Search Results
You are looking at 1 - 6 of 6 items for
- Author: Emily Bergsland x
- Refine by access: All content x
Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, California, USA
Search for other papers by Claire K Mulvey in
Google Scholar
PubMed
Department of Epidemiology and Biostatistics, University of California, San Francisco, California, USA
Search for other papers by Alan Paciorek in
Google Scholar
PubMed
Search for other papers by Farhana Moon in
Google Scholar
PubMed
Search for other papers by Paige Steiding in
Google Scholar
PubMed
Search for other papers by Brandon Shih in
Google Scholar
PubMed
Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, California, USA
Search for other papers by Matthew A Gubens in
Google Scholar
PubMed
Department of Epidemiology and Biostatistics, University of California, San Francisco, California, USA
Search for other papers by Li Zhang in
Google Scholar
PubMed
Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, California, USA
Search for other papers by Emily K Bergsland in
Google Scholar
PubMed
Search for other papers by Iona Cheng in
Google Scholar
PubMed
Lung neuroendocrine tumors (NETs) have few known predictors of survival. We investigated associations of sociodemographic, clinicopathologic, and treatment factors with overall survival (OS) and lung cancer-specific survival (LCSS) for incident lung NET cases (typical or atypical histology) in the California Cancer Registry (CCR) from 1992 to 2019. OS was estimated with the Kaplan–Meier method and compared by sociodemographic and disease factors univariately with the log-rank test. We used sequential Cox proportional hazards regression for multivariable OS analysis. LCSS was estimated using Fine-Gray competing risks regression. There were 6038 lung NET diagnoses (5569 typical, 469 atypical carcinoid); most were women (70%) and non-Hispanic White (73%). In our multivariable model, sociodemographic factors were independently associated with OS, with better survival for women (hazard ratio (HR) 0.62, 95% confidence interval (CI) 0.57–0.68, P < 0.001), married (HR 0.76, 95% CI 0.70–0.84, P < 0.001), and residents of high socioeconomic status (SES) neighborhoods (HRQ5vsQ1 0.73, 95% CI 0.62–0.85, P < 0.001). Compared to cases with private insurance, OS was worse for cases with Medicare (HR 1.24, 95% CI 1.10–1.40, P < 0.001) or Medicaid/other public insurance (HR 1.45, 95% CI 1.24–1.68, P < 0.001). In our univariate model, non-Hispanic Black Californians had worse OS than other racial/ethnic groups, but differences attenuated after adjusting for stage at diagnosis. In our LCSS models, we found similar associations between sex and marital status on survival, but no differences in outcomes by SES or insurance. By race/ethnicity, American Indian cases had worse LCSS. In summary, beyond disease-related and treatment variables, sociodemographic factors were independently associated with survival in lung NETs.
Search for other papers by Matthew H Kulke in
Google Scholar
PubMed
Search for other papers by Thomas O'Dorisio in
Google Scholar
PubMed
Search for other papers by Alexandria Phan in
Google Scholar
PubMed
Search for other papers by Emily Bergsland in
Google Scholar
PubMed
Search for other papers by Linda Law in
Google Scholar
PubMed
Search for other papers by Phillip Banks in
Google Scholar
PubMed
Search for other papers by Joel Freiman in
Google Scholar
PubMed
Search for other papers by Kenny Frazier in
Google Scholar
PubMed
Search for other papers by Jessica Jackson in
Google Scholar
PubMed
Search for other papers by James C Yao in
Google Scholar
PubMed
Search for other papers by Larry Kvols in
Google Scholar
PubMed
Search for other papers by Pablo Lapuerta in
Google Scholar
PubMed
Search for other papers by Brian Zambrowicz in
Google Scholar
PubMed
Search for other papers by Douglas Fleming in
Google Scholar
PubMed
Search for other papers by Arthur Sands in
Google Scholar
PubMed
Serotonin produced by neuroendocrine tumors is believed to be a principal cause of the diarrhea in carcinoid syndrome. We assessed the safety and efficacy of telotristat etiprate, an oral serotonin synthesis inhibitor, in patients with diarrhea associated with carcinoid syndrome. In this prospective, randomized study, patients with evidence of carcinoid tumor and ≥4 bowel movements (BMs)/day despite stable-dose octreotide LAR depot therapy were enrolled in sequential, escalating, cohorts of four patients per cohort. In each cohort, one patient was randomly assigned to placebo and three patients to telotristat etiprate, at 150, 250, 350, or 500 mg three times a day (tid). In a subsequent cohort, one patient was assigned to placebo and six patients to telotristat etiprate 500 mg tid. Patients were assessed for safety, BM frequency (daily diary), 24 h urinary 5-hydroxyindoleacetic acid (u5-HIAA), and adequate relief of carcinoid gastrointestinal symptoms (using a weekly questionnaire). Twenty-three patients were treated: 18 received telotristat etiprate and five received placebo. Adverse events were generally mild. Among evaluable telotristat etiprate-treated patients, 5/18 (28%) experienced a ≥30% reduction in BM frequency for ≥2 weeks, 9/16 (56%) experienced biochemical response (≥50% reduction or normalization in 24-h u5-HIAA) at week 2 or 4, and 10/18 (56%) reported adequate relief during at least 1 of the first 4 weeks of treatment. Similar activity was not observed in placebo-treated patients. Telotristat etiprate was well tolerated. Our observations suggest that telotristat etiprate has activity in controlling diarrhea associated with carcinoid syndrome. Further studies confirming these findings are warranted.
UCSF Helen Diller Family Comprehensive Cancer Center, Department of Surgery, Hormone Research Institute, Department of Medicine, Department of Neurosciences, Diabetes Center
Search for other papers by Yu-cheng Wang in
Google Scholar
PubMed
UCSF Helen Diller Family Comprehensive Cancer Center, Department of Surgery, Hormone Research Institute, Department of Medicine, Department of Neurosciences, Diabetes Center
Search for other papers by Marlene B Zuraek in
Google Scholar
PubMed
Search for other papers by Yasuhiro Kosaka in
Google Scholar
PubMed
Search for other papers by Yasuharu Ota in
Google Scholar
PubMed
UCSF Helen Diller Family Comprehensive Cancer Center, Department of Surgery, Hormone Research Institute, Department of Medicine, Department of Neurosciences, Diabetes Center
Search for other papers by Michael S German in
Google Scholar
PubMed
Search for other papers by Evan S Deneris in
Google Scholar
PubMed
UCSF Helen Diller Family Comprehensive Cancer Center, Department of Surgery, Hormone Research Institute, Department of Medicine, Department of Neurosciences, Diabetes Center
Search for other papers by Emily K Bergsland in
Google Scholar
PubMed
UCSF Helen Diller Family Comprehensive Cancer Center, Department of Surgery, Hormone Research Institute, Department of Medicine, Department of Neurosciences, Diabetes Center
Search for other papers by David B Donner in
Google Scholar
PubMed
UCSF Helen Diller Family Comprehensive Cancer Center, Department of Surgery, Hormone Research Institute, Department of Medicine, Department of Neurosciences, Diabetes Center
Search for other papers by Robert S Warren in
Google Scholar
PubMed
UCSF Helen Diller Family Comprehensive Cancer Center, Department of Surgery, Hormone Research Institute, Department of Medicine, Department of Neurosciences, Diabetes Center
Search for other papers by Eric K Nakakura in
Google Scholar
PubMed
Neuroendocrine (NE) or carcinoid tumors of the small intestine (SI) frequently metastasize and produce the hormone serotonin, causing significant morbidity and mortality. A member of the ETS oncogene family of transcription factors, Fev, acts with the homeodomain transcription factor Nkx2.2 in the development of serotonin neurons in mice. In this study, we investigated the role of Fev in normal and neoplastic SI. In NE tumors (NETs) of the SI, serotonin stimulates tumor growth and causes debilitating symptoms, such as diarrhea, flushing, wheezing, and right-sided valvular heart disease (i.e. carcinoid syndrome). Compared with those in the matched normal human SI, FEV expression levels were significantly elevated in primary NETs (20-fold, P<0.0001), lymph node metastases (35-fold, P=0.004), and NET liver metastases (22-fold, P<0.0001) resected from patients with serotonin excess. Fev is expressed in the wild type but not in Nkx2.2 (−/−) mouse SI, in which cells producing serotonin are absent. Using recombination-based cell lineage tracing, we found that FEV-positive cells give rise to serotonin-producing cells in the SI. In Fev (−/−) mouse SI, we observed no difference in the number of cells producing serotonin or other hormones. We conclude that FEV expression identifies serotonin-producing cells in normal and neoplastic SI and is a novel target for diagnosis of patients with NETs of the SI.
UCSF Helen Diller Family Comprehensive Cancer Center, Surgery, Pathology, Medicine, Hormone Research Institute, Departments of
Search for other papers by Yu-Cheng Wang in
Google Scholar
PubMed
UCSF Helen Diller Family Comprehensive Cancer Center, Surgery, Pathology, Medicine, Hormone Research Institute, Departments of
Search for other papers by Emerick Gallego-Arteche in
Google Scholar
PubMed
Search for other papers by Gioia Iezza in
Google Scholar
PubMed
UCSF Helen Diller Family Comprehensive Cancer Center, Surgery, Pathology, Medicine, Hormone Research Institute, Departments of
Search for other papers by Xiaochen Yuan in
Google Scholar
PubMed
Search for other papers by Mary R Matli in
Google Scholar
PubMed
UCSF Helen Diller Family Comprehensive Cancer Center, Surgery, Pathology, Medicine, Hormone Research Institute, Departments of
Search for other papers by Su-Pin Choo in
Google Scholar
PubMed
UCSF Helen Diller Family Comprehensive Cancer Center, Surgery, Pathology, Medicine, Hormone Research Institute, Departments of
Search for other papers by Marlene B Zuraek in
Google Scholar
PubMed
UCSF Helen Diller Family Comprehensive Cancer Center, Surgery, Pathology, Medicine, Hormone Research Institute, Departments of
Search for other papers by Ravi Gogia in
Google Scholar
PubMed
Search for other papers by Francis C Lynn in
Google Scholar
PubMed
UCSF Helen Diller Family Comprehensive Cancer Center, Surgery, Pathology, Medicine, Hormone Research Institute, Departments of
Search for other papers by Michael S German in
Google Scholar
PubMed
UCSF Helen Diller Family Comprehensive Cancer Center, Surgery, Pathology, Medicine, Hormone Research Institute, Departments of
Search for other papers by Emily K Bergsland in
Google Scholar
PubMed
UCSF Helen Diller Family Comprehensive Cancer Center, Surgery, Pathology, Medicine, Hormone Research Institute, Departments of
Search for other papers by David B Donner in
Google Scholar
PubMed
UCSF Helen Diller Family Comprehensive Cancer Center, Surgery, Pathology, Medicine, Hormone Research Institute, Departments of
Search for other papers by Robert S Warren in
Google Scholar
PubMed
UCSF Helen Diller Family Comprehensive Cancer Center, Surgery, Pathology, Medicine, Hormone Research Institute, Departments of
Search for other papers by Eric K Nakakura in
Google Scholar
PubMed
The homeodomain transcription factor NKX2.2 is necessary for neuroendocrine (NE) differentiation in the central nervous system and pancreas. NE tumors derived from the gut are defined by their NE phenotype, which is used for diagnosis and contributes to tumorigenicity. We hypothesized that NKX2.2 is important for NE differentiation in normal and neoplastic gut. NKX2.2 and NE marker expression was investigated in the small intestine of embryonic and adult mice using immunofluorescence (IF). To determine the role of NKX2.2 in NE differentiation of the intestine, the phenotype of Nkx2.2 (−/−) mice was examined by IF and real-time (RT)-PCR. NKX2.2 and NE marker expression in human NE tumors of the gut and normal tissues were evaluated by immunohistochemistry and qRT-PCR. NKX2.2 expression was detected in the intervillus/crypt regions of embryonic and adult mouse intestine. Co-expression of Nkx2.2 with neurogenin3 (NEUROG3) and hormones was observed in the adult intestinal crypt compartment, suggesting NKX2.2 functions in NEUROG3-positive endocrine progenitors and newly differentiated endocrine cells. In the intestine of Nkx2.2 (−/−) mice, we found a dramatic reduction in the number of cells producing numerous hormones, such as serotonin, gastrin, cholecystokinin, somatostatin, glucagon-like peptide 1 (GLP-1), and secretin, but an increase in cells producing ghrelin. NKX2.2 was expressed in most (24 of 29) human NE tumors derived from diverse primary sites. We conclude NKX2.2 functions in immature endocrine cells to control NE differentiation in normal intestine and is expressed in most NE tumors of the gut, and is therefore a novel target of diagnosis for patients with gastrointestinal NE tumors.
Search for other papers by Jennifer R Eads in
Google Scholar
PubMed
Search for other papers by Thorvardur R Halfdanarson in
Google Scholar
PubMed
Search for other papers by Tim Asmis in
Google Scholar
PubMed
Search for other papers by Andrew M Bellizzi in
Google Scholar
PubMed
Search for other papers by Emily K Bergsland in
Google Scholar
PubMed
Search for other papers by Arvind Dasari in
Google Scholar
PubMed
Search for other papers by Ghassan El-Haddad in
Google Scholar
PubMed
Search for other papers by Michael Frumovitz in
Google Scholar
PubMed
Search for other papers by Joshua Meyer in
Google Scholar
PubMed
Search for other papers by Erik Mittra in
Google Scholar
PubMed
Search for other papers by Sten Myrehaug in
Google Scholar
PubMed
Search for other papers by Eric Nakakura in
Google Scholar
PubMed
Search for other papers by Nitya Raj in
Google Scholar
PubMed
Search for other papers by Heloisa P Soares in
Google Scholar
PubMed
Search for other papers by Brian Untch in
Google Scholar
PubMed
Search for other papers by Namrata Vijayvergia in
Google Scholar
PubMed
Search for other papers by Jennifer A Chan in
Google Scholar
PubMed
High-grade neuroendocrine neoplasms are a rare disease entity and account for approximately 10% of all neuroendocrine neoplasms. Because of their rarity, there is an overall lack of prospectively collected data available to advise practitioners as to how best to manage these patients. As a result, best practices are largely based on expert opinion. Recently, a distinction was made between well-differentiated high-grade (G3) neuroendocrine tumors and poorly differentiated neuroendocrine carcinomas, and with this, pathologic details, appropriate imaging practices and treatment have become more complex. In an effort to provide practitioners with the best guidance for the management of patients with high-grade neuroendocrine neoplasms of the gastrointestinal tract, pancreas, and gynecologic system, the North American Neuroendocrine Tumor Society convened a panel of experts to develop a set of recommendations and a treatment algorithm that may be used by practitioners for the care of these patients. Here, we provide consensus recommendations from the panel on pathology, imaging practices, management of localized disease, management of metastatic disease and surveillance and draw key distinctions as to the approach that should be utilized in patients with well-differentiated G3 neuroendocrine tumors vs poorly differentiated neuroendocrine carcinomas.
Search for other papers by James C Yao in
Google Scholar
PubMed
Search for other papers by Jonathan Strosberg in
Google Scholar
PubMed
Search for other papers by Nicola Fazio in
Google Scholar
PubMed
Search for other papers by Marianne E Pavel in
Google Scholar
PubMed
Search for other papers by Emily Bergsland in
Google Scholar
PubMed
Search for other papers by Philippe Ruszniewski in
Google Scholar
PubMed
Search for other papers by Daniel M Halperin in
Google Scholar
PubMed
Search for other papers by Daneng Li in
Google Scholar
PubMed
Search for other papers by Salvatore Tafuto in
Google Scholar
PubMed
Search for other papers by Nitya Raj in
Google Scholar
PubMed
Search for other papers by Davide Campana in
Google Scholar
PubMed
Search for other papers by Susumu Hijioka in
Google Scholar
PubMed
Search for other papers by Markus Raderer in
Google Scholar
PubMed
Search for other papers by Rosine Guimbaud in
Google Scholar
PubMed
Search for other papers by Pablo Gajate in
Google Scholar
PubMed
Search for other papers by Sara Pusceddu in
Google Scholar
PubMed
Search for other papers by Albert Reising in
Google Scholar
PubMed
Search for other papers by Evgeny Degtyarev in
Google Scholar
PubMed
Search for other papers by Mark Shilkrut in
Google Scholar
PubMed
Search for other papers by Simantini Eddy in
Google Scholar
PubMed
Search for other papers by Simron Singh in
Google Scholar
PubMed
Spartalizumab, a humanized anti-programmed death protein 1 (PD-1) MAB, was evaluated in patients with well-differentiated metastatic grade 1/2 neuroendocrine tumors (NET) and poorly differentiated gastroenteropancreatic neuroendocrine carcinomas (GEP-NEC). In this phase II, multicenter, single-arm study, patients received spartalizumab 400 mg every 4 weeks until confirmed disease progression or unacceptable toxicity. The primary endpoint was confirmed overall response rate (ORR) according to blinded independent review committee using response evaluation criteria in solid tumors 1.1. The study enrolled 95 patients in the NET group (30, 32 and 33 in the thoracic, gastrointestinal, and pancreatic cohorts, respectively), and 21 patients in the GEP-NEC group. The ORR was 7.4% (95% CI: 3.0, 14.6) in the NET group (thoracic, 16.7%; gastrointestinal, 3.1%; pancreatic, 3.0%), which was below the predefined success criterion of ≥10%, and 4.8% (95% CI: 0.1, 23.8) in the GEP-NEC group. In the NET and GEP-NEC groups, the 12-month progression-free survival was 19.5 and 0%, respectively, and the 12-month overall survival was 73.5 and 19.1%, respectively. The ORR was higher in patients with ≥1% PD-L1 expression in immune/tumor cells or ≥1% CD8+ cells at baseline. The most common adverse events considered as spartalizumab-related included fatigue (29.5%) and nausea (10.5%) in the NET group, and increased aspartate and alanine aminotransferases (each 14.3%) in the GEP-NEC group. The efficacy of spartalizumab was limited in this heterogeneous and heavily pre-treated population; however, the results in the thoracic cohort are encouraging and warrants further investigation. Adverse events were manageable and consistent with previous experience.