Skip to main content
Erschienen in: World Journal of Surgery 2/2018

Open Access 22.11.2017 | Original Scientific Report

Radioembolization Versus Bland Embolization for Hepatic Metastases from Small Intestinal Neuroendocrine Tumors: Short-Term Results of a Randomized Clinical Trial

verfasst von: Anna-Karin Elf, Mats Andersson, Olof Henrikson, Oscar Jalnefjord, Maria Ljungberg, Johanna Svensson, Bo Wängberg, Viktor Johanson

Erschienen in: World Journal of Surgery | Ausgabe 2/2018

Abstract

Background

Radioembolization (RE) with intra-arterial administration of 90Y microspheres is a promising technique for the treatment of liver metastases from small intestinal neuroendocrine tumors (SI-NET) not amenable to surgery or local ablation. However, studies comparing RE to other loco-regional therapies are lacking. The aim of this randomized study was to compare the therapeutic response and safety after RE and bland hepatic arterial embolization (HAE), and to investigate early therapy-induced changes with diffusion-weighted MRI (DWI-MRI).

Methods

Eleven patients were included in a prospective randomized controlled pilot study, six assigned to RE and five to HAE. Response according to RECIST 1.1 using MRI or CT at 3 and 6 months post-treatment was recorded as well as changes in DWI-MRI parameters after 1 month. Data on biochemical tumor response, toxicity, and side effects were also collected.

Results

Three months after treatment, all patients in the HAE group showed partial response according to RECIST while none in the RE group did (p = 0.0022). After 6 months, the response rates were 4/5 (80%) and 2/6 (33%) in the HAE and RE groups, respectively (NS). DWI-MRI metrics could not predict RECIST response, but lower pretreatment ADC(120–800) and larger ADC(0–800) increase at 1 month were related to larger decrease in tumor diameter when all tumors were counted.

Conclusion

HAE resulted in significantly higher RECIST response after 3 months, but no difference compared to RE remained after 6 months. These preliminary findings indicate that HAE remains a safe option for the treatment of liver metastases from SI-NET, and further studies are needed to establish the role of RE and the predictive value of MR-DWI.
Hinweise

Electronic supplementary material

The online version of this article (https://​doi.​org/​10.​1007/​s00268-017-4324-9) contains supplementary material, which is available to authorized users.
Anna-Karin Elf and Mats Andersson have contributed equally to the completion of this manuscript.

Introduction

The primary tumors of small intestinal neuroendocrine tumors (SI-NETs) can almost always be radically resected by surgery, often together with the regional lymph node metastases. Hepatic metastases are common, and surgical resection is suitable only in a minority of cases calling for other loco-regional treatment alternatives [1]. Local ablative therapies using radiofrequency or microwaves are an option if the metastases are few, small, and suitably localized. In other cases, loco-regional endovascular therapies, such as hepatic arterial embolization (HAE) or chemoembolization (TACE), are considered. The mainstay for treating widespread liver metastases at our unit has been HAE with polyvinyl alcohol (PVA) particles [2]. However, the treatment is often associated with side effects due to liver ischemia, and recurrence may occur. Recently, radioembolization (RE), involving transcatheter arterial delivery of 20–60 µm microspheres containing 90Y radioisotope into the tumor microvasculature, has emerged as a promising tool in the management of hepatic metastases from NET. RE is considered to have less acute and subacute toxicity than HAE, as it avoids liver ischemia. However, studies documenting the safety and efficacy of RE largely represent retrospective cohort series [39], while prospective studies comparing RE to other loco-regional therapies such as HAE are lacking.
Currently accepted guidelines for assessing treatment response in solid tumors include response evaluation criteria in solid tumors (RECIST) [10, 11]. However, these morphological criteria may not be conclusive until several months following therapy. Earlier evaluation of response to therapy would allow alternative therapy sooner, with optimized management of the individual patient. Diffusion-weighted imaging (DWI) is a functional MR imaging method that provides insight into the tumor microenvironment and can potentially be used as an early surrogate biomarker for tumor response [12, 13].
Here, we present the preliminary results of a randomized phase II study where the primary end point was to compare the treatment response, according to RECIST, of hepatic metastases at 3 months after RE or HAE. Secondary aims were to study the radiological response at 6 months, the biochemical response, and toxicity, and to evaluate the usefulness of early changes in DWI parameters in predicting later treatment response.

Materials and methods

Patients

Inclusion criteria were multiple SI-NET liver metastases, grade 1 or 2, not accessible to curative resection or ablation, and elevated serum chromogranin A (CgA) and/or 24 h urinary 5-HIAA excretion (dU-5HIAA). All patients were under somatostatin analogue (SSA) treatment, and primary surgery with removal of all extrahepatic tumors had been performed in all patients.
Exclusion criteria were remaining extrahepatic metastases, previous loco-regional or systemic anti-tumoral treatment (except SSA), impaired liver function or tumor volume exceeding 50% of total liver volume.
Before treatments, all patients but one were investigated with DWI-MRI. The remaining patient could not undergo MRI due to cardiac pacemaker and was evaluated with CT. DWI-MRI was repeated 1 month after treatment followed by response evaluation with MRI or CT according to RECIST 1.1 at 3 and 6 months.
The study was approved by the Ethical Review Board of Gothenburg University.

RE and HAE procedures

Radioembolization was performed with bilobar infusion in a standard manner in all patients [9]. Protective coil embolization was used when necessary to prevent non-target embolization. The administered activity of 90Y resin microspheres (SIR-spheres™) was calculated using the partition model [14].
HAE was performed by infusion of PVA particles (45–150 μm) into the right or left hepatic artery until stasis was achieved. The right liver lobe was treated first, embolizing the remaining left lobe at a second session, about 6 weeks later. No other anti-tumoral therapy was started during the study period.

Magnetic resonance imaging

The MRI measurements were performed on a Gyroscan Achieva dStream 3 T Release 5.1.7 (Philips Medical Systems, Eindhoven, the Netherlands). The examination included T2W and T1W scans, and a DWI scan with multiple b-values (0, 120, 350, 575, 800 s/mm2). At baseline and at the 3 and 6 months follow-up examinations, a T1-weighted dynamic contrast-enhanced sequence in the late arterial and portal venous phases was added to the protocol (for details see Electronic Supplementary Material).

Image analysis

Up to five liver metastases, with lesion diameters larger than 1 cm, were analyzed per patient. Multiple free-hand regions of interest (ROIs) were drawn to encompass the whole volume of the metastases on DWI-MRI images. The relative ADC values at 1 month, compared to pretreatment baseline values, were calculated. As a control, a circular ROI of minimum 120 mm2 was placed on normal-appearing splenic parenchyma (for details see Electronic Supplementary Material).

Assessment of therapeutic response

Response evaluation was performed by MRI or CT at 3 and 6 months after treatment, according to the RECIST 1.1 guidelines [11]. For every patient, two target lesions, which appeared best measurable and representative of the patient’s overall tumor load at the baseline MRI/CT, were chosen in accordance with RECIST criteria using the “Tumor Tracking” module at the IntelliSpace-Portal workstation.
Biochemical tumor markers CgA in serum and dU-5HIAA were measured at 3 and 6 months after treatment.
Therapeutic response at 3 and 6 months after treatment was also determined on a lesion-by-lesion basis by evaluating changes in size of a total of 39 tumors. The measurements of the lesion diameters (LD = longest diameter) were averaged across all MR sequences. The relative tumor size at different times after treatment was calculated by comparing it with pretreatment baseline values (for details see Electronic Supplementary Material).

Toxicity

Toxicity was assessed weekly during the first month after treatment by measuring hemoglobin, white blood count, and platelets. At the same time points, liver-specific toxicity was assessed by measuring ASAT, ALAT, ALP, and bilirubin. Blood sampling was repeated at 3 and 6 months after treatments.

Statistical analysis

Response rates according to RECIST1.1 were estimated with binomial proportions and compared by Fisher’s exact test using the MedCalc statistics package (MedCalc Software, Ostend, Belgium). Linear mixed models were used to test for difference between changes in ADC parameters and for linear correlation between ADC parameters and change in size following treatment using MATLAB 2016b (MathWorks Inc, Natick, MA, USA). Mann–Whitney U test or Wilcoxon signed-rank test were used to compare differences in continuous variables. A p value of <0.05 was considered statistically significant.

Results

Between January 2014 and September 2016, 11 patients with irresectable liver metastases from SI-NET, not suitable for local ablative treatments, were randomly assigned to RE (n = 6) or HAE (n = 5). Median age at enrollment was 67 years, and three patients were men (Table 1). All patients had > 5 metastases, and a total of 36 lesions, which could be consistently assessed throughout all MRI examinations, were analyzed on a lesion-by-lesion basis. A median activity of 1.4 (range 1.1–1.8) GBq of 90Y was given to patients receiving RE. The first radiological response evaluation, DWI-MRI, was performed after a median time of 32 days (range 22–55 days) after RE or the first HAE treatment (the right lobe). The 2nd and 3rd evaluations were performed after a median time of 90 and 195 days, respectively (range 84–118 and 166–251 days), after treatment. However, one of the patients, with a pacemaker, was assessed with CT only, and another patient treated with HAE failed to undergo DWI-MRI at the 1-month time point.
Table 1
Baseline data: Baseline clinical and tumor characteristics of the study population randomized to RE or HAE
 
All patients (n = 11)
Patients receiving RE (n = 6)
Patients receiving HAE (n = 5)
p value
Age, years, median (range)
67 (40–79)
66.5 (40–79)
67 (51–79)
0.65*
Male sex
3
2
1
1.00**
Number of lesions analyzed, median (range)
4 (1–5)
5 (2–5)
3 (1–5)
0.11*
Median LD, mm (range)
20.3 (13–55)
20.3 (13–50)
19.9 (13–55)
0.92*
Median sum of LD of metastases analyzed, mm (range)
77 (30–170)
89 (35–170)
74 (30–170)
0.27*
Median baseline ADC(120–800), 10−3mm2/s (range)
0.73 (0.5–1.3)
0.78 (0.5–1.3)
0.68 (0.5–1.0)
0.15*
Primary tumor, grade 1 (Ki-67 < 2%) (n)
7
5
2
0.24**
Primary tumor, grade 2 (Ki-67 3–20%) (n)
4
1
3
0.24**
Median dU-5HIAA (µmol/24 h) (range)
110 (21–270)
97 (54–130)
110 (21–270)
0.93*
Median CgA (µg/L) (range)
231 (81–1890)
162 (81–470)
384 (115–1890)
0.36*
* Mann–Whitney U test
** Fisher’s exact test

RECIST response

At 3 months after treatment, no responders were seen in the RE group while all patients in the HAE group showed partial response (PR) (p = 0.0022). After 6 months, two patients in the RE group showed PR while one patient in the HAE group had progressed compared to nadir at 3 months, resulting in no remaining significant difference between the groups (Fisher’s exact test, p = 0.24) (Fig. 1).

Biochemical response

Biochemical markers, CgA and 5HIAA, decreased in most patients after 3 and 6 months. The data were rather divergent, and there were no statistically significant differences between the treatment groups. The median reduction in CgA level after 3 months was 52% in the HAE group and 29% in the RE group. Corresponding values after 6 months were 47 and 44%, respectively (Fig. 2). The median reduction in 5HIAA excretion after 3 months was 43% in the HAE group and 25% in the RE group. Corresponding values after 6 months were 36 and 43%, respectively (Fig. 3).

Tumor size response and correlation with DWI assessment

The median pretreatment LD was 20 mm (range 13–55 mm) when evaluating all 36 tumors. In the 10 patients examined with MRI, pretreatment ADC(120–800) values were significantly negatively correlated with treatment response (decreasing LD) at 6 months (k = 45, p < 0.01). The mean increase in ADC(120–800) at 1 month was 40% after HAE and 11% after RE, although the difference between treatments was not statistically significant (p = 0.08). However, there was a statistically significant correlation between the relative increase in the ADC(0–800) values at 1 month and treatment response (decreasing LD) at 3 months (k = −0.2, p < 0.05) (Fig. 4). After 3 and 6 months, the relative (compared to baseline) median LD was significantly smaller in metastases treated with HAE than in patients treated with RE, 45 versus 89% (p < 0.001) and 39 versus 81% (p < 0.001), respectively (Fig. 5).
No difference in the median ADC(0–800) or ADC(120–800) values measured in the spleen before and after treatment was found (data not shown).

Toxicity

Overall toxicity from both treatments was low, and blood cell count at 6 months did not differ from baseline. Liver-specific evaluation showed that RE patients had slightly increased median levels of alkaline phosphatase compared to HAE patients at 6 months (2.8 µkat/L, range 1.6–15.5 vs. 1.45 µkat/L, range 1.0–2.1, p < 0.05), but no significant differences in liver enzymes or bilirubin values were found.
One patient in the HAE group developed post-treatment cholecystitis, which caused a prolonged hospital stay of 10 days. Median hospital stay after HAE was 4 days (range 4–10 days), which was significantly (p = 0.024) longer than after RE (median 2 days, range 2–6 days).

Discussion

For SI-NET patients with metastases that cannot be resected by surgery, other treatment options are needed. Systemic radiotherapy with radiolabeled somatostatin analogues (PRRT) has recently proved to be superior to treatment with somatostatin analogues alone [15]. But accumulated radiotoxicity is a concern, and repeated full-dose PRRT can rarely be performed. When the metastatic burden is limited to the liver, liver-directed therapy is preferable. At least HAE can be performed repeatedly, and neither HAE nor RE prevents future use of PRRT, if needed [16].
This is the first randomized prospective study that compares HAE and RE for the treatment of hepatic SI-NET metastases. Response evaluation according to RECIST utilizes the longest lesion diameter (LD) and specified threshold diameter changes. However, the appearance and size of liver metastases from NET may vary between MR sequences and phases of contrast administration [17]. It is currently somewhat unclear which sequence is the best for measuring these lesions [18]. Peri-tumoral edema on T2W imaging and peri-tumoral enhancement on dynamic contrast imaging may cause overestimation of the diameter measured. We hypothesized that averaging LD across all imaging sequences at one time point would compensate for these differences, optimizing the conditions for accurate response evaluation.
In this study, the RECIST response rate after radioembolization for metastatic NET was 33% at 6 months, which compares well with other studies that have shown a response rate between 22 and 63% [46, 1922]. We noted that compared to HAE, the response after RE was delayed and that the target lesions continued to decrease in size from 3 to 6 months post-treatment. At 6 months, no significant difference in the RECIST response remained. A similar trend in the response pattern of the biochemical tumor markers CgA and 5HIAA was noted. Such a delayed treatment response has also been observed by Fidelman et al. [23] who reported a median time to maximum response of 11 months in a study using 90Y glass microspheres for the treatment of metastatic NET.
ADC(120–800) primarily reflects diffusion, and both animal models and clinical studies have shown that lower ADC values in tumors indicate high viability [12] and a relative lack of necrosis, facilitating the intratumoral delivery of therapeutic agents [24, 25]. The finding that lower pretreatment ADC(120–800) was related to improved response (larger decrease of LD at 6 months) represents a well-known association between pretreatment ADC and response that has been reported for NET as well as for other tumor entities [2628].
While HAE resulted in a significant increase in ADC(120–800) at 1 month, RE did not. The reason for this is not clear, although the radiotoxic effect certainly is delayed compared to the more immediate ischemic effect of bland embolization. Another study of RE found no early ADC increase in responding colorectal liver metastases, which they associated with cellular edema [29]. Signs of an inflammatory response with peri-tumoral edema after RE have also been described on CT imaging [30]. However, it cannot be ruled out that the lack of a significant increase in diffusion at 1 month after RE was due to inadequate anti-tumor effect or suboptimal timing of the DWI measurement.
DWI analysis at 1 month after treatment could not predict the RECIST response in an individual patient in this small study. A larger increase in ADC(0–800) was, however, related to improved response (larger decrease of LD at 3 months) when analyzing all measured lesions. Since ADC0–800, as opposed to ADC120–800, is affected by both diffusion and pseudodiffusion caused by microscopic circulation, this result may indicate that changes in microperfusion after embolization are involved in the therapeutic process. Although there are contradictory results in the literature [31], this finding is in accordance with the suggestion of Kukuk et al. [26] that there is an increase in microperfusion secondary to a decrease in interstitial fluid pressure (IFP) in responding NET metastases.
This study has several limitations. Being an interim analysis of a pilot study, the study population is small, which limits the conclusions that can be drawn. The effect of inter-observer variation in the ROI positioning was not investigated, but it has been shown that this variation is less important in whole-volume measurements, as in our study [32].
Strength of the study is the prospective randomized design. To our knowledge, this is the first randomized trial comparing outcome after RE versus HAE in patients with metastatic NET.
In conclusion, in this randomized pilot study comparing RE and HAE for liver metastatic SI-NET, all metastases decreased in size after 3 months, but the decrease was significantly larger in patients treated with HAE. However, no significant difference in RECIST response between groups remained after 6 months, suggesting a delayed anatomical response after treatment with RE. Although lower pretreatment ADC(120–800) and larger increase in ADC(0–800) at 1 month were related to improved response (decrease in tumor diameter), at 3 and 6 months respectively, no DWI-MRI threshold for the prediction of response according to RECIST could be defined. These preliminary findings indicate that HAE remains a safe option for treatment of liver metastases from SI-NET, and further studies are needed to establish the role of RE and the predictive value of DWI-MRI in these tumors.

Acknowledgements

90Y resin microspheres (SIR-spheres™) were generously supplied by Sirtex. Professor Peter Bernhardt, Department of Radiation Physics at Sahlgrenska University Hospital is greatly appreciated for his contribution during the planning and performance of this study including responsibility of the dosimetry.

Compliance with ethical standards

Conflict of interest

The authors declare that they have no conflict of interest.
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

Die Chirurgie

Print-Titel

Das Abo mit mehr Tiefe

Mit der Zeitschrift Die Chirurgie erhalten Sie zusätzlich Online-Zugriff auf weitere 43 chirurgische Fachzeitschriften, CME-Fortbildungen, Webinare, Vorbereitungskursen zur Facharztprüfung und die digitale Enzyklopädie e.Medpedia.

Bis 30. April 2024 bestellen und im ersten Jahr nur 199 € zahlen!

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.

Anhänge

Electronic supplementary material

Below is the link to the electronic supplementary material.
Literatur
1.
Zurück zum Zitat Ahlman H, Westeberg G, Wängberg B et al (1996) Treatment of liver metastases of carcinoid tumors. World J Surg 20:196–202CrossRefPubMed Ahlman H, Westeberg G, Wängberg B et al (1996) Treatment of liver metastases of carcinoid tumors. World J Surg 20:196–202CrossRefPubMed
2.
Zurück zum Zitat Swärd C, Johanson V, Nieveen van Dijkum E et al (2009) Prolonged survival after hepatic artery embolization in patients with midgut carcinoid syndrome. Br J Surg 96:517–521CrossRefPubMed Swärd C, Johanson V, Nieveen van Dijkum E et al (2009) Prolonged survival after hepatic artery embolization in patients with midgut carcinoid syndrome. Br J Surg 96:517–521CrossRefPubMed
4.
Zurück zum Zitat Kennedy AS, Dezarn WA, McNeillie P et al (2008) Radioembolization for unresectable neuroendocrine hepatic metastases using resin 90Y-microspheres: early results in 148 patients. Am J Clin Oncol 31:271–279CrossRefPubMed Kennedy AS, Dezarn WA, McNeillie P et al (2008) Radioembolization for unresectable neuroendocrine hepatic metastases using resin 90Y-microspheres: early results in 148 patients. Am J Clin Oncol 31:271–279CrossRefPubMed
5.
Zurück zum Zitat King J, Quinn R, Glenn DM et al (2008) Radioembolization with selective internal radiation microspheres for neuroendocrine liver metastases. Cancer 113:921–929CrossRefPubMed King J, Quinn R, Glenn DM et al (2008) Radioembolization with selective internal radiation microspheres for neuroendocrine liver metastases. Cancer 113:921–929CrossRefPubMed
6.
Zurück zum Zitat Rhee TK, Lewandowski RJ, Liu DM et al (2008) 90Y Radioembolization for metastatic neuroendocrine liver tumors: preliminary results from a multi-institutional experience. Ann Surg 247:1029–1035CrossRefPubMed Rhee TK, Lewandowski RJ, Liu DM et al (2008) 90Y Radioembolization for metastatic neuroendocrine liver tumors: preliminary results from a multi-institutional experience. Ann Surg 247:1029–1035CrossRefPubMed
7.
Zurück zum Zitat Jia Z, Paz-Fumagalli R, Frey G et al (2017) Single-institution experience of radioembolization with yttrium-90 microspheres for unresectable metastatic neuroendocrine liver tumors. J Gastroenterol Hepatol 32:1617–1623CrossRefPubMed Jia Z, Paz-Fumagalli R, Frey G et al (2017) Single-institution experience of radioembolization with yttrium-90 microspheres for unresectable metastatic neuroendocrine liver tumors. J Gastroenterol Hepatol 32:1617–1623CrossRefPubMed
8.
Zurück zum Zitat Memon K, Lewandowski RJ, Mulcahy MF et al (2012) Radioembolization for neuroendocrine liver metastases: safety, imaging, and long-term outcomes. Int J Radiat Oncol Biol Phys 83:887–894CrossRefPubMed Memon K, Lewandowski RJ, Mulcahy MF et al (2012) Radioembolization for neuroendocrine liver metastases: safety, imaging, and long-term outcomes. Int J Radiat Oncol Biol Phys 83:887–894CrossRefPubMed
9.
Zurück zum Zitat Barbier CE, Garske-Román U, Sandström M et al (2016) Selective internal radiation therapy in patients with progressive neuroendocrine liver metastases. Eur J Nucl Med Mol Imaging 43:425–1431CrossRef Barbier CE, Garske-Román U, Sandström M et al (2016) Selective internal radiation therapy in patients with progressive neuroendocrine liver metastases. Eur J Nucl Med Mol Imaging 43:425–1431CrossRef
10.
Zurück zum Zitat Therasse P, Arbuck SG, Eisenhauer EA et al (2000) New guidelines to evaluate the response to treatment in solid tumors: European Organization for Research and Treatment of Cancer, National Cancer Institute of the United States, National Cancer Institute of Canada. J Natl Cancer Inst 92:205–216CrossRefPubMed Therasse P, Arbuck SG, Eisenhauer EA et al (2000) New guidelines to evaluate the response to treatment in solid tumors: European Organization for Research and Treatment of Cancer, National Cancer Institute of the United States, National Cancer Institute of Canada. J Natl Cancer Inst 92:205–216CrossRefPubMed
11.
Zurück zum Zitat Eisenhauer EA, Therasse P, Bogaerts J et al (2009) New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer 45:228–247CrossRefPubMed Eisenhauer EA, Therasse P, Bogaerts J et al (2009) New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer 45:228–247CrossRefPubMed
12.
Zurück zum Zitat Padhani AR, Liu G, Mu-Koh D et al (2009) Diffusion-weighted magnetic resonance imaging as a cancer biomarker: consensus and recommendations. Neoplasia 11:102–125CrossRefPubMedPubMedCentral Padhani AR, Liu G, Mu-Koh D et al (2009) Diffusion-weighted magnetic resonance imaging as a cancer biomarker: consensus and recommendations. Neoplasia 11:102–125CrossRefPubMedPubMedCentral
14.
Zurück zum Zitat Bernardini M, Smadja C, Faraggi M et al (2014) Liver selective internal radiation therapy with 90Y resin microspheres: comparison between pre-treatment activity calculation methods. Phys Med 30:752–764CrossRefPubMed Bernardini M, Smadja C, Faraggi M et al (2014) Liver selective internal radiation therapy with 90Y resin microspheres: comparison between pre-treatment activity calculation methods. Phys Med 30:752–764CrossRefPubMed
15.
Zurück zum Zitat Strosberg J, El-Haddad G, Wolin E et al (2017) Phase 3 Trial of 177Lu-Dotatate for midgut neuroendocrine tumors. N Engl J Med 376:125–135CrossRefPubMed Strosberg J, El-Haddad G, Wolin E et al (2017) Phase 3 Trial of 177Lu-Dotatate for midgut neuroendocrine tumors. N Engl J Med 376:125–135CrossRefPubMed
16.
Zurück zum Zitat Hamiditabar M, Ali M, Bolek L (2017) Safety and effectiveness of 177Lu-DOTATATE peptide receptor radionuclide therapy after regional hepatic embolization in patients with somatostatin-expressing neuroendocrine tumors. Clin Nucl Med 42:822–828CrossRefPubMed Hamiditabar M, Ali M, Bolek L (2017) Safety and effectiveness of 177Lu-DOTATATE peptide receptor radionuclide therapy after regional hepatic embolization in patients with somatostatin-expressing neuroendocrine tumors. Clin Nucl Med 42:822–828CrossRefPubMed
17.
Zurück zum Zitat Luersen GF, Wei W, Tamm EP et al (2016) Evaluation of magnetic resonance (MR) biomarkers for assessment of response with response evaluation criteria in solid tumors: comparison of the measurements of neuroendocrine tumor liver metastases (NETLM) with various MR sequences and at multiple phases of contrast administration. J Comput Assist Tomogr 40:717–722CrossRefPubMedPubMedCentral Luersen GF, Wei W, Tamm EP et al (2016) Evaluation of magnetic resonance (MR) biomarkers for assessment of response with response evaluation criteria in solid tumors: comparison of the measurements of neuroendocrine tumor liver metastases (NETLM) with various MR sequences and at multiple phases of contrast administration. J Comput Assist Tomogr 40:717–722CrossRefPubMedPubMedCentral
18.
Zurück zum Zitat Bhosale P, Kwek JW, Iyer R et al (2011) Follow-up of known carcinoid liver metastases: is respiratory-gated T2 fast spin-echo enough? Neuroendocrinology 93:241–248CrossRefPubMed Bhosale P, Kwek JW, Iyer R et al (2011) Follow-up of known carcinoid liver metastases: is respiratory-gated T2 fast spin-echo enough? Neuroendocrinology 93:241–248CrossRefPubMed
19.
Zurück zum Zitat Paprottka PM, Hoffmann RT, Haug A et al (2012) Radioembolization of symptomatic, unresectable neuroendocrine hepatic metastases using yttrium-90 microspheres. Cardiovasc Interv Radiol 35:334–342CrossRef Paprottka PM, Hoffmann RT, Haug A et al (2012) Radioembolization of symptomatic, unresectable neuroendocrine hepatic metastases using yttrium-90 microspheres. Cardiovasc Interv Radiol 35:334–342CrossRef
20.
Zurück zum Zitat Cao CQ, Yan TD, Bester L et al (2010) Radioembolization with yttrium microspheres for neuroendocrine tumour liver metastases. Br J Surg 297:537–543CrossRef Cao CQ, Yan TD, Bester L et al (2010) Radioembolization with yttrium microspheres for neuroendocrine tumour liver metastases. Br J Surg 297:537–543CrossRef
21.
Zurück zum Zitat Peker A, Cicek O, Soydal C et al (2015) Radioembolization with yttrium-90 resin microspheres for neuroendocrine tumor liver metastases. Diagn Interv Radiol 21:54–59CrossRefPubMed Peker A, Cicek O, Soydal C et al (2015) Radioembolization with yttrium-90 resin microspheres for neuroendocrine tumor liver metastases. Diagn Interv Radiol 21:54–59CrossRefPubMed
22.
Zurück zum Zitat Saxena A, Chua TC, Bester L et al (2010) Factors predicting response and survival after yttrium-90 radioembolization of unresectable neuroendocrine tumor liver metastases: a critical appraisal of 48 cases. Ann Surg 251:910–916CrossRefPubMed Saxena A, Chua TC, Bester L et al (2010) Factors predicting response and survival after yttrium-90 radioembolization of unresectable neuroendocrine tumor liver metastases: a critical appraisal of 48 cases. Ann Surg 251:910–916CrossRefPubMed
23.
Zurück zum Zitat Fidelman N, Kerlan RK Jr, Hawkins RA et al (2016) Radioembolization with 90Y glass microspheres for the treatment of unresectable metastatic liver disease from chemotherapy–refractory gastrointestinal cancers: final report of a prospective pilot study. J Gastrointest Oncol 7:860–874CrossRefPubMedPubMedCentral Fidelman N, Kerlan RK Jr, Hawkins RA et al (2016) Radioembolization with 90Y glass microspheres for the treatment of unresectable metastatic liver disease from chemotherapy–refractory gastrointestinal cancers: final report of a prospective pilot study. J Gastrointest Oncol 7:860–874CrossRefPubMedPubMedCentral
24.
Zurück zum Zitat Deng J, Rhee TK, Sato KT et al (2006) In vivo diffusion-weighted imaging of liver tumor necrosis in the VX2 rabbit model at 1.5 Tesla. Invest Radiol 41:410–414CrossRefPubMed Deng J, Rhee TK, Sato KT et al (2006) In vivo diffusion-weighted imaging of liver tumor necrosis in the VX2 rabbit model at 1.5 Tesla. Invest Radiol 41:410–414CrossRefPubMed
25.
Zurück zum Zitat Herneth AM, Guccione S, Bednarski M (2003) Apparent diffusion coefficient: a quantitative parameter for in vivo tumor characterization. Eur J Radiol 45:208–213CrossRefPubMed Herneth AM, Guccione S, Bednarski M (2003) Apparent diffusion coefficient: a quantitative parameter for in vivo tumor characterization. Eur J Radiol 45:208–213CrossRefPubMed
26.
Zurück zum Zitat Kukuk GM, Murtz P, Traber F et al (2013) Diffusion-weighted imaging with acquisition of three b-values for response evaluation of neuroendocrine liver metastases undergoing selective internal radiotherapy. Eur Radiol 24:267–276CrossRefPubMed Kukuk GM, Murtz P, Traber F et al (2013) Diffusion-weighted imaging with acquisition of three b-values for response evaluation of neuroendocrine liver metastases undergoing selective internal radiotherapy. Eur Radiol 24:267–276CrossRefPubMed
27.
Zurück zum Zitat Cui Y, Zhang XP, Sun YS et al (2008) Apparent diffusion coefficient: potential imaging biomarker for prediction and early detection of response to chemotherapy in hepatic metastases. Radiology 248:894–900CrossRefPubMed Cui Y, Zhang XP, Sun YS et al (2008) Apparent diffusion coefficient: potential imaging biomarker for prediction and early detection of response to chemotherapy in hepatic metastases. Radiology 248:894–900CrossRefPubMed
28.
Zurück zum Zitat Koh DM, Scurr E, Collins D et al (2007) Predicting response of colorectal hepatic metastasis: value of pretreatment apparent diffusion coefficients. AJR Am J Roentgenol 188:1001–1008CrossRefPubMed Koh DM, Scurr E, Collins D et al (2007) Predicting response of colorectal hepatic metastasis: value of pretreatment apparent diffusion coefficients. AJR Am J Roentgenol 188:1001–1008CrossRefPubMed
29.
Zurück zum Zitat Dudeck O, Zeile M, Wybranski C et al (2010) Early prediction of anticancer effects with diffusion-weighted MR imaging in patients with colorectal liver metastases following selective internal radiotherapy. Eur Radiol 20:2699–2706CrossRefPubMed Dudeck O, Zeile M, Wybranski C et al (2010) Early prediction of anticancer effects with diffusion-weighted MR imaging in patients with colorectal liver metastases following selective internal radiotherapy. Eur Radiol 20:2699–2706CrossRefPubMed
30.
Zurück zum Zitat Atassi B, Bangash AK, Bahrani A et al (2008) Multimodality imaging following 90Y radioembolization: a comprehensive review and pictorial essay. Radiographics 28:81–99CrossRefPubMed Atassi B, Bangash AK, Bahrani A et al (2008) Multimodality imaging following 90Y radioembolization: a comprehensive review and pictorial essay. Radiographics 28:81–99CrossRefPubMed
31.
Zurück zum Zitat Pieper CC, Sprinkart AM, Meyer C et al (2016) Evaluation of a simplified intravoxel incoherent motion (IVIM) analysis of diffusion-weighted imaging for prediction of tumor size changes and imaging response in breast cancer liver metastases undergoing radioembolization: a retrospective single center analysis. Medicine 95(e3275):1–9 Pieper CC, Sprinkart AM, Meyer C et al (2016) Evaluation of a simplified intravoxel incoherent motion (IVIM) analysis of diffusion-weighted imaging for prediction of tumor size changes and imaging response in breast cancer liver metastases undergoing radioembolization: a retrospective single center analysis. Medicine 95(e3275):1–9
32.
Zurück zum Zitat Lambregts DMJ, Beets GL, Maas M et al (2011) Tumour ADC measurements in rectal cancer: effect of ROI methods on ADC values and interobserver variability. Eur Radiol 21:2567–2574CrossRefPubMedPubMedCentral Lambregts DMJ, Beets GL, Maas M et al (2011) Tumour ADC measurements in rectal cancer: effect of ROI methods on ADC values and interobserver variability. Eur Radiol 21:2567–2574CrossRefPubMedPubMedCentral
Metadaten
Titel
Radioembolization Versus Bland Embolization for Hepatic Metastases from Small Intestinal Neuroendocrine Tumors: Short-Term Results of a Randomized Clinical Trial
verfasst von
Anna-Karin Elf
Mats Andersson
Olof Henrikson
Oscar Jalnefjord
Maria Ljungberg
Johanna Svensson
Bo Wängberg
Viktor Johanson
Publikationsdatum
22.11.2017
Verlag
Springer International Publishing
Erschienen in
World Journal of Surgery / Ausgabe 2/2018
Print ISSN: 0364-2313
Elektronische ISSN: 1432-2323
DOI
https://doi.org/10.1007/s00268-017-4324-9

Weitere Artikel der Ausgabe 2/2018

World Journal of Surgery 2/2018 Zur Ausgabe

Update Chirurgie

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

S3-Leitlinie „Diagnostik und Therapie des Karpaltunnelsyndroms“

Karpaltunnelsyndrom BDC Leitlinien Webinare
CME: 2 Punkte

Das Karpaltunnelsyndrom ist die häufigste Kompressionsneuropathie peripherer Nerven. Obwohl die Anamnese mit dem nächtlichen Einschlafen der Hand (Brachialgia parästhetica nocturna) sehr typisch ist, ist eine klinisch-neurologische Untersuchung und Elektroneurografie in manchen Fällen auch eine Neurosonografie erforderlich. Im Anfangsstadium sind konservative Maßnahmen (Handgelenksschiene, Ergotherapie) empfehlenswert. Bei nicht Ansprechen der konservativen Therapie oder Auftreten von neurologischen Ausfällen ist eine Dekompression des N. medianus am Karpaltunnel indiziert.

Prof. Dr. med. Gregor Antoniadis
Berufsverband der Deutschen Chirurgie e.V.

S2e-Leitlinie „Distale Radiusfraktur“

Radiusfraktur BDC Leitlinien Webinare
CME: 2 Punkte

Das Webinar beschäftigt sich mit Fragen und Antworten zu Diagnostik und Klassifikation sowie Möglichkeiten des Ausschlusses von Zusatzverletzungen. Die Referenten erläutern, welche Frakturen konservativ behandelt werden können und wie. Das Webinar beantwortet die Frage nach aktuellen operativen Therapiekonzepten: Welcher Zugang, welches Osteosynthesematerial? Auf was muss bei der Nachbehandlung der distalen Radiusfraktur geachtet werden?

PD Dr. med. Oliver Pieske
Dr. med. Benjamin Meyknecht
Berufsverband der Deutschen Chirurgie e.V.

S1-Leitlinie „Empfehlungen zur Therapie der akuten Appendizitis bei Erwachsenen“

Appendizitis BDC Leitlinien Webinare
CME: 2 Punkte

Inhalte des Webinars zur S1-Leitlinie „Empfehlungen zur Therapie der akuten Appendizitis bei Erwachsenen“ sind die Darstellung des Projektes und des Erstellungswegs zur S1-Leitlinie, die Erläuterung der klinischen Relevanz der Klassifikation EAES 2015, die wissenschaftliche Begründung der wichtigsten Empfehlungen und die Darstellung stadiengerechter Therapieoptionen.

Dr. med. Mihailo Andric
Berufsverband der Deutschen Chirurgie e.V.