1
|
Jentus MM, Bakker L, Verstegen M, Pelsma I, van Wezel T, Ruano D, Kapiteijn E, Crobach S, Biermasz N, Morreau H. Chromosomal alteration patterns in PitNETs: massive losses in aggressive tumors. Endocr Relat Cancer 2025; 32:e240070. [PMID: 39441837 PMCID: PMC11729050 DOI: 10.1530/erc-24-0070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 10/23/2024] [Accepted: 10/23/2024] [Indexed: 10/25/2024]
Abstract
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 next-generation sequencing panel targeting 1500 single nucleotide polymorphisms (SNPs) 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 PitNETs by the occurrence of a near-haploid/near-homozygous genome. Furthermore, the presence of neoplastic tissue in the resected material can be potentially confirmed for non-gonadotroph PitNETs under suboptimal histological assessment conditions.
Collapse
Affiliation(s)
- Maaia Margo Jentus
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Leontine Bakker
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
- Center for Endocrine Tumors Leiden (CETL), Pituitary Center, Leiden University Medical Center, Leiden, The Netherlands
| | - Marco Verstegen
- Center for Endocrine Tumors Leiden (CETL), Pituitary Center, Leiden University Medical Center, Leiden, The Netherlands
- Department of Neurosurgery, Center for Endocrine Tumors Leiden (CETL), Leiden University Medical Center, Leiden, The Netherlands
| | - Iris Pelsma
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
- Center for Endocrine Tumors Leiden (CETL), Pituitary Center, Leiden University Medical Center, Leiden, The Netherlands
| | - Tom van Wezel
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Dina Ruano
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Ellen Kapiteijn
- Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - Stijn Crobach
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
- Center for Endocrine Tumors Leiden (CETL), Pituitary Center, Leiden University Medical Center, Leiden, The Netherlands
| | - Nienke Biermasz
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
- Center for Endocrine Tumors Leiden (CETL), Pituitary Center, Leiden University Medical Center, Leiden, The Netherlands
| | - Hans Morreau
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
- Center for Endocrine Tumors Leiden (CETL), Pituitary Center, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
2
|
DE Alcubierre D, Carretti AL, Ducray F, Jouanneau E, Raverot G, Ilie MD. Aggressive pituitary tumors and carcinomas: medical treatment beyond temozolomide. Minerva Endocrinol (Torino) 2024; 49:321-334. [PMID: 38240681 DOI: 10.23736/s2724-6507.23.04058-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2024]
Abstract
Aggressive pituitary tumors are a subset of pituitary neoplasms, characterized by unusually fast growth rate, invasiveness and overall resistance to optimized standard treatment. When metastases are present, the term pituitary carcinoma is employed. After failure of standard treatments, current guidelines recommend first-line temozolomide monotherapy. However, a significant number of patients do not respond to temozolomide, or experience disease progression following its discontinuation; in these latter cases, re-challenge with temozolomide is generally advised, although the reported outcomes have been less satisfactory. Although no alternative therapies have been formally recommended after temozolomide failure, growing evidence regarding potential second- or third-line therapeutic strategies has emerged. In the present work, we reviewed the available evidence published up to April 2023 involving the most relevant therapies employed so far, namely immune checkpoint inhibitors, bevacizumab, peptide radionuclide receptor therapy, tyrosine kinase inhibitors and mTOR inhibitors. For each treatment, we report efficacy and safety outcomes, along with data regarding potential predictors of response. Overall, immune checkpoint inhibitors and bevacizumab are showing the most promise as therapeutic options after temozolomide failure. The former showed better responses in pituitary carcinomas. Peptide radionuclide receptor therapy has also showed some efficacy in these tumors, while tyrosine kinase inhibitors and mTOR inhibitors have exhibited so far limited or no efficacy. Further studies, as well as an individualized, patient-tailored approach, are clearly needed. In addition, we report an unpublished case of a silent corticotroph pituitary carcinoma that progressed under dual immunotherapy, and then showed stable disease under a combination of lomustine and bevacizumab.
Collapse
Affiliation(s)
- Dario DE Alcubierre
- Department of Experimental Medicine, Sapienza University, Rome, Italy
- Inserm U1052, CNRS UMR5286, Cancer Research Center of Lyon, Claude Bernard Lyon1 University, Lyon, France
| | - Anna L Carretti
- Department of Experimental Medicine, Sapienza University, Rome, Italy
- Department of Endocrinology, Reference Center for Rare Pituitary Diseases HYPO, Groupement Hospitalier Est Hospices Civils de Lyon, Bron, France
| | - François Ducray
- Service of Neuro-Oncology, Groupement Hospitalier Est Hospices Civils de Lyon, Bron, France
| | - Emmanuel Jouanneau
- Inserm U1052, CNRS UMR5286, Cancer Research Center of Lyon, Claude Bernard Lyon1 University, Lyon, France
- Department of Pituitary and Skull Base Neurosurgical, Reference Center for Rare Pituitary Diseases HYPO, Groupement Hospitalier Est Hospices Civils de Lyon, Bron, France
| | - Gérald Raverot
- Inserm U1052, CNRS UMR5286, Cancer Research Center of Lyon, Claude Bernard Lyon1 University, Lyon, France -
- Department of Endocrinology, Reference Center for Rare Pituitary Diseases HYPO, Groupement Hospitalier Est Hospices Civils de Lyon, Bron, France
| | - Mirela D Ilie
- Inserm U1052, CNRS UMR5286, Cancer Research Center of Lyon, Claude Bernard Lyon1 University, Lyon, France
- Department of Endocrinology, C.I. Parhon National Institute of Endocrinology, Bucharest, Romania
| |
Collapse
|
3
|
Lopes-Pinto M, Lacerda-Nobre E, Silva AL, Marques P. Therapeutical Usefulness of PD-1/PD-L1 Inhibitors in Aggressive or Metastatic Pituitary Tumours. Cancers (Basel) 2024; 16:3033. [PMID: 39272895 PMCID: PMC11394371 DOI: 10.3390/cancers16173033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 08/25/2024] [Accepted: 08/28/2024] [Indexed: 09/15/2024] Open
Abstract
Therapeutic options for pituitary neuroendocrine tumours (PitNETs) refractory to temozolomide are scarce. Immune checkpoint inhibitors (ICIs), particularly inhibitors of the programmed cell death-1 (PD-1) pathway and its ligand (PD-L1), have been experimentally used in aggressive or metastatic PitNETs. We aimed to study the therapeutic usefulness of anti-PD-1 drugs in patients with aggressive or metastatic PitNETs. Published cases and case series involving patients with PitNETs treated with PD-1/PD-L1 inhibitors were reviewed. Demographic data, clinical-pathological features, previous therapies, drug dosage and posology, and the best radiological and biochemical responses, as well as survival data, were evaluated. We identified 29 cases of aggressive (n = 13) or metastatic (n = 16) PitNETs treated with PD-1/PD-L1 inhibitors. The hypersecretion of adrenocorticotropic hormone (ACTH) was documented in eighteen cases (62.1%), seven were prolactinomas (24.1%), and four were non-functioning PitNETs. All patients underwent various therapies prior to using ICIs. Overall, a positive radiological response (i.e., partial/complete radiological response and stable disease) was observed in eighteen of twenty-nine cases (62.1%), of which ten and four were ACTH- and prolactin-secreting PitNETs, respectively. Hormonal levels reduced or stabilised after using ICIs in 11 of the 17 functioning PitNET cases with available data (64.7%). The median survival of patients treated with ICIs was 13 months, with a maximum of 42 months in two ACTH-secreting tumours. Among 29 patients with PitNETs treated with PD-1/PD-L1 inhibitors, the positive radiological and biochemical response rates were 62.1% and 64.7%, respectively. Altogether, these data suggest a promising role of ICIs in patients with aggressive or metastatic PitNETs refractory to other treatment modalities.
Collapse
Affiliation(s)
- Mariana Lopes-Pinto
- Endocrinology Department, Unidade Local de Saúde de Santa Maria, Hospital de Santa Maria, 1649-035 Lisbon, Portugal
| | - Ema Lacerda-Nobre
- Endocrinology Department, Unidade Local de Saúde de Santa Maria, Hospital de Santa Maria, 1649-035 Lisbon, Portugal
- Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Ana Luísa Silva
- Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
- Instituto de Saúde Ambiental, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
- Faculdade de Medicina, Universidade Católica Portuguesa, 1649-023 Lisbon, Portugal
| | - Pedro Marques
- Faculdade de Medicina, Universidade Católica Portuguesa, 1649-023 Lisbon, Portugal
- Pituitary Tumor Unit, Endocrinology Department, Hospital CUF Descobertas, 1998-018 Lisbon, Portugal
| |
Collapse
|
4
|
Guo X, Yang Y, Qian Z, Chang M, Zhao Y, Ma W, Wang Y, Xing B. Immune landscape and progress in immunotherapy for pituitary neuroendocrine tumors. Cancer Lett 2024; 592:216908. [PMID: 38677640 DOI: 10.1016/j.canlet.2024.216908] [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: 02/07/2024] [Revised: 04/17/2024] [Accepted: 04/18/2024] [Indexed: 04/29/2024]
Abstract
Pituitary neuroendocrine tumors (pitNETs) are the second most common primary brain tumors. Despite their prevalence, the tumor immune microenvironment (TIME) and its clinical implications remain largely unexplored. This review provides a comprehensive overview of current knowledge on the immune landscape and advancements in targeted immunotherapy for pitNETs. Macrophages and T cells are principal immune infiltrates within the TIME. Different subtypes of pitNETs display distinct immune patterns, influencing tumor progressive behaviors. PD-L1, the most extensively studied immune checkpoint, is prominently expressed in hormonal pitNETs and correlates with tumor growth and invasion. Cytokines and chemokines including interleukins, CCLs, and CXCLs have complex correlations with tumor subtypes and immune cell infiltration. Crosstalk between macrophages and pitNET cells highlights bidirectional regulatory roles, suggesting potential macrophage-targeted strategies. Recent preclinical studies have demonstrated the efficacy of anti-PD-L1 therapy in a mouse model of corticotroph pitNET. Moreover, anti-PD-1 and/or anti-CTLA-4 immunotherapy has been applied globally in 28 cases of refractory pitNETs, showing more favorable responses in pituitary carcinomas than aggressive pitNETs. In conclusion, the TIME of pitNETs represents a promising avenue for targeted immunotherapy and warrants further investigation.
Collapse
Affiliation(s)
- Xiaopeng Guo
- Department of Neurosurgery, Key Laboratory of Endocrinology of National Ministry of Health, China Pituitary Adenoma Specialist Council, China Pituitary Disease Registry Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yiying Yang
- Department of Neurosurgery, Key Laboratory of Endocrinology of National Ministry of Health, China Pituitary Adenoma Specialist Council, China Pituitary Disease Registry Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Eight-Year Program of Clinical Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, China
| | - Zhihong Qian
- Department of Neurosurgery, Key Laboratory of Endocrinology of National Ministry of Health, China Pituitary Adenoma Specialist Council, China Pituitary Disease Registry Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Mengqi Chang
- Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuanli Zhao
- Department of Neurosurgery, Key Laboratory of Endocrinology of National Ministry of Health, China Pituitary Adenoma Specialist Council, China Pituitary Disease Registry Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wenbin Ma
- Department of Neurosurgery, Key Laboratory of Endocrinology of National Ministry of Health, China Pituitary Adenoma Specialist Council, China Pituitary Disease Registry Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yu Wang
- Department of Neurosurgery, Key Laboratory of Endocrinology of National Ministry of Health, China Pituitary Adenoma Specialist Council, China Pituitary Disease Registry Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Bing Xing
- Department of Neurosurgery, Key Laboratory of Endocrinology of National Ministry of Health, China Pituitary Adenoma Specialist Council, China Pituitary Disease Registry Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
5
|
Lin AL, Rudneva VA, Richards AL, Zhang Y, Woo HJ, Cohen M, Tisnado J, Majd N, Wardlaw SL, Page-Wilson G, Sengupta S, Chow F, Goichot B, Ozer BH, Dietrich J, Nachtigall L, Desai A, Alano T, Ogilive S, Solit DB, Bale TA, Rosenblum M, Donoghue MTA, Geer EB, Tabar V. Genome-wide loss of heterozygosity predicts aggressive, treatment-refractory behavior in pituitary neuroendocrine tumors. Acta Neuropathol 2024; 147:85. [PMID: 38758238 PMCID: PMC11101347 DOI: 10.1007/s00401-024-02736-8] [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: 02/21/2024] [Revised: 04/21/2024] [Accepted: 04/22/2024] [Indexed: 05/18/2024]
Abstract
Pituitary neuroendocrine tumors (PitNETs) exhibiting aggressive, treatment-refractory behavior are the rare subset that progress after surgery, conventional medical therapies, and an initial course of radiation and are characterized by unrelenting growth and/or metastatic dissemination. Two groups of patients with PitNETs were sequenced: a prospective group of patients (n = 66) who consented to sequencing prior to surgery and a retrospective group (n = 26) comprised of aggressive/higher risk PitNETs. A higher mutational burden and fraction of loss of heterozygosity (LOH) was found in the aggressive, treatment-refractory PitNETs compared to the benign tumors (p = 1.3 × 10-10 and p = 8.5 × 10-9, respectively). Within the corticotroph lineage, a characteristic pattern of recurrent chromosomal LOH in 12 specific chromosomes was associated with treatment-refractoriness (occurring in 11 of 14 treatment-refractory versus 1 of 14 benign corticotroph PitNETs, p = 1.7 × 10-4). Across the cohort, a higher fraction of LOH was identified in tumors with TP53 mutations (p = 3.3 × 10-8). A machine learning approach identified loss of heterozygosity as the most predictive variable for aggressive, treatment-refractory behavior, outperforming the most common gene-level alteration, TP53, with an accuracy of 0.88 (95% CI: 0.70-0.96). Aggressive, treatment-refractory PitNETs are characterized by significant aneuploidy due to widespread chromosomal LOH, most prominently in the corticotroph tumors. This LOH predicts treatment-refractoriness with high accuracy and represents a novel biomarker for this poorly defined PitNET category.
Collapse
Affiliation(s)
- Andrew L Lin
- Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Multidisciplinary Pituitary and Skull Base Tumor Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Vasilisa A Rudneva
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Allison L Richards
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yanming Zhang
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hyung Jun Woo
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Marc Cohen
- Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
- Multidisciplinary Pituitary and Skull Base Tumor Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jamie Tisnado
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nazanin Majd
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sharon L Wardlaw
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Gabrielle Page-Wilson
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Soma Sengupta
- Department of Neurology and Neurosurgery, University of North Carolina, Chapel Hill, NC, USA
| | - Frances Chow
- Department of Neurology, Keck School of Medicine at University of Southern California Medical Center, Los Angeles, CA, USA
| | - Bernard Goichot
- Department of Endocrinology, Les Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Byram H Ozer
- Department of Oncology, Sibley Memorial Hospital/Johns Hopkins, Washington, DC, USA
| | - Jorg Dietrich
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Lisa Nachtigall
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Arati Desai
- Department of Medicine, University of Pennsylvania Medical Center, Philadelphia, PA, USA
| | - Tina Alano
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Shahiba Ogilive
- Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - David B Solit
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Tejus A Bale
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Marc Rosenblum
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mark T A Donoghue
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Eliza B Geer
- Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA.
- Multidisciplinary Pituitary and Skull Base Tumor Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Viviane Tabar
- Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
- Multidisciplinary Pituitary and Skull Base Tumor Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
6
|
Lopes-Pinto M, Lacerda-Nobre E, Silva AL, Tortosa F, Marques P. The Role of Programmed Cell Death Ligand 1 Expression in Pituitary Tumours: Lessons from the Current Literature. Neuroendocrinology 2024; 114:709-720. [PMID: 38754394 DOI: 10.1159/000539345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Accepted: 05/13/2024] [Indexed: 05/18/2024]
Abstract
BACKGROUND Programmed cell death-1 (PD-1) and PD ligand-1 (PD-L1) expression predict the biological behaviour, aggressiveness, and response to immune checkpoint inhibitors in different cancers. We reviewed the published data on PD-L1 expression in pituitary tumours from the perspective of its biological role and prognostic usefulness. SUMMARY A literature review focused on PD-L1 expression in pituitary tumours was performed. Six immunohistochemistry-based studies which assessed PD-L1 positivity in pituitary tumours were included, encompassing 704 patients. The cohort consisted of 384 (54.5%) nonfunctioning tumours and 320 (43.5%) functioning pituitary tumours. PD-L1 expression was positive in 248 cases (35.2%). PD-L1 positivity rate was higher in functioning than in nonfunctioning tumours (46.3% vs. 26.0%; p < 0.001) but also higher in growth hormone-secreting tumours (56.7%) and prolactinomas (53.6%) than in thyrotroph (33.3%) or corticotroph tumours (20.6%). While proliferative pituitary tumours showed higher rate of PD-L1 positivity than non-proliferative tumours (p < 0.001), no association with invasion or recurrence was found. KEY MESSAGES PD-L1 is expressed in a substantial number of pituitary tumours, predominantly in the functioning ones. PD-L1 positivity rates were significantly higher in proliferative pituitary tumours in comparison to non-proliferative tumours, but no differences were found concerning invasive or recurrent pituitary tumours. More studies following homogeneous and standardised methodologies are needed to fully elucidate the role and usefulness of PD-L1 expression in pituitary tumours.
Collapse
Affiliation(s)
- Mariana Lopes-Pinto
- Endocrinology Department, Unidade Local de Saúde de Santa Maria, Hospital de Santa Maria, Lisbon, Portugal
| | - Ema Lacerda-Nobre
- Endocrinology Department, Unidade Local de Saúde de Santa Maria, Hospital de Santa Maria, Lisbon, Portugal
- Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Ana Luísa Silva
- Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Instituto de Saúde Ambiental da Faculdade de Medicina da Universidade de Lisboa (ISAMB-FMUL), Lisbon, Portugal
| | - Francisco Tortosa
- Pituitary Tumor Unit, Pathology Department, Hospital CUF Descobertas, Lisbon, Portugal
| | - Pedro Marques
- Pituitary Tumor Unit, Endocrinology Department, Hospital CUF Descobertas, Lisbon, Portugal
- Faculdade de Medicina, Universidade Católica Portuguesa, Lisbon, Portugal
| |
Collapse
|
7
|
Ilie MD, De Alcubierre D, Carretti AL, Jouanneau E, Raverot G. Therapeutic targeting of the pituitary tumor microenvironment. Pharmacol Ther 2023; 250:108506. [PMID: 37562699 DOI: 10.1016/j.pharmthera.2023.108506] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/28/2023] [Accepted: 08/07/2023] [Indexed: 08/12/2023]
Abstract
The tumor microenvironment (TME), the complex environment in which tumors develop, has been increasingly targeted for cancer treatment in recent years. Aggressive pituitary tumors and pituitary carcinomas have been so far targeted with immune-checkpoint inhibitors (28 cases, including a large cohort), and anti-angiogenic drugs (34 cases), specifically bevacizumab (30 cases), sunitinib (three cases), and apatinib (one case). Here, we reviewed all these cases, reporting tumor response, potential predictors of response, as well as adverse events. Given that the histological type could potentially influence treatment response, we present the existing data separately for each type. Briefly, under ICIs, complete response was noted in one case, partial response in a third of cases, stable disease in 10% of cases, while 54% of tumors progressed. Under BVZ monotherapy, most cases (57%) showed stable disease, while 36% of tumors progressed; partial response was reported in only one case. The three cases treated with sunitinib monotherapy progressed. Regarding predictive factors of response, the tumor type (aggressive pituitary tumor versus pituitary carcinoma) appears as the strongest predictor of response to ICIs. To date, no predictor of response to anti-angiogenic drugs in the treatment of pituitary carcinomas and aggressive pituitary tumors has been identified. The interest of BZV add-on to first- or second-line chemotherapy warrants further investigation. In addition, we discuss perspectives regarding the TME-targeting in aggressive pituitary tumors and pituitary carcinomas, including perspectives on immunotherapy, anti-angiogenic drugs, as well as on other TME components, namely stromal cells, extracellular matrix, and secreted molecules.
Collapse
Affiliation(s)
- Mirela-Diana Ilie
- Inserm U1052, CNRS UMR5286, Cancer Research Center of Lyon, Lyon, France; Lyon 1 University, Villeurbanne, France; Endocrinology Department, "C.I. Parhon" National Institute of Endocrinology, Bucharest, Romania
| | - Dario De Alcubierre
- Inserm U1052, CNRS UMR5286, Cancer Research Center of Lyon, Lyon, France; Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Anna Lucia Carretti
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy; Endocrinology Department, Reference Center for Rare Pituitary Diseases HYPO, "Groupement Hospitalier Est" Hospices Civils de Lyon, Bron, France
| | - Emmanuel Jouanneau
- Inserm U1052, CNRS UMR5286, Cancer Research Center of Lyon, Lyon, France; Lyon 1 University, Villeurbanne, France; Neurosurgery Department, Reference Center for Rare Pituitary Diseases HYPO, "Groupement Hospitalier Est" Hospices Civils de Lyon, Bron, France
| | - Gérald Raverot
- Inserm U1052, CNRS UMR5286, Cancer Research Center of Lyon, Lyon, France; Lyon 1 University, Villeurbanne, France; Endocrinology Department, Reference Center for Rare Pituitary Diseases HYPO, "Groupement Hospitalier Est" Hospices Civils de Lyon, Bron, France.
| |
Collapse
|
8
|
Shi M, Song Y, Zhang Y, Li L, Yu J, Hou A, Han S. PD-L1 and tumor-infiltrating CD8 + lymphocytes are correlated with clinical characteristics in pediatric and adolescent pituitary adenomas. Front Endocrinol (Lausanne) 2023; 14:1151714. [PMID: 37424874 PMCID: PMC10323746 DOI: 10.3389/fendo.2023.1151714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 05/23/2023] [Indexed: 07/11/2023] Open
Abstract
Objective To investigate the levels of tumor-infiltrating CD8+ lymphocytes (CD8+ TILs) and the expression of programmed cell death receptor ligand 1 (PD-L1) in the tumor microenvironment (TME) of pediatric and adolescent pituitary adenomas (PAPAs) and analyze the correlation between their levels and the clinical characteristics. Methods A series of 43 PAPAs cases were enrolled over a period of 5 years. To compare the TME of PAPAs and adult PAs, 43 PAPAs cases were matched with 60 adult PAs cases (30 cases were between 20 and 40 years old, and 30 cases were older than 40 years) for main clinical characteristics. The expression of immune markers in PAPAs was detected by immunohistochemistry, and their correlation with the clinical outcomes was analyzed using statistical methods. Results In the PAPAs group, CD8+ TILs level was significantly lower (3.4 (5.7) vs. 6.1 (8.5), p = 0.001), and PD-L1 expression (0.040 (0.022) vs. 0.024 (0.024), p < 0.0001) was significantly higher as compared with the older group. The level of CD8+ TILs was negatively correlated with the expression of PD-L1 (r = -0.312, p = 0.042). Moreover, CD8+ TILs and PD-L1 levels were associated with Hardy (CD8, p = 0.014; PD-L1, p = 0.018) and Knosp (CD8, p = 0.02; PD-L1, p = 0.017) classification. CD8+ TILs level was associated with high-risk adenomas (p = 0.015), and it was associated with the recurrence of PAPAs (HR = 0.047, 95% CI 0.003-0.632, p = 0.021). Conclusion Compared with the TME in adult PAs, the TME in PAPAs was found to express a significantly altered level of CD8+ TILs and PD-L1. In PAPAs, CD8+ TILs and PD-L1 levels were associated with clinical characteristics.
Collapse
Affiliation(s)
- Mengwu Shi
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, China
| | - Yifu Song
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, China
| | - Yaochuan Zhang
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, China
| | - Longjie Li
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, China
| | - Juanhan Yu
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Ana Hou
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Sheng Han
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
9
|
Burman P, Casar-Borota O, Perez-Rivas LG, Dekkers OM. Aggressive Pituitary Tumors and Pituitary Carcinomas: From Pathology to Treatment. J Clin Endocrinol Metab 2023; 108:1585-1601. [PMID: 36856733 PMCID: PMC10271233 DOI: 10.1210/clinem/dgad098] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 01/13/2023] [Accepted: 02/15/2023] [Indexed: 03/02/2023]
Abstract
Aggressive pituitary tumors (APTs) and pituitary carcinomas (PCs) are heterogeneous with regard to clinical presentation, proliferative markers, clinical course, and response to therapy. Half of them show an aggressive course only many years after the first apparently benign presentation. APTs and PCs share several properties, but a Ki67 index greater than or equal to 10% and extensive p53 expression are more prevalent in PCs. Mutations in TP53 and ATRX are the most common genetic alterations; their detection might be of value for early identification of aggressiveness. Treatment requires a multimodal approach including surgery, radiotherapy, and drugs. Temozolomide is the recommended first-line chemotherapy, with response rates of about 40%. Immune checkpoint inhibitors have emerged as second-line treatment in PCs, with currently no evidence for a superior effect of dual therapy compared to monotherapy with PD-1 blockers. Bevacizumab has resulted in partial response (PR) in few patients; tyrosine kinase inhibitors and everolimus have generally not been useful. The effect of peptide receptor radionuclide therapy is limited as well. Management of APT/PC is challenging and should be discussed within an expert team with consideration of clinical and pathological findings, age, and general condition of the patient. Considering that APT/PCs are rare, new therapies should preferably be evaluated in shared standardized protocols. Prognostic and predictive markers to guide treatment decisions are needed and are the scope of ongoing research.
Collapse
Affiliation(s)
- Pia Burman
- Department of Endocrinology, Skåne University Hospital, Lund
University, 205 02 Malmö, Sweden
| | - Olivera Casar-Borota
- Department of Immunology, Genetics, and Pathology; Uppsala
University, 751 85 Uppsala, Sweden
- Department of Clinical Pathology, Uppsala University
Hospital, 751 85 Uppsala, Sweden
| | - Luis Gustavo Perez-Rivas
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München,
Ludwig-Maximilians-Universität München, 80804
Munich, Germany
| | - Olaf M Dekkers
- Department of Internal Medicine (Section of Endocrinology & Clinical
Epidemiology), Leiden University Medical Centre, 2333 ZA
Leiden, The Netherlands
| |
Collapse
|
10
|
Geer EB. Medical therapy for refractory pituitary adenomas. Pituitary 2023:10.1007/s11102-023-01320-9. [PMID: 37115295 DOI: 10.1007/s11102-023-01320-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/11/2023] [Indexed: 04/29/2023]
Abstract
INTRODUCTION Refractory pituitary adenomas are those that have progressed following standard of care treatments. Medical therapy options for these challenging tumors are limited. PURPOSE To review the landscape of tumor directed medical therapies and off-label investigational approaches for refractory pituitary adenomas. METHODS Literature on medical therapies for refractory adenomas was reviewed. RESULTS The established first-line medical therapy for refractory adenomas is temozolomide, which importantly may increase survival, but clinical trial data are still needed to clearly establish its efficacy, identify biomarkers of response, and clarify eligibility and outcome criteria. Other therapies for refractory tumors have only been described in case reports and small case series. CONCLUSION There are currently no approved non-endocrine medical therapies for refractory pituitary tumors. There is an urgent need for identifying effective medical therapies and studying them in multi-center clinical trials.
Collapse
Affiliation(s)
- Eliza B Geer
- Departments of Medicine and Neurosurgery, Multidisciplinary Pituitary and Skull Base Tumor Center, Memorial Sloan Kettering Cancer Center, David H. Koch Center for Cancer Care, 530 East 74th Street, Box 19, New York, NY, 10021, USA.
| |
Collapse
|
11
|
Updates in neuroendocrine neoplasms: From mechanisms to the clinic. ANNALES D'ENDOCRINOLOGIE 2023; 84:291-297. [PMID: 36690074 DOI: 10.1016/j.ando.2022.12.424] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 12/04/2022] [Indexed: 01/22/2023]
Abstract
Scientific advances constantly improve our understanding of the mechanisms underlying tumorigenesis, allowing us now to analyze cancer in a more precise manner and to identify at an earlier stage the tumors that have greater risk of aggressive behavior. Understanding neuroendocrine neoplasms at molecular level has enabled increasingly targeted treatments, with safety and efficacy validated in large randomized trials. Moreover, the first studies of targeted therapies after molecular profiling of neuroendocrine neoplasms have shown encouraging results, allowing us to foresee ever more personalized medical treatments in the future. This literature review aims to summarize recent advances in the study of neuroendocrine neoplasms and to show how identification of new mechanisms underlying tumorigenesis can be of benefit in clinical practice.
Collapse
|
12
|
Abstract
After temozolomide failure, no evidence-based treatment option is currently available for aggressive pituitary tumors (APTs) and pituitary carcinomas (PCs). Moreover, once temozolomide has failed, the survival of these patients is very poor. The use of immune-checkpoint inhibitors (ICIs) has been so far reported in a large cohort, a small phase 2 clinical trial, and in another five isolated cases (24 cases in total). Here, we review the available evidence on the efficacy and potential predictors of response to ICIs in PCs and APTs, namely the histological type (corticotroph versus lactotroph), the tumor type (PC versus APT), the presence of uncontrolled endogenous hypercortisolism, the type of protocol (combined ICIs versus monotherapy), programmed death-ligand 1 (PD-L1) expression, CD8+ cell infiltration, tumor mutational burden (TMB), microsatellite instability (MSI), and mismatch repair (MMR) status. We also discuss key clinical aspects that can already be implemented in the everyday practice and identify future research needs.
Collapse
Affiliation(s)
- Gérald Raverot
- Inserm U1052, CNRS UMR5286, Claude Bernard Lyon 1 University, Cancer Research Center of Lyon, Lyon, France; Endocrinology Department, Reference Center for Rare Pituitary Diseases HYPO, "Groupement Hospitalier Est" Hospices Civils de Lyon, Bron, France.
| | - Mirela Diana Ilie
- Inserm U1052, CNRS UMR5286, Claude Bernard Lyon 1 University, Cancer Research Center of Lyon, Lyon, France; Endocrinology Department, "C.I. Parhon" National Institute of Endocrinology, Bucharest, Romania.
| |
Collapse
|
13
|
Ilie MD, Villa C, Cuny T, Cortet C, Assie G, Baussart B, Cancel M, Chanson P, Decoudier B, Deluche E, Di Stefano AL, Drui D, Gaillard S, Goichot B, Huillard O, Joncour A, Larrieu-Ciron D, Libe R, Nars G, Vasiljevic A, Raverot G. Real-life efficacy and predictors of response to immunotherapy in pituitary tumors: a cohort study. Eur J Endocrinol 2022; 187:685-696. [PMID: 36111659 DOI: 10.1530/eje-22-0647] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 09/15/2022] [Indexed: 11/08/2022]
Abstract
OBJECTIVE After temozolomide failure, no evidence-based treatment is available for pituitary carcinomas (PCs) and aggressive pituitary tumors (APTs). To date, only 12 cases treated with immune-checkpoint inhibitors (ICIs) have been published, showing encouraging efficacy. Predictive factors of response are lacking. Here, we aimed to assess the real-life efficacy and predictors of response to ICIs in PCs and APTs. DESIGN AND METHODS This study is a multicentric, retrospective, observational cohort study, including all PCs and APTs treated with ICIs in France up to March 2022. PD-L1 immunohistochemistry and CD8+ T cell infiltration were evaluated centrally. RESULTS Six PCs (four corticotroph and two lactotroph) and nine APTs (five corticotroph and four lactotroph) were included. The real-life efficacy of ICIs was lower than previously published data. Three corticotroph tumors (33.3%) showed partial response, one (11.1%) stable disease, while five (55.6%) progressed. One lactotroph tumor (16.7%) showed partial response, one (16.7%) stable disease, while four (66.7%) progressed. PCs responded far better than APTs, with 4/6 PCs showing partial response compared to 0/9 APTs. Corticotroph tumors responded slightly better than lactotroph tumors. In the four responsive corticotroph tumors, PD-L1 staining was negative and CD8+ T cell infiltration attained a maximum of 1% in the tumor center. CONCLUSIONS Confirmation of the presence or absence of metastases is necessary before starting ICIs. After temozolomide failure, ICIs appear as a good therapeutic option for PCs, especially for corticotroph carcinomas. Negative PD-L1 staining and very low CD8+ T cell infiltration in the tumor center should not preclude ICI administration in corticotroph carcinomas. SIGNIFICANCE STATEMENT This is the first study to assess the real-life efficacy of ICIs in pituitary carcinomas (PCs) and aggressive pituitary tumors. We also assessed potential predictors of response and are the first to assess the predictive value of CD8+ cell infiltration. We identified the tumor type as a major predictor, ICIs proving far more effective in treating PCs. Our study provides evidence that ICIs are a good option after temozolomide failure for PCs (four of six responded), especially for corticotroph carcinomas (three of four responded). We also provide evidence that negative PD-L1 staining and very low CD8+ cell infiltration in the tumor center should not preclude ICI administration in corticotroph carcinomas. Moreover, our findings point toward the need to systematically perform extension workup before starting ICIs.
Collapse
Affiliation(s)
- Mirela Diana Ilie
- Inserm U1052, CNRS UMR5286, Claude Bernard Lyon 1 University, Cancer Research Center of Lyon, Lyon, France
- Endocrinology Department, 'C.I. Parhon' National Institute of Endocrinology, Bucharest, Romania
| | - Chiara Villa
- Neuropathology Department, Pitié-Salpêtrière University Hospital, AP-HP-Sorbonne University, Paris, France
- Inserm U1016, CNRS UMR8104, Cochin Institute, Paris, France
| | - Thomas Cuny
- Endocrinology Department, Conception University Hospital, AP-HM, Marseille, France
- Inserm U1251, Marseille Medical Genetics, Aix Marseille University, Marseille, France
| | - Christine Cortet
- Endocrinology Department, Lille University Hospital, Lille, France
| | - Guillaume Assie
- Inserm U1016, CNRS UMR8104, Cochin Institute, Paris, France
- Endocrinology Department, Cochin University Hospital, AP-HP, Paris, France
| | - Bertrand Baussart
- Inserm U1016, CNRS UMR8104, Cochin Institute, Paris, France
- Neurosurgery Department, Pitié-Salpêtrière University Hospital, AP-HP, Paris, France
| | - Mathilde Cancel
- Oncology Department, Tours University Hospital, Tours, France
| | - Philippe Chanson
- Department of Endocrinology and Reproduction Disorders, Bicêtre Hospital, AP-HP, Le Kremlin-Bicêtre, France
- Paris-Saclay University, Inserm, 'Physiologie et Physiopathologie Endocriniennes', Le Kremlin-Bicêtre, France
| | | | - Elise Deluche
- Oncology Department, Limoges University Hospital, Limoges, France
| | - Anna Luisa Di Stefano
- Neurology Department, Foch Hospital, Suresnes, France
- Neurosurgery Unit, Livorno Hospital, Livorno, Italy
| | - Delphine Drui
- Endocrinology Department, Nantes University Hospital, Nantes, France
| | - Stephan Gaillard
- Neurosurgery Department, Pitié-Salpêtrière University Hospital, AP-HP, Paris, France
| | - Bernard Goichot
- Internal Medicine Department, Strasbourg University Hospital, Strasbourg, France
| | - Olivier Huillard
- Medical Oncology Department, Cochin University Hospital, AP-HP, Paris Cancer Institute CARPEM, Paris, France
| | - Anthony Joncour
- Oncology Department, Poitiers University Hospital, Poitiers, France
| | | | - Rossella Libe
- Inserm U1016, CNRS UMR8104, Cochin Institute, Paris, France
- Endocrinology Department, Cochin University Hospital, AP-HP, Paris, France
| | - Guillaume Nars
- Internal Medicine Department, Strasbourg University Hospital, Strasbourg, France
| | - Alexandre Vasiljevic
- Inserm U1052, CNRS UMR5286, Claude Bernard Lyon 1 University, Cancer Research Center of Lyon, Lyon, France
- Pathology Department
| | - Gérald Raverot
- Inserm U1052, CNRS UMR5286, Claude Bernard Lyon 1 University, Cancer Research Center of Lyon, Lyon, France
- Endocrinology Department, Reference Center for Rare Pituitary Diseases HYPO, 'Groupement Hospitalier Est' Hospices Civils de Lyon, Bron, France
| |
Collapse
|
14
|
Burman P, Trouillas J, Losa M, McCormack A, Petersenn S, Popovic V, Theodoropoulou M, Raverot G, Dekkers OM. Aggressive pituitary tumours and carcinomas, characteristics and management of 171 patients. Eur J Endocrinol 2022; 187:593-605. [PMID: 36018781 PMCID: PMC9513638 DOI: 10.1530/eje-22-0440] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Accepted: 08/26/2022] [Indexed: 11/08/2022]
Abstract
OBJECTIVE To describe clinical and pathological characteristics and treatment outcomes in a large cohort of aggressive pituitary tumours (APT)/pituitary carcinomas (PC). DESIGN Electronic survey August 2020-May 2021. RESULTS 96% of 171 (121 APT, 50 PC), initially presented as macro/giant tumours, 6 were microadenomas (5 corticotroph). Ninety-seven tumours, initially considered clinically benign, demonstrated aggressive behaviour after 5.5 years (IQR: 2.8-12). Of the patients, 63% were men. Adrenocorticotrophic hormone (ACTH)-secreting tumours constituted 30% of the APT/PC, and the gonadotroph subtypes were under-represented. Five out of 13 silent corticotroph tumours and 2/6 silent somatotroph tumours became secreting. Metastases were observed after median 6.3 years (IQR 3.7-12.1) from diagnosis. At the first surgery, the Ki67 index was ≥3% in 74/93 (80%) and ≥10% in 38/93 (41%) tumours. An absolute increase of Ki67 ≥ 10% after median of 6 years from the first surgery occurred in 18/49 examined tumours. Tumours with an aggressive course from outset had higher Ki67, mitotic counts, and p53. Temozolomide treatment in 156/171 patients resulted in complete response in 9.6%, partial response in 30.1%, stable disease in 28.1%, and progressive disease in 32.2% of the patients. Treatment with bevacizumab, immune checkpoint inhibitors, and peptide receptor radionuclide therapy resulted in partial regression in 1/10, 1/6, and 3/11, respectively. Median survival in APT and PC was 17.2 and 11.3 years, respectively. Tumours with Ki67 ≥ 10% and ACTH-secretion were associated with worse prognosis. CONCLUSION APT/PCs exhibit a wide and challenging spectrum of behaviour. Temozolomide is the first-line chemotherapy, and other oncological therapies are emerging. Treatment response continues to be difficult to predict with currently studied biomarkers.
Collapse
Affiliation(s)
- Pia Burman
- Department of Endocrinology, Skåne University Hospital Malmö, University of Lund, Lund, Sweden
- Correspondence should be addressed to P Burman;
| | | | - Marco Losa
- Marco Losa Department of Neurosurgery, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Ann McCormack
- St Vincent’s Hospital and Garvan Institute of Medical Research, Sydney, Australia
| | | | | | - Marily Theodoropoulou
- Medizinische Klinik und Poliklinik IV, LMU Klinikum, Ludwig-Maximilians-Universität München, Germany
| | - Gerald Raverot
- Fédération d’Endocrinologie, Groupement Hospitalier Est, Hospices Civils de Lyon, University of Lyon-Est de Lyon, Bron, France
| | - Olaf M Dekkers
- Department of Internal Medicine (Section Endocrinology) & Clinical Epidemiology, Leiden University Medical Centre, Leiden, The Netherlands
| | | |
Collapse
|
15
|
Immunotherapy for Aggressive and Metastatic Pituitary Neuroendocrine Tumors (PitNETs): State-of-the Art. Cancers (Basel) 2022; 14:cancers14174093. [PMID: 36077631 PMCID: PMC9454884 DOI: 10.3390/cancers14174093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/20/2022] [Accepted: 08/21/2022] [Indexed: 11/17/2022] Open
Abstract
Background: Aggressive and metastatic PitNETs are challenging conditions. Immune checkpoint inhibitors (ICIs) are currently considered in cases resistant to temozolomide (TMZ). However, clinical experience is essentially limited to case reports, with variable outcomes. Material and Methods: The effects of ICIs on 12 aggressive/metastatic PitNETs from the literature were reviewed and analyzed according to tumor characteristics, with the additional description of a silent-Pit1 metastatic tumor responding to pembrolizumab. Results: Most cases were metastatic (10/13: 6 corticotroph, 3 lactotroph, 1 silent Pit1); 3 were aggressive (2 corticotroph, 1 lactotroph). ICIS was used either as monotherapy or in combination. At last follow-up on ICI, a complete response (CR) was present in 3 cases and a partial response (PR) in 2 cases (4/5 metastatic). One sustained stable disease (SD) was reported. Progressive disease (PD) was observed in 7 cases, 3 of them after initial SD (n = 1) or PR (n = 3), with 2 reported deaths. PDL1 expression was studied in 10 cases and was high (>95%) in 2 Pit1-derived metastatic PitNETs (1 CR and 1 remarkable PR) but absent/low (<1%) in the remaining cases (including 1 CP and 2 PR). Elevated tumor mutation burden could be informative in corticotroph PitNETs, especially in mismatch repair-deficient tumors. Conclusion: Significant benefits from ICIs were documented in about half of TMZ-resistant PitNETS. High PDL1 expression was associated with remarkable responses but may be dispensable. Based on their acceptable tolerance and awaiting recognized predictors of response, ICIs may be considered a valuable option for such patients.
Collapse
|