1
|
Liu F, Miao W, Nan J, Shi Z, Zhang A, Bo Y, Xu J. Clinical diagnosis and treatment of 37 cases of gallbladder neuroendocrine carcinoma. World J Surg Oncol 2024; 22:157. [PMID: 38877554 PMCID: PMC11177431 DOI: 10.1186/s12957-024-03436-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 06/01/2024] [Indexed: 06/16/2024] Open
Abstract
OBJECTIVE This study aims to investigate the clinical and pathological characteristics, treatment approaches, and prognosis of gallbladder neuroendocrine carcinoma (GB-NEC). METHODS Retrospective analysis was conducted on the clinical data of 37 patients with GB-NEC admitted to Shanxi Cancer Hospital from January 2010 to June 2023. The study included an examination of their general information, treatment regimens, and overall prognosis. RESULTS Twelve cases, either due to distant metastasis or other reasons, did not undergo surgical treatment and received palliative chemotherapy (Group 1). Two cases underwent simple cholecystectomy (Group 2); four patients underwent palliative tumor resection surgery (Group 3), and nineteen patients underwent radical resection surgery (Group 4). Among the 37 GB-NEC patients, the average pre-surgery CA19-9 level was 113.29 ± 138.45 U/mL, and the median overall survival time was 19 months (range 7.89-30.11 months). Of these, 28 cases (75.7%) received systemic treatment, 25 cases (67.6%) underwent surgical intervention, and 16 cases (64.0%) received postoperative adjuvant treatment, including combined radiochemotherapy or chemotherapy alone. The median overall survival time was 4 months (0.61-7.40 months) for Group 1 (n = 12), 8 months for Group 2 (n = 2), 21 months (14.67-43.33 months) for Group 3 (n = 4), and 19 months (range 7.89-30.11 months) for Group 4 (n = 19). A significant difference in median overall survival time was observed between Group 1 and Group 4 (P = 0.004). CONCLUSION Surgery remains the primary treatment for GB-NEC, with radical resection potentially offering greater benefits to patient survival compared to other therapeutic options. Postoperative adjuvant therapy has the potential to extend patient survival, although the overall prognosis remains challenging.
Collapse
Affiliation(s)
- Feng Liu
- First Clinical Medical School, Shanxi Medical University, Taiyuan, 030001, China
- Department of Head and Neck Surgery, Shanxi Provincial Cancer Hospital, Shanxi Hospital Cancer Hospital of Chinese Academy of Medical Sciences, Taiyuan, 030001, China
| | - Wentao Miao
- First Clinical Medical School, Shanxi Medical University, Taiyuan, 030001, China
| | - Jiang Nan
- Department of Plastic Surgery, Taiyuan Maternity and Child Health Care Hospital, Taiyuan Children's Hospital, Taiyuan, 030001, China
| | - Zhiyong Shi
- Department of Hepatobiliary and Pancreatic Surgery, Liver Transplantation Center, The First Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Anhong Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Liver Transplantation Center, The First Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Yunfeng Bo
- Department of Pathology, Shanxi Hospital Affiliated to Cancer Hospital, Shanxi Province Cancer Hospital, Chinese Academy of Medical Sciences, Taiyuan, 030001, China
| | - Jun Xu
- Department of Hepatobiliary and Pancreatic Surgery, Liver Transplantation Center, The First Hospital of Shanxi Medical University, Taiyuan, 030001, China.
| |
Collapse
|
2
|
Wally SF, Albalawi AA, Al Madshush AM, Aljohani M, Alshehri AJ, Alamrani FM, Alyahya M, Aljohani FS, Modrba AY, Albalawi RH, Abo Draa O. Updates on the Diagnostic Use of Ultrasonography Augmented With Perfluorobutane Contrast in Hepatocellular Carcinoma: A Meta-Analysis. Cureus 2024; 16:e60891. [PMID: 38910635 PMCID: PMC11193104 DOI: 10.7759/cureus.60891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/22/2024] [Indexed: 06/25/2024] Open
Abstract
To investigate the diagnostic accuracy of contrast-enhanced ultrasound (CEUS) in the diagnosis of primary hepatocellular carcinoma (HCC), a thorough search was conducted for pertinent literature using PubMed, SCOPUS, Web of Science, Science Direct, and Wiley Library. This was a meta-analysis of diagnostic test accuracy. MetaDiSc 1.4 was used for all analyses and assessed statistical heterogeneity with the I2 index and the chi-square test. The random-effects model was applied where there was considerable heterogeneity. Using the eight elements of the Newcastle-Ottawa Scale (NOS) for cohort and case-control studies, we assessed the quality of the included studies. Our results included nine studies with a total of 2598 patients, and 1607 (61.8%) were males. The pooled overall sensitivity of perfluorobutane with CEUS was 85.6% (95% CI 0.832, -0.878, and P=0.000) and specificity was 91.5% (95% CI 0.899, -0.930, and P=0.000) with significant inter heterogeneity between studies (I2=94.3% and 95.7%), respectively. The pooled positive likelihood ratio was 12.42 (4.59 to 33.61, P=0.000). Our analysis revealed a symmetric summary receiver operating characteristic (SROC) curve and seven of the included studies are near the top left corner of the graph, indicating that this test has a high diagnostic value. The results showed that CEUS augmented with perfluorobutane contrast had good diagnostic accuracy (sensitivity and specificity) for primary HCC. Further real-world data studies are needed to confirm the good diagnosis accuracy of perfluorobutane CEUS in primary HCC.
Collapse
|
3
|
Santero M, de Mas J, Rifà B, Clavero I, Rexach I, Bonfill Cosp X. Assessing the methodological strengths and limitations of the Spanish Society of Medical Oncology (SEOM) guidelines: a critical appraisal using AGREE II and AGREE-REX tool. Clin Transl Oncol 2024; 26:85-97. [PMID: 37368198 PMCID: PMC10761528 DOI: 10.1007/s12094-023-03219-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 05/16/2023] [Indexed: 06/28/2023]
Abstract
BACKGROUND The Spanish Society of Medical Oncology (SEOM) has provided open-access guidelines for cancer since 2014. However, no independent assessment of their quality has been conducted to date. This study aimed to critically evaluate the quality of SEOM guidelines on cancer treatment. METHODS Appraisal of Guidelines for Research and Evaluation II (AGREE II) and AGREE-REX tool was used to evaluate the qualities of the guidelines. RESULTS We assessed 33 guidelines, with 84.8% rated as "high quality". The highest median standardized scores (96.3) were observed in the domain "clarity of presentation", whereas "applicability" was distinctively low (31.4), with only one guideline scoring above 60%. SEOM guidelines did not include the views and preferences of the target population, nor did specify updating methods. CONCLUSIONS Although developed with acceptable methodological rigor, SEOM guidelines could be improved in the future, particularly in terms of clinical applicability and patient perspectives.
Collapse
Affiliation(s)
| | - Júlia de Mas
- Universitat Autònoma Barcelona (UAB), Barcelona, Spain
| | - Berta Rifà
- Universitat Autònoma Barcelona (UAB), Barcelona, Spain
| | - Inés Clavero
- Universitat Autònoma Barcelona (UAB), Barcelona, Spain
| | - Irene Rexach
- Universitat Autònoma Barcelona (UAB), Barcelona, Spain
| | - Xavier Bonfill Cosp
- Universitat Autònoma Barcelona (UAB), Barcelona, Spain
- Iberoamerican Cochrane Centre, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain
- CIBER Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
| |
Collapse
|
4
|
Nibhondhratana P, Watcharadetwittaya S, Sa-ngiamwibool P. CD44v6 Expression in Gastroenteropancreatic Neuroendocrine Neoplasms: Clinicopathological Correlation and Prognosis. Pathol Res Pract 2022; 240:154213. [DOI: 10.1016/j.prp.2022.154213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 10/30/2022] [Accepted: 11/08/2022] [Indexed: 11/13/2022]
|
5
|
Maxwell JE, Naraev B, Halperin DM, Choti MA, Halfdanarson TR. Shifting Paradigms in the Pathophysiology and Treatment of Carcinoid Crisis. Ann Surg Oncol 2022; 29:3072-3084. [PMID: 35165817 DOI: 10.1245/s10434-022-11371-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 01/16/2022] [Indexed: 11/18/2022]
Abstract
Carcinoid crisis is a potentially fatal condition characterized by various symptoms, including hemodynamic instability, flushing, and diarrhea. The incidence of carcinoid crisis is unknown, in part due to inconsistency in definitions across studies. Triggers of carcinoid crisis include general anesthesia and surgical procedures, but drug-induced and spontaneous cases have also been reported. Patients with neuroendocrine tumors (NETs) and carcinoid syndrome are at risk for carcinoid crisis. The pathophysiology of carcinoid crisis has been attributed to secretion of bioactive substances, such as serotonin, histamine, bradykinin, and kallikrein by NETs. The somatostatin analog octreotide has been considered the standard of care for carcinoid crisis due to its inhibitory effect on hormone release and relatively fast resolution of carcinoid crisis symptoms in several case studies. However, octreotide's efficacy in the treatment of carcinoid crisis has been questioned. This is due to a lack of a common definition for carcinoid crisis, the heterogeneity in clinical presentation, the paucity of prospective studies assessing octreotide efficacy in carcinoid crisis, and the lack of understanding of the pathophysiology of carcinoid crisis. These issues challenge the classical physiologic model of carcinoid crisis and its common etiology with carcinoid syndrome and raise questions regarding the utility of somatostatin analogs in its treatment. As surgical procedures and invasive liver-directed therapies remain important treatment modalities in patients with NETs, the pathophysiology of carcinoid crisis, potential benefits of octreotide, and efficacy of alternative treatment modalities must be studied prospectively to develop an effective evidence-based treatment strategy for carcinoid crisis.
Collapse
Affiliation(s)
- Jessica E Maxwell
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Boris Naraev
- Banner MD Anderson Cancer Center, Phoenix, AZ, USA
| | - Daniel M Halperin
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Thorvardur R Halfdanarson
- Division of Medical Oncology, Mayo Clinic, Rochester, MN, USA. .,Mayo Clinic Cancer Center, Rochester, MN, USA.
| |
Collapse
|
6
|
Wang P, Chen J, Jiang Y, Jia C, Pang J, Wang S, Chang X. Neuroendocrine Neoplasms of the Gallbladder: A Clinicopathological Analysis of 13 Patients and a Review of the Literature. Gastroenterol Res Pract 2021; 2021:5592525. [PMID: 34122537 PMCID: PMC8166508 DOI: 10.1155/2021/5592525] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 05/10/2021] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVES Mixed neuroendocrine-non-neuroendocrine neoplasms (MiNENs) are rare gallbladder neuroendocrine neoplasms (GB-NENs). This study is aimed at investigating the clinicopathological features of GB-NENs and identifying prognostic factors related to overall survival (OS) of GB-MiNENs. METHODS The clinical data and pathological features of 13 patients with GB-NENs in our hospital were retrospectively reviewed. Additionally, 41 GB-MiNENs cases reported in English literature were reviewed and survival analysis was performed. RESULTS The mean age of thirteen patients (6 males and 7 females) with GB-NENs was 57.2 years (range: 35-75 years). Two patients were diagnosed with NET grade 1 (G1), two patients with NEC (large cell/small cell = 1/1), and nine patients with MiNENs. Of these 9 patients with MiNENs, 8 had composite tumors and 1 had amphicrine carcinoma. Microscopically, the adenocarcinoma component was located in the surface mucosa, and the neuroendocrine component was in the area of deep invasion, liver infiltration, and lymph node metastasis. Total analysis of 41 GB-MiNENs showed that patients were mainly elderly women (female/male ratio, 2.4 : 1.0; median age, 60 years). Kaplan-Meier's analysis demonstrated that liver metastasis and TNM stage III-IV were associated with decreased OS (P < 0.05), whereas age, sex, tumor size, grade of the neuroendocrine component, lymph node metastasis, and adjuvant chemotherapy were not significantly prognostic indicators of OS. Multivariate analysis identified liver metastasis (hazard ratio = 4.262, 95%confidence interval = 1.066-17.044, P = 0.040) as an independent unfavorable prognostic factor. CONCLUSIONS GB-MiNENs were the most common type of GB-NENs in our case series, and neuroendocrine components exhibited more aggressive lymph node metastasis and local invasion than adenocarcinoma. Liver metastasis was a poor prognostic indicator in GB-MiNENs patients.
Collapse
Affiliation(s)
- Pengyan Wang
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tsinghua University, Beijing 100730, China
| | - Jingci Chen
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tsinghua University, Beijing 100730, China
| | - Ying Jiang
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tsinghua University, Beijing 100730, China
| | - Congwei Jia
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tsinghua University, Beijing 100730, China
| | - Junyi Pang
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tsinghua University, Beijing 100730, China
| | - Shan Wang
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tsinghua University, Beijing 100730, China
| | - Xiaoyan Chang
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tsinghua University, Beijing 100730, China
| |
Collapse
|
7
|
Survival in Patients with Neuroendocrine Tumours of the Small Intestine: Nomogram Validation and Predictors of Survival. J Clin Med 2020; 9:jcm9082502. [PMID: 32756529 PMCID: PMC7464451 DOI: 10.3390/jcm9082502] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 07/24/2020] [Accepted: 07/29/2020] [Indexed: 02/07/2023] Open
Abstract
Neuroendocrine tumours of the small intestine (SI-NETs) are rare and heterogeneous. There is an unmet need for prognostication of disease course and to aid treatment strategies. A previously developed nomogram based on clinical and tumour characteristics aims to predict disease-specific survival (DSS) in patients with a SI-NET. We aimed to validate the nomogram and identify predictors of survival. Four hundred patients with a grade 1 or 2 SI-NET were included, between January 2000 and June 2016. Predicted 5- and 10-year survival was compared to actual DSS. Multivariable analysis identified predictors for actual DSS. We found that in low-, medium- and high-risk groups 5-year nomogram DSS vs. actual DSS was 0.86 vs. 0.82 (p < 0.001), 0.52 vs. 0.71 (p < 0.001) and 0.26 vs. 0.53 (p < 0.001), respectively. Ten-year nomogram DSS vs. actual DSS was 0.68 vs. 0.69 (p < 0.001), 0.40 vs. 0.50 (p < 0.001) and 0.20 vs. 0.35 (p < 0.001), respectively. Age, WHO-performance score of 2, Ki-67 index ≥10, unknown primary tumour, CgA > 6x ULN and elevated liver tests were identified as independent predictors for a worse DSS. This shows that the nomogram was able to differentiate, but underestimated DSS for patients with a SI-NET. Improvement of prognostication incorporating new emerging biomarkers is necessary to adequately estimate survival.
Collapse
|
8
|
Yan S, Liu T, Li Y, Zhu Y, Jiang J, Jiang L, Zhao H. Value of computed tomography evaluation in pathologic classification and prognosis prediction of gastric neuroendocrine tumors. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:545. [PMID: 31807527 DOI: 10.21037/atm.2019.09.114] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Background The study aims to investigate the correlation of CT characteristics with pathological classifications and the prognostic value of CT features in patients with gastric neuroendocrine neoplasms (g-NENs). Methods Ninety-one cases of pathologically diagnosed g-NENs, including 15 cases of well-differentiated neuroendocrine tumors (NETs) (G1 and G2) and 76 cases of poor-differentiated neuroendocrine carcinomas (NECs) (G3 and MANEC) were retrospectively studied. All cases were included in correlation analysis of CT characteristics with pathologic grades. Among them, 76 patients who had fulfilled follow-up data were included for overall survival (OS) and disease-free survival (DFS) analysis. Results CT characteristics that favor poor differentiation include tumor location (fundus and cardia), larger tumor size (>3.0 cm), infiltrative growth, unclear tumor margin, serosa involvement, ulceration and lymph node metastasis (P<0.05). Most variables had sensitivities >80% and specificities >60% to distinguish NECs from NETs. Through log-rank analysis, it was revealed that serosa involvement, cystic degeneration, necrosis, heterogeneous enhancement and lymph node metastasis led to worse DFS and OS for patients with g-NENs (P<0.05). COX regression analysis showed that serosa involvement and lymph node metastasis were independent risk factor for DFS and OS, respectively, despite of grading, staging and therapeutic choices (P<0.05). Moreover, high Ki-67 index (>55%) in G3 g-NENs is in correlation with serosa involvement and lymph node metastasis; accordingly, patients with higher Ki-67 index had worse 1-year DFS (61.7% vs. 92.3%; P<0.05). Conclusions CT characteristics can be useful discriminators and prognostic factors for g-NENs and may help identify G3 g-NEC from G3 g-NEN by revealing its poor differentiation and high invasive potential.
Collapse
Affiliation(s)
- Shida Yan
- Department of Hepatobiliary Surgery, National Cancer Center, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Tongtong Liu
- Department of Radiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Ying Li
- Department of Radiology, National Cancer Center, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yongjian Zhu
- Department of Radiology, National Cancer Center, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Jun Jiang
- Department of Radiology, National Cancer Center, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Liming Jiang
- Department of Radiology, National Cancer Center, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Hong Zhao
- Department of Hepatobiliary Surgery, National Cancer Center, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| |
Collapse
|
9
|
Samsom KG, van Veenendaal LM, Valk GD, Vriens MR, Tesselaar MET, van den Berg JG. Molecular prognostic factors in small-intestinal neuroendocrine tumours. Endocr Connect 2019; 8:906-922. [PMID: 31189127 PMCID: PMC6599083 DOI: 10.1530/ec-19-0206] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 06/10/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND Small-intestinal neuroendocrine tumours (SI-NETs) represent a heterogeneous group of rare tumours. In recent years, basic research in SI-NETs has attempted to unravel the molecular events underlying SI-NET tumorigenesis. AIM We aim to provide an overview of the current literature regarding prognostic and predictive molecular factors in patients with SI-NETs. METHOD A PubMed search was conducted on (epi)genetic prognostic factors in SI-NETs from 2000 until 2019. RESULTS The search yielded 1522 articles of which 20 reviews and 35 original studies were selected for further evaluation. SI-NETs are mutationally quiet tumours with a different genetic make-up compared to pancreatic NETs. Loss of heterozygosity at chromosome 18 is the most frequent genomic aberration (44-100%) followed by mutations of CDKN1B in 8%. Prognostic analyses were performed in 16 studies, of which 8 found a significant (epi)genetic association for survival or progression. Loss of heterozygosity at chromosome 18, gains of chromosome 4, 5, 7, 14 and 20p, copy gain of the SRC gene and low expression of RASSF1A and P16 were associated with poorer survival. In comparison with genetic mutations, epigenetic alterations are significantly more common in SI-NETs and may represent more promising targets in the treatment of SI-NETs. CONCLUSION SI-NETs are mutationally silent tumours. No biomarkers have been identified yet that can easily be adopted into current clinical decision making. SI-NETs may represent a heterogeneous disease and larger international studies are warranted to translate molecular findings into precision oncology.
Collapse
Affiliation(s)
- K G Samsom
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - L M van Veenendaal
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - G D Valk
- Department of Endocrine Oncology, University Medical Centre Utrecht, Utrecht, The Netherlands
- Correspondence should be addressed to G D Valk:
| | - M R Vriens
- Department of Surgical Oncology and Endocrine Surgery, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - M E T Tesselaar
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - J G van den Berg
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| |
Collapse
|
10
|
Herrera-Martínez AD, Gahete MD, Sánchez-Sánchez R, Alors-Perez E, Pedraza-Arevalo S, Serrano-Blanch R, Martínez-Fuentes AJ, Gálvez-Moreno MA, Castaño JP, Luque RM. Ghrelin-O-Acyltransferase (GOAT) Enzyme as a Novel Potential Biomarker in Gastroenteropancreatic Neuroendocrine Tumors. Clin Transl Gastroenterol 2018; 9:196. [PMID: 30297816 PMCID: PMC6175927 DOI: 10.1038/s41424-018-0058-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 08/16/2018] [Accepted: 08/17/2018] [Indexed: 12/18/2022] Open
Abstract
OBJECTIVES The association between the presence and alterations of the components of the ghrelin system and the development and progression of neuroendocrine tumors (NETs) is still controversial and remains unclear. METHODS Here, we systematically evaluated the expression levels (by quantitative-PCR) of key ghrelin system components of in gastroenteropancreatic (GEP)-NETs, as compared to non-tumor adjacent (NTA; n = 42) and normal tissues (NT; n = 14). Then, we analyzed their putative associations with clinical-histological characteristics. RESULTS The results indicate that ghrelin and its receptor GHSR1a are present in a high proportion of normal tissues, while the enzyme ghrelin-O-acyltransferase (GOAT) and the splicing variants In1-ghrelin and GHSR1b were present in a lower proportion of normal tissues. In contrast, all ghrelin system components were present in a high proportion of tumor and NTA tissues. GOAT was significantly overexpressed (by quantitative-PCR (qPCR)) in tumor samples compared to NTA, while a trend was found for ghrelin, In1-ghrelin and GHSR1a. In addition, expression of these components displayed significant correlations with key clinical parameters. The marked overexpression of GOAT in tumor samples compared to NTA regions was confirmed by IHC, revealing that this enzyme is particularly overexpressed in gastrointestinal NETs, where it is directly correlated with tumor diameter. CONCLUSIONS These results provide novel information on the presence and potential pathophysiological implications of the ghrelin system components in GEP-NETs, wherein GOAT might represent a novel diagnostic biomarker.
Collapse
Affiliation(s)
- Aura D Herrera-Martínez
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain.,Endocrinology and Nutrition Service, Reina Sofia University Hospital, Córdoba, Spain
| | - Manuel D Gahete
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain.,Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain.,Reina Sofia University Hospital, Córdoba, Spain
| | - Rafael Sánchez-Sánchez
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain.,Pathology Service, Reina Sofia University Hospital, Córdoba, Spain
| | - Emilia Alors-Perez
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain.,Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain.,Reina Sofia University Hospital, Córdoba, Spain
| | - Sergio Pedraza-Arevalo
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain.,Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain.,Reina Sofia University Hospital, Córdoba, Spain
| | - Raquel Serrano-Blanch
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain.,Medical Oncology Service, Reina Sofia University Hospital, Córdoba, Spain
| | - Antonio J Martínez-Fuentes
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain.,Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain.,Reina Sofia University Hospital, Córdoba, Spain
| | - Maria A Gálvez-Moreno
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain. .,Endocrinology and Nutrition Service, Reina Sofia University Hospital, Córdoba, Spain.
| | - Justo P Castaño
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain. .,Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain. .,CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain. .,Reina Sofia University Hospital, Córdoba, Spain.
| | - Raúl M Luque
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain. .,Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain. .,CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain. .,Reina Sofia University Hospital, Córdoba, Spain.
| |
Collapse
|
11
|
Wagner M, Samaha D, Cuervo J, Patel H, Martinez M, O'Neil WM, Jimenez-Fonseca P. Applying Reflective Multicriteria Decision Analysis (MCDA) to Patient-Clinician Shared Decision-Making on the Management of Gastroenteropancreatic Neuroendocrine Tumors (GEP-NET) in the Spanish Context. Adv Ther 2018; 35:1215-1231. [PMID: 29987525 PMCID: PMC6096971 DOI: 10.1007/s12325-018-0745-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Indexed: 12/18/2022]
Abstract
Introduction Unresectable, well-differentiated nonfunctioning gastroenteropancreatic neuroendocrine tumors (GEP-NETs) can be monitored (watchful waiting, WW) or treated with systemic therapy such as somatostatin analogues (SSAs) to delay progression. We applied a reflective multicriteria decision analysis (MCDA) shared-decision framework (previously developed for the USA) to explore what matters to Spanish patients and clinicians considering GEP-NET treatment options. Methods The EVIDEM-derived framework was updated and adapted to the Spanish context. During a Chatham House session, five patients and six physicians assigned criteria weights using hierarchical point allocation and direct rating scale (alternative analysis). Informed by synthesized evidence embedded in the framework, participants scored how each criterion favored SSA treatment (reference case lanreotide) or WW and shared insights and knowledge. Weights and scores were combined into value contributions (norm. weight × score/5), which were added across criteria to derive the relative benefit–risk balance (RBRB, scale − 1 to + 1). Exploratory comparisons to US study findings were performed. Results Focusing on intervention outcomes (effectiveness, patient-reported, and safety), the mean RBRB favored treatment over WW (+ 0.32 ± 0.24), with the largest contributions from progression-free survival (+ 0.11 ± SD 0.07), fatal adverse events (+ 0.06 ± SD 0.08), and impact on HRQoL (+ 0.04 ± SD 0.04). Consideration of modulating criteria (type of benefit, need, costs, evidence, and feasibility) increased the RBRB to + 0.50 ± 0.14, with type of therapeutic benefit (+ 0.10 ± SD 0.08) and quality of evidence (+ 0.08 ± SD 0.06) contributing most towards treatment. Alternative weighting yielded similar results. Results were broadly comparable to those derived from the US study. Conclusion The multicriteria framework helped Spanish patients and clinicians identify and express what matters to them. The approach is transferable across decision-making contexts. Funding IPSEN Pharma. Electronic supplementary material The online version of this article (10.1007/s12325-018-0745-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Paula Jimenez-Fonseca
- Department of Medical Oncology, Hospital Universitario Central de Asturias, Oviedo, Spain
| |
Collapse
|
12
|
Herrera-Martínez AD, Gahete MD, Pedraza-Arevalo S, Sánchez-Sánchez R, Ortega-Salas R, Serrano-Blanch R, Luque RM, Gálvez-Moreno MA, Castaño JP. Clinical and functional implication of the components of somatostatin system in gastroenteropancreatic neuroendocrine tumors. Endocrine 2018; 59:426-437. [PMID: 29196939 DOI: 10.1007/s12020-017-1482-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2017] [Accepted: 11/20/2017] [Indexed: 02/06/2023]
Abstract
PURPOSE Gastroenteropancreatic neuroendocrine tumors (GEP-NETs) comprise a heterogeneous group of malignancies often presenting with metastasis at diagnosis and whose clinical outcome is difficult to predict. Somatostatin (SST) analogs (SSAs) provide a valuable pharmacological tool to palliate hormonal symptoms, and control progression in some NETs. However, many patients do not respond to SSAs or develop resistance, and there are many uncertainties regarding pathophysiology of SST and its receptors (sst1-sst5) in GEP-NETs. METHODS The expression of SST system components in GEP-NETs was determined, compared with that of non-tumor adjacent and normal tissues and correlated with clinical and histological characteristics. Specifically, 58 patients with GEP-NETs and 14 normal samples were included. Cell viability in NET cell lines was determined in response to specific SSAs. RESULTS Normal samples and non-tumor adjacent tissues presented a similar expression profile, with appreciable expression of sst2 and sst3, and a lower expression of the other receptors. In contrast, cortistatin, sst1, sst4, and sst5 were overexpressed in tumors, while sst3 and sst4 seemed overexpressed in less differentiated tumors. Some SST system components were related to vascular/nerve invasion and metastasis. In vitro, sst1 and sst3 agonists reduced viability in BON-1 cells, while they, similar to octreotide and pasireotide, increased viability in QGP-1 cells. CONCLUSIONS These results provide novel information on SST system pathophysiology in GEP-NETs, including relevant associations with clinical-histological parameters, which might help to better understand the intrinsic heterogeneity of NETs and to identify novel biomarkers and/or targets with potential prognostic and/or therapeutic value for GEP-NETs patients.
Collapse
Affiliation(s)
- Aura D Herrera-Martínez
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain
- Endocrinology and Nutrition Service, Reina Sofia University Hospital, Córdoba, Spain
| | - Manuel D Gahete
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain
- Reina Sofia University Hospital, Córdoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain
- Campus de Excelencia Internacional Agroalimentario (ceiA3), Córdoba, Spain
| | - Sergio Pedraza-Arevalo
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain
- Reina Sofia University Hospital, Córdoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain
- Campus de Excelencia Internacional Agroalimentario (ceiA3), Córdoba, Spain
| | - Rafael Sánchez-Sánchez
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain
- Pathology Service, Reina Sofia University Hospital, Córdoba, Spain
| | - Rosa Ortega-Salas
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain
- Pathology Service, Reina Sofia University Hospital, Córdoba, Spain
| | - Raquel Serrano-Blanch
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain
- Medical Oncology Service, Reina Sofia University Hospital, Córdoba, Spain
| | - Raúl M Luque
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain.
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain.
- Reina Sofia University Hospital, Córdoba, Spain.
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain.
- Campus de Excelencia Internacional Agroalimentario (ceiA3), Córdoba, Spain.
| | - María A Gálvez-Moreno
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain.
- Endocrinology and Nutrition Service, Reina Sofia University Hospital, Córdoba, Spain.
| | - Justo P Castaño
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain.
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain.
- Reina Sofia University Hospital, Córdoba, Spain.
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain.
- Campus de Excelencia Internacional Agroalimentario (ceiA3), Córdoba, Spain.
| |
Collapse
|
13
|
Ligiero Braga T, Santos-Oliveira R. PPoma Review: Epidemiology, Aetiopathogenesis, Prognosis and Treatment. Diseases 2018; 6:diseases6010008. [PMID: 29324681 PMCID: PMC5871954 DOI: 10.3390/diseases6010008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 11/06/2017] [Accepted: 11/13/2017] [Indexed: 02/06/2023] Open
Abstract
Generally, pancreatic polypeptide-secreting tumor of the distal pancreas (PPoma) is classified as a rare tumor, and may occur sporadically or be associated in families or with multiple endocrine neoplasia type 1 (NEM 1). It grows slowly, reaching large dimensions at the time of diagnosis and the symptomatology is fundamentally due to the mass effect, causing either non-specific abdominal pain or symptoms suggestive of obstruction of the pancreatic or biliary duct. Therefore, when detected, they are usually malignant, with metastases mainly in the liver. The combination of serum analysis of increased levels of chromogranin A and pancreatic polypeptide and pancreastatin is very useful with a sensitivity of up to 95%. However, in addition, scintigraphicexams with somatostatin analogues should be performed to better clarify the diagnosis. Surgical resection is the treatment of choice, despite surgical difficulty and because they are generally palliative due to the metastases. Surgeries for tumor volume reduction are also performed to relieve symptoms. Chemotherapy commonly uses streptozotocin and somatostatin analogues to treat residual disease. Unfortunately, the survival rates are still very low, less than 10%, and if metastases already exist, this percentage drops to 3%.
Collapse
Affiliation(s)
- Thais Ligiero Braga
- Brazilian Nuclear Energy Commission, Nuclear Engineering Institute, Rio de Janeiro 21941906, Brazil.
| | - Ralph Santos-Oliveira
- Brazilian Nuclear Energy Commission, Nuclear Engineering Institute, Rio de Janeiro 21941906, Brazil.
| |
Collapse
|
14
|
Petersenn S, Koch CA. Neuroendocrine neoplasms - still a challenge despite major advances in clinical care with the development of specialized guidelines. Rev Endocr Metab Disord 2017; 18:373-378. [PMID: 29480376 DOI: 10.1007/s11154-018-9442-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Stephan Petersenn
- ENDOC Center for Endocrine Tumors, Erik-Blumenfeld-Platz 27a, 22587, Hamburg, Germany.
| | - Christian A Koch
- Medicover Oldenburg MVZ, Oldenburg, Germany
- Department of Medicine III, Technical University of Dresden, Dresden, Germany
- University of Louisville, Louisville, KY, USA
| |
Collapse
|
15
|
Zhang M, Zhao P, Shi X, Zhao A, Zhang L, Zhou L. Clinicopathological features and prognosis of gastroenteropancreatic neuroendocrine neoplasms in a Chinese population: a large, retrospective single-centre study. BMC Endocr Disord 2017; 17:39. [PMID: 28705205 PMCID: PMC5508659 DOI: 10.1186/s12902-017-0190-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 06/30/2017] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Gastroenteropancreatic neuroendocrine neoplasms (GEP-NENs) are the most common type of neuroendocrine tumors, accounting for more than half of neuroendocrine neoplasms (NENs). We performed a retrospective study in our center to investigate the clinicopathological features, risk factors of metastasis, and prognosis of GEP-NENs in a Chinese population. METHODS Four hundred forty patients with GEP-NENs treated at the First Affiliated Hospital of Zhengzhou University between January 2011 and March 2016 were analyzed retrospectively. Multivariate logistic regression was performed to identify independent risk factors for metastasis of the tumors. The Kaplan-Meier method was used for survival analysis, and log-rank tests for comparisons among groups. RESULTS Primary sites were the stomach (24.3%), rectum (24.1%), pancreas (20.5%), esophagus (12.3%), unknown primary origin (UPO-NEN) (8.0%), duodenum (6.1%). Three hundred eighty-nine of the 440 GEP-NENs cases (88.4%) were non-functional tumors, and patients had non-specific symptoms, which could have led to delay in diagnosis and treatment. Neuroendocrine tumor, neuroendocrine carcinoma, and mixed adenoendocrine carcinoma were 56.8%, 33.2% and 3.2%, respectively, of the cases. One hundred thirty (29.5%) of the tumors were G1, 120 (27.3%) G2, and 190 (43.2%) G3. The immunohistochemical positive rate of synaptophysin was 97.7% and of chromogranin 48.7%. Logistic regression analysis revealed that the diameter and pathological classification of tumors were the most important predictors for metastasis. The median survival time was 34 months for patients with well-differentiated neuroendocrine tumors grade G3 and 11 months for poorly differentiated neuroendocrine carcinoma. The median survival time of patients with localized disease, regional disease, and distant disease was 36 months, 15 month, and 6 months, respectively. CONCLUSIONS This study constitutes a comprehensive analysis of the clinicopathological features of GEP-NENs in a Chinese population. GEP-NENs may occur at any part of the digestive system. The diameter and pathological classification of tumor are the most important predictors for metastasis. The prognosis is poor for patients with poorly differentiated neuroendocrine cancers and distant metastases.
Collapse
Affiliation(s)
- Meng Zhang
- Department of Gastroenterology, the First Affiliated Hospital of Zhengzhou University, No.1, East Jianshe Road, Zhengzhou, 450052 China
| | - Ping Zhao
- Department of Gastroenterology, the First Affiliated Hospital of Zhengzhou University, No.1, East Jianshe Road, Zhengzhou, 450052 China
| | - Xiaodan Shi
- Department of Gastroenterology, the First Affiliated Hospital of Zhengzhou University, No.1, East Jianshe Road, Zhengzhou, 450052 China
| | - Ahong Zhao
- Department of Pathology, the First Affiliated Hospital of Zhengzhou University, No.1, East Jianshe Road, Zhengzhou, 450052 China
| | - Lianfeng Zhang
- Department of Gastroenterology, the First Affiliated Hospital of Zhengzhou University, No.1, East Jianshe Road, Zhengzhou, 450052 China
| | - Lin Zhou
- Department of Gastroenterology, the First Affiliated Hospital of Zhengzhou University, No.1, East Jianshe Road, Zhengzhou, 450052 China
| |
Collapse
|
16
|
Management Options for Advanced Low or Intermediate Grade Gastroenteropancreatic Neuroendocrine Tumors: Review of Recent Literature. Int J Surg Oncol 2017; 2017:6424812. [PMID: 28593056 PMCID: PMC5448049 DOI: 10.1155/2017/6424812] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 04/13/2017] [Accepted: 04/26/2017] [Indexed: 02/06/2023] Open
Abstract
Our understanding of the biology, genetics, and natural history of neuroendocrine tumors (NETs) of the gastrointestinal tract and pancreas has improved considerably in the last several decades and the spectrum of available therapeutic options is rapidly expanding. The management of patients with metastatic low or intermediate grade NETs has been revolutionized by the development of new treatment strategies such as molecular targeting therapies with everolimus and sunitinib, somatostatin analogs, tryptophan hydroxylase inhibitors, and peptide receptor radionuclide therapy that can be used alone or as a multimodal approach with or without surgery. To further define and clarify the utility, appropriateness, and the sequence of the growing list of available therapies for this patient population will require more high level evidence; however, data from well-designed randomized phase III clinical trials is rapidly accumulating that will further stimulate development of new management strategies. It is therefore important to thoroughly review emerging evidence and report major findings in frequent updates, which will expand our knowledge and contribute to a better understanding, characterization, and management of advanced NETs.
Collapse
|
17
|
Sevilla I, Segura Á, Capdevila J, López C, García-Carbonero R, Grande E. Management of controversial gastroenteropancreatic neuroendocrine tumour clinical situations with somatostatin analogues: results of a Delphi questionnaire panel from the NETPraxis program. BMC Cancer 2016; 16:858. [PMID: 27821081 PMCID: PMC5100262 DOI: 10.1186/s12885-016-2901-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 10/30/2016] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND There are clinical situations (CS) in which the use of somatostatin analogs (SSAs) in patients with neuroendocrine tumors (NET) is controversial due to lack of evidence. A Delphi study was conducted to develop common treatment guidelines for these CS, based on clinical practice and expert opinion of Spanish oncologists. METHODS A scientific committee identified 5 CS with a common core (c-c) [non-functioning NET, not susceptible of surgery/locoregional therapy, Ki67 < 10 % (except for CS5: >10 %), ECOG ≤ 2], and controversy regarding use of SSAs, and prepared a Delphi questionnaire of 48 treatment statements. Statements were rated on a 1 (completely disagree) to 9 (completely agree) scale. Responses were grouped by tertiles: 1-3: Disagreement, 4-6: Neutral, 7-9: Agreement. Consensus was reached when the responses of ≥2/3 participants were located in the same tertile as the median value of all reported responses for that statement. RESULTS Sixty five (81.2 %) of 80 invited oncologists with experience in the management of NETs answered a first round of the questionnaire and 57 (87.7 %) of those 65 answered a second round (mean age 43.5 years; 53.8 % women; median time of experience 9 years). Consensus was obtained in 42 (36 agreement and 6 disagreement) of the 48 statements (87.5 %). Regarding CS1 (Enteropancreatic NET, c-c, non-progressive in the last 3-6 months), overall, SSA treatment is recommended (a wait and see approach is anecdotal and reserved for fragile patients or with low tumor load or ki-67 < 2 %); CS2 (Pancreatic NET, c-c), overall, SSA monotherapy is recommended, except when high tumor load or tumor progression exists, where combination therapy would be considered; CS3 [Gastroenteropancreatic (GEP)-NET, c-c, in treatment with anti-proliferative dose of SSA and progressing], overall, SSA maintenance is recommended at the time of progression, with or without adding molecular targeted drugs; CS4 (GEP-NET, c-c, and negative octreoscan®), SSA in monotherapy is only considered in low-risk patients (low tumor load and Ki-67 < 5 %); CS5 [GEP-NET, c-c (ki67 > 10 %), and positive octreoscan®], monotherapy with SSA is mainly considered in patients with comorbidities. CONCLUSION Several recommendations regarding use of SSAs in controversial NET CS were reached in consensus and might be considered as treatment guideline.
Collapse
Affiliation(s)
- Isabel Sevilla
- Oncology Unit. Hospital Clínico y Regional de Málaga, Colonia Santa Inés s/n, Málaga, 29010 Spain
| | - Ángel Segura
- Oncology Unit. Hospital Universitario La Fe, Avda. de Fernando Abril Martorell 106, 46026 Valencia, Spain
| | - Jaume Capdevila
- Oncology Unit. Hospital Vall d’Hebron, Pg de la Vall d’Hebron 119-129, 08035 Barcelona, Spain
| | - Carlos López
- Oncology Unit. Hospital Marqués de Valdecilla, Avda. Valdecilla 25, 39008 Santander, Spain
| | - Rocío García-Carbonero
- Oncology Unit. Hospital Universitario 12 de Octubre, Avda. de Córdoba s/n, 28041 Madrid, Spain
| | - Enrique Grande
- Oncology Unit. Hospital Universitario Ramón y Cajal, Ctra. de Colmenar Viejo km. 9.100, 28034 Madrid, Spain
| | - On behalf of GETNE (Spanish Group of NeuroEndocrine Tumors)
- Oncology Unit. Hospital Clínico y Regional de Málaga, Colonia Santa Inés s/n, Málaga, 29010 Spain
- Oncology Unit. Hospital Universitario La Fe, Avda. de Fernando Abril Martorell 106, 46026 Valencia, Spain
- Oncology Unit. Hospital Vall d’Hebron, Pg de la Vall d’Hebron 119-129, 08035 Barcelona, Spain
- Oncology Unit. Hospital Marqués de Valdecilla, Avda. Valdecilla 25, 39008 Santander, Spain
- Oncology Unit. Hospital Universitario 12 de Octubre, Avda. de Córdoba s/n, 28041 Madrid, Spain
- Oncology Unit. Hospital Universitario Ramón y Cajal, Ctra. de Colmenar Viejo km. 9.100, 28034 Madrid, Spain
| |
Collapse
|
18
|
Fotouhi O, Kjellin H, Larsson C, Hashemi J, Barriuso J, Juhlin CC, Lu M, Höög A, Pastrián LG, Lamarca A, Soto VH, Zedenius J, Mendiola M, Lehtiö J, Kjellman M. Proteomics Suggests a Role for APC-Survivin in Response to Somatostatin Analog Treatment of Neuroendocrine Tumors. J Clin Endocrinol Metab 2016; 101:3616-3627. [PMID: 27459532 PMCID: PMC5052342 DOI: 10.1210/jc.2016-2028] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
CONTEXT Somatostatin analogs are established in the treatment of neuroendocrine tumors (NETs) including small intestinal NET; however, the molecular mechanisms are not well known. Here, we examined the direct effects of lanreotide in NET cell line models. SETTING AND DESIGN The cell lines HC45 and H727 were treated with 10nM lanreotide for different time periods and alterations of the proteome were analyzed by in-depth high-resolution isoelectric focusing tandem liquid chromatography-mass spectrometry. We next investigated whether the observed suppression of survivin was mediated by adenomatous polyposis coli (APC) and possible effects on tumor proliferation in vitro. Expression of survivin was assessed by immunohistochemistry in 112 NET cases and compared with patient outcome. RESULTS We quantified 6451 and 7801 proteins in HC45 and H727, respectively. After short time lanreotide treatment APC was increased and survivin reduced. Overexpression of APC in H727 cells decreased, and APC knock-down elevated the survivin level. The lanreotide regulation of APC-survivin could be suppressed by small interfering RNA against somatostatin receptor 2. Although lanreotide only gave slight inhibition of proliferation, targeting of survivin with the small molecule YM155 dramatically reduced proliferation. Moderate or high as compared with low or absent total survivin expression was associated with shorter progression-free survival, independent of tumor stage, grade, and localization. CONCLUSIONS We report a proteome-wide analysis of changes in response to lanreotide in NET cell lines. This analysis suggests a connection between somatostatin analog, APC, and survivin levels. Survivin is a possible prognostic factor and a new potential therapeutic target in NETs.
Collapse
Affiliation(s)
- Omid Fotouhi
- Departments of Oncology-Pathology (O.F., C.L., J.H., C.C.J., M.L., A.H., J.L.) and Molecular Medicine and Surgery (H.K., J.Z., M.K.), Karolinska Institutet, Stockholm, Sweden; Cancer Center Karolinska (O.F., C.L., J.H., C.C.J., M.L., A.H.), Karolinska University Hospital, Stockholm, Sweden; Cancer Proteomics Mass Spectrometry (H.K., J.L.), Science for Life Laboratory, Stockholm, Sweden SE-171 76; Faculty of Biology (J.B.), Medicine and Health, University of Manchester, M13 9PT, Manchester, United Kingdom; Laboratory of Molecular Pathology and Therapeutic Targets, and Translational Oncology Research Group (J.B., A.L., V.H.S., M.M.), Instituto de Investigación; Department of Pathology (L.G.P.); and Molecular Pathology Section (M.M.), Instituto de Genética Médica, Hospital Universitario La Paz 28046, Madrid, Spain; and Department of Medical Oncology (A.L.), The Christie NHS Trust, M20 4BX, Manchester, United Kingdom
| | - Hanna Kjellin
- Departments of Oncology-Pathology (O.F., C.L., J.H., C.C.J., M.L., A.H., J.L.) and Molecular Medicine and Surgery (H.K., J.Z., M.K.), Karolinska Institutet, Stockholm, Sweden; Cancer Center Karolinska (O.F., C.L., J.H., C.C.J., M.L., A.H.), Karolinska University Hospital, Stockholm, Sweden; Cancer Proteomics Mass Spectrometry (H.K., J.L.), Science for Life Laboratory, Stockholm, Sweden SE-171 76; Faculty of Biology (J.B.), Medicine and Health, University of Manchester, M13 9PT, Manchester, United Kingdom; Laboratory of Molecular Pathology and Therapeutic Targets, and Translational Oncology Research Group (J.B., A.L., V.H.S., M.M.), Instituto de Investigación; Department of Pathology (L.G.P.); and Molecular Pathology Section (M.M.), Instituto de Genética Médica, Hospital Universitario La Paz 28046, Madrid, Spain; and Department of Medical Oncology (A.L.), The Christie NHS Trust, M20 4BX, Manchester, United Kingdom
| | - Catharina Larsson
- Departments of Oncology-Pathology (O.F., C.L., J.H., C.C.J., M.L., A.H., J.L.) and Molecular Medicine and Surgery (H.K., J.Z., M.K.), Karolinska Institutet, Stockholm, Sweden; Cancer Center Karolinska (O.F., C.L., J.H., C.C.J., M.L., A.H.), Karolinska University Hospital, Stockholm, Sweden; Cancer Proteomics Mass Spectrometry (H.K., J.L.), Science for Life Laboratory, Stockholm, Sweden SE-171 76; Faculty of Biology (J.B.), Medicine and Health, University of Manchester, M13 9PT, Manchester, United Kingdom; Laboratory of Molecular Pathology and Therapeutic Targets, and Translational Oncology Research Group (J.B., A.L., V.H.S., M.M.), Instituto de Investigación; Department of Pathology (L.G.P.); and Molecular Pathology Section (M.M.), Instituto de Genética Médica, Hospital Universitario La Paz 28046, Madrid, Spain; and Department of Medical Oncology (A.L.), The Christie NHS Trust, M20 4BX, Manchester, United Kingdom
| | - Jamileh Hashemi
- Departments of Oncology-Pathology (O.F., C.L., J.H., C.C.J., M.L., A.H., J.L.) and Molecular Medicine and Surgery (H.K., J.Z., M.K.), Karolinska Institutet, Stockholm, Sweden; Cancer Center Karolinska (O.F., C.L., J.H., C.C.J., M.L., A.H.), Karolinska University Hospital, Stockholm, Sweden; Cancer Proteomics Mass Spectrometry (H.K., J.L.), Science for Life Laboratory, Stockholm, Sweden SE-171 76; Faculty of Biology (J.B.), Medicine and Health, University of Manchester, M13 9PT, Manchester, United Kingdom; Laboratory of Molecular Pathology and Therapeutic Targets, and Translational Oncology Research Group (J.B., A.L., V.H.S., M.M.), Instituto de Investigación; Department of Pathology (L.G.P.); and Molecular Pathology Section (M.M.), Instituto de Genética Médica, Hospital Universitario La Paz 28046, Madrid, Spain; and Department of Medical Oncology (A.L.), The Christie NHS Trust, M20 4BX, Manchester, United Kingdom
| | - Jorge Barriuso
- Departments of Oncology-Pathology (O.F., C.L., J.H., C.C.J., M.L., A.H., J.L.) and Molecular Medicine and Surgery (H.K., J.Z., M.K.), Karolinska Institutet, Stockholm, Sweden; Cancer Center Karolinska (O.F., C.L., J.H., C.C.J., M.L., A.H.), Karolinska University Hospital, Stockholm, Sweden; Cancer Proteomics Mass Spectrometry (H.K., J.L.), Science for Life Laboratory, Stockholm, Sweden SE-171 76; Faculty of Biology (J.B.), Medicine and Health, University of Manchester, M13 9PT, Manchester, United Kingdom; Laboratory of Molecular Pathology and Therapeutic Targets, and Translational Oncology Research Group (J.B., A.L., V.H.S., M.M.), Instituto de Investigación; Department of Pathology (L.G.P.); and Molecular Pathology Section (M.M.), Instituto de Genética Médica, Hospital Universitario La Paz 28046, Madrid, Spain; and Department of Medical Oncology (A.L.), The Christie NHS Trust, M20 4BX, Manchester, United Kingdom
| | - C Christofer Juhlin
- Departments of Oncology-Pathology (O.F., C.L., J.H., C.C.J., M.L., A.H., J.L.) and Molecular Medicine and Surgery (H.K., J.Z., M.K.), Karolinska Institutet, Stockholm, Sweden; Cancer Center Karolinska (O.F., C.L., J.H., C.C.J., M.L., A.H.), Karolinska University Hospital, Stockholm, Sweden; Cancer Proteomics Mass Spectrometry (H.K., J.L.), Science for Life Laboratory, Stockholm, Sweden SE-171 76; Faculty of Biology (J.B.), Medicine and Health, University of Manchester, M13 9PT, Manchester, United Kingdom; Laboratory of Molecular Pathology and Therapeutic Targets, and Translational Oncology Research Group (J.B., A.L., V.H.S., M.M.), Instituto de Investigación; Department of Pathology (L.G.P.); and Molecular Pathology Section (M.M.), Instituto de Genética Médica, Hospital Universitario La Paz 28046, Madrid, Spain; and Department of Medical Oncology (A.L.), The Christie NHS Trust, M20 4BX, Manchester, United Kingdom
| | - Ming Lu
- Departments of Oncology-Pathology (O.F., C.L., J.H., C.C.J., M.L., A.H., J.L.) and Molecular Medicine and Surgery (H.K., J.Z., M.K.), Karolinska Institutet, Stockholm, Sweden; Cancer Center Karolinska (O.F., C.L., J.H., C.C.J., M.L., A.H.), Karolinska University Hospital, Stockholm, Sweden; Cancer Proteomics Mass Spectrometry (H.K., J.L.), Science for Life Laboratory, Stockholm, Sweden SE-171 76; Faculty of Biology (J.B.), Medicine and Health, University of Manchester, M13 9PT, Manchester, United Kingdom; Laboratory of Molecular Pathology and Therapeutic Targets, and Translational Oncology Research Group (J.B., A.L., V.H.S., M.M.), Instituto de Investigación; Department of Pathology (L.G.P.); and Molecular Pathology Section (M.M.), Instituto de Genética Médica, Hospital Universitario La Paz 28046, Madrid, Spain; and Department of Medical Oncology (A.L.), The Christie NHS Trust, M20 4BX, Manchester, United Kingdom
| | - Anders Höög
- Departments of Oncology-Pathology (O.F., C.L., J.H., C.C.J., M.L., A.H., J.L.) and Molecular Medicine and Surgery (H.K., J.Z., M.K.), Karolinska Institutet, Stockholm, Sweden; Cancer Center Karolinska (O.F., C.L., J.H., C.C.J., M.L., A.H.), Karolinska University Hospital, Stockholm, Sweden; Cancer Proteomics Mass Spectrometry (H.K., J.L.), Science for Life Laboratory, Stockholm, Sweden SE-171 76; Faculty of Biology (J.B.), Medicine and Health, University of Manchester, M13 9PT, Manchester, United Kingdom; Laboratory of Molecular Pathology and Therapeutic Targets, and Translational Oncology Research Group (J.B., A.L., V.H.S., M.M.), Instituto de Investigación; Department of Pathology (L.G.P.); and Molecular Pathology Section (M.M.), Instituto de Genética Médica, Hospital Universitario La Paz 28046, Madrid, Spain; and Department of Medical Oncology (A.L.), The Christie NHS Trust, M20 4BX, Manchester, United Kingdom
| | - Laura G Pastrián
- Departments of Oncology-Pathology (O.F., C.L., J.H., C.C.J., M.L., A.H., J.L.) and Molecular Medicine and Surgery (H.K., J.Z., M.K.), Karolinska Institutet, Stockholm, Sweden; Cancer Center Karolinska (O.F., C.L., J.H., C.C.J., M.L., A.H.), Karolinska University Hospital, Stockholm, Sweden; Cancer Proteomics Mass Spectrometry (H.K., J.L.), Science for Life Laboratory, Stockholm, Sweden SE-171 76; Faculty of Biology (J.B.), Medicine and Health, University of Manchester, M13 9PT, Manchester, United Kingdom; Laboratory of Molecular Pathology and Therapeutic Targets, and Translational Oncology Research Group (J.B., A.L., V.H.S., M.M.), Instituto de Investigación; Department of Pathology (L.G.P.); and Molecular Pathology Section (M.M.), Instituto de Genética Médica, Hospital Universitario La Paz 28046, Madrid, Spain; and Department of Medical Oncology (A.L.), The Christie NHS Trust, M20 4BX, Manchester, United Kingdom
| | - Angela Lamarca
- Departments of Oncology-Pathology (O.F., C.L., J.H., C.C.J., M.L., A.H., J.L.) and Molecular Medicine and Surgery (H.K., J.Z., M.K.), Karolinska Institutet, Stockholm, Sweden; Cancer Center Karolinska (O.F., C.L., J.H., C.C.J., M.L., A.H.), Karolinska University Hospital, Stockholm, Sweden; Cancer Proteomics Mass Spectrometry (H.K., J.L.), Science for Life Laboratory, Stockholm, Sweden SE-171 76; Faculty of Biology (J.B.), Medicine and Health, University of Manchester, M13 9PT, Manchester, United Kingdom; Laboratory of Molecular Pathology and Therapeutic Targets, and Translational Oncology Research Group (J.B., A.L., V.H.S., M.M.), Instituto de Investigación; Department of Pathology (L.G.P.); and Molecular Pathology Section (M.M.), Instituto de Genética Médica, Hospital Universitario La Paz 28046, Madrid, Spain; and Department of Medical Oncology (A.L.), The Christie NHS Trust, M20 4BX, Manchester, United Kingdom
| | - Victoria Heredia Soto
- Departments of Oncology-Pathology (O.F., C.L., J.H., C.C.J., M.L., A.H., J.L.) and Molecular Medicine and Surgery (H.K., J.Z., M.K.), Karolinska Institutet, Stockholm, Sweden; Cancer Center Karolinska (O.F., C.L., J.H., C.C.J., M.L., A.H.), Karolinska University Hospital, Stockholm, Sweden; Cancer Proteomics Mass Spectrometry (H.K., J.L.), Science for Life Laboratory, Stockholm, Sweden SE-171 76; Faculty of Biology (J.B.), Medicine and Health, University of Manchester, M13 9PT, Manchester, United Kingdom; Laboratory of Molecular Pathology and Therapeutic Targets, and Translational Oncology Research Group (J.B., A.L., V.H.S., M.M.), Instituto de Investigación; Department of Pathology (L.G.P.); and Molecular Pathology Section (M.M.), Instituto de Genética Médica, Hospital Universitario La Paz 28046, Madrid, Spain; and Department of Medical Oncology (A.L.), The Christie NHS Trust, M20 4BX, Manchester, United Kingdom
| | - Jan Zedenius
- Departments of Oncology-Pathology (O.F., C.L., J.H., C.C.J., M.L., A.H., J.L.) and Molecular Medicine and Surgery (H.K., J.Z., M.K.), Karolinska Institutet, Stockholm, Sweden; Cancer Center Karolinska (O.F., C.L., J.H., C.C.J., M.L., A.H.), Karolinska University Hospital, Stockholm, Sweden; Cancer Proteomics Mass Spectrometry (H.K., J.L.), Science for Life Laboratory, Stockholm, Sweden SE-171 76; Faculty of Biology (J.B.), Medicine and Health, University of Manchester, M13 9PT, Manchester, United Kingdom; Laboratory of Molecular Pathology and Therapeutic Targets, and Translational Oncology Research Group (J.B., A.L., V.H.S., M.M.), Instituto de Investigación; Department of Pathology (L.G.P.); and Molecular Pathology Section (M.M.), Instituto de Genética Médica, Hospital Universitario La Paz 28046, Madrid, Spain; and Department of Medical Oncology (A.L.), The Christie NHS Trust, M20 4BX, Manchester, United Kingdom
| | - Marta Mendiola
- Departments of Oncology-Pathology (O.F., C.L., J.H., C.C.J., M.L., A.H., J.L.) and Molecular Medicine and Surgery (H.K., J.Z., M.K.), Karolinska Institutet, Stockholm, Sweden; Cancer Center Karolinska (O.F., C.L., J.H., C.C.J., M.L., A.H.), Karolinska University Hospital, Stockholm, Sweden; Cancer Proteomics Mass Spectrometry (H.K., J.L.), Science for Life Laboratory, Stockholm, Sweden SE-171 76; Faculty of Biology (J.B.), Medicine and Health, University of Manchester, M13 9PT, Manchester, United Kingdom; Laboratory of Molecular Pathology and Therapeutic Targets, and Translational Oncology Research Group (J.B., A.L., V.H.S., M.M.), Instituto de Investigación; Department of Pathology (L.G.P.); and Molecular Pathology Section (M.M.), Instituto de Genética Médica, Hospital Universitario La Paz 28046, Madrid, Spain; and Department of Medical Oncology (A.L.), The Christie NHS Trust, M20 4BX, Manchester, United Kingdom
| | - Janne Lehtiö
- Departments of Oncology-Pathology (O.F., C.L., J.H., C.C.J., M.L., A.H., J.L.) and Molecular Medicine and Surgery (H.K., J.Z., M.K.), Karolinska Institutet, Stockholm, Sweden; Cancer Center Karolinska (O.F., C.L., J.H., C.C.J., M.L., A.H.), Karolinska University Hospital, Stockholm, Sweden; Cancer Proteomics Mass Spectrometry (H.K., J.L.), Science for Life Laboratory, Stockholm, Sweden SE-171 76; Faculty of Biology (J.B.), Medicine and Health, University of Manchester, M13 9PT, Manchester, United Kingdom; Laboratory of Molecular Pathology and Therapeutic Targets, and Translational Oncology Research Group (J.B., A.L., V.H.S., M.M.), Instituto de Investigación; Department of Pathology (L.G.P.); and Molecular Pathology Section (M.M.), Instituto de Genética Médica, Hospital Universitario La Paz 28046, Madrid, Spain; and Department of Medical Oncology (A.L.), The Christie NHS Trust, M20 4BX, Manchester, United Kingdom
| | - Magnus Kjellman
- Departments of Oncology-Pathology (O.F., C.L., J.H., C.C.J., M.L., A.H., J.L.) and Molecular Medicine and Surgery (H.K., J.Z., M.K.), Karolinska Institutet, Stockholm, Sweden; Cancer Center Karolinska (O.F., C.L., J.H., C.C.J., M.L., A.H.), Karolinska University Hospital, Stockholm, Sweden; Cancer Proteomics Mass Spectrometry (H.K., J.L.), Science for Life Laboratory, Stockholm, Sweden SE-171 76; Faculty of Biology (J.B.), Medicine and Health, University of Manchester, M13 9PT, Manchester, United Kingdom; Laboratory of Molecular Pathology and Therapeutic Targets, and Translational Oncology Research Group (J.B., A.L., V.H.S., M.M.), Instituto de Investigación; Department of Pathology (L.G.P.); and Molecular Pathology Section (M.M.), Instituto de Genética Médica, Hospital Universitario La Paz 28046, Madrid, Spain; and Department of Medical Oncology (A.L.), The Christie NHS Trust, M20 4BX, Manchester, United Kingdom
| |
Collapse
|
19
|
C8-T1 Radiculopathy Due to an Intradural Extramedullary Metastasis of a Pancreatic Neuroendocrine Tumor: Case Report and Review of the Literature. Pancreas 2016; 45:772-9. [PMID: 27077714 DOI: 10.1097/mpa.0000000000000515] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Pancreatic neuroendocrine tumors (pNETs) are usually well-to-moderately differentiated neuroendocrine tumors (NETs) that most often metastasize to the liver and lymph nodes with other locations being uncommon. We present a case of intradural pNET metastasis and conduct a review of the literature. Forty-five cases, including the case presently reported, of spinal cord compression due to well-differentiated NETs were found: carcinoid (80%), pNET (13.3%), and NETs of unknown primary (6.7%). Seventy-eight percent of cases consisted of extradural compressions from vertebral bone metastases, whereas there were only 5 cases of intradural extramedullary spinal cord compression. Most cases were managed with surgery and/or radiotherapy with a good clinical outcome in the majority. We report the first case of a pNET intradural extramedullary metastasis and conduct the largest review to date of an infrequent complication of well-differentiated NETs such as malignant spinal cord compression. Aggressive local treatment is warranted in most cases because it usually achieves neurologic improvement and symptomatic relief in patients who may still have a long life expectancy.
Collapse
|
20
|
Treatment of neuroendocrine tumors: new recommendations based on the CLARINET study. Contemp Oncol (Pozn) 2015; 19:345-9. [PMID: 26793016 PMCID: PMC4709401 DOI: 10.5114/wo.2015.56006] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 11/24/2015] [Indexed: 12/29/2022] Open
Abstract
Somatostatin analogs (SSAs), including lanreotide, play a fundamental role in treatment of neuroendocrine tumors (NETs) of the gastrointestinal tract. SSAs control the clinical symptoms and are the treatment of choice in functioning NETs. Data indicating that SSAs have anti-proliferative activity has mainly come from prospective or retrospective observational studies. A recently published CLARINET study confirmed the anti-proliferative effect of lanreotide in a much broader range of NET patients than previously reported. As a result, it is now possible for clinicians to use lanreotide to treat patients with well-differentiated metastatic grade 1 and grade 2 GEP NETs (i.e., with a Ki-67 proliferative index < 10%) located in the pancreas, small intestine, or of unknown primary location, regardless of the degree of liver involvement. The results of the CLARINET study also challenge the current “wait and watch” strategy for NET treatment. Instead, it is proposed that SSAs are considered at an early stage of NET management, as already suggested by many organizations and scientific societies.
Collapse
|
21
|
Alonso-Gordoa T, Díez JJ, Molina J, Reguera P, Martínez-Sáez O, Grande E. An Overview on the Sequential Treatment of Pancreatic Neuroendocrine Tumors (pNETs). ACTA ACUST UNITED AC 2015; 3:13-33. [PMID: 27182476 PMCID: PMC4837935 DOI: 10.1007/s40487-015-0007-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Indexed: 01/10/2023]
Abstract
Patients suffering from pancreatic neuroendocrine tumors (pNETs) are now candidates to receive novel approved drugs that have demonstrated benefit in disease control rate and delay the time taken for tumor progression in Phase III clinical trials; for example, sunitinib, everolimus and lanreotide. Though pNETs represent a rare and heterogeneous disease, recent approaches are being taken to better understand the molecular pathways involved in carcinogenesis. Consequently, new treatment strategies are now available and others still under investigation show promising results. However, some questions around how to approach patients with pNETs are still unresolved, such as what the best sequence of treatments we can offer to each of our patients in the clinic at any time of their disease would be. Therapeutic decisions are, at the moment, guided by clinical judgment, based on different parameters coming from retrospective analysis and non-randomized clinical trials. However, advances in genomic research would lead to a more precise approach using therapeutic targets that would also allow the development of new agents, prognostic or predictive biomarkers and a better understanding of resistance mechanisms. The following article is a comprehensive review of the approved and investigational drugs in pNET, and highlights the current concerns about treatment sequencing, but also provides an update of some of the present and future efforts for an improvement in the therapeutic algorithm of the disease.
Collapse
Affiliation(s)
- Teresa Alonso-Gordoa
- Medical Oncology Department, Ramón y Cajal University Hospital, Ctra. Colmenar Viejo km 9100, 28034 Madrid, Spain
| | - Juan José Díez
- Endocrinology and Nutrition Department, Ramón y Cajal University Hospital, Madrid, Spain
| | - Javier Molina
- Medical Oncology Department, Ramón y Cajal University Hospital, Ctra. Colmenar Viejo km 9100, 28034 Madrid, Spain
| | - Pablo Reguera
- Medical Oncology Department, Ramón y Cajal University Hospital, Ctra. Colmenar Viejo km 9100, 28034 Madrid, Spain
| | - Olga Martínez-Sáez
- Medical Oncology Department, Ramón y Cajal University Hospital, Ctra. Colmenar Viejo km 9100, 28034 Madrid, Spain
| | - Enrique Grande
- Medical Oncology Department, Ramón y Cajal University Hospital, Ctra. Colmenar Viejo km 9100, 28034 Madrid, Spain
| |
Collapse
|
22
|
Garrido P, Rodríguez CA. SEOM guidelines 2014: improving quality to increase its usefulness. Clin Transl Oncol 2014; 16:1023-4. [PMID: 25392081 PMCID: PMC4239804 DOI: 10.1007/s12094-014-1246-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Accepted: 10/07/2014] [Indexed: 11/28/2022]
Affiliation(s)
- P. Garrido
- Department of Medical Oncology, Spanish Society for Clinical Oncology, Hospital Universitario Ramon y Cajal, Madrid, Spain
| | - C. A. Rodríguez
- Department of Medical Oncology, Spanish Society for Clinical Oncology, Hospital Universitario de Salamanca-IBSAL, Salamanca, Spain
| |
Collapse
|