Browse

You are looking at 21 - 30 of 2,623 items for

  • Refine by access: All content x
Clear All
Dominique D. Liddy D Liddy, Surgery, University of Florida, Gainesville, United States

Search for other papers by Dominique D. Liddy in
Google Scholar
PubMed
Close
,
Zhongyue Zhang Z Zhang, Biostatistics, University of Florida, Gainesville, United States

Search for other papers by Zhongyue Zhang in
Google Scholar
PubMed
Close
,
Kalyanee Shirlekar K Shirlekar, Biostatistics, University of Florida, Gainesville, United States

Search for other papers by Kalyanee Shirlekar in
Google Scholar
PubMed
Close
,
Zhongping He Z He, Biostatistics, University of Florida, Gainesville, United States

Search for other papers by Zhongping He in
Google Scholar
PubMed
Close
,
Kelly M Herremans K Herremans, Surgery, University of Florida, Gainesville, United States

Search for other papers by Kelly M Herremans in
Google Scholar
PubMed
Close
,
Song Han S Han, Surgery, University of Florida, Gainesville, United States

Search for other papers by Song Han in
Google Scholar
PubMed
Close
,
Jason O Brant J Brant, Biostatistics, University of Florida, Gainesville, United States

Search for other papers by Jason O Brant in
Google Scholar
PubMed
Close
,
Francis D Moore F Moore, Surgery, University of Florida, Gainesville, United States

Search for other papers by Francis D Moore in
Google Scholar
PubMed
Close
,
Steven J Hughes S Hughes, Surgery, University of Florida, Gainesville, United States

Search for other papers by Steven J Hughes in
Google Scholar
PubMed
Close
, and
Aditya S. Shirali A Shirali, Surgery, University of Florida, Gainesville, United States

Search for other papers by Aditya S. Shirali in
Google Scholar
PubMed
Close

Differentiated thyroid cancer in older adults has been linked to alterations in the mutational landscape and tumor immune cell infiltration that create a tumor-permissive microenvironment. We sought to determine the impact of age on genomic alterations and immune cell composition in papillary thyroid cancer (PTC). Genomic alterations, immune cell composition and clinical data were obtained using The Cancer Genome Atlas (TCGA) and computational immunogenomic analyses. Disease severity was recoded into 3 groups: Group A (T1-2N0M0), Group B (T1-3N1a-1bM0), and Group C (T4NxMx or TxNxM1). Histopathologic subtypes included conventional, follicular-variant, and tall cell variant PTC. Spearman’s rank correlation, ANOVA, t-test, and multivariable linear regression were performed. 470 PTC samples were retrieved from the TCGA portal with genomic alteration and immune cell composition data. TERT promoter alterations were more common in patients ≥ 65 years (26% vs 4%, p<0.0001). Tumor mutational burden increased with increasing age (r=0.463, p<0.0001). Increasing age was associated with decreased CD8+ T cells (r=-0.15, p=0.01) using CIBERSORT and decreased B cells (r=-0.13), CD8+ T cells (r=-0.19) and neutrophils (r=-0.14, p<0.05) using TIMER. Multivariate regression found that increasing age was independently associated with increased resting NK cells and resting dendritic cells, and decreased naïve B cells and CD8+ T cells (p<0.05). PTC tumors of older adults are characterized by increased TERT promoter alterations, increased tumor mutational burden, and a decreased cytotoxic CD8+ T and increased resting dendritic cell immune infiltrate. Further studies are needed to determine if these changes in immune cell infiltrate are associated with compromised outcomes.

Restricted access
Paul Benjamin Loughrey Patrick G Johnston Centre for Cancer Research, Queen’s University Belfast, Belfast, UK
Regional Centre for Endocrinology and Diabetes, Belfast Health and Social Care Trust, Belfast, UK

Search for other papers by Paul Benjamin Loughrey in
Google Scholar
PubMed
Close
,
Brian Herron Department of Cellular Pathology, Belfast Health and Social Care Trust, Belfast, UK

Search for other papers by Brian Herron in
Google Scholar
PubMed
Close
,
Stephen Cooke Department of Neurosurgery, Belfast Health and Social Care Trust, Belfast, UK

Search for other papers by Stephen Cooke in
Google Scholar
PubMed
Close
,
Philip Weir Department of Neurosurgery, Belfast Health and Social Care Trust, Belfast, UK

Search for other papers by Philip Weir in
Google Scholar
PubMed
Close
,
Jayna Elizabeth Smyth Regional Centre for Endocrinology and Diabetes, Belfast Health and Social Care Trust, Belfast, UK

Search for other papers by Jayna Elizabeth Smyth in
Google Scholar
PubMed
Close
,
Karen R Mullan Regional Centre for Endocrinology and Diabetes, Belfast Health and Social Care Trust, Belfast, UK

Search for other papers by Karen R Mullan in
Google Scholar
PubMed
Close
,
Estelle G Healy Department of Cellular Pathology, Belfast Health and Social Care Trust, Belfast, UK

Search for other papers by Estelle G Healy in
Google Scholar
PubMed
Close
,
Jane Evanson Department of Radiology, Barts Health NHS Trust, London, UK

Search for other papers by Jane Evanson in
Google Scholar
PubMed
Close
,
Stephanie G Craig Patrick G Johnston Centre for Cancer Research, Queen’s University Belfast, Belfast, UK

Search for other papers by Stephanie G Craig in
Google Scholar
PubMed
Close
,
Jacqueline A James Patrick G Johnston Centre for Cancer Research, Queen’s University Belfast, Belfast, UK
Department of Cellular Pathology, Belfast Health and Social Care Trust, Belfast, UK
Northern Ireland Biobank, Belfast Health and Social Care Trust, Belfast, UK

Search for other papers by Jacqueline A James in
Google Scholar
PubMed
Close
,
Márta Korbonits Department of Endocrinology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK

Search for other papers by Márta Korbonits in
Google Scholar
PubMed
Close
, and
Steven J Hunter Regional Centre for Endocrinology and Diabetes, Belfast Health and Social Care Trust, Belfast, UK

Search for other papers by Steven J Hunter in
Google Scholar
PubMed
Close

Cushing’s disease is a rare condition that occurs due to an adrenocorticotrophin-producing corticotrophinoma arising from the pituitary gland. The consequent hypercortisolaemia results in multisystem morbidity and mortality. This study aims to report incidence, clinicopathological characteristics, remission outcomes and mortality in a regional pituitary neurosurgical cohort of patients diagnosed with Cushing’s disease in Northern Ireland (NI) from 2000 to 2019. Clinical, biochemical and radiological data from a cohort of patients operated for Cushing’s disease were retrospectively collected and analysed. Fifty-three patients were identified, resulting in an estimated annual incidence of Cushing’s disease of 1.39–1.57 per million population per year. Females accounted for 72% (38/53) of the cohort. The majority (74%, 39/53) of corticotrophinomas were microadenomas and in 44% (17/39) of these no tumour was identified on preoperative magnetic resonance imaging. Histopathological characterisation was similarly difficult, with no tumour being identified in the histopathological specimen in 40% (21/53) of cases. Immediate postoperative remission rates were 53% and 66% when considering serum morning cortisol cut-offs of ≤ 50 nmol/L (1.8 µg/dL) and ≤ 138 nmol/L (5 µg/dL), respectively, in the week following pituitary surgery. Approximately 70% (37/53) of patients achieved longer-term remission with a single pituitary surgery. Three patients had recurrent disease. Patients with Cushing’s disease had a significantly higher mortality rate compared to the NI general population (standardised mortality ratio 8.10, 95% CI 3.3–16.7, P < 0.001). Annual incidence of Cushing’s disease in NI is consistent with other Northern European cohorts. Functioning corticotrophinomas are a clinically, radiologically and histopathologically elusive disease with increased mortality compared to the general population.

Open access
E Baudin Endocrine Oncology Unit, Imaging Department, Gustave Roussy, Villejuif, France

Search for other papers by E Baudin in
Google Scholar
PubMed
Close
,
J Capdevila Medical Oncology Department, Vall d’Hebron University Hospital, Vall d’Hebron Institute of Oncology (VHIO), IOB Quirón-Teknon, Barcelona, Spain

Search for other papers by J Capdevila in
Google Scholar
PubMed
Close
,
D Hörsch ENETS Center of Excellence, Zentralklinik Bad Berka GmbH, Bad Berka, Germany

Search for other papers by D Hörsch in
Google Scholar
PubMed
Close
,
S Singh Division of Medical Oncology, University of Toronto, Sunnybrook Odette Cancer Center, Sunnybrook HSC, Toronto, Ontario, Canada

Search for other papers by S Singh in
Google Scholar
PubMed
Close
,
M E Caplin Neuroendocrine Tumour Unit, Royal Free Hospital School of Medicine, London, UK

Search for other papers by M E Caplin in
Google Scholar
PubMed
Close
,
E M Wolin Division of Hematology and Oncology, Icahn School of Medicine at Mount Sinai, New York, New York, USA

Search for other papers by E M Wolin in
Google Scholar
PubMed
Close
,
W Buikhuisen Department of Thorax Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands

Search for other papers by W Buikhuisen in
Google Scholar
PubMed
Close
,
M Raderer Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria

Search for other papers by M Raderer in
Google Scholar
PubMed
Close
,
E Dansin Thoracic Oncology Unit, Centre Oscar Lambret, Lille, France

Search for other papers by E Dansin in
Google Scholar
PubMed
Close
,
C Grohe Department of Respiratory Diseases, Evangelische Lungenklinik, Berlin, Germany

Search for other papers by C Grohe in
Google Scholar
PubMed
Close
,
D Ferone Neuroendocrine Tumour Unit, Department of Internal Medicine and Medical Specialties, University of Genova, Genova, Italy

Search for other papers by D Ferone in
Google Scholar
PubMed
Close
,
A Houchard Data and Insights Generation and Strategy, Ipsen, Boulogne-Billancourt, France

Search for other papers by A Houchard in
Google Scholar
PubMed
Close
,
X-M Truong-Thanh Medical Affairs, Ipsen, Boulogne-Billancourt, France

Search for other papers by X-M Truong-Thanh in
Google Scholar
PubMed
Close
,
D Reidy-Lagunes Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical Center, New York, New York, USA

Search for other papers by D Reidy-Lagunes in
Google Scholar
PubMed
Close
, and
the SPINET Study Group
Search for other papers by the SPINET Study Group in
Google Scholar
PubMed
Close
the SPINET Study Group

Prospective data are lacking on early somatostatin analog (SSA) therapy in bronchopulmonary neuroendocrine tumors (BP-NETs; typical carcinoids and atypical carcinoids (TCs and ACs)). SPINET (EudraCT: 2015-004992-62; NCT02683941) was a phase III, double-blind study of lanreotide autogel/depot (LAN; 120 mg every 28 days) plus best supportive care (BSC) vs placebo plus BSC, with an optional open-label treatment phase (LAN plus BSC). Patients had metastatic/unresectable, somatostatin receptor (SSTR)-positive TCs or ACs. Recruitment was stopped early owing to slow accrual; eligible patients from the double-blind phase transitioned to open-label LAN. The adapted primary endpoint was progression-free survival (PFS) during either phase for patients receiving LAN. Seventy-seven patients were randomized (LAN, n = 51 (TCs, n = 29; ACs, n = 22); placebo, n = 26 (TCs, n = 16; ACs, n = 10)). Median (95% CI) PFS during double-blind and open-label phases in patients receiving LAN was 16.6 (11.3; 21.9) months overall (primary endpoint), 21.9 (12.8, not calculable (NC)) months in TCs, and 13.8 (5.4; 16.6) months in ACs. During double-blind treatment, median (95% CI) PFS was 16.6 (11.3; 21.9) months for LAN vs 13.6 (8.3; NC) months for placebo (not significant); corresponding values were 21.9 (13.8; NC) and 13.9 (13.4; NC) months, respectively, in TCs and 13.8 (5.4; 16.6) and 11.0 (2.8; 16.9) months, respectively, in ACs. Patients’ quality of life did not deteriorate and LAN was well tolerated. Although recruitment stopped early and the predefined sample size was not met, SPINET is the largest prospective study to date of SSA therapy in SSTR-positive TCs and ACs and suggests clinical benefit in TCs.

Open access
Judith Favier Université Paris Cité, Inserm UMR970 PARCC, Equipe Labellisée par la Ligue contre le cancer, Paris, France

Search for other papers by Judith Favier in
Google Scholar
PubMed
Close
,
Karel Pacak Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA

Search for other papers by Karel Pacak in
Google Scholar
PubMed
Close
, and
Roderick Clifton-Bligh Department of Endocrinology Royal North Shore Hospital, University of Sydney, Sydney, Australia

Search for other papers by Roderick Clifton-Bligh in
Google Scholar
PubMed
Close
Restricted access
Giulia Vocino Trucco Pathology Unit, SS. Annunziata Hospital, Savigliano, Italy

Search for other papers by Giulia Vocino Trucco in
Google Scholar
PubMed
Close
and
Marco Volante Department of Oncology, University of Turin, Orbassano, Turin, Italy

Search for other papers by Marco Volante in
Google Scholar
PubMed
Close

Graphical abstract

Abstract

The 5th edition of the World Health Organization (WHO) classification of neuroendocrine neoplasms (NENs) is built to achieve a uniform terminology and to define a similar diagnostic scheme across different anatomic locations. Since the 4th edition, a chapter discussing NENs in nonneuroendocrine organs has been introduced, which proposes a binary system for classification segregating well-differentiated neoplasms, termed neuroendocrine tumors (NETs), and poorly differentiated neoplasms, termed neuroendocrine carcinomas (NECs). A grading system for NETs is based on mitotic index and/or Ki-67 index and/or necrosis, depending on the different locations. Although this approach has been already well established in the digestive system, it modifies and homogenizes the classification of NENs in the urinary tract, in female genital organs, and in the male genital system. In the lung and thymus, the double terminology of carcinoid/NET, already introduced in the 5th edition of the WHO classification of thoracic tumors, is endorsed. This approach undoubtedly helps the multidisciplinary approach for the diagnosis and clinical management of patients affected by these neoplasms, without losing site-specific characteristics that influence the clinical and biological behavior of tumors in different anatomical sites. Other major advances of the new WHO scheme are the homogenization of epidemiological data and the correct integration of data from prospective future studies aimed at the definition of molecular profiles and at the identification of tumor type-specific and patient-specific therapeutic approaches.

Restricted access
Ali S Alzahrani Department of Medicine, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
Department of Molecular Oncology, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia

Search for other papers by Ali S Alzahrani in
Google Scholar
PubMed
Close
,
Meshael Alswailem Department of Molecular Oncology, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia

Search for other papers by Meshael Alswailem in
Google Scholar
PubMed
Close
,
Alexandre Buffet Université Paris Cité, Inserm, Paris Centre de Recherche Cardiovasculaire (PARCC), Equipe Labellisée Ligue contre le Cancer, Paris, France
Département de Médecine Génomique des Tumeurs et des Cancers, Fédération de Génétique et de Médecine Génomique, Assistance Publique-Hôpitaux de Paris (AP-HP) Centre, Hôpital Européen Georges Pompidou, Paris, France

Search for other papers by Alexandre Buffet in
Google Scholar
PubMed
Close
,
Balgees Alghamdi Department of Molecular Oncology, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia

Search for other papers by Balgees Alghamdi in
Google Scholar
PubMed
Close
,
Lulu Alobaid Department of Medicine, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia

Search for other papers by Lulu Alobaid in
Google Scholar
PubMed
Close
,
Osamah Alsagheir Department of Medicine, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia

Search for other papers by Osamah Alsagheir in
Google Scholar
PubMed
Close
,
Hindi Al-Hindi Department of Pathology and Laboratory Medicine, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia

Search for other papers by Hindi Al-Hindi in
Google Scholar
PubMed
Close
, and
Karel Pacak Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA

Search for other papers by Karel Pacak in
Google Scholar
PubMed
Close

In 2012, somatic EPAS1 pathogenic variants were found to cause a triad of pheochromocytoma/paragangliomas (PPGLs), polycythemia, and somatostatinoma. Since then, a limited number of studies on this subject have been reported, and data on the long-term outcome of metastatic disease are not available on this rare syndrome. We comprehensively reviewed EPAS1-related PPGL and describe an unusual patient who has been living with an EPAS1-related metastatic PPGL for 47 years. The results of this work show that EPAS1 pathogenic variants are rare, more in females and patients without pathogenic variants in other PPGL susceptibility genes. PPGLs are the most common manifestation followed by polycythemia and somatostatinoma. The EPAS1 pathogenic variants are often postzygotic, and the timing of their acquirement during embryonic development seems to correlate with the number and timing of development of the disease manifestations. Although recurrent and multifocal disease is common in EPAS1-related PPGL, distant metastases are uncommon and usually indolent. This is illustrated by a case of a man who was diagnosed at the age of 9 years and is currently 56 years old, alive, and well for 47 years with these metastases. He was found to have a somatic EPAS1 pathogenic variant (c.1592C>A, p.Pro531His) in bilateral pheochomocytoma and a pancreatic NET (somatostatinoma) but not in genomic DNA isolated from peripheral leukocytes. This and previous reports suggest that distant metastases are uncommon and less aggressive in EPAS1-related PPGLs compared to those found in other hereditary PPGLs.

Restricted access
Mijin Kim Department of Internal Medicine, Pusan National University School of Medicine, Yangsan, Korea
Biomedical Research Institute, Pusan National University Hospital, Busan, Korea

Search for other papers by Mijin Kim in
Google Scholar
PubMed
Close
,
Chae Hwa Kwon Biomedical Research Institute, Pusan National University Hospital, Busan, Korea

Search for other papers by Chae Hwa Kwon in
Google Scholar
PubMed
Close
, and
Bo Hyun Kim Department of Internal Medicine, Pusan National University School of Medicine, Yangsan, Korea
Biomedical Research Institute, Pusan National University Hospital, Busan, Korea

Search for other papers by Bo Hyun Kim in
Google Scholar
PubMed
Close

The genetic alterations currently identified in papillary thyroid microcarcinomas (PTMCs) are insufficient for distinguishing tumors with aggressive features. We aimed to identify candidate markers associated with lateral lymph node metastasis (LLNM, N1sb disease) in patients with PTMC using transcriptomic analysis. RNA sequencing was performed on 26 matched tumor and normal thyroid tissue samples (N0, n = 14; N1b, n = 12), followed by functional enrichment analyses of differentially expressed genes (DEGs). EcoTyper was used to explore the distinct tumor microenvironment (TME). We identified 631 DEGs (213 upregulated and 418 downregulated) between N1b and N0 PTMCs. The most significantly upregulated genes in N1b were associated with tumorigenesis, adhesion, migration, and invasion. DEGs were mainly enriched in the pathways of idiopathic pulmonary fibrosis, TME, wound healing, and inhibition of matrix metalloproteases. We predicted the activation of these pathways in N1b PTMCs. N1b PTMCs had a unique TME with abundant fibroblasts and epithelial cells, associated with an increased risk of disease progression. Fibroblast marker genes, including POSTN, MMP11, TNFAIP6,and FN1, and epithelial cell marker genes, including NOX4, MFAP2, TGFVBI,and TNC, were selected. POSTN and FN1, fibroblast cell-specific genes, and NOX4 and TNC, epithelial cell-specific genes, were promising biomarkers for predicting LLNM development and recurrence in patients with PTMC. We delineated the cellular ecotypes within the TME of patients with N1b PTMC and revealed potential markers for predicting LLNM and the prognosis of PTMC. These findings provide valuable insights into the contributions of cancer-associated fibroblasts and epithelial cells to PTMC progression and metastasis.

Restricted access
Marcia S Brose Department of Medical Oncology, Sidney Kimmel Cancer Center at Thomas Jefferson University, Philadelphia, Pennsylvania, USA

Search for other papers by Marcia S Brose in
Google Scholar
PubMed
Close
,
Jaume Capdevila Gastrointestinal and Endocrine Tumor Unit, Medical Oncology Department, Vall d’Hebron University Hospital, Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain

Search for other papers by Jaume Capdevila in
Google Scholar
PubMed
Close
,
Rossella Elisei Unit of Endocrinology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy

Search for other papers by Rossella Elisei in
Google Scholar
PubMed
Close
,
Lars Bastholt Department of Clinical Oncology, Odense University Hospital, Odense, Denmark

Search for other papers by Lars Bastholt in
Google Scholar
PubMed
Close
,
Dagmar Führer-Sakel Department of Endocrinology, Diabetes and Metabolism and Clinical Chemistry, University Hospital Essen, Essen, Germany

Search for other papers by Dagmar Führer-Sakel in
Google Scholar
PubMed
Close
,
Sophie Leboulleux Department of Nuclear Medicine and Endocrine Oncology, Gustave Roussy and Université Paris Saclay, Villejuif, France
Department of Endocrinology, Diabetology, Nutrition and Therapeutic Education, Hôpitaux Universitaires de Genève, Geneve, Switzerland

Search for other papers by Sophie Leboulleux in
Google Scholar
PubMed
Close
,
Iwao Sugitani Department of Endocrine Surgery, Nippon Medical School Graduate School of Medicine, Tokyo, Japan

Search for other papers by Iwao Sugitani in
Google Scholar
PubMed
Close
,
Matthew H Taylor Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, Oregon, USA

Search for other papers by Matthew H Taylor in
Google Scholar
PubMed
Close
,
Zhuoying Wang Department of Head Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
Department of Head Neck Surgery, Renji Hospital Affiliated to Jiaotong University School of Medicine, Shanghai, China

Search for other papers by Zhuoying Wang in
Google Scholar
PubMed
Close
,
Lori J Wirth Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA

Search for other papers by Lori J Wirth in
Google Scholar
PubMed
Close
,
Francis P Worden Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan, USA

Search for other papers by Francis P Worden in
Google Scholar
PubMed
Close
,
John Bernard Sanofi, Cambridge, Massachusetts, USA

Search for other papers by John Bernard in
Google Scholar
PubMed
Close
,
Paolo Caferra Sanofi, Amsterdam, The Netherlands
Department of Pharmacy, University of Pisa, Pisa, Italy

Search for other papers by Paolo Caferra in
Google Scholar
PubMed
Close
,
Raffaella M Colzani Sanofi, Cambridge, Massachusetts, USA

Search for other papers by Raffaella M Colzani in
Google Scholar
PubMed
Close
,
Shiguang Liu Sanofi, Cambridge, Massachusetts, USA

Search for other papers by Shiguang Liu in
Google Scholar
PubMed
Close
, and
Martin Schlumberger Department of Nuclear Medicine and Endocrine Oncology, Gustave Roussy and Université Paris Saclay, Villejuif, France

Search for other papers by Martin Schlumberger in
Google Scholar
PubMed
Close

The VERIFY study aimed to determine the efficacy of vandetanib in patients with differentiated thyroid cancer (DTC) that is either locally advanced or metastatic and refractory to radioiodine (RAI) therapy. Specifically, VERIFY is a randomized, double-blind, multicenter phase III trial aimed to determine the efficacy and safety of vandetanib in tyrosine kinase inhibitor-naive patients with locally advanced or metastatic RAI-refractory DTC with documented progression (NCT01876784). Patients were randomized 1:1 to vandetanib or placebo. The primary endpoint was progression-free survival (PFS). Secondary endpoints included best objective response rate, overall survival (OS), safety, and tolerability. Patients continued to receive randomized treatment until disease progression or for as long as they were receiving clinical benefit unless criteria for treatment discontinuation were met. Following randomization, 117 patients received vandetanib, and 118 patients received a placebo. Median PFS was 10.0 months in the vandetanib group and 5.7 months in the placebo group (hazard ratio: 0.75; 95% CI: 0.55–1.03; P = 0.080). OS was not significantly different between treatment arms. Common Terminology Criteria for Adverse Events (CTCAE) of grade ≥3 were reported in 55.6% of patients in the vandetanib arm and 25.4% in the placebo arm. Thirty-three deaths (28.2%; one related to study treatment) occurred in the vandetanib arm compared with 16 deaths (13.6%; two related to treatment) in the placebo arm. No statistically significant improvement was observed in PFS in treatment versus placebo in patients with locally advanced or metastatic, RAI-refractory DTC. Moreover, active treatment was associated with more adverse events and more deaths than placebo, though the difference in OS was not statistically significant.

Open access
Andreas Machens Medical Faculty, Department of Visceral, Vascular and Endocrine Surgery, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany

Search for other papers by Andreas Machens in
Google Scholar
PubMed
Close
,
Kerstin Lorenz Medical Faculty, Department of Visceral, Vascular and Endocrine Surgery, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany

Search for other papers by Kerstin Lorenz in
Google Scholar
PubMed
Close
,
Frank Weber Department of General, Visceral and Transplantation Surgery, Division of Endocrine Surgery, University of Duisburg-Essen, Essen, Germany

Search for other papers by Frank Weber in
Google Scholar
PubMed
Close
,
Tim Brandenburg Department of Endocrinology, Diabetology and Metabolism, University of Duisburg-Essen, Essen, Germany

Search for other papers by Tim Brandenburg in
Google Scholar
PubMed
Close
,
Dagmar Führer-Sakel Department of Endocrinology, Diabetology and Metabolism, University of Duisburg-Essen, Essen, Germany

Search for other papers by Dagmar Führer-Sakel in
Google Scholar
PubMed
Close
, and
Henning Dralle Medical Faculty, Department of Visceral, Vascular and Endocrine Surgery, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
Department of General, Visceral and Transplantation Surgery, Division of Endocrine Surgery, University of Duisburg-Essen, Essen, Germany

Search for other papers by Henning Dralle in
Google Scholar
PubMed
Close

The age-specific development of the three constituent components of multiple endocrine neoplasia type 2 (MEN 2) is incompletely characterized for many of the >30 causative rearranged during transfection (RET) mutations, which this genetic association study aimed to specify. Included in the study were 683 carriers of heterogeneous RET germline mutations: 53 carriers with 1 highest-risk mutation (codon 918); 240 carriers with 8 different high-risk mutations (codon 634); 176 carriers with 16 different intermediate-risk mutations (codon 609, 611, 618, 620, or 630); and 214 carriers with 6 different low-risk mutations (codon 768, 790, 804, or 891).There was a strong genotype-specific development of MEN 2 constituent components, with distinct age gradients from C cell disease to node negative medullary thyroid cancer (MTC), from node negative to node positive MTC, from node positive MTC to pheochromocytoma, and from pheochromocytoma to primary hyperparathyroidism. Primary hyperparathyroidism was not observed among the 53 MEN 2B patients who carried highest-risk mutations (age range: 0.5–50 years), of whom no more than 12 (23%) and 3 (6%) carriers were older than age 30 years and 35 years, respectively. The age-specific development of MTC differed significantly between the four RET risk categories, whereas the age-specific development of pheochromocytoma differed significantly only between the two strongest RET risk categories. No significant differences were noted in the development of primary hyperparathyroidism. These findings delineate age-specific disease manifestation corridors for the three constituent components of MEN 2 by RET genotype. These corridors are useful for initial risk assessment and organ-specific surveillance of newly identified RET carriers going forward.

Restricted access
Masaki Shiota Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan

Search for other papers by Masaki Shiota in
Google Scholar
PubMed
Close
,
Satoshi Endo United Graduate School of Medical Information Sciences, Gifu University, Gifu, Japan
Center for One Medicine Innovative Translational Research (COMIT), Gifu University, Gifu, Japan

Search for other papers by Satoshi Endo in
Google Scholar
PubMed
Close
,
Shigehiro Tsukahara Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan

Search for other papers by Shigehiro Tsukahara in
Google Scholar
PubMed
Close
,
Tokiyoshi Tanegashima Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan

Search for other papers by Tokiyoshi Tanegashima in
Google Scholar
PubMed
Close
,
Satoshi Kobayashi Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan

Search for other papers by Satoshi Kobayashi in
Google Scholar
PubMed
Close
,
Takashi Matsumoto Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan

Search for other papers by Takashi Matsumoto in
Google Scholar
PubMed
Close
, and
Masatoshi Eto Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan

Search for other papers by Masatoshi Eto in
Google Scholar
PubMed
Close

Androgen receptor signaling is crucial for the development of treatment resistance in prostate cancer. Among steroidogenic enzymes, 3β-hydroxysteroid dehydrogenases (3βHSDs) play critical roles in extragonadal androgen synthesis, especially 3βHSD1. Increased expression of 3βHSDs is observed in castration-resistant prostate cancer tumors compared with primary prostate tumors, indicating their involvement in castration resistance. Recent studies link 3βHSD1 to resistance to androgen receptor signaling inhibitors. The regulation of 3βHSD1 expression involves various factors, including transcription factors, microenvironmental influences, and posttranscriptional modifications. Additionally, the clinical significance of HSD3B1 genotypes, particularly the rs1047303 variant, has been extensively studied. The impact of HSD3B1 genotypes on treatment outcomes varies according to the therapy administered, suggesting the potential of HSD3B1 genotyping for personalized medicine. Targeting 3βHSDs may be a promising strategy for prostate cancer management. Overall, understanding the roles of 3βHSDs and their genetic variations may enable the development and optimization of novel treatments for prostate cancer.

Restricted access