Elahe Mostaghel, Victoria Wang, Brett T Marck, Nima Sharifi, Alvin M Matsumoto, Christopher J Sweeney
The CHAARTED study showed that adding docetaxel (Doc) to androgen deprivation therapy (ADT) in men initiating treatment for metastatic hormone sensitive prostate cancer (mHSPC) prolongs survival, particularly in high-volume disease. Androgens drive both mHSPC and metastatic castration resistant prostate cancer (mCRPC). Lower nadir serum testosterone (T) concentrations are associated with better outcomes in men treated with ADT for biochemical relapse, while higher androgens at mCRPC are associated with better prognosis and increased benefit from abiraterone. We evaluated the association of serum steroids at 24 weeks with overall survival (OS) and time to CRPC (TTCRPC) in 588 men with available samples from the CHAARTED study. Steroid concentrations were measured using mass spectrometry. The median T concentration at 24 weeks was 8 ng/dl and did not differ in ADT alone vs ADT plus Doc arms. Achieving nadir T below 20ng/dl was not associated with OS or TTCRPC in either arm. In high volume disease, Doc conferred an OS and TTCRPC benefit regardless of steroid concentrations. In low volume disease, steroid concentrations in the lowest quartile at week 24 identified a subset of men with poor survival outcomes more like high volume disease, and in whom Doc was also associated with improved OS and TTCRPC. The known OS benefit of Doc in high volume mHSPC is not modified by serum steroid concentrations achieved on treatment. In low volume disease, steroid concentrations in the lowest quartile may identify a poor prognosis subset in whom Doc also confers OS benefit.
{"title":"Association of serum steroids with survival in metastatic hormone-sensitive prostate cancer.","authors":"Elahe Mostaghel, Victoria Wang, Brett T Marck, Nima Sharifi, Alvin M Matsumoto, Christopher J Sweeney","doi":"10.1530/ERC-24-0140","DOIUrl":"10.1530/ERC-24-0140","url":null,"abstract":"<p><p>The CHAARTED study showed that adding docetaxel (Doc) to androgen deprivation therapy (ADT) in men initiating treatment for metastatic hormone sensitive prostate cancer (mHSPC) prolongs survival, particularly in high-volume disease. Androgens drive both mHSPC and metastatic castration resistant prostate cancer (mCRPC). Lower nadir serum testosterone (T) concentrations are associated with better outcomes in men treated with ADT for biochemical relapse, while higher androgens at mCRPC are associated with better prognosis and increased benefit from abiraterone. We evaluated the association of serum steroids at 24 weeks with overall survival (OS) and time to CRPC (TTCRPC) in 588 men with available samples from the CHAARTED study. Steroid concentrations were measured using mass spectrometry. The median T concentration at 24 weeks was 8 ng/dl and did not differ in ADT alone vs ADT plus Doc arms. Achieving nadir T below 20ng/dl was not associated with OS or TTCRPC in either arm. In high volume disease, Doc conferred an OS and TTCRPC benefit regardless of steroid concentrations. In low volume disease, steroid concentrations in the lowest quartile at week 24 identified a subset of men with poor survival outcomes more like high volume disease, and in whom Doc was also associated with improved OS and TTCRPC. The known OS benefit of Doc in high volume mHSPC is not modified by serum steroid concentrations achieved on treatment. In low volume disease, steroid concentrations in the lowest quartile may identify a poor prognosis subset in whom Doc also confers OS benefit.</p>","PeriodicalId":93989,"journal":{"name":"Endocrine-related cancer","volume":" ","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142718039","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Catrin Lutz, Hendrik A Messal, Damir Vareslija, Stefan Prekovic
The breast epithelium, vital for mammary gland function, is influenced by oestrogen through the ER signalling pathway. Luminal breast cancer, characterised by ER expression, comprises the majority of all breast cancers and presents significant clinical challenges due to therapy resistance and recurrence. Despite advancements in understanding luminal disease, improving long-term survival and reducing relapse of breast cancer patients by predicting therapy efficacy and understanding resistance mechanisms remain critical challenges. This review discusses luminal breast cancer biology, focusing on molecular classification of the primary disease, metastatic spread, and experimental models.
乳腺上皮对乳腺功能至关重要,它通过 ER 信号通路受到雌激素的影响。以ER表达为特征的腔隙型乳腺癌占所有乳腺癌的大多数,由于耐药和复发,给临床带来了巨大挑战。尽管在了解腔隙性疾病方面取得了进展,但通过预测疗效和了解耐药机制来提高乳腺癌患者的长期生存率和减少复发仍然是严峻的挑战。本综述讨论腔隙性乳腺癌生物学,重点是原发疾病的分子分类、转移扩散和实验模型。
{"title":"The complex landscape of luminal breast cancer.","authors":"Catrin Lutz, Hendrik A Messal, Damir Vareslija, Stefan Prekovic","doi":"10.1530/ERC-24-0201","DOIUrl":"https://doi.org/10.1530/ERC-24-0201","url":null,"abstract":"<p><p>The breast epithelium, vital for mammary gland function, is influenced by oestrogen through the ER signalling pathway. Luminal breast cancer, characterised by ER expression, comprises the majority of all breast cancers and presents significant clinical challenges due to therapy resistance and recurrence. Despite advancements in understanding luminal disease, improving long-term survival and reducing relapse of breast cancer patients by predicting therapy efficacy and understanding resistance mechanisms remain critical challenges. This review discusses luminal breast cancer biology, focusing on molecular classification of the primary disease, metastatic spread, and experimental models.</p>","PeriodicalId":93989,"journal":{"name":"Endocrine-related cancer","volume":" ","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142585298","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Maaia Margo Jentus, Leontine Bakker, Marco Verstegen, Iris Pelsma, Tom van Wezel, Dina Ruano, Ellen Kapiteijn, Stijn Crobach, Nienke R Biermasz, Hans Morreau
The molecular biology of pituitary neuroendocrine tumors (PitNETs) revealed few recurrent mutations and extensive chromosomal alterations, with the latter being the driving force in a subset of these lesions. Addressing the need for an easily applicable diagnostic tool, we conducted a retrospective study of 61 PitNETs operated at a tertiary care center. All cases were subtyped according to the 2022 WHO classification of endocrine tumors. A genome wide NGS panel targeting 1500 single-nucleotide polymorphisms (SNP) was used to classify chromosomal imbalances, loss of heterozygosity and copy number variations in DNA from formalin fixed paraffin embedded tissues. We identified four distinct chromosomal patterns, with varying distribution among different tumor lineages. Forty-two of 61 (69%) PitNETs showed chromosomal alterations. Gonadotroph PitNETs showed mostly quiet genomes. The majority of lactotroph PitNETs (19/20, 95%) were altered, exhibiting a gained genome and a remarkably low recurrence rate. Nine of ten (90%) corticotroph PitNETs harbored chromosomal alterations, of which two aggressive corticotroph tumors and one metastatic corticotroph PitNET showed massive chromosomal losses leading to near haploid/near homozygous genomes. The comparison of the molecular profile of primary and recurrent PitNETs of five patients showed no significant accumulation of alterations over time. A simple genome wide 1500 SNP test can be used in the identification of outspoken aggressive subsets of PitNET by the occurrence of a near haploid/near homozygous genome. Furthermore, the presence of neoplastic tissue in resected material can be potentially confirmed for non-gonadotroph PitNETs in suboptimal histological assessment conditions.
垂体神经内分泌肿瘤(PitNETs)的分子生物学研究发现,复发性突变很少,而染色体改变却很广泛,后者是这些病变中一部分的驱动力。为了满足对易于应用的诊断工具的需求,我们对在一家三级医疗中心手术的 61 例 PitNET 进行了回顾性研究。所有病例都根据 2022 年世界卫生组织内分泌肿瘤分类进行了亚型划分。研究人员利用针对 1500 个单核苷酸多态性(SNP)的全基因组 NGS 面板对福尔马林固定石蜡包埋组织 DNA 中的染色体失衡、杂合性缺失和拷贝数变异进行了分类。我们发现了四种不同的染色体模式,它们在不同肿瘤系中的分布各不相同。61例PitNET中有42例(69%)出现染色体改变。促性腺激素型PitNET的基因组大多是安静的。大多数泌乳型PitNET(19/20,95%)的基因组发生了改变,表现为基因组增殖,且复发率极低。10个皮质营养型PitNET中有9个(90%)存在染色体改变,其中2个侵袭性皮质营养型肿瘤和1个转移性皮质营养型PitNET出现大量染色体缺失,导致近单倍体/近同源基因组。对五名患者的原发性和复发性 PitNET 的分子图谱进行比较后发现,随着时间的推移,这些改变并没有明显的累积。一个简单的全基因组 1500 SNP 检测可用于通过近单倍体/近同源基因组的出现来鉴别 PitNET 的明显侵袭性亚群。此外,在组织学评估条件不理想的情况下,切除材料中是否存在肿瘤组织也有可能被确认为非促性腺激素PitNET。
{"title":"Chromosomal Alteration Patterns in PitNETs: Massive Losses in Aggressive Tumors.","authors":"Maaia Margo Jentus, Leontine Bakker, Marco Verstegen, Iris Pelsma, Tom van Wezel, Dina Ruano, Ellen Kapiteijn, Stijn Crobach, Nienke R Biermasz, Hans Morreau","doi":"10.1530/ERC-24-0070","DOIUrl":"https://doi.org/10.1530/ERC-24-0070","url":null,"abstract":"<p><p>The molecular biology of pituitary neuroendocrine tumors (PitNETs) revealed few recurrent mutations and extensive chromosomal alterations, with the latter being the driving force in a subset of these lesions. Addressing the need for an easily applicable diagnostic tool, we conducted a retrospective study of 61 PitNETs operated at a tertiary care center. All cases were subtyped according to the 2022 WHO classification of endocrine tumors. A genome wide NGS panel targeting 1500 single-nucleotide polymorphisms (SNP) was used to classify chromosomal imbalances, loss of heterozygosity and copy number variations in DNA from formalin fixed paraffin embedded tissues. We identified four distinct chromosomal patterns, with varying distribution among different tumor lineages. Forty-two of 61 (69%) PitNETs showed chromosomal alterations. Gonadotroph PitNETs showed mostly quiet genomes. The majority of lactotroph PitNETs (19/20, 95%) were altered, exhibiting a gained genome and a remarkably low recurrence rate. Nine of ten (90%) corticotroph PitNETs harbored chromosomal alterations, of which two aggressive corticotroph tumors and one metastatic corticotroph PitNET showed massive chromosomal losses leading to near haploid/near homozygous genomes. The comparison of the molecular profile of primary and recurrent PitNETs of five patients showed no significant accumulation of alterations over time. A simple genome wide 1500 SNP test can be used in the identification of outspoken aggressive subsets of PitNET by the occurrence of a near haploid/near homozygous genome. Furthermore, the presence of neoplastic tissue in resected material can be potentially confirmed for non-gonadotroph PitNETs in suboptimal histological assessment conditions.</p>","PeriodicalId":93989,"journal":{"name":"Endocrine-related cancer","volume":" ","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142515229","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Multiple endocrine neoplasia type 2 (MEN2) is the collective term for 2 distinct types of autosomal dominantly inherited neuroendocrine neoplasm (NEN) syndromes (Barakat, 2004); MEN2A and MEN 2B (or MEN3). MEN2 is characterised by medullary thyroid cancer (99%) and phaeochromocytoma (50%) but also other conditions according to specific genotype. MEN2A also includes a 25% risk of developing parathyroid hyperplasia and is now recognised as 4 separate syndromes: Classic MEN2A, MEN2A with cutaneous lichen amyloidosis (CLA), MEN2A with Hirschsprung's disease (HD) and Familial MTC (Wells, 2015). MEN2B accounts for around 5% of all MEN2 cases and predisposes patients to diffuse intestinal ganglioneuromatosis (Goncharova, 2020), mucosal neuromas, and musculoskeletal abnormalities (Henderson, 2022). MEN2 is autosomal dominantly inherited meaning that several generations in a single family may be affected by the same syndrome. We present a mini review of 4 case studies (x2 MEN2A, x2 MEN2B) that illustrate the advantages of RET testing, as well as some of the likely obstacles that must be overcome to receive a diagnosis of MEN2A or MEN2B. In addition, despite improved genotype/phenotype correlation in MEN2, we highlight that not all cases are 'typical' which emphasises the need for all MEN2 patients to be cared for in a centre of expertise and experience. Some of our case study patients or parents also took this opportunity to personally tell us more about their lives with MEN2, illustrating the need for more research into the psychosocial impact of these hereditary diseases.
{"title":"Living with a RET gene mutation: patient perspectives.","authors":"Caroline Brain, Joanna Grey, Kirstie Purnell","doi":"10.1530/ERC-24-0130","DOIUrl":"https://doi.org/10.1530/ERC-24-0130","url":null,"abstract":"<p><p>Multiple endocrine neoplasia type 2 (MEN2) is the collective term for 2 distinct types of autosomal dominantly inherited neuroendocrine neoplasm (NEN) syndromes (Barakat, 2004); MEN2A and MEN 2B (or MEN3). MEN2 is characterised by medullary thyroid cancer (99%) and phaeochromocytoma (50%) but also other conditions according to specific genotype. MEN2A also includes a 25% risk of developing parathyroid hyperplasia and is now recognised as 4 separate syndromes: Classic MEN2A, MEN2A with cutaneous lichen amyloidosis (CLA), MEN2A with Hirschsprung's disease (HD) and Familial MTC (Wells, 2015). MEN2B accounts for around 5% of all MEN2 cases and predisposes patients to diffuse intestinal ganglioneuromatosis (Goncharova, 2020), mucosal neuromas, and musculoskeletal abnormalities (Henderson, 2022). MEN2 is autosomal dominantly inherited meaning that several generations in a single family may be affected by the same syndrome. We present a mini review of 4 case studies (x2 MEN2A, x2 MEN2B) that illustrate the advantages of RET testing, as well as some of the likely obstacles that must be overcome to receive a diagnosis of MEN2A or MEN2B. In addition, despite improved genotype/phenotype correlation in MEN2, we highlight that not all cases are 'typical' which emphasises the need for all MEN2 patients to be cared for in a centre of expertise and experience. Some of our case study patients or parents also took this opportunity to personally tell us more about their lives with MEN2, illustrating the need for more research into the psychosocial impact of these hereditary diseases.</p>","PeriodicalId":93989,"journal":{"name":"Endocrine-related cancer","volume":" ","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142549666","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-09-21Print Date: 2024-10-01DOI: 10.1530/ERC-24-0025
A Mohamed, M Trybula, S L Asa, T R Halfdanarson, M B Sonbol
The classification and management of neuroendocrine neoplasms (NENs) arising in the tubular gastrointestinal (GI) tract and pancreas have significantly evolved over the last decades. In the latest WHO classification published in 2022, NENs are separated regardless of their primary origin into two main groups: well-differentiated neuroendocrine tumors (NETs) and poorly differentiated neuroendocrine carcinomas (NECs). The substantial changes in the grading system changed the definition of grade 3 to include high-grade well-differentiated NETs (G3-NETs), and poorly differentiated NECs (-NECs). Although these two subgroups are considered high grades with Ki-67 >20%, they have different genomic profiles, prognosis, and clinical behavior, which critically influence their treatment strategies. The available clinical trial data to guide therapy of these high-grade subgroups are extremely limited, which impacts their management. In this review, we will summarize the current advances in the multidisciplinary approach for the management of high-grade gastroenteropancreatic NENs (GEP-NENs) including G3-NETs and NECs.
{"title":"Management of advanced high grade gastroenteropancreatic neuroendocrine neoplasms (GEP-NENs): comprehensive review of the current literature.","authors":"A Mohamed, M Trybula, S L Asa, T R Halfdanarson, M B Sonbol","doi":"10.1530/ERC-24-0025","DOIUrl":"10.1530/ERC-24-0025","url":null,"abstract":"<p><p>The classification and management of neuroendocrine neoplasms (NENs) arising in the tubular gastrointestinal (GI) tract and pancreas have significantly evolved over the last decades. In the latest WHO classification published in 2022, NENs are separated regardless of their primary origin into two main groups: well-differentiated neuroendocrine tumors (NETs) and poorly differentiated neuroendocrine carcinomas (NECs). The substantial changes in the grading system changed the definition of grade 3 to include high-grade well-differentiated NETs (G3-NETs), and poorly differentiated NECs (-NECs). Although these two subgroups are considered high grades with Ki-67 >20%, they have different genomic profiles, prognosis, and clinical behavior, which critically influence their treatment strategies. The available clinical trial data to guide therapy of these high-grade subgroups are extremely limited, which impacts their management. In this review, we will summarize the current advances in the multidisciplinary approach for the management of high-grade gastroenteropancreatic NENs (GEP-NENs) including G3-NETs and NECs.</p>","PeriodicalId":93989,"journal":{"name":"Endocrine-related cancer","volume":" ","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-09-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141918391","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-09-21Print Date: 2024-10-01DOI: 10.1530/ERC-24-0111
Adam I Kaplan, Trisha Dwight, Catherine Luxford, Diana E Benn, Roderick J Clifton-Bligh
Phaeochromocytomas and paragangliomas (collectively termed PPGL) are rare yet highly heritable neuroendocrine tumours, with over one-third of cases associated with germline pathogenic variants (PVs) in numerous genes. PVs in the succinate dehydrogenase subunit-A gene (SDHA) were initially implicated in hereditary PPGL in 2010, and SDHA has since become an important susceptibility gene accounting for up to 2.8% of cases. However, it remains poorly understood, particularly regarding the clinical nature of SDHA PPGL, rates of recurrence and metastasis, and the nature of metastatic disease. We present a narrative review of SDHA-related PPGL, covering pathophysiology, relevance to current clinical practice, and considerations for clinical genetics. We analyse a pool of 107 previously reported cases of SDHA-associated PPGL to highlight the spectrum of SDHA-related PPGL. Our analysis demonstrates that SDHA PPGL occurs across a wide age range (11-81 years) and affects men and women equally. SDHA PPGL typically presents as single tumours (91%), usually occurring in the head and neck (46%) or abdomen (43%, including 15% with phaeochromocytomas). Metastatic disease was reported in 25.5% of cases, with bone (82%) and lymph nodes (71%) being the most common sites of metastasis, often identified many years after the initial diagnosis. A family history of SDHA-related neoplasia was rare, reported in only 4% of cases. Understanding the clinical nature and risks associated with SDHA PVs is essential for facilitating the optimal management of patients and their families.
{"title":"SDHA-related phaeochromocytoma and paraganglioma: review and clinical management.","authors":"Adam I Kaplan, Trisha Dwight, Catherine Luxford, Diana E Benn, Roderick J Clifton-Bligh","doi":"10.1530/ERC-24-0111","DOIUrl":"10.1530/ERC-24-0111","url":null,"abstract":"<p><p>Phaeochromocytomas and paragangliomas (collectively termed PPGL) are rare yet highly heritable neuroendocrine tumours, with over one-third of cases associated with germline pathogenic variants (PVs) in numerous genes. PVs in the succinate dehydrogenase subunit-A gene (SDHA) were initially implicated in hereditary PPGL in 2010, and SDHA has since become an important susceptibility gene accounting for up to 2.8% of cases. However, it remains poorly understood, particularly regarding the clinical nature of SDHA PPGL, rates of recurrence and metastasis, and the nature of metastatic disease. We present a narrative review of SDHA-related PPGL, covering pathophysiology, relevance to current clinical practice, and considerations for clinical genetics. We analyse a pool of 107 previously reported cases of SDHA-associated PPGL to highlight the spectrum of SDHA-related PPGL. Our analysis demonstrates that SDHA PPGL occurs across a wide age range (11-81 years) and affects men and women equally. SDHA PPGL typically presents as single tumours (91%), usually occurring in the head and neck (46%) or abdomen (43%, including 15% with phaeochromocytomas). Metastatic disease was reported in 25.5% of cases, with bone (82%) and lymph nodes (71%) being the most common sites of metastasis, often identified many years after the initial diagnosis. A family history of SDHA-related neoplasia was rare, reported in only 4% of cases. Understanding the clinical nature and risks associated with SDHA PVs is essential for facilitating the optimal management of patients and their families.</p>","PeriodicalId":93989,"journal":{"name":"Endocrine-related cancer","volume":" ","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-09-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11466202/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141918392","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Marta Araujo-Castro, Betina Biagetti, Edelmiro Menéndez Torre, Iría Novoa-Testa, Fernando Cordido, Eider Pascual-Corrales, Víctor Rodríguez Berrocal, Fernando Guerrero-Pérez, Almudena Vicente, Juan Carlos Percovich Hualpa, Rogelio García-Centeno, Laura González-Fernández, María Dolores Ollero García, Ana Irigaray Echarri, María Dolores Moure Rodríguez, Cristina Novo-Rodríguez, María Calatayud, Rocío Villar-Taibo, Ignacio Bernabéu, Cristina Alvarez-Escola, Pamela Benítez Valderrama, Carmen Tenorio-Jiménez, Pablo Abellán Galiana, Eva Venegas, Inmaculada González-Molero, Pedro Iglesias, Concepción Blanco-Carrera, Fernando Vidal-Ostos De Lara, Paz de Miguel Novoa, Elena López Mezquita, Felicia Alexandra Hanzu, Iban Aldecoa, Silvia Aznar, Cristina Lamas, Anna Aulinas, Queralt Asla, Paola Gracia Gimeno, José María Recio-Córdova, María Dolores Avilés-Pérez, Diego Asensio-Wandosell, Miguel Sampedro-Núñez, Rosa Cámara, Miguel Paja Fano, Ignacio Ruz-Caracuel, Carmen Fajardo, Mónica Marazuela, Manel Puig-Domingo
{"title":"ERRATUM: Pegvisomant and pasireotide in PRL and GH co-secreting vs GH-secreting Pit-NETs.","authors":"Marta Araujo-Castro, Betina Biagetti, Edelmiro Menéndez Torre, Iría Novoa-Testa, Fernando Cordido, Eider Pascual-Corrales, Víctor Rodríguez Berrocal, Fernando Guerrero-Pérez, Almudena Vicente, Juan Carlos Percovich Hualpa, Rogelio García-Centeno, Laura González-Fernández, María Dolores Ollero García, Ana Irigaray Echarri, María Dolores Moure Rodríguez, Cristina Novo-Rodríguez, María Calatayud, Rocío Villar-Taibo, Ignacio Bernabéu, Cristina Alvarez-Escola, Pamela Benítez Valderrama, Carmen Tenorio-Jiménez, Pablo Abellán Galiana, Eva Venegas, Inmaculada González-Molero, Pedro Iglesias, Concepción Blanco-Carrera, Fernando Vidal-Ostos De Lara, Paz de Miguel Novoa, Elena López Mezquita, Felicia Alexandra Hanzu, Iban Aldecoa, Silvia Aznar, Cristina Lamas, Anna Aulinas, Queralt Asla, Paola Gracia Gimeno, José María Recio-Córdova, María Dolores Avilés-Pérez, Diego Asensio-Wandosell, Miguel Sampedro-Núñez, Rosa Cámara, Miguel Paja Fano, Ignacio Ruz-Caracuel, Carmen Fajardo, Mónica Marazuela, Manel Puig-Domingo","doi":"10.1530/ERC-24-0043e","DOIUrl":"10.1530/ERC-24-0043e","url":null,"abstract":"","PeriodicalId":93989,"journal":{"name":"Endocrine-related cancer","volume":"31 9","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-07-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141768316","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-07-22Print Date: 2024-09-01DOI: 10.1530/ERC-23-0337
E Baudin, J Capdevila, D Hörsch, S Singh, M E Caplin, E M Wolin, W Buikhuisen, M Raderer, E Dansin, C Grohe, D Ferone, A Houchard, X-M Truong-Thanh, D Reidy-Lagunes
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.
{"title":"Treatment of advanced BP-NETS with lanreotide autogel/depot vs placebo: the phase III SPINET study.","authors":"E Baudin, J Capdevila, D Hörsch, S Singh, M E Caplin, E M Wolin, W Buikhuisen, M Raderer, E Dansin, C Grohe, D Ferone, A Houchard, X-M Truong-Thanh, D Reidy-Lagunes","doi":"10.1530/ERC-23-0337","DOIUrl":"10.1530/ERC-23-0337","url":null,"abstract":"<p><p>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.</p>","PeriodicalId":93989,"journal":{"name":"Endocrine-related cancer","volume":" ","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-07-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11301421/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141447868","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-07-02Print Date: 2024-08-01DOI: 10.1530/ERC-23-0354
Marcia S Brose, Jaume Capdevila, Rossella Elisei, Lars Bastholt, Dagmar Führer-Sakel, Sophie Leboulleux, Iwao Sugitani, Matthew H Taylor, Zhuoying Wang, Lori J Wirth, Francis P Worden, John Bernard, Paolo Caferra, Raffaella M Colzani, Shiguang Liu, Martin Schlumberger
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.
{"title":"Vandetanib in locally advanced or metastatic differentiated thyroid cancer refractory to radioiodine therapy.","authors":"Marcia S Brose, Jaume Capdevila, Rossella Elisei, Lars Bastholt, Dagmar Führer-Sakel, Sophie Leboulleux, Iwao Sugitani, Matthew H Taylor, Zhuoying Wang, Lori J Wirth, Francis P Worden, John Bernard, Paolo Caferra, Raffaella M Colzani, Shiguang Liu, Martin Schlumberger","doi":"10.1530/ERC-23-0354","DOIUrl":"10.1530/ERC-23-0354","url":null,"abstract":"<p><p>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.</p>","PeriodicalId":93989,"journal":{"name":"Endocrine-related cancer","volume":" ","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-07-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11301419/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141201610","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}