denoting the morphological features, but leaving the histogenesis open.

Clinical Findings

The SPN is a rare pancreatic tumor: during 10 to 35 years of period various institutes have reported 9–187 pathologically diagnosed cases [25]. It may occur in both sexes, with a female predominance. Familiarity is not a feature. The median age is about 25–35 years, but children and old patients are equally affected; it may occur in an 8-year-old child up to 75 years of age. (Between 2007 and 2018 at our institute 13 SPNs have been diagnosed, all of them were female (11–59 years). The clinical signs are typically insignificant: it may be painless, or the patients may just complain of an undefined, slight, upper abdominal pain. It is not accompanied by general symptoms, hormonal activity is absent, the lab findings are normal; therefore, early diagnosis is usually not possible. An exceptional childhood case (a ruptured SPN following a blunt abdominal trauma) was reported by Tajima et al., but later on this tumor proved to be malignant [9]. A spontaneous rupture in a pregnant woman was also documented [10], but there is no evidence that the pregnancy itself could facilitate breakup in SPN. Regarding the localization, there is no preference of it; any part of the pancreas can be the site of origin. Although it is a primary pancreatic neoplasm, exceedingly rarely it may also occur in extrapancreatic places (omentum, adrenal or mesentery) [1113]. As a rule, the neoplasm presents as a solitary lesion, the multicentric manifestation is a curiosity [14].
Although SPN does occur in females and males, some gender differences are observed. The affected males are usually older, but the tumor size, the location or the clinical symptoms do not differ significantly [3, 15].

Pathological Characteristics

Because the clinical symptoms are usually vague or even the tumor can be an incidental finding, at the time of discovery it may be bulky; the mean size is about 6–8 cm, and the diameter may reach up to 15–22 cm [68]. Macroscopically, the sharp demarcation from the pancreatic tissue is typical, making the surgical removal easy (Fig. 1a) Sometimes it can be surrounded by a delicate or thick capsule. By palpation it has a rubbery feeling, and the cut surface is rather characteristically spongy in appearance. When the tumor is voluminous, extensive necrotic and hemorrhagic areas may be seen. (Fig. 1b).
Histologically, according to the terminology, two tissue patterns are observed: large areas of solid sheets of cells are randomly mixed with pseuodopapillary structures (Figs. 1e-f). The separation from the pancreatic tissue is sharp, or it displays a collagenous capsule (Fig. 1c). Plenty of vascular channels are seen, and in the stroma variable amount of hyalinized areas are noted (Figs.1d, g). The cells are pale, roundish, characteristically monomorphous, the nuclei are oval in shape and are frequently grooved, the nucleoli are marginated (Fig. 1h). In some areas the cells may have a foamy cytoplasm. A frequent, rather typical feature is the presence of PAS-positive globules in grouping, and the cholesterol clefts are also commonly seen (Figs. 1i-j). Mitotic figures are rarely seen (0–6/20 HPF) with no occurrence of atypical forms, and the Ki-67 score is very low (Fig. 2a). Presence of psammoma bodies is a rare finding.
A conspicuous but unexplained phenomenon was reported by Japanese authors comparing SPN in females and males. They could not observe fibrous capsule and cholesterol clefts in the male tumors, but these histological signs were seen in more than 60% among females. There were slight, but not significant differences regarding the capillary density, but the cystic degenerations or the necrotic areas occurred at the same frequency [15].
Very rarely, peculiar, pigmented variants may also occur [16, 17]. In these cases either a large amount of lipofuscin or melanin is accumulated.

Molecular Characteristics

Several studies are available about the molecular alterations in SPN [1823]. The tumor has complex karyotypic changes involving chromosomes 2, 4 or X, including breakpoints, bands or monosomy. Loss of heterozygosity for HRAS was also identified [18]. Hundreds to thousands of genes are differently expressed among them tumor associated genes. Most papers underline the importance of disrupted Wnt/β-catenin signaling pathways with concomitant cyclin D1 overexpression. Characteristic finding is the mutation in exon 3 of CTNNB1, but activated Hedgehog, androgen receptor, epithelial-mesenchymal transition (EMT)-coupled genes have also been identified, and several, closely associated miRNAs, especially the miR-200 family. Upregulated p27, p21 are typical, but no p53 or K-ras mutations are present. The ErbB and GnRH signaling pathways are also disturbed.
Proteomic profiles were also examined with high-resolution mass spectrometry [24]. More than 300 differentially expressed (both up- and downregulated) proteins have been identified. In addition to the proteins involved in Wnt-signaling like FUS or NONO, overexpressed molecules involved in glycolysis, including HK1, ENO2, PKM2 were found in accordance with their mRNA levels. The presented data indicate that SPN is a distinct pancreatic entity.

Biological Behavior

In the earlier international classifications SPN was positioned into the group of borderline tumors, but recently the WHO categorizes it as a malignant neoplasm. The scene is, however, intriguing, because the expected biology is unpredictable.
It is generally accepted that the majority of the tumor runs a benign course. In large-scale studies 2.1–22.8% of the treated SPNs proved to be malignant based on local recurrences or metastases [5, 2528]. The 5-year survival is excellent, reaching 93.6–98.8% [26, 29, 30], and even the 10-year disease-specific survival rate is 96% [31]. The local recurrence rate is low (less than 10% after 5 years of resection) [31]. Women and men have the same prognosis [3] and the multicentric forms have similar clinical and pathologic features to the solitary ones [14]. However, when the tumors are located in the head, the disease-free and the overall survivals are significantly shorter than those of other locations [32]. When metastases develop, they usually occur late, 8 to 15.8 years after complete resection of the primary [21, 31, 33, 34]. This indolent behavior might be due to diploid DNA content and a long (765 days) doubling time [11, 35].

Solid-Pseudopapillary Carcinoma. Signs of Malignancy

A minority of SPN recurs or even metastasizes despite the bland histological appearance. Although the local recurrence may lead to death, but most of fatal cases arise from liver metastases. Interestingly, the bones, the lungs are not involved, the peritoneal carcinosis is also rarely seen [36, 37]. Widespread metastatic spread (liver, stomach, adrenal gland, lymph node, peritoneum) is even more infrequent event [38].
A burning issue is how to predict the potentially malignant behavior of this tumor, in absence of frankly worrisome histological signs? Unfortunately, no clear-cut answer exists, there are various approaches in the literature, sometimes with alternate results. Some authors suggested that the large tumor size (> 5 cm) and the young age increase the risk of recurrence [28, 39, 40]. It seems logical that the inadequate resection, the positive surgical margins, capsular invasion or even the tumor rupture would be an important predictor [28, 31, 4042], but some other authors have found that these findings have no predictive value [8, 39]. While the perineural invasion is a classical pathohistological sign in the malignant tumors, this finding is not a reliable indicator of aggressiveness in SPN [8, 39]. Similarly, the mitotic rate or the cellular atypia do not necessarily forecast the recurrence [8].
The tumor typically shows a very low proliferative activity; the Ki-67 rate is 1–2%. Some authors have claimed, when the neoplasm displays an elevated Ki-67 score, a malignant course is expected: high values were detected in 66.7% in malignant cases, but only 8.4% when the tumor was classified as benign (p < 0.001) [27]. Yang et al. have found that the ≥4% score was associated with poorer recurrence-free survival and in these patients an adverse outcome was expected [29]. In another study this cut-off level was 5% [43].
Some other, potential predictive signs have been published, but still they need further reinforcement. Such a marker could be the galectin-3, which is useful to distinguish SPN from neuroendocrine tumors, but it is also interesting that its low immunohistochemical expression is associated with metastatic spreading [44]. Although the molecular mutation profiles are rarely studied at this context, in a single malignant SPN an uncommon EGFR mutation was identified at L861Q by pyrosequencing [45].
Despite all the above mentioned findings, the assessment of the potentially malignant behavior of SPN remains unpredictable. Even the most careful pathological examination cannot exactly identify patients who may develop recurrence after curative surgical operation. Suggestive signs are capsular infiltration, perineural or vascular invasion, brisk mitotic activity, striking polymorphism, but the absence of these features is not exclusive for malignancy [46, 47].

Immune Profile

The classical histopathological appearance usually allows the correct diagnosis, but some pancreatic tumors may show similarities and for differential diagnostic reason various complex immunohistochemical examinations are needed. In addition, these markers may serve aids to the possible histogenesis of SPN.
In the literature a large number of antibodies have been checked, but sometimes the results seem to be conflicting. There is a general agreement that practically all of these tumors are positive for vimentin [7. 31, 48, 49], β-catenin [7, 31, 43, 49, 50] (Fig. 2b), cyclin D1 [31, 50] (Fig. 2c), alpha-1-antitrypsin (AAT) [11, 48] (Fig. 2d) or CD56 [12, 17, 31, 51] (Fig. 2e). The loss of E-cadherin expression is also a typical finding [49, 52] (Fig. 2f). Presence of progesterone receptor is regularly detected [8, 12, 31, 48, 50] (Fig. 2g), androgen receptor is expressed in about 80% of cases [43], but the estrogen receptor is usually negative [48, 50, 53]. The cytokeratin expression is highly variable, between 28 to 70% [7, 8, 48], but the CK7 is negative [49]. Most SPNs are positive for CD10, usually with a perinuclear dot pattern [7, 8, 31, 51], and the CD99 was also reported as a reliable positive marker [49, 52] (Fig. 2h). Among the neuroendocrine markers the chromogranin-A is usually absent [7, 48, 50], the NSE (although its specificity is questionable) may either be negative or consistently positive [6, 48], while in a small percentage of tumors synaptophysin is detectable [8, 48]. Because SPN is not regarded as a hormonally active tumor, specific pancreatic hormones are not found in the cells [50]. The TFE3, a transcription factor promoting several genes involved in cell proliferation and growth, proved to be a highly sensitive marker (75–96% of cases), and can be used for reinforcement of the SPN diagnosis [43, 54, 55].
There are some other, seemingly unrelated immune positivities (LEF-1, FUS, WIF-1, CD200) that are expressed with a high frequency [43, 56], but their significance and applicability are far from obvious in this tumor.
The SPN, the neuroendocrine tumors and the acinar cell carcinomas sometimes may show similar histological patterns making their distinction difficult. Based on the immunohistochemical staining properties, some differential diagnostic approaches have been advised. TFE3 expression, for example, is very frequent (94%) in SPN, while the pancreatic neuroendocrine tumors (PNET) are positive just in 25%, and the acinar carcinomas are negative [55]. Similarly, β-catenin, glypican-3 or galectin-3 are positive in SPN, but the neuroendocrine tumors are negative [44, 54, 57]. A new finding was recently reported by Shen et al., that the α-Methylacyl-CoA racemase (AMACR, P504S), which marker is a very useful diagnostic tool in urological malignancies, is positively stained in SPN, but not in the others [58]. CD200, however, is highly expressed either in SPN or in PNET (100% vs. 96%), so it cannot be used for differential diagnosis [56].

Histogenesis

Despite the extensive investigations, the histogenesis has remained obscure and still hypothetical, because the results from different studies are conflicting. Although vimentin, AAT or NSE are positive in most cases, this phenotype in not specific and does not define a specific lineage [59]. The electron microscopical (EM) studies would be a good tool in this context, however, the findings are incongruent. The presence of zymogen granules and the positive trypsin-stain would indicate an acinar character [6, 60, 61], but other authors failed to detect them [53, 62]. In the vast majority of studies, no neurosecretory granules were found [53, 60, 62], only occasional studies claimed their presence in about 50% of the cells [63]. The strong galectin-3 expression also differentiates it from the galectin-3-negative neuroendocrine tumors [44]. The ductal origin is similarly very unlikely: ultrastructurally the tumors cells do not display ductal cell character [61], K-ras mutation is not detectable [22], the CK19 – which is a characteristically positive marker in conventional adenocarcinoma – is also absent [64]. Based on both immunohistochemical and ultrastructural features, Kallichandra et al. proposed that the centroacinar cells would be origin of SPN [65, 66], although the CK7 is negative in this tumor, and among our cases only 1 out of 13 displayed just 10% positivity of the cells. The neural crest [16] or the genital ridge/ovarian anlage attached to the pancreatic tissue [48] have also been proposed. Because of the highly divergent immunohistochemical findings, it fails to reveal a definite phenotypic relationship with any clearly defined lineage [48, 50], some authors suggest that SPN may originate from totipotent primordial cells that would further differentiate toward duct epithelial, acinar or endocrine patterns [55, 66]. This would explain the different immune profiles behind the identical morphology. This hypothesis, however, does not lie on solid evidence, because the genes expressed during pancreatic organogenesis and determine the lineage developmental routes are not regularly identifiable in SPN. So, the histogenesis still remained enigmatic.

Therapeutic Considerations

Because the SPN mostly appears as a localized tumor, the surgery remains the mainstay of therapy, because the well-circumscribed tumors usually show a favorable prognosis [67]. In the recurrent or metastatic cases, however, various other, but not standardized techniques are available. Among them a tyrosine kinase inhibitor sunitinib and hepatic arterial embolisation [68], hyperthermic intraperitoneal chemotherapy are reported [33, 40], but there are occasional limited experience about the palliative radiotherapy or even liver transplantation [69, 70].

Important Practical Pathological Considerations

Although in the majority of cases the pathological diagnosis of SPN is quite easy, the major differential diagnostic challenges are the neuroendocrine tumor and the acinar cell carcinoma, because their morphology may sometimes overlap. Although the aspecific NSE is usually positive, but the chromogranin-A or synaptophysin are negative, moreover, no hormonal activity is identified in this tumor. The nuclear expression of β-catenin, SOX-11 and TFE3 positivities are characteristic findings in SPN [71], but not in PNET and in acinar cell carcinoma (Fig. 2i). Similarly, the rarely used claudin 5 and 7 can also distinguish these entities [72]. The loss of E-cadherin immune staining is also a reliable, typical reaction in SPN. In addition, Geers et al. have found galectin-3 as a useful positive marker, which is not found in the neuroendocrine tumors [44], similarly to CD99 [49. 52]. The major points are presented in Table 1.
Table 1
Differential diagnostic approaches among the look-alike pancreatic lesions
 
SPN
PNET
ACC
β-catenin
nuclear, +++
negative
negative
TFE3
94%
25%
negative
E-cadherin
loss of positivity
positive
positive
galectin-3
positive
negative
negative
CD56
positive
positive
negative
claudin 5
positive
negative
negative
claudin 7
±
+++
+++
SOX-11
positive
negative
negative
Pdx-1
negative
positive
positive
CD200
100%
96%
12,5% (focal)
SPN solid-pseudopapillary neoplasm, PNET pancreatic neuroendocrine tumor, ACC acinar cell carcinoma
Another pathological challenge is the assessment of its biological behavior, because despite the bland histology it may run a malignant course, and the classical signs of malignancy are not always present. The age, gender or even multiplicity do not seem to be decisive factors [3, 14, 73], but they should also be taken into account. Although it is not possible to foresee the expected prognosis with absolute certainty, the histological report should contain the (a) size, (b) presence/absence of capsule, (c) free/infiltrated surgical margins, (d) cellular atypia, (e) mitotic activity, (f) perineural/vascular infiltration, (g) expression of galectin-3, and the (h) Ki-67 score.

Conclusion

The rare solid-pseudopapillary neoplasm may develop in any age and in both sexes, but mainly young women are affected. Although the survival rate is usually high, however, the histological picture does not allow precisely predicting the expected biological behavior. There are some suspicious morphological signs, however, late recurrence, metastases or even death can occur with totally bland appearance. The exact histogenesis remains indeterminate, probably primordial cell underwent divergent differentiation; however, SPN is still an enigmatic tumor warranting further elucidation.

Compliance with Ethical Standards

Conflict of Interest

The authors declare no conflict of interest.
Open Access This 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.

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.

Literatur
1.
Zurück zum Zitat Frantz VK (1959) Tumors of the pancreas. Armed forces Institute of Pathology, Washington, DC Frantz VK (1959) Tumors of the pancreas. Armed forces Institute of Pathology, Washington, DC
2.
Zurück zum Zitat Yoon DY, Hines OJ, Bilchik AJ, Lewin K, Cortina G, Reber HA (2001) Solid and papillary epithelial neoplasms of the pancreas: aggressive resection for cure. Am Surg 67:1195–1199PubMed Yoon DY, Hines OJ, Bilchik AJ, Lewin K, Cortina G, Reber HA (2001) Solid and papillary epithelial neoplasms of the pancreas: aggressive resection for cure. Am Surg 67:1195–1199PubMed
3.
Zurück zum Zitat Cai YQ, Xie SM, Ran X, Wang X, Mai G, Liu XB (2014) Solid pseudopapillary tumor of the pancreas in male patients: report of 16 cases. World J Gastroenterol 20:6939–6945PubMedPubMedCentralCrossRef Cai YQ, Xie SM, Ran X, Wang X, Mai G, Liu XB (2014) Solid pseudopapillary tumor of the pancreas in male patients: report of 16 cases. World J Gastroenterol 20:6939–6945PubMedPubMedCentralCrossRef
4.
Zurück zum Zitat Shorter NA, Glick RD, Klimstra DS, Brennan MF, Laquaglia MP (2002) Malignant pancreatic tumors in childhood and adolescence: the memorial Sloan-Kettering experience, 1967 to present. J Pediatr Surg 37:887–892PubMedCrossRef Shorter NA, Glick RD, Klimstra DS, Brennan MF, Laquaglia MP (2002) Malignant pancreatic tumors in childhood and adolescence: the memorial Sloan-Kettering experience, 1967 to present. J Pediatr Surg 37:887–892PubMedCrossRef
5.
Zurück zum Zitat Wang WB, Zhang TP, Sun MQ, Peng Z, Chen G, Zhao YP (2014) Solid pseudopapillary tumor of the pancreas with liver metastasis: clinical features and management. Eur J Surg Oncol 40:1572–1577PubMedCrossRef Wang WB, Zhang TP, Sun MQ, Peng Z, Chen G, Zhao YP (2014) Solid pseudopapillary tumor of the pancreas with liver metastasis: clinical features and management. Eur J Surg Oncol 40:1572–1577PubMedCrossRef
6.
Zurück zum Zitat Lieber MR, Lack EE, Roberts JR Jr, Merino MJ, Patterson K, Restrepo C, Solomon D, Chandra R, Triche TJ (1987) Solid and papillary epithelial neoplasm of the pancreas. An ultrastructural and immunocytochemical study of six cases. Am J Surg Pathol 11:85–93PubMedCrossRef Lieber MR, Lack EE, Roberts JR Jr, Merino MJ, Patterson K, Restrepo C, Solomon D, Chandra R, Triche TJ (1987) Solid and papillary epithelial neoplasm of the pancreas. An ultrastructural and immunocytochemical study of six cases. Am J Surg Pathol 11:85–93PubMedCrossRef
7.
Zurück zum Zitat Uppin SG, Hui M, Thumma V, Challa S, Uppin MS, Bheerappa N, Sastry RA, Paul TR, Prayaga AK (2015) Solid-pseudopapillary neoplasm of the pancreas: a clinicopathological and immunohistochemical study of 33 cases from a single institution in southern India. Indian J Pathol Microbiol 58:163–169PubMedCrossRef Uppin SG, Hui M, Thumma V, Challa S, Uppin MS, Bheerappa N, Sastry RA, Paul TR, Prayaga AK (2015) Solid-pseudopapillary neoplasm of the pancreas: a clinicopathological and immunohistochemical study of 33 cases from a single institution in southern India. Indian J Pathol Microbiol 58:163–169PubMedCrossRef
8.
Zurück zum Zitat Nguyen NQ, Johns AL, Gill AJ, Ring N, Chang DK, Clarkson A, Merrett ND, Kench JG, Colvin EK, Scarlett CJ, Biankin AV (2011) Clinical and immunohistochemical features of 34 solid pseudopapillary tumors of the pancreas. J Gastroenterol Hepatol 26:267–274PubMedCrossRef Nguyen NQ, Johns AL, Gill AJ, Ring N, Chang DK, Clarkson A, Merrett ND, Kench JG, Colvin EK, Scarlett CJ, Biankin AV (2011) Clinical and immunohistochemical features of 34 solid pseudopapillary tumors of the pancreas. J Gastroenterol Hepatol 26:267–274PubMedCrossRef
9.
Zurück zum Zitat Tajima Y, Kohara N, Maeda J, Inoue K, Kitasato A, Natsuda K, Irie J, Adachi T, Kuroki T, Eguchi S, Kanematsu T (2012) Peritoneal and nodal recurrence 7 years after the excision of a ruptured solid pseudopapillary neoplasm of the pancreas: report of a case. Surg Today 42:776–780PubMedCrossRef Tajima Y, Kohara N, Maeda J, Inoue K, Kitasato A, Natsuda K, Irie J, Adachi T, Kuroki T, Eguchi S, Kanematsu T (2012) Peritoneal and nodal recurrence 7 years after the excision of a ruptured solid pseudopapillary neoplasm of the pancreas: report of a case. Surg Today 42:776–780PubMedCrossRef
10.
Zurück zum Zitat Huang SC, Wu TH, Chen CC, Chen TC (2013) Spontaneous rupture of solid pseudopapillary neoplasm of the pancreas during pregnancy. Obstet Gynecol 121(2 Pt 2 Suppl 1):486–488PubMed Huang SC, Wu TH, Chen CC, Chen TC (2013) Spontaneous rupture of solid pseudopapillary neoplasm of the pancreas during pregnancy. Obstet Gynecol 121(2 Pt 2 Suppl 1):486–488PubMed
11.
Zurück zum Zitat Fukunaga M (2001) Pseudopapillary solid cystic tumor arising from an extrapancreatic site. Arch Pathol Lab Med 125:1368–1371PubMed Fukunaga M (2001) Pseudopapillary solid cystic tumor arising from an extrapancreatic site. Arch Pathol Lab Med 125:1368–1371PubMed
12.
Zurück zum Zitat Zhu H, Xia D, Wang B, Meng H (2013) Extrapancreatic solid pseudopapillary neoplasm: report of a case of primary retroperitoneal origin and review of the literature. Oncol Lett 5:1501–1504PubMedPubMedCentralCrossRef Zhu H, Xia D, Wang B, Meng H (2013) Extrapancreatic solid pseudopapillary neoplasm: report of a case of primary retroperitoneal origin and review of the literature. Oncol Lett 5:1501–1504PubMedPubMedCentralCrossRef
13.
Zurück zum Zitat Wu H, Huang YF, Liu XH, Xu MH (2017) Extrapancreatic solid pseudopapillary neoplasm followed by multiple metastases: case report. World J Gastrointest Oncol 9:497–501PubMedPubMedCentralCrossRef Wu H, Huang YF, Liu XH, Xu MH (2017) Extrapancreatic solid pseudopapillary neoplasm followed by multiple metastases: case report. World J Gastrointest Oncol 9:497–501PubMedPubMedCentralCrossRef
14.
Zurück zum Zitat Li HX, Zhang Y, Du ZG, Tang F, Qi XQ, Yin B, Jiang YJ, Yang F, Subedi S (2013) Multi-centric solid-pseudopapillary neoplasm of the pancreas. Med Oncol 30:330PubMedCrossRef Li HX, Zhang Y, Du ZG, Tang F, Qi XQ, Yin B, Jiang YJ, Yang F, Subedi S (2013) Multi-centric solid-pseudopapillary neoplasm of the pancreas. Med Oncol 30:330PubMedCrossRef
15.
Zurück zum Zitat Hirabayashi K, Kurokawa S, Maruno A, Yamada M, Kawaguchi Y, Nakagohri T, Mine T, Sugiyama T, Tajiri T, Nakamura N (2015) Sex differences in immunohistochemical expression and capillary density in pancreatic solid pseudopapillary neoplasm. Ann Diagn Pathol 19:45–49PubMedCrossRef Hirabayashi K, Kurokawa S, Maruno A, Yamada M, Kawaguchi Y, Nakagohri T, Mine T, Sugiyama T, Tajiri T, Nakamura N (2015) Sex differences in immunohistochemical expression and capillary density in pancreatic solid pseudopapillary neoplasm. Ann Diagn Pathol 19:45–49PubMedCrossRef
16.
Zurück zum Zitat Chen C, Jing W, Gulati P, Vargas H, French SW (2004) Melanocytic differentiation in a solid pseudopapillary tumor of the pancreas. J Gastroenterol 39:579–583PubMedCrossRef Chen C, Jing W, Gulati P, Vargas H, French SW (2004) Melanocytic differentiation in a solid pseudopapillary tumor of the pancreas. J Gastroenterol 39:579–583PubMedCrossRef
17.
Zurück zum Zitat Daum O, Sima R, Mukensnabl P, Vanecek T, Brouckova M, Benes Z, Michal M (2005) Pigmented solid-pseudopapillary neoplasm of the pancreas. Pathol Int 55:280–284PubMedCrossRef Daum O, Sima R, Mukensnabl P, Vanecek T, Brouckova M, Benes Z, Michal M (2005) Pigmented solid-pseudopapillary neoplasm of the pancreas. Pathol Int 55:280–284PubMedCrossRef
18.
Zurück zum Zitat Kempski HM, Austin N, Chatters SJ, Toomey SM, Chalker J, Anderson J, Sebire NJ (2006) Previously unidentified complex cytogenetic changes found in a pediatric case of solid-pseudopapillary neoplasm of the pancreas. Cancer Genet Cytogenet 164:54–60PubMedCrossRef Kempski HM, Austin N, Chatters SJ, Toomey SM, Chalker J, Anderson J, Sebire NJ (2006) Previously unidentified complex cytogenetic changes found in a pediatric case of solid-pseudopapillary neoplasm of the pancreas. Cancer Genet Cytogenet 164:54–60PubMedCrossRef
19.
Zurück zum Zitat Tiemann K, Heitling U, Kosmahl M, Klöppel G (2007) Solid pseudopapillary neoplasms of the pancreas show an interruption of the Wnt-signaling pathway and express gene products of 11q. Mod Pathol 20:955–960PubMedCrossRef Tiemann K, Heitling U, Kosmahl M, Klöppel G (2007) Solid pseudopapillary neoplasms of the pancreas show an interruption of the Wnt-signaling pathway and express gene products of 11q. Mod Pathol 20:955–960PubMedCrossRef
20.
Zurück zum Zitat Park M, Kim M, Hwang D, Park M, Kim WK, Kim SK, Shin J, Park ES, Kang CM, Paik YK, Kim H (2014) Characterization of gene expression and activated signaling pathways in solid-pseudopapillary neoplasm of pancreas. Mod Pathol 27:580–593PubMedCrossRef Park M, Kim M, Hwang D, Park M, Kim WK, Kim SK, Shin J, Park ES, Kang CM, Paik YK, Kim H (2014) Characterization of gene expression and activated signaling pathways in solid-pseudopapillary neoplasm of pancreas. Mod Pathol 27:580–593PubMedCrossRef
21.
Zurück zum Zitat Müller-Höcker J, Zietz CH, Sendelhofert A (2001) Deregulated expression of cell cycle-associated proteins in solid pseudopapillary tumor of the pancreas. Mod Pathol 14:47–53PubMedCrossRef Müller-Höcker J, Zietz CH, Sendelhofert A (2001) Deregulated expression of cell cycle-associated proteins in solid pseudopapillary tumor of the pancreas. Mod Pathol 14:47–53PubMedCrossRef
22.
Zurück zum Zitat Abraham SC, Klimstra DS, Wilentz RE, Yeo CJ, Conlon K, Brennan M, Cameron JL, Wu TT, Hruban RH (2002) Solid-pseudopapillary tumors of the pancreas are genetically distinct from pancreatic ductal adenocarcinomas and almost always harbor beta-catenin mutations. Am J Pathol 160:1361–1369PubMedPubMedCentralCrossRef Abraham SC, Klimstra DS, Wilentz RE, Yeo CJ, Conlon K, Brennan M, Cameron JL, Wu TT, Hruban RH (2002) Solid-pseudopapillary tumors of the pancreas are genetically distinct from pancreatic ductal adenocarcinomas and almost always harbor beta-catenin mutations. Am J Pathol 160:1361–1369PubMedPubMedCentralCrossRef
23.
Zurück zum Zitat Zhang Y, Han X, Wu H, Zhou Y (2017) Bioinformatics analysis of transcription profiling of solid pseudopapillary neoplasm of the pancreas. Mol Med Rep 16:1635–1642PubMedPubMedCentralCrossRef Zhang Y, Han X, Wu H, Zhou Y (2017) Bioinformatics analysis of transcription profiling of solid pseudopapillary neoplasm of the pancreas. Mol Med Rep 16:1635–1642PubMedPubMedCentralCrossRef
24.
Zurück zum Zitat Park M, Lim JS, Lee HJ, Na K, Lee MJ, Kang CM, Paik YK, Kim H (2015) Distinct protein expression profiles of solid-Pseudopapillary neoplasms of the pancreas. J Proteome Res 14:3007–3014PubMedCrossRef Park M, Lim JS, Lee HJ, Na K, Lee MJ, Kang CM, Paik YK, Kim H (2015) Distinct protein expression profiles of solid-Pseudopapillary neoplasms of the pancreas. J Proteome Res 14:3007–3014PubMedCrossRef
25.
Zurück zum Zitat Xu Y, Zhao G, Pu N, Nuerxiati A, Ji Y, Zhang L, Rong Y, Lou W, Wang D, Kuang T, Xu X, Wu W (2017) One hundred twenty-one resected solid Pseudopapillary tumors of the pancreas: an 8-year single-institution experience at Zhongshan hospital, Shanghai, China. Pancreas 46:1023–1028PubMedCrossRef Xu Y, Zhao G, Pu N, Nuerxiati A, Ji Y, Zhang L, Rong Y, Lou W, Wang D, Kuang T, Xu X, Wu W (2017) One hundred twenty-one resected solid Pseudopapillary tumors of the pancreas: an 8-year single-institution experience at Zhongshan hospital, Shanghai, China. Pancreas 46:1023–1028PubMedCrossRef
26.
Zurück zum Zitat Yu PF, Hu ZH, Wang XB, Guo JM, Cheng XD, Zhang YL, Xu Q (2010) Solid pseudopapillary tumor of the pancreas: a review of 553 cases in Chinese literature. World J Gastroenterol 16:1209–1214PubMedPubMedCentralCrossRef Yu PF, Hu ZH, Wang XB, Guo JM, Cheng XD, Zhang YL, Xu Q (2010) Solid pseudopapillary tumor of the pancreas: a review of 553 cases in Chinese literature. World J Gastroenterol 16:1209–1214PubMedPubMedCentralCrossRef
27.
Zurück zum Zitat Yu P, Cheng X, Du Y, Yang L, Xu Z, Yin W, Zhong Z, Wang X, Xu H, Hu C (2015) Solid Pseudopapillary neoplasms of the pancreas: a 19-year multicenter experience in China. J Gastrointest Surg 19:1433–1440PubMedCrossRef Yu P, Cheng X, Du Y, Yang L, Xu Z, Yin W, Zhong Z, Wang X, Xu H, Hu C (2015) Solid Pseudopapillary neoplasms of the pancreas: a 19-year multicenter experience in China. J Gastrointest Surg 19:1433–1440PubMedCrossRef
28.
Zurück zum Zitat Kim CW, Han DJ, Kim J, Kim YH, Park JB, Kim SC (2011) Solid pseudopapillary tumor of the pancreas: can malignancy be predicted? Surgery 149:625–634PubMedCrossRef Kim CW, Han DJ, Kim J, Kim YH, Park JB, Kim SC (2011) Solid pseudopapillary tumor of the pancreas: can malignancy be predicted? Surgery 149:625–634PubMedCrossRef
29.
Zurück zum Zitat Yang F, Yu X, Bao Y, Du Z, Jin C, Fu D (2016) Prognostic value of Ki-67 in solid pseudopapillary tumor of the pancreas: Huashan experience and systematic review of the literature. Surgery 159:1023–1031PubMedCrossRef Yang F, Yu X, Bao Y, Du Z, Jin C, Fu D (2016) Prognostic value of Ki-67 in solid pseudopapillary tumor of the pancreas: Huashan experience and systematic review of the literature. Surgery 159:1023–1031PubMedCrossRef
30.
Zurück zum Zitat Hanada K, Kurihara K, Itoi T, Katanuma A, Sasaki T, Hara K, Nakamura M, Kimura W, Suzuki Y, Sugiyama M, Ohike N, Fukushima N, Shimizu M, Ishigami K, Gabata T, Okazaki K (2018) Clinical and pathological features of solid pseudopapillary neoplasms of the pancreas: a nationwide multicenter study in Japan. Pancreas 47:1019–1026PubMedCrossRef Hanada K, Kurihara K, Itoi T, Katanuma A, Sasaki T, Hara K, Nakamura M, Kimura W, Suzuki Y, Sugiyama M, Ohike N, Fukushima N, Shimizu M, Ishigami K, Gabata T, Okazaki K (2018) Clinical and pathological features of solid pseudopapillary neoplasms of the pancreas: a nationwide multicenter study in Japan. Pancreas 47:1019–1026PubMedCrossRef
31.
Zurück zum Zitat Serrano PE, Serra S, Al-Ali H, Gallinger S, Greig PD, McGilvray ID, Moulton CA, Wei AC, Cleary SP (2014) Risk factors associated with recurrence in patients with solid pseudopapillary tumors of the pancreas. JOP 15:561–568PubMed Serrano PE, Serra S, Al-Ali H, Gallinger S, Greig PD, McGilvray ID, Moulton CA, Wei AC, Cleary SP (2014) Risk factors associated with recurrence in patients with solid pseudopapillary tumors of the pancreas. JOP 15:561–568PubMed
33.
Zurück zum Zitat Honore C, Goere D, Dartigues P, Burtin P, Dumont F, Elias D (2012) Peritoneal carcinomatosis from solid pseudopapillary neoplasm (Frantz's tumour) of the pancreas treated with HIPEC. Anticancer Res 32:1069–1073PubMed Honore C, Goere D, Dartigues P, Burtin P, Dumont F, Elias D (2012) Peritoneal carcinomatosis from solid pseudopapillary neoplasm (Frantz's tumour) of the pancreas treated with HIPEC. Anticancer Res 32:1069–1073PubMed
34.
Zurück zum Zitat Gomez P, Yorke R, Ayala AG, Ro JY (2012) Solid-pseudopapillary neoplasm of pancreas with long delayed liver metastasis. Ann Diagn Pathol 16:380–384PubMedCrossRef Gomez P, Yorke R, Ayala AG, Ro JY (2012) Solid-pseudopapillary neoplasm of pancreas with long delayed liver metastasis. Ann Diagn Pathol 16:380–384PubMedCrossRef
35.
Zurück zum Zitat Kato T, Egawa N, Kamisawa T, Tu Y, Sanaka M, Sakaki N, Okamoto A, Bando N, Funata N, Isoyama T (2002) A case of solid pseudopapillary neoplasm of the pancreas and tumor doubling time. Pancreatology 2:495–498PubMedCrossRef Kato T, Egawa N, Kamisawa T, Tu Y, Sanaka M, Sakaki N, Okamoto A, Bando N, Funata N, Isoyama T (2002) A case of solid pseudopapillary neoplasm of the pancreas and tumor doubling time. Pancreatology 2:495–498PubMedCrossRef
36.
Zurück zum Zitat Estrella JS, Li L, Rashid A, Wang H, Katz MH, Fleming JB, Abbruzzese JL, Wang H (2014) Solid pseudopapillary neoplasm of the pancreas: clinicopathologic and survival analyses of 64 cases from a single institution. Am J Surg Pathol 38:147–157PubMedCrossRef Estrella JS, Li L, Rashid A, Wang H, Katz MH, Fleming JB, Abbruzzese JL, Wang H (2014) Solid pseudopapillary neoplasm of the pancreas: clinicopathologic and survival analyses of 64 cases from a single institution. Am J Surg Pathol 38:147–157PubMedCrossRef
37.
Zurück zum Zitat Ercelep O, Ozdemir N, Turan N, Topcu TO, Uysal M, Tanriverdi O, Demirci U, Taskoylu BY, Urakcı Z, Duran AO, Aksoy A, Menekse S, Ozcelik M, Gumus M (2019) Retrospective evaluation of patients diagnosed with solid pseudopapillary neoplasms of the pancreas. Curr Probl Cancer 43:26–32 Ercelep O, Ozdemir N, Turan N, Topcu TO, Uysal M, Tanriverdi O, Demirci U, Taskoylu BY, Urakcı Z, Duran AO, Aksoy A, Menekse S, Ozcelik M, Gumus M (2019) Retrospective evaluation of patients diagnosed with solid pseudopapillary neoplasms of the pancreas. Curr Probl Cancer 43:26–32
38.
Zurück zum Zitat Prakash PS, Chan DYS, Madhavan K (2018) The stomach: a rare site for metastatic solid pseudopapillary neoplasm of the pancreas. J Gastrointest Surg 22:759–760PubMedCrossRef Prakash PS, Chan DYS, Madhavan K (2018) The stomach: a rare site for metastatic solid pseudopapillary neoplasm of the pancreas. J Gastrointest Surg 22:759–760PubMedCrossRef
39.
Zurück zum Zitat Gao H, Gao Y, Yin L, Wang G, Wei J, Jiang K, Miao Y (2018) Risk factors of the recurrences of pancreatic solid Pseudopapillary tumors: a systematic review and meta-analysis. J Cancer 9:1905–1914PubMedPubMedCentralCrossRef Gao H, Gao Y, Yin L, Wang G, Wei J, Jiang K, Miao Y (2018) Risk factors of the recurrences of pancreatic solid Pseudopapillary tumors: a systematic review and meta-analysis. J Cancer 9:1905–1914PubMedPubMedCentralCrossRef
40.
Zurück zum Zitat Irtan S, Galmiche-Rolland L, Elie C, Orbach D, Sauvanet A, Elias D, Guérin F, Coze C, Faure-Conter C, Becmeur F, Demarche M, Galifer RB, Galloy MA, Podevin G, Aubert D, Piolat C, De Lagausie P, Sarnacki S (2016) Recurrence of solid Pseudopapillary neoplasms of the pancreas: results of a Nationwide study of risk factors and treatment modalities. Pediatr Blood Cancer 63:1515–1521PubMedCrossRef Irtan S, Galmiche-Rolland L, Elie C, Orbach D, Sauvanet A, Elias D, Guérin F, Coze C, Faure-Conter C, Becmeur F, Demarche M, Galifer RB, Galloy MA, Podevin G, Aubert D, Piolat C, De Lagausie P, Sarnacki S (2016) Recurrence of solid Pseudopapillary neoplasms of the pancreas: results of a Nationwide study of risk factors and treatment modalities. Pediatr Blood Cancer 63:1515–1521PubMedCrossRef
41.
Zurück zum Zitat Song H, Dong M, Zhou J, Sheng W, Zhong B, Gao W (2017) Solid Pseudopapillary neoplasm of the pancreas: Clinicopathologic feature, risk factors of malignancy, and survival analysis of 53 cases from a single center. Biomed Res Int 2017:5465261PubMedPubMedCentral Song H, Dong M, Zhou J, Sheng W, Zhong B, Gao W (2017) Solid Pseudopapillary neoplasm of the pancreas: Clinicopathologic feature, risk factors of malignancy, and survival analysis of 53 cases from a single center. Biomed Res Int 2017:5465261PubMedPubMedCentral
42.
Zurück zum Zitat Shimizu M, Matsumoto T, Hirokawa M, Monobe Y, Iwamoto S, Tsunoda T, Manabe T (1999) Solid-pseudopapillary carcinoma of the pancreas. Pathol Int 49:231–234PubMedCrossRef Shimizu M, Matsumoto T, Hirokawa M, Monobe Y, Iwamoto S, Tsunoda T, Manabe T (1999) Solid-pseudopapillary carcinoma of the pancreas. Pathol Int 49:231–234PubMedCrossRef
43.
Zurück zum Zitat Kim EK, Jang M, Park M, Kim H (2017) LEF1, TFE3 and AR are putative diagnostic markers of solid pseudopapillary neoplasms. Oncotarget. 8:93404–93413PubMedPubMedCentralCrossRef Kim EK, Jang M, Park M, Kim H (2017) LEF1, TFE3 and AR are putative diagnostic markers of solid pseudopapillary neoplasms. Oncotarget. 8:93404–93413PubMedPubMedCentralCrossRef
44.
Zurück zum Zitat Geers C, Moulin P, Gigot JF, Weynand B, Deprez P, Rahier J, Sempoux C (2006) Solid and pseudopapillary tumor of the pancreas--review and new insights into pathogenesis. Am J Surg Pathol 30:1243–1249PubMedCrossRef Geers C, Moulin P, Gigot JF, Weynand B, Deprez P, Rahier J, Sempoux C (2006) Solid and pseudopapillary tumor of the pancreas--review and new insights into pathogenesis. Am J Surg Pathol 30:1243–1249PubMedCrossRef
45.
Zurück zum Zitat Neill KG, Saller J, Al Diffalha S, Centeno BA, Malafa MP, Coppola D (2018) EGFR L861Q mutation in a metastatic solid-pseudopapillary neoplasm of the pancreas. Cancer Genomics Proteomics 15:201–205PubMedPubMedCentral Neill KG, Saller J, Al Diffalha S, Centeno BA, Malafa MP, Coppola D (2018) EGFR L861Q mutation in a metastatic solid-pseudopapillary neoplasm of the pancreas. Cancer Genomics Proteomics 15:201–205PubMedPubMedCentral
46.
Zurück zum Zitat Liszka L, Mrowiec S, Pająk J, Kostrząb-Zdebel A, Lampe P, Kajor M (2014) Limited usefulness of histopathological features in identification of a clinically aggressive solid-pseudopapillary neoplasm of the pancreas. Pol J Pathol 65:182–193PubMedCrossRef Liszka L, Mrowiec S, Pająk J, Kostrząb-Zdebel A, Lampe P, Kajor M (2014) Limited usefulness of histopathological features in identification of a clinically aggressive solid-pseudopapillary neoplasm of the pancreas. Pol J Pathol 65:182–193PubMedCrossRef
47.
Zurück zum Zitat Kim JH, Lee JM (2016) Clinicopathologic review of 31 cases of solid Pseudopapillary pancreatic tumors: can we use the scoring system of microscopic features for suggesting clinically malignant potential? Am Surg 82:308–313PubMed Kim JH, Lee JM (2016) Clinicopathologic review of 31 cases of solid Pseudopapillary pancreatic tumors: can we use the scoring system of microscopic features for suggesting clinically malignant potential? Am Surg 82:308–313PubMed
48.
Zurück zum Zitat Kosmahl M, Seada LS, Jänig U, Harms D, Klöppel G (2000) Solid-pseudopapillary tumor of the pancreas: its origin revisited. Virchows Arch 436:473–480PubMedCrossRef Kosmahl M, Seada LS, Jänig U, Harms D, Klöppel G (2000) Solid-pseudopapillary tumor of the pancreas: its origin revisited. Virchows Arch 436:473–480PubMedCrossRef
49.
Zurück zum Zitat Park JY, Kim SG, Park J (2014) Solid pseudopapillary tumor of the pancreas in children: 15-year experience at a single institution with assays using an immunohistochemical panel. Ann Surg Treat Res 86:130–135PubMedPubMedCentralCrossRef Park JY, Kim SG, Park J (2014) Solid pseudopapillary tumor of the pancreas in children: 15-year experience at a single institution with assays using an immunohistochemical panel. Ann Surg Treat Res 86:130–135PubMedPubMedCentralCrossRef
50.
Zurück zum Zitat Calvani J, Lopez P, Sarnacki S, Molina TJ, Gibault L, Fabre M, Scharfmann R, Capito C, Galmiche L (2019) Solid pseudopapillary neoplasms of the pancreas do not express major pancreatic markers in pediatric patients. Hum Pathol 83:29–31PubMedCrossRef Calvani J, Lopez P, Sarnacki S, Molina TJ, Gibault L, Fabre M, Scharfmann R, Capito C, Galmiche L (2019) Solid pseudopapillary neoplasms of the pancreas do not express major pancreatic markers in pediatric patients. Hum Pathol 83:29–31PubMedCrossRef
51.
Zurück zum Zitat Notohara K, Hamazaki S, Tsukayama C, Nakamoto S, Kawabata K, Mizobuchi K, Sakamoto K, Okada S (2000) Solid-pseudopapillary tumor of the pancreas: immunohistochemical localization of neuroendocrine markers and CD10. Am J Surg Pathol 24:1361–1371PubMedCrossRef Notohara K, Hamazaki S, Tsukayama C, Nakamoto S, Kawabata K, Mizobuchi K, Sakamoto K, Okada S (2000) Solid-pseudopapillary tumor of the pancreas: immunohistochemical localization of neuroendocrine markers and CD10. Am J Surg Pathol 24:1361–1371PubMedCrossRef
52.
Zurück zum Zitat Li L, Li J, Hao C, Zhang C, Mu K, Wang Y, Zhang T (2011) Immunohistochemical evaluation of solid pseudopapillary tumors of the pancreas: the expression pattern of CD99 is highly unique. Cancer Lett 310:9–14PubMedCrossRef Li L, Li J, Hao C, Zhang C, Mu K, Wang Y, Zhang T (2011) Immunohistochemical evaluation of solid pseudopapillary tumors of the pancreas: the expression pattern of CD99 is highly unique. Cancer Lett 310:9–14PubMedCrossRef
53.
54.
Zurück zum Zitat Harrison G, Hemmerich A, Guy C, Perkinson K, Fleming D, McCall S, Cardona D, Zhang X (2017) Overexpression of SOX11 and TFE3 in solid-Pseudopapillary neoplasms of the pancreas. Am J Clin Pathol 149:67–75PubMedCrossRef Harrison G, Hemmerich A, Guy C, Perkinson K, Fleming D, McCall S, Cardona D, Zhang X (2017) Overexpression of SOX11 and TFE3 in solid-Pseudopapillary neoplasms of the pancreas. Am J Clin Pathol 149:67–75PubMedCrossRef
55.
Zurück zum Zitat Jiang Y, Xie J, Wang B, Mu Y, Liu P (2018) TFE3 is a diagnostic marker for solid pseudopapillary neoplasms of the pancreas. Hum Pathol 81:166–175PubMedCrossRef Jiang Y, Xie J, Wang B, Mu Y, Liu P (2018) TFE3 is a diagnostic marker for solid pseudopapillary neoplasms of the pancreas. Hum Pathol 81:166–175PubMedCrossRef
56.
Zurück zum Zitat Lawlor RT, Daprà V, Girolami I, Pea A, Pilati C, Nottegar A, Piccoli P, Parolini C, Sperandio N, Capelli P, Scarpa A, Luchini C (2019) CD200 expression is a feature of solid pseudopapillary neoplasms of the pancreas. Virchows Arch 474:105–109PubMedCrossRef Lawlor RT, Daprà V, Girolami I, Pea A, Pilati C, Nottegar A, Piccoli P, Parolini C, Sperandio N, Capelli P, Scarpa A, Luchini C (2019) CD200 expression is a feature of solid pseudopapillary neoplasms of the pancreas. Virchows Arch 474:105–109PubMedCrossRef
57.
Zurück zum Zitat Hav M, De Potter A, Ferdinande L, Van Bockstal M, Lem D, Eav S, Pattyn P, Praet M, Cuvelier C, Libbrecht L (2011) Glypican-3 is a marker for solid pseudopapillary neoplasm of the pancreas. Histopathology. 59:1278–1279PubMedCrossRef Hav M, De Potter A, Ferdinande L, Van Bockstal M, Lem D, Eav S, Pattyn P, Praet M, Cuvelier C, Libbrecht L (2011) Glypican-3 is a marker for solid pseudopapillary neoplasm of the pancreas. Histopathology. 59:1278–1279PubMedCrossRef
58.
Zurück zum Zitat Shen Y, Wang Z, Zhu J, Chen Y, Gu W, Liu Q (2014) α-Methylacyl-CoA racemase (P504S) is a useful marker for the differential diagnosis of solid pseudopapillary neoplasm of the pancreas. Ann Diagn Pathol 18:146–150PubMedCrossRef Shen Y, Wang Z, Zhu J, Chen Y, Gu W, Liu Q (2014) α-Methylacyl-CoA racemase (P504S) is a useful marker for the differential diagnosis of solid pseudopapillary neoplasm of the pancreas. Ann Diagn Pathol 18:146–150PubMedCrossRef
59.
Zurück zum Zitat Klimstra DS, Wenig BM, Heffess CS (2000) Solid-pseudopapillary tumor of the pancreas: a typically cystic carcinoma of low malignant potential. Semin Diagn Pathol 17:66–80PubMed Klimstra DS, Wenig BM, Heffess CS (2000) Solid-pseudopapillary tumor of the pancreas: a typically cystic carcinoma of low malignant potential. Semin Diagn Pathol 17:66–80PubMed
60.
Zurück zum Zitat Wrba F, Chott A, Ludvik B, Schratter M, Spona J, Reiner A, Scjernthaner G, Krisch K (1988) Solid and cystic tumour of the pancreas; a hormonal-dependent neoplasm? Histopathology 12:338–340PubMed Wrba F, Chott A, Ludvik B, Schratter M, Spona J, Reiner A, Scjernthaner G, Krisch K (1988) Solid and cystic tumour of the pancreas; a hormonal-dependent neoplasm? Histopathology 12:338–340PubMed
61.
Zurück zum Zitat Ueda N, Nagakawa T, Ohta T, Nakamura T, Ueno K, Miyazaki I, Kurachi M, Konishi I, Hirono T, Takayanagi N, Sodani H, Kanno S (1991) Clinicopathological studies on solid and cystic tumors of the pancreas. Gastroenterol Jpn 26:497–502PubMedCrossRef Ueda N, Nagakawa T, Ohta T, Nakamura T, Ueno K, Miyazaki I, Kurachi M, Konishi I, Hirono T, Takayanagi N, Sodani H, Kanno S (1991) Clinicopathological studies on solid and cystic tumors of the pancreas. Gastroenterol Jpn 26:497–502PubMedCrossRef
62.
Zurück zum Zitat Hamoudi AB, Misugi K, Grosfeld JL, Reiner CB (1970) Papillary epithelial neoplasms of pancreas in a child. Cancer 26:1126–1134PubMedCrossRef Hamoudi AB, Misugi K, Grosfeld JL, Reiner CB (1970) Papillary epithelial neoplasms of pancreas in a child. Cancer 26:1126–1134PubMedCrossRef
63.
Zurück zum Zitat Schlosnagle DC, Campbell WG (1981) The papillary and solid neoplasm of the pancreas. A report ot two cases with electron microscopy, one containing neurosecretory granules. Cancer 47:2603–2610PubMedCrossRef Schlosnagle DC, Campbell WG (1981) The papillary and solid neoplasm of the pancreas. A report ot two cases with electron microscopy, one containing neurosecretory granules. Cancer 47:2603–2610PubMedCrossRef
64.
Zurück zum Zitat Cao D, Maitra A, Saavedra JA, Klimstra DS, Adsay NV, Hruban RH (2005) Expression of novel markers of pancreatic ductal adenocarcinoma in pancreatic nonductal neoplasms: additional evidence of different genetic pathways. Mod Pathol 18:752–761PubMedCrossRef Cao D, Maitra A, Saavedra JA, Klimstra DS, Adsay NV, Hruban RH (2005) Expression of novel markers of pancreatic ductal adenocarcinoma in pancreatic nonductal neoplasms: additional evidence of different genetic pathways. Mod Pathol 18:752–761PubMedCrossRef
65.
Zurück zum Zitat Kallichandra N, Tsai S, Stabile BE, Buslon V, Delgado DL, French SW (2006) Histogenesis of solid pseudopapillary tumor of the pancreas: the case for the centroacinar cell of origin. Exp Mol Pathol 81:101–107 Kallichandra N, Tsai S, Stabile BE, Buslon V, Delgado DL, French SW (2006) Histogenesis of solid pseudopapillary tumor of the pancreas: the case for the centroacinar cell of origin. Exp Mol Pathol 81:101–107
66.
Zurück zum Zitat Matsunou H, Konishi F (1990) Papillary-cystic neoplasm of the pancreas. A clinicopathologic study concerning the tumor aging and malignancy of nine cases. Cancer 65:283–291PubMedCrossRef Matsunou H, Konishi F (1990) Papillary-cystic neoplasm of the pancreas. A clinicopathologic study concerning the tumor aging and malignancy of nine cases. Cancer 65:283–291PubMedCrossRef
67.
Zurück zum Zitat Sun CD, Lee WJ, Choi JS, Oh JT, Choi SH (2005) Solid-pseudopapillary tumours of the pancreas: 14 years experience. ANZ J Surg 75:684–689PubMedCrossRef Sun CD, Lee WJ, Choi JS, Oh JT, Choi SH (2005) Solid-pseudopapillary tumours of the pancreas: 14 years experience. ANZ J Surg 75:684–689PubMedCrossRef
68.
Zurück zum Zitat Escobar MA Jr, McClellan JM, Thomas W (2017) Solid pseudopapillary tumour (Frantz's tumour) of the pancreas in childhood: successful management of late liver metastases with sunitinib and chemoembolisation. BMJ Case Rep pii: bcr-2017-221906. https://doi.org/10.1136/bcr-2017-221906 Escobar MA Jr, McClellan JM, Thomas W (2017) Solid pseudopapillary tumour (Frantz's tumour) of the pancreas in childhood: successful management of late liver metastases with sunitinib and chemoembolisation. BMJ Case Rep pii: bcr-2017-221906. https://​doi.​org/​10.​1136/​bcr-2017-221906
69.
Zurück zum Zitat Łągiewska B, Pacholczyk M, Lisik W, Cichocki A, Nawrocki G, Trzebicki J, Chmura A (2013) Liver transplantation for nonresectable metastatic solid pseudopapillary pancreatic cancer. Ann Transplant 18:651–653PubMedCrossRef Łągiewska B, Pacholczyk M, Lisik W, Cichocki A, Nawrocki G, Trzebicki J, Chmura A (2013) Liver transplantation for nonresectable metastatic solid pseudopapillary pancreatic cancer. Ann Transplant 18:651–653PubMedCrossRef
70.
Zurück zum Zitat Wójciak M, Gozdowska J, Pacholczyk M, Lisik W, Kosieradzki M, Cichocki A, Tronina O, Durlik M (2018) Liver transplantation for a metastatic pancreatic solid-Pseudopapillary tumor (Frantz tumor): a case report. Ann Transplant 23:520–523PubMedPubMedCentralCrossRef Wójciak M, Gozdowska J, Pacholczyk M, Lisik W, Kosieradzki M, Cichocki A, Tronina O, Durlik M (2018) Liver transplantation for a metastatic pancreatic solid-Pseudopapillary tumor (Frantz tumor): a case report. Ann Transplant 23:520–523PubMedPubMedCentralCrossRef
71.
Zurück zum Zitat Foo WC, Harrison G, Zhang X (2017) Immunocytochemistry for SOX-11 and TFE3 as diagnostic markers for solid pseudopapillary neoplasms of the pancreas in FNA biopsies. Cancer Cytopathol 125:831–837PubMedCrossRef Foo WC, Harrison G, Zhang X (2017) Immunocytochemistry for SOX-11 and TFE3 as diagnostic markers for solid pseudopapillary neoplasms of the pancreas in FNA biopsies. Cancer Cytopathol 125:831–837PubMedCrossRef
72.
Zurück zum Zitat Comper F, Antonello D, Beghelli S, Gobbo S, Montagna L, Pederzoli P, Chilosi M, Scarpa A (2009) Expression pattern of claudins 5 and 7 distinguishes solid-pseudopapillary from pancreatoblastoma, acinar cell and endocrine tumors of the pancreas. Am J Surg Pathol 33:768–774PubMedCrossRef Comper F, Antonello D, Beghelli S, Gobbo S, Montagna L, Pederzoli P, Chilosi M, Scarpa A (2009) Expression pattern of claudins 5 and 7 distinguishes solid-pseudopapillary from pancreatoblastoma, acinar cell and endocrine tumors of the pancreas. Am J Surg Pathol 33:768–774PubMedCrossRef
73.
Zurück zum Zitat Bouassida M, Mighri MM, Bacha D, Chtourou MF, Touinsi H, Azzouz MM, Sassi S (2012) Solid pseudopapillary neoplasm of the pancreas in an old man: age does not matter. Pan Afr Med J 13:8PubMedPubMedCentral Bouassida M, Mighri MM, Bacha D, Chtourou MF, Touinsi H, Azzouz MM, Sassi S (2012) Solid pseudopapillary neoplasm of the pancreas in an old man: age does not matter. Pan Afr Med J 13:8PubMedPubMedCentral
Metadaten
Titel
Solid-pseudopapillary Neoplasms of the Pancreas is still an Enigma: a Clinicopathological Review
verfasst von
Attila Zalatnai
Viktória Kis-Orha
Publikationsdatum
17.06.2019
Verlag
Springer Netherlands
Erschienen in
Pathology & Oncology Research / Ausgabe 2/2020
Print ISSN: 1219-4956
Elektronische ISSN: 1532-2807
DOI
https://doi.org/10.1007/s12253-019-00671-8

Weitere Artikel der Ausgabe 2/2020

Pathology & Oncology Research 2/2020 Zur Ausgabe

Adjuvante Immuntherapie verlängert Leben bei RCC

25.04.2024 Nierenkarzinom Nachrichten

Nun gibt es auch Resultate zum Gesamtüberleben: Eine adjuvante Pembrolizumab-Therapie konnte in einer Phase-3-Studie das Leben von Menschen mit Nierenzellkarzinom deutlich verlängern. Die Sterberate war im Vergleich zu Placebo um 38% geringer.

Alectinib verbessert krankheitsfreies Überleben bei ALK-positivem NSCLC

25.04.2024 NSCLC Nachrichten

Das Risiko für Rezidiv oder Tod von Patienten und Patientinnen mit reseziertem ALK-positivem NSCLC ist unter einer adjuvanten Therapie mit dem Tyrosinkinase-Inhibitor Alectinib signifikant geringer als unter platinbasierter Chemotherapie.

Bei Senioren mit Prostatakarzinom auf Anämie achten!

24.04.2024 DGIM 2024 Nachrichten

Patienten, die zur Behandlung ihres Prostatakarzinoms eine Androgendeprivationstherapie erhalten, entwickeln nicht selten eine Anämie. Wer ältere Patienten internistisch mitbetreut, sollte auf diese Nebenwirkung achten.

ICI-Therapie in der Schwangerschaft wird gut toleriert

Müssen sich Schwangere einer Krebstherapie unterziehen, rufen Immuncheckpointinhibitoren offenbar nicht mehr unerwünschte Wirkungen hervor als andere Mittel gegen Krebs.

Update Onkologie

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